1
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [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: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
2
|
Yuba E, Gupta RK. Preparation of glycopeptide-modified pH-sensitive liposomes for promoting antigen cross-presentation and induction of antigen-specific cellular immunity. Biomater Sci 2024; 12:1490-1501. [PMID: 38329387 DOI: 10.1039/d3bm01746j] [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: 02/09/2024]
Abstract
Cross-presentation, exogenous antigen presentation onto major histocompatibility complex class I molecules on antigen presenting cells, is crucially important for inducing antigen-specific cellular immune responses for cancer immunotherapy and for the treatment of infectious diseases. One strategy to induce cross-presentation is cytosolic delivery of an exogenous antigen using fusogenic or endosomolytic molecule-introduced nanocarriers. Earlier, we reported liposomes modified with pH-responsive polymers to achieve cytosolic delivery of an antigen. Polyglycidol-based or polysaccharide-based pH-responsive polymers can provide liposomes with delivery performance of antigenic proteins into cytosol via membrane fusion with endosomes responding to acidic pH, leading to induction of cross-presentation. Mannose residue was introduced to pH-responsive polysaccharides to increase uptake selectivity to antigen presenting cells and to improve cross-presentation efficiency. However, direct introduction of mannose residue into pH-responsive polysaccharides suppressed cytoplasmic delivery performance of liposomes. To avoid such interference, for this study, mannose-containing glycans were incorporated separately into pH-responsive polysaccharide-modified liposomes. Soybean agglutinin-derived glycopeptide was used as a ligand for lectins on antigen presenting cells. Incorporation of glycopeptide significantly increased the cellular uptake of liposomes by dendritic cell lines and increased cross-presentation efficiency. Liposomes incorporated both glycopeptide and pH-responsive polysaccharides exhibited strong adjuvant effects in vitro and induced the increase of dendritic cells, M1 macrophages, and effector T cells in the spleen. Subcutaneous administration of these liposomes induced antigen-specific cellular immunity, resulting in strong therapeutic effects in tumor-bearing mice. These results suggest that separate incorporation of glycopeptides and pH-responsive polysaccharides into antigen-loaded liposomes is an effective strategy to produce liposome-based nanovaccines to achieve antigen cross-presentation and induction of cellular immunity towards cancer immunotherapy.
Collapse
Affiliation(s)
- Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr D. Y. Patil Biotechnology and Bioinformatics Institute, Dr D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India.
| |
Collapse
|
3
|
Yanagihara S, Kitayama Y, Yuba E, Harada A. Preparing Size-Controlled Liposomes Modified with Polysaccharide Derivatives for pH-Responsive Drug Delivery Applications. Life (Basel) 2023; 13:2158. [PMID: 38004298 PMCID: PMC10672248 DOI: 10.3390/life13112158] [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: 10/09/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
The liposome particle size is an important parameter because it strongly affects content release from liposomes as a result of different bilayer curvatures and lipid packing. Earlier, we developed pH-responsive polysaccharide-derivative-modified liposomes that induced content release from the liposomes under weakly acidic conditions. However, the liposome used in previous studies size was adjusted to 100-200 nm. The liposome size effects on their pH-responsive properties were unclear. For this study, we controlled the polysaccharide-derivative-modified liposome size by extrusion through polycarbonate membranes having different pore sizes. The obtained liposomes exhibited different average diameters, in which the diameters mostly corresponded to the pore sizes of polycarbonate membranes used for extrusion. The amounts of polysaccharide derivatives per lipid were identical irrespective of the liposome size. Introduction of cholesterol within the liposomal lipid components suppressed the size increase in these liposomes for at least three weeks. These liposomes were stable at neutral pH, whereas the content release from liposomes was induced at weakly acidic pH. Smaller liposomes exhibited highly acidic pH-responsive content release compared with those from large liposomes. However, liposomes with 50 mol% cholesterol were not able to induce content release even under acidic conditions. These results suggest that control of the liposome size and cholesterol content is important for preparing stable liposomes at physiological conditions and for preparing highly pH-responsive liposomes for drug delivery applications.
Collapse
Affiliation(s)
- Shin Yanagihara
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan; (S.Y.); (Y.K.); (A.H.)
| | - Yukiya Kitayama
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan; (S.Y.); (Y.K.); (A.H.)
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan; (S.Y.); (Y.K.); (A.H.)
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Atsushi Harada
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan; (S.Y.); (Y.K.); (A.H.)
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| |
Collapse
|
4
|
Yuba E, Kado Y, Kasho N, Harada A. Cationic lipid potentiated the adjuvanticity of polysaccharide derivative-modified liposome vaccines. J Control Release 2023; 362:767-776. [PMID: 36244508 DOI: 10.1016/j.jconrel.2022.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022]
Abstract
Antigen carriers that can selectively deliver antigens to antigen presenting cells and which can simultaneously activate these cells (adjuvant property) are necessary for efficient cancer immunotherapy or vaccination. Delivery of a model antigen into dendritic cell cytosol has been achieved by pH-responsive polymer-modified liposomes via destabilization of endosomal membranes responding to acidic pH, which impelled antigen-specific cellular immunity. Furthermore, β-glucan-based pH-responsive polysaccharides have shown not only cytosolic antigen delivery performance but also adjuvant property, which further heightened cellular immune responses. Because pH-responsive polysaccharides have anionic carboxy groups, cationic lipid was introduced to liposomes in this study to improve the modification efficiency of pH-responsive polysaccharides and to improve their adjuvanticity and immunity-inducing functions. Introduction of cationic lipids increased the amounts of polysaccharide derivatives on the liposome and increased the cellular association of the liposomes to dendritic cells. Liposomes containing β-glucan-based pH-responsive polysaccharides and cationic lipids increased cytokine production from dendritic cells much more than other polysaccharide derivatives did. Furthermore, through improvement of intra-tumoral immunosuppression and induction of antigen-specific cellular immunity, administering these liposomes impelled tumor suppression even with a small antigen dose. These results suggest that introducing cationic lipids and using pH-responsive polysaccharides having intrinsically adjuvant function are effective for producing liposomal nanovaccines showing strong immunity-inducing function.
Collapse
Affiliation(s)
- Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan; Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Yuna Kado
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan
| | - Nozomi Kasho
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan
| | - Atsushi Harada
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan; Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| |
Collapse
|
5
|
Hao Y, Ji Z, Zhou H, Wu D, Gu Z, Wang D, ten Dijke P. Lipid-based nanoparticles as drug delivery systems for cancer immunotherapy. MedComm (Beijing) 2023; 4:e339. [PMID: 37560754 PMCID: PMC10407046 DOI: 10.1002/mco2.339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 08/11/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have shown remarkable success in cancer treatment. However, in cancer patients without sufficient antitumor immunity, numerous data indicate that blocking the negative signals elicited by immune checkpoints is ineffective. Drugs that stimulate immune activation-related pathways are emerging as another route for improving immunotherapy. In addition, the development of nanotechnology presents a promising platform for tissue and cell type-specific delivery and improved uptake of immunomodulatory agents, ultimately leading to enhanced cancer immunotherapy and reduced side effects. In this review, we summarize and discuss the latest developments in nanoparticles (NPs) for cancer immuno-oncology therapy with a focus on lipid-based NPs (lipid-NPs), including the characteristics and advantages of various types. Using the agonists targeting stimulation of the interferon genes (STING) transmembrane protein as an exemplar, we review the potential of various lipid-NPs to augment STING agonist therapy. Furthermore, we present recent findings and underlying mechanisms on how STING pathway activation fosters antitumor immunity and regulates the tumor microenvironment and provide a summary of the distinct STING agonists in preclinical studies and clinical trials. Ultimately, we conduct a critical assessment of the obstacles and future directions in the utilization of lipid-NPs to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Yang Hao
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
- Department of Basic MedicineChangzhi Medical CollegeChangzhiChina
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Zhonghao Ji
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
- Department of Basic MedicineChangzhi Medical CollegeChangzhiChina
| | - Hengzong Zhou
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
| | - Dongrun Wu
- Departure of Philosophy, Faculty of HumanitiesLeiden UniversityLeidenThe Netherlands
| | - Zili Gu
- Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Dongxu Wang
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
6
|
Li Q, Liu X, Yan C, Zhao B, Zhao Y, Yang L, Shi M, Yu H, Li X, Luo K. Polysaccharide-Based Stimulus-Responsive Nanomedicines for Combination Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206211. [PMID: 36890780 DOI: 10.1002/smll.202206211] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/09/2023] [Indexed: 06/08/2023]
Abstract
Cancer immunotherapy is a promising antitumor approach, whereas nontherapeutic side effects, tumor microenvironment (TME) intricacy, and low tumor immunogenicity limit its therapeutic efficacy. In recent years, combination immunotherapy with other therapies has been proven to considerably increase antitumor efficacy. However, achieving codelivery of the drugs to the tumor site remains a major challenge. Stimulus-responsive nanodelivery systems show controlled drug delivery and precise drug release. Polysaccharides, a family of potential biomaterials, are widely used in the development of stimulus-responsive nanomedicines due to their unique physicochemical properties, biocompatibility, and modifiability. Here, the antitumor activity of polysaccharides and several combined immunotherapy strategies (e.g., immunotherapy combined with chemotherapy, photodynamic therapy, or photothermal therapy) are summarized. More importantly, the recent progress of polysaccharide-based stimulus-responsive nanomedicines for combination cancer immunotherapy is discussed, with the focus on construction of nanomedicine, targeted delivery, drug release, and enhanced antitumor effects. Finally, the limitations and application prospects of this new field are discussed.
