1
|
Zhang Y, Li Y, Xu Z, Xu L, Wang Y, Li N, Solek NC, Wang Y, Li B, Liu H. PPS-TLR7/8 agonist nanoparticles equip robust anticancer immunity by selectively prolonged activation of dendritic cells. Biomaterials 2025; 316:123032. [PMID: 39705927 DOI: 10.1016/j.biomaterials.2024.123032] [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: 07/12/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Checkpoint inhibitor therapies do not benefit all patients, and adjuvants play a critical role in boosting immune responses for effective cancer immunotherapy. However, their systemic toxicity and suboptimal activation kinetics pose significant challenges. Here, this study presented a linker-based strategy to modulate the activation kinetics of Toll-like receptor 7/8 (TLR7/8) agonists delivered via poly (propylene sulfide) nanoparticles (PPS NPs). By covalently binding small molecule TLR7/8 agonists to PPS NPs with different linkers, enhanced therapeutic efficacy is achieved while abrogating systemic toxicity. These results showed that an alkyl linker selectively prolong the activation of DCs. It avoided the extensive activation of other APCs, favoring the limitation of immune-related toxicities. This strategy exhibited significant anti-tumor activity in alkyl linked nano-TLR7/8 agonists treatment alone, and cytokine and immune cell profiling provided evidence of prolonged immune cell activation in the tumor microenvironment, with evidence of an increase in the frequency of tumor antigen-specific CD8+ T cells. This linker-based approach offers a promising strategy to optimize the delivery of nano-TLR7/8 agonists for cancer immunotherapy, potentially advancing the field toward improved clinical outcomes.
Collapse
Affiliation(s)
- Yingxi Zhang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Yicheng Li
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Zhaochu Xu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Linyi Xu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Yue Wang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Ning Li
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Nicholas C Solek
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Yongjun Wang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Bowen Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada; Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C1, Canada.
| | - Hongzhuo Liu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
2
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
3
|
Paranandi KS, Amar-Lewis E, Mirkin CA, Artzi N. Nomadic Nanomedicines: Medicines Enabled by the Paracrine Transfer Effect. ACS NANO 2025; 19:21-30. [PMID: 39746105 DOI: 10.1021/acsnano.4c15052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
In nanomedicine, the cellular export of nanomaterials has been less explored than uptake. Traditionally viewed in a negative light, recent findings highlight the potential of nanomedicine export to enhance therapeutic effects. This Perspective examines key pathways for export and how nanomaterial design affects removal rates. We present the idea of the "paracrine transfer effect" (PTE), where nanomaterials are first internalized by a "waypoint" cell and then exported to a "destination" cell, influencing both in potentially exploitable ways. Essential characteristics for nanomedicines to leverage the PTE are discussed, along with two case studies: STING-stimulating polymeric nanoparticles and TLR9-stimulating liposomal spherical nucleic acids. We propose future research directions to better understand and utilize the PTE in developing more effective nanomedicines.
Collapse
Affiliation(s)
- Krishna S Paranandi
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Eliz Amar-Lewis
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| | - Chad A Mirkin
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| |
Collapse
|
4
|
Shao L, Zhang H, Sun L, Ning L, Sun X, Qin C, Xu W, Xu R, Jia F. Responsive Degradable Bottlebrush Polymers Enable Drugs With Superior Efficacy and Minimal Systemic Toxicity. Adv Healthc Mater 2025:e2405202. [PMID: 39757488 DOI: 10.1002/adhm.202405202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Bottlebrush polymers (BBPs) have garnered significant attention as advanced drug delivery systems, capable of transporting a diverse range of therapeutic agents, including both chemical drugs and biologics. Despite their effectiveness, the empty BBP vectors post-drug release may pose long-term safety risks due to their difficult systemic clearance. Here, a responsive degradable BBP platform for cancer therapy is developed, featuring a poly(disulfide) backbone grafted with fluorine-terminated zwitterionic side chains. Anti-cancer drugs are tethered to the backbone via a clinically approved valine-citrulline (VC) linker. This design leverages the tumor's reductive environment and Cathepsin B overexpression for BBP rapid degradation and precise drug release restricted within tumor cells, thereby addressing systemic safety concerns over synthetic BBP and expanding the therapeutic window of anti-cancer drugs simultaneously. Surface fluorination of BBP further enhances tumor accumulation and deep penetration. In vivo studies with monomethyl auristatin E (MMAE)-loaded BBP in tumor-bearing mice demonstrate substantial tumor suppression with minimal side effects. Together, these findings highlight the potential of responsive degradable BBP as a versatile unimolecular platform for cancer drug delivery, addressing existing challenges associated with synthetic BBP nanomedicines.
