Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 327
Filtrer
1.
Mol Ther Oncol ; 32(3): 200821, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39021370

RÉSUMÉ

Bispecific T cell engagers are a promising class of therapeutic proteins for cancer therapy. Their potency and small size often come with systemic toxicity and short half-life, making intravenous administration cumbersome. These limitations can be overcome by tumor-specific in situ expression, allowing high local accumulation while reducing systemic concentrations. However, encoding T cell engagers in viral or non-viral vectors and expressing them in situ ablates all forms of quality control performed during recombinant protein production. It is therefore vital to design constructs that feature minimal domain mispairing, and increased homogeneity of the therapeutic product. Here, we report a T cell engager architecture specifically designed for vector-mediated immunotherapy. It is based on a fusion of a designed ankyrin repeat protein (DARPin) to a CD3-targeting single-chain antibody fragment, termed DATE (DARPin-fused T cell Engager). The DATE induces potent T cell-mediated killing of HER2+ cancer cells, both as recombinantly produced therapeutic protein and as in situ expressed payload from a HER2+-retargeted high-capacity adenoviral vector (HC-AdV). We report remarkable tumor remission, DATE accumulation, and T cell infiltration through in situ expression mediated by a HER2+-retargeted HC-AdV in vivo. Our results support further investigations and developments of DATEs as payloads for vector-mediated immunotherapy.

2.
J Neural Eng ; 21(4)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38885674

RÉSUMÉ

Objective.To develop a clinically relevant injectable hydrogel derived from decellularized porcine peripheral nerves and with mechanical properties comparable to native central nervous system (CNS) tissue to be used as a delivery vehicle for Schwann cell transplantation to treat spinal cord injury (SCI).Approach.Porcine peripheral nerves (sciatic and peroneal) were decellularized by chemical decellularization using a sodium deoxycholate and DNase (SDD) method previously developed by our group. The decellularized nerves were delipidated using dichloromethane and ethanol solvent and then digested using pepsin enzyme to form injectable hydrogel formulations. Genipin was used as a crosslinker to enhance mechanical properties. The injectability, mechanical properties, and gelation kinetics of the hydrogels were further analyzed using rheology. Schwann cells encapsulated within the injectable hydrogel formulations were passed through a 25-gauge needle and cell viability was assessed using live/dead staining. The ability of the hydrogel to maintain Schwann cell viability against an inflammatory milieu was assessedin vitrousing inflamed astrocytes co-cultured with Schwann cells.Mainresults. The SDD method effectively removes cells and retains extracellular matrix in decellularized tissues. Using rheological studies, we found that delipidation of decellularized porcine peripheral nerves using dichloromethane and ethanol solvent improves gelation kinetics and mechanical strength of hydrogels. The delipidated and decellularized hydrogels crosslinked using genipin mimicked the mechanical strength of CNS tissue. The hydrogels were found to have shear thinning properties desirable for injectable formulations and they also maintained higher Schwann cell viability during injection compared to saline controls. Usingin vitroco-culture experiments, we found that the genipin-crosslinked hydrogels also protected Schwann cells from astrocyte-mediated inflammation.Significance. Injectable hydrogels developed using delipidated and decellularized porcine peripheral nerves are a potential clinically relevant solution to deliver Schwann cells, and possibly other therapeutic cells, at the SCI site by maintaining higher cellular viability and increasing therapeutic efficacy for SCI treatment.


Sujet(s)
Hydrogels , Nerfs périphériques , Cellules de Schwann , Traumatismes de la moelle épinière , Animaux , Cellules de Schwann/physiologie , Cellules de Schwann/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Hydrogels/administration et posologie , Suidae , Traumatismes de la moelle épinière/thérapie , Nerfs périphériques/physiologie , Nerfs périphériques/effets des médicaments et des substances chimiques , Régénération de la moelle épinière/physiologie , Régénération de la moelle épinière/effets des médicaments et des substances chimiques , Cellules cultivées , Survie cellulaire/physiologie , Survie cellulaire/effets des médicaments et des substances chimiques
3.
Int J Pharm ; 660: 124332, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-38866085