Collapse
Affiliation(s)
- Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Chunmei Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Bolin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yuxin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Lu Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Mingyi Shi
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hua Yu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao SAR, 999078, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Kaipei Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| |
Collapse
|
7
|
Makandar AI, Jain M, Yuba E, Sethi G, Gupta RK. Canvassing Prospects of Glyco-Nanovaccines for Developing Cross-Presentation Mediated Anti-Tumor Immunotherapy. Vaccines (Basel) 2022; 10:vaccines10122049. [PMID: 36560459 PMCID: PMC9784904 DOI: 10.3390/vaccines10122049] [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: 09/28/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
In view of the severe downsides of conventional cancer therapies, the quest of developing alternative strategies still remains of critical importance. In this regard, antigen cross-presentation, usually employed by dendritic cells (DCs), has been recognized as a potential solution to overcome the present impasse in anti-cancer therapeutic strategies. It has been established that an elevated cytotoxic T lymphocyte (CTL) response against cancer cells can be achieved by targeting receptors expressed on DCs with specific ligands. Glycans are known to serve as ligands for C-type lectin receptors (CLRs) expressed on DCs, and are also known to act as a tumor-associated antigen (TAA), and, thus, can be harnessed as a potential immunotherapeutic target. In this scenario, integrating the knowledge of cross-presentation and glycan-conjugated nanovaccines can help us to develop so called 'glyco-nanovaccines' (GNVs) for targeting DCs. Here, we briefly review and analyze the potential of GNVs as the next-generation anti-tumor immunotherapy. We have compared different antigen-presenting cells (APCs) for their ability to cross-present antigens and described the potential nanocarriers for tumor antigen cross-presentation. Further, we discuss the role of glycans in targeting of DCs, the immune response due to pathogens, and imitative approaches, along with parameters, strategies, and challenges involved in cross-presentation-based GNVs for cancer immunotherapy. It is known that the effectiveness of GNVs in eradicating tumors by inducing strong CTL response in the tumor microenvironment (TME) has been largely hindered by tumor glycosylation and the expression of different lectin receptors (such as galectins) by cancer cells. Tumor glycan signatures can be sensed by a variety of lectins expressed on immune cells and mediate the immune suppression which, in turn, facilitates immune evasion. Therefore, a sound understanding of the glycan language of cancer cells, and glycan-lectin interaction between the cancer cells and immune cells, would help in strategically designing the next-generation GNVs for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Amina I. Makandar
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Mannat Jain
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| |
Collapse
|
8
|
Diep YN, Kim TJ, Cho H, Lee LP. Nanomedicine for advanced cancer immunotherapy. J Control Release 2022; 351:1017-1037. [DOI: 10.1016/j.jconrel.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/09/2022]
|
9
|
Mukherjee A, Bisht B, Dutta S, Paul MK. Current advances in the use of exosomes, liposomes, and bioengineered hybrid nanovesicles in cancer detection and therapy. Acta Pharmacol Sin 2022; 43:2759-2776. [PMID: 35379933 PMCID: PMC9622806 DOI: 10.1038/s41401-022-00902-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/15/2022] [Indexed: 12/17/2022] Open
Abstract
Three major approaches of cancer therapy can be enunciated as delivery of biotherapeutics, tumor image analysis, and immunotherapy. Liposomes, artificial fat bubbles, are long known for their capacity to encapsulate a diverse range of bioactive molecules and release the payload in a sustained, stimuli-responsive manner. They have already been widely explored as a delivery vehicle for therapeutic drugs as well as imaging agents. They are also extensively being used in cancer immunotherapy. On the other hand, exosomes are naturally occurring nanosized extracellular vesicles that serve an important role in cell-cell communication. Importantly, the exosomes also have proven their capability to carry an array of active pharmaceuticals and diagnostic molecules to the tumor cells. Exosomes, being enriched with tumor antigens, have numerous immunomodulatory effects. Much to our intrigue, in recent times, efforts have been directed toward developing smart, bioengineered, exosome-liposome hybrid nanovesicles, which are augmented by the benefits of both vesicular systems. This review attempts to summarize the contemporary developments in the use of exosome and liposome toward cancer diagnosis, therapy, as a vehicle for drug delivery, diagnostic carrier for tumor imaging, and cancer immunotherapy. We shall also briefly reflect upon the recent advancements of the exosome-liposome hybrids in cancer therapy. Finally, we put forward future directions for the use of exosome/liposome and/or hybrid nanocarriers for accurate diagnosis and personalized therapies for cancers.
Collapse
Affiliation(s)
| | - Bharti Bisht
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Suman Dutta
- International Institute of Innovation and Technology, New Town, Kolkata, 700156, India
| | - Manash K Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
10
|
Das A, Ringu T, Ghosh S, Pramanik N. A comprehensive review on recent advances in preparation, physicochemical characterization, and bioengineering applications of biopolymers. Polym Bull (Berl) 2022; 80:7247-7312. [PMID: 36043186 PMCID: PMC9409625 DOI: 10.1007/s00289-022-04443-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/20/2022] [Accepted: 08/15/2022] [Indexed: 12/01/2022]
Abstract
Biopolymers are mainly the polymers which are created or obtained from living creatures such as plants and bacteria rather than petroleum, which has traditionally been the source of polymers. Biopolymers are chain-like molecules composed of repeated chemical blocks derived from renewable resources that may decay in the environment. The usage of biomaterials is becoming more popular as a means of reducing the use of non-renewable resources and reducing environmental pollution produced by synthetic materials. Biopolymers' biodegradability and non-toxic nature help to maintain our environment clean and safe. This study discusses how to improve the mechanical and physical characteristics of biopolymers, particularly in the realm of bioengineering. The paper begins with a fundamental introduction and progresses to a detailed examination of synthesis and a unique investigation of several recent focused biopolymers with mechanical, physical, and biological characterization. Biopolymers' unique non-toxicity, biodegradability, biocompatibility, and eco-friendly features are boosting their applications, especially in bioengineering fields, including agriculture, pharmaceuticals, biomedical, ecological, industrial, aqua treatment, and food packaging, among others, at the end of this paper. The purpose of this paper is to provide an overview of the relevance of biopolymers in smart and novel bioengineering applications. Graphical abstract The Graphical abstract represents the biological sources and applications of biopolymers. Plants, bacteria, animals, agriculture wastes, and fossils are all biological sources for biopolymers, which are chemically manufactured from biological monomer units, including sugars, amino acids, natural fats and oils, and nucleotides. Biopolymer modification (chemical or physical) is recognized as a crucial technique for modifying physical and chemical characteristics, resulting in novel materials with improved capabilities and allowing them to be explored to their full potential in many fields of application such as tissue engineering, drug delivery, agriculture, biomedical, food industries, and industrial applications.