Collapse
Affiliation(s)
- Liming Shao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Hongrui Zhang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Lei Sun
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Lubin Ning
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xiuying Sun
- School of Materials Science and Engineering, Tianjin University, Tianjin, 300350, China
| | - Chaoke Qin
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Wenhua Xu
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Rui Xu
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Fei Jia
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
5
|
Zhao Y, Zhao X, Wang X, Ma Z, Yan J, Li S, Wang N, Jiao J, Cui J, Zhang G. Polyphenol-mediated assembly of toll-like receptor 7/8 agonist nanoparticles for effective tumor immunotherapy. Acta Biomater 2024:S1742-7061(24)00780-3. [PMID: 39746528 DOI: 10.1016/j.actbio.2024.12.060] [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: 10/07/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Toll-like receptor (TLR) 7/8 agonists have shown significant potential in tumor immunotherapy. However, the limited pharmacokinetic properties and systemic toxicity resulting from off-target effects limits their biomedical applications. We here report the polyphenol-mediated assembly of resiquimod (R848, a TLR7/8 agonist) nanoparticles (RTP NPs) to achieve tumor-selective immunotherapy while avoiding systemic adverse effects. Upon intravenous administration, the prepared RTP NPs are effectively accumulated at tumor sites, which increase their bioavailability and reduce systemic inflammation. RTP NPs can trigger a potent antitumor immune response in a mouse tumor model to inhibit tumor growth. Additionally, after subcutaneous injection at the tail base, RTP NPs efficiently migrate to the lymph nodes, where they elicit immune memory to prevent tumorigenesis. This study underscores the potential application of polyphenol-mediated assembly in developing nanomedicines with reduced toxicity for tumor-specific immunotherapy. STATEMENT OF SIGNIFICANCE: Toll-like receptor agonist (R848) nanoparticles for tumor-selective immunotherapy were synthesized through polyphenol-mediated assembly, a method that simplifies preparation process and minimizes potential side effects. Intravenously administered these nanoparticles effectively extended circulation time, enhanced tumor enrichment, and reduced systemic inflammation, thus augmenting the bioavailability and minimizing the side effects of R848. The nanoparticles significantly inhibited tumor growth by triggering a potent antitumor immune response, including dendritic cell maturation, macrophage polarization, T-cell infiltration, and cytokine secretion. Moreover, after subcutaneous injection at the tail base, they can elicit immune memory to prevent tumorigenesis.
Collapse
Affiliation(s)
- Yilei Zhao
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xiaonan Zhao
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xuechun Wang
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zilin Ma
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jie Yan
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Songyan Li
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ning Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology Chinese Academy of Sciences, Beijing 100101, China.
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Guiqiang Zhang
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
6
|
Niu G, Song G, Kang Y, Zhai Y, Fan Y, Ye J, Li R, Li R, Zhang Y, Wang H, Chen Y, Ji X. Quinoidal Semiconductor Nanoparticles for NIR-II Photoacoustic Imaging and Photoimmunotherapy of Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2415189. [PMID: 39696886 DOI: 10.1002/adma.202415189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Photoagents with ultra-high near-infrared II (NIR-II) light energy conversion efficiency hold great promise in tumor phototherapy due to their ability to penetrate deeper tissues and minimize damage to surrounding healthy cells. However, the development of NIR-II photoagents remain challenging. In this study, an all-fused-ring quinoidal acceptor-donor-acceptor (A-D-A) molecule, SKCN, with a BTP core is synthesized, and nanoparticles named FA-SNPs are prepared. The unique quinoidal structure enhances π-electron delocalization and bond length uniformity, significantly reducing the bandgap of SKCN, resulting in strong NIR-II absorption, a high molar extinction coefficient, and a photothermal conversion efficiency of 75.14%. Enhanced molecular rigidity also facilitates efficient energy transfer to oxygen, boosting reactive oxygen species generation. By incorporating the immunomodulator R848, FA-SRNPs nanoparticles are further developed, effectively modulating the tumor immune microenvironment by reducing Tregs and M-MDSCs infiltration, promoting dendritic cell maturation, M1 macrophage polarization, and activating CD8+ T cells and NK cells. Comprehensive studies using orthotopic ovarian cancer models demonstrated strong tumor targeting, photoacoustic imaging capabilities, and significant tumor suppression and metastasis inhibition, and also showing excellent therapeutic efficacy in an orthotopic breast cancer model. This study provides strong evidence for the potential application of quinoidal A-D-A molecules in cancer photoimmunotherapy.
Collapse
Affiliation(s)
- Gaoli Niu
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
- The First Affiliated Hospital of Henan Polytechnic University, Jiaozuo, 454000, China
| | - Guangkun Song
- State Key Laboratory and Institute of Elemento-Organic Chemistry, The Centre of Nanoscale Science and Technology and Key Laboratory of Functional Polymer Materials, Renewable Energy Conversion and Storage Center (RECAST), Tianjin Key Laboratory of Functional Polymer Materials, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yong Kang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Yanhong Zhai
- The First Affiliated Hospital of Henan Polytechnic University, Jiaozuo, 454000, China
| | - Yueyue Fan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Jiamin Ye
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Ruiyan Li
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Runtan Li
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Yanwei Zhang
- Xiyingmen Subdistrict Community Health Service Center, Xiqing District, Tianjin, 300072, China
| | - Hong Wang
- The First Affiliated Hospital of Henan Polytechnic University, Jiaozuo, 454000, China
| | - Yongsheng Chen
- State Key Laboratory and Institute of Elemento-Organic Chemistry, The Centre of Nanoscale Science and Technology and Key Laboratory of Functional Polymer Materials, Renewable Energy Conversion and Storage Center (RECAST), Tianjin Key Laboratory of Functional Polymer Materials, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
- Medical College, Linyi University, Linyi, 276000, China
| |
Collapse
|
7
|
Xiao W, Sun S, Geng R, Bi D, Liu Y, Zhu J. A General "Two-Lock" Strategy to Enhance Drug Loading and Lysosomal Escape in Intelligent Boronate Ester Polymer Nanoparticles for Improved Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68890-68904. [PMID: 39641664 DOI: 10.1021/acsami.4c15172] [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: 12/07/2024]
Abstract
Boronate ester can be used to prepare intelligent polymer nanoparticles (NPs). However, the traditional boronate ester polymer NPs made of boronic acid and diols using a "single-lock" strategy (B-O NPs) exhibit low drug loading capacity (DLC) and insufficient lysosomal escape ability, resulting in limited antitumor efficacy. We develop a "two-lock" strategy that combines dodecanamine and boronic acid using boron-nitrogen (B ← N) coordination to enhance the formation of a boronate ester polymer. Through this strategy, amphiphilic dextran and poly(vinyl alcohol) are synthesized through conjugation with the phenylboronic acid (PBA)/dodecanamine complex. The amphiphilic dextran encapsulates paclitaxel (PTX) to form B-N-O NPs with a higher DLC than their "single-lock" compartments due to enhanced boronate ester stability and improved hydrophobic drug-polymer interactions. Moreover, the B-N-O NPs release more PTX under acidic conditions compared with the B-O NPs. To demonstrate the generality of this "two-lock" strategy, eight polymer prodrug B-N-O NPs employing poly(vinyl alcohol) or dextran, along with PBA-modified gemcitabine, fluorouracil, and 7-ethyl-10-hydroxycamptothecin, or boronic acid-containing bortezomib and dodecanamine, are prepared, showing overall enhanced DLC and higher responsive drug release efficiency compared to B-O NPs. Importantly, B-N-O NPs with a combination of dodecanamine and boronic acid show a better lysosomal escape capability than B-O NPs. Moreover, B-N-O NPs exhibit stronger cytotoxicity compared to B-O NPs and free drugs in vitro. Their enhanced drug loading, responsive drug release, and lysosomal escape abilities contribute to enhanced antitumor efficacy in vivo. This "two-lock" strategy can be a general and convenient method to prepare responsive polymer NPs with enhanced anticancer efficacy.
