20 results on '"Yoon, Hong Yeol"'
Search Results
2. Dual-Action Protein-siRNA Conjugates for Targeted Disruption of CD47-Signal Regulatory Protein α Axis in Cancer Therapy
- Author
-
Lee, Jong Won, Yoon, Hong Yeol, Ko, Young Ji, Kim, Eun Hye, Song, Sukyung, Hue, Seungmi, Gupta, Nilaksh, Malin, Dmitry, Kim, Jay, Kong, Byoungjae, Kim, Sehoon, Kim, In-San, Kwon, Ick Chan, Yang, Yoosoo, and Kim, Sun Hwa
- Abstract
A series of successes in RNA interference (RNAi) therapies for liver diseases using lipid nanoparticles and N-acetylgalactosamine have heralded a current era of RNA therapeutics. However, alternative delivery strategies are required to take RNAi out of the comfort zone of hepatocytes. Here we report SIRPα IgV/anti-CD47 siRNA (vS-siCD47) conjugates that selectively and persistently disrupt the antiphagocytic CD47/SIRPα axis in solid tumors. Conjugation of the SIRPα IgV domain protein to siRNAs enables tumor dash through CD47-mediated erythrocyte piggyback, primarily blocking the physical interaction between CD47 on cancer cells and SIRPα on phagocytes. After internalization of the vS-siCD47 conjugates within cancer cells, the detached free-standing anti-CD47 siRNAs subsequently attack CD47 through the RNAi mechanism. The dual-action approach of the vS-siCD47 conjugate effectively overcomes the “don’t eat me” barrier and stimulates phagocyte-mediated tumor destruction, demonstrating a highly selective and potent CD47-blocking immunotherapy. This delivery strategy, employing IgV domain protein–siRNA conjugates with a dual mode of target suppression, holds promise for expanding RNAi applications beyond hepatocytes and advancing RNAi-based cancer immunotherapies for solid tumors.
- Published
- 2024
- Full Text
- View/download PDF
3. Photo-induced crosslinked and anti-PD-L1 peptide incorporated liposomes to promote PD-L1 multivalent binding for effective immune checkpoint blockade therapy.
- Author
-
Lee, Youngjoo, Song, Sukyung, Yang, Suah, Kim, Jinseong, Moon, Yujeong, Shim, Nayeon, Yoon, Hong Yeol, Kim, Sehoon, Shim, Man Kyu, and Kim, Kwangmeyung
- Subjects
IMMUNE checkpoint proteins ,PEPTIDES ,PROGRAMMED death-ligand 1 ,LIPOSOMES ,CELLULAR immunity - Abstract
Immune checkpoint blockade (ICB) therapy targeting PD-L1 via monoclonal antibody (mAb) has shown extensive clinical benefits in the diverse types of advanced malignancies. However, most patients are completely refractory to ICB therapy owing to the PD-L1 recycling mechanism. Herein, we propose photo-induced crosslinked and anti-PD-L1 peptide incorporated liposomes (immune checkpoint blockade liposomes; ICB-LPs) to promote PD-L1 multivalent binding for inducing lysosomal degradation of PD-L1 in tumor cells. The ICB-LPs are prepared by formulation of DC 8,9 PC with photo-polymerized diacetylenic moiety, 1,2-dipalmitoylphosphatidylcholine (DPPC) and anti-PD-L1 peptide (D-form NYSKPTDRQYHF)-conjugated DSPE-PEG 2k (anti-PD-L1-DSPE-PEG 2k) in a molar ratio of 45:45:10, followed by cross-linking of liposomal bilayer upon UV irradiation. The 10 mol% anti-PD-L1-DSPE-PEG 2k incorporated ICB-LPs have a nano-sized lipid bilayer structure with an average diameter of 137.7 ± 1.04 nm, showing a high stability in serum condition. Importantly, the ICB-LPs efficiently promote the multivalent binding with PD-L1 on the tumor cell membrane, which are endocytosed with aim to deliver PD-L1 to the lysosomes, wherein the durable PD-L1 degradation is observed for 72 h, in contrast to anti PD-L1 mAbs showing the rapid PD-L1 recycling within 9 h. The in vitro co-culture experiments with CD8