Collapse
Affiliation(s)
- Abinash Das
- Department of Chemistry, National Institute of Technology, Arunachal Pradesh, Jote, Arunachal Pradesh 791113 India
| | - Togam Ringu
- Department of Chemistry, National Institute of Technology, Arunachal Pradesh, Jote, Arunachal Pradesh 791113 India
| | - Sampad Ghosh
- Department of Chemistry, Nalanda College of Engineering, Nalanda, Bihar 803108 India
| | - Nabakumar Pramanik
- Department of Chemistry, National Institute of Technology, Arunachal Pradesh, Jote, Arunachal Pradesh 791113 India
| |
Collapse
|
11
|
Tang J, Yang Y, Qu J, Ban W, Song H, Gu Z, Yang Y, Cai L, Theivendran S, Wang Y, Zhang M, Yu C. Mesoporous sodium four-coordinate aluminosilicate nanoparticles modulate dendritic cell pyroptosis and activate innate and adaptive immunity. Chem Sci 2022; 13:8507-8517. [PMID: 35974763 PMCID: PMC9337734 DOI: 10.1039/d1sc05319a] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 06/20/2022] [Indexed: 11/23/2022] Open
Abstract
Pyroptosis is a programmed cell death widely studied in cancer cells for tumour inhibition, but rarely in dendritic cell (DC) activation for vaccine development. Here, we report the synthesis of sodium stabilized mesoporous aluminosilicate nanoparticles as DC pyroptosis modulators and antigen carriers. By surface modification of sodium-stabilized four-coordinate aluminium species on dendritic mesoporous silica nanoparticles, the resultant Na-IVAl-DMSN significantly activated DC through caspase-1 dependent pyroptosis via pH responsive intracellular ion exchange. The released proinflammatory cellular contents further mediated DC hyperactivation with prolonged cytokine release. In vivo studies showed that Na-IVAl-DMSN induced enhanced cellular immunity mediated by natural killer (NK) cells, cytotoxic T cells, and memory T cells as well as humoral immune response. Our results provide a new principle for the design of next-generation nanoadjuvants for vaccine applications.
Collapse
Affiliation(s)
- Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Yang Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Jingjing Qu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Wenhuang Ban
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Zhengying Gu
- School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Larry Cai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Shevanuja Theivendran
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
| | - Min Zhang
- School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland St Lucia Brisbane QLD 4072 Australia
- School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| |
Collapse
|
12
|
A comprehensive review on different approaches for tumor targeting using nanocarriers and recent developments with special focus on multifunctional approaches. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
13
|
Nadukkandy AS, Ganjoo E, Singh A, Dinesh Kumar L. Tracing New Landscapes in the Arena of Nanoparticle-Based Cancer Immunotherapy. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.911063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Over the past two decades, unique and comprehensive cancer treatment has ushered new hope in the holistic management of the disease. Cancer immunotherapy, which harnesses the immune system of the patient to attack the cancer cells in a targeted manner, scores over others by being less debilitating compared to the existing treatment strategies. Significant advancements in the knowledge of immune surveillance in the last few decades have led to the development of several types of immune therapy like monoclonal antibodies, cancer vaccines, immune checkpoint inhibitors, T-cell transfer therapy or adoptive cell therapy (ACT) and immune system modulators. Intensive research has established cancer immunotherapy to be a safe and effective method for improving survival and the quality of a patient’s life. However, numerous issues with respect to site-specific delivery, resistance to immunotherapy, and escape of cancer cells from immune responses, need to be addressed for expanding and utilizing this therapy as a regular mode in the clinical treatment. Development in the field of nanotechnology has augmented the therapeutic efficiency of treatment modalities of immunotherapy. Nanocarriers could be used as vehicles because of their advantages such as increased surface areas, targeted delivery, controlled surface and release chemistry, enhanced permeation and retention effect, etc. They could enhance the function of immune cells by incorporating immunomodulatory agents that influence the tumor microenvironment, thus enabling antitumor immunity. Robust validation of the combined effect of nanotechnology and immunotherapy techniques in the clinics has paved the way for a better treatment option for cancer than the already existing procedures such as chemotherapy and radiotherapy. In this review, we discuss the current applications of nanoparticles in the development of ‘smart’ cancer immunotherapeutic agents like ACT, cancer vaccines, monoclonal antibodies, their site-specific delivery, and modulation of other endogenous immune cells. We also highlight the immense possibilities of using nanotechnology to accomplish leveraging the coordinated and adaptive immune system of a patient to tackle the complexity of treating unique disease conditions and provide future prospects in the field of cancer immunotherapy.
Collapse
|
14
|
Advancement of cancer immunotherapy using nanoparticles-based nanomedicine. Semin Cancer Biol 2022; 86:624-644. [DOI: 10.1016/j.semcancer.2022.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022]
|
15
|
Polysaccharide hydrogels: Functionalization, construction and served as scaffold for tissue engineering. Carbohydr Polym 2022; 278:118952. [PMID: 34973769 DOI: 10.1016/j.carbpol.2021.118952] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/07/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023]
Abstract
Polysaccharide hydrogels have been widely utilized in tissue engineering. They interact with the organismal environments, modulating the cargos release and realizing of long-term survival and activations of living cells. In this review, the potential strategies for modification of polysaccharides were introduced firstly. It is not only used to functionalize the polysaccharides for the consequent formation of hydrogels, but also used to introduce versatile side groups for the regulation of cell behavior. Then, techniques and underlying mechanisms in inducing the formation of hydrogels by polysaccharides or their derivatives are briefly summarized. Finally, the applications of polysaccharide hydrogels in vivo, mainly focus on the performance for alleviation of foreign-body response (FBR) and as cell scaffolds for tissue regeneration, are exemplified. In addition, the perspectives and challenges for further research are addressed. It aims to provide a comprehensive framework about the potentials and challenges that the polysaccharide hydrogels confronting in tissue engineering.
Collapse
|
16
|
Volovat SR, Ursulescu CL, Moisii LG, Volovat C, Boboc D, Scripcariu D, Amurariti F, Stefanescu C, Stolniceanu CR, Agop M, Lungulescu C, Volovat CC. The Landscape of Nanovectors for Modulation in Cancer Immunotherapy. Pharmaceutics 2022; 14:397. [PMID: 35214129 PMCID: PMC8875018 DOI: 10.3390/pharmaceutics14020397] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy represents a promising strategy for the treatment of cancer, which functions via the reprogramming and activation of antitumor immunity. However, adverse events resulting from immunotherapy that are related to the low specificity of tumor cell-targeting represent a limitation of immunotherapy's efficacy. The potential of nanotechnologies is represented by the possibilities of immunotherapeutical agents being carried by nanoparticles with various material types, shapes, sizes, coated ligands, associated loading methods, hydrophilicities, elasticities, and biocompatibilities. In this review, the principal types of nanovectors (nanopharmaceutics and bioinspired nanoparticles) are summarized along with the shortcomings in nanoparticle delivery and the main factors that modulate efficacy (the EPR effect, protein coronas, and microbiota). The mechanisms by which nanovectors can target cancer cells, the tumor immune microenvironment (TIME), and the peripheral immune system are also presented. A possible mathematical model for the cellular communication mechanisms related to exosomes as nanocarriers is proposed.
Collapse
Affiliation(s)
- Simona-Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
| | - Corina Lupascu Ursulescu
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.L.U.); (L.G.M.); (C.C.V.)
| | - Liliana Gheorghe Moisii
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.L.U.); (L.G.M.); (C.C.V.)
| | - Constantin Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
- Department of Medical Oncology, “Euroclinic” Center of Oncology, 2 Vasile Conta Str., 700106 Iaşi, Romania
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
| | - Dragos Scripcariu
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania;
| | - Florin Amurariti
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.S.); (C.R.S.)
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.S.); (C.R.S.)
| | - Maricel Agop
- Physics Department, “Gheorghe Asachi” Technical University, Prof. Dr. Docent Dimitrie Mangeron Rd., No. 59A, 700050 Iaşi, Romania;
| | - Cristian Lungulescu
- Department of Medical Oncology, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Cristian Constantin Volovat
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.L.U.); (L.G.M.); (C.C.V.)
| |
Collapse
|
17
|
Yang Y, Guo T, Xu J, Xiong Y, Cui X, Ke Y, Wang C. Micelle nanovehicles for co-delivery of Lepidium meyenii Walp. (maca) polysaccharide and chloroquine to tumor-associated macrophages for synergistic cancer immunotherapy. Int J Biol Macromol 2021; 189:577-589. [PMID: 34450149 DOI: 10.1016/j.ijbiomac.2021.08.155] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/18/2023]
Abstract
Here, we fabricated amphiphilic polysaccharide micelles for synergistic cancer immunotherapy targeting tumor-associated macrophages (TAMs). Lepidium meyenii Walp. (maca) polysaccharide (MP), a naturally derived macromolecule with a strong TAM-remodeling effect, was grafted on a hydrophobic poly(lactic-co-glycolic acid) (PLGA) segment, with a disulfide bond for redox-sensitive linkage. The amphiphilic polysaccharide derivatives could self-assemble into core (PLGA)-shell (MP)-structured micelles and encapsulate chloroquine (CQ) into the hydrophobic core. By using a 4T1-M2 macrophage co-culture model and a 4T1 tumor xenograft mouse model, we showed that the prepared micelles could co-deliver MP and CQ to the tumor sites and selectively accumulate at TAMs because of the specific properties of MP. Furthermore, the nanoparticles exerted synergistic tumor immunotherapeutic and antimetastatic effects, which might be attributable to the enhanced cell internalization of the micelles and the multiple regulatory mechanisms of MP and CQ. Thus, immunomodulatory MP may be a promising biomaterial for cancer immunotherapy.
