1
|
Ma M, Li X, Zhong M, Li X, Yu J, Wang Z, Lv Q, Li X, He Z, Liu H, Wang Y. Galloylated Toll-Like Receptor 7/8 Agonist Nanovaccine for Enhanced Tumor Antigen Delivery in Personalized Immunotherapy. ACS NANO 2025; 19:11900-11912. [PMID: 40102033 DOI: 10.1021/acsnano.4c15442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Cancer vaccines, a critical technology in cancer immunotherapy, have shown great therapeutic potential. However, traditional vaccines based on tumor cell lysates (TCLs) have shown disappointing results in early clinical trials due to low immunogenicity, in vivo instability, and the inability to codeliver with adjuvants. To address these issues, we developed a nanoparticle vaccine, R848-GA@TCLs, by modifying the toll-like receptor 7/8 (TLR7/8) agonist R848 with gallic acid. This nanovaccine leverages the "capturing" ability of the galloyl moiety to coload TCLs and R848, forming stable nanoparticles. R848-GA@TCLs efficiently target lymph nodes, increasing TCL accumulation 10-fold, and enable the synchronized release of antigens and adjuvants within dendritic cells (DCs). Our results show that R848-GA@TCLs increase with respect to the cross-presentation of tumor antigens, promote the production of pro-inflammatory cytokines, and activate DCs, leading to a significant increase in effector T cells, natural killer (NK) cells, and M1 macrophages. This strong immune response resulted in potent antitumor effects, with R848-GA@TCLs demonstrating efficacy in multiple tumor models by significantly inhibiting tumor growth and metastasis. In conclusion, R848-GA@TCLs represent a personalized cancer vaccine capable of codelivering TCLs and adjuvants, eliciting robust antitumor immune responses, and hold great potential for clinical applications.
Collapse
Affiliation(s)
- Mengyao Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Ximu Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Mingyuan Zhong
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuejing Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhaomeng Wang
- Department of Oncology, Innovative Cancer Drug Research and Engineering Center of Liaoning Province, Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Qingzhi Lv
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Xin Li
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| |
Collapse
|
2
|
Chen D, Ling X, Wang Y, Zhang Q, He X, Dong Z, Li M, He Q. Autophagy-activating aluminum hydroxide nanovaccine for enhanced antigen presentation and anti-tumor immunity. J Control Release 2025; 377:223-235. [PMID: 39547420 DOI: 10.1016/j.jconrel.2024.11.018] [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: 06/11/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
Lymph node (LN) targeting and antigen presentation by antigen-presenting cells (APCs) are critical factors affecting the immune responses induced by tumor vaccines. Autophagy activation promotes MHC class I and II antigen presentation in APCs. To enhance antigen presentation in LNs, we developed an aluminum hydroxide nanovaccine that simultaneously incorporates the autophagy-activating peptide Beclin-1 and the antigenic protein OVA (B/O@AN nanovaccine) through layer-by-layer electrostatic interaction. B/O@AN has a particle size of approximately 80 nm and efficiently targets lymph nodes following subcutaneous administration. The combination of the Beclin-1 peptide with the aluminum hydroxide nanovaccine promotes dendritic cell (DC) maturation. More importantly, B/O@AN facilitates antigen cross-presentation by promoting lysosomal escape and autophagy induction. After immunization, compared to O/@AN without Beclin-1, B/O@AN significantly augments antigen-specific cellular immune responses, leading to substantial increases in cytotoxic T lymphocytes (CTLs), T-helper 1 (Th1) cells, as well as serum antibody levels, thereby impeding melanoma development and progression in both prophylactic and therapeutic settings. These results provide evidence that autophagy activation strengthens antigen presentation and augments the antigen-specific immune responses of the aluminum hydroxide nanovaccine.
Collapse
Affiliation(s)
- Dong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Xiaoli Ling
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Yashi Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Qiang Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Xuan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Ziyan Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Baselga M, Güemes A, Arruebo M, Yus C, Alejo T, Sebastián V, Martínez G, Arribas D, Mendoza G, Junquera C, Monleón E. Preclinical evaluation of polymer encapsulated carbon-based nano and microparticles for sentinel lymph node tattooing. Sci Rep 2024; 14:29512. [PMID: 39604460 PMCID: PMC11603039 DOI: 10.1038/s41598-024-80931-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Selective sentinel lymph node biopsy (SNLB) is the standard method for detecting regional metastases in breast cancer patients. Identifying affected axillary lymph nodes before neoadjuvant treatment is crucial, as such treatment may alter drainage pathways and lymph node morphology, hindering the identification of sentinel lymph nodes. The use of carbon-based tattooing on sentinel lymph nodes (SLN) has been employed as a permanent tattooing method in clinical studies of Targeted Axillary Dissection (TAD), aiding in the SLN identification during surgery. Our study introduces a new method of lymph node tattooing based on poly lactic-co-glycolic (PLGA) particles with encapsulated carbon. This strategy substantially improves tattooing efficiency over single carbon suspensions currently used in clinical studies. We synthesized and characterized carbon-loaded PLGA micro- and nanoparticles, experimentally assessing their biological impact on porcine lymph nodes. The effect of particles' size and concentration was evaluated over time (from 1 to 16 weeks). Light and electron microscopy studies were conducted to characterize the cellular effects induced by the presence of these particles. Our findings reveal that the diverse physicochemical parameters of the particles interact differently with the lymphatic tissue, influencing their biodistribution within the lymph nodes and the intensity of the inflammatory response.
Collapse
Affiliation(s)
- Marta Baselga
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
| | - Antonio Güemes
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, 50009, Spain
| | - Manuel Arruebo
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Zaragoza, 50009, Spain
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro, Zaragoza, 50018, Spain
| | - Cristina Yus
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain.
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Zaragoza, 50009, Spain.
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro, Zaragoza, 50018, Spain.
| | - Teresa Alejo
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Zaragoza, 50009, Spain
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro, Zaragoza, 50018, Spain
| | - Víctor Sebastián
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Zaragoza, 50009, Spain
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro, Zaragoza, 50018, Spain
- Networking Res. Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de 13 Salud Carlos III, Madrid, 28029, Spain
| | - Gema Martínez
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Zaragoza, 50009, Spain
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro, Zaragoza, 50018, Spain
- Networking Res. Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de 13 Salud Carlos III, Madrid, 28029, Spain
| | - Dolores Arribas
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, 50009, Spain
| | - Gracia Mendoza
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
| | - Concepción Junquera
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Department of Human Anatomy and Histology, University of Zaragoza, Zaragoza, 50009, Spain
| | - Eva Monleón
- Institute for Health Research Aragon (IIS Aragón), Zaragoza, 50009, Spain
- Department of Human Anatomy and Histology, University of Zaragoza, Zaragoza, 50009, Spain
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, University of Zaragoza, Zaragoza, 50009, Spain
| |
Collapse
|
4
|
Archer PA, Heiler AJ, Bourque AR, Alapan Y, Thomas SN. Different leukocyte subsets are targeted by systemic and locoregional administration despite conserved nanomaterial characteristics optimal for lymph node delivery. Biomater Sci 2024; 12:5582-5597. [PMID: 39318195 PMCID: PMC11422756 DOI: 10.1039/d4bm00910j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024]
Abstract
Lymph nodes (LNs) house a large proportion of the body's leukocytes. Accordingly, engineered nanomaterials are increasingly developed to direct therapeutics to LNs to enhance their efficacy. Yet while lymphatic delivery of nanomaterials to LNs upon locoregional injection has been extensively evaluated, nanomaterial delivery to LN-localized leukocytes after intravenous administration has not been systematically explored nor benchmarked. In this work, a panel of inert, fluorescent nanoscale tracers and drug delivery vehicles were utilized to interrogate intravenous versus locoregionally administered nanomaterial access to LNs and leukocyte subsets therein. Hydrodynamic size and material effects on LN accumulation extents were similar between intravenous versus intradermal injection routes. Nanomaterial distribution to various LN leukocyte subsets differed substantially with injection route, however, in a manner not proportional to total LN accumulation. While intravenously administered nanomaterials accumulated in LNs lowly compared to systemic tissues, in sharp contrast to locoregional delivery, they exhibited size-dependent but material-independent access to immune cells within the LN parenchyma, which are not easily accessed with locoregional delivery.
Collapse
Affiliation(s)
- Paul A Archer
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alexander J Heiler
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alisyn R Bourque
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yunus Alapan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Lu R, Lee BJ, Lee E. Three-Dimensional Lymphatics-on-a-Chip Reveals Distinct, Size-Dependent Nanoparticle Transport Mechanisms in Lymphatic Drug Delivery. ACS Biomater Sci Eng 2024; 10:5752-5763. [PMID: 39176471 DOI: 10.1021/acsbiomaterials.4c01005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Although nanoparticle-based lymphatic drug delivery systems promise better treatment of cancer, infectious disease, and immune disease, their clinical translations are limited by low delivery efficiencies and unclear transport mechanisms. Here, we employed a three-dimensional (3D) lymphatics-on-a-chip featuring an engineered lymphatic vessel (LV) capable of draining interstitial fluids including nanoparticles. We tested lymphatic drainage of different sizes (30, 50, and 70 nm) of PLGA-b-PEG nanoparticles (NPs) using the lymphatics-on-a-chip device. In this study, we discovered that smaller NPs (30 and 50 nm) transported faster than larger NPs (70 nm) through the interstitial space, as expected, but the smaller NPs were captured by lymphatic endothelial cells (LECs) and accumulated within their cytosol, delaying NP transport into the lymphatic lumen, which was not observed in larger NPs. To examine the mechanisms of size-dependent NP transports, we employed four inhibitors, dynasore, nystatin, amiloride, and adrenomedullin, to selectively block dynamin-, caveolin-, macropinocytosis-mediated endocytosis-, and cell junction-mediated paracellular transport. Inhibiting dynamin using dynasore enhanced the transport of smaller NPs (30 and 50 nm) into the lymphatic lumen, minimizing cytosolic accumulation, but showed no effect on larger NP transport. Interestingly, the inhibition of caveolin by nystatin decreased the lymphatic transport of larger NPs without affecting the smaller NP transport, indicating distinct endocytosis mechanisms used by different sizes of NPs. Macropinocytosis inhibition by amiloride did not change the drainage of all sizes of NPs; however, paracellular transport inhibition by adrenomedullin blocked the lymphatic transport of NPs of all sizes. We further revealed that smaller NPs were captured in the Rab7-positive late-stage lymphatic endosomes to delay their lymphatic drainage, which was reversed by dynamin inhibition, suggesting that Rab7 is a potential target to enhance the lymphatic delivery of smaller NPs. Together, our 3D lymphatics-on-a-chip model unveils size-dependent NP transport mechanisms in lymphatic drug delivery.
