1
|
Li M, Gao Z, Wang N, Sekhar KPC, Hao J, Cui J. Targeting of Low-Immunogenic Poly(ethylene glycol) Nanoparticles for Photothermal-Enhanced Immunotherapy. Adv Healthc Mater 2024:e2402954. [PMID: 39676379 DOI: 10.1002/adhm.202402954] [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: 08/08/2024] [Revised: 11/06/2024] [Indexed: 12/17/2024]
Abstract
The assembly of low-immunogenic poly(ethylene glycol) nanoparticles (PEG NPs) for targeted delivery of therapeutics (i.e., mitoxantrone and imidazoquinoline) and improved photothermal-immunotherapy is reported. The targeted PEG NPs incorporating targeting molecules of hyaluronic acid are engineered via the templating of metal-organic frameworks, which can circumvent accelerated blood clearance and exhibit prolonged circulation time as well as improved accumulation of therapeutics at tumor sites. The targeted delivery of mitoxantrone under laser radiation induces immunogenic cell death of tumor cells, which is combined with toll-like receptor 7/8 agonists of imidazoquinoline to trigger immune responses of cytotoxic T lymphocytes for the eradication of tumor cells. Furthermore, the treatment can induce tumor-specific immune responses that inhibit metastatic lung tumor growth. This reported targeted PEG NPs provide a rational design for cancer immunotherapy.
Collapse
Affiliation(s)
- Mengqi Li
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Zhiliang Gao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Ning Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Kanaparedu P C Sekhar
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong University, Jinan, 250100, China
| |
Collapse
|
2
|
Liu YC, Ansaryan S, Tan J, Broguiere N, Lorenzo-Martín LF, Homicsko K, Coukos G, Lütolf MP, Altug H. Nanoplasmonic Single-Tumoroid Microarray for Real-Time Secretion Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401539. [PMID: 38924371 PMCID: PMC11425908 DOI: 10.1002/advs.202401539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/30/2024] [Indexed: 06/28/2024]
Abstract
Organoid tumor models have emerged as a powerful tool in the fields of biology and medicine as such 3D structures grown from tumor cells recapitulate better tumor characteristics, making these tumoroids unique for personalized cancer research. Assessment of their functional behavior, particularly protein secretion, is of significant importance to provide comprehensive insights. Here, a label-free spectroscopic imaging platform is presented with advanced integrated optofluidic nanoplasmonic biosensor that enables real-time secretion analysis from single tumoroids. A novel two-layer microwell design isolates tumoroids, preventing signal interference, and the microarray configuration allows concurrent analysis of multiple tumoroids. The dual imaging capability combining time-lapse plasmonic spectroscopy and bright-field microscopy facilitates simultaneous observation of secretion dynamics, motility, and morphology. The integrated biosensor is demonstrated with colorectal tumoroids derived from both cell lines and patient samples to investigate their vascular endothelial growth factor A (VEGF-A) secretion, growth, and movement under various conditions, including normoxia, hypoxia, and drug treatment. This platform, by offering a label-free approach with nanophotonics to monitor tumoroids, can pave the way for new applications in fundamental biological studies, drug screening, and the development of therapies.
Collapse
Affiliation(s)
- Yen-Cheng Liu
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Saeid Ansaryan
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Jiayi Tan
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Luis Francisco Lorenzo-Martín
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Krisztian Homicsko
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, Lausanne, 1005, Switzerland
- Ludwig Institute for Cancer Research, Ludwig Lausanne Branch, Chem. des Boveresses 155, Epalinges, 1066, Switzerland
- Swiss Cancer Center Leman, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
- Agora Translational Research Center, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
| | - George Coukos
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, Lausanne, 1005, Switzerland
- Ludwig Institute for Cancer Research, Ludwig Lausanne Branch, Chem. des Boveresses 155, Epalinges, 1066, Switzerland
- Swiss Cancer Center Leman, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
- Agora Translational Research Center, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
| | - Matthias P Lütolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Hatice Altug
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| |
Collapse
|
3
|
Barz M, Parak WJ, Zentel R. Concepts and Approaches to Reduce or Avoid Protein Corona Formation on Nanoparticles: Challenges and Opportunities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402935. [PMID: 38976560 PMCID: PMC11425909 DOI: 10.1002/advs.202402935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/19/2024] [Indexed: 07/10/2024]
Abstract
This review describes the formation of a protein corona (or its absence) on different classes of nanoparticles, its basic principles, and its consequences for nanomedicine. For this purpose, it describes general concepts to control (guide/minimize) the interaction between artificial nanoparticles and plasma proteins to reduce protein corona formation. Thereafter, methods for the qualitative or quantitative determination of protein corona formation are presented, as well as the properties of nanoparticle surfaces, which are relevant for protein corona prevention (or formation). Thereby especially the role of grafting density of hydrophilic polymers on the surface of the nanoparticle is discussed to prevent the formation of a protein corona. In this context also the potential of detergents (surfactants) for a temporary modification as well as grafting-to and grafting-from approaches for a permanent modification of the surface are discussed. The review concludes by highlighting several promising avenues. This includes (i) the use of nanoparticles without protein corona for active targeting, (ii) the use of synthetic nanoparticles without protein corona formation to address the immune system, (iii) the recollection of nanoparticles with a defined protein corona after in vivo application to sample the blood proteome and (iv) further concepts to reduce protein corona formation.
Collapse
Affiliation(s)
- Matthias Barz
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, NL-2333 CC, Netherlands
| | - Wolfgang J Parak
- Institut für Nanostruktur- und Festkörperphysik, Universität Hamburg, Luruper Chaussee 149, D-22761, Hamburg, Germany
| | - Rudolf Zentel
- Department of Chemistry, Johannes Gutenberg-University of Mainz, Duesbergweg 10-14, D-55128, Mainz, Germany
| |
Collapse
|
4
|
Dang BTN, Kwon TK, Lee S, Jeong JH, Yook S. Nanoparticle-based immunoengineering strategies for enhancing cancer immunotherapy. J Control Release 2024; 365:773-800. [PMID: 38081328 DOI: 10.1016/j.jconrel.2023.12.007] [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: 09/07/2023] [Revised: 11/27/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
Cancer immunotherapy is a groundbreaking strategy that has revolutionized the field of oncology compared to other therapeutic strategies, such as surgery, chemotherapy, or radiotherapy. However, cancer complexity, tumor heterogeneity, and immune escape have become the main hurdles to the clinical application of immunotherapy. Moreover, conventional immunotherapies cause many harmful side effects owing to hyperreactivity in patients, long treatment durations and expensive cost. Nanotechnology is considered a transformative approach that enhances the potency of immunotherapy by capitalizing on the superior physicochemical properties of nanocarriers, creating highly targeted tissue delivery systems. These advantageous features include a substantial specific surface area, which enhances the interaction with the immune system. In addition, the capability to finely modify surface chemistry enables the achievement of controlled and sustained release properties. These advances have significantly increased the potential of immunotherapy, making it more powerful than ever before. In this review, we introduce recent nanocarriers for application in cancer immunotherapy based on strategies that target different main immune cells, including T cells, dendritic cells, natural killer cells, and tumor-associated macrophages. We also provide an overview of the role and significance of nanotechnology in cancer immunotherapy.