Collapse
Affiliation(s)
- Ye Yang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Tingting Guo
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Junwei Xu
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Yin Xiong
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Xiuming Cui
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Yang Ke
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Kunming Medical University, Kunming 650500, China.
| | - Chengxiao Wang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China.
| |
Collapse
|
18
|
Yanagihara S, Kasho N, Sasaki K, Shironaka N, Kitayama Y, Yuba E, Harada A. pH-Sensitive branched β-glucan-modified liposomes for activation of antigen presenting cells and induction of antitumor immunity. J Mater Chem B 2021; 9:7713-7724. [PMID: 34545900 DOI: 10.1039/d1tb00786f] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Induction of cellular immunity is important for effective cancer immunotherapy. Although various antigen carriers for cancer immunotherapy have been developed to date, balancing efficient antigen delivery to antigen presenting cells (APCs) and their activation via innate immune receptors, both of which are crucially important for the induction of strong cellular immunity, remains challenging. For this study, branched β-glucan was selected as an intrinsically immunity-stimulating and biocompatible material. It was engineered to develop multifunctional liposomal cancer vaccines capable of efficient interactions with APCs and subsequent activation of the cells. Hydroxy groups of branched β-glucan (Aquaβ) were modified with 3-methylglutaric acid ester and decyl groups, respectively, to provide pH-sensitivity and anchoring capability to the liposomal membrane. The modification efficiency of Aquaβ derivatives to the liposomes was significantly high compared with linear β-glucan (curdlan) derivatives. Aquaβ derivative-modified liposomes released their contents in response to weakly acidic pH. As a model antigenic protein, ovalbumin (OVA)-loaded liposomes modified with Aquaβ derivatives interacted efficiently with dendritic cells, and induced inflammatory cytokine secretion from the cells. Subcutaneous administration of Aquaβ derivative-modified liposomes suppressed the growth of the E.G7-OVA tumor significantly compared with curdlan derivative-modified liposomes. Aquaβ derivative-modified liposomes induced the increase of CD8+ T cells, and polarized macrophages to the antitumor M1-phenotype within the tumor microenvironment. Therefore, pH-sensitive Aquaβ derivatives can be promising materials for liposomal antigen delivery systems to induce antitumor immune responses efficiently.
Collapse
Affiliation(s)
- Shin Yanagihara
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Nozomi Kasho
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Koichi Sasaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Naoto Shironaka
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Yukiya Kitayama
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Atsushi Harada
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| |
Collapse
|
19
|
Hyun J, Eom J, Song J, Seo I, Um SH, Park KM, Bhang SH. Poly(amino ester)-Based Polymers for Gene and Drug Delivery Systems and Further Application toward Cell Culture System. Macromol Biosci 2021; 21:e2100106. [PMID: 34117832 DOI: 10.1002/mabi.202100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/20/2021] [Indexed: 11/10/2022]
Abstract
Various synthetic polymers based on poly(amino ester) (PAE) are suggested as candidates for gene and drug delivery owing to their pH-responsiveness, which contributes to efficient delivery performance. PAE-based pH-responsive polymers are more biodegradable and hydrophilic than other types of pH-responsive polymers. The functionality of PAE-based polymers can be reinforced by using different chemical modifications to improve the efficiency of gene and drug delivery. Additionally, PAE-based polymers are used in many ways in the biomedical field, such as in transdermal delivery and stem cell culture systems. Here, the recent novel PAE-based polymers designed for gene and drug delivery systems along with their further applications toward adult stem cell culture systems are reviewed. The synthetic tactics are contemplated and pros and cons of each type of polymer are analyzed, and detailed examples of the different types are analyzed.
Collapse
Affiliation(s)
- Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jiin Eom
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihun Song
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Inwoo Seo
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyung Min Park
- Division of Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
20
|
Volovat SR, Negru S, Stolniceanu CR, Volovat C, Lungulescu C, Scripcariu D, Cobzeanu BM, Stefanescu C, Grigorescu C, Augustin I, Lupascu Ursulescu C, Volovat CC. Nanomedicine to modulate immunotherapy in cutaneous melanoma (Review). Exp Ther Med 2021; 21:535. [PMID: 33815608 PMCID: PMC8014970 DOI: 10.3892/etm.2021.9967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy has shifted the paradigm in cancer treatment in recent years. Immune checkpoint blockage (ICB), the active cancer vaccination and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer represent the main developments, achieving a surprising increased survival in patients included in clinical trials. In spite of these results, the current state-of-the-art immunotherapy has its limitations in efficacy. The existence of an interdisciplinary interface involving current knowledge in biology, immunology, bioengineering and materials science represents important progress in increasing the effectiveness of immunotherapy in cancer. Cutaneous melanoma remains a difficult cancer to treat, in which immunotherapy is a major therapeutic option. In fact, enhancing immunotherapy is possible using sophisticated biomedical nanotechnology platforms of organic or inorganic materials or engineering various immune cells to enhance the immune system. In addition, biological devices have developed, changing the approach to and treatment results in melanoma. In this review, we present different modalities to modulate the immune system, as well as opportunities and challenges in melanoma treatment.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Serban Negru
- Department of Medical Oncology, 'Victor Babes' University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Constantin Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Cristian Lungulescu
- Department of Medical Oncology, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | - Dragos Scripcariu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Mihail Cobzeanu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristina Grigorescu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Corina Lupascu Ursulescu
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristian Constantin Volovat
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Radiology, 'Sf. Spiridon' Emergency Clinic Hospital, 700111 Iasi, Romania
| |
Collapse
|
21
|
Kong X, Cheng R, Wang J, Fang Y, Hwang KC. Nanomedicines inhibiting tumor metastasis and recurrence and their clinical applications. NANO TODAY 2021; 36:101004. [DOI: 10.1016/j.nantod.2020.101004] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
22
|
Masuelli L, Benvenuto M, Focaccetti C, Ciuffa S, Fazi S, Bei A, Miele MT, Piredda L, Manzari V, Modesti A, Bei R. Targeting the tumor immune microenvironment with "nutraceuticals": From bench to clinical trials. Pharmacol Ther 2020; 219:107700. [PMID: 33045254 DOI: 10.1016/j.pharmthera.2020.107700] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2020] [Indexed: 02/06/2023]
Abstract
The occurrence of immune effector cells in the tissue microenvironment during neoplastic progression is critical in determining tumor growth outcomes. On the other hand, tumors may also avoid immune system-mediated elimination by recruiting immunosuppressive leukocytes and soluble factors, which coordinate a tumor microenvironment that counteracts the efficiency of the antitumor immune response. Checkpoint inhibitor therapy results have indicated a way forward via activation of the immune system against cancer. Widespread evidence has shown that different compounds in foods, when administered as purified substances, can act as immunomodulators in humans and animals. Although there is no universally accepted definition of nutraceuticals, the term identifies a wide category of natural compounds that may impact health and disease statuses and includes purified substances from natural sources, plant extracts, dietary supplements, vitamins, phytonutrients, and various products with combinations of functional ingredients. In this review, we summarize the current knowledge on the immunomodulatory effects of nutraceuticals with a special focus on the cancer microenvironment, highlighting the conceptual benefits or drawbacks and subtle cell-specific effects of nutraceuticals for envisioning future therapies employing nutraceuticals as chemoadjuvants.
Collapse
Affiliation(s)
- Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Monica Benvenuto
- Saint Camillus International University of Health and Medical Sciences, via di Sant'Alessandro 8, 00131 Rome, Italy; Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; Department of Human Science and Promotion of the Quality of Life, San Raffaele University Rome, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Sara Ciuffa
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Sara Fazi
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Arianna Bei
- Medical School, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Lucia Piredda
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; CIMER, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
23
|
Yuba E, Fukaya Y, Yanagihara S, Kasho N, Harada A. Development of Mannose-Modified Carboxylated Curdlan-Coated Liposomes for Antigen Presenting Cell Targeted Antigen Delivery. Pharmaceutics 2020; 12:pharmaceutics12080754. [PMID: 32796567 PMCID: PMC7465930 DOI: 10.3390/pharmaceutics12080754] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 02/03/2023] Open
Abstract
Specific delivery to antigen presenting cells (APC) and precise control of the intracellular fate of antigens are crucial to induce cellular immunity that directly and specifically attacks cancer cells. We previously achieved cytoplasmic delivery of antigen and activation of APC using carboxylated curdlan-modified liposomes, which led to the induction of cellular immunity in vivo. APCs express mannose receptors on their surface to recognize pathogen specifically and promote cross-presentation of antigen. In this study, mannose-residue was additionally introduced to carboxylated curdlan as a targeting moiety to APC for further improvement of polysaccharide-based antigen carriers. Mannose-modified curdlan derivatives were synthesized by the condensation between amino group-introduced mannose and carboxy group in pH-sensitive curdlan. Mannose residue-introduced carboxylated curdlan-modified liposomes showed higher pH-sensitivity than that of liposomes modified with conventional carboxylated curdlan. The introduction of mannose-residue to the liposomes induced aggregation in the presence of Concanavalin A, indicating that mannose residues were presented onto liposome surface. Mannose residue-introduced carboxylated curdlan-modified liposomes exhibited high and selective cellular association to APC. Furthermore, mannose residue-introduced carboxylated curdlan-modified liposomes promoted cross-presentation of antigen and induced strong antitumor effects on tumor-bearing mice. Therefore, these liposomes are promising as APC-specific antigen delivery systems for the induction of antigen-specific cellular immunity.