Collapse
Affiliation(s)
- Renhao Lu
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Benjamin J Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
6
|
Zhang W, Lu L, Zhu Z, Deng F, Zhang W, Wang F, Zeng P, Shi H, Wang T, Chen Y, Song Y, Liu Y, Kang T, Li K, Mao J, Liu Z, Zhang L. A Manganese-Based Nanodriver Coordinates Tumor Prevention and Suppression through STING Activation in Glioblastoma. Adv Healthc Mater 2024; 13:e2400421. [PMID: 38576069 DOI: 10.1002/adhm.202400421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/23/2024] [Indexed: 04/06/2024]
Abstract
Glioblastoma (GBM), the most prevalent and aggressive primary malignant brain tumor, exhibits profound immunosuppression and demonstrates a low response rate to current immunotherapy strategies. Manganese cations (Mn2+) directly activate the cGAS/STING pathway and induce the unique catalytic synthesis of 2'3'-cGAMP to facilitate type I IFN production, thereby enhancing innate immunity. Here, a telodendrimer and Mn2+-based nanodriver (PLHM) with a small size is developed, which effectively target lymph nodes through the blood circulation and exhibit tumor-preventive effects at low doses of Mn2+ (3.7 mg kg-1). On the other hand, the PLHM nanodriver also exhibits apparent antitumor effects in GBM-bearing mice via inducing in vivo innate immune responses. The combination of PLHM with doxorubicin nanoparticles (PLHM-DOX NPs) results in superior inhibition of tumor growth in GBM-bearing mice due to the synergistic potentiation of STING pathway functionality by Mn2+ and the presence of cytoplasmic DNA. These findings demonstrate that PLHM-DOX NPs effectively stimulate innate immunity, promote dendritic cell maturation, and orchestrate cascaded infiltration of CD8 cytotoxic T lymphocytes within glioblastomas characterized by low immunogenicity. These nanodivers chelated with Mn2+ show promising potential for tumor prevention and antitumor effects on glioblastoma by activating the STING pathway.
Collapse
Affiliation(s)
- Wenyuan Zhang
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Liejing Lu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, 710032, China
| | - Fuan Deng
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wenchang Zhang
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, China
| | - Fengyi Wang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ping Zeng
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Haonan Shi
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Tong Wang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yichi Chen
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yue Song
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yiping Liu
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, China
| | - Tianze Kang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Kai Li
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jie Mao
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, China
| | - Zhengwei Liu
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, China
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
7
|
Wang J, Zhang Z, Liang R, Chen W, Li Q, Xu J, Zhao H, Xing D. Targeting lymph nodes for enhanced cancer vaccination: From nanotechnology to tissue engineering. Mater Today Bio 2024; 26:101068. [PMID: 38711936 PMCID: PMC11070719 DOI: 10.1016/j.mtbio.2024.101068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/02/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024] Open
Abstract
Lymph nodes (LNs) occupy a critical position in initiating and augmenting immune responses, both spatially and functionally. In cancer immunotherapy, tumor-specific vaccines are blooming as a powerful tool to suppress the growth of existing tumors, as well as provide preventative efficacy against tumorigenesis. Delivering these vaccines more efficiently to LNs, where antigen-presenting cells (APCs) and T cells abundantly reside, is under extensive exploration. Formulating vaccines into nanomedicines, optimizing their physiochemical properties, and surface modification to specifically bind molecules expressed on LNs or APCs, are common routes and have brought encouraging outcomes. Alternatively, porous scaffolds can be engineered to attract APCs and provide an environment for them to mature, proliferate and migrate to LNs. A relatively new research direction is inducing the formation of LN-like organoids, which have shown positive relevance to tumor prognosis. Cutting-edge advances in these directions and discussions from a future perspective are given here, from which the up-to-date pattern of cancer vaccination will be drawn to hopefully provide basic guidance to future studies.
Collapse
Affiliation(s)
- Jie Wang
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Zongying Zhang
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Rongxiang Liang
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, 266033, China
| | - Wujun Chen
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Qian Li
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Jiazhen Xu
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Hongmei Zhao
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Dongming Xing
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
8
|
Hartmeier PR, Ostrowski SM, Busch EE, Empey KM, Meng WS. Lymphatic distribution considerations for subunit vaccine design and development. Vaccine 2024; 42:2519-2529. [PMID: 38494411 DOI: 10.1016/j.vaccine.2024.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/30/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Subunit vaccines are an important platform for controlling current and emerging infectious diseases. The lymph nodes are the primary site generating the humoral response and delivery of antigens to these sites is critical to effective immunization. Indeed, the duration of antigen exposure within the lymph node is correlated with the antibody response. While current licensed vaccines are typically given through the intramuscular route, injecting vaccines subcutaneously allows for direct access to lymphatic vessels and therefore can enhance the transfer of antigen to the lymph nodes. However, protein subunit antigen uptake into the lymph nodes is inefficient, and subunit vaccines require adjuvants to stimulate the initial immune response. Therefore, formulation strategies have been developed to enhance the exposure of subunit proteins and adjuvants to the lymph nodes by increasing lymphatic uptake or prolonging the retention at the injection site. Given that lymph node exposure is a crucial consideration in vaccine design, in depth analyses of the pharmacokinetics of antigens and adjuvants should be the focus of future preclinical and clinical studies. This review will provide an overview of formulation strategies for targeting the lymphatics and prolonging antigen exposure and will discuss pharmacokinetic evaluations which can be applied toward vaccine development.
Collapse
Affiliation(s)
- Paul R Hartmeier
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Sarah M Ostrowski
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA 15213, USA
| | - Emelia E Busch
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Kerry M Empey
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, PA 15213, USA; Department of Immunology, School of Medicine University of Pittsburgh, PA 15213, USA
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15219, USA.
| |
Collapse
|
9
|
Chen H, Wang L, Zhao X, Jiang H, Wu M, Ding Y, Jia X, Zhang Y, Li T, Zhang Y, Zhou W, Zheng P, Yang Y, Du J. A Polymer-Based Antigen Carrier Activates Two Innate Immune Pathways for Adjuvant-Free Subunit Vaccines. ACS NANO 2024; 18:9160-9175. [PMID: 38478910 DOI: 10.1021/acsnano.4c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
The activation of multiple Pattern Recognition Receptors (PRRs) has been demonstrated to trigger inflammatory responses and coordinate the host's adaptive immunity during pathogen infections. The use of PRR agonists as vaccine adjuvants has been reported to synergistically induce specific humoral and cellular immune responses. However, incorporating multiple PRR agonists as adjuvants increases the complexity of vaccine design and manufacturing. In this study, we discovered a polymer that can activate both the Toll-like receptor (TLR) pathway and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. The polymer was then conjugated to protein antigens, creating an antigen delivery system for subunit vaccines. Without additional adjuvants, the antigen-polymer conjugates elicited strong antigen-specific humoral and cellular immune responses. Furthermore, the antigen-polymer conjugates, containing the Receptor Binding Domain (RBD) of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Protein or the Monkeypox Antigen M1R as the antigens, were found to induce potent antigen-specific antibodies, neutralizing antibodies, and cytotoxic T cells. Immunization with M1R-polymer also resulted in effective protection in a lethal challenge model. In conclusion, this vaccine delivery platform offers an effective, safe, and simple strategy for inducing antigen-specific immunity against infectious diseases.
Collapse
Affiliation(s)
- Hang Chen
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Luyao Wang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Xiaofan Zhao
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Haolin Jiang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
- Academy for Advanced Interdisciplinary Studies (AAIS), Peking University-Tsinghua University-National Institute Biological Sciences (PTN) Joint Graduate Program, Peking University, Beijing 100871, China
| | - Mengling Wu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yanchao Ding
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Xiangqian Jia
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yaning Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Tiantian Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yue Zhang
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Wen Zhou
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Peiyuan Zheng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yilong Yang
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Juanjuan Du
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
10
|
Jha SK, Imran M, Jha LA, Hasan N, Panthi VK, Paudel KR, Almalki WH, Mohammed Y, Kesharwani P. A Comprehensive review on Pharmacokinetic Studies of Vaccines: Impact of delivery route, carrier-and its modulation on immune response. ENVIRONMENTAL RESEARCH 2023; 236:116823. [PMID: 37543130 DOI: 10.1016/j.envres.2023.116823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
The lack of knowledge about the absorption, distribution, metabolism, and excretion (ADME) of vaccines makes former biopharmaceutical optimization difficult. This was shown during the COVID-19 immunization campaign, where gradual booster doses were introduced.. Thus, understanding vaccine ADME and its effects on immunization effectiveness could result in a more logical vaccine design in terms of formulation, method of administration, and dosing regimens. Herein, we will cover the information available on vaccine pharmacokinetics, impacts of delivery routes and carriers on ADME, utilization and efficiency of nanoparticulate delivery vehicles, impact of dose level and dosing schedule on the therapeutic efficacy of vaccines, intracellular and endosomal trafficking and in vivo fate, perspective on DNA and mRNA vaccines, new generation sequencing and mathematical models to improve cancer vaccination and pharmacology, and the reported toxicological study of COVID-19 vaccines. Altogether, this review will enhance the reader's understanding of the pharmacokinetics of vaccines and methods that can be implied in delivery vehicle design to improve the absorption and distribution of immunizing agents and estimate the appropriate dose to achieve better immunogenic responses and prevent toxicities.
Collapse
Affiliation(s)
- Saurav Kumar Jha
- Department of Biomedicine, Health & Life Convergence Sciences, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea; Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India.
| | - Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Laxmi Akhileshwar Jha
- H. K. College of Pharmacy, Mumbai University, Pratiksha Nagar, Jogeshwari, West Mumbai, 400102, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Vijay Kumar Panthi
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Science, Centenary Institute and University of Technology Sydney, Sydney, 2007, Australia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 24381, Saudi Arabia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
11
|
Martinis E, Ricci C, Trevisan C, Tomadini G, Tonon S. Cancer Vaccines: From the State of the Art to the Most Promising Frontiers in the Treatment of Colorectal Cancer. Pharmaceutics 2023; 15:1969. [PMID: 37514155 PMCID: PMC10383643 DOI: 10.3390/pharmaceutics15071969] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Colorectal cancer represents 10% of all new cancer cases each year and accounts for almost 10% of all cancer deaths. According to the WHO, by 2040 there will be a 60% increase in colorectal cancer cases. These data highlight the need to explore new therapeutic strategies. Classical interventions include surgical resection, chemotherapy and radiotherapy, which are invasive strategies that have many side effects on the patients and greatly affect their quality of life. A great advance in the treatment of this cancer type, as well as of all the others, could be the development of a vaccination strategy preventing the onset, the progression or the relapse of the pathology. In this review, we summarize the main vaccination strategies that are being studied for the treatment of colorectal cancer (CRC) and finally explore the possibility of using B-cells for the development of a new type of vaccine.
Collapse
Affiliation(s)
- Eleonora Martinis
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Carolina Ricci
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Caterina Trevisan
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Gaia Tomadini
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Silvia Tonon
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| |
Collapse
|
12
|
Ivanov KI, Samuilova OV, Zamyatnin AA. The emerging roles of long noncoding RNAs in lymphatic vascular development and disease. Cell Mol Life Sci 2023; 80:197. [PMID: 37407839 PMCID: PMC10322780 DOI: 10.1007/s00018-023-04842-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/06/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023]
Abstract
Recent advances in RNA sequencing technologies helped uncover what was once uncharted territory in the human genome-the complex and versatile world of long noncoding RNAs (lncRNAs). Previously thought of as merely transcriptional "noise", lncRNAs have now emerged as essential regulators of gene expression networks controlling development, homeostasis and disease progression. The regulatory functions of lncRNAs are broad and diverse, and the underlying molecular mechanisms are highly variable, acting at the transcriptional, post-transcriptional, translational, and post-translational levels. In recent years, evidence has accumulated to support the important role of lncRNAs in the development and functioning of the lymphatic vasculature and associated pathological processes such as tumor-induced lymphangiogenesis and cancer metastasis. In this review, we summarize the current knowledge on the role of lncRNAs in regulating the key genes and pathways involved in lymphatic vascular development and disease. Furthermore, we discuss the potential of lncRNAs as novel therapeutic targets and outline possible strategies for the development of lncRNA-based therapeutics to treat diseases of the lymphatic system.
