51
|
Zhang Y, Dong P, Yang L. The role of nanotherapy in head and neck squamous cell carcinoma by targeting tumor microenvironment. Front Immunol 2023; 14:1189323. [PMID: 37292204 PMCID: PMC10244756 DOI: 10.3389/fimmu.2023.1189323] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) refers to a group of highly malignant and pathogenically complex tumors. Traditional treatment methods include surgery, radiotherapy, and chemotherapy. However, with advancements in genetics, molecular medicine, and nanotherapy, more effective and safer treatments have been developed. Nanotherapy, in particular, has the potential to be an alternative therapeutic option for HNSCC patients, given its advantageous targeting capabilities, low toxicity and modifiability. Recent research has highlighted the important role of the tumor microenvironment (TME) in the development of HNSCC. The TME is composed of various cellular components, such as fibroblasts, vascular endothelial cells, and immune cells, as well as non-cellular agents such as cytokines, chemokines, growth factors, extracellular matrix (ECM), and extracellular vesicles (EVs). These components greatly influence the prognosis and therapeutic efficacy of HNSCC, making the TME a potential target for treatment using nanotherapy. By regulating angiogenesis, immune response, tumor metastasis and other factors, nanotherapy can potentially alleviate HNSCC symptoms. This review aims to summarize and discuss the application of nanotherapy that targets HNSCC's TME. We highlight the therapeutic value of nanotherapy for HNSCC patients.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Radiation Oncology, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Pengbo Dong
- School of Energy and Power Engineering, Dalian University of Technology, Dalian, China
| | - Lu Yang
- Department of Internal Medicine, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
52
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Bradu P, Biswas A, Ganesan R, Renu K, Dey A, Vellingiri B, El Allali A, Alsamman AM, Zayed H, George Priya Doss C. Evolving strategies and application of proteins and peptide therapeutics in cancer treatment. Biomed Pharmacother 2023; 163:114832. [PMID: 37150032 DOI: 10.1016/j.biopha.2023.114832] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/18/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023] Open
Abstract
Several proteins and peptides have therapeutic potential and can be used for cancer therapy. By binding to cell surface receptors and other indicators uniquely linked with or overexpressed on tumors compared to healthy tissue, protein biologics enhance the active targeting of cancer cells, as opposed to the passive targeting of cells by conventional small-molecule chemotherapeutics. This study focuses on peptide medications that exist to slow or stop tumor growth and the spread of cancer, demonstrating the therapeutic potential of peptides in cancer treatment. As an alternative to standard chemotherapy, peptides that selectively kill cancer cells while sparing healthy tissue are developing. A mountain of clinical evidence supports the efficacy of peptide-based cancer vaccines. Since a single treatment technique may not be sufficient to produce favourable results in the fight against cancer, combination therapy is emerging as an effective option to generate synergistic benefits. One example of this new area is the use of anticancer peptides in combination with nonpeptidic cytotoxic drugs or the combination of immunotherapy with conventional therapies like radiation and chemotherapy. This review focuses on the different natural and synthetic peptides obtained and researched. Discoveries, manufacture, and modifications of peptide drugs, as well as their contemporary applications, are summarized in this review. We also discuss the benefits and difficulties of potential advances in therapeutic peptides.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| | - Pragya Bradu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Antara Biswas
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, South Korea
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077 Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco.
| | - Alsamman M Alsamman
- Department of Genome Mapping, Molecular Genetics, and Genome Mapping Laboratory, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - C George Priya Doss
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
53
|
Jiang T, Gonzalez KM, Cordova LE, Lu J. Nanotechnology-enabled gene delivery for cancer and other genetic diseases. Expert Opin Drug Deliv 2023; 20:523-540. [PMID: 37017558 PMCID: PMC10164135 DOI: 10.1080/17425247.2023.2200246] [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: 08/22/2022] [Accepted: 04/04/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Despite gene therapy is ideal for genetic abnormality-related diseases, the easy degradation, poor targeting, and inefficiency in entering targeted cells are plaguing the effective delivery of gene therapy. Viral and non-viral vectors have been used for delivering gene therapeutics in vivo by safeguarding nucleic acid agents to target cells and to reach the specific intracellular location. A variety of nanotechnology-enabled safe and efficient systems have been successfully developed to improve the targeting ability for effective therapeutic delivery of genetic drugs. AREAS COVERED In this review, we outline the multiple biological barriers associated with gene delivery process, and highlight recent advances to gene therapy strategy in vivo, including gene correction, gene silencing, gene activation and genome editing. We point out current developments and challenges exist of non-viral and viral vector systems in association with chemical and physical gene delivery technologies and their potential for the future. EXPERT OPINION This review focuses on the opportunities and challenges to various gene therapy strategy, with specific emphasis on overcoming the challenges through the development of biocompatibility and smart gene vectors for potential clinical application.
Collapse
Affiliation(s)
- Tong Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Leyla Estrella Cordova
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, Arizona, 85721, United States
- BIO5 Institute, The University of Arizona, Tucson, Arizona, 85721, United States
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, United States
| |
Collapse
|
54
|
Chen LH, Liang NW, Huang WY, Liu YC, Ho CY, Kuan CH, Huang YF, Wang TW. Supramolecular hydrogel for programmable delivery of therapeutics to cancer multidrug resistance. BIOMATERIALS ADVANCES 2023; 146:213282. [PMID: 36634378 DOI: 10.1016/j.bioadv.2023.213282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 12/25/2022] [Accepted: 01/02/2023] [Indexed: 01/08/2023]
Abstract
Multidrug resistance (MDR) has been considered as a major adversary in oncologic chemotherapy. To simultaneously overcome drug resistance and inhibit tumor growth, it is essential to develop a drug delivery system that can carry and release multiple therapeutic agents with spatiotemporal control. In this study, we developed a hydrogel containing an enzyme-cleavable peptide motif, with a network structure formed by 4-armed polyethylene glycol (PEG) crosslinked by complementary nucleic acid sequences. Hydrogen bond formation between nucleobase pairing allows the hydrogel to be injectable, and the peptide motif grants deliberate control over hydrogel degradation and the responsive drug release. Moreover, MDR-targeted siRNAs are complexed with stearyl-octaarginine (STR-R8), while doxorubicin (Dox) is intercalated with DNA and nanoclay structures in this hydrogel to enhance therapeutic efficacy and overcome MDR. The results show a successful configuration of a hydrogel network with in situ gelation property, injectability, and degradability in the presence of tumor-associated enzyme, MMP-2. The synergistic effect by combining MDR-targeted siRNAs and Dox manifests with the enhanced anti-cancer effect on drug resistant breast cancer cells in both in vitro and in vivo tumor models. We suggest that with the tailor-designed hydrogel system, multidrug resistance in tumor cells can be significantly inhibited by the co-delivery of multiple therapeutics with spatial-temporal control release.
Collapse
Affiliation(s)
- Liang-Hsin Chen
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan
| | - Nai-Wen Liang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan
| | - Wei-Yuan Huang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan
| | - Yu-Chung Liu
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan
| | - Chia-Yu Ho
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan
| | - Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei 10002, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10617, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Yu-Fen Huang
- Department of Biomedical Engineering and Environmental Sciences, and Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan.
| |
Collapse
|
55
|
Chang M. Matrix metalloproteinase profiling and their roles in disease. RSC Adv 2023; 13:6304-6316. [PMID: 36825288 PMCID: PMC9942564 DOI: 10.1039/d2ra07005g] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Matrix metalloproteinases (MMPs) play roles in remodelling of the extracellular matrix that occurs during morphogenesis, repair, and angiogenesis. Dysregulation of extracellular matrix remodelling can lead to cell proliferation, invasion, and tissue fibrosis. Identification of a specific MMP(s) in a disease has been challenging due to the presence of 24 closely-related human MMPs, each existing in three forms, of which only one is active and capable of catalysis. This review focuses on methods for MMP profiling, with particular emphasis on the batimastat affinity resin that binds only to the active forms of MMPs and related ADAMs (a disintegrin and metalloproteinases), which are then identified by mass spectrometry. Use of the batimastat affinity resin has identified targets for intervention in several human diseases.
Collapse
Affiliation(s)
- Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame Notre Dame IN 46556 USA
| |
Collapse
|
56
|
Yang Y, Zheng W, Tan W, Wu X, Dai Z, Li Z, Yan Z, Ji Y, Wang Y, Su W, Zhong S, Li Y, Sun Y, Li S, Huang W. Injectable MMP1-sensitive microspheres with spatiotemporally controlled exosome release promote neovascularized bone healing. Acta Biomater 2023; 157:321-336. [PMID: 36481504 DOI: 10.1016/j.actbio.2022.11.065] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Bone marrow mesenchymal stromal cell-derived exosomes (BMSC-Exos) can recruit stem cells for bone repair, with neovessels serving as the main migratory channel for stem cells to the injury site. However, existing exosome (Exo) delivery strategies cannot reach the angiogenesis phase following bone injury. To that end, an enzyme-sensitive Exo delivery material that responds to neovessel formation during the angiogenesis phase was designed in the present study to achieve spatiotemporally controlled Exo release. Herein, matrix metalloproteinase-1 (MMP1) was found to be highly expressed in neovascularized bone; as a result, we proposed an injectable MMP1-sensitive hydrogel microspheres (KGE) made using a microfluidic chip prepared by mixing self-assembling peptide (KLDL-MMP1), GelMA, and BMSC-Exos. The results revealed that KGE microspheres had a uniform diameter of 50-70 µm, ideal for minimally invasive injection and could release exosomes in response to MMP1 expression. In vitro experiments demonstrated that KGE had less cytotoxicity and could promote the migration and osteodifferentiation of BMSCs. Furthermore, in vivo experiments confirmed that KGE could promote bone repair during angiogenesis by recruiting CD90+ stem cells via neovessels. Collectively, our results suggest that injectable enzyme-responsive KGE microspheres could be a promising Exo-secreting material for accelerating neovascularized bone healing. STATEMENT OF SIGNIFICANCE: Exosomes can spread through blood vessels and activate stem cells to participate in bone repair, but under normal circumstances, exosomes lacking sustained-release delivery materials cannot be maintained until the angiogenesis phase. In this study, we found that MMP1 was highly expressed in neovascularized bone, then we proposed an MMP1-sensitive injectable microsphere that carries exosomes and responds temporally and spatially to neovascularization, which maximizes the ability of exosomes to recruit stem cells. Different from previous strategies that focus on promoting angiogenesis to accelerate bone healing, this is a brand new delivery strategy that is stimuli-responsive to neovessel formation. In addition, the preparation of self-assembled peptide microspheres by a microfluidic chip is also proposed for the first time.
