Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Expert Opin Drug Deliv ; 20(11): 1531-1552, 2023.
Article in English | MEDLINE | ID: mdl-37946533

ABSTRACT

INTRODUCTION: Numerous delivery strategies, primarily novel nucleic acid delivery carriers, have been developed and explored to enable therapeutically relevant lung gene therapy. However, its clinical translation is yet to be achieved despite over 30 years of efforts, which is attributed to the inability to overcome a series of biological barriers that hamper efficient nucleic acid transfer to target cells in the lung. AREAS COVERED: This review is initiated with the fundamentals of nucleic acid therapy and a brief overview of previous and ongoing efforts on clinical translation of lung gene therapy. We then walk through the nature of biological barriers encountered by nucleic acid carriers administered via respiratory and/or systemic routes. Finally, we introduce advanced strategies developed to overcome those barriers to achieve therapeutically relevant nucleic acid delivery efficiency in the lung. EXPERT OPINION: We are now stepping close to the clinical translation of lung gene therapy, thanks to the discovery of novel delivery strategies that overcome biological barriers via comprehensive preclinical studies. However, preclinical findings should be cautiously interpreted and validated to ultimately realize meaningful therapeutic outcomes with newly developed delivery strategies in humans. In particular, individual strategies should be selected, tailored, and implemented in a manner directly relevant to specific therapeutic applications and goals.


Subject(s)
Nucleic Acids , Humans , Genetic Therapy , Lung , Drug Delivery Systems
2.
ACS Biomater Sci Eng ; 9(8): 4567-4572, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37523785

ABSTRACT

We here introduce a novel bioreducible polymer-based gene delivery platform enabling widespread transgene expression in multiple brain regions with therapeutic relevance following intracranial convection-enhanced delivery. Our bioreducible nanoparticles provide markedly enhanced gene delivery efficacy in vitro and in vivo compared to nonbiodegradable nanoparticles primarily due to the ability to release gene payloads preferentially inside cells. Remarkably, our platform exhibits competitive gene delivery efficacy in a neuron-rich brain region compared to a viral vector under previous and current clinical investigations with demonstrated positive outcomes. Thus, our platform may serve as an attractive alternative for the intracranial gene therapy of neurological disorders.


Subject(s)
Gene Transfer Techniques , Polymers , Polymers/metabolism , Genetic Therapy , Brain/metabolism
3.
J Extracell Vesicles ; 12(6): e12324, 2023 06.
Article in English | MEDLINE | ID: mdl-37272896

ABSTRACT

Adeno-associated virus (AAV) vector has shown multiple clinical breakthroughs, but its clinical implementation in inhaled gene therapy remains elusive due to difficulty in transducing lung airway cells. We demonstrate here AAV serotype 6 (AAV6) associated with extracellular vesicles (EVs) and secreted from vector-producing HEK-293 cells during vector preparation (EVAAV6) as a safe and highly efficacious gene delivery platform for inhaled gene therapy applications. Specifically, we discovered that EVAAV6 provided markedly enhanced reporter transgene expression in mucus-covered air-liquid interface (ALI) cultures of primary human bronchial and nasal epithelial cells as well as in mouse lung airways compared to standard preparations of AAV6 alone. Of note, AAV6 has been previously shown to outperform other clinically tested AAV serotypes, including those approved by the FDA for treating non-lung diseases, in transducing ALI cultures of primary human airway cells. We provide compelling experimental evidence that the superior performance of EVAAV6 is attributed to the ability of EV to facilitate mucus penetration and cellular entry/transduction of AAV6. The tight and stable linkage between AAV6 and EVs appears essential to exploit the benefits of EVs given that a physical mixture of individually prepared EVs and AAV6 failed to mediate EV-AAV6 interactions or to enhance gene transfer efficacy.