Collapse
Affiliation(s)
- Wanyue Xiao
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shaoxing Sun
- Department of Radiation and Medical Oncology; Cancer Clinical Study Center of Hubei Province; Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Rui Geng
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Duohang Bi
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yijing Liu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518057, China
| | - Jintao Zhu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
8
|
Beilharz S, Debnath MK, Vinella D, Shoffstall AJ, Karayilan M. Advances in Injectable Polymeric Biomaterials and Their Contemporary Medical Practices. ACS APPLIED BIO MATERIALS 2024; 7:8076-8101. [PMID: 39471414 DOI: 10.1021/acsabm.4c01001] [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: 11/01/2024]
Abstract
Injectable biomaterials have been engineered to operate within the human body, offering versatile solutions for minimally invasive therapies and meeting several stringent requirements such as biocompatibility, biodegradability, low viscosity for ease of injection, mechanical strength, rapid gelation postinjection, controlled release of therapeutic agents, hydrophobicity/hydrophilicity balance, stability under physiological conditions, and the ability to be sterilized. Their adaptability and performance in diverse clinical settings make them invaluable for modern medical treatments. This article reviews recent advancements in the design, synthesis, and characterization of injectable polymeric biomaterials, providing insights into their emerging applications. We discuss a broad spectrum of these materials, including natural, synthetic, hybrid, and composite types, that are being applied in targeted drug delivery, cell and protein transport, regenerative medicine, tissue adhesives, injectable implants, bioimaging, diagnostics, and 3D bioprinting. Ultimately, the review highlights the critical role of injectable polymeric biomaterials in shaping the future of medical treatments and improving patient outcomes across a wide range of therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Sophia Beilharz
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Mithun Kumar Debnath
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Daniele Vinella
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Metin Karayilan
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
9
|
Hauck AV, Komforth P, Erlenbusch J, Stickdorn J, Radacki K, Braunschweig H, Besenius P, Van Herck S, Nuhn L. Aliphatic polycarbonates with acid degradable ketal side groups as multi-pH-responsive immunodrug nanocarriers. Biomater Sci 2024. [PMID: 39575699 DOI: 10.1039/d4bm00949e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Pharmacokinetics and biodistribution profiles of active substances are crucial aspects for their safe and successful administration. Since many immunogenic compounds do not meet all requirements for safe and effective administration, well-defined drug nanocarrier systems are necessary with a stimuli-responsive drug-release profile. For this purpose, a novel pH-responsive aliphatic cyclic carbonate is introduced with benzyl ketal side chains and polymerized onto a poly(ethylene glycol) macroinitiator. The resulting block copolymers could be formulated via a solvent-evaporation method into well-defined polymeric micelles. The hydrophobic carbonate block was equipped with an acid degradable ketal side group that served as an acid-responsive functional group. Already subtle pH alternations led to micelle disassembly and the release of the active cargo. Furthermore, basic carbonate backbone degradation assured the pH responsiveness of the nanocarriers in both acidic and basic conditions. To investigate the delivery capacity of polymeric micelles, the model small molecule compound CL075, which serves as an immunotherapeutic TLR7/8 agonist, was encapsulated. Incubation studies with human blood plasma revealed the absence of undesirable protein adsorption on the drug-loaded nanoparticles. Furthermore, in vitro applications confirmed cell uptake of the nanodrug formulations by macrophages and the induction of payload-mediated immune stimulation. Altogether, these results underline the huge potential of the developed multi-pH-responsive polymeric nanocarrier for immunodrug delivery.
Collapse
Affiliation(s)
- Adrian V Hauck
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Julius-Maximilians-Universität Würzburg, 97070 Würzburg, Germany.
| | - Patric Komforth
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Jessica Erlenbusch
- Department of Chemistry, Johannes-Gutenberg-Universität Mainz, 55122 Mainz, Germany
| | | | - Krzysztof Radacki
- Institute for Sustainable Chemistry and Catalysis with Boron, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Holger Braunschweig
- Institute for Sustainable Chemistry and Catalysis with Boron, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Pol Besenius
- Department of Chemistry, Johannes-Gutenberg-Universität Mainz, 55122 Mainz, Germany
| | - Simon Van Herck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Lutz Nuhn
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Julius-Maximilians-Universität Würzburg, 97070 Würzburg, Germany.
