1
|
Cai Z, Liu B, Cai Q, Gou J, Tang X. Advances in microsphere-based therapies for peritoneal carcinomatosis: challenges, innovations, and future prospects. Expert Opin Drug Deliv 2025; 22:31-46. [PMID: 39641971 DOI: 10.1080/17425247.2024.2439462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/09/2024] [Accepted: 12/04/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Clinical outcomes for the treatment of peritoneal carcinomatosis (PC) have remained suboptimal. Microsphere-based intraperitoneal chemotherapy has shown considerable potential in preclinical studies. However, due to the complications associated with peritoneal adhesions, there has been a lack of comprehensive reviews focusing on the progress of microsphere applications in the treatment of PC. AREAS COVERED We provide an overview of the current clinical treatment strategies for PC and analyze the potential advantages of microspheres in this context. Regarding the issue of peritoneal adhesions induced by microspheres, we investigate the underlying mechanisms and propose possible solutions. Furthermore, we outline the future directions for the development of microsphere-based therapies in the treatment of PC. EXPERT OPINION Microspheres formulated with highly biocompatible materials to the peritoneum, such as sodium alginate, gelatin, or genipin, or with an optimal particle size (4 ~ 30 μm) and lower molecular weights (10 ~ 57 kDa), can prevent peritoneal adhesions and improve drug distribution. To further enhance the antitumor efficacy, enhancing the tumor penetration capability and specificity of microspheres, optimizing intraperitoneal distribution, and addressing tumor resistance have demonstrated significant potential in preclinical studies, offering new therapeutic prospects for the treatment of PC.
Collapse
Affiliation(s)
- Zhitao Cai
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Boyuan Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qing Cai
- Department of Formulation, Zhuhai Livzon Microsphere Technology Co. Ltd, Zhuhai, China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
2
|
Singh N, Anand SK, Sharma A, Singh S, Kakkar P, Srivastava V. Chitosan/alginate nanogel potentiate berberine uptake and enhance oxidative stress mediated apoptotic cell death in HepG2 cells. Int J Biol Macromol 2024; 257:128717. [PMID: 38081485 DOI: 10.1016/j.ijbiomac.2023.128717] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/18/2023]
Abstract
Biopolymer-based nanoscale drug delivery systems have become a promising approach to overcome the limitations associated with conventional chemotherapeutics used for cancer treatment. Herein, we reported to develop a hydrophilic nanogel (NG) composed of Chitosan (Chi) and sodium alginate (Alg) using the ion gelation method for delivering Berberine hydrochloride (BBR), an alkaloid obtained from Berberis aristata roots. The use of different nanocarriers for BBR delivery has been reported previously, but the bioavailability of these carriers was limited due to phagocytic uptake and poor systemic delivery. The developed NG showed enhanced stability and efficient entrapment of BBR ∼92 %, resulting in a significant increase in bioavailability. The pH-dependent release behavior demonstrated sustained and effective release of ∼86 %, ∼74 % and, ∼53 % BBR at pH 5.5, 6.6, and 7.4 respectively after 72h, indicating its potential as a drug carrier. Additionally, the cellular uptake of BBR was significantly higher ∼19 % in the BBR-NG (25 μM) than in bulk BBR (100 μM), leading to enhanced ROS generation, mitochondrial depolarisation, and inhibition of cell proliferation and colony formation in HepG2 cells. In summary, the results suggest that the Chi/Alg biopolymer-based nano-formulation could be an effective approach for delivering BBR and enhancing its cellular uptake, efficacy, and cytotoxicity.