Subject(s)
Extracellular Vesicles , Satellite Viruses , Mice , Animals , Humans , Satellite Viruses/genetics , Transduction, Genetic , Dependovirus/genetics , HEK293 Cells
4.
Bioact Mater ; 25: 527-540, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37056267

ABSTRACT

Highly immunosuppressive tumor microenvironment containing various protumoral immune cells accelerates malignant transformation and treatment resistance. In particular, tumor-associated macrophages (TAMs), as the predominant infiltrated immune cells in a tumor, play a pivotal role in regulating the immunosuppressive tumor microenvironment. As a potential therapeutic strategy to counteract TAMs, here we explore an exosome-guided in situ direct reprogramming of tumor-supportive M2-polarized TAMs into tumor-attacking M1-type macrophages. Exosomes derived from M1-type macrophages (M1-Exo) promote a phenotypic switch from anti-inflammatory M2-like TAMs toward pro-inflammatory M1-type macrophages with high conversion efficiency. Reprogrammed M1 macrophages possessing protein-expression profiles similar to those of classically activated M1 macrophages display significantly increased phagocytic function and robust cross-presentation ability, potentiating antitumor immunity surrounding the tumor. Strikingly, these M1-Exo also lead to the conversion of human patient-derived TAMs into M1-like macrophages that highly express MHC class II, offering the clinical potential of autologous and allogeneic exosome-guided direct TAM reprogramming for arming macrophages to join the fight against cancer.

5.
Small ; 19(11): e2207278, 2023 03.
Article in English | MEDLINE | ID: mdl-36651002

ABSTRACT

Drug delivery nanoparticles (NPs) based entirely on materials generally recognized as safe that provide widespread parenchymal distribution following intracranial administration via convection-enhanced delivery (CED) are introduced. Poly(lactic-co-glycolic acid) (PLGA) NPs are coated with various poloxamers, including F68, F98, or F127, via physical adsorption to render particle surfaces non-adhesive, thereby resisting interactions with brain extracellular matrix. F127-coated PLGA (F127/PLGA) NPs provide markedly greater distribution in healthy rat brains compared to uncoated NPs and widespread coverage in orthotopically-established brain tumors. Distribution analysis of variously-sized F127/PLGA NPs determines the average rat brain tissue porosity to be between 135 and 170 nm while revealing unprecedented brain coverage of larger F127/PLGA NPs with an aid of hydraulic pressure provided by CED. Importantly, F127/PLGA NPs can be lyophilized for long-term storage without compromising their ability to penetrate the brain tissue. Further, 65- and 200-nm F127/PLGA NPs lyophilized-reconstituted and administered in a moderately hyperosmolar infusate solution show further enhance particle dissemination in the brain via osmotically-driven enlargement of the brain tissue porosity. Combination of F127/PLGA NPs and osmotic tissue modulation provides a means with a clear regulatory path to maximize the brain distribution of large NPs that enable greater drug loading and prolong drug release.


Subject(s)
Nanoparticles , Neoplasms , Rats , Animals , Polylactic Acid-Polyglycolic Acid Copolymer , Polyglycolic Acid , Lactic Acid , Drug Carriers , Brain , Particle Size
6.
Curr Protoc ; 2(12): e607, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36469609

ABSTRACT

The advent of the first-ever retinal gene therapy product, involving subretinal administration of a virus-based gene delivery platform, has garnered hope that this state-of-the-art therapeutic modality may benefit a broad spectrum of patients with diverse retinal disorders. On the other hand, clinical studies have revealed limitations of the applied delivery strategy that may restrict its universal use. To this end, intravitreal administration of synthetic gene-delivery platforms, such as polymer-based nanoparticles (PNPs), has emerged as an attractive alternative to the current mainstay. To achieve success, however, it is imperative that synthetic platforms overcome key biological barriers in human eyes encountered following intravitreal administration, including the vitreous gel and inner limiting membrane (ILM). Here, we introduce a series of experiments, from the fabrication of PNPs to a comprehensive evaluation in relevant experimental models, to determine whether PNPs overcome these barriers and efficiently deliver therapeutic gene payloads to retinal cells. We conclude the article by discussing a few important considerations for successful implementation of the strategy. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Preparation and characterization of PNPs Basic Protocol 2: Evaluation of in vitro transfection efficacy Basic Protocol 3: Evaluation of PNP diffusion in vitreous gel Basic Protocol 4: Ex vivo assessment of PNP penetration within vitreoretinal explant culture Basic Protocol 5: Assessment of in vivo transgene expression mediated by intravitreally administered PNPs.