| |
Collapse
|
10
|
Lou Z, Shi Y, Guo X, Jin Z, Huang J, Hu Y, Liu X, Zhu J, Kuang R, You J. Chronological Management of Adjuvant Effect for Optimized mRNA Vaccine Inspired by Natural Virus Infection. ACS NANO 2024. [PMID: 39011561 DOI: 10.1021/acsnano.4c04953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
The efficacy and safety of mRNA vaccines both rely on a fine-tuning of specific humoral and cellular immune responses. Instead of adjustments in vaccine component, we proposed a concept of chronological management of adjuvant effect to modulate the adaptive immune potency and preference inspired by natural virus infection. By simulating type I interferon expression dynamics during viral infection, three vaccine strategies employing distinct exposure sequences of adjuvant and mRNA have been developed, namely Precede, Coincide, and Follow. Follow, the strategy of adjuvant administration following mRNA, effectively suppressed tumor progression, which was attributed to enhanced mRNA translation, augmented p-MHC I expression, and elevated CD8+ T cell response. Meanwhile, Follow exhibited improved biosafety, characterized by reduced incidences of cardiac and liver toxicity, owing to its alteration to the vaccination microenvironment between successive injections. Our strategy highlights the importance of fine-tuning adjuvant effect dynamics in optimizing mRNA vaccines for clinical application.
Collapse
Affiliation(s)
- Zeliang Lou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Xuemeng Guo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Zhaolei Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jiaxin Huang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yilong Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Xu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jiang Zhu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P. R. China
| | - Rong Kuang
- Zhejiang Institute for Food and Drug Control, 325 Pingle Street, Hangzhou, Zhejiang 310004, P. R. China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang 310006, P. R. China
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310006, P. R. China
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang 321299, P. R. China
| |
Collapse
|
11
|
Leo CM, Jang J, Corey EJ, Neary WJ, Bowman JI, Kennemur JG. Comparison of Polypentenamer and Polynorbornene Bottlebrushes in Dilute Solution. ACS POLYMERS AU 2024; 4:235-246. [PMID: 38882033 PMCID: PMC11177302 DOI: 10.1021/acspolymersau.3c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 06/18/2024]
Abstract
Bottlebrush (BB) polymers were synthesized via grafting-from-atom transfer radical polymerization (ATRP) of styrene on polypentenamer and polynorbornene macroinitiators with matched grafting density (n g = 4) and backbone degrees of polymerization (122 ≥ N bb ≥ 61) to produce a comparative study on their respective dilute solution properties as a function of increasing side chain degree of polymerization (116 ≥ N sc ≥ 5). The grafting-from technique produced near quantitative grafting efficiency and narrow dispersity N sc as evidenced by spectroscopic analysis and ring closing metathesis depolymerization of the polypentenamer BBs. The versatility of this synthetic approach permitted a comprehensive survey of power law expressions that arise from monitoring intrinsic viscosity, hydrodynamic radius, and radius of gyration as a function of increasing the molar mass of the BBs by increasing N sc. These values were compared to a series of linear (nongrafted, N sc = 0) macroinitiators in addition to linear grafts. This unique study allowed elucidation of the onset of bottlebrush behavior for two different types of bottlebrush backbones with identical grafting density but inherently different flexibility. In addition, grafting-from ATRP of methyl acrylate on a polypentenamer macroinitiator allowed the observation of the effects of graft chemistry in comparison to polystyrene. Differences in the observed scaling relationships in dilute solution as a function of each of these synthetic variants are discussed.
Collapse
Affiliation(s)
- Courtney M Leo
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32303, United States
| | - Jaehoon Jang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32303, United States
| | - Ethan J Corey
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32303, United States
| | - William J Neary
- Department of Chemistry, University of California at Riverside, Riverside, California 92521, United States
| | - Jared I Bowman
- George and Josephine Butler Polymer Research Laboratory, Center for Macromolecular Science & Engineering, Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Justin G Kennemur
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32303, United States
| |
Collapse
|
12
|
Xu Y, Yang J, Meng X, Meng S, Sun T, Ding N, Zong C. The synthesis and preliminary immunological evaluation of a dual-adjuvant SARS-CoV-2 RBD vaccine: Covalent integration of TLR7/8 and iNKT cell agonists. Int J Biol Macromol 2024; 270:132258. [PMID: 38735613 DOI: 10.1016/j.ijbiomac.2024.132258] [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: 02/29/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
Covalently linking an adjuvant to an antigenic protein enhances its immunogenicity by ensuring a synergistic delivery to the immune system, fostering a more robust and targeted immune response. Most adjuvant-protein conjugate vaccines incorporate only one adjuvant due to the difficulties in its synthesis. However, there is a growing interest in developing vaccines with multiple adjuvants designed to elicit a more robust and targeted immune response by engaging different aspects of the immune system for complex diseases where traditional vaccines fall short. Here, we pioneer the synthesis of a dual-adjuvants protein conjugate Vaccine 1 by assembling a toll-like receptor 7/8 (TLR7/8) agonist, an invariant natural killer T cell (iNKT) agonist with a clickable bicyclononyne (BCN). The BCN group can bio-orthogonally react with azide-modified severe acute respiratory syndrome coronavirus-2 receptor-binding domain (SARS-CoV-2 RBD) trimer antigen to give the three-component Vaccine 1. Notably, with a mere 3 μg antigen, it elicited a balanced subclass of IgG titers and 20-fold more IgG2a than control vaccines, highlighting its potential for enhancing antibody-dependent cellular cytotoxicity. This strategy provides a practicable way to synthesize covalently linked dual immunostimulants. It expands the fully synthetic self-adjuvant protein vaccine that uses a single adjuvant to include two different types of adjuvants.