RÉSUMÉ

Surface functionalization of nano drug carriers allows for precise delivery of therapeutic molecules to the target site. This technique involves attaching targeting molecules to the nanoparticle surface, facilitating selective interaction. In this study, we engineered virus-like particles (VLPs) to enhance their targeting capabilities. Azide groups incorporated on the lipid membranes of VLPs enabled bioorthogonal click reactions for conjugation with cycloalkyne-bearing molecules, providing efficient conjugation with high specificity. HIV-1 Gag VLPs were chosen due to their envelope, which allows host membrane component incorporation, and the Gag protein, which serves as a recognition motif for human T cells. This combination, along with antibody-mediated targeting, addresses the limitations of intracellular delivery to T cells, which typically exhibit low uptake of exogenous materials. The selective uptake of azide VLPs by CD3-positive T cells was evaluated in a co-culture system. Even without antibody conjugation, VLP uptake was enhanced in T cells, indicating their intrinsic targeting potential. Antibody conjugation further amplified this effect, demonstrating the synergistic benefits of the combined targeting approach. Our study shows that recombinant production of azide functionalized VLPs results in engineered nanoparticles that can be easily modified using bioorthogonal click reactions, providing high specificity and versatility for conjugation with various molecules, making it applicable to a wide range of biological products.


Sujet(s)
Azotures , Chimie click , Lymphocytes T , Humains , Azotures/composition chimique , Lymphocytes T/immunologie , Nanoparticules/composition chimique , Produits du gène gag du virus de l'immunodéficience humaine , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Techniques de coculture , Systèmes de délivrance de médicaments , Propriétés de surface
4.
Sci Rep ; 14(1): 14665, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918510

RÉSUMÉ

Delivery of therapeutic stem cells to treat bone tissue damage is a promising strategy that faces many hurdles to clinical translation. Among them is the design of a delivery vehicle which promotes desired cell behavior for new bone formation. In this work, we describe the use of an injectable microporous hydrogel, made of crosslinked gelatin microgels, for the encapsulation and delivery of human mesenchymal stem cells (MSCs) and compared it to a traditional nonporous injectable hydrogel. MSCs encapsulated in the microporous hydrogel showed rapid cell spreading with direct cell-cell connections whereas the MSCs in the nonporous hydrogel were entrapped by the surrounding polymer mesh and isolated from each other. On a per-cell basis, encapsulation in microporous hydrogel induced a 4 × increase in alkaline phosphatase (ALP) activity and calcium mineral deposition in comparison to nonporous hydrogel, as measured by ALP and calcium assays, which indicates more robust osteogenic differentiation. RNA-seq confirmed the upregulation of the genes and pathways that are associated with cell spreading and cell-cell connections, as well as the osteogenesis in the microporous hydrogel. These results demonstrate that microgel-based injectable hydrogels can be useful tools for therapeutic cell delivery for bone tissue repair.


Sujet(s)
Différenciation cellulaire , Hydrogels , Cellules souches mésenchymateuses , Ostéogenèse , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Humains , Hydrogels/composition chimique , Porosité , Phosphatase alcaline/métabolisme , Cellules cultivées , Encapsulation de cellules/méthodes , Transplantation de cellules souches mésenchymateuses/méthodes , Injections
5.
Adv Mater ; 36(30): e2401667, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38843541

RÉSUMÉ

The efficacy of adoptive T cell therapy (ACT) for the treatment of solid tumors remains challenging. In addition to the poor infiltration of effector T (Teff) cells limited by the physical barrier surrounding the solid tumor, another major obstacle is the extensive infiltration of regulatory T (Treg) cells, a major immunosuppressive immune cell subset, in the tumor microenvironment. Here, this work develops a grooved microneedle patch for augmenting ACT, aiming to simultaneously overcome physical and immunosuppressive barriers. The microneedles are engineered through an ice-templated method to generate the grooved structure for sufficient T-cell loading. In addition, with the surface modification of chemokine CCL22, the MNs could not only directly deliver tumor-specific T cells into solid tumors through physical penetration, but also specifically divert Treg cells from the tumor microenvironment to the surface of the microneedles via a cytokine concentration gradient, leading to an increase in the ratio of Teff cells/Treg cells in a mouse melanoma model. Consequently, this local delivery strategy of both T cell receptor T cells and chimeric antigen receptor T cells via the CCL22-modified grooved microneedles as a local niche could significantly enhance the antitumor efficacy and reduce the on-target off-tumor toxicity of ACT.