Collapse
Affiliation(s)
- Neha Singh
- CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sumit Kumar Anand
- CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India; Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA-71103, USA
| | - Ankita Sharma
- CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226 001, Uttar Pradesh, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India
| | - Sukhveer Singh
- CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Poonam Kakkar
- CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| | - Vikas Srivastava
- CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
3
|
Niu H, Zhao P, Sun W. Biomaterials for chimeric antigen receptor T cell engineering. Acta Biomater 2023; 166:1-13. [PMID: 37137403 DOI: 10.1016/j.actbio.2023.04.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/05/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cells have achieved breakthrough efficacies against hematological malignancies, but their unsatisfactory efficacies in solid tumors limit their applications. The prohibitively high prices further restrict their access to broader populations. Novel strategies are urgently needed to address these challenges, and engineering biomaterials can be one promising approach. The established process for manufacturing CAR-T cells involves multiple steps, and biomaterials can help simplify or improve several of them. In this review, we cover recent progress in engineering biomaterials for producing or stimulating CAR-T cells. We focus on the engineering of non-viral gene delivery nanoparticles for transducing CAR into T cells ex vivo/in vitro or in vivo. We also dive into the engineering of nano-/microparticles or implantable scaffolds for local delivery or stimulation of CAR-T cells. These biomaterial-based strategies can potentially change the way CAR-T cells are manufactured, significantly reducing their cost. Modulating the tumor microenvironment with the biomaterials can also considerably enhance the efficacy of CAR-T cells in solid tumors. We pay special attention to progress made in the past five years, and perspectives on future challenges and opportunities are also discussed. STATEMENT OF SIGNIFICANCE: Chimeric antigen receptor T (CAR-T) cell therapies have revolutionized the field of cancer immunotherapy with genetically engineered tumor recognition. They are also promising for treating many other diseases. However, the widespread application of CAR-T cell therapy has been hampered by the high manufacturing cost. Poor penetration of CAR-T cells into solid tissues further restricted their use. While biological strategies have been explored to improve CAR-T cell therapies, such as identifying new cancer targets or integrating smart CARs, biomaterial engineering provides alternative strategies toward better CAR-T cells. In this review, we summarize recent advances in engineering biomaterials for CAR-T cell improvement. Biomaterials ranging from nano-, micro-, and macro-scales have been developed to assist CAR-T cell manufacturing and formulation.
Collapse
Affiliation(s)
- Huanqing Niu
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA; State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu 211816, China
| | - Penghui Zhao
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Wujin Sun
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA; Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; Center for Emerging, Zoonotic, and Arthropod-Born Pathogens, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
4
|
Lu L, Wang L, Zhao L, Liao J, Zhao C, Xu X, Wang F, Zhang X. A Novel Blood-Brain Barrier-Penetrating and Vascular-Targeting Chimeric Peptide Inhibits Glioma Angiogenesis. Int J Mol Sci 2023; 24:ijms24108753. [PMID: 37240099 DOI: 10.3390/ijms24108753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The high vascularization of glioma highlights the potential value of anti-angiogenic therapeutics for glioma treatment. Previously, we designed a novel vascular-targeting and blood-brain barrier (BBB)-penetrating peptide, TAT-AT7, by attaching the cell-penetrating peptide TAT to a vascular-targeting peptide AT7, and we demonstrated that TAT-AT7 could target binding to the vascular endothelial growth factor receptor 2 (VEGFR-2) and Neuropilin-1 (NRP-1), which are both highly expressed in endothelial cells. TAT-AT7 has been proven to be a good targeting peptide which could effectively deliver the secretory endostatin gene to treat glioma via the TAT-AT7-modified polyethyleneimine (PEI) nanocomplex. In the current study, we further explored the molecular binding mechanisms of TAT-AT7 to VEGFR-2 and NRP-1 and its anti-glioma effects. Accordingly, TAT-AT7 was proven to competitively bind to VEGFR-2 and NRP-1 and prevent VEGF-A165 binding to the receptors by the surface plasmon resonance (SPR) assay. TAT-AT7 inhibited endothelial cells' proliferation, migration, invasion, and tubule formation, as well as promoted endothelial cells' apoptosis in vitro. Further research revealed that TAT-AT7 inhibited the phosphorylation of VEGFR-2 and its downstream PLC-γ, ERK1/2, SRC, AKT, and FAK kinases. Additionally, TAT-AT7 significantly inhibited angiogenesis of zebrafish embryo. Moreover, TAT-AT7 had a better penetrating ability and could penetrate the BBB into glioma tissue and target glioma neovascularization in an orthotopic U87-glioma-bearing nude mice model, and exhibited the effect of inhibiting glioma growth and angiogenesis. Taken together, the binding and function mechanisms of TAT-AT7 were firstly revealed, and TAT-AT7 was proven to be an effective and promising peptide for the development of anti-angiogenic drugs for targeted treatment of glioma.