Collapse
Affiliation(s)
- Ying Xu
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Jing Yang
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Xiongyan Meng
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Shuai Meng
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Tiantian Sun
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China.
| | - Ning Ding
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| | - Chengli Zong
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China.
| |
Collapse
|
13
|
Gu L, Kong X, Li M, Chen R, Xu K, Li G, Qin Y, Wu L. Molecule engineering strategy of toll-like receptor 7/8 agonists designed for potentiating immune stimuli activation. Chem Commun (Camb) 2024; 60:5474-5485. [PMID: 38712400 DOI: 10.1039/d4cc00792a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Toll-like receptor 7/8 (TLR-7/8) agonists serve as a promising class of pattern recognition receptors that effectively evoke the innate immune response, making them promising immunomodulatory agents for tumor immunotherapy. However, the uncontrollable administration of TLR-7/8 agonists frequently leads to the occurrence of severe immune-related adverse events (irAEs). Thus, it is imperative to strategically design tumor-microenvironment-associated biomarkers or exogenous stimuli responsive TLR-7/8 agonists in order to accurately evaluate and activate innate immune responses. No comprehensive elucidation has been documented thus far regarding TLR-7/8 immune agonists that are specifically engineered to enhance immune activation. In this feature article, we provide an overview of the advancements in TLR-7/8 agonists, aiming to enhance the comprehension of their mechanisms and promote the clinical progression through nanomedicine strategies. The current challenges and future directions of cancer immunotherapy are also discussed, with the hope that this work will inspire researchers to explore innovative applications for triggering immune responses through TLR-7/8 agonists.
Collapse
Affiliation(s)
- Liuwei Gu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Xiaojie Kong
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Mengyan Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Rui Chen
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Ke Xu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Guo Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Yulin Qin
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Li Wu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| |
Collapse
|
14
|
Zafar H, Liu B, Nguyen HVT, Johnson JA. Caspase-3-Responsive, Fluorogenic Bivalent Bottlebrush Polymers. ACS Macro Lett 2024; 13:571-576. [PMID: 38647178 DOI: 10.1021/acsmacrolett.4c00119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Controlling the access of proteases to cleavable peptides placed at specific locations within macromolecular architectures represents a powerful strategy for biologically responsive materials design. Here, we report the synthesis of peptide-containing bivalent bottlebrush (co)polymers (BBPs) featuring polyethylene glycol (PEG) and 7-amino-4-methylcoumarin (AMC) pendants on each backbone repeat unit. The AMCs are linked via caspase-3-cleavable peptides which, upon enzymatic cleavage, provide a "turn-on" fluorescence signal due to the release of free AMC. Time-dependent fluorscence measurements demonstrate that the caspase-3-induced peptide cleavage and AMC release from BBPs is strongly dependent on the BBP backbone length and the AMC-peptide linker location within the BBP architecture, revealing fundamental insights into the interactions of enzymes with BBPs.
Collapse
Affiliation(s)
- Hadiqa Zafar
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bin Liu
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Hung V-T Nguyen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Wilson J, Kimmel B, Arora K, Chada N, Bharti V, Kwiatkowski A, Finklestein J, Hanna A, Arner E, Sheehy T, Pastora L, Yang J, Pagendarm H, Stone P, Taylor B, Hubert L, Gibson-Corley K, May J, McLean J, Rathmell J, Richmond A, Rathmell W, Balko J, Fingleton B, Hargrove-Wiley E. Programable Albumin-Hitchhiking Nanobodies Enhance the Delivery of STING Agonists to Potentiate Cancer Immunotherapy. RESEARCH SQUARE 2024:rs.3.rs-3243545. [PMID: 38766114 PMCID: PMC11100900 DOI: 10.21203/rs.3.rs-3243545/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Stimulator of interferon genes (STING) is a promising target for potentiating antitumor immunity, but multiple pharmacological barriers limit the clinical utility, efficacy, and/or safety of STING agonists. Here we describe a modular platform for systemic administration of STING agonists based on nanobodies engineered for in situ hitchhiking of agonist cargo on serum albumin. Using site-selective bioconjugation chemistries to produce molecularly defined products, we found that covalent conjugation of a STING agonist to anti-albumin nanobodies improved pharmacokinetics and increased cargo accumulation in tumor tissue, stimulating innate immune programs that increased the infiltration of activated natural killer cells and T cells, which potently inhibited tumor growth in multiple mouse tumor models. We also demonstrated the programmability of the platform through the recombinant integration of a second nanobody domain that targeted programmed cell death ligand-1 (PD-L1), which further increased cargo delivery to tumor sites while also blocking immunosuppressive PD-1/PD-L1 interactions. This bivalent nanobody carrier for covalently conjugated STING agonists stimulated robust antigen-specific T cell responses and long-lasting immunological memory, conferred enhanced therapeutic efficacy, and was effective as a neoadjuvant treatment for improving responses to adoptive T cell transfer therapy. Albumin-hitchhiking nanobodies thus offer an enabling, multimodal, and programmable platform for systemic delivery of STING agonists with potential to augment responses to multiple immunotherapeutic modalities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ann Hanna
- Vanderbilt University Medical Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Zhang W, Xu Y, Guo R, Zhuang P, Hong H, Tan H, Wang M. Theranostic Bottle-Brush Polymers Tailored for Universal Solid-Tumor Targeting. ACS NANO 2024; 18:11688-11702. [PMID: 38665009 DOI: 10.1021/acsnano.3c11755] [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: 05/08/2024]
Abstract
Designing an efficient nanocarrier to target multiple types of cancer remains a major challenge in the development of cancer nanomedicines. The majority of systemically administered nanoparticles (NPs) are rapidly cleared by the liver, resulting in poor tumor-targeting efficiency and severe side effects. Here, we present a delicately tailored design and synthesis of fluorescent bottle-brush polymers and screen nine derived NPs, each varying in size and surface coatings, for tumor imaging and targeted delivery. Our optimized polymer bearing (oligo(ethylene glycol) methyl ether methacrylate) in the side chains shows reduced macrophage uptake, prolonged blood-circulation time (up to 27 h), and exceptionally high accumulation in the tumor compared to the liver, elucidating an immune-evasion-induced tumor-targeting mechanism. High tumor accumulation significantly improved the antitumor efficacy. The outstanding tumor-targeting ability has been further validated across five distinct tumor models, including orthotopic glioblastoma and pancreatic cancer, which demonstrate the universality of our polymeric nanocarrier for tumor-targeting delivery.
