1
|
Kim KR, Lee AS, Heo HR, Park SY, Kim CS. Bioinspired synthesis of virus-like particle-templated thin silica-layered nanocages with enhanced biocompatibility and cellular uptake as drug delivery carriers. Colloids Surf B Biointerfaces 2024; 247:114418. [PMID: 39642678 DOI: 10.1016/j.colsurfb.2024.114418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
The bioinspired synthesis of virus-like silica nanoparticles in biomedical applications makes it possible to utilize the cellular delivery capabilities of viruses while minimizing the cytotoxicity of inorganic silica. In this study, we developed a diatom-inspired method for synthesizing silica-layered nanocages utilizing R5 peptide-functionalized virus-like particles (VLPs). R5 peptides were genetically inserted into the F-G loop of human papillomavirus 16 L1 proteins (HPV16 L1-R5). HPV16 L1-R5 was self-assembled into VLPs under an acidic pH similar to native ones and exhibited ∼65 % drug encapsulation efficiency. The HPV16 L1-R5 VLP@silica nanocages (SiNPs) were synthesized through diatom-inspired silicification of HPV16 L1-R5 VLPs via intermolecular interaction of the R5 peptide and polyol. HPV16L1-R5 VLP@SiNPs displayed uniform, monodisperse particles with approximately 10 nm silica layer compared to HPV16 L1-R5 VLPs. HPV16 L1-R5 VLP@SiNPs showed high biocompatibility at high concentrations, unlike commercial mesoporous SiNPs. Furthermore, the virus-like HPV16 L1-R5 VLP@SiNPs resulted in approximately 2.5-fold increased cellular uptake efficiency compared to commercial mesoporous SiNPs. These results suggest that the thin silica layer on HPV16 L1-R5 VLPs retains cellular delivery capacity while reducing cytotoxicity. Our strategy presents an innovative method for synthesizing virus-like nanoparticles in biomedical applications, enhancing cellular delivery capacity and biocompatibility.
Collapse
Affiliation(s)
- Kyeong Rok Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Ae Sol Lee
- Graduate School of Chemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Hye Ryoung Heo
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu 42415, Republic of Korea.
| | - Chang Sup Kim
- Graduate School of Chemical Engineering, Dongguk University, Seoul 04620, Republic of Korea; Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
2
|
Yuan X, Yang X. CAPRIN1 Transcriptionally Activated PLPP4 to Inhibit DOX Sensitivity and Promote Breast Cancer Progression. Cell Biochem Biophys 2024:10.1007/s12013-024-01614-0. [PMID: 39556159 DOI: 10.1007/s12013-024-01614-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Phospholipid phosphatase 4 (PLPP4) has been identified as a potential regulator of cancer cell dynamics, however, the role of PLPP4 in breast cancer (BC) progression and the sensitivity of BC cells to doxorubicin (DOX) remain elusive. METHODS The study analyzed the expression of PLPP4 and cell cycle-associated protein 1 (CAPRIN1) expression in BC tissues and cells using quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and western blotting assays. Functional assays including colony formation, EdU, Transwell, and flow cytometry were employed to assess cellular behaviors. The sensitivity of BC cells to DOX was analyzed by CCK-8 assay and an in vivo xenograft model assay. The association between PLPP4 and CAPRIN1 was investigated using RNA immunoprecipitation assay and dual-luciferase reporter assay. RESULTS Upregulation of PLPP4 expression was observed in BC tissues and cells. Downregulation of PLPP4 expression in BC cells resulted in a suppression of their proliferative capacity, as well as a reduction in migratory and invasive capabilities. Additionally, this manipulation enhanced cell susceptibility to apoptosis and improved the sensitivity of these cells to DOX. When PLPP4 was knocked down in vivo in transplantable tumors, there was a marked enhancement in the responsiveness to DOX treatment. The transcription factor CAPRIN1 was found to regulate the expression of PLPP4 in the HCC1937 and MDA-MB-231 cell lines. Upregulation of CAPRIN1 was observed in both BC tissues and cells, and overexpression of PLPP4 reversed the effects of CAPRIN1 silencing on BC cell proliferation, migration, invasion, apoptosis, and DOX sensitivity. CONCLUSION This study demonstrates that CAPRIN1 transcriptionally activates PLPP4 to inhibit DOX sensitivity and promote BC progression. Targeting PLPP4 may represent a novel therapeutic strategy to enhance the efficacy of DOX in BC patients.