Collapse
Affiliation(s)
- Konstantin I Ivanov
- Research Center for Translational Medicine, Sirius University of Science and Technology, Sochi, Russian Federation.
- Department of Microbiology, University of Helsinki, Helsinki, Finland.
| | - Olga V Samuilova
- Department of Biochemistry, Sechenov First Moscow State Medical University, Moscow, Russian Federation
- HSE University, Moscow, Russian Federation
| | - Andrey A Zamyatnin
- Research Center for Translational Medicine, Sirius University of Science and Technology, Sochi, Russian Federation
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
13
|
Maisel K, McClain CA, Bogseth A, Thomas SN. Nanotechnologies for Physiology-Informed Drug Delivery to the Lymphatic System. Annu Rev Biomed Eng 2023; 25:233-256. [PMID: 37000965 PMCID: PMC10879987 DOI: 10.1146/annurev-bioeng-092222-034906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2023]
Abstract
Accompanying the increasing translational impact of immunotherapeutic strategies to treat and prevent disease has been a broadening interest across both bioscience and bioengineering in the lymphatic system. Herein, the lymphatic system physiology, ranging from its tissue structures to immune functions and effects, is described. Design principles and engineering approaches to analyze and manipulate this tissue system in nanoparticle-based drug delivery applications are also elaborated.
Collapse
Affiliation(s)
- Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA;
| | - Claire A McClain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA;
| | - Amanda Bogseth
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA;
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA;
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Xia Y, Fu S, Ma Q, Liu Y, Zhang N. Application of Nano-Delivery Systems in Lymph Nodes for Tumor Immunotherapy. NANO-MICRO LETTERS 2023; 15:145. [PMID: 37269391 PMCID: PMC10239433 DOI: 10.1007/s40820-023-01125-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/07/2023] [Indexed: 06/05/2023]
Abstract
Immunotherapy has become a promising research "hotspot" in cancer treatment. "Soldier" immune cells are not uniform throughout the body; they accumulate mostly in the immune organs such as the spleen and lymph nodes (LNs), etc. The unique structure of LNs provides the microenvironment suitable for the survival, activation, and proliferation of multiple types of immune cells. LNs play an important role in both the initiation of adaptive immunity and the generation of durable anti-tumor responses. Antigens taken up by antigen-presenting cells in peripheral tissues need to migrate with lymphatic fluid to LNs to activate the lymphocytes therein. Meanwhile, the accumulation and retaining of many immune functional compounds in LNs enhance their efficacy significantly. Therefore, LNs have become a key target for tumor immunotherapy. Unfortunately, the nonspecific distribution of the immune drugs in vivo greatly limits the activation and proliferation of immune cells, which leads to unsatisfactory anti-tumor effects. The efficient nano-delivery system to LNs is an effective strategy to maximize the efficacy of immune drugs. Nano-delivery systems have shown beneficial in improving biodistribution and enhancing accumulation in lymphoid tissues, exhibiting powerful and promising prospects for achieving effective delivery to LNs. Herein, the physiological structure and the delivery barriers of LNs were summarized and the factors affecting LNs accumulation were discussed thoroughly. Moreover, developments in nano-delivery systems were reviewed and the transformation prospects of LNs targeting nanocarriers were summarized and discussed.
Collapse
Affiliation(s)
- Yiming Xia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
15
|
Xu R, Bhangu SK, Sourris KC, Vanni D, Sani MA, Karas JA, Alt K, Niego B, Ale A, Besford QA, Dyett B, Patrick J, Carmichael I, Shaw JE, Caruso F, Cooper ME, Hagemeyer CE, Cavalieri F. An Engineered Nanosugar Enables Rapid and Sustained Glucose-Responsive Insulin Delivery in Diabetic Mice. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210392. [PMID: 36908046 DOI: 10.1002/adma.202210392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/11/2023] [Indexed: 05/26/2023]
Abstract
Glucose-responsive insulin-delivery platforms that are sensitive to dynamic glucose concentration fluctuations and provide both rapid and prolonged insulin release have great potential to control hyperglycemia and avoid hypoglycemia diabetes. Here, biodegradable and charge-switchable phytoglycogen nanoparticles capable of glucose-stimulated insulin release are engineered. The nanoparticles are "nanosugars" bearing glucose-sensitive phenylboronic acid groups and amine moieties that allow effective complexation with insulin (≈95% loading capacity) to form nanocomplexes. A single subcutaneous injection of nanocomplexes shows a rapid and efficient response to a glucose challenge in two distinct diabetic mouse models, resulting in optimal blood glucose levels (below 200 mg dL-1 ) for up to 13 h. The morphology of the nanocomplexes is found to be key to controlling rapid and extended glucose-regulated insulin delivery in vivo. These studies reveal that the injected nanocomplexes enabled efficient insulin release in the mouse, with optimal bioavailability, pharmacokinetics, and safety profiles. These results highlight a promising strategy for the development of a glucose-responsive insulin delivery system based on a natural and biodegradable nanosugar.
Collapse
Affiliation(s)
- Rong Xu
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Sukhvir Kaur Bhangu
- School of Science, RMIT University, Melbourne, Victoria, 3000, Australia
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3000, Australia
| | - Karly C Sourris
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Domitilla Vanni
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3000, Australia
- Dipartimento di Scienze e Tecnologie Chimiche Universita' di Roma "Tor Vergata", Via della Ricerca Scientifica 1, Rome, 00133, Italy
| | - Marc-Antoine Sani
- School of Chemistry, The Bio21 Institute, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - John A Karas
- School of Chemistry, The Bio21 Institute, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Karen Alt
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Be'eri Niego
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Anukreity Ale
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Quinn A Besford
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3000, Australia
| | - Brendan Dyett
- School of Science, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Joshua Patrick
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Irena Carmichael
- Monash Micro Imaging, Monash University, Melbourne, Victoria, 3004, Australia
| | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3000, Australia
| | - Mark E Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Christoph E Hagemeyer
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, Victoria, 3000, Australia
- Dipartimento di Scienze e Tecnologie Chimiche Universita' di Roma "Tor Vergata", Via della Ricerca Scientifica 1, Rome, 00133, Italy
| |
Collapse
|
16
|
Sestito LF, To KH, Cribb MT, Archer PA, Thomas SN, Dixon JB. Lymphatic-draining nanoparticles deliver Bay K8644 payload to lymphatic vessels and enhance their pumping function. SCIENCE ADVANCES 2023; 9:eabq0435. [PMID: 36827374 PMCID: PMC9956116 DOI: 10.1126/sciadv.abq0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Dysfunction of collecting lymphatic vessel pumping is associated with an array of pathologies. S-(-)-Bay K8644 (BayK), a small-molecule agonist of L-type calcium channels, improves vessel contractility ex vivo but has been left unexplored in vivo because of poor lymphatic access and risk of deleterious off-target effects. When formulated within lymph-draining nanoparticles (NPs), BayK acutely improved lymphatic vessel function, effects not seen from treatment with BayK in its free form. By preventing rapid drug access to the circulation, NP formulation also reduced BayK's dose-limiting side effects. When applied to a mouse model of lymphedema, treatment with BayK formulated in lymph-draining NPs, but not free BayK, improved pumping pressure generated by intact lymphatic vessels and tissue remodeling associated with the pathology. This work reveals the utility of a lymph-targeting NP platform to pharmacologically enhance lymphatic pumping in vivo and highlights a promising approach to treating lymphatic dysfunction.
Collapse
Affiliation(s)
- Lauren F. Sestito
- Wallace H. Coulter School of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Mechanical Engineering and Bioengineering, Valparaiso University, 1900 Chapel Dr, Valparaiso, IN 46383, USA
| | - Kim H. T. To
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Matthew T. Cribb
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Paul A. Archer
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Susan N. Thomas
- Wallace H. Coulter School of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - J. Brandon Dixon
- Wallace H. Coulter School of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
17
|
Fu X, Fu S, Cai Z, Jin R, Xia C, Lui S, Song B, Gong Q, Ai H. Manganese porphyrin/ICG nanoparticles as magnetic resonance/fluorescent dual-mode probes for imaging of sentinel lymph node metastasis. J Mater Chem B 2022; 10:10065-10074. [PMID: 36454208 DOI: 10.1039/d2tb01885c] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Diagnosis of sentinel lymph node (SLN) metastasis and its status are key parameters for predicting overall disease prognosis. In this work, Pluronic F127 stabilized ICG/tetra(4-carboxyphenyl)porphyrin-Mn(III) (TCPP(Mn)) nanoparticles (F127-ICG/Mn NPs) as fluorescent/magnetic resonance (FL/MR) dual-modality probes were prepared. The application of F127-ICG/Mn NPs in SLN imaging was mainly evaluated from two perspectives: the difference between the normal LN and the metastatic SLN and the difference between micrometastasis and macrometastasis. Normal and metastatic SLNs and micro- and macro-SLN metastasis were successfully distinguished through fluorescence and MR imaging with the help of F127-ICG/Mn NPs. In contrast, for the ICG group, the micro- and macro-SLN metastasis status could not be differentiated by fluorescence imaging. Besides, the lymph nodes can be stained green by the F127-ICG/Mn NPs and clearly visualized by the naked eye. In general, F127-ICG/Mn NPs demonstrated the potential of the preoperative diagnosis of SLN metastasis and its status, as well as intraoperative navigation by green-stained SLN and NIR FL imaging. This work provides a reference for developing multimodal nanoparticles for SLN metastasis diagnosis.
Collapse
Affiliation(s)
- Xiaomin Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Shengxiang Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Zhongyuan Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Chunchao Xia
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Su Lui
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Bing Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China.,Psychoradiology Research Unit of Chinese Academy of Medical Sciences, Sichuan University, Chengdu, China
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China.,Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
18
|
Cho KJ, Cho YE, Kim J. Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy. Pharmaceutics 2022; 14:2752. [PMID: 36559246 PMCID: PMC9788085 DOI: 10.3390/pharmaceutics14122752] [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: 10/28/2022] [Revised: 11/22/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
The lymphatic system has gained significant interest as a target tissue to control cancer progress, which highlights its central role in adaptive immune response. Numerous mechanistic studies have revealed the benefits of nano-sized materials in the transport of various cargos to lymph nodes, overcoming barriers associated with lymphatic physiology. The potential of sustained drug delivery systems in improving the therapeutic index of various immune modulating agents is also being actively discussed. Herein, we aim to discuss design rationales and principles of locoregional lymphatic drug delivery systems for invigorating adaptive immune response for efficient antitumor immunotherapy and provide examples of various advanced nanoparticle- and hydrogel-based formulations.