Collapse
Affiliation(s)
- Eiji Yuba
- Correspondence: (E.Y.); (A.H.); Tel.: +81-72-254-9330 (E.Y.); Fax: +81-72-254-9330 (E.Y.)
| | | | | | | | - Atsushi Harada
- Correspondence: (E.Y.); (A.H.); Tel.: +81-72-254-9330 (E.Y.); Fax: +81-72-254-9330 (E.Y.)
| |
Collapse
|
24
|
Kim K, Khang D. Past, Present, and Future of Anticancer Nanomedicine. Int J Nanomedicine 2020; 15:5719-5743. [PMID: 32821098 PMCID: PMC7418170 DOI: 10.2147/ijn.s254774] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
This review aims to summarize the methods that have been used till today, highlight methods that are currently being developed, and predict the future roadmap for anticancer therapy. In the beginning of this review, established approaches for anticancer therapy, such as conventional chemotherapy, hormonal therapy, monoclonal antibodies, and tyrosine kinase inhibitors are summarized. To counteract the side effects of conventional chemotherapy and to increase limited anticancer efficacy, nanodrug- and stem cell-based therapies have been introduced. However, current level of understanding and strategies of nanodrug and stem cell-based therapies have limitations that make them inadequate for clinical application. Subsequently, this manuscript reviews methods with fewer side effects compared to those of the methods mentioned above which are currently being investigated and are already being applied in the clinic. The newer strategies that are already being clinically applied include cancer immunotherapy, especially T cell-mediated therapy and immune checkpoint inhibitors, and strategies that are gaining attention include the manipulation of the tumor microenvironment or the activation of dendritic cells. Tumor-associated macrophage repolarization is another potential strategy for cancer immunotherapy, a method which activates macrophages to immunologically attack malignant cells. At the end of this review, we discuss combination therapies, which are the future of cancer treatment. Nanoparticle-based anticancer immunotherapies seem to be effective, in that they effectively use nanodrugs to elicit a greater immune response. The combination of these therapies with others, such as photothermal or tumor vaccine therapy, can result in a greater anticancer effect. Thus, the future of anticancer therapy aims to increase the effectiveness of therapy using various therapies in a synergistic combination rather than individually.
Collapse
Affiliation(s)
- Kyungeun Kim
- College of Medicine, Gachon University, Incheon 21999, South Korea
| | - Dongwoo Khang
- College of Medicine, Gachon University, Incheon 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea.,Gachon Advanced Institute for Health Science & Technology (GAIHST), Gachon University, Incheon 21999, South Korea.,Department of Physiology, School of Medicine, Gachon University, Incheon 21999, South Korea
| |
Collapse
|
25
|
Hu F, Yue H, Lu T, Ma G. Cytosolic delivery of HBsAg and enhanced cellular immunity by pH-responsive liposome. J Control Release 2020; 324:460-470. [DOI: 10.1016/j.jconrel.2020.05.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 01/10/2023]
|
26
|
Engineering nanoparticulate vaccines for enhancing antigen cross-presentation. Curr Opin Biotechnol 2020; 66:113-122. [PMID: 32745889 DOI: 10.1016/j.copbio.2020.06.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 12/13/2022]
Abstract
Efficient cross-presentation is pivotal for vaccination against cancer and infection by intracellular virus and bacteria. Recently, various types of nanoparticle vaccines have been developed and investigated for efficiently and specifically improving cross-presentation and CD8+ T cell priming. In this review, we will summarize the known intracellular pathways involved in cross-presentation, and focus on several nanoparticle strategies that have been reported for enhancing cross-presentation, including designing multifunctional nano-vaccines for increasing endosomal escape, designing nano-vaccines that can target lymph nodes to improve antigen uptake by lymph node resident CD8α+ dendritic cells, and co-delivering immune modulators for upregulating cross-presentation related intracellular components. We will also briefly discuss the future prospects of cross-presentation based nano-vaccine strategy for curing diseases.
Collapse
|
27
|
Yang F, Shi K, Jia YP, Hao Y, Peng JR, Qian ZY. Advanced biomaterials for cancer immunotherapy. Acta Pharmacol Sin 2020; 41:911-927. [PMID: 32123302 PMCID: PMC7468530 DOI: 10.1038/s41401-020-0372-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/27/2020] [Indexed: 02/05/2023] Open
Abstract
Immunotherapy, as a powerful strategy for cancer treatment, has achieved tremendous efficacy in clinical trials. Despite these advancements, there is much to do in terms of enhancing therapeutic benefits and decreasing the side effects of cancer immunotherapy. Advanced nanobiomaterials, including liposomes, polymers, and silica, play a vital role in the codelivery of drugs and immunomodulators. These nanobiomaterial-based delivery systems could effectively promote antitumor immune responses and simultaneously reduce toxic adverse effects. Furthermore, nanobiomaterials may also combine with each other or with traditional drugs via different mechanisms, thus giving rise to more accurate and efficient tumor treatment. Here, an overview of the latest advancement in these nanobiomaterials used for cancer immunotherapy is given, describing outstanding systems, including lipid-based nanoparticles, polymer-based scaffolds or micelles, inorganic nanosystems, and others.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Yan-Peng Jia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Ying Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Jin-Rong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zhi-Yong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
Nishiguchi A, Taguchi T. Sustained‐immunostimulatory nanocellulose scaffold to enhance vaccine efficacy. J Biomed Mater Res A 2020; 108:1159-1170. [DOI: 10.1002/jbm.a.36890] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 01/28/2023]
Affiliation(s)
- Akihiro Nishiguchi
- Biomaterials Field, Research Center for Functional MaterialsNational Institute for Materials Science Tsukuba Japan
| | - Tetsushi Taguchi
- Biomaterials Field, Research Center for Functional MaterialsNational Institute for Materials Science Tsukuba Japan
| |
Collapse
|
29
|
Abstract
Vaccines are powerful tools that can activate the immune system for protection against various diseases. As carbohydrates can play important roles in immune recognition, they have been widely applied in vaccine development. Carbohydrate antigens have been investigated in vaccines against various pathogenic microbes and cancer. Polysaccharides such as dextran and β-glucan can serve as smart vaccine carriers for efficient antigen delivery to immune cells. Some glycolipids, such as galactosylceramide and monophosphoryl lipid A, are strong immune stimulators, which have been studied as vaccine adjuvants. In this review, we focus on the current advances in applying carbohydrates as vaccine delivery carriers and adjuvants. We will discuss the examples that involve chemical modifications of the carbohydrates for effective antigen delivery, as well as covalent antigen-carbohydrate conjugates for enhanced immune responses.
Collapse
Affiliation(s)
- Shuyao Lang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
30
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
31
|
Plant lectins and their usage in preparing targeted nanovaccines for cancer immunotherapy. Semin Cancer Biol 2020; 80:87-106. [PMID: 32068087 DOI: 10.1016/j.semcancer.2020.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/30/2020] [Accepted: 02/06/2020] [Indexed: 01/06/2023]
Abstract
Plant lectins, a natural source of glycans with a therapeutic potential may lead to the discovery of new targeted therapies. Glycans extracted from plant lectins are known to act as ligands for C-type lectin receptors (CLRs) that are primarily present on immune cells. Plant-derived glycosylated lectins offer diversity in their N-linked oligosaccharide structures that can serve as a unique source of homogenous and heterogenous glycans. Among the plant lectins-derived glycan motifs, Man9GlcNAc2Asn exhibits high-affinity interactions with CLRs that may resemble glycan motifs of pathogens. Thus, such glycan domains when presented along with antigens complexed with a nanocarrier of choice may bewilder the immune cells and direct antigen cross-presentation - a cytotoxic T lymphocyte immune response mediated by CD8+ T cells. Glycan structure analysis has attracted considerable interest as glycans are looked upon as better therapeutic alternatives than monoclonal antibodies due to their cost-effectiveness, reduced toxicity and side effects, and high specificity. Furthermore, this approach will be useful to understand whether the multivalent glycan presentation on the surface of nanocarriers can overcome the low-affinity lectin-ligand interaction and thereby modulation of CLR-dependent immune response. Besides this, understanding how the heterogeneity of glycan structure impacts the antigen cross-presentation is pivotal to develop alternative targeted therapies. In the present review, we discuss the findings on structural analysis of glycans from natural lectins performed using GlycanBuilder2 - a software tool based on a thorough literature review of natural lectins. Additionally, we discuss how multiple parameters like the orientation of glycan ligands, ligand density, simultaneous targeting of multiple CLRs and design of antigen delivery nanocarriers may influence the CLR targeting efficacy. Integrating this information will eventually set the ground for new generation immunotherapeutic vaccine design for the treatment of various human malignancies.