Collapse
Affiliation(s)
- Lu Lu
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Longkun Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lin Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jing Liao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chunqian Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaohan Xu
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
5
|
Li T, Wu M, Wei Q, Xu D, He X, Wang J, Wu J, Chen L. Conjugated Polymer Nanoparticles for Tumor Theranostics. Biomacromolecules 2023; 24:1943-1979. [PMID: 37083404 DOI: 10.1021/acs.biomac.2c01446] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Water-dispersible conjugated polymer nanoparticles (CPNs) have demonstrated great capabilities in biological applications, such as in vitro cell/subcellular imaging and biosensing, or in vivo tissue imaging and disease treatment. In this review, we summarized the recent advances of CPNs used for tumor imaging and treatment during the past five years. CPNs with different structures, which have been applied to in vivo solid tumor imaging (fluorescence, photoacoustic, and dual-modal) and treatment (phototherapy, drug carriers, and synergistic therapy), are discussed in detail. We also demonstrated the potential of CPNs as cancer theranostic nanoplatforms. Finally, we discussed current challenges and outlooks in this field.
Collapse
Affiliation(s)
- Tianyu Li
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Mengqi Wu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Qidong Wei
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Dingshi Xu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Xuehan He
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Jiasi Wang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong 999077, SAR, China
| | - Lei Chen
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
6
|
Chi XK, Xu XL, Chen BY, Su J, Du YZ. Combining nanotechnology with monoclonal antibody drugs for rheumatoid arthritis treatments. J Nanobiotechnology 2023; 21:105. [PMID: 36964609 PMCID: PMC10039584 DOI: 10.1186/s12951-023-01857-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic immune disease characterized by synovial inflammation. Patients with RA commonly experience significant damage to their hand and foot joints, which can lead to joint deformities and even disability. Traditional treatments have several clinical drawbacks, including unclear pharmacological mechanisms and serious side effects. However, the emergence of antibody drugs offers a promising approach to overcome these limitations by specifically targeting interleukin-1 (IL-1), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and other cytokines that are closely related to the onset of RA. This approach reduces the incidence of adverse effects and contributes to significant therapeutic outcomes. Furthermore, combining these antibody drugs with drug delivery nanosystems (DDSs) can improve their tissue accumulation and bioavailability.Herein, we provide a summary of the pathogenesis of RA, the available antibody drugs and DDSs that improve the efficacy of these drugs. However, several challenges need to be addressed in their clinical applications, including patient compliance, stability, immunogenicity, immunosupression, target and synergistic effects. We propose strategies to overcome these limitations. In summary, we are optimistic about the prospects of treating RA with antibody drugs, given their specific targeting mechanisms and the potential benefits of combining them with DDSs.
Collapse
Affiliation(s)
- Xiao-Kai Chi
- College of Pharmacy, Jiamusi University, 258 Xuefu Road, Jiamusi, 154007, China
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China.
| | - Bang-Yao Chen
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China
| | - Jin Su
- College of Pharmacy, Jiamusi University, 258 Xuefu Road, Jiamusi, 154007, China.
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China.
| |
Collapse
|
7
|
Fluorinated Human Serum Albumin as Potential 19F Magnetic Resonance Imaging Probe. Molecules 2023; 28:molecules28041695. [PMID: 36838682 PMCID: PMC9959765 DOI: 10.3390/molecules28041695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Fluorinated human serum albumin conjugates were prepared and tested as potential metal-free probes for 19F magnetic resonance imaging (MRI). Each protein molecule was modified by several fluorine-containing compounds via the N-substituted natural acylating reagent homocysteine thiolactone. Albumin conjugates retain the protein's physical and biological properties, such as its 3D dimensional structure, aggregation ability, good solubility, proteolysis efficiency, biocompatibility, and low cytotoxicity. A dual-labeled with cyanine 7 fluorescence dye and fluorine reporter group albumin were synthesized for simultaneous fluorescence imaging and 19F MRI. The preliminary in vitro studies show the prospects of albumin carriers for multimodal imaging.