Sujet(s)
Immunothérapie adoptive , Aiguilles , Lymphocytes T régulateurs , Animaux , Lymphocytes T régulateurs/immunologie , Souris , Immunothérapie adoptive/méthodes , Microenvironnement tumoral , Lignée cellulaire tumorale , Chimiokine CCL22/métabolisme , Humains , Souris de lignée C57BL , Tumeurs/thérapie , Tumeurs/immunologie
6.
Nanomicro Lett ; 16(1): 218, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38884868

RÉSUMÉ

Microgels prepared from natural or synthetic hydrogel materials have aroused extensive attention as multifunctional cells or drug carriers, that are promising for tissue engineering and regenerative medicine. Microgels can also be aggregated into microporous scaffolds, promoting cell infiltration and proliferation for tissue repair. This review gives an overview of recent developments in the fabrication techniques and applications of microgels. A series of conventional and novel strategies including emulsification, microfluidic, lithography, electrospray, centrifugation, gas-shearing, three-dimensional bioprinting, etc. are discussed in depth. The characteristics and applications of microgels and microgel-based scaffolds for cell culture and delivery are elaborated with an emphasis on the advantages of these carriers in cell therapy. Additionally, we expound on the ongoing and foreseeable applications and current limitations of microgels and their aggregate in the field of biomedical engineering. Through stimulating innovative ideas, the present review paves new avenues for expanding the application of microgels in cell delivery techniques.

7.
Adv Healthc Mater ; 13(19): e2304397, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38684223

RÉSUMÉ

A zwitterionic injectable and degradable hydrogel based on hydrazide and aldehyde-functionalized [2-(methacryloyloxy)ethyl] dimethyl-(3-sulfopropyl)ammonium hydroxide (DMAPS) precursor polymers that can address practical in vivo needs is reported. Zwitterion fusion interactions between the zwitterionic precursor polymers create a secondary physically crosslinked network to enable much more rapid gelation than previously reported with other synthetic polymers, facilitating rapid gelation at much lower polymer concentrations or degrees of functionalization than previously accessible in addition to promoting zero swelling and long-term degradation responses and significantly stiffer mechanics than are typically accessed with previously reported low-viscosity precursor gelation systems. The hydrogels maintain the highly anti-fouling properties of conventional zwitterionic hydrogels against proteins, mammalian cells, and bacteria while also promoting anti-fibrotic tissue responses in vivo. Furthermore, the use of the hydrogels for effective delivery and subsequent controlled release of viable cells with tunable profiles both in vitro and in vivo is demonstrated, including the delivery of myoblasts in a mouse skeletal muscle defect model for reducing the time between injury and functional mobility recovery. The combination of the injectability, degradability, and tissue compatibility achieved offers the potential to expand the utility of zwitterionic hydrogels in minimally invasive therapeutic applications.


Sujet(s)
Hydrogels , Hydrogels/composition chimique , Hydrogels/pharmacologie , Animaux , Souris , Régénération/effets des médicaments et des substances chimiques , Réactifs réticulants/composition chimique , Muscles squelettiques/effets des médicaments et des substances chimiques , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Myoblastes/effets des médicaments et des substances chimiques , Myoblastes/cytologie
8.
Bioengineering (Basel) ; 11(4)2024 Apr 13.
Article de Anglais | MEDLINE | ID: mdl-38671796

RÉSUMÉ

Mesenchymal stromal cells (MSCs) have displayed potential in regenerating organ function due to their anti-fibrotic, anti-inflammatory, and regenerative properties. However, there is a need for delivery systems to enhance MSC retention while maintaining their anti-fibrotic characteristics. This study investigates the feasibility of using alginate hydrogel microstrands as a cell delivery vehicle to maintain MSC viability and phenotype. To accommodate cell implantation needs, we invented a Syringe-in-Syringe approach to reproducibly fabricate microstrands in small numbers with a diameter of around 200 µm and a porous structure, which would allow for transporting nutrients to cells by diffusion. Using murine NIH 3T3 fibroblasts and primary embryonic 16 (E16) salivary mesenchyme cells as primary stromal cell models, we assessed cell viability, growth, and expression of mesenchymal and fibrotic markers in microstrands. Cell viability remained higher than 90% for both cell types. To determine cell number within the microstrands prior to in vivo implantation, we have further optimized the alamarBlue assay to measure viable cell growth in microstrands. We have shown the effect of initial cell seeding density and culture period on cell viability and growth to accommodate future stromal cell delivery and implantation. Additionally, we confirmed homeostatic phenotype maintenance for E16 mesenchyme cells in microstrands.