Collapse
Affiliation(s)
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
5
|
Heck AG, Stickdorn J, Rosenberger LJ, Scherger M, Woller J, Eigen K, Bros M, Grabbe S, Nuhn L. Polymerizable 2-Propionic-3-methylmaleic Anhydrides as a Macromolecular Carrier Platform for pH-Responsive Immunodrug Delivery. J Am Chem Soc 2023; 145:27424-27436. [PMID: 38054646 DOI: 10.1021/jacs.3c08511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
The design of functional polymers coupled with stimuli-triggered drug release mechanisms is a promising achievement to overcome various biological barriers. pH trigger methods yield significant potential for controlled targeting and release of therapeutics due to their simplicity and relevance, especially upon cell internalization. Here, we introduce reactive polymers that conjugate primary or secondary amines and release potential drugs under acidic conditions. For that purpose, we introduced methacrylamide-based monomers with pendant 2-propionic-3-methylmaleic anhydride groups. Such groups allow the conjugation of primary and secondary amines but are resistant to radical polymerization conditions. We, therefore, polymerized 2-propionic-3-methylmaleic anhydride amide-based methacrylates via reversible addition-fragmentation chain transfer (RAFT) polymerization. Their amine-reactive anhydrides could sequentially be derivatized by primary or secondary amines into hydrophilic polymers. Acidic pH-triggered drug release from the polymeric systems was fine-tuned by comparing different amines. Thereby, the conjugation of primary amines led to the formation of irreversible imide bonds in dimethyl sulfoxide, while secondary amines could quantitatively be released upon acidification. In vitro, this installed pH-responsiveness can contribute to an effective release of conjugated immune stimulatory drugs under endosomal pH conditions. Interestingly, the amine-modified polymers generally showed no toxicity and a high cellular uptake. Furthermore, secondary amine-modified immune stimulatory drugs conjugated to the polymers yielded better receptor activity and immune cell maturation than their primary amine derivatives due to their pH-sensitive drug release mechanism. Consequently, 2-propionic-3-methylmaleic anhydride-based polymers can be considered as a versatile platform for pH-triggered delivery of various (immuno)drugs, thus enabling new strategies in macromolecule-assisted immunotherapy.
Collapse
Affiliation(s)
- Alina G Heck
- Max Planck Institute for Polymer Research, Mainz 55128, Germany
| | | | - Laura J Rosenberger
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | | | - Jonas Woller
- Max Planck Institute for Polymer Research, Mainz 55128, Germany
| | - Katharina Eigen
- Institute of Functional Materials and Biofabrication, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Würzburg 97070, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research, Mainz 55128, Germany
- Institute of Functional Materials and Biofabrication, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Würzburg 97070, Germany
| |
Collapse
|
6
|
Lybaert L, Thielemans K, Feldman SA, van der Burg SH, Bogaert C, Ott PA. Neoantigen-directed therapeutics in the clinic: where are we? Trends Cancer 2023; 9:503-519. [PMID: 37055237 PMCID: PMC10414146 DOI: 10.1016/j.trecan.2023.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 04/15/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cell therapy have brought immunotherapy to the forefront of cancer treatment; however, only subsets of patients benefit from current approaches. Neoantigen-driven therapeutics specifically redirect the immune system of the patient to enable or reinduce its ability to recognize and eliminate cancer cells. The tumor specificity of this strategy spares healthy and normal cells from being attacked. Consistent with this concept, initial clinical trials have demonstrated the feasibility, safety, and immunogenicity of neoantigen-directed personalized vaccines. We review neoantigen-driven therapy strategies as well as their promise and clinical successes to date.
Collapse
Affiliation(s)
| | | | - Steven A Feldman
- Center for Cancer Cell Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Sjoerd H van der Burg
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
7
|
Ramos E, Egea J, López-Muñoz F, Gil-Martín E, Romero A. Therapeutic Potential of Melatonin Counteracting Chemotherapy-Induced Toxicity in Breast Cancer Patients: A Systematic Review. Pharmaceutics 2023; 15:1616. [PMID: 37376065 DOI: 10.3390/pharmaceutics15061616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/05/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
The purpose of this systematic review is to provide an overview of the existing knowledge on the therapeutic potential of melatonin to counteract the undesirable effects of chemotherapy in breast cancer patients. To this aim, we summarized and critically reviewed preclinical- and clinical-related evidence according to the PRISMA guidelines. Additionally, we developed an extrapolation of melatonin doses in animal studies to the human equivalent doses (HEDs) for randomized clinical trials (RCTs) with breast cancer patients. For the revision, 341 primary records were screened, which were reduced to 8 selected RCTs that met the inclusion criteria. We assembled the evidence drawn from these studies by analyzing the remaining gaps and treatment efficacy and suggested future translational research and clinical trials. Overall, the selected RCTs allow us to conclude that melatonin combined with standard chemotherapy lines would derive, at least, a better quality of life for breast cancer patients. Moreover, regular doses of 20 mg/day seemed to increase partial response and 1-year survival rates. Accordingly, this systematic review leads us to draw attention to the need for more RCTs to provide a comprehensive view of the promising actions of melatonin in breast cancer and, given the safety profile of this molecule, adequate translational doses should be established in further RCTs.