Subject(s)
Nanoparticles , Polymers , Humans , Polymers/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Retina/metabolism
7.
J Control Release ; 345: 62-74, 2022 05.
Article in English | MEDLINE | ID: mdl-35263615

ABSTRACT

Upregulation of oncogenic miRNA21 (miR-21) plays a pivotal role in proliferation, migration and invasion of cancer cells. In addition to cancer cells, tumor-associated macrophages (TAMs) also have high abundance of miR-21, which accelerates malignant progression of tumors in the late stages of carcinogenesis. Despite of the pro-tumorigenic functions of miR-21 in TAMs and cancer cells, reliable therapeutic strategies to simultaneously inhibit miR-21 activity in both types of cell have not yet been developed. In this study, we designed a dual-targeting drug delivery system of miR-21 inhibitors that could bind to both tumor cells and macrophages with overexpressed PD-L1 receptors. This peptide-oligonucleotide conjugate (Pep-21) consists of a PDL1-binding peptide covalently linked with an anti-miR-21 inhibitor via click chemistry. Pep-21 was preferentially internalized in both cell types, consequently depleting endogenous miR-21. Our studies found that Pep-21 treatment reduced tumor cell migration, reprogrammed immunosuppressive M2-type TAMs into M1-type macrophages, and restrained tumor progression. Collectively, neutralization of miR-21 activity in both cancer cells and TAMs can be a promising strategy for effective antitumor responses.


Subject(s)
MicroRNAs , Neoplasms , B7-H1 Antigen/metabolism , Humans , Neoplasms/drug therapy , Peptides , Tumor Microenvironment , Tumor-Associated Macrophages
8.
Small ; 18(15): e2200060, 2022 04.
Article in English | MEDLINE | ID: mdl-35229462

ABSTRACT

Macrophages (Mφs) are characterized by remarkable plasticity, an essential component of chronic inflammation. Thus, an appropriate and timely transition from proinflammatory (M1) to anti-inflammatory (M2) Mφs during wound healing is vital to promoting resolution of acute inflammation and enhancing tissue repair. Herein, exosomes derived from M2-Mφs (M2-Exos), which contain putative key regulators driving Mφ polarization, are used as local microenvironmental cues to induce reprogramming of M1-Mφs toward M2-Mφs for effective wound management. As an injectable controlled release depot for exosomes, hydrolytically degradable poly(ethylene glycol) (PEG) hydrogels (Exogels) are designed and employed for encapsulating M2-Exos to maximize their therapeutic effects in cutaneous wound healing. The degradation time of the hydrogels is adjustable from 6 days or up to 27 days by controlling the crosslinking density and tightness. The localization of M2-Exos leads to a successful local transition from M1-Mφs to M2-Mφs within the lesion for more than 6 days, followed by enhanced therapeutic effects including rapid wound closure and increased healing quality in an animal model for cutaneous wound healing. Collectively, the hydrolytically degradable PEG hydrogel-based exosome delivery system may serve as a potential tool in regulating local polarization state of Mφs, which is crucial for tissue homeostasis and wound repair.


Subject(s)
Exosomes , MicroRNAs , Animals , Biocompatible Materials/metabolism , Delayed-Action Preparations , Exosomes/metabolism , Hydrogels , Inflammation/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Wound Healing/physiology
9.
Thorax ; 77(8): 812-820, 2022 08.
Article in English | MEDLINE | ID: mdl-34697091