Collapse
Affiliation(s)
- Kyeong Jin Cho
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong 36729, Republic of Korea
| | - Jihoon Kim
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| |
Collapse
|
19
|
Yu M, Yang W, Yue W, Chen Y. Targeted Cancer Immunotherapy: Nanoformulation Engineering and Clinical Translation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2204335. [PMID: 36257824 PMCID: PMC9762307 DOI: 10.1002/advs.202204335] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/17/2022] [Indexed: 05/09/2023]
Abstract
With the rapid growth of advanced nanoengineering strategies, there are great implications for therapeutic immunostimulators formulated in nanomaterials to combat cancer. It is crucial to direct immunostimulators to the right tissue and specific immune cells at the right time, thereby orchestrating the desired, potent, and durable immune response against cancer. The flexibility of nanoformulations in size, topology, softness, and multifunctionality allows precise regulation of nano-immunological activities for enhanced therapeutic effect. To grasp the modulation of immune response, research efforts are needed to understand the interactions of immune cells at lymph organs and tumor tissues, where the nanoformulations guide the immunostimulators to function on tissue specific subsets of immune cells. In this review, recent advanced nanoformulations targeting specific subset of immune cells, such as dendritic cells (DCs), T cells, monocytes, macrophages, and natural killer (NK) cells are summarized and discussed, and clinical development of nano-paradigms for targeted cancer immunotherapy is highlighted. Here the focus is on the targeting nanoformulations that can passively or actively target certain immune cells by overcoming the physiobiological barriers, instead of directly injecting into tissues. The opportunities and remaining obstacles for the clinical translation of immune cell targeting nanoformulations in cancer therapy are also discussed.
Collapse
Affiliation(s)
- Meihua Yu
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Wei Yang
- Department of UrologyXinhua HospitalSchool of MedicineShanghai Jiaotong University1665 Kongjiang RoadShanghai200092P. R. China
| | - Wenwen Yue
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentDepartment of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University Cancer CenterTongji University School of MedicineShanghai200072P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| |
Collapse
|
20
|
Banerjee P, Roy S, Chakraborty S. Recent advancement of imaging strategies of the lymphatic system: Answer to the decades old questions. Microcirculation 2022; 29:e12780. [PMID: 35972391 DOI: 10.1111/micc.12780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/22/2022] [Accepted: 08/10/2022] [Indexed: 12/30/2022]
Abstract
The role of the lymphatic system in maintaining tissue homeostasis and a number of different pathophysiological conditions has been well established. The complex and delicate structure of the lymphatics along with the limitations of conventional imaging techniques make lymphatic imaging particularly difficult. Thus, in-depth high-resolution imaging of lymphatic system is key to understanding the progression of lymphatic diseases and cancer metastases and would greatly benefit clinical decisions. In recent years, the advancement of imaging technologies and development of new tracers suitable for clinical applications has enabled imaging of the lymphatic system in both clinical and pre-clinical settings. In this current review, we have highlighted the advantages and disadvantages of different modern techniques such as near infra-red spectroscopy (NIRS), positron emission tomography (PET), computed tomography (CT), magnetic resonance imaging (MRI) and fluorescence optical imaging, that has significantly impacted research in this field and has led to in-depth insights into progression of pathological states. This review also highlights the use of current imaging technologies, and tracers specific for immune cell markers to identify and track the immune cells in the lymphatic system that would help understand disease progression and remission in immune therapy regimen.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Sukanya Roy
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
21
|
Russell PS, Velivolu R, Maldonado Zimbrón VE, Hong J, Kavianinia I, Hickey AJR, Windsor JA, Phillips ARJ. Fluorescent Tracers for In Vivo Imaging of Lymphatic Targets. Front Pharmacol 2022; 13:952581. [PMID: 35935839 PMCID: PMC9355481 DOI: 10.3389/fphar.2022.952581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
The lymphatic system continues to gain importance in a range of conditions, and therefore, imaging of lymphatic vessels is becoming more widespread for research, diagnosis, and treatment. Fluorescent lymphatic imaging offers advantages over other methods in that it is affordable, has higher resolution, and does not require radiation exposure. However, because the lymphatic system is a one-way drainage system, the successful delivery of fluorescent tracers to lymphatic vessels represents a unique challenge. Each fluorescent tracer used for lymphatic imaging has distinct characteristics, including size, shape, charge, weight, conjugates, excitation/emission wavelength, stability, and quantum yield. These characteristics in combination with the properties of the target tissue affect the uptake of the dye into lymphatic vessels and the fluorescence quality. Here, we review the characteristics of visible wavelength and near-infrared fluorescent tracers used for in vivo lymphatic imaging and describe the various techniques used to specifically target them to lymphatic vessels for high-quality lymphatic imaging in both clinical and pre-clinical applications. We also discuss potential areas of future research to improve the lymphatic fluorescent tracer design.
Collapse
Affiliation(s)
- P. S. Russell
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - R. Velivolu
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - V. E. Maldonado Zimbrón
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - J. Hong
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - I. Kavianinia
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
- School of Chemical Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - A. J. R. Hickey
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - J. A. Windsor
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - A. R. J. Phillips
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
McCright J, Naiknavare R, Yarmovsky J, Maisel K. Targeting Lymphatics for Nanoparticle Drug Delivery. Front Pharmacol 2022; 13:887402. [PMID: 35721179 PMCID: PMC9203826 DOI: 10.3389/fphar.2022.887402] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/25/2022] Open
Abstract
The lymphatics transport material from peripheral tissues to lymph nodes, where immune responses are formed, before being transported into systemic circulation. With key roles in transport and fluid homeostasis, lymphatic dysregulation is linked to diseases, including lymphedema. Fluid within the interstitium passes into initial lymphatic vessels where a valve system prevents fluid backflow. Additionally, lymphatic endothelial cells produce key chemokines, such as CCL21, that direct the migration of dendritic cells and lymphocytes. As a result, lymphatics are an attractive delivery route for transporting immune modulatory treatments to lymph nodes where immunotherapies are potentiated in addition to being an alternative method of reaching systemic circulation. In this review, we discuss the physiology of lymphatic vessels and mechanisms used in the transport of materials from peripheral tissues to lymph nodes. We then summarize nanomaterial-based strategies to take advantage of lymphatic transport functions for delivering therapeutics to lymph nodes or systemic circulation. We also describe opportunities for targeting lymphatic endothelial cells to modulate transport and immune functions.
Collapse
Affiliation(s)
| | | | | | - Katharina Maisel
- Department of Bioengineering, University of Maryland College Park, College Park, MD, United States
| |
Collapse
|
23
|
Feng C, Li Y, Ferdows BE, Patel DN, Ouyang J, Tang Z, Kong N, Chen E, Tao W. Emerging vaccine nanotechnology: From defense against infection to sniping cancer. Acta Pharm Sin B 2022; 12:2206-2223. [PMID: 35013704 PMCID: PMC8730377 DOI: 10.1016/j.apsb.2021.12.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/24/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
Looking retrospectively at the development of humanity, vaccination is an unprecedented medical landmark that saves lives by harnessing the human immune system. During the ongoing coronavirus disease 2019 (COVID-19) pandemic, vaccination is still the most effective defense modality. The successful clinical application of the lipid nanoparticle-based Pfizer/BioNTech and Moderna mRNA COVID-19 vaccines highlights promising future of nanotechnology in vaccine development. Compared with conventional vaccines, nanovaccines are supposed to have advantages in lymph node accumulation, antigen assembly, and antigen presentation; they also have, unique pathogen biomimicry properties because of well-organized combination of multiple immune factors. Beyond infectious diseases, vaccine nanotechnology also exhibits considerable potential for cancer treatment. The ultimate goal of cancer vaccines is to fully mobilize the potency of the immune system as a living therapeutic to recognize tumor antigens and eliminate tumor cells, and nanotechnologies have the requisite properties to realize this goal. In this review, we summarize the recent advances in vaccine nanotechnology from infectious disease prevention to cancer immunotherapy and highlight the different types of materials, mechanisms, administration methods, as well as future perspectives.
Collapse
Affiliation(s)
- Chan Feng
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Bijan Emiliano Ferdows
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dylan Neal Patel
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jiang Ouyang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zhongmin Tang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Enguo Chen
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Corresponding authors. Fax: +001 857 307 2337 (Wei Tao).
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Corresponding authors. Fax: +001 857 307 2337 (Wei Tao).
| |
Collapse
|
24
|
Chen H, Liu H, Liu L, Chen Y. Fabrication of subunit nanovaccines by physical interaction. SCIENCE CHINA. TECHNOLOGICAL SCIENCES 2022; 65:989-999. [PMID: 35432491 PMCID: PMC9004205 DOI: 10.1007/s11431-021-2011-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
Vaccines can improve the quality of human life by preventing the burden of infectious diseases. Also, vaccination is becoming a powerful medication for preventing and treating tumors. Various vaccines have been developed based on the origin of the antigens. Herein, we focus on the subunit vaccines whose antigens are proteins or peptides. The advantage of subunit vaccines is safety for recipients; however, the immunogenicity of subunit antigens is relatively low. Nanoparticular delivery systems have been applied to improve the immunocompetence of subunit vaccines by targeting lymph nodes, and effectively present antigens to immune cells. Moreover, adding appropriate molecular adjuvants may strengthen the antigens to elicit immune response. In this perspective article, we first elucidate the characteristics of immunity induced by subunit nanovaccines and then summarize the strategies to fabricate subunit nanovaccines with delivering materials. Herein we highlight non-covalent interaction to fabricate nanoparticular subunit vaccines.
Collapse
Affiliation(s)
- HaoLin Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275 China
| | - Hong Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Jinan University, Zhuhai, 519000 China
| | - LiXin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275 China
| | - YongMing Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275 China
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630 China
| |
Collapse
|
25
|
Duan M, Han D, Shen W, Chang K, Wang X, Gao N, Du J. Preparation of the Biodegradable Lymphatic Targeting Imaging Agent Based on the Indocyanine Green Mesoporous Silicon System. Front Chem 2022; 10:847929. [PMID: 35273951 PMCID: PMC8902163 DOI: 10.3389/fchem.2022.847929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/21/2022] [Indexed: 12/04/2022] Open
Abstract
The lymphatic system plays a crucial role in the immune system’s recognition and response to disease. Therefore, the imaging of the lymphatic system, especially lymphatic vessels, has emerged as a valuable tool for the diagnosis of metastasis. FDA-approved small-molecule dyes, namely, indocyanine green (ICG), have been widely applied to lymphatic vessels imaging. However, due to the small physical size, such molecule-based agents show no selectivity, and rapid clearance from lymph nodes. Herein, a biodegradable lymphatic targeting imaging agent based on the ICG-mesoporous silicon system (ICG@HMONs-HA) was obtained, which not only could target lymph vessels but also had a long residence time. The reported work provides a practical way for lymph vessel fluorescence imaging and paves the way for clinical translation of nanomaterial-based tracers.