Collapse
Affiliation(s)
- Wei Zhang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Yanwen Xu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518000, Guangdong, China
| | - Rongjun Guo
- Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518000, Guangdong, China
| | - Peiling Zhuang
- Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518000, Guangdong, China
| | - Huixia Hong
- College of Chemistry, Xinjiang University, Urumqi 830046, China
| | - Hui Tan
- Shenzhen Children's Hospital of Shantou University Medical College, Shenzhen Second People's Hospital, Shenzhen University Health Science Center, Shenzhen 518026, Guangdong, China
| | - Mingfeng Wang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| |
Collapse
|
17
|
Kawai T, Ikegawa M, Ori D, Akira S. Decoding Toll-like receptors: Recent insights and perspectives in innate immunity. Immunity 2024; 57:649-673. [PMID: 38599164 DOI: 10.1016/j.immuni.2024.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024]
Abstract
Toll-like receptors (TLRs) are an evolutionarily conserved family in the innate immune system and are the first line of host defense against microbial pathogens by recognizing pathogen-associated molecular patterns (PAMPs). TLRs, categorized into cell surface and endosomal subfamilies, recognize diverse PAMPs, and structural elucidation of TLRs and PAMP complexes has revealed their intricate mechanisms. TLRs activate common and specific signaling pathways to shape immune responses. Recent studies have shown the importance of post-transcriptional regulation in TLR-mediated inflammatory responses. Despite their protective functions, aberrant responses of TLRs contribute to inflammatory and autoimmune disorders. Understanding the delicate balance between TLR activation and regulatory mechanisms is crucial for deciphering their dual role in immune defense and disease pathogenesis. This review provides an overview of recent insights into the history of TLR discovery, elucidation of TLR ligands and signaling pathways, and their relevance to various diseases.
Collapse
Affiliation(s)
- Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan; Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan.
| | - Moe Ikegawa
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Daisuke Ori
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Shizuo Akira
- Center for Advanced Modalities and DSS (CAMaD), Osaka University, Osaka 565-0871, Japan; Laboratory of Host Defense, Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan; Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
18
|
Song K, Pun SH. Design and Evaluation of Synthetic Delivery Formulations for Peptide-Based Cancer Vaccines. BME FRONTIERS 2024; 5:0038. [PMID: 38515636 PMCID: PMC10956738 DOI: 10.34133/bmef.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
With the recent advances in neoantigen identification, peptide-based cancer vaccines offer substantial potential in the field of immunotherapy. However, rapid clearance, low immunogenicity, and insufficient antigen-presenting cell (APC) uptake limit the efficacy of peptide-based cancer vaccines. This review explores the barriers hindering vaccine efficiency, highlights recent advancements in synthetic delivery systems, and features strategies for the key delivery steps of lymph node (LN) drainage, APC delivery, cross-presentation strategies, and adjuvant incorporation. This paper also discusses the design of preclinical studies evaluating vaccine efficiency, including vaccine administration routes and murine tumor models.
Collapse
Affiliation(s)
- Kefan Song
- Department of Bioengineering, University of Washington, USA
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, USA
- Molecular Engineering & Sciences Institute, University of Washington, USA
| |
Collapse
|
19
|
Sun J, Yao H, Ren X, Cui L, Liu L, Wang G, Tang Z. Radiation-Activated Resiquimod Prodrug Nanomaterials for Enhancing Immune Checkpoint Inhibitor Therapy. NANO LETTERS 2024; 24:2921-2930. [PMID: 38411094 DOI: 10.1021/acs.nanolett.4c00114] [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: 02/28/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapy is effectively employed in treating various malignancies. However, the response rate is constrained to 5-30%, which is attributed to differences in immune responses across different tumors. Overcoming all obstacles of multistep immune activation with monotherapy is difficult. Here, maleimide-modified resiquimod (R848) prodrug nanoparticles (MAL-NPs) are reported and combined with radiotherapy (RT) and anti-PD1 to enhance ICI therapy. MAL-NPs can promote antigen endocytosis by dendritic cells and are radio-reduced to produce R848. When combined with RT, MAL-NPs can augment the concentration of nanoparticles at tumor sites and be selectively radio-reduced within the tumor, thereby triggering a potent antitumor immune response. The systemic immune response and long-term memory efficacy induced by MAL-NPs + RT + anti-PD1 significantly inhibit the abscopal tumor growth and prevent tumor recurrence. This strategy can achieve systemic therapy through selective training of the tumor immune microenvironment, offering a new approach to overcome the obstacles of ICI therapy.