ABSTRACT

INTRODUCTION: Inhaled gene therapy of muco-obstructive lung diseases requires a strategy to achieve therapeutically relevant gene transfer to airway epithelium covered by particularly dehydrated and condensed mucus gel layer. Here, we introduce a synthetic DNA-loaded mucus-penetrating particle (DNA-MPP) capable of providing safe, widespread and robust transgene expression in in vivo and in vitro models of muco-obstructive lung diseases. METHODS: We investigated the ability of DNA-MPP to mediate reporter and/or therapeutic transgene expression in lung airways of a transgenic mouse model of muco-obstructive lung diseases (ie, Scnn1b-Tg) and in air-liquid interface cultures of primary human bronchial epithelial cells harvested from an individual with cystic fibrosis. A plasmid designed to silence epithelial sodium channel (ENaC) hyperactivity, which causes airway surface dehydration and mucus stasis, was intratracheally administered via DNA-MPP to evaluate therapeutic effects in vivo with or without pretreatment with hypertonic saline, a clinically used mucus-rehydrating agent. RESULTS: DNA-MPP exhibited marked greater reporter transgene expression compared with a mucus-impermeable formulation in in vivo and in vitro models of muco-obstructive lung diseases. DNA-MPP carrying ENaC-silencing plasmids provided efficient downregulation of ENaC and reduction of mucus burden in the lungs of Scnn1b-Tg mice, and synergistic impacts on both gene transfer efficacy and therapeutic effects were achieved when DNA-MPP was adjuvanted with hypertonic saline. DISCUSSION: DNA-MPP constitutes one of the rare gene delivery systems providing therapeutically meaningful gene transfer efficacy in highly relevant in vivo and in vitro models of muco-obstructive lung diseases due to its unique ability to efficiently penetrate airway mucus.


Subject(s)
Lung Diseases, Obstructive , Nanoparticles , Animals , DNA , Genetic Therapy , Humans , Lung/metabolism , Lung Diseases, Obstructive/therapy , Mice , Mucus/metabolism
10.
ACS Nano ; 15(7): 11369-11384, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34191497

ABSTRACT

MicroRNAs (miRNAs), a recently discovered class of noncoding RNAs, play pivotal roles in regulating fundamental biological processes by suppressing the expression of target genes. Aberrant miRNA expression is commonly correlated with human diseases, including cancers. Anti-miRNA oligonucleotides provide an innovative therapeutic strategy for silencing disease-associated miRNAs. However, the clinical application of anti-miRNA therapy has been limited by formulation challenges and physiological delivery barriers. Here, to provide the safe and effective tumor-targeted delivery of anti-miRNAs, we designed carrier-free maleimide-functionalized anti-miRNAs (MI-Anti-miRNAs) that enable "piggybacking" onto albumin in vivo. These functionalized MI-Anti-miRNAs covalently bind to cysteine-34 of endogenous albumin within minutes. In addition to resulting in a markedly extended blood circulation lifetime, this strategy allows MI-Anti-miRNAs to "hitchhike" to the tumor site. Importantly, in situ-generated albumin-Anti-miRNAs are capable of intracellularly internalizing highly negatively charged anti-miRNA molecules and knocking down target miRNAs. In particular, MI-Anti-miRNAs that targeted miRNA-21, which is involved in tumor initiation, progression, invasion, and metastasis in several types of cancer, successfully repressed miRNA-21 activity, resulting in a superior antitumor activity in both solid and metastatic tumor models without causing systemic toxicity. This endogenous albumin-piggybacking approach using MI-Anti-miRNAs provides a simple and broadly applicable platform strategy for the systemic delivery of anti-miRNA therapeutics.


Subject(s)
MicroRNAs , Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Carcinogenesis , Neoplasms/genetics , Neoplasms/pathology , Oligonucleotides , Albumins
11.
Angew Chem Int Ed Engl ; 60(28): 15225-15229, 2021 07 05.
Article in English | MEDLINE | ID: mdl-33855792