Collapse
|
8
|
Deng G, Zha H, Luo H, Zhou Y. Aptamer-conjugated gold nanoparticles and their diagnostic and therapeutic roles in cancer. Front Bioeng Biotechnol 2023; 11:1118546. [PMID: 36741760 PMCID: PMC9892635 DOI: 10.3389/fbioe.2023.1118546] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
The burden of incidence rate and mortality of cancer is increasing rapidly, and the development of precise intervention measures for cancer detection and treatment will help reduce the burden and pain of cancer. At present, the sensitivity and specificity of tumor markers such as CEA and CA-125 used clinically are low, while PET, SPECT, and other imaging diagnoses with high sensitivity possess shortcomings, including long durations to obtain formal reports and the inability to identify the molecular pathological type of cancer. Cancer surgery is limited by stage and easy to recur. Radiotherapy and chemotherapy often cause damage to normal tissues, leading to evident side effects. Aptamers can selectively and exclusively bind to biomarkers and have, therefore, gained attention as ligands to be targeted for cancer detection and treatment. Gold nanoparticles (AuNPs) are considered as promising nano carriers for cancer diagnosis and treatment due to their strong light scattering characteristics, effective biocompatibility, and easy surface modification with targeted agents. The aptamer-gold nanoparticles targeting delivery system developed herein can combine the advantages of aptamers and gold nanoparticles, and shows excellent targeting, high specificity, low immunogenicity, minor side effects, etc., which builds a bridge for cancer markers to be used in early and efficient diagnosis and precise treatment. In this review, we summarize the latest progress in the application of aptamer-modified gold nanoparticles in cancer targeted diagnosis and delivery of therapeutic agents to cancer cells and emphasize the prospects and challenges of transforming these studies into clinical applications.
Collapse
Affiliation(s)
- Guozhen Deng
- Department of Laboratory Medicine, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - He Zha
- Department of Laboratory Medicine, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Hongzhi Luo
- Department of Laboratory Medicine, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Yi Zhou
- Department of Orthopaedics, Jian Yang Hospital of Traditional Chinese Medicine, JianYang, Sichuan, China
| |
Collapse
|
9
|
Chen Z, Yue Z, Yang K, Li S. Nanomaterials: small particles show huge possibilities for cancer immunotherapy. J Nanobiotechnology 2022; 20:484. [DOI: 10.1186/s12951-022-01692-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022] Open
Abstract
AbstractWith the economy's globalization and the population's aging, cancer has become the leading cause of death in most countries. While imposing a considerable burden on society, the high morbidity and mortality rates have continuously prompted researchers to develop new oncology treatment options. Anti-tumor regimens have evolved from early single surgical treatment to combined (or not) chemoradiotherapy and then to the current stage of tumor immunotherapy. Tumor immunotherapy has undoubtedly pulled some patients back from the death. However, this strategy of activating or boosting the body's immune system hardly benefits most patients. It is limited by low bioavailability, low response rate and severe side effects. Thankfully, the rapid development of nanotechnology has broken through the bottleneck problem of anti-tumor immunotherapy. Multifunctional nanomaterials can not only kill tumors by combining anti-tumor drugs but also can be designed to enhance the body's immunity and thus achieve a multi-treatment effect. It is worth noting that the variety of nanomaterials, their modifiability, and the diversity of combinations allow them to shine in antitumor immunotherapy. In this paper, several nanobiotics commonly used in tumor immunotherapy at this stage are discussed, and they activate or enhance the body's immunity with their unique advantages. In conclusion, we reviewed recent advances in tumor immunotherapy based on nanomaterials, such as biological cell membrane modification, self-assembly, mesoporous, metal and hydrogels, to explore new directions and strategies for tumor immunotherapy.
Collapse
|
10
|
Szymborski J, Emad A. RAPPPID: towards generalizable protein interaction prediction with AWD-LSTM twin networks. Bioinformatics 2022; 38:3958-3967. [PMID: 35771595 DOI: 10.1093/bioinformatics/btac429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 04/30/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Computational methods for the prediction of protein-protein interactions (PPIs), while important tools for researchers, are plagued by challenges in generalizing to unseen proteins. Datasets used for modelling protein-protein predictions are particularly predisposed to information leakage and sampling biases. RESULTS In this study, we introduce RAPPPID, a method for the Regularized Automatic Prediction of Protein-Protein Interactions using Deep Learning. RAPPPID is a twin Averaged Weight-Dropped Long Short-Term memory network which employs multiple regularization methods during training time to learn generalized weights. Testing on stringent interaction datasets composed of proteins not seen during training, RAPPPID outperforms state-of-the-art methods. Further experiments show that RAPPPID's performance holds regardless of the particular proteins in the testing set and its performance is higher for experimentally supported edges. This study serves to demonstrate that appropriate regularization is an important component of overcoming the challenges of creating models for PPI prediction that generalize to unseen proteins. Additionally, as part of this study, we provide datasets corresponding to several data splits of various strictness, in order to facilitate assessment of PPI reconstruction methods by others in the future. AVAILABILITY AND IMPLEMENTATION Code and datasets are freely available at https://github.com/jszym/rapppid and Zenodo.org. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Joseph Szymborski