+ T cells show that ICB-LPs effectively enhance the T cell-mediated antitumor immune responses against tumor cells by blocking the PD-L1/PD-1 axis. When ICB-LPs are intravenously injected into colon tumor-bearing mice, they efficiently accumulate within the targeted tumor tissues via both passive and active tumor targeting, inducing a potent T cell-mediated antitumor immune response by effective and durable PD-L1 degradation. Collectively, this study demonstrates the superior antitumor efficacy of crosslinked and anti-PD-L1 peptide incorporated liposome formulation that promotes PD-L1 multivalent binding for trafficking of PD-L1 toward the lysosomes instead of the recycling endosomes. Photo-induced crosslinked and anti-PD-L1 peptide incorporated liposomes promote multivalent binding with PD-L1 on the tumor cell surface, thereby promoting lysosomal PD-L1 degradation and enhancing T cell-mediated antitumor immunity. [Display omitted] [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. All-in-one glycol chitosan nanoparticles for co-delivery of doxorubicin and anti-PD-L1 peptide in cancer immunotherapy
- Author
-
Song, Sukyung, Shim, Man Kyu, Yang, Suah, Lee, Jaewan, Yun, Wan Su, Cho, Hanhee, Moon, Yujeong, Min, Jin Young, Han, Eun Hee, Yoon, Hong Yeol, and Kim, Kwangmeyung
- Abstract
Synergistic immunotherapy of immune checkpoint blockade (ICB) and immunogenic cell death (ICD) has shown remarkable therapeutic efficacy in various cancers. However, patients show low response rates and undesirable outcomes to these combination therapies owing to the recycling mechanism of programmed death-ligand 1 (PD-L1) and the systemic toxicity of ICD-inducing chemotherapeutic drugs. Herein, we propose all-in-one glycol chitosan nanoparticles (CNPs) that can deliver anti-PD-L1 peptide (PP) and doxorubicin (DOX) to targeted tumor tissues for a safe and more effective synergistic immunotherapy. The PP-CNPs, which are prepared by conjugating ᴅ-form PP (NYSKPTDRQYHF) to CNPs, form stable nanoparticles that promote multivalent binding with PD-L1 proteins on the targeted tumor cell surface, resulting in effective lysosomal PD-L1 degradation in contrast with anti-PD-L1 antibody, which induces recycling of endocytosed PD-L1. Consequently, PP-CNPs prevent subcellular PD-L1 recycling and eventually destruct immune escape mechanism in CT26 colon tumor-bearing mice. Moreover, the ICD inducer, DOX is loaded into PP-CNPs (DOX-PP-CNPs) for synergistic ICD and ICB therapy, inducing a large number of damage-associated molecular patterns (DAMPs) in targeted tumor tissues with minimal toxicity in normal tissues. When the DOX-PP-CNPs are intravenously injected into CT26 colon tumor-bearing mice, PP and DOX are efficiently delivered to the tumor tissues viananoparticle-derived passive and active targeting, which eventually induce both lysosomal PD-L1 degradation and substantial ICD, resulting in a high rate of complete tumor regression (CR: 60%) by a strong antitumor immune response. Collectively, this study demonstrates the superior efficacy of synergistic immunotherapy using all-in-one nanoparticles to deliver PP and DOX to targeted tumor tissues.