Collapse
Affiliation(s)
- Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain
- Institute Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Francisco López-Muñoz
- Faculty of Health, Camilo José Cela University of Madrid (UCJC), 28692 Madrid, Spain
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute, 28041 Madrid, Spain
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, 36310 Vigo, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
8
|
Mao L, Ma P, Luo X, Cheng H, Wang Z, Ye E, Loh XJ, Wu YL, Li Z. Stimuli-Responsive Polymeric Nanovaccines Toward Next-Generation Immunotherapy. ACS NANO 2023. [PMID: 37207347 DOI: 10.1021/acsnano.3c02273] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The development of nanovaccines that employ polymeric delivery carriers has garnered substantial interest in therapeutic treatment of cancer and a variety of infectious diseases due to their superior biocompatibility, lower toxicity and reduced immunogenicity. Particularly, stimuli-responsive polymeric nanocarriers show great promise for delivering antigens and adjuvants to targeted immune cells, preventing antigen degradation and clearance, and increasing the uptake of specific antigen-presenting cells, thereby sustaining adaptive immune responses and improving immunotherapy for certain diseases. In this review, the most recent advances in the utilization of stimulus-responsive polymer-based nanovaccines for immunotherapeutic applications are presented. These sophisticated polymeric nanovaccines with diverse functions, aimed at therapeutic administration for disease prevention and immunotherapy, are further classified into several active domains, including pH, temperature, redox, light and ultrasound-sensitive intelligent nanodelivery systems. Finally, the potential strategies for the future design of multifunctional next-generation polymeric nanovaccines by integrating materials science with biological interface are proposed.
Collapse
Affiliation(s)
- Liuzhou Mao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Panqin Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xi Luo
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, 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 361102, China
| | - Zhanxiang Wang
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Enyi Ye
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| |
Collapse
|
9
|
Tian H, Cao J, Li B, Nice EC, Mao H, Zhang Y, Huang C. Managing the immune microenvironment of osteosarcoma: the outlook for osteosarcoma treatment. Bone Res 2023; 11:11. [PMID: 36849442 PMCID: PMC9971189 DOI: 10.1038/s41413-023-00246-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 03/01/2023] Open
Abstract
Osteosarcoma, with poor survival after metastasis, is considered the most common primary bone cancer in adolescents. Notwithstanding the efforts of researchers, its five-year survival rate has only shown limited improvement, suggesting that existing therapeutic strategies are insufficient to meet clinical needs. Notably, immunotherapy has shown certain advantages over traditional tumor treatments in inhibiting metastasis. Therefore, managing the immune microenvironment in osteosarcoma can provide novel and valuable insight into the multifaceted mechanisms underlying the heterogeneity and progression of the disease. Additionally, given the advances in nanomedicine, there exist many advanced nanoplatforms for enhanced osteosarcoma immunotherapy with satisfactory physiochemical characteristics. Here, we review the classification, characteristics, and functions of the key components of the immune microenvironment in osteosarcoma. This review also emphasizes the application, progress, and prospects of osteosarcoma immunotherapy and discusses several nanomedicine-based options to enhance the efficiency of osteosarcoma treatment. Furthermore, we examine the disadvantages of standard treatments and present future perspectives for osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Hailong Tian
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Jiangjun Cao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Bowen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Edouard C. Nice
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315020, People's Republic of China.
| | - Yi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
10
|
Zhou K, Cai C, Ding G, He Y, Hu D. A signature of six-hypoxia-related genes to evaluate the tumor immune microenvironment and predict prognosis in gastric cancer. BMC Med Genomics 2022; 15:261. [PMID: 36527012 PMCID: PMC9755770 DOI: 10.1186/s12920-022-01411-9] [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: 04/01/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Hypoxia will trigger a series of immunosuppressive process in tumor microenvironment, leading to the progression in gastric cancer (GC). This research aims to establish a prognostic model made up of hypoxia-risk-related genes in GC. METHODS Hypoxic genes were outlined via the protein-protein interaction network. And a prognostic model was developed using univariate cox analysis and lasso regression from data in TCGA. Two independent queues of GEO were used for validation. RESULTS We set up a hypoxic model presented as an independent prognostic factor for GC. And a nomogram combined this model with clinical features can predict OS with great performance. Furthermore, DNA methylation, IHC and cell line analyses validated the expression of hypoxic genes in GC. CONCLUSIONS In summary, we proposed and verified a hypoxia-risk-related model, which could reflect the immune microenvironment and predict prognosis in GC.
Collapse
Affiliation(s)
- Kena Zhou
- Gastroenterology Department of Ningbo No. 9 Hospital, No.68 Xiangbei Road, Jiangbei District, Ningbo, 315000 Zhejiang China
| | - Congbo Cai
- Emergency Department of Yinzhou No.2 Hospital, Ningbo, 315000 Zhejiang China
| | - Guanjun Ding
- General Surgery Department of Ningbo No. 9 Hospital, Ningbo, 315000 Zhejiang China
| | - Yi He
- Gastroenterology Department of Ningbo No. 9 Hospital, No.68 Xiangbei Road, Jiangbei District, Ningbo, 315000 Zhejiang China
| | - Di Hu
- Gastroenterology Department of Ningbo No. 9 Hospital, No.68 Xiangbei Road, Jiangbei District, Ningbo, 315000 Zhejiang China
| |
Collapse
|
11
|
Barz M, Nuhn L, Hörpel G, Zentel R. From Self-Organization to Tumor-Immune Therapy: How Things Started and How They Evolved. Macromol Rapid Commun 2022; 43:e2100829. [PMID: 35729069 DOI: 10.1002/marc.202100829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Indexed: 11/08/2022]
Affiliation(s)
- Matthias Barz
- Leiden Academic Center for Drug Research (LACDR), Einsteinweg 55, 2333 CC Leiden, The Netherlands.,Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Gerhard Hörpel
- GBH Gesellschaft für Batterie Know-how mbH, Lerchenhain 84, 48301, Nottuln, Germany
| | - Rudolf Zentel
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| |
Collapse
|
12
|
Stickdorn J, Stein L, Arnold-Schild D, Hahlbrock J, Medina-Montano C, Bartneck J, Ziß T, Montermann E, Kappel C, Hobernik D, Haist M, Yurugi H, Raabe M, Best A, Rajalingam K, Radsak MP, David SA, Koynov K, Bros M, Grabbe S, Schild H, Nuhn L. Systemically Administered TLR7/8 Agonist and Antigen-Conjugated Nanogels Govern Immune Responses against Tumors. ACS NANO 2022; 16:4426-4443. [PMID: 35103463 PMCID: PMC8945363 DOI: 10.1021/acsnano.1c10709] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The generation of specific humoral and cellular immune responses plays a pivotal role in the development of effective vaccines against tumors. Especially the presence of antigen-specific, cytotoxic T cells influences the outcome of therapeutic cancer vaccinations. Different strategies, ranging from delivering antigen-encoding mRNAs to peptides or full antigens, are accessible but often suffer from insufficient immunogenicity and require immune-boosting adjuvants as well as carrier platforms to ensure stability and adequate retention. Here, we introduce a pH-responsive nanogel platform as a two-component antitumor vaccine that is safe for intravenous application and elicits robust immune responses in vitro and in vivo. The underlying chemical design allows for straightforward covalent attachment of a model antigen (ovalbumin) and an immune adjuvant (imidazoquinoline-type TLR7/8 agonist) onto the same nanocarrier system. In addition to eliciting antigen-specific T and B cell responses that outperform mixtures of individual components, our two-component nanovaccine leads in prophylactic and therapeutic studies to an antigen-specific growth reduction of different tumors expressing ovalbumin intracellularly or on their surface. Regarding the versatile opportunities for functionalization, our nanogels are promising for the development of highly customized and potent nanovaccines.