Collapse
Affiliation(s)
- Jiali Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, 130021 Jilin, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, China
| | - Haochen Yao
- Hepatobiliary and Pancreatic Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Xitong Ren
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026 Anhui, China
| | - Linjie Cui
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026 Anhui, China
| | - Linlin Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033 Jilin, China
| | - Guoqing Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, 130021 Jilin, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026 Anhui, China
| |
Collapse
|
20
|
Shin HS, Kim S, Jin SM, Yoo YJ, Heo JH, Lim YT. Molecular Masking of Synthetic Immunomodulator Evokes Antitumor Immunity With Reduced Immune Tolerance and Systemic Toxicity by Temporal Activity Recovery and Sustained Stimulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309039. [PMID: 37903320 DOI: 10.1002/adma.202309039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/17/2023] [Indexed: 11/01/2023]
Abstract
Activation of the innate immune system counteracts tumor-induced immunosuppression. Hence, small molecule-based toll-like receptor 7/8 agonists (TLR7/8a), which can modulate immunosuppression in the tumor microenvironment along with the activation of innate immunity, are emerging as essential components of cancer immunotherapy. However, the clinical application of synthetic TLR7/8a therapies is limited by systemic immune-associated toxicity and immune tolerance induced by uncontrolled stimulatory activities and repeated treatments. To address these limitations, a dynamic immunomodulation strategy incorporating masking and temporal recovery of the activity of TLR7/8a through prodrug-like TLR7/8a (pro-TLR7/8a) at the molecular level and a sustained and controlled release of active TLR7/8a from nanoliposome (pro-TLR7/8a) (NL(pro-TLR7/8)) in a macroscale depot are designed. Immunization with cationic NL(pro-TLR7/8) and anionic antigens triggers robust activation of innate immune cells as well as antigen-specific T cell responses, eliciting reprogramming of immunosuppressive cells into tumor-suppressive cells, with decreased systemic adverse effects and immune tolerance. Combination treatment with NL(pro-TLR7/8a) and immune checkpoint inhibitors (anti-CTLA-4 plus anti-PD-L1) or nanoliposomes (Doxorubicin) has synergistic effects on antitumor immunity in various tumor models. The concept of pro-TLR7/8a suggested herein may facilitate the advancement of small-molecule-based immunomodulators for clinical translation and safe and effective cancer immunotherapy.
Collapse
Affiliation(s)
- Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sohyun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, 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 Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Yeon Jeong Yoo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jang Hun Heo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, 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 Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| |
Collapse
|
21
|
Lensch V, Gabba A, Hincapie R, Bhagchandani SH, Basak A, Alam MM, Irvine DJ, Shalek AK, Johnson JA, Finn MG, Kiessling LL. Glycan-costumed virus-like particles promote type 1 anti-tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.575711. [PMID: 38293025 PMCID: PMC10827186 DOI: 10.1101/2024.01.18.575711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Cancer vaccine development is inhibited by a lack of strategies for directing dendritic cell (DC) induction of effective tumor-specific cellular immunity. Pathogen engagement of DC lectins and toll-like receptors (TLRs) shapes immunity by directing T cell function. Strategies to activate specific DC signaling pathways via targeted receptor engagement are crucial to unlocking type 1 cellular immunity. Here, we engineered a glycan-costumed virus-like particle (VLP) vaccine that delivers programmable peptide antigens to induce tumor-specific cellular immunity in vivo. VLPs encapsulating TLR7 agonists and decorated with a selective mannose-derived ligand for the lectin DC-SIGN induced robust DC activation and type 1 cellular immunity, whereas VLPs lacking this key DC-SIGN ligand failed to promote DC-mediated immunity. Vaccination with glycan-costumed VLPs generated tumor antigen-specific Th1 CD4+ and CD8+ T cells that infiltrated solid tumors, inhibiting tumor growth in a murine melanoma model. Thus, VLPs employing lectin-driven immune reprogramming provide a framework for advancing cancer immunotherapies.
Collapse
Affiliation(s)
- Valerie Lensch
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adele Gabba
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Hincapie
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sachin H. Bhagchandani
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ankit Basak
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | | | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Alex K. Shalek
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jeremiah A. Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - M. G. Finn
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Laura L. Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
22
|
Anfray C, Varela CF, Ummarino A, Maeda A, Sironi M, Gandoy S, Brea J, Loza MI, León S, Calvo A, Correa J, Fernandez-Megia E, Alonso MJ, Allavena P, Crecente-Campo J, Andón FT. Polymeric nanocapsules loaded with poly(I:C) and resiquimod to reprogram tumor-associated macrophages for the treatment of solid tumors. Front Immunol 2024; 14:1334800. [PMID: 38259462 PMCID: PMC10800412 DOI: 10.3389/fimmu.2023.1334800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Background In the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer. Toll-like receptors (TLRs) ligands, such as poly(I:C) or resiquimod (R848) are able to reprogram TAMs towards M1-like antitumor effector cells. The objective of our work has been to develop and evaluate polymeric nanocapsules (NCs) loaded with poly(I:C)+R848, to improve drug stability and systemic toxicity, and evaluate their targeting and therapeutic activity towards TAMs in the TME of solid tumors. Methods NCs were developed by the solvent displacement and layer-by-layer methodologies and characterized by dynamic light scattering and nanoparticle tracking analysis. Hyaluronic acid (HA) was chemically functionalized with mannose for the coating of the NCs to target TAMs. NCs loaded with TLR ligands were evaluated in vitro for toxicity and immunostimulatory activity by Alamar Blue, ELISA and flow cytometry, using primary human monocyte-derived macrophages. For in vivo experiments, the CMT167 lung cancer model and the MN/MCA1 fibrosarcoma model metastasizing to lungs were used; tumor-infiltrating leukocytes were evaluated by flow cytometry and multispectral immunophenotyping. Results We have developed polymeric NCs loaded with poly(I:C)+R848. Among a series of 5 lead prototypes, protamine-NCs were selected based on their physicochemical properties (size, charge, stability) and in vitro characterization, showing good biocompatibility on primary macrophages and ability to stimulate their production of T-cell attracting chemokines (CXCL10, CCL5) and to induce M1-like macrophages cytotoxicity towards tumor cells. In mouse tumor models, the intratumoral injection of poly(I:C)+R848-protamine-NCs significantly prevented tumor growth and lung metastasis. In an orthotopic murine lung cancer model, the intravenous administration of poly(I:C)+R848-prot-NCs, coated with an additional layer of HA-mannose to improve TAM-targeting, resulted in good antitumoral efficacy with no apparent systemic toxicity. While no significant alterations were observed in T cell numbers (CD8, CD4 or Treg), TAM-reprogramming in treated mice was confirmed by the relative decrease of interstitial versus alveolar macrophages, having higher CD86 expression but lower CD206 and Arg1 expression in the same cells, in treated mice. Conclusion Mannose-HA-protamine-NCs loaded with poly(I:C)+R848 successfully reprogram TAMs in vivo, and reduce tumor progression and metastasis spread in mouse tumors.