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC H3A 0G4, Canada.,Mila, Québec AI Institute, Montréal, QC H2S 3H1, Canada
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC H3A 0G4, Canada.,Mila, Québec AI Institute, Montréal, QC H2S 3H1, Canada.,The Rosalind and Morris Goodman Cancer Institute, Montréal, QC H3A 1A3, Canada
| |
Collapse
|
11
|
Jiang M, Fang X, Ma L, Liu M, Chen M, Liu J, Yang Y, Wang C. A nucleus-targeting peptide antagonist towards EZH2 displays therapeutic efficacy for lung cancer. Int J Pharm 2022; 622:121894. [DOI: 10.1016/j.ijpharm.2022.121894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 10/18/2022]
|
12
|
Berti C, Boarino A, Graciotti M, Bader LPE, Kandalaft LE, Klok HA. Reduction-Sensitive Protein Nanogels Enhance Uptake of Model and Tumor Lysate Antigens In Vitro by Mouse- and Human-Derived Dendritic Cells. ACS APPLIED BIO MATERIALS 2021; 4:8291-8300. [PMID: 35005925 DOI: 10.1021/acsabm.1c00828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Peptides and proteins represent an emerging class of powerful therapeutics. Peptide and protein nanogels are attractive carriers for the transport and delivery of biologically active peptides and proteins because they allow essentially quantitative encapsulation of these biologics. One interesting field of use of peptide and protein nanogels is the transport of antigens and adjuvants in cancer immunotherapy. This study demonstrates the use of reduction-sensitive protein nanogels for the delivery of ovalbumin and oxidized tumor lysate-based antigens to mouse and human-donor-derived dendritic cells. Challenging mouse-derived and human dendritic cells with reduction-sensitive ovalbumin nanogels was found to significantly enhance antigen uptake as compared to the use of the corresponding free protein antigen. The experiments with mouse-derived dendritic cells further showed that the administration of ovalbumin in the form of reduction-sensitive nanogels enhanced dendritic cell maturation as well as the presentation of the SIINFEKL epitope as compared to experiments that use free ovalbumin. In addition to ovalbumin as a model antigen, the feasibility of reduction-sensitive nanogels was also demonstrated for the delivery of oxidized, whole tumor lysate-based cancer antigens. In experiments with dendritic cells harvested from human donors, dendritic cell uptake of the oxidized tumor lysate antigen was significantly enhanced in experiments that used oxidized tumor lysate nanogels as compared to the free antigen.
Collapse
Affiliation(s)
- Cristiana Berti
- Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Alice Boarino
- Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Michele Graciotti
- Ludwig Cancer Research Center─Lausanne Branch, Department of Oncology, University Hospital of Lausanne, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lisa P E Bader
- Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Lana E Kandalaft
- Ludwig Cancer Research Center─Lausanne Branch, Department of Oncology, University Hospital of Lausanne, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Harm-Anton Klok
- Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|
13
|
Li R, Chen J, Gao X, Jiang G. Transcription factor KLF2 enhances the sensitivity of breast cancer cells to cisplatin by suppressing kinase WEE1. Cancer Biol Ther 2021; 22:465-477. [PMID: 34486497 DOI: 10.1080/15384047.2021.1949228] [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: 01/10/2023] Open
Abstract
Cisplatin is an effective chemotherapeutic agent in facilitating the inhibition of proliferation, migration, and invasion in cancerous cells. However, the detailed mechanism of the regulation by cisplatin of human breast cancer cells is still unclear. This study aimed to investigate the mechanism of kruppel-like factor 2 (KLF2) transcription factor in cisplatin therapy for breast cancer. RT-qPCR was performed to quantify the expression of KLF2 and WEE1 in clinical tissue samples from breast cancer patients and in MDA-MB-231 cells. ChIP assay and dual-luciferase reporter assay were used to analyze the potential-binding sites of KLF2 and WEE1 promoter. Gain- or loss-of-function approaches were used to manipulate KLF2 and WEE1 in cisplatin-treated MDA-MB-231 cells, and the mechanism of KLF2 in breast cancer was evaluated both via CCK-8 assay, flow cytometry, Transwell assay, and Western blot. Further validation of the KLF2 was performed on nude mouse models. Breast cancer tissues and cells showed a relative decline of KLF2 expression and abundant WEE1 expression. Cisplatin inhibited the proliferation, migration, and invasion of MDA-MB-231 cells. Overexpression of KLF2 enhanced the inhibitory effect of cisplatin on the malignant characteristics of MDA-MB-231 cells in vitro. KLF2 targeted WEE1 and negatively regulated its expression, thus enhancing the sensitivity to cisplatin of breast cancer cells as well as tumor-bearing mice. Overall, these results suggest that KLF2 can potentially inhibit WEE1 expression and sensitize breast cancer cells to cisplatin, thus presenting a promising adjunct treatment.