Collapse
Affiliation(s)
- Judith Stickdorn
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Lara Stein
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Danielle Arnold-Schild
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Jennifer Hahlbrock
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Carolina Medina-Montano
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Joschka Bartneck
- III Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University
Mainz, Langenbeckstraße
1, 55131 Mainz, Germany
| | - Tanja Ziß
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Evelyn Montermann
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Cinja Kappel
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Dominika Hobernik
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Maximilian Haist
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Hajime Yurugi
- Cell
Biology Unit, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Marco Raabe
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Andreas Best
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Krishnaraj Rajalingam
- Cell
Biology Unit, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Markus P. Radsak
- III Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University
Mainz, Langenbeckstraße
1, 55131 Mainz, Germany
| | - Sunil A. David
- ViroVax,
LLC, 2029 Becker Drive
Suite 100E, Lawrence 66047-1620, Kansas. United States
| | - Kaloian Koynov
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Matthias Bros
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stephan Grabbe
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Hansjörg Schild
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Lutz Nuhn
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
13
|
Berti C, Graciotti M, Boarino A, Yakkala C, Kandalaft LE, Klok HA. Polymer Nanoparticle-Mediated Delivery of Oxidized Tumor Lysate-Based Cancer Vaccines. Macromol Biosci 2021; 22:e2100356. [PMID: 34822219 DOI: 10.1002/mabi.202100356] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/09/2021] [Indexed: 12/17/2022]
Abstract
Cancer vaccination is a powerful strategy to combat cancer. A very attractive approach to prime the immune system against cancer cells involves the use of tumor lysate as antigen source. The immunogenicity of tumor lysate can be further enhanced by treatment with hypochlorous acid. This study explores poly(lactic-co-glycolic acid) (PLGA) nanoparticles to enhance the delivery of oxidized tumor lysate to dendritic cells. Using human donor-derived dendritic cells, it is found that the use of PLGA nanoparticles enhances antigen uptake and dendritic cell maturation, as compared to the use of the free tumor lysate. The ability of the activated dendritic cells to stimulate autologous peripheral blood mononuclear cells (PBMCs) is assessed in vitro by coculturing PBMCs with A375 melanoma cells. Live cell imaging analysis of this experiment highlights the potential of nanoparticle-mediated dendritic-cell-based vaccination approaches. Finally, the efficacy of the PLGA nanoparticle formulation is evaluated in vivo in a therapeutic vaccination study using B16F10 tumor-bearing C57BL/6J mice. Animals that are challenged with the polymer nanoparticle-based oxidized tumor lysate formulation survive for up to 50 days, in contrast to a maximum of 41 days for the group that receives the corresponding free oxidized tumor lysate-based vaccine.
Collapse
Affiliation(s)
- Cristiana Berti
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, Lausanne, CH-1015, Switzerland
| | - Michele Graciotti
- Ludwig Cancer Research Center - Lausanne Branch, Department of Oncology, University Hospital of Lausanne, University of Lausanne, Lausanne, CH-1011, Switzerland
| | - Alice Boarino
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, Lausanne, CH-1015, Switzerland
| | - Chakradhar Yakkala
- Ludwig Cancer Research Center - Lausanne Branch, Department of Oncology, University Hospital of Lausanne, University of Lausanne, Lausanne, CH-1011, Switzerland
| | - Lana E Kandalaft
- Ludwig Cancer Research Center - Lausanne Branch, Department of Oncology, University Hospital of Lausanne, University of Lausanne, Lausanne, CH-1011, Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, Lausanne, CH-1015, Switzerland
| |
Collapse
|
14
|
Gao S, Yang X, Xu J, Qiu N, Zhai G. Nanotechnology for Boosting Cancer Immunotherapy and Remodeling Tumor Microenvironment: The Horizons in Cancer Treatment. ACS NANO 2021; 15:12567-12603. [PMID: 34339170 DOI: 10.1021/acsnano.1c02103] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunotherapy that harnesses the human immune system to fight cancer has received widespread attention and become a mainstream strategy for cancer treatment. Cancer immunotherapy not only eliminates primary tumors but also treats metastasis and recurrence, representing a major advantage over traditional cancer treatments. Recently with the development of nanotechnology, there exists much work applying nanomaterials to cancer immunotherapy on the basis of their excellent physiochemical properties, such as efficient tissue-specific delivery function, huge specific surface area, and controllable surface chemistry. Consequently, nanotechnology holds significant potential in improving the efficacy of cancer immunotherapy. Nanotechnology-based immunotherapy mainly manifests its inhibitory effect on tumors via two different approaches: one is to produce an effective anti-tumor immune response during tumorigenesis, and the other is to enhance tumor immune defense ability by modulating the immune suppression mechanism in the tumor microenvironment. With the success of tumor immunotherapy, understanding the interaction between the immune system and smart nanomedicine has provided vigorous vitality for the development of cancer treatment. This review highlights the application, progress, and prospect of nanomedicine in the process of tumor immunoediting and also discusses several engineering methods to improve the efficiency of tumor treatment.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Xiaoye Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Jiangkang Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Na Qiu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| |
Collapse
|
15
|
Huppertsberg A, Kaps L, Zhong Z, Schmitt S, Stickdorn J, Deswarte K, Combes F, Czysch C, De Vrieze J, Kasmi S, Choteschovsky N, Klefenz A, Medina-Montano C, Winterwerber P, Chen C, Bros M, Lienenklaus S, Sanders NN, Koynov K, Schuppan D, Lambrecht BN, David SA, De Geest BG, Nuhn L. Squaric Ester-Based, pH-Degradable Nanogels: Modular Nanocarriers for Safe, Systemic Administration of Toll-like Receptor 7/8 Agonistic Immune Modulators. J Am Chem Soc 2021; 143:9872-9883. [PMID: 34166595 PMCID: PMC8267846 DOI: 10.1021/jacs.1c03772] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 12/25/2022]
Abstract