Collapse
Affiliation(s)
- Yang Yang
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weihan Zheng
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei Tan
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
| | - Xiaoqi Wu
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Zhenning Dai
- Department of Stomatology, Guangdong Key Laboratory of Traditional Chinese Medicine Research and Development, Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou 510095, China
| | - Ziyue Li
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zi Yan
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuelun Ji
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
| | - Yilin Wang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weiwei Su
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shu Zhong
- Department of orthopedic, Dongguan People's Hospital, Dongguan 523058, China
| | - Yanbing Li
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongjian Sun
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China.
| | - Shiyu Li
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Wenhua Huang
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
57
|
Su Y, Jin G, Zhou H, Yang Z, Wang L, Mei Z, Jin Q, Lv S, Chen X. Development of stimuli responsive polymeric nanomedicines modulating tumor microenvironment for improved cancer therapy. MEDICAL REVIEW (2021) 2023; 3:4-30. [PMID: 37724108 PMCID: PMC10471091 DOI: 10.1515/mr-2022-0048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 02/16/2023] [Indexed: 09/20/2023]
Abstract
The complexity of the tumor microenvironment (TME) severely hinders the therapeutic effects of various cancer treatment modalities. The TME differs from normal tissues owing to the presence of hypoxia, low pH, and immune-suppressive characteristics. Modulation of the TME to reverse tumor growth equilibrium is considered an effective way to treat tumors. Recently, polymeric nanomedicines have been widely used in cancer therapy, because their synthesis can be controlled and they are highly modifiable, and have demonstrated great potential to remodel the TME. In this review, we outline the application of various stimuli responsive polymeric nanomedicines to modulate the TME, aiming to provide insights for the design of the next generation of polymeric nanomedicines and promote the development of polymeric nanomedicines for cancer therapy.
Collapse
Affiliation(s)
- Yuanzhen Su
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Materials Science and Engineering, Peking University, Beijing, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Guanyu Jin
- School of Materials Science and Engineering, Peking University, Beijing, China
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Huicong Zhou
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Zhaofan Yang
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Lanqing Wang
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Zi Mei
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Qionghua Jin
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Materials Science and Engineering, Peking University, Beijing, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
58
|
Noddeland HK, Lind M, Jensen LB, Petersson K, Skak-Nielsen T, Larsen FH, Malmsten M, Heinz A. Design and characterization of matrix metalloproteinase-responsive hydrogels for the treatment of inflammatory skin diseases. Acta Biomater 2023; 157:149-161. [PMID: 36526241 DOI: 10.1016/j.actbio.2022.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/23/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022]
Abstract
Enzyme-responsive hydrogels, formed by step growth photopolymerization of biscysteine peptide linkers with alkene functionalized polyethylene glycol, provide interesting opportunities as biomaterials and drug delivery systems. In this study, we developed stimuli-responsive, specific, and cytocompatible hydrogels for delivery of anti-inflammatory drugs for the treatment of inflammatory skin diseases. We designed peptide linkers with optimized sensitivity towards matrix metalloproteinases, a family of proteolytic enzymes overexpressed in the extracellular matrix of the skin during inflammation. The peptide linkers were crosslinked with branched 4-arm and 8-arm polyethylene glycols by thiol-norbornene photopolymerization, leading to the formation of a hydrogel network, in which the anti-inflammatory Janus kinase inhibitor tofacitinib citrate was incorporated. The hydrogels were extensively characterized by physical properties, in vitro release studies, cytocompatibility with fibroblasts, and anti-inflammatory efficacy testing in both an atopic dermatitis-like keratinocyte assay and an activated T-cell assay. The drug release was studied after single and multiple-time exposure to matrix metalloproteinase 9 to mimic inflammatory flare-ups. Drug release was found to be triggered by matrix metalloproteinase 9 and to depend on type of crosslinker and of the polyethylene glycol polymer, due to differences in architecture and swelling behavior. Moreover, swollen hydrogels showed elastic properties similar to those of extracellular matrix proteins in the dermis. Cell studies revealed limited cytotoxicity when fibroblasts and keratinocytes were exposed to the hydrogels or their enzymatic cleavage products. Taken together, our results suggest multi-arm polyethylene glycol hydrogels as promising matrix metalloproteinase-responsive drug delivery systems, with potential in the treatment of inflammatory skin disease. STATEMENT OF SIGNIFICANCE: Smart responsive drug delivery systems such as matrix metalloproteinase-responsive hydrogels are excellent candidates for the treatment of inflammatory skin diseases including psoriasis. Their release profile can be optimized to correspond to the patient's individual disease state by tuning formulation parameters and disease-related stimuli, providing personalized treatment solutions. However, insufficient cross-linking efficiency, low matrix metalloproteinase sensitivity, and undesirable drug release kinetics remain major challenges in the development of such drug delivery systems. In this study, we address shortcomings of previous work by designing peptide linkers with optimized sensitivity towards matrix metalloproteinases and high cross-linking efficiencies. We further provide a proof-of-concept for the usability of the hydrogels in inflammatory skin conditions by employing a drug release set-up simulating inflammatory flare-ups.
Collapse
Affiliation(s)
- Heidi Kyung Noddeland
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark; Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Marianne Lind
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Louise Bastholm Jensen
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Karsten Petersson
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Tine Skak-Nielsen
- Cells & Assays, In vitro Biology, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Flemming Hofmann Larsen
- Advanced Analytical and Structural Chemistry, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Martin Malmsten
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark; Department of Physical Chemistry 1, University of Lund, SE-22100 Lund, Sweden
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark.
| |
Collapse
|
59
|
Wei Z, Zheng D, Pi W, Qiu Y, Xia K, Guo W. Isoquercitrin restrains the proliferation and promotes apoptosis of human osteosarcoma cells by inhibiting the Wnt/β-catenin pathway. J Bone Oncol 2023; 38:100468. [PMID: 36685044 PMCID: PMC9846017 DOI: 10.1016/j.jbo.2023.100468] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/26/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Currently, chemotherapeutic drugs are widely used for the treatment of osteosarcoma. However, many of these drugs exhibit shortcomings such as poor efficacy, high toxicity, and tolerance. Isoquercitrin (ISO) is a traditional Chinese medicine that has been proved to exert good therapeutic effects on various tumors; however, its role in osteosarcoma has not been reported. Here, we observed that ISO exerted a marked inhibitory effect on the occurrence and development of osteosarcoma in a time- and dose-dependent manner. First, we determined that ISO significantly inhibited proliferation, induced EMT-related migration and invasion and induced apoptosis of osteosarcoma cells in vitro. Concurrently, we also observed that both β-catenin and its downstream genes (c-Myc, CyclinD1, and Survivin) were significantly down-regulated. To verify if the anti-tumor effect of ISO was related to the Wnt/β-catenin signaling pathway, we altered the protein expression level of β-catenin using recombinant lentivirus, then we observed that the effects of ISO on the proliferation, metastasis, and apoptosis of osteosarcoma cells were significantly reversed. Additionally, we used a nude mouse xenograft model and observed that ISO significantly inhibited the growth of osteosarcoma and improved the survival rate of the animal models. In conclusion, this study demonstrates that ISO can exert anti-tumor effects in part by inhibiting the Wnt/β-catenin signaling pathway, thus providing a new potential therapeutic strategy for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Zhun Wei
- Department of Orthopaedics, Renmin Hospital of Wuhan University, Hubei Province, Wuhan 430060, China
| | - Di Zheng
- Department of Orthopaedics, Renmin Hospital of Wuhan University, Hubei Province, Wuhan 430060, China
| | - Wenfeng Pi
- Department of Orthopaedics, Chinese Traditional Medicine of Daye Hospital, Hubei Province, Daye 435100, China
| | - Yonglong Qiu
- Department of Orthopaedics, Renmin Hospital of Wuhan University, Hubei Province, Wuhan 430060, China
| | - Kezhou Xia
- Department of Orthopaedics, Renmin Hospital of Wuhan University, Hubei Province, Wuhan 430060, China,Corresponding authors at: Department of Orthopaedics, Renmin hospital of Wuhan university, 238 Jiefang Road, Wuhan 430060, China.
| | - Weichun Guo
- Department of Orthopaedics, Renmin Hospital of Wuhan University, Hubei Province, Wuhan 430060, China,Corresponding authors at: Department of Orthopaedics, Renmin hospital of Wuhan university, 238 Jiefang Road, Wuhan 430060, China.
| |
Collapse
|
60
|
Cheng D, Wen Z, Chen H, Lin S, Zhang W, Tang X, Wu W. Hepatocyte-targeting and tumor microenvironment-responsive liposomes for enhanced anti-hepatocarcinoma efficacy. Drug Deliv 2022; 29:2995-3008. [PMID: 36104946 PMCID: PMC9487930 DOI: 10.1080/10717544.2022.2122635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
To increase the antitumor drug concentration in the liver tumor site and improve the therapeutic effects, a functionalized liposome (PPP-LIP) with tumor targetability and enhanced internalization after matrix metalloproteinase-2 (MMP2)-triggered cell-penetrating peptide (TATp) exposure was modified with myrcludex B (a synthetic HBV preS-derived lipopeptide endowed with compelling liver tropism) for liver tumor-specific delivery. After intravenous administration, PPP-LIP was mediated by myrcludex B to reach the hepatocyte surface. The MMP2-overexpressing tumor microenvironment deprotected PEG, exposing it to TATp, facilitating tumor penetration and subsequent efficient destruction of tumor cells. In live imaging of small animals and cellular uptake, PPP-LIP was taken up much more than typical unmodified liposomes in the ICR mouse liver and liver tumor cells. Hydroxycamptothecin (HCPT)-loaded PPP-LIP showed a better antitumor effect than commercially available HCPT injections among MTT, three-dimensional (3 D) tumor ball, and tumor-bearing nude mouse experiments. Our findings indicated that PPP-LIP nanocarriers could be a promising tumor-targeted medication delivery strategy for treating liver cancers with elevated MMP2 expression.