9.
Cureus ; 16(3): e56958, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38665717

RÉSUMÉ

Corneal endothelial dysfunction poses significant challenges in ophthalmology, leading to corneal edema and vision loss. Traditional treatments, including corneal transplantation, are limited by donor scarcity and potential complications. Nanoparticle-mediated cell delivery emerges as a promising approach for corneal endothelial regeneration, offering targeted and minimally invasive solutions. This comprehensive review provides insights into the role of nanoparticles in enhancing cell survival, integration, and therapeutic efficacy. We discuss the current understanding of corneal endothelial dysfunction, emphasizing the importance of regeneration. Furthermore, we explore the potential implications of nanoparticle-mediated approaches in clinical practice, highlighting opportunities for personalized treatment strategies. Future directions are also discussed, including optimization of nanoparticle design and exploration of combination therapies. Overall, this review elucidates the promising advancements in nanoparticle-mediated cell delivery for corneal endothelial regeneration and underscores the importance of continued research efforts in this evolving field.

10.
Protein Sci ; 33(4): e4944, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38501479

RÉSUMÉ

Antibody (Ab)-based drugs have been widely used in targeted therapies and immunotherapies, leading to significant improvements in tumor therapy. However, the failure of Ab therapy due to the loss of target antigens or Ab modifications that affect its function limits its application. In this study, we expanded the application of antibodies (Abs) by constructing a fusion protein as a versatile tool for Ab-based target cell detection, delivery, and therapy. We first constructed a SpaC Catcher (SpaCC for short) fusion protein that included the C domains of Staphylococcal protein A (SpaC) and the SpyCatcher. SpaCC conjugated with SpyTag-X (S-X) to form the SpaCC-S-X complex, which binds non-covalently to an Ab to form the Ab-SpaCC-S-X protein complex. The "X" can be a variety of small molecules such as fluoresceins, cell-penetrating peptide TAT, Monomethyl auristatin E (MMAE), and DNA. We found that Ab-SpaCC-S-FITC(-TAT) could be used for target cell detection and delivery. Besides, we synthesized the Ab-SpaCC-SN3-MMAE complex by linking Ab with MMAE by SpaCC, which improved the cytotoxicity of small molecule toxins. Moreover, we constructed an Ab-DNA complex by conjugating SpaCC with the aptamer (Ap) and found that Ab-SpaCC-SN3-Ap boosted the tumor-killing function of T-cells by retargeting tumor cells. Thus, we developed a multifunctional tool that could be used for targeted therapies and immunotherapies, providing a cheap and convenient novel drug development strategy.


Sujet(s)
Peptides de pénétration cellulaire , Tumeurs , Humains , Tumeurs/génétique , Tumeurs/thérapie , Immunothérapie , Anticorps , ADN , Lignée cellulaire tumorale
11.
Nanomedicine ; 57: 102742, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38460654

RÉSUMÉ

Modification of T-lymphocytes, which are capable of paracellular transmigration is a promising trend in modern personalized medicine. However, the delivery of required concentrations of functionalized T-cells to the target tissues remains a problem. We describe a novel method to functionalize T-cells with magnetic nanocapsules and target them with electromagnetic tweezers. T-cells were modified with the following magnetic capsules: Parg/DEX (150 nm), BSA/TA (300 nm), and BSA/TA (500 nm). T-cells were magnetonavigated in a phantom blood vessel capillary in cultural medium and in whole blood. The permeability of tumor tissues to captured T-cells was analyzed by magnetic delivery of modified T-cells to spheroids formed from 4T1 breast cancer cells. The dynamics of T-cell motion under a magnetic field gradient in model environments were analyzed by particle image velocimetry. The magnetic properties of the nanocomposite capsules and magnetic T-cells were measured. The obtained results are promising for biomedical applications in cancer immunotherapy.