Collapse
Affiliation(s)
- Clément Anfray
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Carmen Fernández Varela
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Aldo Ummarino
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Akihiro Maeda
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Marina Sironi
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Sara Gandoy
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- BioFarma Research Group, CIMUS, Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose Brea
- BioFarma Research Group, CIMUS, Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María Isabel Loza
- BioFarma Research Group, CIMUS, Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Sergio León
- Navarra Institute for Health Research (IdiSNA), Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Alfonso Calvo
- Navarra Institute for Health Research (IdiSNA), Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Juan Correa
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Eduardo Fernandez-Megia
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paola Allavena
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - José Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Fernando Torres Andón
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Oncology Department, Complexo Hospitalario de A Coruña (CHUAC), A Coruña, Spain
| |
Collapse
|
23
|
Prange CJ, Hu X, Tang L. Smart chemistry for traceless release of anticancer therapeutics. Biomaterials 2023; 303:122353. [PMID: 37925794 DOI: 10.1016/j.biomaterials.2023.122353] [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: 07/11/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
In the design of delivery strategies for anticancer therapeutics, the controlled release of intact cargo at the destined tumor and metastasis locations is of particular importance. To this end, stimuli-responsive chemical linkers have been extensively investigated owing to their ability to respond to tumor-specific physiological stimuli, such as lowered pH, altered redox conditions, increased radical oxygen species and pathological enzymatic activities. To prevent premature action and off-target effects, anticancer therapeutics are chemically modified to be transiently inactivated, a strategy known as prodrug development. Prodrugs are reactivated upon stimuli-dependent release at the sites of interest. As most drugs and therapeutic proteins have the optimal activity when released from carriers in their native and original forms, traceless release mechanisms are increasingly investigated. In this review, we summarize the chemical toolkit for developing innovative traceless prodrug strategies for stimuli-responsive drug delivery and discuss the applications of these chemical modifications in anticancer treatment including cancer immunotherapy.
Collapse
Affiliation(s)
- Céline Jasmin Prange
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland
| | - Xile Hu
- Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Materials Science & Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| |
Collapse
|
24
|
Davis MA, Cho E, Teplensky MH. Harnessing biomaterial architecture to drive anticancer innate immunity. J Mater Chem B 2023; 11:10982-11005. [PMID: 37955201 DOI: 10.1039/d3tb01677c] [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] [Indexed: 11/14/2023]
Abstract
Immunomodulation is a powerful therapeutic approach that harnesses the body's own immune system and reprograms it to treat diseases, such as cancer. Innate immunity is key in mobilizing the rest of the immune system to respond to disease and is thus an attractive target for immunomodulation. Biomaterials have widely been employed as vehicles to deliver immunomodulatory therapeutic cargo to immune cells and raise robust antitumor immunity. However, it is key to consider the design of biomaterial chemical and physical structure, as it has direct impacts on innate immune activation and antigen presentation to stimulate downstream adaptive immunity. Herein, we highlight the widespread importance of structure-driven biomaterial design for the delivery of immunomodulatory cargo to innate immune cells. The incorporation of precise structural elements can be harnessed to improve delivery kinetics, uptake, and the targeting of biomaterials into innate immune cells, and enhance immune activation against cancer through temporal and spatial processing of cargo to overcome the immunosuppressive tumor microenvironment. Structural design of immunomodulatory biomaterials will profoundly improve the efficacy of current cancer immunotherapies by maximizing the impact of the innate immune system and thus has far-reaching translational potential against other diseases.
Collapse
Affiliation(s)
- Meredith A Davis
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Ezra Cho
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Michelle H Teplensky
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
- Department of Materials Science and Engineering, Boston University, Boston, Massachusetts, 02215, USA
| |
Collapse
|
25
|
Brown CM, Husted KEL, Wang Y, Kilgallon LJ, Shieh P, Zafar H, Lundberg DJ, Johnson JA. Thiol-triggered deconstruction of bifunctional silyl ether terpolymers via an S NAr-triggered cascade. Chem Sci 2023; 14:8869-8877. [PMID: 37621440 PMCID: PMC10445473 DOI: 10.1039/d3sc02868b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/12/2023] [Indexed: 08/26/2023] Open
Abstract
While Si-containing polymers can often be deconstructed using chemical triggers such as fluoride, acids, and bases, they are resistant to cleavage by mild reagents such as biological nucleophiles, thus limiting their end-of-life options and potential environmental degradability. Here, using ring-opening metathesis polymerization, we synthesize terpolymers of (1) a "functional" monomer (e.g., a polyethylene glycol macromonomer or dicyclopentadiene); (2) a monomer containing an electrophilic pentafluorophenyl (PFP) substituent; and (3) a cleavable monomer based on a bifunctional silyl ether . Exposing these polymers to thiols under basic conditions triggers a cascade of nucleophilic aromatic substitution (SNAr) at the PFP groups, which liberates fluoride ions, followed by cleavage of the backbone Si-O bonds, inducing polymer backbone deconstruction. This method is shown to be effective for deconstruction of polyethylene glycol (PEG) based graft terpolymers in organic or aqueous conditions as well as polydicyclopentadiene (pDCPD) thermosets, significantly expanding upon the versatility of bifunctional silyl ether based functional polymers.
Collapse
Affiliation(s)
- Christopher M Brown
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Keith E L Husted
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Yuyan Wang
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Landon J Kilgallon
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Peyton Shieh
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Hadiqa Zafar
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - David J Lundberg
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
- Department of Chemical Engineering, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge Massachusetts 02139 USA
| |
Collapse
|