Collapse
Affiliation(s)
- Xiaorong Yuan
- Department of Lymphatic Breast Oncology, Baotou Cancer Hospital, Baotou, 014030, China
| | - Xuejie Yang
- Department of Lymphatic Breast Oncology, Baotou Cancer Hospital, Baotou, 014030, China.
| |
Collapse
|
3
|
Jiang Y, Lu H, Lei L, Yuan X, Scherman D, Liu Y. MOF-derived cobalt-iron containing nanocomposite with cascade-catalytic activities for multimodal synergistic tumor therapy. Colloids Surf B Biointerfaces 2024; 240:113981. [PMID: 38815310 DOI: 10.1016/j.colsurfb.2024.113981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024]
Abstract
Reactive oxygen species (ROS)-driven chemodynamic therapy has emerged as a promising anti-tumor strategy. However, the insufficient hydrogen peroxide (H2O2) supply in tumor microenvironment results in a low Fenton reaction rate and subsequently poor ROS production and therapeutic efficacy. Herein, we report on a new nanocomposite MIL-53@ZIF-67/S loaded with doxorubicin and glucose oxidase, which is decomposed under the acidic tumor microenvironment to release Fe3+, Co3+, glucose oxidase, and doxorubicin. The released content leads to synergistic anti-tumor effect through the following manners: 1) doxorubicin is directly used for chemotherapy; 2) Fe3+and Co3+ result in glutathione depletion and Fenton reaction activation through Fe2+ and Co2+ generation to achieve chemodynamic therapy; 3) glucose oxidase continuously catalyzes glucose consumption to induce starvation of the cancer cells, and 4) at the same time the produced gluconic acid and H2O2 significantly promote Fenton reaction and further boost chemodynamic therapy. This work not only demonstrates the high anti-tumor effect of the new nanocomposite, but also provides an innovative strategy for the development of a multi-in-one nanoplatform for cancer therapy.
Collapse
Affiliation(s)
- Ying Jiang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China; Department of Mechanics and Engineering Science, Beijing Innovation Centre for Engineering Science and Advanced Technology, College of Engineering, Peking University, Beijing 100871, China
| | - Hao Lu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Lingli Lei
- College of Pharmacy, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Xiangyang Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Daniel Scherman
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China; Université Paris Cité, CNRS, INSERM, UTCBS, Unité de Technologies Chimiques et Biologiques pour la Santé, Paris F-75006, France.
| | - Yingshuai Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China.
| |
Collapse
|
4
|
Liu Y, Cao R, Yang J, Chen H, Zhang J, Feng X. Self-assembly of enzymes and prodrugs with clickable amino acids for nucleus-targeted cancer therapy. Chem Commun (Camb) 2024; 60:7335-7338. [PMID: 38915280 DOI: 10.1039/d4cc02377c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
A nucleus-targeted nanocomposite was prepared by clickable amino acid-tuned one-step co-assembly of proteins and chemotherapeutics. The nanocomposite with favorable pharmacokinetic behavior can effectively accumulate in the nucleus, thereby significantly enhancing the anticancer therapeutic effect both in vitro and in vivo.
Collapse
Affiliation(s)
- Ye Liu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | - Rumeng Cao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | - Jieyu Yang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | - Hui Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | - Jiumeng Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | - Xuli Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
5
|
Liu J, Hu X, Yu G, Wang Q, Gu L, Shen J, Zhao Q, Sun H, Wang S, Guo Z, Zhao Y, Ma H. Doxorubicin-based ENO1 targeted drug delivery strategy enhances therapeutic efficacy against colorectal cancer. Biochem Pharmacol 2024; 224:116220. [PMID: 38641307 DOI: 10.1016/j.bcp.2024.116220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Alpha-enolase (ENO1), a multifunctional protein with carcinogenic properties, has emerged as a promising cancer biomarker because of its differential expression in cancer and normal cells. On the basis of this characteristic, we designed a cell-targeting peptide that specifically targets ENO1 and connected it with the drug doxorubicin (DOX) by aldehyde-amine condensation. A surface plasmon resonance (SPR) assay showed that the affinity for ENO1 was stronger (KD = 2.5 µM) for the resulting cell-targeting drug, DOX-P, than for DOX. Moreover, DOX-P exhibited acid-responsive capabilities, enabling precise release at the tumor site under the guidance of the homing peptide and alleviating DOX-induced cardiotoxicity. An efficacy experiment confirmed that, the targeting ability of DOX-P toward ENO1 demonstrated superior antitumor activity against colorectal cancer than that of DOX, while reducing its toxicity to cardiomyocytes. Furthermore, in vivo metabolic distribution results indicated low accumulation of DOX-P in nontumor sites, further validating its targeting ability. These results showed that the ENO1-targeted DOX-P peptide has great potential for application in targeted drug-delivery systems for colorectal cancer therapy.