Sujet(s)
Nanocapsules , Nanocomposites , Systèmes de délivrance de médicaments/méthodes , Lymphocytes T , Phénomènes électromagnétiques , Capsules
12.
Small Methods ; 8(7): e2301495, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38308323

RÉSUMÉ

Field-driven transport systems offer great promise for use as biofunctionalized carriers in microrobotics, biomedicine, and cell delivery applications. Despite the construction of artificial microtubules using several micromagnets, which provide a promising transport pathway for the synchronous delivery of microrobotic carriers to the targeted location inside microvascular networks, the selective transport of different microrobotic carriers remains an unexplored challenge. This study demonstrated the selective manipulation and transport of microrobotics along a patterned micromagnet using applied magnetic fields. Owing to varied field strengths, the magnetic beads used as the microrobotic carriers with different sizes revealed varied locomotion, including all of them moving along the same direction, selective rotation, bidirectional locomotion, and all of them moving in a reversed direction. Furthermore, cells immobilized with magnetic beads and nanoparticles also revealed varied locomotion. It is expected that such steering strategies of microrobotic carriers can be used in microvascular channels for the targeted delivery of drugs or cells in an organized manner.

13.
Small Methods ; 8(7): e2301197, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38376006

RÉSUMÉ

Safe and accurate in situ delivery of biocompatible materials is a fundamental requirement for many biomedical applications. These include sustained and local drug release, implantation of acellular biocompatible scaffolds, and transplantation of cells and engineered tissues for functional restoration of damaged tissues and organs. The common practice today includes highly invasive operations with major risks of surgical complications including adjacent tissue damage, infections, and long healing periods. In this work, a novel non-invasive delivery method is presented for scaffold, cells, and drug delivery deep into the body to target inner tissues. This technology is based on acousto-sensitive materials which are polymerized by ultrasound induction through an external transducer in a rapid and local fashion without additional photoinitiators or precursors. The applicability of this technology is demonstrated for viable and functional cell delivery, for drug delivery with sustained release profiles, and for 3D printing. Moreover, the mechanical properties of the delivered scaffold can be tuned to the desired target tissue as well as controlling the drug release profile. This promising technology may shift the paradigm for local and non-invasive material delivery approach in many clinical applications as well as a new printing method - "acousto-printing" for 3D printing and in situ bioprinting.


Sujet(s)
Matériaux biocompatibles , Systèmes de délivrance de médicaments , Polymérisation , Impression tridimensionnelle , Structures d'échafaudage tissulaires , Structures d'échafaudage tissulaires/composition chimique , Humains , Matériaux biocompatibles/composition chimique , Ingénierie tissulaire , Animaux , Ondes ultrasonores , Souris
14.
Discov Med ; 36(181): 294-307, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38409835

RÉSUMÉ

BACKGROUND: Stem cell-based therapies display immense potential in regenerative medicine, highlighting the crucial significance of devising efficient delivery methods. This study centers on a pioneering approach that utilizes Pluronic F127 (PF127) as a thermoresponsive and injectable hydrogel designed for the encapsulation of adipose-derived mesenchymal stem cells (AdMSCs). METHODS: The degradation profile, gelation time, and microstructure of the PF127 hydrogel were thoroughly examined. AdMSCs were isolated, expanded, and characterized based on their multi-lineage differentiation potential. AdMSCs from the third passage were specifically employed for encapsulation within the PF127 hydrogel. Subsequently, the cytotoxicity of the AdMSC-loaded PF127 hydrogel was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and apoptosis assays. RESULTS: Characterized by scanning electron microscopy (SEM), the PF127 hydrogel exhibited a porous structure, indicating its suitability for accommodating AdMSCs and facilitating wound healing. The PF127 hydrogel demonstrated reversible phase transitions, rendering it suitable for in vivo applications. Studies on the gelation time of PF127 hydrogel unveiled a concentration-dependent decrease in gelation time, offering adaptability for diverse medical applications. Analysis of the degradation profile showcased a seven-day degradation period, leading to the decision for weekly topical applications. Cytotoxicity assessments confirmed that AdMSCs loaded into the PF127 hydrogel maintained heightened metabolic activity for up to one week, affirming the safety and appropriateness of the PF127 hydrogel for encapsulating cellular therapeutics. Furthermore, cell apoptosis assays consistently indicated low rates of apoptosis, emphasizing the viability and robust health of AdMSCs when delivered within the hydrogel. CONCLUSIONS: These findings underscore the vast potential of PF127 hydrogel as a versatile and biocompatible delivery system for AdMSCs in the realm of regenerative medicine. Boasting adjustable gelation properties and a remarkable capacity for cell encapsulation, this pioneering delivery system presents a promising path for applications in tissue engineering and wound healing. Ultimately, these advancements propel and elevate the landscape of regenerative medicine.