Collapse
|
32
|
Yuba E. Development of functional liposomes by modification of stimuli-responsive materials and their biomedical applications. J Mater Chem B 2020; 8:1093-1107. [PMID: 31960007 DOI: 10.1039/c9tb02470k] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Liposomes are a promising nanocarrier for drug delivery because of their biocompatibility and the encapsulation capacity of drugs. Liposomes can be functionalized easily by introduction of functional materials such as stimulus-responsive materials. Temperature-responsive liposomes and pH-responsive liposomes are representative stimulus-responsive liposomes that can deliver drugs to locally heated target tissues and intracellular organelles. Here, temperature-responsive liposomes for the selective release of cargo and pH-responsive liposomes for the induction of antigen-specific immunity are overviewed. Temperature-responsive polymer-modified liposomes immediately released drugs in response to heating, which achieved selective drug release at a tumour after topical heating of tumour-bearing mice. Introduction of MR-detectable molecules enabled the tracing of liposome accumulation into target sites to optimize the heating timing. These liposomes can also be combined with magnetic nanoparticles or carbon nanomaterials to attain magnetic field-responsive, electric field-responsive and light-responsive properties to support on-demand drug release or control of biological reactions using these external stimuli. pH-Responsive liposomes were produced by modification of poly(carboxylic acid) derivatives or by pH-responsive amphiphiles. These liposomes delivered antigenic proteins into the cytosol of antigen presenting cells, which induced cross-presentation and antigen-specific cellular immunity. Adjuvant molecules or bioactive polysaccharide-based pH-responsive polymers improved their immunity-inducing effect further, leading to tumour regression in tumour-bearing mice. Precise design and control of the structures of stimulus-responsive materials and combination with functional materials are expected to create novel methodologies to control biological functions and to produce highly potent liposomal drugs that can achieve selective release of bioactive molecules.
Collapse
Affiliation(s)
- Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.
| |
Collapse
|
33
|
Zhang C, Pu K. Molecular and nanoengineering approaches towards activatable cancer immunotherapy. Chem Soc Rev 2020; 49:4234-4253. [DOI: 10.1039/c9cs00773c] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review summarizes the development of activatable immunotherapeutic nanoagents that activate antitumor immunity only in response to internal or external stimuli, which potentially enhance patient response rates while reducing immune-related adverse events during cancer immunotherapy.
Collapse
Affiliation(s)
- Chi Zhang
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
| |
Collapse
|
34
|
Miura R, Sawada SI, Mukai SA, Sasaki Y, Akiyoshi K. Antigen Delivery to Antigen-Presenting Cells for Adaptive Immune Response by Self-Assembled Anionic Polysaccharide Nanogel Vaccines. Biomacromolecules 2019; 21:621-629. [PMID: 31800235 DOI: 10.1021/acs.biomac.9b01351] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although current vaccine technology induces sufficient antibody responses to prophylactically ward off viral infections, an anticancer vaccine that directs the patient's immune system to directly fight extant malignant cells will require inducing Th1 and cytotoxic T lymphocyte responses in addition to antibody-mediated activities. Thus, new mechanisms are necessary to deliver antigen to cells in the lymphatic system that will induce these responses. To this end, we have developed a cholesterol-bearing pullulan (CHP) self-assembly nanogel of less than 100 nm, which we have now further modified to be anionic by carboxyl group substitution. Overall, the nanogel-protected antigens during transport to the lymphatic system and converting the vehicle to an anionic charge improved interactions with antigen-presenting cells. We further show that these modified nanogels are a more efficient system for delivering antigen to antigen-presenting cells, particularly langerin-expressing cells, and that this induced significant adaptive immunity. Therefore, we think that this technology could be used to improve anticancer immunotherapies.
Collapse
Affiliation(s)
- Risako Miura
- Department of Polymer Chemistry, Graduate School of Engineering , Kyoto University , Kyoto 615-8510 , Japan
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering , Kyoto University , Kyoto 615-8510 , Japan
| | - Sada-Atsu Mukai
- Department of Polymer Chemistry, Graduate School of Engineering , Kyoto University , Kyoto 615-8510 , Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering , Kyoto University , Kyoto 615-8510 , Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering , Kyoto University , Kyoto 615-8510 , Japan
| |
Collapse
|
35
|
Miyazaki M, Yuba E, Hayashi H, Harada A, Kono K. Development of pH-Responsive Hyaluronic Acid-Based Antigen Carriers for Induction of Antigen-Specific Cellular Immune Responses. ACS Biomater Sci Eng 2019; 5:5790-5797. [PMID: 33405671 DOI: 10.1021/acsbiomaterials.9b01278] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Maiko Miyazaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Hiroshi Hayashi
- Sciencelin, 1-1-35, Nishiawaji, Higashiyodogawa-ku, Osaka, Osaka 533-0031, Japan
| | - Atsushi Harada
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Kenji Kono
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
36
|
Lv J, Fan Q, Wang H, Cheng Y. Polymers for cytosolic protein delivery. Biomaterials 2019; 218:119358. [DOI: 10.1016/j.biomaterials.2019.119358] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/11/2019] [Accepted: 07/13/2019] [Indexed: 12/31/2022]
|
37
|
Augustine R, Kalva N, Kim HA, Zhang Y, Kim I. pH-Responsive Polypeptide-Based Smart Nano-Carriers for Theranostic Applications. Molecules 2019; 24:E2961. [PMID: 31443287 PMCID: PMC6719039 DOI: 10.3390/molecules24162961] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
Smart nano-carriers have attained great significance in the biomedical field due to their versatile and interesting designs with different functionalities. The initial stages of the development of nanocarriers mainly focused on the guest loading efficiency, biocompatibility of the host and the circulation time. Later the requirements of less side effects with more efficacy arose by attributing targetability and stimuli-responsive characteristics to nano-carriers along with their bio- compatibility. Researchers are utilizing many stimuli-responsive polymers for the better release of the guest molecules at the targeted sites. Among these, pH-triggered release achieves increasing importance because of the pH variation in different organ and cancer cells of acidic pH. This specific feature is utilized to release the guest molecules more precisely in the targeted site by designing polymers having specific functionality with the pH dependent morphology change characteristics. In this review, we mainly concert on the pH-responsive polypeptides and some interesting nano-carrier designs for the effective theranostic applications. Also, emphasis is made on pharmaceutical application of the different nano-carriers with respect to the organ, tissue and cellular level pH environment.
Collapse
Affiliation(s)
- Rimesh Augustine
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Nagendra Kalva
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Ho An Kim
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Yu Zhang
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Il Kim
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea.
| |
Collapse
|
38
|
Liu J, Zhang R, Xu ZP. Nanoparticle-Based Nanomedicines to Promote Cancer Immunotherapy: Recent Advances and Future Directions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900262. [PMID: 30908864 DOI: 10.1002/smll.201900262] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/21/2019] [Indexed: 05/27/2023]
Abstract
Cancer immunotherapy is a promising cancer terminator by directing the patient's own immune system in the fight against this challenging disorder. Despite the monumental therapeutic potential of several immunotherapy strategies in clinical applications, the efficacious responses of a wide range of immunotherapeutic agents are limited in virtue of their inadequate accumulation in the tumor tissue and fatal side effects. In the last decades, increasing evidences disclose that nanotechnology acts as an appealing solution to address these technical barriers via conferring rational physicochemical properties to nanomaterials. In this Review, an imperative emphasis will be drawn from the current understanding of the effect of a nanosystem's structure characteristics (e.g., size, shape, surface charge, elasticity) and its chemical modification on its transport and biodistribution behavior. Subsequently, rapid-moving advances of nanoparticle-based cancer immunotherapies are summarized from traditional vaccine strategies to recent novel approaches, including delivery of immunotherapeutics (such as whole cancer cell vaccines, immune checkpoint blockade, and immunogenic cell death) and engineered immune cells, to regulate tumor microenvironment and activate cellular immunity. The future prospects may involve in the rational combination of a few immunotherapies for more efficient cancer inhibition and elimination.