Collapse
Affiliation(s)
- Jun Liu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Xiaoyu Hu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Guanghao Yu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Qingrong Wang
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Liwei Gu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Jianying Shen
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Qinghe Zhao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Hao Sun
- Nanjing Agricultural University, Nanjing 210009, China
| | - Shi Wang
- Nanjing Agricultural University, Nanjing 210009, China
| | - Zhongyuan Guo
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China; College of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Yu Zhao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China.
| | - Hai Ma
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China.
| |
Collapse
|
6
|
Wu D, Wu Z, Yao H, Yan X, Jiao Z, Liu Y, Zhang M, Wang D. Doxorubicin induces deglycosylation of cancer cell-intrinsic PD-1 by NGLY1. FEBS Lett 2024; 598:1543-1553. [PMID: 38782868 DOI: 10.1002/1873-3468.14935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Tumor cells can express the immune checkpoint protein programmed death-1 (PD-1), but how cancer cell-intrinsic PD-1 is regulated in response to cellular stresses remains largely unknown. Here, we uncover a unique mechanism by which the chemotherapy drug doxorubicin (Dox) regulates cancer cell-intrinsic PD-1. Dox upregulates PD-1 mRNA while reducing PD-1 protein levels in tumor cells. Although Dox shortens the PD-1 half-life, it fails to directly induce PD-1 degradation. Instead, we observe that Dox promotes the interaction between peptide-N(4)-(N-acetyl-beta-glucosaminyl)asparagine amidase (NGLY1) and PD-1, facilitating NGLY1-mediated PD-1 deglycosylation and destabilization. The maintenance of PD-1 sensitizes tumor cells to Dox-mediated antiproliferative effects. Our study unveils a regulatory mechanism of PD-1 in response to Dox and highlights a potential role of cancer cell-intrinsic PD-1 in Dox-mediated antitumor effects.
Collapse
Affiliation(s)
- Dexuan Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhen Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Han Yao
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaojun Yan
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zishan Jiao
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yajing Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meng Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Donglai Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Kashani GK, Naghib SM, Soleymani S, Mozafari MR. A review of DNA nanoparticles-encapsulated drug/gene/protein for advanced controlled drug release: Current status and future perspective over emerging therapy approaches. Int J Biol Macromol 2024; 268:131694. [PMID: 38642693 DOI: 10.1016/j.ijbiomac.2024.131694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
In the last ten years, the field of nanomedicine has experienced significant progress in creating novel drug delivery systems (DDSs). An effective strategy involves employing DNA nanoparticles (NPs) as carriers to encapsulate drugs, genes, or proteins, facilitating regulated drug release. This abstract examines the utilization of DNA NPs and their potential applications in strategies for controlled drug release. Researchers have utilized the distinctive characteristics of DNA molecules, including their ability to self-assemble and their compatibility with living organisms, to create NPs specifically for the purpose of delivering drugs. The DNA NPs possess numerous benefits compared to conventional drug carriers, such as exceptional stability, adjustable dimensions and structure, and convenient customization. Researchers have successfully achieved a highly efficient encapsulation of different therapeutic agents by carefully designing their structure and composition. This advancement enables precise and targeted delivery of drugs. The incorporation of drugs, genes, or proteins into DNA NPs provides notable advantages in terms of augmenting therapeutic effectiveness while reducing adverse effects. DNA NPs serve as a protective barrier for the enclosed payloads, preventing their degradation and extending their duration in the body. The protective effect is especially vital for delicate biologics, such as proteins or gene-based therapies that could otherwise be vulnerable to enzymatic degradation or quick elimination. Moreover, the surface of DNA NPs can be altered to facilitate specific targeting towards particular tissues or cells, thereby augmenting the accuracy of delivery. A significant benefit of DNA NPs is their capacity to regulate the kinetics of drug release. Through the manipulation of the DNA NPs structure, scientists can regulate the rate at which the enclosed cargo is released, enabling a prolonged and regulated dispensation of medication. This control is crucial for medications with limited therapeutic ranges or those necessitating uninterrupted administration to attain optimal therapeutic results. In addition, DNA NPs have the ability to react to external factors, including alterations in temperature, pH, or light, which can initiate the release of the payload at precise locations or moments. This feature enhances the precision of drug release control. The potential uses of DNA NPs in the controlled release of medicines are extensive. The NPs have the ability to transport various therapeutic substances, for example, drugs, peptides, NAs (NAs), and proteins. They exhibit potential for the therapeutic management of diverse ailments, including cancer, genetic disorders, and infectious diseases. In addition, DNA NPs can be employed for targeted drug delivery, traversing biological barriers, and surpassing the constraints of conventional drug administration methods.