- Published
- 2023
- Full Text
- View/download PDF
5. Improved survival rate and minimal side effects of doxorubicin for lung metastasis using engineered discoidal polymeric particlesElectronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d2bm00718e
- Author
-
Park, Sanghyo, Park, Hyungkyu, Park, Chaewon, Yun, Wan Su, Hwang, Soonjae, Yoon, Hong Yeol, Kwon, Ick Chan, Kim, Kwangmeyung, and Key, Jaehong
- Abstract
Despite advances in cancer therapy, the discovery of effective cancer treatments remains challenging. In this study, a simple method was developed to increase the efficiency of doxorubicin (DOX) delivery in a lung metastasis model. This method comprises a simple configuration to increase the delivery efficiency viaprecise engineering of the size, shape, loading content, and biodegradability of the drug delivery system. This system had a 3 μm discoidal shape and exerted approximately 90% burst release of the drug within the first 24 h. There was no cytotoxicity of the drug carrier up to a concentration of 1 mg ml−1, and DOX from the carrier was delivered into the cancer cells, exhibiting an anticancer effect comparable to that of the free drug. The ex vivoresults revealed a strong correlation between the location of cancer cells in the lung and the location of DOX delivered by this drug delivery system. These drug carriers were confirmed to intensively deliver DOX to cancer cells in the lung, with minimal off-target effects. These findings indicate that this delivery system can be a new approach to improving the survival rate and reducing the side effects caused by anticancer drugs without the use of targeting ligands and polyethylene glycol.
- Published
- 2022
- Full Text
- View/download PDF
6. Intracellular Uptake Mechanism of Bioorthogonally Conjugated Nanoparticles on Metabolically Engineered Mesenchymal Stem Cells.
- Author
-
Lim, Seungho, Kim, Woojun, Song, Sukyung, Shim, Man Kyu, Yoon, Hong Yeol, Kim, Byung-Soo, Kwon, Ick Chan, and Kim, Kwangmeyung
- Published
- 2021
- Full Text
- View/download PDF
7. Heat-Generating Iron Oxide Multigranule Nanoclusters for Enhancing Hyperthermic Efficacy in Tumor Treatment.
- Author
-
Jeon, Sangmin, Park, Bum Chul, Lim, Seungho, Yoon, Hong Yeol, Jeon, Yoo Sang, Kim, Byung-Soo, Kim, Young Keun, and Kim, Kwangmeyung
- Published
- 2020
- Full Text
- View/download PDF
8. Heat-Generating Iron Oxide Multigranule Nanoclusters for Enhancing Hyperthermic Efficacy in Tumor Treatment
- Author
-
Jeon, Sangmin, Park, Bum Chul, Lim, Seungho, Yoon, Hong Yeol, Jeon, Yoo Sang, Kim, Byung-Soo, Kim, Young Keun, and Kim, Kwangmeyung
- Abstract
The development of heat-generating magnetic nanostructures is critical for the effective management of tumors using magnetic hyperthermia. Herein, we demonstrate that polyethylene glycol (PEG)-coated iron oxide (magnetite, Fe3O4) multigranule nanoclusters (PEG-MGNCs) can enhance the efficiency of hyperthermia-based tumor suppression in vitroand in vivo. MGNCs consisting of granules (crystallites) measuring 22.9 nm in diameter were prepared viathe hydrothermal polyol method, followed by the surface modification of MGNCs with PEG-dopamine. The freshly prepared PEG-MGNCs exhibit 145.9 ± 10.2 nm diameter on average under aqueous conditions. The three-dimensional structures of PEG-MGNCs enhance the hyperthermic efficacy compared with PEGylated single iron-oxide nanoparticles (NPs), resulting in severe heat damage to tumor cells in vitro. In the SCC7 tumor-bearing mice, near-infrared fluorescence dye (Cy5.5)-labeled PEG-MGNCs are successfully accumulated in the tumor tissues because of NP-derived enhanced permeation and retention effect. Finally, the tumor growth is significantly suppressed in PEG-MGNC-treated mice after two-times heat generation by using a longitudinal solenoid, which can generate an alternating magnetic field under high-frequency (19.5 kA/m, 389 kHz) induction. This study shows for the first time that the PEG-MGNCs greatly enhance the hyperthermic efficacy of tumor treatment both in vitroand in vivo.