Collapse
Affiliation(s)
- Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
39
|
Liposome and immune system interplay: Challenges and potentials. J Control Release 2019; 305:194-209. [DOI: 10.1016/j.jconrel.2019.05.030] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 01/20/2023]
|
40
|
Li C, Lu Y, Chen Q, Hu H, Zhao X, Qiao M, Chen D. Tailored Polymers with Complement Activation Ability To Improve Antitumor Immunity. Mol Pharm 2019; 16:2648-2660. [PMID: 31046290 DOI: 10.1021/acs.molpharmaceut.9b00195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The complement system plays an important role in host innate immunity, and its activation can be exploited as a potential strategy for vaccine adjuvants. Herein, a pH-responsive micellar vaccine platform (COOH-NPs) was developed using a carboxyl-modified diblock copolymer of poly(2-ethyl-2-oxazoline)-poly(d,l-lactide) (COOH-PEOz-PLA). The copolymer self-assembled into micelles with hydroxyl groups shielding on the surface, which activated the complement system for the enhanced immune responses. Compared with the control nanoparticles (OCH3-NPs), COOH-NPs significantly enhanced lymph node-resident dendritic cell maturation, antigen-specific IgG production, antigen-specific CD4+ and CD8+ T-cell activation, and the amount of memory T-cell generation in vivo. Furthermore, immunization with COOH-NPs/OVA in E.G7-OVA tumor-bearing mice not only remarkably inhibited tumor growth but also prolonged the survival of tumor-bearing mice. These results indicated that COOH-NPs with the capability of complement activation efficiently boosted the immune responses for the antitumor effect. The study demonstrated the significance of taking advantage of a complement-activating vaccine platform for cancer immunotherapy.
Collapse
Affiliation(s)
- Chenxi Li
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Yue Lu
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Qing Chen
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Haiyang Hu
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Xiuli Zhao
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Mingxi Qiao
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Dawei Chen
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China.,School of Pharmacy , Soochow University , Suzhou 215123 , China
| |
Collapse
|
41
|
Liu B, Zhang Q, Zhou F, Ren L, Zhao Y, Yuan X. Enhancing Membrane-Disruptive Activity via Hydrophobic Phenylalanine and Lysine Tethered to Poly(aspartic acid). ACS APPLIED MATERIALS & INTERFACES 2019; 11:14538-14547. [PMID: 30933470 DOI: 10.1021/acsami.8b22721] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Amphiphilic polymers with pH-responsive abilities have been widely used as carriers for intracellular delivery of bioactive substances, while their membrane-disruptive activity exerted on cells is a critical characteristic that determines delivery efficiency. Herein, we present a novel method to prepare amphiphilic and pH-responsive polymers by chemically tethering l-phenylalanine methyl ester and followed by Nε-carbobenzyloxy-l-lysine benzyl ester to the side carboxylic acid groups of poly(aspartic acid). The obtained phenylalanine- and lysine-grafted polymer (PAsp- g-Phe)- g-Lys demonstrated enhanced membrane-disruptive activity at pH 7.4 in comparison with that of PAsp- g-Phe. Moreover, the pH-responsive behavior of the grafted polymers caused by the significantly intensified hydrophobicity could be modulated by the tethered amount of hydrophobic amino acids with phenyl groups. The prepared amphiphilic (PAsp- g-Phe)- g-Lys could facilitate entry of calcein into NIH/3T3 and HeLa cells at physiological pH values, possibly due to local chemical destabilization of cell membranes by the interaction between the polymer and membrane bilayers. Therefore, we have provided a feasible approach to prepare pH-responsive polymers with enhanced membrane-disruptive activity, and the phenylalanine- and lysine-grafted polymers could be a potential candidate for intracellular delivery of bioactive molecules in biomedical applications.
Collapse
Affiliation(s)
- Bo Liu
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Qifa Zhang
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Fang Zhou
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Lixia Ren
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Yunhui Zhao
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Xiaoyan Yuan
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| |
Collapse
|
42
|
Okubo M, Miyazaki M, Yuba E, Harada A. Chondroitin Sulfate-Based pH-Sensitive Polymer-Modified Liposomes for Intracellular Antigen Delivery and Induction of Cancer Immunity. Bioconjug Chem 2019; 30:1518-1529. [DOI: 10.1021/acs.bioconjchem.9b00221] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Minori Okubo
- Department of Applied Chemistry Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Maiko Miyazaki
- Department of Applied Chemistry Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Eiji Yuba
- Department of Applied Chemistry Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Atsushi Harada
- Department of Applied Chemistry Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
43
|
Mi Y, Hagan CT, Vincent BG, Wang AZ. Emerging Nano-/Microapproaches for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801847. [PMID: 30937265 PMCID: PMC6425500 DOI: 10.1002/advs.201801847] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/01/2018] [Indexed: 05/15/2023]
Abstract
Cancer immunotherapy has achieved remarkable clinical efficacy through recent advances such as chimeric antigen receptor-T cell (CAR-T) therapy, immune checkpoint blockade (ICB) therapy, and neoantigen vaccines. However, application of immunotherapy in a clinical setting has been limited by low durable response rates and immune-related adverse events. The rapid development of nano-/microtechnologies in the past decade provides potential strategies to improve cancer immunotherapy. Advances of nano-/microparticles such as virus-like size, high surface to volume ratio, and modifiable surfaces for precise targeting of specific cell types can be exploited in the design of cancer vaccines and delivery of immunomodulators. Here, the emerging nano-/microapproaches in the field of cancer vaccines, immune checkpoint blockade, and adoptive or indirect immunotherapies are summarized. How nano-/microparticles improve the efficacy of these therapies, relevant immunological mechanisms, and how nano-/microparticle methods are able to accelerate the clinical translation of cancer immunotherapy are explored.
Collapse
Affiliation(s)
- Yu Mi
- Laboratory of Nano‐ and Translational MedicineCarolina Center for Cancer Nanotechnology ExcellenceCarolina Institute of NanomedicineLineberger Comprehensive Cancer CenterDepartment of Radiation OncologyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - C. Tilden Hagan
- Laboratory of Nano‐ and Translational MedicineCarolina Center for Cancer Nanotechnology ExcellenceCarolina Institute of NanomedicineLineberger Comprehensive Cancer CenterDepartment of Radiation OncologyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer CenterDepartment of Microbiology & ImmunologyCurriculum in Bioinformatics and Computational BiologyDivision of Hematology/OncologyDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - Andrew Z. Wang
- Laboratory of Nano‐ and Translational MedicineCarolina Center for Cancer Nanotechnology ExcellenceCarolina Institute of NanomedicineLineberger Comprehensive Cancer CenterDepartment of Radiation OncologyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| |
Collapse
|
44
|
Yang Y, Wang Z, Peng Y, Ding J, Zhou W. A Smart pH-Sensitive Delivery System for Enhanced Anticancer Efficacy via Paclitaxel Endosomal Escape. Front Pharmacol 2019; 10:10. [PMID: 30733675 PMCID: PMC6353802 DOI: 10.3389/fphar.2019.00010] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/07/2019] [Indexed: 12/17/2022] Open
Abstract
Micelles are highly attractive nano-drug delivery systems for targeted cancer therapy. While they have been demonstrated to significantly alleviate the side-effects of their cargo drugs, the therapy outcomes are usually suboptimal partially due to ineffective drug release and endosome entrapment. Stimulus-responsive nanoparticles have allowed controlled drug release in a smart fashion, and we want to use this concept to design novel micelles. Herein, we reported pH-sensitive paclitaxel (PTX)-loaded poly (ethylene glycol)-phenylhydrazone-dilaurate (PEG-BHyd-dC12) micelles (PEG-BHyd-dC12/PTX). The micelles were spherical, with an average particle size of ∼135 nm and a uniform size distribution. The pH-responsive properties of the micelles were certified by both colloidal stability and drug release profile, where the particle size was strikingly increased accompanied by faster drug release as pH decreased from 7.4 to 5.5. As a result, the micelles exhibited much stronger cytotoxicity than the pH-insensitive counterpart micelles against various types of cancer cells due to the hydrolysis of the building block polymers and subsequent rapid PTX release. Overall, these results demonstrate that the PEG-BHyd-dC12 micelle is a promising drug delivery system for cancer therapy.