ABSTRACT

Inhaled gene therapy poses a unique potential of curing chronic lung diseases, which are currently managed primarily by symptomatic treatments. However, it has been challenging to achieve therapeutically relevant gene transfer efficacy in the lung due to the presence of numerous biological delivery barriers. Here, we introduce a simple approach that overcomes both extracellular and cellular barriers to enhance gene transfer efficacy in the lung in vivo. We endowed tetra(piperazino)fullerene epoxide (TPFE)-based nanoparticles with non-adhesive surface polyethylene glycol (PEG) coatings, thereby enabling the nanoparticles to cross the airway mucus gel layer and avoid phagocytic uptake by alveolar macrophages. In parallel, we utilized a hypotonic vehicle to facilitate endocytic uptake of the PEGylated nanoparticles by lung parenchymal cells via the osmotically driven regulatory volume decrease (RVD) mechanism. We demonstrate that this two-pronged delivery strategy provides safe, wide-spread and high-level transgene expression in the lungs of both healthy mice and mice with chronic lung diseases characterized by reinforced delivery barriers.


Subject(s)
Epoxy Compounds/chemistry , Fullerenes/chemistry , Gene Transfer Techniques , Lung Diseases/therapy , Nanoparticles/chemistry , Chronic Disease , Humans , Lung Diseases/metabolism
12.
Int J Mol Sci ; 22(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33374978

ABSTRACT

Exosomes are cell-secreted nanovesicles that naturally contain biomolecular cargoes such as lipids, proteins, and nucleic acids. Exosomes mediate intercellular communication, enabling the transfer biological signals from the donor cells to the recipient cells. Recently, exosomes are emerging as promising drug delivery vehicles due to their strong stability in blood circulation, high biocompatibility, low immunogenicity, and natural targeting ability. In particular, exosomes derived from specific types of cells can carry endogenous signaling molecules with therapeutic potential for cancer treatment, thus presenting a significant impact on targeted drug delivery and therapy. Furthermore, exosomes can be engineered to display targeting moieties on their surface or to load additional therapeutic agents. Therefore, a comprehensive understanding of exosome biogenesis and the development of efficient exosome engineering techniques will provide new avenues to establish convincing clinical therapeutic strategies based on exosomes. This review focuses on the therapeutic applications of exosomes derived from various cells and the exosome engineering technologies that enable the accurate delivery of various types of cargoes to target cells for cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Derived Microparticles/metabolism , Drug Delivery Systems/methods , Exosomes/metabolism , Nanoparticles/metabolism , Neoplasms/drug therapy , Animals , Drug Carriers/metabolism , Humans , Molecular Targeted Therapy/methods
13.
Adv Mater ; 32(16): e1907953, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32125731

ABSTRACT

Necroptosis, caspase-independent programmed necrosis, has emerged as a therapeutic target to make dying cancer cells stimulants for antitumor immune responses. The clinical translations exploiting necroptosis, however, have been limited since most cancer cells downregulate receptor-interacting protein kinase 3 (RIPK3) as a key enzyme for necroptosis. Herein, nanobubbles (NBs) that can trigger RIPK3-independent necroptosis, facilitating cell-membrane rupture via the acoustic cavitation effect are reported. The NBs, imbibing perfluoropentane as the gas precursor, are prepared using an amphiphilic polymer conjugate, composed of PEGylated carboxymethyl dextran as the hydrophilic backbone and chlorin e6 as the hydrophobic sonosensitizer. When exposed to ultrasound, the NBs efficiently promote the release of biologically active damage-associated molecular patterns by inducing burst-mediated cell-membrane disintegration. Consequently, the necroptosis-inducible NBs significantly improve antitumor immunity by maturation of dendritic cells and activation of CD8+ cytotoxic T cells both in vitro and in vivo. In addition, the combination of NBs and immune checkpoint blockade leads to complete regression of the primary tumor and beneficial therapeutic activity against metastatic tumors in an RIPK3-deficient CT26 tumor-bearing mouse model. Overall, the innovative NB that causes immunogenic cell death of cancer via RIPK3-independent necroptosis is a promising enhancer for cancer immunotherapy.