Collapse
Affiliation(s)
- Ruiqing Li
- Department of Throat and Breast Surgery, The Second Affiliated Hospital of Soochow University, Soochow P.R. China
| | - Jiejing Chen
- Department of Throat and Breast Surgery, Affiliated Hospital of Yangzhou University, Yangzhou P.R. China
| | - Xiaokang Gao
- Department of Throat and Breast Surgery, Affiliated Hospital of Yangzhou University, Yangzhou P.R. China
| | - Guoqin Jiang
- Department of Throat and Breast Surgery, The Second Affiliated Hospital of Soochow University, Soochow P.R. China
| |
Collapse
|
14
|
Sokolov AV, Limareva LV, Iliasov PV, Gribkova OV, Sustretov AS. Methods of Encapsulation of Biomacromolecules and Living Cells. Prospects of Using Metal–Organic Frameworks. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2021. [PMCID: PMC8141827 DOI: 10.1134/s1070428021040011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The review discusses different methods of encapsulation and biomineralization of macromolecules and living cells. Main advantages and disadvantages of most commonly used carriers, matrices, and materials for immobilization of proteins, enzymes, nucleic acids, and living cells are briefly surveyed. Examples of delivery vehicles for multifunctional encapsulation of protein-like substances are presented. Particular attention is paid to prospects of using metal–organic frameworks in medicine and biotechnology.
Collapse
Affiliation(s)
- A. V. Sokolov
- Institute of Experimental Medicine and Biotechnologies, Samara State Medical University, Ministry of Health of the Russian Federation, 443099 Samara, Russia
| | - L. V. Limareva
- Institute of Experimental Medicine and Biotechnologies, Samara State Medical University, Ministry of Health of the Russian Federation, 443099 Samara, Russia
| | - P. V. Iliasov
- Institute of Experimental Medicine and Biotechnologies, Samara State Medical University, Ministry of Health of the Russian Federation, 443099 Samara, Russia
| | - O. V. Gribkova
- Institute of Experimental Medicine and Biotechnologies, Samara State Medical University, Ministry of Health of the Russian Federation, 443099 Samara, Russia
| | - A. S. Sustretov
- Institute of Experimental Medicine and Biotechnologies, Samara State Medical University, Ministry of Health of the Russian Federation, 443099 Samara, Russia
| |
Collapse
|
15
|
Mishra S, Reshma G B, Pal S, Bano S, Gupta A, Kumari A, Ganguli M. Topical Application of Peptide-Chondroitin Sulfate Nanoparticles Allows Efficient Photoprotection in Skin. ACS APPLIED MATERIALS & INTERFACES 2021; 13:2382-2398. [PMID: 33406837 DOI: 10.1021/acsami.0c22011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In this article, we describe a method of delivery of chondroitin sulfate to skin as nanoparticles and demonstrate its anti-inflammatory and antioxidant role using UV irradiation as a model condition. These nanoparticles, formed through electrostatic interactions of chondroitin sulfate with a skin-penetrating peptide, were found to be homogenous with positive surface charges and stable at physiological and acidic pH under certain conditions. They were able to enter into the human keratinocyte cell line (HaCaT), artificial skin membrane (mimicking the human skin), and mouse skin tissue unlike free chondroitin sulfate. The preapplication of nanoparticles also exhibited reduced levels of oxidative stress, cyclobutane pyrimidine dimer formation, TNF-α, and so on in UV-B-irradiated HaCaT cells. In an acute UV-B irradiation mouse model, their topical application resulted in reduced epidermal thickness and sunburn cells, unlike in the case of free chondroitin sulfate. Thus, a completely noninvasive method was used to deliver a bio-macromolecule into the skin without using injections or abrasive procedures.
Collapse
Affiliation(s)
- Sarita Mishra
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Betsy Reshma G
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Simanti Pal
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Subia Bano
- Elvesys Microfluidics Innovation Centre, Paris 75011, France
| | - Aanchal Gupta
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anupama Kumari
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|