Collapse
Affiliation(s)
- Dongliang Cheng
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Hui Chen
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Shiyuan Lin
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Xin Tang
- School of Public Health, Guilin Medical University, Guilin, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin, China
| |
Collapse
|
61
|
Shao Y, Xiang L, Zhang W, Chen Y. Responsive shape-shifting nanoarchitectonics and its application in tumor diagnosis and therapy. J Control Release 2022; 352:600-618. [PMID: 36341936 DOI: 10.1016/j.jconrel.2022.10.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Nanodrug delivery system has a great application in the treatment of solid tumors by virtue of EPR effect, though its success in clinics is still limited by its poor extravasation, small intratumoral accumulation, and weak tumor penetration. The shape of nanoparticles (NPs) greatly affects their circulation time, flow behavior, intratumoral amassing, cell internalization as well as tumor tissue penetration. Generally, short nanorods and 100-200 nm spherical nanocarriers possess nice circulation behaviors, nanorods and nanofibers with a large aspect ratio (AR) cumulate well at tumor sites, and tiny nanospheres/disks (< 50 nm) and short nanorods with a low AR achieve a favorable tumor tissue penetration. The AR and surface evenness of NPs also tune their cell contact, cell ingestion, and drug accumulation at tumor sites. Therefore, adopting stimulus-responsive shape-switching (namely, shape-shifting nanoarchitectonics) can not only ensure a good circulation and extravasation for NPs, but also and more importantly, promote their amassing, retention, and penetration in tumor tissues to maximize therapeutic efficacy. Here we review the recently developed shape-switching nanoarchitectonics of antitumoral NPs based on stimulus-responsiveness, demonstrate how successful they are in tumor shrinking and elimination, and provide new ideas for the optimization of anticancer nanotherapeutics.
Collapse
Affiliation(s)
- Yaru Shao
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Li Xiang
- Hengyang Medical School, University of South China, Hengyang 410001, China
| | - Wenhui Zhang
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Yuping Chen
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| |
Collapse
|
62
|
Developments on the Smart Hydrogel-Based Drug Delivery System for Oral Tumor Therapy. Gels 2022; 8:gels8110741. [PMID: 36421563 PMCID: PMC9689473 DOI: 10.3390/gels8110741] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
At present, an oral tumor is usually treated by surgery combined with preoperative or postoperative radiotherapies and chemotherapies. However, traditional chemotherapies frequently result in substantial toxic side effects, including bone marrow suppression, malfunction of the liver and kidneys, and neurotoxicity. As a new local drug delivery system, the smart drug delivery system based on hydrogel can control drug release in time and space, and effectively alleviate or avoid these problems. Environmentally responsive hydrogels for smart drug delivery could be triggered by temperature, photoelectricity, enzyme, and pH. An overview of the most recent research on smart hydrogels and their controlled-release drug delivery systems for the treatment of oral cancer is given in this review. It is anticipated that the local drug release method and environment-responsive benefits of smart hydrogels will offer a novel technique for the low-toxicity and highly effective treatment of oral malignancy.
Collapse
|
63
|
Zhou Q, Huang J, Hao L, Geng Y, Xu C, Zhou Z, Tang J, Zhou R, Shen Y. Hydrophobicity Effects of γ-Glutamyl Transpeptidase-Responsive Polymers on the Catalytic Activity and Transcytosis Efficacy. Bioconjug Chem 2022; 33:2132-2142. [DOI: 10.1021/acs.bioconjchem.2c00391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianxiang Huang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lingqiao Hao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yu Geng
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Changhuo Xu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
64
|
Ding L, Liang M, Li C, Ji X, Zhang J, Xie W, Reis RL, Li FR, Gu S, Wang Y. Design Strategies of Tumor-Targeted Delivery Systems Based on 2D Nanomaterials. SMALL METHODS 2022; 6:e2200853. [PMID: 36161304 DOI: 10.1002/smtd.202200853] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/11/2022] [Indexed: 06/16/2023]
Abstract
Conventional chemotherapy and radiotherapy are nonselective and nonspecific for cell killing, causing serious side effects and threatening the lives of patients. It is of great significance to develop more accurate tumor-targeting therapeutic strategies. Nanotechnology is in a leading position to provide new treatment options for cancer, and it has great potential for selective targeted therapy and controlled drug release. 2D nanomaterials (2D NMs) have broad application prospects in the field of tumor-targeted delivery systems due to their special structure-based functions and excellent optical, electrical, and thermal properties. This review emphasizes the design strategies of tumor-targeted delivery systems based on 2D NMs from three aspects: passive targeting, active targeting, and tumor-microenvironment targeting, in order to promote the rational application of 2D NMs in clinical practice.
Collapse
Affiliation(s)
- Lin Ding
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
- The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, 518055, China
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, Guangdong, 518055, China
- Guangdong Engineering Technology Research Centerof Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Minli Liang
- The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, 518055, China
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, Guangdong, 518055, China
- Guangdong Engineering Technology Research Centerof Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Chenchen Li
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinting Ji
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Weifen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials Biodegradables and Biomimetics, University of Minho, Guimarães, 4805-017, Portugal
| | - Fu-Rong Li
- The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, 518055, China
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, Guangdong, 518055, China
- Guangdong Engineering Technology Research Centerof Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Shuo Gu
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
| | - Yanli Wang
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
| |
Collapse
|
65
|
Zheng X, Zhao J, Wang S, Hu L. Research Progress of Antioxidant Nanomaterials for Acute Pancreatitis. Molecules 2022; 27:7238. [PMID: 36364064 PMCID: PMC9658789 DOI: 10.3390/molecules27217238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
Acute pancreatitis (AP) is a complex inflammatory disease caused by multiple etiologies, the pathogenesis of which has not been fully elucidated. Oxidative stress is important for the regulation of inflammation-related signaling pathways, the recruitment of inflammatory cells, the release of inflammatory factors, and other processes, and plays a key role in the occurrence and development of AP. In recent years, antioxidant therapy that suppresses oxidative stress by scavenging reactive oxygen species has become a research highlight of AP. However, traditional antioxidant drugs have problems such as poor drug stability and low delivery efficiency, which limit their clinical translation and applications. Nanomaterials bring a brand-new opportunity for the antioxidant treatment of AP. This review focuses on the multiple advantages of nanomaterials, including small size, good stability, high permeability, and long retention effect, which can be used not only as effective carriers of traditional antioxidant drugs but also directly as antioxidants. In this review, after first discussing the association between oxidative stress and AP, we focused on summarizing the literature related to antioxidant nanomaterials for the treatment of AP and highlighting the effects of these nanomaterials on the indicators related to oxidative stress in pathological states, aiming to provide references for follow-up research and promote clinical application.
Collapse
Affiliation(s)
- Xiaoyi Zheng
- Ningxia Medical University, Postgraduate Training Base in Shanghai Gongli Hospital, Pudong New Area, No. 219 Miao Pu Road, Shanghai 200135, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Jiulong Zhao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China
| | - Lianghao Hu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
66
|
Austin MJ, Schunk H, Watkins C, Ling N, Chauvin J, Morton L, Rosales AM. Fluorescent Peptomer Substrates for Differential Degradation by Metalloproteases. Biomacromolecules 2022; 23:4909-4923. [PMID: 36269900 DOI: 10.1021/acs.biomac.2c01077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Proteases, especially MMPs, are attractive biomarkers given their central role in both physiological and pathological processes. Distinguishing MMP activity with degradable substrates, however, is a difficult task due to overlapping substrate specificity profiles. Here, we developed a system of peptomers (peptide-peptoid hybrids) to probe the impact of non-natural residues on MMP specificity for an MMP peptide consensus sequence. Peptoids are non-natural, N-substituted glycines with a large side-chain diversity. Given the presence of a hallmark proline residue in the P3 position of MMP consensus sequences, we hypothesized that peptoids may offer N-substituted alternatives to generate differential interactions with MMPs. To investigate this hypothesis, peptomer substrates were exposed to five different MMPs, as well as bacterial collagenase, and monitored by fluorescence resonance energy transfer and liquid chromatography-mass spectrometry to determine the rate of cleavage and the composition of degraded fragments, respectively. We found that peptoid residues are well tolerated in the P3 and P3' substrate sites and that the identity of the peptoid in these sites displays a moderate influence on the rate of cleavage. However, peptoid residues were even better tolerated in the P1 substrate site where activity was more strongly correlated with side-chain identity than side-chain position. All MMPs explored demonstrated similar trends in specificity for the peptomers but exhibited different degrees of variability in proteolytic rate. These kinetic profiles served as "fingerprints" for the proteases and yielded separation by multivariate data analysis. To further demonstrate the practical application of this tunability in degradation kinetics, peptomer substrates were tethered into hydrogels and released over distinct timescales. Overall, this work represents a significant step toward the design of probes that maximize differential MMP behavior and presents design rules to tune degradation kinetics with peptoid substitutions, which has promising implications for diagnostic and prognostic applications using array-based sensors.
Collapse
Affiliation(s)
- Mariah J Austin
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas78712, United States
| | - Hattie Schunk
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas78712, United States.,Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas78712, United States
| | - Carolyn Watkins
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas78712, United States
| | - Natalie Ling
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas78712, United States
| | - Jeremy Chauvin
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas78712, United States
| | - Logan Morton
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas78712, United States
| | - Adrianne M Rosales
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas78712, United States
| |
Collapse
|
67
|
Wan D, Zhu Q, Zhang J, Chen X, Li F, Liu Y, Pan J. Intracellular and extracellular enzymatic responsive micelle for intelligent therapy of cancer. NANO RESEARCH 2022; 16:2851-2858. [PMID: 36258757 PMCID: PMC9561310 DOI: 10.1007/s12274-022-4967-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 05/07/2023]
Abstract
Recently, the incidence of cancer keeps increasing, seriously endangers human health, and has evolved into the main culprit of human death. Conventional chemotherapeutic drugs, such as paclitaxel and doxorubicin (DOX), have some disadvantages, including low therapeutic effect, poor water solubility, high toxic side effects, short blood circulation time in the body, and so on. To improve the anti-tumor effect of the drug in vivo and reduce its side effects on the body, researchers have designed and developed a variety of responsive nanocarriers. In this work, we synthesized D-α-tocopherol polyethylene glycol 3350 succinate (TPGS3350)-Gly-Pro-Leu-Gly-Val-Arg (GPLGVR)-DOX (TPD) prodrugs in response to extracellular enzymes of matrix metalloproteinase (MMP-9) in the tumor microenvironment and FA-Asp-Glu-Val-Asp (DEVD)-DOX (FPD) prodrugs responsive to intracellular enzymes of caspase-3. Then, intracellular and extracellular enzyme-responsive TPD&FPD micelles with DOX (TPD&FPD&D) were successfully prepared through dialysis method. The outer layer of TPGS3350 can prolong the blood circulation time of micelles in vivo, followed by accumulation of micelles at tumor tissue through enhanced permeability and retention (EPR) effect. The peptide of GPLGVR can be cleaved by MMP-9 enzymes to remove the outer layer of TPGS3350, exposing the targeting molecule of folate, and then the micelles are engulfed by tumor cells through folate receptor-mediated endocytosis. After entering the tumor cells, the free DOX loaded in the micelles is released, which induces tumor cell apoptosis to activate caspase-3 in the cells, cutting the peptide DEVD to accelerate the intracellular release of the DOX, which further enhances cytotoxicity to improve antitumor effect. Electronic Supplementary Material Supplementary material () is available in the online version of this article at 10.1007/s12274-022-4967-1.