Small-molecular Toll-like receptor 7/8 (TLR7/8) agonists hold promise as immune modulators for a variety of immune therapeutic purposes including cancer therapy or vaccination. However, due to their rapid systemic distribution causing difficult-to-control inflammatory off-target effects, their application is still problematic, in particular systemically. To address this problem, we designed and robustly fabricated pH-responsive nanogels serving as versatile immunodrug nanocarriers for safe delivery of TLR7/8-stimulating imidazoquinolines after intravenous administration. To this aim, a primary amine-reactive methacrylamide monomer bearing a pendant squaric ester amide is introduced, which is polymerized under controlled RAFT polymerization conditions. Corresponding PEG-derived squaric ester amide block copolymers self-assemble into precursor micelles in polar protic solvents. Their cores are amine-reactive and can sequentially be transformed by acid-sensitive cross-linkers, dyes, and imidazoquinolines. Remaining squaric ester amides are hydrophilized affording fully hydrophilic nanogels with profound stability in human plasma but stimuli-responsive degradation upon exposure to endolysosomal pH conditions. The immunomodulatory behavior of the imidazoquinolines alone or conjugated to the nanogels was demonstrated by macrophages in vitro. In vivo, however, we observed a remarkable impact of the nanogel: After intravenous injection, a spatially controlled immunostimulatory activity was evident in the spleen, whereas systemic off-target inflammatory responses triggered by the small-molecular imidazoquinoline analogue were absent. These findings underline the potential of squaric ester-based, pH-degradable nanogels as a promising platform to permit intravenous administration routes of small-molecular TLR7/8 agonists and, thus, the opportunity to explore their adjuvant potency for systemic vaccination or cancer immunotherapy purposes.
Collapse
Affiliation(s)
| | - Leonard Kaps
- Institute
for Translational Immunology and Research Center for Immune Therapy,
University Medical Center, Johannes Gutenberg-University
Mainz, 55131 Mainz, Germany
- Department
of Internal Medicine I, University Medical
Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Zifu Zhong
- Department
of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Sascha Schmitt
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | | | - Kim Deswarte
- Department
of Internal Medicine and Pediatrics, Ghent
University, VIB Center for Inflammation Research, Ghent 9052, Belgium
| | - Francis Combes
- Laboratory
of Gene Therapy, Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke 9820, Belgium
| | | | - Jana De Vrieze
- Department
of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Sabah Kasmi
- Department
of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Niklas Choteschovsky
- Institute
for Translational Immunology and Research Center for Immune Therapy,
University Medical Center, Johannes Gutenberg-University
Mainz, 55131 Mainz, Germany
| | - Adrian Klefenz
- Institute
for Translational Immunology and Research Center for Immune Therapy,
University Medical Center, Johannes Gutenberg-University
Mainz, 55131 Mainz, Germany
| | - Carolina Medina-Montano
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | | | - Chaojian Chen
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Matthias Bros
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Stefan Lienenklaus
- Institute
for Laboratory Animal Science and Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Niek N. Sanders
- Laboratory
of Gene Therapy, Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke 9820, Belgium
| | - Kaloian Koynov
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Detlef Schuppan
- Institute
for Translational Immunology and Research Center for Immune Therapy,
University Medical Center, Johannes Gutenberg-University
Mainz, 55131 Mainz, Germany
- Division
of Gastroenterology, Beth Israel Deaconess
Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Bart N. Lambrecht
- Department
of Internal Medicine and Pediatrics, Ghent
University, VIB Center for Inflammation Research, Ghent 9052, Belgium
- Department
of Pulmonary Medicine, Erasmus University
Medical Center, Rotterdam 3015, Netherlands
| | | | - Bruno G. De Geest
- Department
of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Lutz Nuhn
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| |
Collapse
|
16
|
Zhao Y, Li A, Jiang L, Gu Y, Liu J. Hybrid Membrane-Coated Biomimetic Nanoparticles (HM@BNPs): A Multifunctional Nanomaterial for Biomedical Applications. Biomacromolecules 2021; 22:3149-3167. [PMID: 34225451 DOI: 10.1021/acs.biomac.1c00440] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The application of nanoparticles in the diagnosis and treatment of diseases has undergone different developmental stages, but phagocytosis and nonspecific distribution have been the main factors restricting the transformation of nanobased drugs into clinical practice. In the past decade, the design of membrane-coated nanoparticles has gained increasing attention. It is hoped that the combination of the cell membrane's natural biological properties and the functional integration of synthetic nanoparticle systems can compensate for the shortage of traditional nanoparticles. The membrane coating gives the nanoparticles unique biological functions such as immune evasion and targeting capability. However, when the encapsulation of monotypic membranes does not meet the diverse demands of biomedicine, the combination of different cell membranes may offer more possibilities. In this review, the composition, preparation, and advantages of biomimetic nanoparticles coated with hybrid cell membranes are summarized, and the applications of hybrid membrane-coated biomimetic nanoparticles (HM@BNPs) in drug delivery, phototherapy, liquid biopsy, tumor vaccines, immune therapy, and detoxification are reviewed. Finally, the current challenges and opportunities with regard to HM@BNPs are discussed.
Collapse
Affiliation(s)
- Yunan Zhao
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Aixue Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Liangdi Jiang
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Wu H, Fu X, Zhai Y, Gao S, Yang X, Zhai G. Development of Effective Tumor Vaccine Strategies Based on Immune Response Cascade Reactions. Adv Healthc Mater 2021; 10:e2100299. [PMID: 34021717 DOI: 10.1002/adhm.202100299] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/11/2021] [Indexed: 12/13/2022]
Abstract
To solve the problems of high toxicity and poor efficacy of existing tumor treatment methods, researchers have developed a variety of tumor immunotherapies. Among them, tumor vaccines activate antigen-presenting cells and T lymphocytes upstream of the cancer-immunity cycle are considered the most promising therapy to activate the immune system. Nanocarriers are considered the most promising tumor vaccine delivery vehicles, including polymer nanocarriers, lipid nanocarriers, inorganic nanocarriers, and biomimetic nanocarriers that have been developed for vaccine delivery. Based on the cascade reaction for tumor vaccines to exert their effects, this review summarizes the four key factors for the design and construction of nano-tumor vaccines. The composition and functional characteristics of the corresponding preferred nanocarriers are illustrated to provide a reference for the development of effective tumor vaccines. Finally, potential challenges and perspectives are illustrated in the hope of improving the efficacy of tumor vaccine immunotherapy and accelerating the clinical transformation of next-generation tumor vaccines.