Sujet(s)
Hydrogels , Cellules souches mésenchymateuses , Humains , Hydrogels/composition chimique , Poloxamère/composition chimique
15.
Polymers (Basel) ; 16(1)2024 Jan 04.
Article de Anglais | MEDLINE | ID: mdl-38201830

RÉSUMÉ

Poly(lactic-glycolic acid) (PLGA) is a biocompatible bio-scaffold material, but its own hydrophobic and electrically neutral surface limits its application as a cell scaffold. Polymer materials, mimics ECM materials, and organic material have often been used as coating materials for PLGA cell scaffolds to improve the poor cell adhesion of PLGA and enhance tissue adaptation. These coating materials can be modified on the PLGA surface via simple physical or chemical methods, and coating multiple materials can simultaneously confer different functions to the PLGA scaffold; not only does this ensure stronger cell adhesion but it also modulates cell behavior and function. This approach to coating could facilitate the production of more PLGA-based cell scaffolds. This review focuses on the PLGA surface-modified materials, methods, and applications, and will provide guidance for PLGA surface modification.

16.
Int J Pharm ; 652: 123801, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38244647

RÉSUMÉ

Immune cell delivery using injectable hydrogel attracts much attention for improving its therapeutic effect. Specifically, dendritic cells (DCs) are the trigger cells for immune responses, and DC vaccines are studied for improving cancer immunotherapy. Hydrogel-assisted cell delivery is expected to enhance the viability of the implanted cells. We recently reported temperature-responsive biodegradable injectable polymer (IP) formulation utilizing poly(ε-caprolactone-co-glycolide)-b-poly(ethylene glycol)(PEG)-b-poly(ε-caprolactone-co-glycolide) (tri-PCG). Tri-PCG-based IP was reported to exhibit immediate sol-to-gel transition in response to temperature increase, in vivo biodegradability, and excellent biocompatibility. In this study, tri-PCG-based IP was applied to DC delivery. IP encapsulated live DCs, and the DCs incorporated ovalbumin (OVA) as a model antigen and CpG-DNA (oligo DNA with adjuvant effect) in IP hydrogel. Results suggested that DCs encapsulated in IP hydrogel internalized OVA and CpG-DNA and DCs were maturated to present antigens to T cells. Moreover, subcutaneously injected tri-PCG-based IP prolonged the retention period of cell accumulation at injected sites. Tri-PCG IP hydrogel could release matured DCs as the degradation of the hydrogel progressed. Tri-PCG IP formulation improved treatment efficacy of OVA transfected mouse lymphoma (E.G7-OVA) tumor. Hence, tri-PCG IP is a promising platform for immune cell delivery.


Sujet(s)
Caproates , Lactones , Tumeurs , Polymères , Souris , Animaux , Polymères/métabolisme , Polyéthylène glycols , Hydrogels , Antigènes , ADN , Cellules dendritiques , Immunothérapie , Tumeurs/thérapie
17.
Adv Sci (Weinh) ; 11(13): e2307030, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38279587

RÉSUMÉ

Allogeneic tumor cell vaccines provide off-the-shelf convenience but lack patient specificity due to heterogeneity in tumor antigens. Here, allogeneic tumor cell corpses are converted into "zombie cells" capable of assimilating heterogeneous tumor by seizing cancer cells and spreading adjuvant infection. This causes pseudo-oncolysis of tumors, transforming them into immunogenic targets for enhanced phagocytosis. It is shown that in postoperative tumor models, localized delivery of premade "zombie cells" through stepwise gelation in resection cavity consolidates tumor surgery. Compared to analogous vaccines lacking "seizing" or "assimilating" capability, "zombie cell" platform effectively mobilizes T cell response against residual tumors, and establishes immunological memory against tumor re-challenge, showing less susceptibility to immune evasion. Despite using allogeneic sources, "zombie cell" platform functions as generalizable framework to produce long-term antitumor immunity in different tumor models, showing comparable effect to autologous vaccine. Together, with the potential of off-the-shelf availability and personalized relevance to heterogenous tumor antigens, this study suggests an alternative strategy for timely therapy after tumor surgery.