Collapse
Affiliation(s)
- Dong Wan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387 China
| | - Qinan Zhu
- School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387 China
| | - Jianxin Zhang
- School of Chemistry, Tiangong University, Tianjin, 300387 China
| | - Xi Chen
- School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387 China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190 China
| | - Yi Liu
- School of Chemistry, Tiangong University, Tianjin, 300387 China
| | - Jie Pan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387 China
| |
Collapse
|
68
|
Lee H, Park G, Kim S, Son B, Joo J, Park HH, Park TH. Enhancement of anti-tumor activity in melanoma using arginine deiminase fused with 30Kc19α protein. Appl Microbiol Biotechnol 2022; 106:7531-7545. [PMID: 36227339 DOI: 10.1007/s00253-022-12218-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022]
Abstract
Arginine deiminase (ADI) is a microbial-derived enzyme which catalyzes the conversion of L-arginine into L-citrulline. ADI originating from Mycoplasma has been reported to present anti-tumor activity against arginine-auxotrophic tumors, including melanoma. Melanoma cells are sensitive to arginine depletion due to reduced expression of argininosuccinate synthase 1 (ASS1), a key enzyme for arginine biosynthesis. However, clinical applications of recombinant ADI for melanoma treatment present some limitations. Since recombinant ADI is not human-derived, it shows instability, proteolytic degradation, and antigenicity in human serum. In addition, there is a problem of drug resistance issue due to the intracellular expression of once-silenced ASS1. Moreover, recombinant ADI proteins are mainly expressed as inclusion body forms in Escherichia coli and require a time-consuming refolding process to turn them back into active form. Herein, we propose fusion of recombinant ADI from Mycoplasma hominis and 30Kc19α, a cell-penetrating protein which also increases stability and soluble expression of cargo proteins, to overcome these problems. We inserted matrix metalloproteinase-2 cleavable linker between ADI and 30Kc19α to increase enzyme activity in melanoma cells. Compared to ADI, ADI-LK-30Kc19α showed enhanced solubility, stability, and cell penetration. The fusion protein demonstrated selective cytotoxicity and reduced drug resistance in melanoma cells, thus would be a promising strategy for the improved efficacy in melanoma treatment. KEY POINTS: • Fusion of ADI with 30Kc19α enhances soluble expression and productivity of recombinant ADI in E. coli • 30Kc19α protects ADI from the proteolytic degradation by shielding effect, helping ADI to remain active • Intracellular delivery of ADI by 30Kc19α overcomes ADI resistance in melanoma cells by degrading intracellularly expressed arginine.
Collapse
Affiliation(s)
- Haein Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea
| | - Geunhwa Park
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Seulha Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea
| | - Boram Son
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea. .,Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea.
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea. .,Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea. .,BioMax/N-Bio Institute, Institute of Bioengineering, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
69
|
Quader S, Van Guyse JFR. Bioresponsive Polymers for Nanomedicine-Expectations and Reality! Polymers (Basel) 2022; 14:3659. [PMID: 36080733 PMCID: PMC9460233 DOI: 10.3390/polym14173659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 12/18/2022] Open
Abstract
Bioresponsive polymers in nanomedicine have been widely perceived to selectively activate the therapeutic function of nanomedicine at diseased or pathological sites, while sparing their healthy counterparts. This idea can be described as an advanced version of Paul Ehrlich's magic bullet concept. From that perspective, the inherent anomalies or malfunction of the pathological sites are generally targeted to allow the selective activation or sensory function of nanomedicine. Nonetheless, while the primary goals and expectations in developing bioresponsive polymers are to elicit exclusive selectivity of therapeutic action at diseased sites, this remains difficult to achieve in practice. Numerous research efforts have been undertaken, and are ongoing, to tackle this fine-tuning. This review provides a brief introduction to key stimuli with biological relevance commonly featured in the design of bioresponsive polymers, which serves as a platform for critical discussion, and identifies the gap between expectations and current reality.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan
| | - Joachim F. R. Van Guyse
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan
- Leiden Academic Center for Drug Research (LACDR), Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
70
|
Wan D, Liu Y, Guo X, Zhang J, Pan J. Intelligent Drug Delivery by Peptide-Based Dual-Function Micelles. Int J Mol Sci 2022; 23:ijms23179698. [PMID: 36077102 PMCID: PMC9456463 DOI: 10.3390/ijms23179698] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
To endow the polymeric prodrug with smart properties through a safe and simple method, matrix metalloproteinase (MMPs) responsive peptide GPLGVRGDG was introduced into the block copolymer to prepare TPGS3350-GPLGVRGDG-DOX&DOX micelles, where TPGS3350 is D-α-tocopheryl polyethylene glycol 3350 succinate. During the doxorubicin delivery, the cleavage of the peptide chain triggers de-PEGylation, and the remaining VRGDG sequence was retained on the surface of the micelles, which can act as a ligand to facilitate cell uptake. Moreover, the cytotoxicity of TPGS3350-GPLGVRGDG-DOX&DOX micelles against 4T1 cells was significantly improved, compared with TPGS3350-GPLGVRG-DOX&DOX micelles and TPGS3350-DOX&DOX micelles. During in vivo studies, TPGS3350-GPLGVRGDG-DOX&DOX micelles exhibited good anticancer efficacy with long circulation in the body and more efficient accumulation at the tumor site. Therefore, TPGS3350-GPLGVRGDG-DOX&DOX micelles have improved antitumor activity and reduced toxic side effects. This work opens new potential for exploring the strategy of drug delivery in clinical applications.
Collapse
Affiliation(s)
- Dong Wan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Yujun Liu
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Xinhao Guo
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Jianxin Zhang
- School of Chemistry, Tiangong University, Tianjin 300387, China
- Correspondence: (J.Z.); (J.P.)
| | - Jie Pan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
- Correspondence: (J.Z.); (J.P.)
| |
Collapse
|
71
|
Fang T, Cao X, Ibnat M, Chen G. Stimuli-responsive nanoformulations for CRISPR-Cas9 genome editing. J Nanobiotechnology 2022; 20:354. [PMID: 35918694 PMCID: PMC9344766 DOI: 10.1186/s12951-022-01570-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/22/2022] [Indexed: 12/07/2022] Open
Abstract
The CRISPR-Cas9 technology has changed the landscape of genome editing and has demonstrated extraordinary potential for treating otherwise incurable diseases. Engineering strategies to enable efficient intracellular delivery of CRISPR-Cas9 components has been a central theme for broadening the impact of the CRISPR-Cas9 technology. Various non-viral delivery systems for CRISPR-Cas9 have been investigated given their favorable safety profiles over viral systems. Many recent efforts have been focused on the development of stimuli-responsive non-viral CRISPR-Cas9 delivery systems, with the goal of achieving efficient and precise genome editing. Stimuli-responsive nanoplatforms are capable of sensing and responding to particular triggers, such as innate biological cues and external stimuli, for controlled CRISPR-Cas9 genome editing. In this Review, we overview the recent advances in stimuli-responsive nanoformulations for CRISPR-Cas9 delivery, highlight the rationale of stimuli and formulation designs, and summarize their biomedical applications.
Collapse
Affiliation(s)
- Tianxu Fang
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Xiaona Cao
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada.,School of Nursing, Tianjin Medical University, Tianjin, China
| | - Mysha Ibnat
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada. .,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
72
|
Chaves Filho AJM, Mottin M, Lós DB, Andrade CH, Macedo DS. The tetrapartite synapse in neuropsychiatric disorders: Matrix metalloproteinases (MMPs) as promising targets for treatment and rational drug design. Biochimie 2022; 201:79-99. [PMID: 35931337 DOI: 10.1016/j.biochi.2022.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 06/26/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023]
Abstract
Inflammation and an exacerbated immune response are widely accepted contributing mechanisms to the genesis and progression of major neuropsychiatric disorders. However, despite the impressive advances in understanding the neurobiology of these disorders, there is still no approved drug directly linked to the regulation of inflammation or brain immune responses. Importantly, matrix metalloproteinases (MMPs) comprise a group of structurally related endopeptidases primarily involved in remodeling extracellular matrix (ECM). In the central nervous system (CNS), these proteases control synaptic plasticity and strength, patency of the blood-brain barrier, and glia-neuron interactions through cleaved and non-cleaved mediators. Several pieces of evidence have pointed to a complex scenario of MMPs dysregulation triggered by neuroinflammation. Furthermore, major psychiatric disorders' affective symptoms and neurocognitive abnormalities are related to MMPs-mediated ECM changes and neuroglia activation. In the past decade, research efforts have been directed to broad-spectrum MMPs inhibitors with frustrating clinical results. However, in the light of recent advances in combinatorial chemistry and drug design technologies, specific and CNS-oriented MMPs modulators have been proposed as a new frontier of therapy for regulating ECM properties in the CNS. Therefore, here we aim to discuss the state of the art of MMPs and ECM abnormalities in major neuropsychiatric disorders, namely depression, bipolar disorder, and schizophrenia, the possible neuro-immune interactions involved in this complex scenario of MMPs dysregulation and propose these endopeptidases as promising targets for rational drug design.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Melina Mottin
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Deniele Bezerra Lós
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
73
|
Li Z, Lai X, Fu S, Ren L, Cai H, Zhang H, Gu Z, Ma X, Luo K. Immunogenic Cell Death Activates the Tumor Immune Microenvironment to Boost the Immunotherapy Efficiency. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201734. [PMID: 35652198 PMCID: PMC9353475 DOI: 10.1002/advs.202201734] [Citation(s) in RCA: 259] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/21/2022] [Indexed: 02/05/2023]
Abstract
Tumor immunotherapy is only effective in a fraction of patients due to a low response rate and severe side effects, and these challenges of immunotherapy in clinics can be addressed through induction of immunogenic cell death (ICD). ICD is elicited from many antitumor therapies to release danger associated molecular patterns (DAMPs) and tumor-associated antigens to facilitate maturation of dendritic cells (DCs) and infiltration of cytotoxic T lymphocytes (CTLs). The process can reverse the tumor immunosuppressive microenvironment to improve the sensitivity of immunotherapy. Nanostructure-based drug delivery systems (NDDSs) are explored to induce ICD by incorporating therapeutic molecules for chemotherapy, photosensitizers (PSs) for photodynamic therapy (PDT), photothermal conversion agents for photothermal therapy (PTT), and radiosensitizers for radiotherapy (RT). These NDDSs can release loaded agents at a right dose in the right place at the right time, resulting in greater effectiveness and lower toxicity. Immunotherapeutic agents can also be combined with these NDDSs to achieve the synergic antitumor effect in a multi-modality therapeutic approach. In this review, NDDSs are harnessed to load multiple agents to induce ICD by chemotherapy, PDT, PTT, and RT in combination of immunotherapy to promote the therapeutic effect and reduce side effects associated with cancer treatment.