Collapse
Affiliation(s)
- Ghazal Kadkhodaie Kashani
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Sina Soleymani
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran; Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia; Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Iran University of Science and Technology (IUST), Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
8
|
Seo HS, Han JH, Lim J, Bae GH, Byun MJ, Wang CPJ, Han J, Park J, Park HH, Shin M, Park TE, Kim TH, Kim SN, Park W, Park CG. Enhanced Postsurgical Cancer Treatment Using Methacrylated Glycol Chitosan Hydrogel for Sustained DNA/Doxorubicin Delivery and Immunotherapy. Biomater Res 2024; 28:0008. [PMID: 38532906 PMCID: PMC10964224 DOI: 10.34133/bmr.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/09/2024] [Indexed: 03/28/2024] Open
Abstract
Background: Cancer recurrence and metastasis are major contributors to treatment failure following tumor resection surgery. We developed a novel implantable drug delivery system utilizing glycol chitosan to address these issues. Glycol chitosan is a natural adjuvant, inducing dendritic cell activation to promote T helper 1 cell immune responses, macrophage activation, and cytokine production. Effective antigen production by dendritic cells initiates T-cell-mediated immune responses, aiding tumor growth control. Methods: In this study, we fabricated multifunctional methacrylated glycol chitosan (MGC) hydrogels with extended release of DNA/doxorubicin (DOX) complex for cancer immunotherapy. We constructed the resection model of breast cancer to verify the anticancer effects of MGC hydrogel with DNA/DOX complex. Results: This study demonstrated the potential of MGC hydrogel with extended release of DNA/DOX complex for local and efficient cancer therapy. The MGC hydrogel was implanted directly into the surgical site after tumor resection, activating tumor-related immune cells both locally and over a prolonged period of time through immune-reactive molecules. Conclusions: The MGC hydrogel effectively suppressed tumor recurrence and metastasis while enhancing immunotherapeutic efficacy and minimizing side effects. This biomaterial-based drug delivery system, combined with cancer immunotherapy, can substantial improve treatment outcomes and patient prognosis.
Collapse
Affiliation(s)
- Hee Seung Seo
- Department of Biomedical Engineering,
SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence,
Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Jun-Hyeok Han
- Department of Intelligent Precision Healthcare Convergence,
Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering,
SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Jaesung Lim
- Department of Biomedical Engineering,
SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence,
Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Ga-Hyun Bae
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering,
SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth,
SKKU Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Min Ji Byun
- Department of Biomedical Engineering,
SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence,
Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Chi-Pin James Wang
- Department of Biomedical Engineering,
SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence,
Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Jieun Han
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering,
SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Institute of Biotechnology and Bioengineering, College of Biotechnology and Bioengineering, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School Medicine,
University of Hawai'i at Manoa, Honolulu, HI 96813, USA
| | - Hee Ho Park
- Department of Bioengineering,
Hanyang University, 222, Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Mikyung Shin
- Department of Biomedical Engineering,
SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence,
Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering,
Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulsan 44919, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering,
Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Se-Na Kim
- Research and Development Center,
MediArk Inc., 1, Chungdae-ro, Seowon-gu, Cheongju, Chungcheongbuk 28644, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering,
SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth,
SKKU Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Institute of Biotechnology and Bioengineering, College of Biotechnology and Bioengineering, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Biomaterials Research Center,
Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering,
SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence,
Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
- Biomedical Institute for Convergence, SKKU, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi 16419, Republic of Korea
| |
Collapse
|
9
|
Li X, Zhang Y, Wang C, Wang L, Ye Y, Xue R, Shi Y, Su Q, Zhu Y, Wang L. Drug-Loaded Biomimetic Carriers for Non-Hodgkin's Lymphoma Therapy: Advances and Perspective. ACS Biomater Sci Eng 2024; 10:723-742. [PMID: 38296812 DOI: 10.1021/acsbiomaterials.3c01480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Chemotherapy remains the mainstay of treatment for the lymphoma patient population, despite its relatively poor therapeutic results, high toxicity, and low specificity. With the advancement of biotechnology, the significance of drug-loading biomimetic materials in the medical field has become increasingly evident, attracting extensive attention from the scientific community and the pharmaceutical industry. Given that they can cater to the particular requirements of lymphoma patients, drug-loading biomimetic materials have recently become a potent and promising delivery approach for various applications. This review mainly reviews the recent advancements in the treatment of tumors with biological drug carrier-loaded drugs, outlines the mechanisms of lymphoma development and the diverse treatment modalities currently available, and discusses the merits and limitations of biological drug carriers. What is more, the practical application of biocarriers in tumors is explored by providing examples, and the possibility of loading such organisms with antilymphoma drugs for the treatment of lymphoma is conceived.