Sujet(s)
Vaccins anticancéreux , Transplantation de cellules souches hématopoïétiques , Tumeurs , Humains , Tumeurs/thérapie , Immunothérapie , Antigènes néoplasiques
18.
Macromol Biosci ; 24(3): e2300475, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37955619

RÉSUMÉ

Hydrogels loaded with biologics hold great potential for various biomedical applications such as regenerative medicine. However, biologics may lose bioactivity during hydrogel preparation, shipping, and storage. While many injectable hydrogels do not have this issue, they face a dilemma between fast gelation causing the difficulty of injection and slow gelation causing the escape of solutions from an injection site. The purpose of this study is to develop an affinity hydrogel by integrating a pre-formed elastic macroporous matrix and an injectable hydrogel. The data shows that the macroporous hydrogel matrix can hold a large volume of solutions for the formation of in situ injectable hydrogels loaded with growth factors or living cells. The cells can proliferate in the composite hydrogels. The growth factors can be stably sequestered and sustainably released due to the presence of aptamers. When both living cells and growth factors are loaded together into the hydrogels, cells can proliferate under culture conditions with a reduced serum level. Therefore, a macroporous and elastic matrix-supported formation of aptamer-functionalized injectable hydrogels is a promising method for developing the carriers of biologics.


Sujet(s)
Produits biologiques , Hydrogels , Hydrogels/pharmacologie , Protéines et peptides de signalisation intercellulaire , Médecine régénérative , Matrice extracellulaire
19.
Macromol Biosci ; 24(1): e2200562, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-36974501

RÉSUMÉ

Scaffold-based cell delivery can improve therapeutic effects of transplanted cells in cell therapy. Biomaterial scaffolds serveas niche for cell growth and proliferation which improves cell survival and overall function post cell delivery. In this study, gelatin methacryloyl based injectable scaffolds made using poly(ethylene)glycol as a sacrificial polymer and cryogelation as a technique, are demonstrated to have tunable degradability and porosity that is required for cell and drug delivery applications. The pore size (10-142 µm) of these gels makes them suitable for loading different cell types as per the application. In vitro studies using mammalian cells confirm that these cryogels are cytocompatible. These cell-laden scaffolds are injectable and have a cell retention ability of up to 90% after injection. Rheology is done to evaluate stiffness and shape recovery property, and it is found that these gels can maintain their original shape even after applying 7 cycles of strain from 0.1% to 20%. Furthermore, their degradability can be modulated between 6 and 10 days by changing the overall polymer composition. Thus, injectability and degradability of these cryogels can circumvent invasive surgical procedures, thereby making them useful for a variety of applications including delivery of cells and bioactive factors.


Sujet(s)
Matériaux biocompatibles , Cryogels , Méthacrylates , Animaux , Cryogels/pharmacologie , Matériaux biocompatibles/pharmacologie , Gélatine/pharmacologie , Gels/pharmacologie , Polymères/pharmacologie , Structures d'échafaudage tissulaires , Porosité , Ingénierie tissulaire/méthodes , Prolifération cellulaire , Mammifères
20.
Adv Mater ; 36(8): e2309332, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37934114

RÉSUMÉ

Bacterial-based delivery strategies have recently emerged as a unique research direction in the field of drug delivery. However, bacterial vectors are quickly phagocytosed by immune cells after entering the bloodstream. Taking advantage of this phenomenon, herein, this work seeks to harness the potential of immune cells to delivery micron-sized bacterial vectors, and find that inactivated bacterial can accumulate at tumor-site after intravenous injection through CD11b+ cells hitchhiking. To this end, this work then designs a gold-platinum bimetallic nanozyme coated bacterial vector (Au-Pt@VNP20009, APV). Utilizing strong tumor inflammatory response induced by low dose X-rays, this work further heightens the ability of CD11b+ immune cells to assist APV hitchhiking for tumor-targeted delivery, which can significantly relieve tumor hypoxia and immunosuppression, and inhibit tumor growth and metastasis. This work elucidates the potential mechanisms of bacterial vector targeted delivery, opening up new horizons for bacterial vector delivery strategies and clinical tumor radioimmunotherapy.


Sujet(s)
Tumeurs , Radioimmunothérapie , Humains , Bactéries , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments , Immunothérapie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE