1
|
Shalmani AA, Daware R, Elshafei AS, De Lorenzi F, Moeckel D, Buhl EM, Klinkhammer BM, Boor P, Banala S, Adams A, Kiessling F, Storm G, Metselaar JM, Sofias AM, Shi Y, Lammers T, Peña Q, Ojha T. Hydrophobic ion pairing enables co-loading of water-soluble drugs in polymeric micelles. J Control Release 2025; 382:113748. [PMID: 40252978 DOI: 10.1016/j.jconrel.2025.113748] [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: 10/28/2024] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Polymeric micelles are widely used for the delivery of hydrophobic drugs. However, several highly potent and ubiquitously used anticancer drugs are water-soluble, complicating their (co-)formulation in polymeric micelles without having to chemically modify them. We here explore hydrophobic ion pairing to enable the co-delivery of the water-soluble anthracycline chemotherapy drug doxorubicin and the angiotensin II receptor antagonist telmisartan (a clinically used antihypertensive drug that has shown promising (pre-) clinical outcomes in combination with anthracyclines). We show that hydrophobic ion pairing of doxorubicin and telmisartan promotes the co-encapsulation of both drugs in π electron-stabilized [PEG-b-p(HPMAm-Bz)]-based polymeric micelles. The cytotoxic activity of doxorubicin is retained, and the dual drug-loaded micelles display enhanced antitumor activity in vivo as compared to the combination of the free drugs, while also exhibiting good tolerability. Taken together, this work provides proof-of-concept for hydrophobic ion pairing as a promising formulation strategy to promote multidrug nanomedicine and drug combination therapy.
Collapse
Affiliation(s)
- Armin Azadkhah Shalmani
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Rasika Daware
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Asmaa Said Elshafei
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Federica De Lorenzi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Diana Moeckel
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Eva M Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | | | - Peter Boor
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Srinivas Banala
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alina Adams
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584CG Utrecht, the Netherlands; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
| | - Josbert M Metselaar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Yang Shi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | - Quim Peña
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | - Tarun Ojha
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584CG Utrecht, the Netherlands.
| |
Collapse
|
2
|
Han Y, Jiang M, Sun Y, Chen W, Zhao Y, Guan X, Zhang W. Efficient chemo-immunotherapy leveraging minimalist electrostatic complex nanoparticle as "in situ" vaccine integrated tumor ICD and immunoagonist. J Adv Res 2025; 69:169-179. [PMID: 38499244 PMCID: PMC11954839 DOI: 10.1016/j.jare.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
INTRODUCTION Immunotherapy has unprecedentedly opened up a series of neoteric tactics for cancer treatment. As a burgeoning approach, chemo-immunotherapy has innovatively expanded the accomplishments of conventional chemotherapeutic agents for cancer governing. OBJECTIVES An efficacious chemo-immunotherapy leveraging minimalist electrostatic complex nanoparticle (NP) integrated tumor immunogenic cell death (ICD) and immunoagonist was developed as a watertight "in situ" vaccine for cancer therapy through convenient intratumoral administration with minimized systemic toxicity. METHODS Chemical-modified pH-sensitive cis-aconityl-doxorubicin (CAD) and immunoadjuvant unmethylated cytosine-phosphate-guanine (CpG) were co-packaged by polycationic polyethylenimine (PEI) though electrostatic-interaction to construct PEI/CpG/CAD NP. By intratumoral injection, this positively charged NP could be detained at tumor site and endocytosed by tumor cells effortlessly. Then, doxorubicin was released through cis-aconityl cleavage induced by endosomal-acidity and further triggered tumor ICD, the moribund tumor cells could release damage-associated molecular patterns (DAMPs) to recruit dendritic cells (DCs). Meanwhile, the entire tumor debris derived into diversified antigens and cooperated with immunostimulatory CpG to excite DC maturation and activated comprehensive antitumor immunity. RESULTS Prominent tumor suppression was achieved in aggressive mouse melanoma tumor model, which verified the feasibility and effectiveness of this minimalist CAD/CpG-codelivered NP. CONCLUSION This study has provided a convenient and promising paradigm for potent cancer chemo-immunotherapy.
Collapse
Affiliation(s)
- Yunfei Han
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Mingxia Jiang
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Yanju Sun
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Wenqiang Chen
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Yanli Zhao
- Shouguang Market Supervision and Administration Bureau, Shouguang 262700, China
| | - Xiuwen Guan
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China; Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang 261053, China.
| | - Weifen Zhang
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China; Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang 261053, China.
| |
Collapse
|
3
|
Heck AG, Medina-Montano C, Zhong Z, Deswarte K, Eigen K, Stickdorn J, Kockelmann J, Scherger M, Sanders NN, Lienenklaus S, Lambrecht BN, Grabbe S, De Geest BG, Nuhn L. PH-Triggered, Lymph Node Focused Immunodrug Release by Polymeric 2-Propionic-3-Methyl-maleic Anhydrides with Cholesteryl End Groups. Adv Healthc Mater 2024; 13:e2402875. [PMID: 39313985 DOI: 10.1002/adhm.202402875] [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: 08/17/2024] [Indexed: 09/25/2024]
Abstract
Gaining spatial control over innate immune activation is of great relevance during vaccine delivery and anticancer therapy, where one aims at activating immune cells at draining lymphoid tissue while avoiding systemic off-target innate immune activation. Lipid-polymer amphiphiles show high tendency to drain to lymphoid tissue upon local administration. Here, pH-sensitive, cholesteryl end group functionalized polymers as stimuli-responsive carriers are introduced for controlled immunoactivation of draining lymph nodes. Methacrylamide-based monomers bearing pendant 2-propionic-3-methylmaleic anhydride groups are polymerized by Reversible Addition-Fragmentation Chain Transfer (RAFT) polymerization using a cholesterol chain-transfer agent (chol-CTA). The amine-reactive anhydrides are conjugated with various amines, however, while primary amines afforded irreversible imides, secondary amines provided pH-responsive conjugates that are released upon acidification. This can be applied to fluorescent dyes for irreversibly carrier labeling or immunostimulatory Toll-like receptor (TLR) 7/8 agonists as cargos for pH-responsive delivery. Hydrophilization of remaining anhydride repeating units with short PEG-chains yielded cholesteryl-polymer amphiphiles that showed efficient cellular uptake and increased drug release at endosomal pH. Moreover, reversibly conjugated TLR 7/8 agonist amphiphiles efficiently drained to lymph nodes and increased the number of effectively maturated antigen-presenting cells after subcutaneous injection in vivo. Consequently, cholesteryl-linked methacrylamide-based polymers with pH-sensitive 2-propionic-3-methylmaleic anhydride side groups provide ideal features for immunodrug delivery.
Collapse
Affiliation(s)
- Alina G Heck
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Zifu Zhong
- Department of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, 9000, Belgium
| | - Kim Deswarte
- Department of Internal Medicine and Pediatrics, VIB Center for Inflammation Research, Ghent University, Ghent, 9052, Belgium
| | - Katharina Eigen
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
| | - Judith Stickdorn
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Johannes Kockelmann
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
| | | | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, 9820, Belgium
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science and Institute of Immunology, Hannover Medical School, 30625, Hanover, Germany
| | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, VIB Center for Inflammation Research, Ghent University, Ghent, 9052, Belgium
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Bruno G De Geest
- Department of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, 9000, Belgium
| | - Lutz Nuhn
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| |
Collapse
|
4
|
Li HX, Gong YW, Yan PJ, Xu Y, Qin G, Wen WP, Teng FY. Revolutionizing head and neck squamous cell carcinoma treatment with nanomedicine in the era of immunotherapy. Front Immunol 2024; 15:1453753. [PMID: 39676875 PMCID: PMC11638222 DOI: 10.3389/fimmu.2024.1453753] [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: 06/24/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a prevalent malignant tumor globally. Despite advancements in treatment methods, the overall survival rate remains low due to limitations such as poor targeting and low bioavailability, which result in the limited efficacy of traditional drug therapies. Nanomedicine is considered to be a promising strategy in tumor therapy, offering the potential for maximal anti-tumor effects. Nanocarriers can overcome biological barriers, enhance drug delivery efficiency to targeted sites, and minimize damage to normal tissues. Currently, various nano-carriers for drug delivery have been developed to construct new nanomedicine. This review aims to provide an overview of the current status of HNSCC treatment and the necessity of nanomedicine in improving treatment outcomes. Moreover, it delves into the research progress of nanomedicine in HNSCC treatment, with a focus on enhancing radiation sensitivity, improving the efficacy of tumor immunotherapy, effectively delivering chemotherapy drugs, and utilizing small molecule inhibitors. Finally, this article discussed the challenges and prospects of applying nanomedicine in cancer treatment.
Collapse
Affiliation(s)
- Hong-Xia Li
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Yu-Wen Gong
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Pi-Jun Yan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Yong Xu
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei-Ping Wen
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fang-Yuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| |
Collapse
|
5
|
Park S, Jin SM, Kim S, Cho JH, Hong J, Bae YS, Lim YT. Bioconjugated Antibody-Trojan Immune Converter Enhance Cancer Immunotherapy with Minimized Toxicity by Programmed Two-Step Immunomodulation of Myeloid Cells. Adv Healthc Mater 2024; 13:e2401270. [PMID: 38801164 DOI: 10.1002/adhm.202401270] [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: 04/07/2024] [Indexed: 05/29/2024]
Abstract
Current immune checkpoint blockade therapy (ICBT) predominantly targets T cells to harness the antitumor effects of adaptive immune system. However, the effectiveness of ICBT is reduced by immunosuppressive innate myeloid cells in tumor microenvironments (TMEs). Toll-like receptor 7/8 agonists (TLR7/8a) are often used to address this problem because they can reprogram myeloid-derived suppressor cells (MDSCs) and tumor-associated M2 macrophages, and boost dendritic cell (DC)-based T-cell generation; however, the systemic toxicity of TLR7/8a limits its clinical translation. Here, to address this limitation and utilize the effectiveness of TLR7/8a, this work suggests a programmed two-step activation strategy via Antibody-Trojan Immune Converter Conjugates (ATICC) that specifically targets myeloid cells by anti-SIRPα followed by reactivation of transiently inactivated Trojan TLR7/8a after antibody-mediated endocytosis. ATICC blocks the CD47-SIRPα ("don't eat me" signal), enhances phagocytosis, reprograms M2 macrophages and MDSCs, and increases cross-presentation by DCs, resulting in antigen-specific CD8+ T-cell generation in tumor-draining lymph nodes and TME while minimizing systemic toxicity. The local or systemic administration of ATICC improves ICBT responsiveness through reprogramming of the immunosuppressive TME, increased infiltration of antigen-specific CD8+ T cells, and antibody-dependent cellular phagocytosis. These results highlight the programmed and target immunomodulation via ATICC could enhance cancer immunotherapy with minimized systemic toxicities.
Collapse
Affiliation(s)
- Soyoung Park
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, 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, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Suhyeon Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Ju Hee Cho
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - JungHyub Hong
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Department of Biological Science, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Department of Biological Science, Sungkyunkwan University, 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, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| |
Collapse
|
6
|
Yu J, Yu X, Sun X, Wang Q, Long S, Ren R, Guan Z, Yang Z. Bis-2'-F-cG SA SMP isomers encapsulated in cytidinyl/cationic lipids act as potent in situ autologous tumor vaccines. Mol Ther 2024; 32:1917-1933. [PMID: 38637990 PMCID: PMC11184333 DOI: 10.1016/j.ymthe.2024.04.023] [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: 07/04/2023] [Revised: 10/23/2023] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
Cancer immunotherapy has greatly improved the prognosis of tumor-bearing patients. Nevertheless, cancer patients exhibit low response rates to current immunotherapy drugs, such as PD1 and PDL1 antibodies. Cyclic dinucleotide analogs are a promising class of immunotherapeutic agents. In this study, in situ autologous tumor vaccines, composed of bis-2'-F-cGSASMP phosphonothioate isomers (FGA-di-pS-2 or FGA-di-pS-4) and cytidinyl/cationic lipids (Mix), were constructed. Intravenous and intratumoral injection of FGA-di-pS-2/Mix or FGA-di-pS-4/Mix enhanced the immunogenic cell death of tumor cells in vivo, leading to the exposure and presentation of whole tumor antigens, inhibiting tumor growth in both LLC and EO771 tumor in situ murine models and increasing their survival rates to 50% and 23%, respectively. Furthermore, the tumor-bearing mice after treatment showed potent immune memory efficacy and exhibited 100% protection against tumor rechallenge. Intravenous administration of FGA-di-pS-2/Mix potently promoted DC maturation, M1 macrophage polarization and CD8+ T cell activation and decreased the proportion of Treg cells in the tumor microenvironment. Notably, two doses of ICD-debris (generated by FGA-di-pS-2 or 4/Mix-treated LLC cells) protected 100% of mice from tumor growth. These tumor vaccines showed promising results and may serve as personalized cancer vaccinations in the future.