Collapse
Affiliation(s)
- Zhilin Li
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiaoqin Lai
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Shiqin Fu
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Long Ren
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Hao Cai
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Hu Zhang
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Amgen Bioprocessing CentreKeck Graduate InstituteClaremontCA91711USA
| | - Zhongwei Gu
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xuelei Ma
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Kui Luo
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Functional and Molecular Imaging Key Laboratory of Sichuan Provinceand Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengdu610041China
| |
Collapse
|
74
|
Jiao L, Dong Q, Zhai W, Zhao W, Shi P, Wu Y, Zhou X, Gao Y. A PD-L1 and VEGFR2 dual targeted peptide and its combination with irradiation for cancer immunotherapy. Pharmacol Res 2022; 182:106343. [DOI: 10.1016/j.phrs.2022.106343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/03/2022] [Accepted: 07/01/2022] [Indexed: 10/17/2022]
|
75
|
Mansurov A, Hosseinchi P, Chang K, Lauterbach AL, Gray LT, Alpar AT, Budina E, Slezak AJ, Kang S, Cao S, Solanki A, Gomes S, Williford JM, Swartz MA, Mendoza JL, Ishihara J, Hubbell JA. Masking the immunotoxicity of interleukin-12 by fusing it with a domain of its receptor via a tumour-protease-cleavable linker. Nat Biomed Eng 2022; 6:819-829. [PMID: 35534574 DOI: 10.1038/s41551-022-00888-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 03/29/2022] [Indexed: 02/05/2023]
Abstract
Immune-checkpoint inhibitors have shown modest efficacy against immunologically 'cold' tumours. Interleukin-12 (IL-12)-a cytokine that promotes the recruitment of immune cells into tumours as well as immune cell activation, also in cold tumours-can cause severe immune-related adverse events in patients. Here, by exploiting the preferential overexpression of proteases in tumours, we show that fusing a domain of the IL-12 receptor to IL-12 via a linker cleavable by tumour-associated proteases largely restricts the pro-inflammatory effects of IL-12 to tumour sites. In mouse models of subcutaneous adenocarcinoma and orthotopic melanoma, masked IL-12 delivered intravenously did not cause systemic IL-12 signalling and eliminated systemic immune-related adverse events, led to potent therapeutic effects via the remodelling of the immune-suppressive microenvironment, and rendered cold tumours responsive to immune-checkpoint inhibition. We also show that masked IL-12 is activated in tumour lysates from patients. Protease-sensitive masking of potent yet toxic cytokines may facilitate their clinical translation.
Collapse
Affiliation(s)
- Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Peyman Hosseinchi
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kevin Chang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Abigail L Lauterbach
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Laura T Gray
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aaron T Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Erica Budina
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Anna J Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Seounghun Kang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL, USA
| | - Suzana Gomes
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | - Melody A Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| | - Juan L Mendoza
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Department of Biochemistry and Molecular Biology, Chicago, IL, USA
| | - Jun Ishihara
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Department of Bioengineering, Imperial College London, London, UK.
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
76
|
Cong X, Chen J, Xu R. Recent Progress in Bio-Responsive Drug Delivery Systems for Tumor Therapy. Front Bioeng Biotechnol 2022; 10:916952. [PMID: 35845404 PMCID: PMC9277442 DOI: 10.3389/fbioe.2022.916952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/09/2022] [Indexed: 12/24/2022] Open
Abstract
Spatially- and/or temporally-controlled drug release has always been the pursuit of drug delivery systems (DDSs) to achieve the ideal therapeutic effect. The abnormal pathophysiological characteristics of the tumor microenvironment, including acidosis, overexpression of special enzymes, hypoxia, and high levels of ROS, GSH, and ATP, offer the possibility for the design of stimulus-responsive DDSs for controlled drug release to realize more efficient drug delivery and anti-tumor activity. With the help of these stimulus signals, responsive DDSs can realize controlled drug release more precisely within the local tumor site and decrease the injected dose and systemic toxicity. This review first describes the major pathophysiological characteristics of the tumor microenvironment, and highlights the recent cutting-edge advances in DDSs responding to the tumor pathophysiological environment for cancer therapy. Finally, the challenges and future directions of bio-responsive DDSs are discussed.
Collapse
Affiliation(s)
- Xiufeng Cong
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ran Xu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Ran Xu,
| |
Collapse
|
77
|
Fan Z, Jiang C, Wang Y, Wang K, Marsh J, Zhang D, Chen X, Nie L. Engineered extracellular vesicles as intelligent nanosystems for next-generation nanomedicine. NANOSCALE HORIZONS 2022; 7:682-714. [PMID: 35662310 DOI: 10.1039/d2nh00070a] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Extracellular vesicles (EVs), as natural carriers of bioactive cargo, have a unique micro/nanostructure, bioactive composition, and characteristic morphology, as well as fascinating physical, chemical and biochemical features, which have shown promising application in the treatment of a wide range of diseases. However, native EVs have limitations such as lack of or inefficient cell targeting, on-demand delivery, and therapeutic feedback. Recently, EVs have been engineered to contain an intelligent core, enabling them to (i) actively target sites of disease, (ii) respond to endogenous and/or exogenous signals, and (iii) provide treatment feedback for optimal function in the host. These advances pave the way for next-generation nanomedicine and offer promise for a revolution in drug delivery. Here, we summarise recent research on intelligent EVs and discuss the use of "intelligent core" based EV systems for the treatment of disease. We provide a critique about the construction and properties of intelligent EVs, and challenges in their commercialization. We compare the therapeutic potential of intelligent EVs to traditional nanomedicine and highlight key advantages for their clinical application. Collectively, this review aims to provide a new insight into the design of next-generation EV-based theranostic platforms for disease treatment.
Collapse
Affiliation(s)
- Zhijin Fan
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P. R. China
| | - Cheng Jiang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Yichao Wang
- Department of Clinical Laboratory Medicine, Tai Zhou Central Hospital (Taizhou University Hospital), Taizhou 318000, P. R. China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Jade Marsh
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an 710049, P. R. China.
| | - Liming Nie
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P. R. China
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| |
Collapse
|
78
|
Gonzalez-Avila G, Sommer B, García-Hernandez AA, Ramos C, Flores-Soto E. Nanotechnology and Matrix Metalloproteinases in Cancer Diagnosis and Treatment. Front Mol Biosci 2022; 9:918789. [PMID: 35720130 PMCID: PMC9198274 DOI: 10.3389/fmolb.2022.918789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/18/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer is still one of the leading causes of death worldwide. This great mortality is due to its late diagnosis when the disease is already at advanced stages. Although the efforts made to develop more effective treatments, around 90% of cancer deaths are due to metastasis that confers a systemic character to the disease. Likewise, matrix metalloproteinases (MMPs) are endopeptidases that participate in all the events of the metastatic process. MMPs’ augmented concentrations and an increased enzymatic activity have been considered bad prognosis markers of the disease. Therefore, synthetic inhibitors have been created to block MMPs’ enzymatic activity. However, they have been ineffective in addition to causing considerable side effects. On the other hand, nanotechnology offers the opportunity to formulate therapeutic agents that can act directly on a target cell, avoiding side effects and improving the diagnosis, follow-up, and treatment of cancer. The goal of the present review is to discuss novel nanotechnological strategies in which MMPs are used with theranostic purposes and as therapeutic targets to control cancer progression.
Collapse
Affiliation(s)
- Georgina Gonzalez-Avila
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Ciudad de México, Mexico
- *Correspondence: Georgina Gonzalez-Avila,
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Ciudad de México, Mexico
| | - A. Armando García-Hernandez
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Ciudad de México, Mexico
| | - Carlos Ramos
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Ciudad de México, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
79
|
Li H, Zhang Y, Xu M, Yang D. Current trends of targeted therapy for oral squamous cell carcinoma. J Cancer Res Clin Oncol 2022; 148:2169-2186. [PMID: 35501496 DOI: 10.1007/s00432-022-04028-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/15/2022] [Indexed: 10/18/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a malignant disease in the world which has a profound effect on human health and life quality. According to tumor stage and pathological diagnosis, OSCC is mainly treated by combinations of surgery, radiotherapy and chemotherapy. However, traditional treatment methods suffer from some limitations, such as systemic toxicity, limited therapeutic effect and drug resistance. With the rapid development of nanotechnology, nanodrug delivery systems (DDSs) and intelligent DDSs have been widely used in targeted therapy for OSCC. Meanwhile, the newly developed therapeutic techniques such as immunotherapy, gene therapy and bionic technology provide the possibility to realize the active targeted therapy. Here, the latest advances of target therapy for OSCC are reviewed, and their therapeutic remarks, current limits and future prospects are also systematically interpreted. It is believed that active and passive targeted therapies have great potentials for clinical transformation and application of OSCC, which will greatly improve human quality of life.
Collapse
Affiliation(s)
- Hongjiao Li
- School and Hospital of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
| | - Yao Zhang
- School and Hospital of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
| | - Mengmeng Xu
- School and Hospital of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
| | - Deqin Yang
- School and Hospital of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.
| |
Collapse
|
80
|
Wang C, Sani ES, Gao W. Wearable Bioelectronics for Chronic Wound Management. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2111022. [PMID: 36186921 PMCID: PMC9518812 DOI: 10.1002/adfm.202111022] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Indexed: 05/05/2023]
Abstract
Chronic wounds are a major healthcare issue and can adversely affect the lives of millions of patients around the world. The current wound management strategies have limited clinical efficacy due to labor-intensive lab analysis requirements, need for clinicians' experiences, long-term and frequent interventions, limiting therapeutic efficiency and applicability. The growing field of flexible bioelectronics enables a great potential for personalized wound care owing to its advantages such as wearability, low-cost, and rapid and simple application. Herein, recent advances in the development of wearable bioelectronics for monitoring and management of chronic wounds are comprehensively reviewed. First, the design principles and the key features of bioelectronics that can adapt to the unique wound milieu features are introduced. Next, the current state of wound biosensors and on-demand therapeutic systems are summarized and highlighted. Furthermore, we discuss the design criteria of the integrated closed loop devices. Finally, the future perspectives and challenges in wearable bioelectronics for wound care are discussed.