Subject(s)
Acoustics , Immunotherapy/methods , Nanostructures/chemistry , Necroptosis/drug effects , Necroptosis/immunology , Polymers/chemistry , Polymers/pharmacology , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Mice
14.
Theranostics ; 10(7): 3240-3253, 2020.
Article in English | MEDLINE | ID: mdl-32194865

ABSTRACT

Rationale: Of the regulatory microRNAs expressed in the wounded skin, microRNA-21 (miR21) plays a pivotal role in wound repair by stimulating re-epithelialization, an essential feature to facilitate healing and reduce scar formation. Despite their crucial roles in wound healing, synthetic exogenous microRNAs have limited applications owing to the lack of an appropriate delivery system. Herein, we designed an miR21 mimic nanocarrier system using facial amphipathic bile acid-conjugated polyethyleneimines (BA-PEI) for the intracellular and transdermal delivery of synthetic miR21 molecules to accelerate wound repair. Methods: To design miR21 mimic nanocarriers, BA-conjugated PEIs prepared from three different types of BA at molar feed ratios of 1 and 3 were synthesized. The intracellular uptake efficiency of synthetic miR21 mimics was studied using confocal laser scanning microscopy and flow cytometry analysis. The optimized miR21/BA nanocarrier system was used to evaluate the wound healing effects induced by miR21 mimics in human HaCaT keratinocytes in vitro and a murine excisional acute wound model in vivo. Results: The cell uptake efficiency of miR21 complexed with BA-conjugated PEI was dramatically higher than that of miR21 complexed with PEI alone. Deoxycholic acid (DA)-modified PEI at a molar feed ratio of 3:1 (DA3-PEI) showed the highest transfection efficiency for miR21 without any increase in toxicity. After effective transdermal and intracellular delivery of miR21/DA3 nanocarriers, miR21 mimics promoted cell migration and proliferation through the post-transcriptional regulation of programmed cell death protein 4 (PDCD4) and matrix metalloproteinases. Thus, miR21 mimic nanocarriers improved both the rate and quality of wound healing, as evident from enhanced collagen synthesis and accelerated wound re-epithelialization. Conclusion: Our miRNA nanocarrier systems developed using DA3-PEI conjugates may be potentially useful for the delivery of synthetic exogenous miRNAs in various fields.


Subject(s)
Bile Acids and Salts/administration & dosage , Drug Carriers/administration & dosage , MicroRNAs/administration & dosage , Nanoconjugates/administration & dosage , Polyethyleneimine/administration & dosage , Skin/injuries , Wound Healing/drug effects , Administration, Cutaneous , Animals , Bile Acids and Salts/chemistry , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Cell Division/drug effects , Cell Line , Cell Movement/drug effects , Drug Design , Drug Liberation , Gene Expression Profiling , Humans , Hydrophobic and Hydrophilic Interactions , Keratinocytes , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/therapeutic use , Molecular Mimicry , Signal Transduction/drug effects , Skin Absorption
15.
Cancer Lett ; 472: 19-28, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31857157

ABSTRACT

Cancer immunotherapy is a powerful approach for cancer treatment, but its clinical effects rely on the tumor's immune conditions. In particular, low response rates to PD-1 blockades are highly correlated with impaired T cell priming. Here, we demonstrate that E. coli-derived monophosphoryl lipid A (EcML) activates dendritic cells in a toll-like receptor-4 (TLR-4)-dependent manner and increases the sensitivity of cancer cells to anti-PD-1 immunotherapy. EcML is a mixture of 4'-monophosphoryl lipids A (MPLAs) produced directly by an engineered Escherichia coli strain; it has a unique congener composition that differentiates it from the well-established MPLA adjuvants, 3-O-desacyl-4'-monophosphoryl lipid A and glucopyranosyl lipid A. Given that active dendritic cells initiate adaptive immune responses, we investigated the anti-tumor activity of an aqueous formulation of EcML. Upon sensing EcML via TLR-4, dendritic cells matured into powerful antigen-presenting cells that could stimulate naïve T cells. EcML reduced tumor growth in the B16F10 mouse model via dendritic cell activation and potentiated PD-1 blockade therapy in the B16F10-OVA melanoma model. These data identify EcML as a promising TLR-4 agonist that can induce anti-tumor immune responses and potentiate PD-1 blockade therapy against tumors.