Collapse
Affiliation(s)
- Xiaoqi Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong China
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
- Linyi Key Laboratory of Nanomedicine, Linyi 276000, Shandong China
| | - Yu Zhang
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
- Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong China
| | - Chao Wang
- Department of Hematology, Linyi People's Hospital, Linyi 276000, Shandong China
| | - Liyuan Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong China
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
- Linyi Key Laboratory of Nanomedicine, Linyi 276000, Shandong China
| | - Yufu Ye
- Department of Hepatobiliary and Pancreatic Surgery, the First Affliliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, Zhejiang China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Hangzhou310000, Zhejiang China
| | - Renyu Xue
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
| | - Yuanwei Shi
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong China
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
| | - Quanping Su
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
| | - Yanxi Zhu
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
- Linyi Key Laboratory of Nanomedicine, Linyi 276000, Shandong China
- Key Laboratory for Translational Oncology, Xuzhou Medical University, Xuzhou 221000, Jiangsu China
| | - Lijuan Wang
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong China
- Linyi Key Laboratory of Tumor Biology, Linyi 276000, Shandong China
- Linyi Key Laboratory of Nanomedicine, Linyi 276000, Shandong China
- Key Laboratory for Translational Oncology, Xuzhou Medical University, Xuzhou 221000, Jiangsu China
| |
Collapse
|
10
|
Liu Y, Liu Y, Sun X, Wang Y, Du C, Bai J. Morphologically transformable peptide nanocarriers coloaded with doxorubicin and curcumin inhibit the growth and metastasis of hepatocellular carcinoma. Mater Today Bio 2024; 24:100903. [PMID: 38130427 PMCID: PMC10733681 DOI: 10.1016/j.mtbio.2023.100903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/03/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
In tumor treatment, the highly disordered vascular system and lack of accumulation of chemotherapeutic drugs in tumors severely limit the therapeutic role of nanocarriers. Smaller drug-containing nanoparticles (NPs) can better penetrate the tumor but are easily removed, which severely limits the tumor-killing properties of the drug. The chemotherapeutic medication doxorubicin (DOX) is highly toxic to the heart, but this toxicity can be effectively mitigated and the combined anticancer effect can be enhanced by clinically incorporating curcumin (CUR) as part of the dual therapy. We designed a small-molecule peptide, Pep1, containing a targeting peptide (CREKA) and a pH-responsive moiety. These NPs can target the blood vessels in tumor microthrombi and undergo a morphological shift in the tumor microenvironment. This process enhances the penetration and accumulation of drugs, ultimately improving the effectiveness of cancer treatment. In vitro and in vivo experiments demonstrated that this morphological transformation allowed rapid and effective drug release into tumors, the effective inhibition of tumor angiogenesis, and the promotion of tumor cell apoptosis, thus effectively killing tumor cells. Our findings provide a novel and simple approach to nhibit the growth and metastasis of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yun Liu
- School of Stomatology, Weifang Medical University, Weifang, 261053, China
| | - Yunxia Liu
- School of Stomatology, Weifang Medical University, Weifang, 261053, China
- Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, 261035, China
| | - Xinyu Sun
- School of Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Yue Wang
- School of Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Changqing Du
- Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, 261035, China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, China
| |
Collapse
|