Collapse
Affiliation(s)
- Yihua Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmaceutical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Zhe Wang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, China
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
45
|
Fundamentals of Polymers Science Applied in Pharmaceutical Product Development. BASIC FUNDAMENTALS OF DRUG DELIVERY 2019. [DOI: 10.1016/b978-0-12-817909-3.00003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
46
|
Sang W, Zhang Z, Dai Y, Chen X. Recent advances in nanomaterial-based synergistic combination cancer immunotherapy. Chem Soc Rev 2019; 48:3771-3810. [DOI: 10.1039/c8cs00896e] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This review aims to summarize various synergistic combination cancer immunotherapy strategies based on nanomaterials.
Collapse
Affiliation(s)
- Wei Sang
- Cancer Centre
- Faculty of Health Sciences
- University of Macau
- Macau SAR 999078
- China
| | - Zhan Zhang
- Cancer Centre
- Faculty of Health Sciences
- University of Macau
- Macau SAR 999078
- China
| | - Yunlu Dai
- Cancer Centre
- Faculty of Health Sciences
- University of Macau
- Macau SAR 999078
- China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine
- National Institute of Biomedical Imaging and Bioengineering
- National Institutes of Health
- Bethesda
- USA
| |
Collapse
|
47
|
Wang YY, Wang WL, Shen XC, Zhou B, Chen T, Guo ZX, Wen CC, Jiang BP, Liang H. Combination-Responsive MoO 3- x-Hybridized Hyaluronic Acid Hollow Nanospheres for Cancer Phototheranostics. ACS APPLIED MATERIALS & INTERFACES 2018; 10:42088-42101. [PMID: 30408413 DOI: 10.1021/acsami.8b15818] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
It is of extreme importance to reduce side effects resulting from the nonspecific uptake of phototherapeutic agents by normal tissues. Currently, the single responsive strategy still cannot entirely satisfy the requirements of practical applications. In this study, we developed one kind of combination-responsive phototherapeutic nanoplatforms, where oxygen-deficient molybdenum oxide (MoO3- x) hybridized hyaluronic acid (HA) hollow nanospheres, namely, MoO3- x@HA HNSs, were constructed via a facile one-step method. In MoO3- x@HA HNSs, the reasonable combination of actively targeted specificity endowed by the HA component and tumor microenvironment-responsive phototherapy activity induced by the MoO3- x component can effectively improve the precision of phototherapy. The in vitro and in vivo experimental results confirm that MoO3- x@HA HNSs can selectively kill CD44-overexpressing cancer cells and inhibit tumor growth under an 808 nm laser irradiation, revealing their remarkable synergistic photothermal therapy/photodynamic therapy effect with CD44 receptor-targeted specificity and pH responsiveness in treating cancer. We also prove that MoO3- x@HA HNSs can serve as one kind of contrast agent to achieve the computed tomography/photoacoustic imaging. Encouraged by these results, it is anticipated that the reasonable combination of active targeting and tumor microenvironment responsiveness can be a promising strategy to develop phototherapeutic nanoplatforms for precise multimodality cancer theranostics.
Collapse
Affiliation(s)
- Yuan-Yuan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Wen-Long Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Bo Zhou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Ting Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Zheng-Xi Guo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Chang-Chun Wen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Bang-Ping Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| |
Collapse
|
48
|
Surendran SP, Moon MJ, Park R, Jeong YY. Bioactive Nanoparticles for Cancer Immunotherapy. Int J Mol Sci 2018; 19:E3877. [PMID: 30518139 PMCID: PMC6321368 DOI: 10.3390/ijms19123877] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 11/30/2018] [Accepted: 12/02/2018] [Indexed: 12/18/2022] Open
Abstract
Currently, immunotherapy is considered to be one of the effective treatment modalities for cancer. All the developments and discoveries in this field up to the recent Nobel Prize add to the interest for research into this vast area of study. Targeting tumor environment as well as the immune system is a suitable strategy to be applied for cancer treatment. Usage of nanoparticle systems for delivery of immunotherapeutic agents to the body being widely studied and found to be a promising area of research to be considered and investigated further. Nanoparticles for immunotherapy would be one of the effective treatment options for cancer therapy in the future due to their high specificity, efficacy, ability to diagnose, imaging, and therapeutic effect. Among the many nanoparticle systems, polylactic-co-glycolic acid (PLGA) nanoparticles, liposomes, micelles, gold nanoparticles, iron oxide, dendrimers, and artificial exosomes are widely used for immunotherapy of cancer. Moreover, the combination therapy found to be the more effective way of treating the tumor. Here, we review the current trends in nanoparticle therapy and efficiency of these nanosystems in delivering antigens, adjuvants, therapeutic drugs, and other immunotherapeutic agents. This review summarizes the currently available bioactive nanoparticle systems for cancer immunotherapy.
Collapse
Affiliation(s)
- Suchithra Poilil Surendran
- Department of Biomedical Sciences, Biomolecular Theranostics (BiT) Lab, Chonnam National University Medical School, Hwasun 58128, South Korea.
| | - Myeong Ju Moon
- Department of Radiology, Biomolecular Theranostics (BiT) Lab, Chonnam National University Medical School, Hwasun 58128, South Korea.
| | - Rayoung Park
- Department of Radiology, Biomolecular Theranostics (BiT) Lab, Chonnam National University Medical School, Hwasun 58128, South Korea.
| | - Yong Yeon Jeong
- Department of Radiology, Biomolecular Theranostics (BiT) Lab, Chonnam National University Medical School, Hwasun 58128, South Korea.
| |
Collapse
|
49
|
Reprogramming Tumor Associated Macrophage Phenotype by a Polysaccharide from Ilex asprella for Sarcoma Immunotherapy. Int J Mol Sci 2018; 19:ijms19123816. [PMID: 30513582 PMCID: PMC6320939 DOI: 10.3390/ijms19123816] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 12/25/2022] Open
Abstract
We report here the discovery of an acidic polysaccharide, namely IAPS-2, from the root of Ilex asprella, with anti-tumor activity via a repolarizing tumor associated macrophages (TAMs) phenotype. We obtained IAPS-2 polysaccharide from this herb based on acidity and found that IAPS-2 expressed the activity of promoting the secretion of anti-tumor cytokines in macrophages. Furthermore, we evaluated its anti-tumor effect on TAM cells, through the activation of nuclear factor-κB (NF-κB) and signal transducer and activator of transcription (STAT) signaling. In particular, in the tumor murine model, IAPS-2 demonstrated that it could significantly inhibit the growth of tumors via modulating the function of TAMs and increase the animal survival rate. In summary, IAPS-2, with a clearly illustrated chemical composition, potent anti-tumor activity, and a solid mechanism of action, may be developed into a valuable therapeutic tool for cancer immunotherapy.
Collapse
|
50
|
Miura R, Tahara Y, Sawada SI, Sasaki Y, Akiyoshi K. Structural effects and lymphocyte activation properties of self-assembled polysaccharide nanogels for effective antigen delivery. Sci Rep 2018; 8:16464. [PMID: 30405172 PMCID: PMC6220277 DOI: 10.1038/s41598-018-34885-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/28/2018] [Indexed: 12/27/2022] Open
Abstract
The success of immunotherapeutic vaccines is often limited by their inability to activate the cytotoxic T lymphocyte (CTL)-inducing Th1 pathway. We investigated the ability of self-assembled nanogels (CHP or CH-CDex) to activate this pathway, and characterised them chemically and biologically. Once loaded with antigen (ovalbumin, OVA) their OVA encapsulation and dissociation rates suggested the possibility of effective antigen delivery. The DC2.4 dendritic cell line took up either vaccine time-dependently, but both vaccines required CpG DNA for class I MHC presentation. The nanogel vaccines interacted with RAW264.7, a Balb/c mouse-derived macrophage cell line, and co-localised with lysosomes, suggesting their endocytotic internalization in RAW264.7. Both vaccines activated CTLs better than OVA alone. Unlike OVA alone, the nanogel vaccines induced IgG2a antibody production in mice, whereas the former induced IgG1 antibodies. OVA-nanogel delivery to the draining lymph nodes (DLNs) was higher than that for OVA alone, reaching a deeper medullary area. Furthermore, Langerin+ CD103+ DCs interacted with the nanogel vaccines effectively, which is a subset of cross-presentation DC, in the DLNs. The nanogel vaccines each had good anti-tumour efficacy in OVA tumour-bearing mice compared with the OVA alone. Thus, CHP and CH-CDex nanogels should be investigated further because of the great potential they offer for immunotherapy.
Collapse
Affiliation(s)
- Risako Miura
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Yoshiro Tahara
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan.,Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, Motooka 744, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan.
| |
Collapse
|