- Published
- 2020
- Full Text
- View/download PDF
9. In Situ One-Step Fluorescence Labeling Strategy of Exosomes via Bioorthogonal Click Chemistry for Real-Time Exosome Tracking In Vitro and In Vivo
- Author
-
Song, Sukyung, Shim, Man Kyu, Lim, Seungho, Moon, Yujeong, Yang, Suah, Kim, Jinseong, Hong, Yeonsun, Yoon, Hong Yeol, Kim, In-San, Hwang, Kwang Yeon, and Kim, Kwangmeyung
- Abstract
Exosomes are cellular components with promising uses in cancer diagnostics and therapeutics, and their imaging and tracking are essential to study their biological properties. Herein, we report on an in situ one-step fluorescence labeling strategy for exosomes via bioorthogonal click chemistry. First, exosome donor cancer cells were treated with tetraacetylated N-azidoacetyl-d-mannosamine (Ac4ManNAz) to generate unnatural azide groups (−N3) on their surface via metabolic glycoengineering. Then, the azide groups were labeled with near-infrared fluorescent dye-conjugated dibenzylcyclooctyne (DBCO-Cy5) via bioorthogonal click chemistry. After 2 days of incubation, the DBCO-Cy5-labeled exosomes (Cy5-Exo) were successfully secreted from the donor cancer cells and were isolated via classical ultracentrifugation, providing a high-yield of fluorescent dye-labeled exosomes. This in situ one-step bioorthogonal click chemistry offers improved labeling efficiency, biocompatibility, and imaging sensitivy compared to standard exosomes (ST-Exo), purified with classical ultracentrifugation or carbocyanine lipophilic dye (DiD)-labeled exosomes (DiD-Exo) in vitro. In particular, the Cy5-Exo were successfully taken up by A549 cells in a time-dependent manner, and they could escape from lysosome confinement, showing their possible use as a delivery carrier of therapeutic drugs or imaging agents. Finally, intraveneously injected Cy5-Exo were noninvasively tracked and imaged via near-infrared fluorescence (NIRF) imaging in tumor-bearing mice. This new fluorescence labeling strategy for natural exosomes may be useful to provide better understanding of their theranostic effects in many biomedical applications.
- Published
- 2020
- Full Text
- View/download PDF
10. Recent Trends in In SituEnzyme-Activatable Prodrugs for Targeted Cancer Therapy
- Author
-
Sun, In-Cheol, Yoon, Hong Yeol, Lim, Dong-Kwon, and Kim, Kwangmeyung
- Abstract
Enzyme-activatable anticancer prodrugs are modified medications that are composed of an anticancer drug, cleavable linker, and functional moiety. The purpose of such a prodrug structure is to generate multipurpose functions that traditional drugs cannot perform and to reduce the toxicity of conventional anticancer drugs by the mask of the cleavable linker. Once the cleavable linker is degraded via a specific chemical reaction in the cancer microenvironment, the cytotoxicity of the degraded prodrugs is selectively recovered. Among many factors that cleave the linker, we focus on the overexpressed enzymes in cancer. Because of the selective enzymatic degradation of the cleavable linker and the high local concentration of specific enzymes in cancer, the enzyme-activatable prodrugs could show low toxicity in normal tissues, while showing comparable anticancer effect in tumors. In addition, some prodrugs provide additional features, such as cancer imaging, drug release monitoring, tumor targeting, and enhanced stability, which conventional anticancer drugs cannot possess. In this review, we summarize currently developed enzyme-activatable prodrugs according to their activating enzymes, and categorize them by their additional functions, e.g. targeting, imaging, and delivery. This summary of enzyme-activatable prodrugs may help in the design of anticancer prodrugs, and in the establishment of a personalized cancer treatment strategy.