Collapse
Affiliation(s)
- Canran Wang
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ehsan Shirzaei Sani
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| | - Wei Gao
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
81
|
Mai R, Deng B, Zhao H, Li L, Fang Y, Li S, Deng X, Chen J. Design, Synthesis, and Bioevaluation of Novel Enzyme-Triggerable Cell Penetrating Peptide-Based Dendrimers for Targeted Delivery of Camptothecin and Cancer Therapy. J Med Chem 2022; 65:5850-5865. [PMID: 35380045 DOI: 10.1021/acs.jmedchem.2c00287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Novel enzyme-triggerable cell penetrating peptide (ETCPP) dendrimers with a camptothecin (CPT) warhead were designed and synthesized based on an amphiphilic penetrating peptide (FKKFFRKLL, discovered by us before). Among the newly synthesized ETCPP dendrimer conjugates, BL_Oc-SS-CPT (a high-generation dendrimer) exhibited the highest activity with IC50s in the nanomolar range (31-747 nM) against a panel of cancer cells, which is 3-10 times better than that of CPT. BL_Oc-SS-CPT remained intact during transit to target cells and in normal tissues with a plasma half-life of 4.2 h, 2.3-fold longer than that of the monomer (1.8 h). Once reaching the tumor site, BL_Oc-SS-CPT gradually released CPT in the presence of excessive matrix metalloproteinase-2/9 and GSH in cancer cells. Importantly, BL_Oc-SS-CPT exhibited excellent in vivo tumor targeting capability and antitumor efficacy with benign toxicity profiles. Thus, the novel ETCPP dendrimer-based drug delivery system (e.g., BL_Oc-SS-CPT) represents a safe and effective strategy for targeted cancer therapy.
Collapse
Affiliation(s)
- Ruiyao Mai
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Bulian Deng
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Huiting Zhao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Ling Li
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yuyu Fang
- Department of Nephrology, First People's Hospital of Pingjiang County, Yueyang 414500, China
| | - Siming Li
- Analytical Applications Center, Shimadzu (China) Co., Ltd. Guangzhou Branch, 230 Gaotang Road, Guangzhou 510656, China
| | - Xin Deng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| |
Collapse
|
82
|
Kyu Shim M, Yang S, Sun IC, Kim K. Tumor-activated carrier-free prodrug nanoparticles for targeted cancer Immunotherapy: Preclinical evidence for safe and effective drug delivery. Adv Drug Deliv Rev 2022; 183:114177. [PMID: 35245568 DOI: 10.1016/j.addr.2022.114177] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/27/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
Abstract
As immunogenic cell death (ICD) inducers initiating antitumor immune responses, certain chemotherapeutic drugs have shown considerable potential to reverse the immunosuppressive tumor microenvironment (ITM) into immune-responsive tumors. The application of these drugs in nanomedicine provides a more enhanced therapeutic index by improving unfavorable pharmacokinetic (PK) profiles and inefficient tumor targeting. However, the clinical translation of conventional nanoparticles is restricted by fundamental problems, such as risks of immunogenicity and potential toxicity by carrier materials, premature drug leakage in off-target sites during circulation, low drug loading contents, and complex structure and synthetic processes that hinder quality control (QC) and scale-up industrial production. To address these limitations, tumor-activated carrier-free prodrug nanoparticles (PDNPs), constructed only by the self-assembly of prodrugs without any additional carrier materials, have been widely investigated with distinct advantages for safe and more effective drug delivery. In addition, combination immunotherapy based on PDNPs with other diverse modalities has efficiently reversed the ITM to immune-responsive tumors, potentiating the response to immune checkpoint blockade (ICB) therapy. In this review, the trends and advances in PDNPs are outlined, and each self-assembly mechanism is discussed. In addition, various combination immunotherapies based on PDNPs are reviewed. Finally, a physical tumor microenvironment remodeling strategy to maximize the potential of PDNPs, and key considerations for clinical translation are highlighted.
Collapse
|
83
|
Nance E, Pun SH, Saigal R, Sellers DL. Drug delivery to the central nervous system. NATURE REVIEWS. MATERIALS 2022; 7:314-331. [PMID: 38464996 PMCID: PMC10923597 DOI: 10.1038/s41578-021-00394-w] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 03/12/2024]
Abstract
Despite the rising global incidence of central nervous system (CNS) disorders, CNS drug development remains challenging, with high costs, long pathways to clinical use and high failure rates. The CNS is highly protected by physiological barriers, in particular, the blood-brain barrier and the blood-cerebrospinal fluid barrier, which limit access of most drugs. Biomaterials can be designed to bypass or traverse these barriers, enabling the controlled delivery of drugs into the CNS. In this Review, we first examine the effects of normal and diseased CNS physiology on drug delivery to the brain and spinal cord. We then discuss CNS drug delivery designs and materials that are administered systemically, directly to the CNS, intranasally or peripherally through intramuscular injections. Finally, we highlight important challenges and opportunities for materials design for drug delivery to the CNS and the anticipated clinical impact of CNS drug delivery.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Rajiv Saigal
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| |
Collapse
|
84
|
β-Sheet to Random Coil Transition in Self-Assembling Peptide Scaffolds Promotes Proteolytic Degradation. Biomolecules 2022; 12:biom12030411. [PMID: 35327603 PMCID: PMC8945919 DOI: 10.3390/biom12030411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/25/2022] [Accepted: 03/05/2022] [Indexed: 12/20/2022] Open
Abstract
One of the most desirable properties that biomaterials designed for tissue engineering or drug delivery applications should fulfill is biodegradation and resorption without toxicity. Therefore, there is an increasing interest in the development of biomaterials able to be enzymatically degraded once implanted at the injury site or once delivered to the target organ. In this paper, we demonstrate the protease sensitivity of self-assembling amphiphilic peptides, in particular, RAD16-I (AcN-RADARADARADARADA-CONH2), which contains four potential cleavage sites for trypsin. We detected that when subjected to thermal denaturation, the peptide secondary structure suffers a transition from β-sheet to random coil. We also used Matrix-Assisted Laser Desorption/Ionization-Time-Of-Flight (MALDI-TOF) to detect the proteolytic breakdown products of samples subjected to incubation with trypsin as well as atomic force microscopy (AFM) to visualize the effect of the degradation on the nanofiber scaffold. Interestingly, thermally treated samples had a higher extent of degradation than non-denatured samples, suggesting that the transition from β-sheet to random coil leaves the cleavage sites accessible and susceptible to protease degradation. These results indicate that the self-assembling peptide can be reduced to short peptide sequences and, subsequently, degraded to single amino acids, constituting a group of naturally biodegradable materials optimal for their application in tissue engineering and regenerative medicine.
Collapse
|
85
|
Li Y, Chen Z, Gu L, Duan Z, Pan D, Xu Z, Gong Q, Li Y, Zhu H, Luo K. Anticancer nanomedicines harnessing tumor microenvironmental components. Expert Opin Drug Deliv 2022; 19:337-354. [PMID: 35244503 DOI: 10.1080/17425247.2022.2050211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Small-molecular drugs are extensively used in cancer therapy, while they have issues of nonspecific distribution and consequent side effects. Nanomedicines that incorporate chemotherapeutic drugs have been developed to enhance the therapeutic efficacy of these drugs and reduce their side effects. One of the promising strategies is to prepare nanomedicines by harnessing the unique tumor microenvironment (TME). AREAS COVERED The TME contains numerous cell types that specifically express specific antibodies on the surface including tumor vascular endothelial cells, tumor-associated adipocytes, tumor-associated fibroblasts, tumor-associated immune cells and cancer stem cells. The physicochemical environment is characterized with a low pH, hypoxia, and a high redox potential resulting from tumor-specific metabolism. The intelligent nanomedicines can be categorized into two groups: the first group which is rapidly responsive to extracellular chemical/biological factors in the TME and the second one which actively and/or specifically targets cellular components in the TME. EXPERT OPINION In this paper, we review recent progress of nanomedicines by harnessing the TME and illustrate the principles and advantages of different strategies for designing nanomedicines, which are of great significance for exploring novel nanomedicines or translating current nanomedicines into clinical practice. We will discuss the challenges and prospects of preparing nanomedicines to utilize or alter the TME for achieving effective, safe anticancer treatment.
Collapse
Affiliation(s)
- Yinggang Li
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhonglan Chen
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Gu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyu Duan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dayi Pan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuping Xu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Youping Li
- Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
86
|
|
87
|
Chen R, Zhai YY, Sun L, Wang Z, Xia X, Yao Q, Kou L. Alantolactone-loaded chitosan/hyaluronic acid nanoparticles suppress psoriasis by deactivating STAT3 pathway and restricting immune cell recruitment. Asian J Pharm Sci 2022; 17:268-283. [PMID: 35582636 PMCID: PMC9091614 DOI: 10.1016/j.ajps.2022.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/19/2022] [Indexed: 02/07/2023] Open
|
88
|
Kahle ER, Han B, Chandrasekaran P, Phillips ER, Mulcahey MK, Lu XL, Marcolongo MS, Han L. Molecular Engineering of Pericellular Microniche via Biomimetic Proteoglycans Modulates Cell Mechanobiology. ACS NANO 2022; 16:1220-1230. [PMID: 35015500 PMCID: PMC9271520 DOI: 10.1021/acsnano.1c09015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Molecular engineering of biological tissues using synthetic mimics of native matrix molecules can modulate the mechanical properties of the cellular microenvironment through physical interactions with existing matrix molecules, and in turn, mediate the corresponding cell mechanobiology. In articular cartilage, the pericellular matrix (PCM) is the immediate microniche that regulates cell fate, signaling, and metabolism. The negatively charged osmo-environment, as endowed by PCM proteoglycans, is a key biophysical cue for cell mechanosensing. This study demonstrated that biomimetic proteoglycans (BPGs), which mimic the ultrastructure and polyanionic nature of native proteoglycans, can be used to molecularly engineer PCM micromechanics and cell mechanotransduction in cartilage. Upon infiltration into bovine cartilage explant, we showed that localization of BPGs in the PCM leads to increased PCM micromodulus and enhanced chondrocyte intracellular calcium signaling. Applying molecular force spectroscopy, we revealed that BPGs integrate with native PCM through augmenting the molecular adhesion of aggrecan, the major PCM proteoglycan, at the nanoscale. These interactions are enabled by the biomimetic "bottle-brush" ultrastructure of BPGs and facilitate the integration of BPGs within the PCM. Thus, this class of biomimetic molecules can be used for modulating molecular interactions of pericellular proteoglycans and harnessing cell mechanosensing. Because the PCM is a prevalent feature of various cell types, BPGs hold promising potential for improving regeneration and disease modification for not only cartilage-related healthcare but many other tissues and diseases.