Collapse
Affiliation(s)
- Hang Wu
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Xianglei Fu
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry University of Utah Salt Lake City UT 84124 USA
| | - Shan Gao
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Xiaoye Yang
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| |
Collapse
|
18
|
Han L, Peng K, Qiu LY, Li M, Ruan JH, He LL, Yuan ZX. Hitchhiking on Controlled-Release Drug Delivery Systems: Opportunities and Challenges for Cancer Vaccines. Front Pharmacol 2021; 12:679602. [PMID: 34040536 PMCID: PMC8141731 DOI: 10.3389/fphar.2021.679602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer vaccines represent among the most promising strategies in the battle against cancers. However, the clinical efficacy of current cancer vaccines is largely limited by the lack of optimized delivery systems to generate strong and persistent antitumor immune responses. Moreover, most cancer vaccines require multiple injections to boost the immune responses, leading to poor patient compliance. Controlled-release drug delivery systems are able to address these issues by presenting drugs in a controlled spatiotemporal manner, which allows co-delivery of multiple drugs, reduction of dosing frequency and avoidance of significant systemic toxicities. In this review, we outline the recent progress in cancer vaccines including subunit vaccines, genetic vaccines, dendritic cell-based vaccines, tumor cell-based vaccines and in situ vaccines. Furthermore, we highlight the efforts and challenges of controlled or sustained release drug delivery systems (e.g., microparticles, scaffolds, injectable gels, and microneedles) in ameliorating the safety, effectiveness and operability of cancer vaccines. Finally, we briefly discuss the correlations of vaccine release kinetics and the immune responses to enlighten the rational design of the next-generation platforms for cancer therapy.
Collapse
Affiliation(s)
- Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Ke Peng
- School of pharmacy, Queen's University Belfast, Belfast, United Kingdom
| | - Li-Ying Qiu
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Meng Li
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Jing-Hua Ruan
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Li-Li He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| |
Collapse
|
19
|
Stevens D, Ingels J, Van Lint S, Vandekerckhove B, Vermaelen K. Dendritic Cell-Based Immunotherapy in Lung Cancer. Front Immunol 2021; 11:620374. [PMID: 33679709 PMCID: PMC7928408 DOI: 10.3389/fimmu.2020.620374] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide. The advent of immune checkpoint inhibitors has led to a paradigm shift in the treatment of metastatic non-small cell and small cell lung cancer. However, despite prolonged overall survival, only a minority of the patients derive clinical benefit from these treatments suggesting that the full anti-tumoral potential of the immune system is not being harnessed yet. One way to overcome this problem is to combine immune checkpoint blockade with different strategies aimed at inducing or restoring cellular immunity in a tumor-specific, robust, and durable way. Owing to their unique capacity to initiate and regulate T cell responses, dendritic cells have been extensively explored as tools for immunotherapy in many tumors, including lung cancer. In this review, we provide an update on the nearly twenty years of experience with dendritic cell-based immunotherapy in lung cancer. We summarize the main results from the early phase trials and give an overview of the future perspectives within this field.
Collapse
Affiliation(s)
- Dieter Stevens
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Sandra Van Lint
- Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium.,GMP Cell Therapy Unit, Department of Regenerative Medicine, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Karim Vermaelen
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| |
Collapse
|
20
|
Yang S, Liu Q, Liao Q. Tumor-Associated Macrophages in Pancreatic Ductal Adenocarcinoma: Origin, Polarization, Function, and Reprogramming. Front Cell Dev Biol 2021; 8:607209. [PMID: 33505964 PMCID: PMC7829544 DOI: 10.3389/fcell.2020.607209] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy. PDAC is only cured by surgical resection in its early stage, but there remains a relatively high possibility of recurrence. The development of PDAC is closely associated with the tumor microenvironment. Tumor-associated macrophages (TAMs) are one of the most abundant immune cell populations in the pancreatic tumor stroma. TAMs are inclined to M2 deviation in the tumor microenvironment, which promotes and supports tumor behaviors, including tumorigenesis, immune escape, metastasis, and chemotherapeutic resistance. Herein, we comprehensively reviewed the latest researches on the origin, polarization, functions, and reprogramming of TAMs in PDAC.
Collapse
Affiliation(s)
- Sen Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Fontana F, Bartolo R, Santos HA. Biohybrid Nanosystems for Cancer Treatment: Merging the Best of Two Worlds. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:135-162. [PMID: 33543459 DOI: 10.1007/978-3-030-58174-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
During the last 20+ years, research into the biomedical application of nanotechnology has helped in reshaping cancer treatment. The clinical use of several passively targeted nanosystems resulted in improved quality of care for patients. However, the therapeutic efficacy of these systems is not superior to the original drugs. Moreover, despite extensive investigations into actively targeted nanocarriers, numerous barriers still remain before their successful clinical translation, including sufficient bloodstream circulation time and efficient tumor targeting. The combination of synthetic nanomaterials with biological elements (e.g., cells, cell membranes, and macromolecules) is presently the cutting-edge research in cancer nanotechnology. The features provided by the biological moieties render the particles with prolonged bloodstream circulation time and homotopic targeting to the tumor site. Moreover, cancer cell membranes serve as sources of neoantigens, useful in the formulation of nanovaccines. In this chapter, we will discuss the advantages of biohybrid nanosystems in cancer chemotherapy, immunotherapy, and combined therapy, as well as highlight their preparation methods and clinical translatability.