- Published
- 2020
- Full Text
- View/download PDF
11. A Comparative Study on Albumin-Binding Molecules for Targeted Tumor Delivery through Covalent and Noncovalent Approach.
- Author
-
Um, Wooram, Park, Jooho, Youn, Ahye, Cho, Hanhee, Lim, Seungho, Lee, Jong Won, Yoon, Hong Yeol, Lim, Dong-Kwon, Park, Jae Hyung, and Kim, Kwangmeyung
- Published
- 2019
- Full Text
- View/download PDF
12. A Comparative Study on Albumin-Binding Molecules for Targeted Tumor Delivery through Covalent and Noncovalent Approach
- Author
-
Um, Wooram, Park, Jooho, Youn, Ahye, Cho, Hanhee, Lim, Seungho, Lee, Jong Won, Yoon, Hong Yeol, Lim, Dong-Kwon, Park, Jae Hyung, and Kim, Kwangmeyung
- Abstract
Various types of albumin-binding molecules have been conjugated to anticancer drugs, and these modified prodrugs could be effective in cancer treatments compared to free anticancer drugs. However, the tumor targeting of albumin-binding prodrugs has not been clearly investigated. Herein, we examined the in vitro and in vivo tumor-targeting efficiency of three different albumin-binding molecules including albumin-binding peptide (DICLPRWGCLW: PEP), fatty acid (palmitic acid: PA), and maleimide (MI), respectively. In order to characterize the different targeting efficiency of albumin-binding molecules, PEP, PA, or MI was chemically labeled with near-infrared fluorescence (NIRF) dye, Cy5.5, in resulting PEP-Cy5.5, PA-Cy5.5, and MI-Cy5.5. These NIRF dye-labeled albumin-binding molecules were physically or chemically bound to albumin via gentle incubation in aqueous conditions in vitro. Notably, PA-Cy5.5 with reversible and multivalent binding affinities formed stable albumin complexes, compared to PEP-Cy5.5 and MI-Cy5.5, confirmed via surface plasmon resonance measurement, gel electrophoresis assay, and albumin-bound column-binding test. In tumor-bearing mice model, the different albumin-binding affinities of PA-Cy5.5, PEP-Cy5.5, and MI-Cy5.5 greatly contributed to their tumor-targeting ability. Even though the binding affinity of PEP-Cy5.5 and MI-Cy5.5 to albumin is higher than that of PA-Cy5.5 in vitro, intravenous PA-Cy5.5 showed a higher tumor-targeting efficiency in tumor-bearing mice compared to that of PEP-Cy5.5 and MI-Cy5.5. The reversible and multivalent affinities of albumin-binding molecules to native serum albumin greatly increased the pharmacokinetics and tumor-targeting efficiency in vivo.
- Published
- 2019
- Full Text
- View/download PDF
13. Dual-Modal Imaging-Guided Precise Tracking of Bioorthogonally Labeled Mesenchymal Stem Cells in Mouse Brain Stroke
- Author
-
Lim, Seungho, Yoon, Hong Yeol, Jang, Hee Jeong, Song, Sukyung, Kim, Woojun, Park, Jooho, Lee, Kyung Eun, Jeon, Sangmin, Lee, Sangmin, Lim, Dong-Kwon, Kim, Byung-Soo, Kim, Dong-Eog, and Kim, Kwangmeyung
- Abstract
Noninvasive and precise stem cell tracking after transplantation in living subject is very important to monitor both stem cell destinations and their in vivofate, which is closely related to their therapeutic efficacy. Herein, we developed bicyclo[6.1.0]nonyne (BCN)-conjugated glycol chitosan nanoparticles (BCN-NPs) as a delivery system of dual-modal stem cell imaging probes. Near-infrared fluorescent (NIRF) dye Cy5.5 was chemically conjugated to the BCN-NPs, and then oleic acid-coated superparamagnetic iron oxide nanoparticles (OA-Fe3O4NPs) were encapsulated into BCN-NPs, resulting in Cy5.5-labeled and OA-Fe3O4NP-encapsulated BCN-NPs (BCN-dual-NPs). For bioorthogonal labeling of human adipose-derived mesenchymal stem cells (hMSCs), first, hMSCs were treated with tetra-acetylated N-azidoacetyl-d-mannosamine (Ac4ManNAz) for generating azide (−N3) groups onto their surface viametabolic glycoengineering. Second, azide groups on the cell surface were successfully chemically labeled with BCN-dual-NPs viabioorthogonal click chemistry in vitro. This bioorthogonal labeling of hMSCs could greatly increase the cell labeling efficiency, safety, and imaging sensitivity, compared to only nanoparticle-derived labeling technology. The dual-modal imaging-guided precise tracking of bioorthogonally labeled hMSCs was tested in the photothrombotic stroke mouse model viaintraparenchymal injection. Finally, BCN-dual-NPs-labeled hMSCs could be effectively tracked by their migration from the implanted site to the brain stroke lesion using NIRF/T2-weighted magnetic resonance (MR) dual-modal imaging for 14 days. Our observation would provide a potential application of bioorthogonally labeled stem cell imaging in regenerative medicine by providing safety and high labeling efficiency in vitroand in vivo.