Subject(s)
Lipid A/analogs & derivatives , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/immunology , Toll-Like Receptor 4/genetics , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Resistance, Neoplasm/immunology , Escherichia coli/genetics , Glucosides/pharmacology , Humans , Immunotherapy/methods , Lipid A/pharmacology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptor 4/antagonists & inhibitors
16.
Adv Sci (Weinh) ; 6(20): 1900513, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31637157

ABSTRACT

Macrophages (Mϕs) critically contribute to wound healing by coordinating inflammatory, proliferative, and angiogenic processes. A proper switch from proinflammatory M1 to anti-inflammatory M2 dominant Mϕs accelerates the wound healing processes leading to favorable wound-care outcomes. Herein, an exosome-guided cell reprogramming technique is proposed to directly convert M1 to M2 Mϕs for effective wound management. The M2 Mϕ-derived exosomes (M2-Exo) induce a complete conversion of M1 to M2 Mϕs in vitro. The reprogrammed M2 Mϕs turn Arginase (M2-marker) and iNOS (M1-marker) on and off, respectively, and exhibit distinct phenotypic and functional features of M2 Mϕs. M2-Exo has not only Mϕ reprogramming factors but also various cytokines and growth factors promoting wound repair. After subcutaneous administration of M2-Exo into the wound edge, the local populations of M1 and M2 Mϕs are markedly decreased and increased, respectively, showing a successful exosome-guided switch to M2 Mϕ polarization. The direct conversion of M1 to M2 Mϕs at the wound site accelerates wound healing by enhancing angiogenesis, re-epithelialization, and collagen deposition. The Mϕ phenotype switching induced by exosomes possessing the excellent cell reprogramming capability and innate biocompatibility can be a promising therapeutic approach for various inflammation-associated disorders by regulating the balance between pro- versus anti-inflammatory Mϕs.

17.
Biomaterials ; 213: 119207, 2019 08.
Article in English | MEDLINE | ID: mdl-31136910

ABSTRACT

Theranostic biomaterials have been creating great new opportunities in developing precision medicine in cancer treatment. Moreover, there exist up-coming potentials to find a new paradigm change in cancer treatment when theranostic biomaterials can precisely image a biological process at the exact tumor sites. With aid of molecular imaging technology, theranostic biomaterials can visualize targets, monitor efficiency of delivery carriers as well as therapeutic responses at tumor site. Major obstacles in designing theranostic biomaterials might be what providing target-specificity to biomaterials for improving therapeutic efficacy as well as visualization in cancer treatment. By which, we can find ways to minimize unwanted side effects to normal tissues in cancer therapy. This review focuses on how theranostic biomaterials can be designed with tumor-specific fluorescence imaging probes, especially tumor-specific activatable imaging probes, which in turn can be switched to therapeutic design with identical principle. Highlights such as designing by characteristics of cancer, finding ways to monitor therapeutic efficacy, potential benefits in theranostic biomaterials are also discussed.


Subject(s)
Biocompatible Materials/therapeutic use , Neoplasms/therapy , Precision Medicine , Theranostic Nanomedicine , Animals , Biocompatible Materials/chemistry , Humans , Hydrogen-Ion Concentration , Mice , Nanoparticles/administration & dosage , Neoplasm Transplantation , Oxidation-Reduction , Tumor Microenvironment
18.
Adv Sci (Weinh) ; 5(11): 1800852, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30479928