Collapse
Affiliation(s)
- Elizabeth R. Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Evan R. Phillips
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
| | - Mary K. Mulcahey
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Michele S. Marcolongo
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
- Department of Mechanical Engineering, Villanova University, Villanova, PA 19085, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
89
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
90
|
Cao L, Zhu Y, Wang W, Wang G, Zhang S, Cheng H. Emerging Nano-Based Strategies Against Drug Resistance in Tumor Chemotherapy. Front Bioeng Biotechnol 2021; 9:798882. [PMID: 34950650 PMCID: PMC8688801 DOI: 10.3389/fbioe.2021.798882] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/19/2021] [Indexed: 02/05/2023] Open
Abstract
Drug resistance is the most significant causes of cancer chemotherapy failure. Various mechanisms of drug resistance include tumor heterogeneity, tumor microenvironment, changes at cellular levels, genetic factors, and other mechanisms. In recent years, more attention has been paid to tumor resistance mechanisms and countermeasures. Nanomedicine is an emerging treatment platform, focusing on alternative drug delivery and improved therapeutic effectiveness while reducing side effects on normal tissues. Here, we reviewed the principal forms of drug resistance and the new possibilities that nanomaterials offer for overcoming these therapeutic barriers. Novel nanomaterials based on tumor types are an excellent modality to equalize drug resistance that enables gain more rational and flexible drug selectivity for individual patient treatment. With the emergence of advanced designs and alternative drug delivery strategies with different nanomaterials, overcome of multidrug resistance shows promising and opens new horizons for cancer therapy. This review discussed different mechanisms of drug resistance and recent advances in nanotechnology-based therapeutic strategies to improve the sensitivity and effectiveness of chemotherapeutic drugs, aiming to show the advantages of nanomaterials in overcoming of drug resistance for tumor chemotherapy, which could accelerate the development of personalized medicine.
Collapse
Affiliation(s)
- Lei Cao
- Department of Pathology, Quanzhou Women’s and Children’s Hospital, Quanzhou, China
| | - Yuqin Zhu
- Department of Pathology, Quanzhou Women’s and Children’s Hospital, Quanzhou, China
| | - Weiju Wang
- Department of Pathology, Qingyuan Maternal and Child Health Hospital, Qingyuan, China
| | - Gaoxiong Wang
- Department of Pathology, Quanzhou Women’s and Children’s Hospital, Quanzhou, China
| | - Shuaishuai Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
91
|
Caballero D, Abreu CM, Lima AC, Neves NN, Reis RL, Kundu SC. Precision biomaterials in cancer theranostics and modelling. Biomaterials 2021; 280:121299. [PMID: 34871880 DOI: 10.1016/j.biomaterials.2021.121299] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
Despite significant achievements in the understanding and treatment of cancer, it remains a major burden. Traditional therapeutic approaches based on the 'one-size-fits-all' paradigm are becoming obsolete, as demonstrated by the increasing number of patients failing to respond to treatments. In contrast, more precise approaches based on individualized genetic profiling of tumors have already demonstrated their potential. However, even more personalized treatments display shortcomings mainly associated with systemic delivery, such as low local drug efficacy or specificity. A large amount of effort is currently being invested in developing precision medicine-based strategies for improving the efficiency of cancer theranostics and modelling, which are envisioned to be more accurate, standardized, localized, and less expensive. To this end, interdisciplinary research fields, such as biomedicine, material sciences, pharmacology, chemistry, tissue engineering, and nanotechnology, must converge for boosting the precision cancer ecosystem. In this regard, precision biomaterials have emerged as a promising strategy to detect, model, and treat cancer more efficiently. These are defined as those biomaterials precisely engineered with specific theranostic functions and bioactive components, with the possibility to be tailored to the cancer patient needs, thus having a vast potential in the increasing demand for more efficient treatments. In this review, we discuss the latest advances in the field of precision biomaterials in cancer research, which are expected to revolutionize disease management, focusing on their uses for cancer modelling, detection, and therapeutic applications. We finally comment on the needed requirements to accelerate their application in the clinic to improve cancer patient prognosis.
Collapse
Affiliation(s)
- David Caballero
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Catarina M Abreu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ana C Lima
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Nuno N Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
92
|
Liang J, Yang B, Zhou X, Han Q, Zou J, Cheng L. Stimuli-responsive drug delivery systems for head and neck cancer therapy. Drug Deliv 2021; 28:272-284. [PMID: 33501883 PMCID: PMC7850355 DOI: 10.1080/10717544.2021.1876182] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Head and neck cancer (HNC) is among the most common malignancy that has a profound impact on human health and life quality. The treatment for HNC, especially for the advanced cancer is stage-dependent and in need of combined therapies. Various forms of adjuvant treatments such as chemotherapy, phototherapy, hyperthermia, gene therapy have been included in the HNC therapy. However, there are still restrictions with traditional administration such as limited in situ therapeutic effect, systemic toxicity, drug resistance, etc. In recent years, stimuli-responsive drug delivery systems (DDSs) have attracted the great attention in HNC therapy. These intelligent DDSs could respond to unique tumor microenvironment, external triggers or dual/multi stimulus with more specific drug delivery and release, leading to enhanced treatment efficiency and less reduced side effects. In this article, recent studies on stimuli-responsive DDSs for HNC therapy were summarized, which could respond to endogenous and exogenous triggers including pH, matrix metalloproteinases (MMPs), reactive oxygen species (ROS), redox condition, light, magnetic field and multi stimuli. Their therapeutic remarks, current limits and future prospect for these intelligent DDSs were discussed. Furthermore, multifunctional stimuli-responsive DDSs have also been reviewed. With the modification of drug carriers or co-loading with therapeutic agents. Those intelligent DDSs showed more biofunctions such as combined therapeutic effects or integration of diagnosis and treatment for HNC. It is believed that stimuli-responsive drug delivery systems showed great potential for future clinic translation and application for the treatment of HNC.
Collapse
Affiliation(s)
- Jingou Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Bina Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Qi Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jing Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
93
|
Martu MA, Maftei GA, Luchian I, Stefanescu OM, Scutariu MM, Solomon SM. The Effect of Acknowledged and Novel Anti-Rheumatic Therapies on Periodontal Tissues-A Narrative Review. Pharmaceuticals (Basel) 2021; 14:1209. [PMID: 34959607 PMCID: PMC8705490 DOI: 10.3390/ph14121209] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) and periodontal disease (PD) are chronic complex inflammatory diseases with several common susceptibility factors, especially genetic and environmental risk factors. Although both disorders involve a perturbation of the immune-inflammatory response at multiple levels, one major difference between the two is the different locations in which they develop. RA is triggered by an exaggerated autoimmune response that targets joints, while periodontal disease occurs as a consequence of the subgingival periodontopathogenic microbiota. Current treatment models in both pathologies involve the stratification of patients to allow therapeutic individualization according to disease stage, complexity, progression, lifestyle, risk factors, and additional systemic diseases. Therapeutic guidelines for RA comprise of five main classes of drugs: non-steroidal anti-inflammatory drugs (NSAIDs), analgesics, glucocorticoids, and disease-modifying anti-rheumatic drugs (DMARDs): biologic and non-biologic. Although various treatment options are available, a definitive treatment remains elusive, therefore research is ongoing in this area. Several alternatives are currently being tested, such as matrix metalloproteinases (MMP) inhibitors, toll-like receptors (TLR) blockers, pro-resolution mediators, anti-hypoxia inducing factors, stem cell therapy, NLRP3 inhibitors and even natural derived compounds. Although the link between PD and rheumatoid arthritis has been investigated by multiple microbiology and immunology studies, the precise influence and causality is still debated in the literature. Furthermore, the immunomodulatory effect of anti-rheumatic drugs on the periodontium is still largely unknown. In this narrative review, we explore the mechanisms of interaction and the potential influence that anti-rheumatoid medication, including novel treatment options, has on periodontal tissues and whether periodontal health status and treatment can improve the prognosis of an RA patient.
Collapse
Affiliation(s)
- Maria-Alexandra Martu
- Periodontology Department, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.-A.M.); (S.M.S.)
| | - George-Alexandru Maftei
- Oral Pathology Department, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania
| | - Ionut Luchian
- Periodontology Department, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.-A.M.); (S.M.S.)
| | - Ovidiu Mihail Stefanescu
- Dento-Alveolar Surgery and Anesthesiology Department, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania;
| | - Mihaela Monica Scutariu
- Oro-Dental Diagnostics Department, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania;
| | - Sorina Mihaela Solomon
- Periodontology Department, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.-A.M.); (S.M.S.)
| |
Collapse
|
94
|
Battistella C, Liang Y, Gianneschi NC. Innovations in Disease State Responsive Soft Materials for Targeting Extracellular Stimuli Associated with Cancer, Cardiovascular Disease, Diabetes, and Beyond. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007504. [PMID: 34145625 PMCID: PMC9836048 DOI: 10.1002/adma.202007504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/20/2021] [Indexed: 05/10/2023]
Abstract
Recent advances in polymer chemistry, materials sciences, and biotechnology have allowed the preclinical development of sophisticated programmable nanomedicines and materials that are able to precisely respond to specific disease-associated triggers and microenvironments. These stimuli, endogenous to the targeted diseases, include pH, redox-state, small molecules, and protein upregulation. Herein, recent advances and innovative approaches in programmable soft materials capable of sensing the aforementioned disease-associated stimuli and responding via a range of dynamic processes including morphological and size transitions, changes in mobility and retention, as well as disassembly are described. In this field generally, the majority of ongoing and past research effort has focused on oncology. Given this interest, examples of the latest innovative approaches to chemo- and immunotherapy treatment strategies for cancer are presented. Moreover, as the field broadens its attention, applications of programmable materials in other diseases are highlighted, with a special focus on cardiovascular disease and diabetes mellitus, where limited attention is paid by the field, but where many promising avenues exist with high potential impact.