Collapse
Affiliation(s)
- Man Duan
- Key Laboratory of Lymphatic Surgery Jilin Province, Jilin Engineering Laboratory for Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dongmei Han
- Key Laboratory of Lymphatic Surgery Jilin Province, Jilin Engineering Laboratory for Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wenbin Shen
- Department of Lymphology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Kun Chang
- Department of Lymphology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinyu Wang
- Key Laboratory of Lymphatic Surgery Jilin Province, Jilin Engineering Laboratory for Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Nan Gao
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Faculty of Chemistry, Northeast Normal University, Changchun, China
- *Correspondence: Nan Gao, ; Jianshi Du,
| | - Jianshi Du
- Key Laboratory of Lymphatic Surgery Jilin Province, Jilin Engineering Laboratory for Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Nan Gao, ; Jianshi Du,
| |
Collapse
|
26
|
Ho MJ, Jeong MY, Jeong HT, Kim MS, Park HJ, Kim DY, Lee HC, Song WH, Kim CH, Lee CH, Choi YW, Choi YS, Han YT, Kang MJ. Effect of particle size on in vivo performances of long-acting injectable drug suspension. J Control Release 2021; 341:533-547. [PMID: 34902451 DOI: 10.1016/j.jconrel.2021.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/16/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022]
Abstract
Herein, entecavir-3-palmitate (EV-P), an ester prodrug of entecavir (EV), was employed as a model drug, and the effect of drug particle size on in vivo pharmacokinetic profiles and local inflammatory responses, and those associations were evaluated following intramuscular (IM) injection. EV-P crystals with different median diameters (0.8, 2.3, 6.3, 15.3 and 22.6 μm) were prepared using the anti-solvent crystallization method, with analogous surface charges (-10.7 ~ -4.7 mV), and crystallinity (melting point, 160-170 °C). EV-P particles showed size-dependent in vitro dissolution profiles under sink conditions, exhibiting a high correlation between the median diameter and Hixon-Crowell's release rate constant (r2 = 0.94). Following IM injection in rats (1.44 mg/kg as EV), the pharmacokinetic profile of EV exhibited marked size-dependency; 0.8 μm-sized EV-P particles about 1.6-, 3.6-, and 5.6-folds higher systemic exposure, compared to 6.3, 15.3, and 22.6 μm-sized particles, respectively. This pharmacokinetic pattern, depending on particle size, was also highly associated with histopathological responses in the injected tissue. The smaller EV-P particles (0.8 or 2.3 μm) imparted the larger inflammatory lesion after 3 days, lower infiltration of inflammatory cells, and thinner fibroblastic bands around depots after 4 weeks. Conversely, severe fibrous isolation with increasing particle size augmented the drug remaining at injection site over 4 weeks, impeding the dissolution and systemic exposure. These findings regarding the effects of formulation variable on the in vivo behaviors of long-acting injectable suspension, provide constructive knowledge toward the improved design in poorly water-soluble compounds.
Collapse
Affiliation(s)
- Myoung Jin Ho
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Min Young Jeong
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Hoe Taek Jeong
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Min Seob Kim
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Hyun Jin Park
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Dong Yoon Kim
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Hyo Chun Lee
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Woo Heon Song
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Chang Hyun Kim
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Republic of Korea
| | - Choong Hyun Lee
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Young Wook Choi
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Republic of Korea
| | - Yong Seok Choi
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Young Taek Han
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea
| | - Myung Joo Kang
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Republic of Korea.
| |
Collapse
|
27
|
Archer PA, Sestito LF, Manspeaker MP, O'Melia MJ, Rohner NA, Schudel A, Mei Y, Thomas SN. Quantitation of lymphatic transport mechanism and barrier influences on lymph node-resident leukocyte access to lymph-borne macromolecules and drug delivery systems. Drug Deliv Transl Res 2021; 11:2328-2343. [PMID: 34165731 PMCID: PMC8571034 DOI: 10.1007/s13346-021-01015-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 02/04/2023]
Abstract
Lymph nodes (LNs) are tissues of the immune system that house leukocytes, making them targets of interest for a variety of therapeutic immunomodulation applications. However, achieving accumulation of a therapeutic in the LN does not guarantee equal access to all leukocyte subsets. LNs are structured to enable sampling of lymph draining from peripheral tissues in a highly spatiotemporally regulated fashion in order to facilitate optimal adaptive immune responses. This structure results in restricted nanoscale drug delivery carrier access to specific leukocyte targets within the LN parenchyma. Herein, a framework is presented to assess the manner in which lymph-derived macromolecules and particles are sampled in the LN to reveal new insights into how therapeutic strategies or drug delivery systems may be designed to improve access to dLN-resident leukocytes. This summary analysis of previous reports from our group assesses model nanoscale fluorescent tracer association with various leukocyte populations across relevant time periods post administration, studies the effects of bioactive molecule NO on access of lymph-borne solutes to dLN leukocytes, and illustrates the benefits to leukocyte access afforded by lymphatic-targeted multistage drug delivery systems. Results reveal trends consistent with the consensus view of how lymph is sampled by LN leukocytes resulting from tissue structural barriers that regulate inter-LN transport and demonstrate how novel, engineered delivery systems may be designed to overcome these barriers to unlock the therapeutic potential of LN-resident cells as drug delivery targets.
Collapse
Affiliation(s)
- Paul A Archer
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Lauren F Sestito
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Margaret P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Meghan J O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Nathan A Rohner
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, GA, 30332, Atlanta, USA
| | - Alex Schudel
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- School of Materials Science & Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Yajun Mei
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, GA, 30332, Atlanta, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
28
|
Kim J, Archer PA, Thomas SN. Innovations in lymph node targeting nanocarriers. Semin Immunol 2021; 56:101534. [PMID: 34836772 DOI: 10.1016/j.smim.2021.101534] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022]
Abstract
Lymph nodes are secondary lymphoid tissues in the body that facilitate the co-mingling of immune cells to enable and regulate the adaptive immune response. They are also tissues implicated in a variety of diseases, including but not limited to malignancy. The ability to access lymph nodes is thus attractive for a variety of therapeutic and diagnostic applications. As nanotechnologies are now well established for their potential in translational biomedical applications, their high relevance to applications that involve lymph nodes is highlighted. Herein, established paradigms of nanocarrier design to enable delivery to lymph nodes are discussed, considering the unique lymph node tissue structure as well as lymphatic system physiology. The influence of delivery mechanism on how nanocarrier systems distribute to different compartments and cells that reside within lymph nodes is also elaborated. Finally, current advanced nanoparticle technologies that have been developed to enable lymph node delivery are discussed.
Collapse
Affiliation(s)
- Jihoon Kim
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Paul A Archer
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Emory University, 201 Dowman Drive, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road NE, Atlanta, GA 30322, USA.
| |
Collapse
|
29
|
Maulloo CD, Cao S, Watkins EA, Raczy MM, Solanki AS, Nguyen M, Reda JW, Shim HN, Wilson DS, Swartz MA, Hubbell JA. Lymph Node-Targeted Synthetically Glycosylated Antigen Leads to Antigen-Specific Immunological Tolerance. Front Immunol 2021; 12:714842. [PMID: 34630389 PMCID: PMC8498032 DOI: 10.3389/fimmu.2021.714842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Inverse vaccines that tolerogenically target antigens to antigen-presenting cells (APCs) offer promise in prevention of immunity to allergens and protein drugs and treatment of autoimmunity. We have previously shown that targeting hepatic APCs through intravenous injection of synthetically glycosylated antigen leads to effective induction of antigen-specific immunological tolerance. Here, we demonstrate that targeting these glycoconjugates to lymph node (LN) APCs under homeostatic conditions leads to local and increased accumulation in the LNs compared to unmodified antigen and induces a tolerogenic state both locally and systemically. Subcutaneous administration directs the polymeric glycoconjugate to the draining LN, where the glycoconjugated antigen generates robust antigen-specific CD4+ and CD8+ T cell tolerance and hypo-responsiveness to antigenic challenge via a number of mechanisms, including clonal deletion, anergy of activated T cells, and expansion of regulatory T cells. Lag-3 up-regulation on CD4+ and CD8+ T cells represents an essential mechanism of suppression. Additionally, presentation of antigen released from the glycoconjugate to naïve T cells is mediated mainly by LN-resident CD8+ and CD11b+ dendritic cells. Thus, here we demonstrate that antigen targeting via synthetic glycosylation to impart affinity for APC scavenger receptors generates tolerance when LN dendritic cells are the cellular target.
Collapse
Affiliation(s)
- Chitavi D. Maulloo
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Michal M. Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Ani. S. Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, United States
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Joseph W. Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - D. Scott Wilson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, United States
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Committee on Immunology, University of Chicago, Chicago, IL, United States
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Committee on Immunology, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
30
|
Rezzola S, Sigmund EC, Halin C, Ronca R. The lymphatic vasculature: An active and dynamic player in cancer progression. Med Res Rev 2021; 42:576-614. [PMID: 34486138 PMCID: PMC9291933 DOI: 10.1002/med.21855] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/29/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022]
Abstract
The lymphatic vasculature has been widely described and explored for its key functions in fluid homeostasis and in the organization and modulation of the immune response. Besides transporting immune cells, lymphatic vessels play relevant roles in tumor growth and tumor cell dissemination. Cancer cells that have invaded into afferent lymphatics are propagated to tumor‐draining lymph nodes (LNs), which represent an important hub for metastatic cell arrest and growth, immune modulation, and secondary dissemination to distant sites. In recent years many studies have reported new mechanisms by which the lymphatic vasculature affects cancer progression, ranging from induction of lymphangiogenesis to metastatic niche preconditioning or immune modulation. In this review, we provide an up‐to‐date description of lymphatic organization and function in peripheral tissues and in LNs and the changes induced to this system by tumor growth and progression. We will specifically focus on the reported interactions that occur between tumor cells and lymphatic endothelial cells (LECs), as well as on interactions between immune cells and LECs, both in the tumor microenvironment and in tumor‐draining LNs. Moreover, the most recent prognostic and therapeutic implications of lymphatics in cancer will be reported and discussed in light of the new immune‐modulatory roles that have been ascribed to LECs.