- Published
- 2019
- Full Text
- View/download PDF
14. Tumor-targeting glycol chitosan nanocarriers: overcoming the challenges posed by chemotherapeutics
- Author
-
Choi, Yongwhan, Lim, Seungho, Yoon, Hong Yeol, Kim, Byung-Soo, Kwon, Ick Chan, and Kim, Kwangmeyung
- Abstract
ABSTRACTIntroduction: In the past decade, the glycol chitosan nanocarriers (GCNCs) have been widely used for tumor-targeted delivery of anticancer agents such as chemo drugs, peptides, and genes due to their biocompatibility, biodegradability, and easy fabrication. In particular, GCNCs can effectively solubilize water-insoluble chemo drugs as well as improve the delivery efficiency of chemo drugs to the tumor, resulting in maximizing therapeutic efficacy and minimizing side effect. In this review, we introduce the various applications of GCNCs for cancer treatment and these GCNCs demonstrate the great potential in overcoming challenges of chemotherapeutics.Areas covered: Various designs of GC nanocarriers have been reviewed. The current state of GC nanocarriers for delivering chemotherapeutics with a focus on their physicochemical properties including solubilization of anti-cancer drugs, sustained release, and tumor-selectivity. Furthermore, state of the art in delivery and therapeutic strategy using GC nanocarriers also introduced for overcoming challenges of chemotherapeutics.Expert opinion: Based on the reviewed literature, physicochemical properties of GC nanocarriers will have a great potential to overcome challenges posed by chemotherapeutics.
- Published
- 2019
- Full Text
- View/download PDF
15. Artificial Chemical Reporter Targeting Strategy Using Bioorthogonal Click Reaction for Improving Active-Targeting Efficiency of Tumor
- Author
-
Yoon, Hong Yeol, Shin, Min Lee, Shim, Man Kyu, Lee, Sangmin, Na, Jin Hee, Koo, Heebeom, Lee, Hyukjin, Kim, Jong-Ho, Lee, Kuen Yong, Kim, Kwangmeyung, and Kwon, Ick Chan
- Abstract
Biological ligands such as aptamer, antibody, glucose, and peptide have been widely used to bind specific surface molecules or receptors in tumor cells or subcellular structures to improve tumor-targeting efficiency of nanoparticles. However, this active-targeting strategy has limitations for tumor targeting due to inter- and intraheterogeneity of tumors. In this study, we demonstrated an alternative active-targeting strategy using metabolic engineering and bioorthogonal click reaction to improve tumor-targeting efficiency of nanoparticles. We observed that azide-containing chemical reporters were successfully generated onto surface glycans of various tumor cells such as lung cancer (A549), brain cancer (U87), and breast cancer (BT-474, MDA-MB231, MCF-7) via metabolic engineering in vitro. In addition, we compared tumor targeting of artificial azide reporter with bicyclononyne (BCN)-conjugated glycol chitosan nanoparticles (BCN–CNPs) and integrin αvβ3with cyclic RGD-conjugated CNPs (cRGD–CNPs) in vitroand in vivo. Fluorescence intensity of azide-reporter-targeted BCN–CNPs in tumor tissues was 1.6-fold higher and with a more uniform distribution compared to that of cRGD–CNPs. Moreover, even in the isolated heterogeneous U87 cells, BCN–CNPs could bind artificial azide reporters on tumor cells more uniformly (∼92.9%) compared to cRGD–CNPs. Therefore, the artificial azide-reporter-targeting strategy can be utilized for targeting heterogeneous tumor cells via bioorthogonal click reaction and may provide an alternative method of tumor targeting for further investigation in cancer therapy.