ABSTRACT

In the past few years, there have been many efforts underway to develop effective wound healing treatments for traumatic injuries. In particular, wound-healing peptides (WHPs) and peptide-grafted dressings hold great promise for novel therapeutic strategies for wound management. This study reports a topical formulation of a new synthetic WHP (REGRT, REG) embedded in a hyaluronic acid (HA)-based hydrogel dressing for the enhancement of acute excisional wound repair. The copper-free click chemistry is utilized to form biocompatible HA hydrogels by cross-linking dibenzocyclooctyl-functionalized HA with 4-arm poly(ethylene glycol) (PEG) azide. The HA hydrogels are grafted with the REG peptide, a functional derivative of erythroid differentiation regulator1, displaying potent cell motility-stimulating ability, thus sustainably releasing physiologically active peptides for a prolonged period. Combined with the traditional wound healing benefits of HA, the HA hydrogel embedded REG (REG-HAgel) accelerates re-epithelialization in skin wound healing, particularly by promoting migration of fibroblasts, keratinocytes, and endothelial cells. REG-HAgels improve not only rate, but quality of wound healing with higher collagen deposition and more microvascular formation while being nontoxic. The peptide-grafted HA hydrogel system can be considered as a promising new wound dressing formulation strategy for the treatment of different types of wounds with combinations of various natural and synthetic WHPs.

19.
ACS Nano ; 11(10): 10135-10146, 2017 10 24.
Article in English | MEDLINE | ID: mdl-28985469

ABSTRACT

Programmed cell death protein-1 (PD-1) is a prominent immune checkpoint receptor interacting with its ligand, programmed cell death protein ligand-1 (PD-L1, B7-H1). The PD-1/PD-L1 interaction induces functional exhaustion of tumor-reactive cytotoxic T cells and, thus, interferes with antitumor T-cell immunity. In addition, PD-1/PD-L1 interaction promotes tumorigenesis via the mTOR signaling pathway in a group of cancers including melanoma. Based on the dual functions of PD-1/PD-L1 interactions in tumor progression, we hypothesize that siRNA targeting PD-L1 (siPD-L1) will suppress melanoma growth, acting on both immune checkpoint and intrinsic tumorigenesis pathways. We tested this hypothesis by delivering siPD-L1 with a polymeric carrier ("pd") consisting of disulfide-cross-linked polyethylenimine (CLPEI) and dermatan sulfate (DS), which we previously found to have a specific interaction with CD146-positive B16F10 melanoma cells. The siPD-L1/pd suppressed the expression of PD-L1 in the interferon-γ (IFN-γ)-challenged B16F10 melanoma cells in a cell-type dependent manner and attenuated the expression of tumor-specific genes in B16F10 cells. siPD-L1/pd suppressed the B16F10 melanoma growth in C57BL/6 immune-competent mice with increased tumor-specific immunity. siPD-L1/pd also suppressed melanoma growth in immune-compromised nude mice. Both animals showed a positive correlation between PD-L1 and p-S6k (a marker of mTOR pathway activation) expression in tumors. These results indicate that the siPD-L1/pd complex attenuates melanoma growth in both T-cell-dependent and independent mechanisms.


Subject(s)
Antineoplastic Agents/pharmacology , Dermatan Sulfate/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Polyethyleneimine/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Dermatan Sulfate/chemistry , Drug Screening Assays, Antitumor , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , NIH 3T3 Cells , Polyethyleneimine/chemistry , Programmed Cell Death 1 Receptor/metabolism
20.
Adv Sci (Weinh) ; 4(8): 1600523, 2017 08.
Article in English | MEDLINE | ID: mdl-28852615

ABSTRACT

As ribonucleic acid (RNA) nanotechnology has advanced, it has been applied widely in RNA-based therapeutics. Among the range of approaches, enzymatically synthesized RNA structures for inducing RNA interference in cancer cells have potential for silencing genes in a target-specific manner. On the other hand, the efficiency of gene silencing needs to be improved to utilize the RNA-based system for RNAi therapeutics. This paper introduces a new approach for efficient generation of siRNA from bubbled RNA-based cargo (BRC). The presence of bubbles in between to avoid nonfunctional short dsRNAs allows the RNA-based cargoes to contain multiple Dicer-cleavage sites to release the functional siRNAs when introduced to cells. BRCs can be synthesized easily in a one-pot process and be purified by simple centrifugation. Furthermore, efficient target gene silencing by the bubbled structure is confirmed both in vitro and in vivo. Therefore, this bubbled RNA cargo system can be utilized for target-specific RNAi therapeutics with high efficiency in the generation of functional siRNAs in the target cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...