Collapse
Affiliation(s)
- Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elena C Sigmund
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
31
|
Fries CN, Chen JL, Dennis ML, Votaw NL, Eudailey J, Watts BE, Hainline KM, Cain DW, Barfield R, Chan C, Moody MA, Haynes BF, Saunders KO, Permar SR, Fouda GG, Collier JH. HIV envelope antigen valency on peptide nanofibers modulates antibody magnitude and binding breadth. Sci Rep 2021; 11:14494. [PMID: 34262096 PMCID: PMC8280189 DOI: 10.1038/s41598-021-93702-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/23/2021] [Indexed: 01/02/2023] Open
Abstract
A major challenge in developing an effective vaccine against HIV-1 is the genetic diversity of its viral envelope. Because of the broad range of sequences exhibited by HIV-1 strains, protective antibodies must be able to bind and neutralize a widely mutated viral envelope protein. No vaccine has yet been designed which induces broadly neutralizing or protective immune responses against HIV in humans. Nanomaterial-based vaccines have shown the ability to generate antibody and cellular immune responses of increased breadth and neutralization potency. Thus, we have developed supramolecular nanofiber-based immunogens bearing the HIV gp120 envelope glycoprotein. These immunogens generated antibody responses that had increased magnitude and binding breadth compared to soluble gp120. By varying gp120 density on nanofibers, we determined that increased antigen valency was associated with increased antibody magnitude and germinal center responses. This study presents a proof-of-concept for a nanofiber vaccine platform generating broad, high binding antibody responses against the HIV-1 envelope glycoprotein.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maria L Dennis
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Nicole L Votaw
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Joshua Eudailey
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brian E Watts
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin O Saunders
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, New York-Presbyterian/Weill Cornell Medicine, New York, NY, 10065, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
32
|
Song T, Xia Y, Du Y, Chen MW, Qing H, Ma G. Engineering the Deformability of Albumin-Stabilized Emulsions for Lymph-Node Vaccine Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2100106. [PMID: 34013604 DOI: 10.1002/adma.202100106] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/12/2021] [Indexed: 06/12/2023]
Abstract
A major challenge in vaccine delivery is to achieve robust lymph-node (LN) accumulation, which can capitalize on concentrated immunocytes and cytokines in LNs to stimulate the onset and persistence of adaptive immune responses. Previous attempts at developing vaccine delivery systems have focused on the sizes, charges, or surface ligands but not on their deformability. In fact, the LN homing of antigen-presenting cells depends on deformability to pass through the cellular gaps. Herein, the deformability of albumin-stabilized emulsions is engineered. Owing to self-adaptive deformability, the droplets (≈330 nm) can attach to and deform between cells and adjust their sizes to pass through the endothelial gaps (20-100 nm), favoring direct LN transfer (intercellular pathway). Additionally, owing to relatively large sizes, some emulsions can be retained at the administration sites for potent antigen uptake and activation of APCs as well as LN-targeted delivery of vaccines (intracellular pathway). Compared with solid particles, the dual LN transfer strategy evidently enhances antigen accumulation and activation of LN drainage, potently stimulates cellular immune responses, and increases the survival rate of tumor-bearing mice. Thus, the deformability of albumin-stabilized droplets may offer an efficient strategy for potent LN targeting and enhanced vaccinations.
Collapse
Affiliation(s)
- Tiantian Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yufei Xia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yiqun Du
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, P. R. China
| | - Michael W Chen
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, P. R. China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| |
Collapse
|
33
|
Qin L, Zhang H, Zhou Y, Umeshappa CS, Gao H. Nanovaccine-Based Strategies to Overcome Challenges in the Whole Vaccination Cascade for Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006000. [PMID: 33768693 DOI: 10.1002/smll.202006000] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/26/2020] [Indexed: 06/12/2023]
Abstract
Nanovaccine-based immunotherapy (NBI) has received greater attention recently for its potential to prime tumor-specific immunity and establish a long-term immune memory that prevents tumor recurrence. Despite encouraging results in the recent studies, there are still numerous challenges to be tackled for eliciting potent antitumor immunity using NBI strategies. Based on the principles that govern immune response, here it is proposed that these challenges need to be addressed at the five critical cascading events: Loading tumor-specific antigens by nanoscale drug delivery systems (L); Draining tumor antigens to lymph nodes (D); Internalization by dendritic cells (DCs) (I); Maturation of DCs by costimulatory signaling (M); and Presenting tumor-peptide-major histocompatibility complexes to T cells (P) (LDIMP cascade in short). This review provides a detailed and objective overview of emerging NBI strategies to improve the efficacy of nanovaccines in each step of the LDIMP cascade. It is concluded that the balance between each step must be optimized by delicate designing and modification of nanovaccines and by combining with complementary approaches to provide a synergistic immunity in the fight against cancer.
Collapse
Affiliation(s)
- Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Chongqing Vocational College of Transportation, Chongqing, 400715, China
| | - Huilin Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Channakeshava Sokke Umeshappa
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
34
|
Wu H, Fu X, Zhai Y, Gao S, Yang X, Zhai G. Development of Effective Tumor Vaccine Strategies Based on Immune Response Cascade Reactions. Adv Healthc Mater 2021; 10:e2100299. [PMID: 34021717 DOI: 10.1002/adhm.202100299] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/11/2021] [Indexed: 12/13/2022]
Abstract
To solve the problems of high toxicity and poor efficacy of existing tumor treatment methods, researchers have developed a variety of tumor immunotherapies. Among them, tumor vaccines activate antigen-presenting cells and T lymphocytes upstream of the cancer-immunity cycle are considered the most promising therapy to activate the immune system. Nanocarriers are considered the most promising tumor vaccine delivery vehicles, including polymer nanocarriers, lipid nanocarriers, inorganic nanocarriers, and biomimetic nanocarriers that have been developed for vaccine delivery. Based on the cascade reaction for tumor vaccines to exert their effects, this review summarizes the four key factors for the design and construction of nano-tumor vaccines. The composition and functional characteristics of the corresponding preferred nanocarriers are illustrated to provide a reference for the development of effective tumor vaccines. Finally, potential challenges and perspectives are illustrated in the hope of improving the efficacy of tumor vaccine immunotherapy and accelerating the clinical transformation of next-generation tumor vaccines.
Collapse
Affiliation(s)
- Hang Wu
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Xianglei Fu
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry University of Utah Salt Lake City UT 84124 USA
| | - Shan Gao
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Xiaoye Yang
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| |
Collapse
|
35
|
Fries CN, Curvino EJ, Chen JL, Permar SR, Fouda GG, Collier JH. Advances in nanomaterial vaccine strategies to address infectious diseases impacting global health. NATURE NANOTECHNOLOGY 2021; 16:1-14. [PMID: 32807876 DOI: 10.1038/s41565-020-0739-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 06/23/2020] [Indexed: 05/18/2023]
Abstract
Despite the overwhelming success of vaccines in preventing infectious diseases, there remain numerous globally devastating diseases without fully protective vaccines, particularly human immunodeficiency virus (HIV), malaria and tuberculosis. Nanotechnology approaches are being developed both to design new vaccines against these diseases as well as to facilitate their global implementation. The reasons why a given pathogen may present difficulties for vaccine design are unique and tied to the co-evolutionary history of the pathogen and humans, but there are common challenges that nanotechnology is beginning to help address. In each case, a successful vaccine will need to raise immune responses that differ from the immune responses raised by normal infection. Nanomaterials, with their defined compositions, commonly modular construction, and length scales allowing the engagement of key immune pathways, collectively facilitate the iterative design process necessary to identify such protective immune responses and achieve them reliably. Nanomaterials also provide strategies for engineering the trafficking and delivery of vaccine components to key immune cells and lymphoid tissues, and they can be highly multivalent, improving their engagement with the immune system. This Review will discuss these aspects along with recent nanomaterial advances towards vaccines against infectious disease, with a particular emphasis on HIV/AIDS, malaria and tuberculosis.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
36
|
Polomska AK, Proulx ST. Imaging technology of the lymphatic system. Adv Drug Deliv Rev 2021; 170:294-311. [PMID: 32891679 DOI: 10.1016/j.addr.2020.08.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/16/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022]
Abstract
The lymphatic system plays critical roles in tissue fluid homeostasis and immunity and has been implicated in the development of many different pathologies, ranging from lymphedema, the spread of cancer to chronic inflammation. In this review, we first summarize the state-of-the-art of lymphatic imaging in the clinic and the advantages and disadvantages of these existing techniques. We then detail recent progress on imaging technology, including advancements in tracer design and injection methods, that have allowed visualization of lymphatic vessels with excellent spatial and temporal resolution in preclinical models. Finally, we describe the different approaches to quantifying lymphatic function that are being developed and discuss some emerging topics for lymphatic imaging in the clinic. Continued advancements in lymphatic imaging technology will be critical for the optimization of diagnostic methods for lymphatic disorders and the evaluation of novel therapies targeting the lymphatic system.
Collapse
Affiliation(s)
- Anna K Polomska
- ETH Zürich, Institute of Pharmaceutical Sciences, Vladimir-Prelog Weg 1-5/10, 8093 Zürich, Switzerland
| | - Steven T Proulx
- University of Bern, Theodor Kocher Institute, Freiestrasse 1, 3012 Bern, Switzerland.
| |
Collapse
|
37
|
Abstract
The lymphatic system has received increasing scientific and clinical attention because a wide variety of diseases are linked to lymphatic pathologies and because the lymphatic system serves as an ideal conduit for drug delivery. Lymphatic vessels exert heterogeneous roles in different organs and vascular beds, and consequently, their dysfunction leads to distinct organ-specific outcomes. Although studies in animal model systems have led to the identification of crucial lymphatic genes with potential therapeutic benefit, effective lymphatic-targeted therapeutics are currently lacking for human lymphatic pathological conditions. Here, we focus on the therapeutic roles of lymphatic vessels in diseases and summarize the promising therapeutic targets for modulating lymphangiogenesis or lymphatic function in preclinical or clinical settings. We also discuss considerations for drug delivery or targeting of lymphatic vessels for treatment of lymphatic-related diseases. The lymphatic vasculature is rapidly emerging as a critical system for targeted modulation of its function and as a vehicle for innovative drug delivery.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA;
| | - Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA;
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA;
| |
Collapse
|
38
|
O'Melia MJ, Manspeaker MP, Thomas SN. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol Immunother 2021; 70:2179-2195. [PMID: 33459842 DOI: 10.1007/s00262-020-02792-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/07/2020] [Indexed: 12/21/2022]
Abstract
Triple negative breast cancer (TNBC) is a significant clinical problem to which immunotherapeutic strategies have been applied with limited success. Using the syngeneic E0771 TNBC mouse model, this work explores the potential for antitumor CD8+ T cell immunity to be primed extratumorally in lymphoid tissues and therapeutically leveraged. CD8+ T cell viability and responses within the tumor microenvironment (TME) were found to be severely impaired, effects coincident with local immunosuppression that is recapitulated in lymphoid tissues in late stage disease. Prior to onset of a locally suppressed immune microenvironment, however, CD8+ T cell priming within lymph nodes (LN) that depended on tumor lymphatic drainage remained intact. These results demonstrate tumor-draining LNs (TdLN) to be lymphoid tissue niches that support the survival and antigenic priming of CD8+ T lymphocytes against lymph-draining antigen. The therapeutic effects of and CD8+ T cells response to immune checkpoint blockade were furthermore improved when directed to LNs within the tumor-draining lymphatic basin. Therefore, TdLNs represent a unique potential tumor immunity reservoir in TNBC for which strategies may be developed to improve the effects of ICB immunotherapy.