Collapse
Affiliation(s)
- Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Raquél Bartolo
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
22
|
Fuchs N, Meta M, Schuppan D, Nuhn L, Schirmeister T. Novel Opportunities for Cathepsin S Inhibitors in Cancer Immunotherapy by Nanocarrier-Mediated Delivery. Cells 2020; 9:E2021. [PMID: 32887380 PMCID: PMC7565055 DOI: 10.3390/cells9092021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Cathepsin S (CatS) is a secreted cysteine protease that cleaves certain extracellular matrix proteins, regulates antigen presentation in antigen-presenting cells (APC), and promotes M2-type macrophage and dendritic cell polarization. CatS is overexpressed in many solid cancers, and overall, it appears to promote an immune-suppressive and tumor-promoting microenvironment. While most data suggest that CatS inhibition or knockdown promotes anti-cancer immunity, cell-specific inhibition, especially in myeloid cells, appears to be important for therapeutic efficacy. This makes the design of CatS selective inhibitors and their targeting to tumor-associated M2-type macrophages (TAM) and DC an attractive therapeutic strategy compared to the use of non-selective immunosuppressive compounds or untargeted approaches. The selective inhibition of CatS can be achieved through optimized small molecule inhibitors that show good pharmacokinetic profiles and are orally bioavailable. The targeting of these inhibitors to TAM is now more feasible using nanocarriers that are functionalized for a directed delivery. This review discusses the role of CatS in the immunological tumor microenvironment and upcoming possibilities for a nanocarrier-mediated delivery of potent and selective CatS inhibitors to TAM and related APC to promote anti-tumor immunity.
Collapse
Affiliation(s)
- Natalie Fuchs
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University of Mainz, Staudingerweg 5, D, 55128 Mainz, Germany; (N.F.); (M.M.)
| | - Mergim Meta
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University of Mainz, Staudingerweg 5, D, 55128 Mainz, Germany; (N.F.); (M.M.)
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 63, 55131 Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research Ackermannweg 10, 55128 Mainz, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University of Mainz, Staudingerweg 5, D, 55128 Mainz, Germany; (N.F.); (M.M.)
| |
Collapse
|
23
|
Kockelmann J, Stickdorn J, Kasmi S, De Vrieze J, Pieszka M, Ng DYW, David SA, De Geest BG, Nuhn L. Control over Imidazoquinoline Immune Stimulation by pH-Degradable Poly(norbornene) Nanogels. Biomacromolecules 2020; 21:2246-2257. [PMID: 32255626 PMCID: PMC7304817 DOI: 10.1021/acs.biomac.0c00205] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
The
reactivation of the innate immune system by toll-like receptor
(TLR) agonists holds promise for anticancer immunotherapy. Severe
side effects caused by unspecific and systemic activation of the immune
system upon intravenous injection prevent the use of small-molecule
TLR agonists for such purposes. However, a covalent attachment of
small-molecule imidazoquinoline (IMDQ) TLR7/8 agonists to pH-degradable
polymeric nanogels could be shown to drastically reduce the systemic
inflammation but retain the activity to tumoral tissues and their
draining lymph nodes. Here, we introduce the synthesis of poly(norbornene)-based,
acid-degradable nanogels for the covalent ligation of IMDQs. While
the intact nanogels trigger sufficient TLR7/8 receptor stimulation,
their degraded version of soluble, IMDQ-conjugated poly(norbornene)
chains hardly activates TLR7/8. This renders their clinical safety
profile, as degradation products are obtained, which would not only
circumvent nanoparticle accumulation in the body but also provide
nonactive, polymer-bound IMDQ species. Their immunologically silent
behavior guarantees both spatial and temporal control over immune
activity and, thus, holds promise for improved clinical applications.
Collapse
Affiliation(s)
- Johannes Kockelmann
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Judith Stickdorn
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Sabah Kasmi
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Jana De Vrieze
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Michaela Pieszka
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - David Yuen W Ng
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Sunil A David
- Department of Medicinal Chemistry, University of Minnesota, 2231 Sixth Street SE, Minneapolis, Minnesota 55455, United States
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
24
|
|
25
|
De Vrieze J, Louage B, Deswarte K, Zhong Z, De Coen R, Van Herck S, Nuhn L, Kaas Frich C, Zelikin AN, Lienenklaus S, Sanders NN, Lambrecht BN, David SA, De Geest BG. Potent Lymphatic Translocation and Spatial Control Over Innate Immune Activation by Polymer-Lipid Amphiphile Conjugates of Small-Molecule TLR7/8 Agonists. Angew Chem Int Ed Engl 2019; 58:15390-15395. [PMID: 31397948 DOI: 10.1002/anie.201905687] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/25/2019] [Indexed: 12/16/2022]
Abstract
Uncontrolled systemic inflammatory immune triggering has hampered the clinical translation of several classes of small-molecule immunomodulators, such as imidazoquinoline TLR7/8 agonists for vaccine design and cancer immunotherapy. By taking advantage of the inherent serum-protein-binding property of lipid motifs and their tendency to accumulate in lymphoid tissue, we designed amphiphilic lipid-polymer conjugates that suppress systemic inflammation but provoke potent lymph-node immune activation. This work provides a rational basis for the design of lipid-polymer amphiphiles for optimized lymphoid targeting.