Collapse
Affiliation(s)
- Claudia Battistella
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Department of Pharmacology, Northwestern University, Evanston, IL, 60208, USA
| | - Yifei Liang
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Nathan C Gianneschi
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Department of Pharmacology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
95
|
Wan D, Li S, Zhang J, Ma G, Pan J. Intelligent self-assembly prodrug micelles loading doxorubicin in response to tumor microenvironment for targeted tumors therapy. Chin J Chem Eng 2021. [DOI: 10.1016/j.cjche.2021.06.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
96
|
A Multifunctional Polymeric Micelle for Targeted Delivery of Paclitaxel by the Inhibition of the P-Glycoprotein Transporters. NANOMATERIALS 2021; 11:nano11112858. [PMID: 34835622 PMCID: PMC8618187 DOI: 10.3390/nano11112858] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/17/2022]
Abstract
P-glycoprotein (P-gP) efflux-mediated multidrug resistance is a fundamental aspect of chemotherapeutic failure in oncology. The current study aims to deliver paclitaxel (PTX) specifically at the target site with improved in vivo efficacy of poorly permeable PTX against solid tumors. Multifunctional polymeric micelles as targeted delivery have been devised for loading and release of PTX. Mucoadhesion, permeation enhancement, oral pharmacokinetics, biodistribution, and toxicological studies were carried out to fully elucidate the therapeutic outcomes of the polymeric micelles. Ex vivo permeation studies indicated a 7.89-fold enhancement in the permeation of PTX with mucopermeating papain functionalized thiolated redox micelles (PT-R-Ms) compared to the pure PTX. Moreover, PT-R-Ms exhibited a higher percentage of apoptotic cells (42.9 ± 0.07%) compared to pure PTX. Biodistribution studies revealed that fluorotagged PT-RMs accumulated in excised tumors and organs. The higher fluorescence intensity indicated the mucopermeation of micelles across the intestine. The orally administered PT-R-Ms efficiently overcome intestinal barriers and inhibit the P-gP efflux pump, resulting in increased bioavailability of PTX (up to 8-fold) in comparison to pure PTX. The enhanced anti-tumor efficacy and reduced toxic effects are key aspects of efficient cancer therapy. This study demonstrates that the use of mucopermeating PT-R-Ms is an encouraging approach to overwhelm the permeation barrier in cancer treatment.
Collapse
|
97
|
Spiller S, Wippold T, Bellmann-Sickert K, Franz S, Saalbach A, Anderegg U, Beck-Sickinger AG. Protease-Triggered Release of Stabilized CXCL12 from Coated Scaffolds in an Ex Vivo Wound Model. Pharmaceutics 2021; 13:pharmaceutics13101597. [PMID: 34683890 PMCID: PMC8539926 DOI: 10.3390/pharmaceutics13101597] [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: 08/27/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Biomaterials are designed to improve impaired healing of injured tissue. To accomplish better cell integration, we suggest to coat biomaterial surfaces with bio-functional proteins. Here, a mussel-derived surface-binding peptide is used and coupled to CXCL12 (stromal cell-derived factor 1α), a chemokine that activates CXCR4 and consequently recruits tissue-specific stem and progenitor cells. CXCL12 variants with either non-releasable or protease-mediated-release properties were designed and compared. Whereas CXCL12 was stabilized at the N-terminus for protease resistance, a C-terminal linker was designed that allowed for specific cleavage-mediated release by matrix metalloproteinase 9 and 2, since both enzymes are frequently found in wound fluid. These surface adhesive CXCL12 derivatives were produced by expressed protein ligation. Functionality of the modified chemokines was assessed by inositol phosphate accumulation and cell migration assays. Increased migration of keratinocytes and primary mesenchymal stem cells was demonstrated. Immobilization and release were studied for bioresorbable PCL-co-LC scaffolds, and accelerated wound closure was demonstrated in an ex vivo wound healing assay on porcine skin grafts. After 24 h, a significantly improved CXCL12-specific growth stimulation of the epithelial tips was already observed. The presented data display a successful application of protein-coated biomaterials for skin regeneration.
Collapse
Affiliation(s)
- Sabrina Spiller
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
| | - Tom Wippold
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
| | - Sandra Franz
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Anja Saalbach
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
- Correspondence: (U.A.); (A.G.B.-S.); Tel.: +49-341-972-5881 (U.A.); +49-341-973-6900 (A.G.B.-S.); Fax: +49-341-972-5878 (U.A.); +49-341-973-6909 (A.G.B.-S.)
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
- Correspondence: (U.A.); (A.G.B.-S.); Tel.: +49-341-972-5881 (U.A.); +49-341-973-6900 (A.G.B.-S.); Fax: +49-341-972-5878 (U.A.); +49-341-973-6909 (A.G.B.-S.)
| |
Collapse
|
98
|
Lv S, Sylvestre M, Prossnitz AN, Yang LF, Pun SH. Design of Polymeric Carriers for Intracellular Peptide Delivery in Oncology Applications. Chem Rev 2021; 121:11653-11698. [PMID: 33566580 DOI: 10.1021/acs.chemrev.0c00963] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent decades, peptides, which can possess high potency, excellent selectivity, and low toxicity, have emerged as promising therapeutics for cancer applications. Combined with an improved understanding of tumor biology and immuno-oncology, peptides have demonstrated robust antitumor efficacy in preclinical tumor models. However, the translation of peptides with intracellular targets into clinical therapies has been severely hindered by limitations in their intrinsic structure, such as low systemic stability, rapid clearance, and poor membrane permeability, that impede intracellular delivery. In this Review, we summarize recent advances in polymer-mediated intracellular delivery of peptides for cancer therapy, including both therapeutic peptides and peptide antigens. We highlight strategies to engineer polymeric materials to increase peptide delivery efficiency, especially cytosolic delivery, which plays a crucial role in potentiating peptide-based therapies. Finally, we discuss future opportunities for peptides in cancer treatment, with an emphasis on the design of polymer nanocarriers for optimized peptide delivery.
Collapse
Affiliation(s)
| | | | - Alexander N Prossnitz
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | | | | |
Collapse
|
99
|
Li L, Wang J, Feng L, Fan J, Wang J, Tan N, Wang Z. Rubioncolin C, a natural naphthohydroquinone dimer isolated from Rubia yunnanensis, inhibits the proliferation and metastasis by inducing ROS-mediated apoptotic and autophagic cell death in triple-negative breast cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114184. [PMID: 33961996 DOI: 10.1016/j.jep.2021.114184] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rubia yunnanensis Diels is a traditional Chinese medicine that has diverse pharmacological activities, including antituberculosis, antirheumatism and anticancers. Rubioncolin C (RC), a natural naphthohydroquinone dimer isolated from the roots and rhizomes of R. yunnanensis Diels, has shown potent antitumor activity. However, the antitumor activity and its potential mechanism of RC in triple-negative breast cancer (TNBC) cell lines remained unclear. AIM OF THE STUDY This study was aim to investigate the anti-proliferation and anti-metastasis activity as well as the potential mechanism of RC on triple-negative breast cancer cells in vitro and in vivo. MATERIALS AND METHODS The sulforhodamine B assay, colony formation assay and cell cycle analysis were used to determine the anti-proliferative activity of RC on TNBC. The anti-metastatic activity in vitro of RC was detected through the scratch wound assay, cell migration and invasion assays and gelatin zymography. The flow cytometry, JC-1, GFP-LC3B plasmid transfection, MDC, Lysotracker red and Carboxy-H2DCFDA, DHE, and MitoSOX™ Red staining were performed to investigate the effect of RC on apoptosis, autophagy and ROS level. The apoptosis inhibitor, autophagy inhibitors and ROS inhibitors were used to further verify the antitumor mechanism of RC. The protein levels related with cell cycle, apoptosis, and autophagy were examined with western blotting. In addition, the anti-tumor activity of RC in vivo was assessed in an experimental metastatic model. RESULTS In the present study, RC suppressed the proliferation of TNBC cells in a time- and dose-dependent manner via regulating cell cycle. Further experiments showed that RC inhibited the migration and invasion of TNBC cells by downregulating MMPs and inhibiting EMT. Moreover, we demonstrated that RC induced obviously apoptotic and autophagic cell death, activated MAPK signaling pathway and inhibited mTOR/Akt/p70S6K and NF-κB signaling pathways. Furthermore, the excessive ROS was produced after treatment with RC. The antioxygen NAC and GSH could rescue the cell viability and reestablish the ability of cell metastasis, and inhibit the RC-induced apoptosis and autophagy. In a mice lung metastasis model of breast cancer, RC inhibited lung metastasis, and induced autophagy and apoptosis. CONCLUSION These findings clarified the antitumor mechanism of RC on TNBC cell lines and suggested that RC is a key active ingredient for the cancer treatment of R. yunnanensis, which would help RC develop as a new potential chemotherapeutic agent for TNBC treatment.
Collapse
Affiliation(s)
- Ling Li
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Jia Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China.
| | - Li Feng
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Junting Fan
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China.
| | - Jing Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Ninghua Tan
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Zhe Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
100
|
Zhao Y, Xie YQ, Van Herck S, Nassiri S, Gao M, Guo Y, Tang L. Switchable immune modulator for tumor-specific activation of anticancer immunity. SCIENCE ADVANCES 2021; 7:eabg7291. [PMID: 34516776 PMCID: PMC8442900 DOI: 10.1126/sciadv.abg7291] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Immune stimulatory antibodies and cytokines elicit potent antitumor immunity. However, the dose-limiting systemic toxicity greatly hinders their clinical applications. Here, we demonstrate a chemical approach, termed “switchable” immune modulator (Sw-IM), to limit the systemic exposure and therefore ameliorate their toxicities. Sw-IM is a biomacromolecular therapeutic reversibly masked by biocompatible polymers through chemical linkers that are responsive to tumor-specific stimuli, such as high reducing potential and acidic pH. Sw-IMs stay inert (switch off) in the circulation and healthy tissues but get reactivated (switch on) selectively in tumor via responsive removal of the polymer masks, thus focusing the immune boosting activities in the tumor microenvironment. Sw-IMs applied to anti–4-1BB agonistic antibody and IL-15 cytokine led to equivalent antitumor efficacy to the parental IMs with markedly reduced toxicities. Sw-IM provides a highly modular and generic approach to improve the therapeutic window and clinical applicability of potent IMs in mono- and combinational immunotherapies.
Collapse
Affiliation(s)
- Yu Zhao
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland
| | - Yu-Qing Xie
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Simon Van Herck
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Department of Pharmaceutics, Ghent University, 9000 Ghent, Belgium
| | - Sina Nassiri
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Min Gao
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Yugang Guo
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland
- Corresponding author.
| |
Collapse
|