Collapse
Affiliation(s)
- Meghan J O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, IBB 2310, 315 Ferst Drive NW, Atlanta, GA, 30332, USA
| | - Margaret P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, IBB 2310, 315 Ferst Drive NW, Atlanta, GA, 30332, USA. .,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Winship Cancer Institute, Emory University, Atlanta, GA, 30332, USA.
| |
Collapse
|
39
|
Gomi M, Sakurai Y, Okada T, Miura N, Tanaka H, Akita H. Development of Sentinel LN Imaging with a Combination of HAase Based on a Comprehensive Analysis of the Intra-lymphatic Kinetics of LPs. Mol Ther 2021; 29:225-235. [PMID: 32966771 PMCID: PMC7791005 DOI: 10.1016/j.ymthe.2020.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/15/2020] [Accepted: 09/03/2020] [Indexed: 02/08/2023] Open
Abstract
The sentinel lymph node (LN) is the first LN to which lymph fluid flows from tumor tissue. We identified the key parameters of liposomes (LPs) that affect their accumulation in regional (primary) LNs with minimum leakage to its connecting (secondary) LNs by a comprehensive analysis of the LN-to-LN trafficking of LPs with various surface charges and various sizes. We used a lymphatic flow-modified (LFM) mouse that allows for the chronological analysis of inguinal (primary) LN-to-axillary (secondary) LN at the body surface. As a result, the anionic medium-sized LPs (130 nm on average) exhibited the highest accumulation in the primary LNs. A mechanism-based analysis revealed that CD169-positive macrophages in LNs were the dominant cell population that captures anionic LPs. Sentinel LN imaging was also performed by the intratumoral injection of fluorescent medium-sized anionic LPs using a breast cancer orthotopic model. In comparison with the typically used contrast agent indocyanine green, the anionic LPs were detected in sentinel LNs with a high sensitivity. Additionally, the co-injection of hyaluronidase significantly improved the sensitivity of detection of the fluorescent LPs in sentinel LNs. In conclusion, medium-sized anionic LPs combined with hyaluronidase represents a potent strategy for investigating sentinel LNs.
Collapse
Affiliation(s)
- Masaki Gomi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 264-0028, Japan
| | - Yu Sakurai
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 264-0028, Japan.
| | - Takaharu Okada
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Naoya Miura
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 264-0028, Japan
| | - Hiroki Tanaka
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 264-0028, Japan
| | - Hidetaka Akita
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 264-0028, Japan.
| |
Collapse
|
40
|
Wibowo D, Jorritsma SHT, Gonzaga ZJ, Evert B, Chen S, Rehm BHA. Polymeric nanoparticle vaccines to combat emerging and pandemic threats. Biomaterials 2020; 268:120597. [PMID: 33360074 PMCID: PMC7834201 DOI: 10.1016/j.biomaterials.2020.120597] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/30/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Subunit vaccines are more advantageous than live attenuated vaccines in terms of safety and scale-up manufacture. However, this often comes as a trade-off to their efficacy. Over the years, polymeric nanoparticles have been developed to improve vaccine potency, by engineering their physicochemical properties to incorporate multiple immunological cues to mimic pathogenic microbes and viruses. This review covers recent advances in polymeric nanostructures developed toward particulate vaccines. It focuses on the impact of microbe mimicry (e.g. size, charge, hydrophobicity, and surface chemistry) on modulation of the nanoparticles’ delivery, trafficking, and targeting antigen-presenting cells to elicit potent humoral and cellular immune responses. This review also provides up-to-date progresses on rational designs of a wide variety of polymeric nanostructures that are loaded with antigens and immunostimulatory molecules, ranging from particles, micelles, nanogels, and polymersomes to advanced core-shell structures where polymeric particles are coated with lipids, cell membranes, or proteins.
Collapse
Affiliation(s)
- David Wibowo
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia.
| | - Sytze H T Jorritsma
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Zennia Jean Gonzaga
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Benjamin Evert
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Shuxiong Chen
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan QLD, 4111, Australia.
| |
Collapse
|
41
|
O'Melia MJ, Rohner NA, Manspeaker MP, Francis DM, Kissick HT, Thomas SN. Quality of CD8 + T cell immunity evoked in lymph nodes is compartmentalized by route of antigen transport and functional in tumor context. SCIENCE ADVANCES 2020; 6:eabd7134. [PMID: 33310857 PMCID: PMC7732197 DOI: 10.1126/sciadv.abd7134] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/29/2020] [Indexed: 05/05/2023]
Abstract
Revealing the mechanisms that underlie the expansion of antitumor CD8+ T cells that are associated with improved clinical outcomes is critical to improving immunotherapeutic management of melanoma. How the lymphatic system, which orchestrates the complex sensing of antigen by lymphocytes to mount an adaptive immune response, facilitates this response in the context of malignancy is incompletely understood. To delineate the effects of lymphatic transport and tumor-induced lymphatic and lymph node (LN) remodeling on the elicitation of CD8+ T cell immunity within LNs, we designed a suite of nanoscale biomaterial tools enabling the quantification of antigen access and presentation within the LN and resulting influence on T cell functions. The expansion of antigen-specific stem-like and cytotoxic CD8+ T cell pools was revealed to be sensitive to the mechanism of lymphatic transport to LNs, demonstrating the potential for nanoengineering strategies targeting LNs to optimize cancer immunotherapy in eliciting antitumor CD8+ T cell immunity.
Collapse
Affiliation(s)
- M J O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - N A Rohner
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - M P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - D M Francis
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - H T Kissick
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - S N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
42
|
Affinity-Based Polymers Provide Long-Term Immunotherapeutic Drug Delivery Across Particle Size Ranges Optimal for Macrophage Targeting. J Pharm Sci 2020; 110:1693-1700. [PMID: 33127427 DOI: 10.1016/j.xphs.2020.10.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/19/2020] [Accepted: 10/23/2020] [Indexed: 11/23/2022]
Abstract
Drug delivery to specific arms of the immune system can be technically challenging to provide prolonged drug release while limiting off-target toxicity given the limitations of current drug delivery systems. In this work, we test the design of a cyclodextrin (CD) polymer platform to extend immunomodulatory drug delivery via affinity interactions for sustained release at multiple size scales. The parameter space of synthesis variables influencing particle nucleation and growth (pre-incubation time and stirring speed) and post-synthesis grinding effects on resulting particle diameter were characterized. We demonstrate that polymerized CD forms exhibit size-independent release profiles of the small molecule drug lenalidomide (LND) and can provide improved drug delivery profiles versus macro-scale CD polymer disks in part due to increased loading efficiency. CD polymer microparticles and smaller, ground particles demonstrated no significant cytotoxicity as compared to the base CD monomer when co-incubated with fibroblasts. Uptake of ground CD particles was significantly higher following incubation with RAW 264.7 macrophages in culture over standard CD microparticles. Thus, the affinity/structure properties afforded by polymerized CD allow particle size to be modified to affect cellular uptake profiles independently of drug release rate for applications in cell-targeted drug delivery.
Collapse
|
43
|
Chen Y, De Koker S, De Geest BG. Engineering Strategies for Lymph Node Targeted Immune Activation. Acc Chem Res 2020; 53:2055-2067. [PMID: 32910636 DOI: 10.1021/acs.accounts.0c00260] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Development of vaccine technology that induces long lasting and potent adaptive immune responses is of vital importance to combat emerging pathogens and to design the next generation of cancer immunotherapies. Advanced biomaterials such as nanoparticle carriers are intensively explored to increase the efficacy and safety of vaccines and immunotherapies, based on their intrinsic potential to focus the therapeutic payload onto the relevant immune cells and to limit systemic distribution. With adaptive immune responses being primarily initiated in lymph nodes, the potency of nanoparticle vaccines in turn is tightly linked to their capacity to reach and accumulate in the lymph nodes draining the immunization site. Here, we discuss the main strategies applied to increase nanoparticle delivery to lymph nodes: (1) direct lymph node injection, (2) active cell-mediated transport through targeting of peripheral dendritic cells, and (3) exploiting passive transport through the afferent lymphatics.The intralymph nodal injection is obviously the most direct way for nanoparticles to reach lymph nodes, and multiple studies have demonstrated its capability in enhancing immunostimulant drugs' immune activation and increasing the therapeutic window. However, the requirement of using ultrasound guidance for mapping lymph nodes in patients renders intranodal administration unsuited for mass vaccination campaigns. As lymph nodes are fine structured organs with lymphocytes and chemokine gradients arrayed in a highly ordered fashion, the breakdown of such formats by the intralymph nodal injection is another concern. The exploitation of dendritic cells as live vectors for transporting nanoparticles to lymph nodes has intensively been studied both ex vivo and in vivo. While ex vivo engineering of dendritic cells in theory can achieve 100% dendritic cell-specific selectivity, a scenario impossible to be achieved in vivo, this procedure is usually laborious and complicated and entails the participation of professional staff and equipment. In addition, the poor efficiency of dendritic cell migration to the draining lymph node is another significant limitation following the injection of ex vivo cultured dendritic cells. Thus, in vivo targeting of surface receptors, particularly C-type lectin receptors, on dendritic cells by conjugating nanoparticles with antibodies or ligands is intensively studied by both academia and industry. Although such nanoparticles in vivo still face nonspecific engulfment by various phagocytes, multiple studies have shown its feasibility in targeting dendritic cells with high selectivity. Moreover, through optimizing the physicochemical properties of nanoparticles, nanoparticles can passively drain to lymph nodes carried by the interstitial flow. Compared to dendritic cell-mediated transport, passive draining is much faster and of higher efficiency. Of all such properties, size is the most important parameter as large particles (>500 nm) can only reach lymph nodes by an active cell-mediated transport. Other surface properties, such as the charge and the balance of hydrophobicity-vs-hydrophilicity, strongly influence the mobility of nanoparticles in the extracellular space. In addition, albumin, a natural fatty acid transporter, has recently been demonstrated capable of binding the amphiphiles through their lipid moiety and subsequent transporting them to lymph nodes.
Collapse
Affiliation(s)
- Yong Chen
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghemt, Belgium
| | | | - Bruno G. De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghemt, Belgium
| |
Collapse
|
44
|
Lee SN, Jin SM, Shin HS, Lim YT. Chemical Strategies to Enhance the Therapeutic Efficacy of Toll-like Receptor Agonist Based Cancer Immunotherapy. Acc Chem Res 2020; 53:2081-2093. [PMID: 32966047 DOI: 10.1021/acs.accounts.0c00337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent developments in the fields of biomedical chemistry and immune bioengineering have enabled innovative therapeutic approaches that can enhance the efficacy, accuracy, and safety of cancer immunotherapy. Among the numerous strategies utilized in cancer immunotherapy, Toll-like receptor (TLR) agonist-based approaches have been studied for a long time since they trigger the innate immune system and generate antigen-specific T cell responses to fight against tumors. In addition to these immunostimulatory functions, TLR agonists also contribute to the reprogramming of immune suppressive tumor microenvironments. Although TLR agonists are now being intensively studied in clinical trials due to their substantial immunomodulatory properties, they still show a low therapeutic index. Nonspecific and random stimulation of various immune cells produces excess levels of proinflammatory cytokines, resulting in cytokine storms and chronic diseases. Therefore, the development of chemical strategies to enhance the therapeutic efficacy as well as the safety of TLR agonist-based immunotherapy is essential and in high demand.In this Account, we summarize and discuss recent developments in biomedical chemistry and bioengineering techniques for the immunomodulation of TLR agonists that have addressed the limitations in current cancer immunotherapy. Immunomodulation of TLR agonists can be classified into two different approaches: (1) molecular modulation via chemical structure modification and (2) macroscopic modulation via an engineered drug delivery system. In molecular modulation, based on prodrug and antedrug principles, activity is modulated (active or inactive) through immolative chemical linkers that can respond to extrinsic or intrinsic biological stimulation and the plasmatic environment, respectively. To increase the effectiveness of TLR agonists as immunostimulatory agents, researchers have conjugated TLR agonists with other immunotherapeutic moieties (antigen, antibody, other TLR agonist, etc.). For macroscopic modulation, bioengineering of delivery carriers differing in size or with albumin hitchhiking moieties has been utilized to increase the efficiency of the targeting of these carriers to secondary lymphoid organs (lymph nodes (LNs) and spleen). The conjugation of specific targeting ligands and incorporation of stimulus-triggering moieties can promote the delivery of TLR agonists into specific cells or intracellular compartments. Implantable porous scaffolds for specific immune cell recruitment and in situ depot-forming gel systems for controlled release of immunomodulatory drugs can increase the therapeutic efficacy of TLR agonists while reducing systemic toxicity. Taken together, these findings show that well-designed and precisely controlled chemical strategies for the immunomodulation of TLR agonists at both the molecular and macroscopic levels are expected to play key roles in improving the therapeutic efficacy of cancer immunotherapy while minimizing immune-related toxicity.