Collapse
Affiliation(s)
- Jana De Vrieze
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Benoit Louage
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Kim Deswarte
- Department of Internal Medicine and Pediatrics, Ghent University, VIB Center for Inflammation Research, Ghent, Belgium
| | - Zifu Zhong
- Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, Belgium
| | - Ruben De Coen
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Simon Van Herck
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | | | - Stefan Lienenklaus
- Institute for Laboratory Animal Science and Institute of Immunology, Hanover Medical School, Hannover, Germany
| | - Niek N Sanders
- Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, Belgium
| | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sunil A David
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
26
|
De Vrieze J, Louage B, Deswarte K, Zhong Z, De Coen R, Van Herck S, Nuhn L, Kaas Frich C, Zelikin AN, Lienenklaus S, Sanders NN, Lambrecht BN, David SA, De Geest BG. Amphiphile Polymer‐Lipidkonjugate zur potenten lymphatischen Anreicherung von TLR7/8‐Agonisten ermöglichen eine örtlich begrenzte Aktivierung des angeborenen Immunsystems. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201905687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jana De Vrieze
- Faculteit Farmaceutische Wetenschappen Universiteit Gent Ottergemsesteenweg 460 9000 Gent Belgien
| | - Benoit Louage
- Faculteit Farmaceutische Wetenschappen Universiteit Gent Ottergemsesteenweg 460 9000 Gent Belgien
| | - Kim Deswarte
- Department of Internal Medicine and Pediatrics, Ghent University VIB Center for Inflammation Research Technologiepark 71 9052 Gent Belgien
| | - Zifu Zhong
- Vakgroep Voeding, Genetica en Ethologie, Faculteit Diergeneeskunde Universiteit Gent Heidestraat 19 9820 Merelbeke Belgien
| | - Ruben De Coen
- Faculteit Farmaceutische Wetenschappen Universiteit Gent Ottergemsesteenweg 460 9000 Gent Belgien
| | - Simon Van Herck
- Faculteit Farmaceutische Wetenschappen Universiteit Gent Ottergemsesteenweg 460 9000 Gent Belgien
| | - Lutz Nuhn
- Max-Planck-Institut für Polymerforschung Ackermannweg 10 55128 Mainz Deutschland
| | - Camilla Kaas Frich
- Institut for Kemi Aarhus Universitet Langelandsgade 140 8000 Aarhus C Dänemark
| | | | - Stefan Lienenklaus
- Institut für Versuchstierkunde und Zentrales Tierlaboratorium Medizinische Hochschule Hannover Carl-Neuberg-Str.1 30625 Hannover Deutschland
| | - Niek N. Sanders
- Vakgroep Voeding, Genetica en Ethologie, Faculteit Diergeneeskunde Universiteit Gent Heidestraat 19 9820 Merelbeke Belgien
| | - Bart N. Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University VIB Center for Inflammation Research Technologiepark 71 9052 Gent Belgien
- Department of Pulmonary Medicine Erasmus University Medical Center Rotterdam Netherlands
| | - Sunil A. David
- Department of Medicinal Chemistry University of Minnesota 2231 Sixth Street SE Minneapolis MN 55455 USA
| | - Bruno G. De Geest
- Faculteit Farmaceutische Wetenschappen Universiteit Gent Ottergemsesteenweg 460 9000 Gent Belgien
| |
Collapse
|
27
|
Hoffmann PR, Hoffmann FW, Premeaux TA, Fujita T, Soprana E, Panigada M, Chew GM, Richard G, Hindocha P, Menor M, Khadka VS, Deng Y, Moise L, Ndhlovu LC, Siccardi A, Weinberg AD, De Groot AS, Bertino P. Multi-antigen Vaccination With Simultaneous Engagement of the OX40 Receptor Delays Malignant Mesothelioma Growth and Increases Survival in Animal Models. Front Oncol 2019; 9:720. [PMID: 31428586 PMCID: PMC6688537 DOI: 10.3389/fonc.2019.00720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 07/19/2019] [Indexed: 01/09/2023] Open
Abstract
Malignant Mesothelioma (MM) is a rare and highly aggressive cancer that develops from mesothelial cells lining the pleura and other internal cavities, and is often associated with asbestos exposure. To date, no effective treatments have been made available for this pathology. Herein, we propose a novel immunotherapeutic approach based on a unique vaccine targeting a series of antigens that we found expressed in different MM tumors, but largely undetectable in normal tissues. This vaccine, that we term p-Tvax, is comprised of a series of immunogenic peptides presented by both MHC-I and -II to generate robust immune responses. The peptides were designed using in silico algorithms that discriminate between highly immunogenic T cell epitopes and other harmful epitopes, such as suppressive regulatory T cell epitopes and autoimmune epitopes. Vaccination of mice with p-Tvax led to antigen-specific immune responses that involved both CD8+ and CD4+ T cells, which exhibited cytolytic activity against MM cells in vitro. In mice carrying MM tumors, p-Tvax increased tumor infiltration of CD4+ T cells. Moreover, combining p-Tvax with an OX40 agonist led to decreased tumor growth and increased survival. Mice treated with this combination immunotherapy displayed higher numbers of tumor-infiltrating CD8+ and CD4+ T cells and reduced T regulatory cells in tumors. Collectively, these data suggest that the combination of p-Tvax with an OX40 agonist could be an effective strategy for MM treatment.
Collapse
Affiliation(s)
- Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Fukun W Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Thomas A Premeaux
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Tsuyoshi Fujita
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Elisa Soprana
- Department of Molecular Immunology, San Raffaele University and Research Institute, Milan, Italy
| | - Maddalena Panigada
- Department of Molecular Immunology, San Raffaele University and Research Institute, Milan, Italy
| | - Glen M Chew
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | | | | | - Mark Menor
- Bioinformatics Core, Department of Complementary and Integrative Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Vedbar S Khadka
- Bioinformatics Core, Department of Complementary and Integrative Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Youping Deng
- Bioinformatics Core, Department of Complementary and Integrative Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Lenny Moise
- EpiVax, Inc., Providence, RI, United States.,Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Lishomwa C Ndhlovu
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| | - Antonio Siccardi
- Department of Molecular Immunology, San Raffaele University and Research Institute, Milan, Italy
| | - Andrew D Weinberg
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, United States
| | - Anne S De Groot
- EpiVax, Inc., Providence, RI, United States.,Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Pietro Bertino
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, United States
| |
Collapse
|
28
|
Yang G, Chen S, Zhang J. Bioinspired and Biomimetic Nanotherapies for the Treatment of Infectious Diseases. Front Pharmacol 2019; 10:751. [PMID: 31333467 PMCID: PMC6624236 DOI: 10.3389/fphar.2019.00751] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
There are still great challenges for the effective treatment of infectious diseases, although considerable achievement has been made by using antiviral and antimicrobial agents varying from small-molecule drugs, peptides/proteins, to nucleic acids. The nanomedicine approach is emerging as a new strategy capable of overcoming disadvantages of molecular therapeutics and amplifying their anti-infective activities, by localized delivery to infection sites, reducing off-target effects, and/or attenuating resistance development. Nanotechnology, in combination with bioinspired and biomimetic approaches, affords additional functions to nanoparticles derived from synthetic materials. Herein, we aim to provide a state-of-the-art review on recent progress in biomimetic and bioengineered nanotherapies for the treatment of infectious disease. Different biomimetic nanoparticles, derived from viruses, bacteria, and mammalian cells, are first described, with respect to their construction and biophysicochemical properties. Then, the applications of diverse biomimetic nanoparticles in anti-infective therapy are introduced, either by their intrinsic activity or by loading and site-specifically delivering various molecular drugs. Bioinspired and biomimetic nanovaccines for prevention and/or therapy of infectious diseases are also highlighted. At the end, major translation issues and future directions of this field are discussed.
Collapse
Affiliation(s)
- Guoyu Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|