- Published
- 2017
- Full Text
- View/download PDF
16. A Facile, One-Step NanocarbonFunctionalization forBiomedical Applications.
- Author
-
Swierczewska, Magdalena, Choi, Ki Young, Mertz, Edward L., Huang, Xinglu, Zhang, Fan, Zhu, Lei, Yoon, Hong Yeol, Park, Jae Hyung, Bhirde, Ashwinkumar, Lee, Seulki, and Chen, Xiaoyuan
- Published
- 2012
- Full Text
- View/download PDF
17. Bioreducible Block Copolymers Based on Poly(Ethylene Glycol) and Poly(γ-Benzyl l-Glutamate) for Intracellular Delivery of Camptothecin.
- Author
-
Thambi, Thavasyappan, Yoon, Hong Yeol, Kim, Kwangmeyung, Kwon, Ick Chan, Yoo, Chang Kyoo, and Park, Jae Hyung
- Published
- 2011
- Full Text
- View/download PDF
18. Smart Nanocarrier Based on PEGylated Hyaluronic Acid for Cancer Therapy
- Author
-
Choi, Ki Young, Yoon, Hong Yeol, Kim, Jong-Ho, Bae, Sang Mun, Park, Rang-Woon, Kang, Young Mo, Kim, In-San, Kwon, Ick Chan, Choi, Kuiwon, Jeong, Seo Young, Kim, Kwangmeyung, and Park, Jae Hyung
- Abstract
Tumor targetability and site-specific drug release of therapeutic nanoparticles are key factors for effective cancer therapy. In this study, poly(ethylene glycol) (PEG)-conjugated hyaluronic acid nanoparticles (P-HA-NPs) were investigated as carriers for anticancer drugs including doxorubicin and camptothecin (CPT). P-HA-NPs were internalized into cancer cells (SCC7 and MDA-MB-231) viareceptor-mediated endocytosis, but were rarely taken up by normal fibroblasts (NIH-3T3). During in vitrodrug release tests, P-HA-NPs rapidly released drugs when incubated with cancer cells, extracts of tumor tissues, or the enzyme Hyal-1, which is abundant in the intracellular compartments of cancer cells. CPT-loaded P-HA-NPs (CPT-P-HA-NPs) showed dose-dependent cytotoxicity to cancer cells (MDA-MB-231, SCC7, and HCT 116) and significantly lower cytotoxicity against normal fibroblasts (NIH-3T3) than free CPT. Unexpectedly, high concentrations of CPT-P-HA-NPs demonstrated greater cytotoxicity to cancer cells than free CPT. An in vivobiodistribution study indicated that P-HA-NPs selectively accumulated into tumor sites after systemic administration into tumor-bearing mice, primarily due to prolonged circulation in the blood and binding to a receptor (CD44) that was overexpressed on the cancer cells. In addition, when CPT-P-HA-NPs were systemically administrated into tumor-bearing mice, we saw no significant increases in tumor size for at least 35 days, implying high antitumor activity. Overall, P-HA-NPs showed promising potential as a drug carrier for cancer therapy.