Collapse
Affiliation(s)
- Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| |
Collapse
|
45
|
Manspeaker MP, Thomas SN. Lymphatic immunomodulation using engineered drug delivery systems for cancer immunotherapy. Adv Drug Deliv Rev 2020; 160:19-35. [PMID: 33058931 PMCID: PMC7736326 DOI: 10.1016/j.addr.2020.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Though immunotherapy has revolutionized the treatment of cancer to improve disease outcomes, an array of challenges remain that limit wider clinical success, including low rate of response and immune-related adverse events. Targeting immunomodulatory drugs to therapeutically relevant tissues offers a way to overcome these challenges by potentially enabling enhanced therapeutic efficacy and decreased incidence of side effects. Research highlighting the importance of lymphatic tissues in the response to immunotherapy has increased interest in the application of engineered drug delivery systems (DDSs) to enable specific targeting of immunomodulators to lymphatic tissues and cells that they house. To this end, a variety of DDS platforms have been developed that enable more efficient uptake into lymphatic vessels and lymph nodes to provide targeted modulation of the immune response to cancer. This can occur either by delivery of immunotherapeutics to lymphatics tissues or by direct modulation of the lymphatic vasculature itself due to their direct involvement in tumor immune processes. This review will highlight DDS platforms that, by enabling the activities of cancer vaccines, chemotherapeutics, immune checkpoint blockade (ICB) antibodies, and anti- or pro-lymphangiogenic factors to lymphatic tissues through directed delivery and controlled release, augment cancer immunotherapy.
Collapse
Affiliation(s)
- Margaret P Manspeaker
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States of America.
| |
Collapse
|
46
|
Lymph-directed immunotherapy - Harnessing endogenous lymphatic distribution pathways for enhanced therapeutic outcomes in cancer. Adv Drug Deliv Rev 2020; 160:115-135. [PMID: 33039497 DOI: 10.1016/j.addr.2020.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/07/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
The advent of immunotherapy has revolutionised the treatment of some cancers. Harnessing the immune system to improve tumour cell killing is now standard clinical practice and immunotherapy is the first line of defence for many cancers that historically, were difficult to treat. A unifying concept in cancer immunotherapy is the activation of the immune system to mount an attack on malignant cells, allowing the body to recognise, and in some cases, eliminate cancer. However, in spite of a significant proportion of patients that respond well to treatment, there remains a subset who are non-responders and a number of cancers that cannot be treated with these therapies. These limitations highlight the need for targeted delivery of immunomodulators to both tumours and the effector cells of the immune system, the latter being highly concentrated in the lymphatic system. In this context, macromolecular therapies may provide a significant advantage. Macromolecules are too large to easily access blood capillaries and instead typically exhibit preferential uptake via the lymphatic system. In contexts where immune cells are the therapeutic target, particularly in cancer therapy, this may be advantageous. In this review, we examine in brief the current immunotherapy approaches in cancer and how macromolecular and nanomedicine strategies may improve the therapeutic profiles of these drugs. We subsequently discuss how therapeutics directed either by parenteral or mucosal administration, can be taken up by the lymphatics thereby accessing a larger proportion of the body's immune cells. Finally, we detail drug delivery strategies that have been successfully employed to target the lymphatics.
Collapse
|
47
|
Yukuyama MN, de Araujo GLB, de Souza A, Löbenberg R, Barbosa EJ, Henostroza MAB, Rocha NPD, de Oliveira IF, Folchini BR, Peroni CM, Masiero JF, Bou-Chacra NA. Cancer treatment in the lymphatic system: A prospective targeting employing nanostructured systems. Int J Pharm 2020; 587:119697. [PMID: 32750440 DOI: 10.1016/j.ijpharm.2020.119697] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022]
Abstract
Cancer related to lymphangiogenesis has gained a great deal of attention in recent decades ever since specific markers of this intriguing system were discovered. Unlike the blood system, the lymphatic system has unique features that can advance cancer in future metastasis, or, conversely, can provide an opportunity to prevent or treat this disease that affects people worldwide. The aim of this review is to show the recent research of cancer treatment associated with the lymphatic system, considered one of the main gateways for disseminating metastatic cells to distant organs. Nanostructured systems based on theranostics and immunotherapies can offer several options for this complex disease. Precision targeting and accumulation of nanomaterials into the tumor sites and their elimination, or targeting the specific immune defense cells to promote optimal regression of cancer cells are highlighted in this paper. Moreover, therapies based on nanostructured systems through lymphatic systems may reduce the side effects and toxicity, avoid first pass hepatic metabolism, and improve patient recovery. We emphasize the general understanding of the association between the immune and lymphatic systems, their interaction with tumor cells, the mechanisms involved and the recent developments in several nanotechnology treatments related to this disease.
Collapse
Affiliation(s)
- Megumi Nishitani Yukuyama
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Gabriel Lima Barros de Araujo
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil.
| | - Aline de Souza
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Raimar Löbenberg
- Division of Pharmaceutical Sciences, Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy & Health Research, University of Alberta, 11361 - 87 Avenue, Room 3-142-K, Edmonton, AB T6G 2E1, Canada
| | - Eduardo José Barbosa
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Mirla Anali Bazán Henostroza
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Nataly Paredes da Rocha
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Isabela Fernandes de Oliveira
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Beatriz Rabelo Folchini
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Camilla Midori Peroni
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Jessica Fagionato Masiero
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil
| | - Nádia Araci Bou-Chacra
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Sao Paulo, Avenida Professor Lineu Prestes 508, Butantan, Sao Paulo, SP, Brazil.
| |
Collapse
|
48
|
Lymph-directed nitric oxide increases immune cell access to lymph-borne nanoscale solutes. Biomaterials 2020; 265:120411. [PMID: 33080460 DOI: 10.1016/j.biomaterials.2020.120411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
Lymph nodes (LNs) are immune organs housing high concentrations of lymphocytes, making them critical targets for therapeutic immunomodulation in a wide variety of diseases. While there is great interest in targeted drug delivery to LNs, many nanoscale drug delivery carriers have limited access to parenchymal resident immune cells compared to small molecules, limiting their efficacy. Nitric oxide (NO) is a potent regulator of vascular and lymphatic transport and a promising candidate for modulating nanocarrier access to LNs, but its lymphatic accumulation is limited by its low molecular weight and high reactivity. In this work, we employ S-nitrosated nanoparticles (SNO-NP), a lymphatic-targeted delivery system for controlled NO release, to investigate the effect of NO application on molecule accumulation and distribution within the LN. We evaluated the LN accumulation, spatial distribution, and cellular distribution of a panel of fluorescent tracers after intradermal administration alongside SNO-NP or a small molecule NO donor. While SNO-NP did not alter total tracer accumulation in draining lymph nodes (dLNs) or affect active cellular transport of large molecules from the injection site, its application enhanced the penetration of nanoscale 30 nm dextrans into the LN and their subsequent uptake by LN-resident lymphocytes, while nontargeted NO delivery did not. These results further extended to a peptide-conjugated NP drug delivery system, which showed enhanced uptake by B cells and dendritic cells when administered alongside SNO-NP. Together, these results highlight the utility of LN-targeted NO application for the enhancement of nanocarrier access to therapeutically relevant LN-resident immune cells, making NO a potentially useful tool for improving LN drug delivery and immune responses.
Collapse
|
49
|
Shores LS, Kelly SH, Hainline KM, Suwanpradid J, MacLeod AS, Collier JH. Multifactorial Design of a Supramolecular Peptide Anti-IL-17 Vaccine Toward the Treatment of Psoriasis. Front Immunol 2020; 11:1855. [PMID: 32973764 PMCID: PMC7461889 DOI: 10.3389/fimmu.2020.01855] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
Current treatments for chronic immune-mediated diseases such as psoriasis, rheumatoid arthritis, or Crohn's disease commonly rely on cytokine neutralization using monoclonal antibodies; however, such approaches have drawbacks. Frequent repeated dosing can lead to the formation of anti-drug antibodies and patient compliance issues, and it is difficult to identify a single antibody that is broadly efficacious across diverse patient populations. As an alternative to monoclonal antibody therapy, anti-cytokine immunization is a potential means for long-term therapeutic control of chronic inflammatory diseases. Here we report a supramolecular peptide-based approach for raising antibodies against IL-17 and demonstrate its efficacy in a murine model of psoriasis. B-cell epitopes from IL-17 were co-assembled with the universal T-cell epitope PADRE using the Q11 self-assembling peptide nanofiber system. These materials, with or without adjuvants, raised antibody responses against IL-17. Exploiting the modularity of the system, multifactorial experimental designs were used to select formulations maximizing titer and avidity. In a mouse model of psoriasis induced by imiquimod, unadjuvanted nanofibers had therapeutic efficacy, which could be enhanced with alum adjuvant but reversed with CpG adjuvant. Measurements of antibody subclass induced by adjuvanted and unadjuvanted formulations revealed strong correlations between therapeutic efficacy and titers of IgG1 (improved efficacy) or IgG2b (worsened efficacy). These findings have important implications for the development of anti-cytokine active immunotherapies and suggest that immune phenotype is an important metric for eliciting therapeutic anti-cytokine antibody responses.
Collapse
Affiliation(s)
- Lucas S Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Sean H Kelly
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Jutamas Suwanpradid
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States
| | - Amanda S MacLeod
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
50
|
Yin WM, Li YW, Gu YQ, Luo M. Nanoengineered targeting strategy for cancer immunotherapy. Acta Pharmacol Sin 2020; 41:902-910. [PMID: 32398683 PMCID: PMC7470800 DOI: 10.1038/s41401-020-0417-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/12/2020] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy is rapidly changing the paradigm of cancer care and treatment by evoking host immunity to kill cancer cells. As clinical approval of checkpoint inhibitors (e.g., ipilimumab and pembrolizumab) has been accelerated by a dramatic improvement of long-term survival in a small subset of patients compared to conventional chemotherapy, growing interesting research has focused on immunotherapy. However, majority of patients have not benefited from checkpoint therapies that only partially remove the inhibition of T cell functions. Insufficient systemic T cell responses, low immunogenicity and the immunosuppressive environment of tumors, create great challenges on therapeutic efficiency. Nanotechnology can integrate multiple functions within controlled size and shape, and has been explored as a unique avenue for the development of cancer immunotherapy. In this review, we mainly address how nanoengineered vaccines can induce robust T cell responses against tumors, as well as how nanomedicine can remodel the tumor immunosuppressive microenvironment to boost antitumor immune responses.
Collapse
|