- Published
- 2011
- Full Text
- View/download PDF
19. Visible-Light-Triggered Prodrug Nanoparticles Combine Chemotherapy and Photodynamic Therapy to Potentiate Checkpoint Blockade Cancer Immunotherapy
- Author
-
Choi, Jiwoong, Shim, Man Kyu, Yang, Suah, Hwang, Hee Sook, Cho, Hanhee, Kim, Jeongrae, Yun, Wan Su, Moon, Yujeong, Kim, Jinseong, Yoon, Hong Yeol, and Kim, Kwangmeyung
- Abstract
Immune checkpoint blockade is a promising approach for cancer immunotherapy, but many patients do not respond due to the immunosuppressive tumor microenvironment (ITM). Herein, we propose visible-light-triggered prodrug nanoparticles (LT-NPs) for reversing ITM into high immunogenic tumors to potentiate checkpoint blockade immunotherapy. The photosensitizer (verteporfin; VPF), cathepin B-specific cleavable peptide (FRRG), and doxorubicin (DOX) conjugates are self-assembled into LT-NPs without any additional carrier material. The LT-NPs are specifically cleaved to VPF and DOX in cathepsin B-overexpressing cancer cells, thereby inducing cancer-specific cytotoxicity and immunogenic cell death (ICD) upon visible light irradiation. In tumor models, LT-NPs highly accumulate within tumors viathe enhanced permeability and retention effect, and photochemotherapy of VPF and DOX induces effective ICD and maturation of dendritic cells to stimulate cross-presentation of cancer-antigens to T cells. Furthermore, LT-NPs with PD-L1 blockade greatly inhibit tumor growth, tumor recurrence, and lung metastasis by initiating a strong antitumor immune response. The photochemotherapy by LT-NPs provides a promising strategy for effective checkpoint blockade immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
20. Intracellular Uptake Mechanism of Bioorthogonally Conjugated Nanoparticles on Metabolically Engineered Mesenchymal Stem Cells
- Author
-
Lim, Seungho, Kim, Woojun, Song, Sukyung, Shim, Man Kyu, Yoon, Hong Yeol, Kim, Byung-Soo, Kwon, Ick Chan, and Kim, Kwangmeyung
- Abstract
Nanoparticles have been used for effectively delivering imaging agents and therapeutic drugs into stem cells. However, nanoparticles are not sufficiently internalized into stem cells; thus, new delivery method of nanoparticles into stem cells is urgently needed. Herein, we develop bicyclo[6.1.0]nonyne (BCN)-conjugated gold nanoparticles (BCN-AuNPs), which can be bioorthogonally conjugated to azide (−N3) groups on the surface of metabolically engineered stem cells via bioorthogonal click chemistry. For incorporating azide groups on the cell surface, first, human adipose-derived mesenchymal stem cells (hMSCs) were metabolically engineered with N-azidoacetylmannosamine-tetraacylated (Ac4ManNAz). Second, clickable BCN-AuNPs were bioorthogonally conjugated to azide groups on Ac4ManNAz-treated hMSCs. Importantly, a large amount of BCN-AuNPs was specifically conjugated to metabolically engineered hMSCs and then internalized rapidly into stem cells through membrane turnover mechanism, compared to the conventional nanoparticle-derived endocytosis mechanism. Furthermore, BCN-AuNPs entrapped in endosomal/lysosomal compartment could escape efficiently to the cytoplasm of metabolically engineered stem cells. Finally, BCN-AuNPs in stem cells were very safe, and they did not affect stem cell functions, such as self-renewal and differentiation capacity. These bioorthogonally conjugated nanoparticles on metabolically engineered stem cells can enhance the cellular uptake of nanoparticles via bioorthogonal conjugation mechanism.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.