Collapse
Affiliation(s)
- Jing Yu
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Xiaotong Yu
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China; Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing 100191, China
| | - Xudong Sun
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Quanxin Wang
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Sijie Long
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Runan Ren
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Zhu Guan
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Zhenjun Yang
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China.
| |
Collapse
|
7
|
Ku KS, Tang J, Chen Y, Shi Y. Current Advancements in Anti-Cancer Chimeric Antigen Receptor T Cell Immunotherapy and How Nanotechnology May Change the Game. Int J Mol Sci 2024; 25:5361. [PMID: 38791398 PMCID: PMC11120994 DOI: 10.3390/ijms25105361] [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: 03/13/2024] [Revised: 04/27/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell immunotherapy represents a cutting-edge advancement in the landscape of cancer treatment. This innovative therapy has shown exceptional promise in targeting and eradicating malignant tumors, specifically leukemias and lymphomas. However, despite its groundbreaking successes, (CAR)-T cell therapy is not without its challenges. These challenges, particularly pronounced in the treatment of solid tumors, include but are not limited to, the selection of appropriate tumor antigens, managing therapy-related toxicity, overcoming T-cell exhaustion, and addressing the substantial financial costs associated with treatment. Nanomedicine, an interdisciplinary field that merges nanotechnology with medical science, offers novel strategies that could potentially address these limitations. Its application in cancer treatment has already led to significant advancements, including improved specificity in drug targeting, advancements in cancer diagnostics, enhanced imaging techniques, and strategies for long-term cancer prevention. The integration of nanomedicine with (CAR)-T cell therapy could revolutionize the treatment landscape by enhancing the delivery of genes in (CAR)-T cell engineering, reducing systemic toxicity, and alleviating the immunosuppressive effects within the tumor microenvironment. This review aims to explore how far (CAR)-T cell immunotherapy has come alone, and how nanomedicine could strengthen it into the future. Additionally, the review will examine strategies to limit the off-target effects and systemic toxicity associated with (CAR)-T cell therapy, potentially enhancing patient tolerance and treatment outcomes.
Collapse
Affiliation(s)
- Kimberly S. Ku
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (K.S.K.); (J.T.)
| | - Jie Tang
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (K.S.K.); (J.T.)
| | - Yuan Chen
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (K.S.K.); (J.T.)
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA
| |
Collapse
|
8
|
An L, De Bruyn T, Pang J, Ubhayakar S, Salphati L, Zhang X, Liu L, Li R, Chan B, Dey A, Levy ES. Early Stage Preclinical Formulation Strategies to Alter the Pharmacokinetic Profile of Two Small Molecule Therapeutics. Pharmaceuticals (Basel) 2024; 17:179. [PMID: 38399394 PMCID: PMC10892288 DOI: 10.3390/ph17020179] [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: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Early stage chemical development presents numerous challenges, and achieving a functional balance is a major hurdle, with many early compounds not meeting the clinical requirements for advancement benchmarks due to issues like poor oral bioavailability. There is a need to develop strategies for achieving the desired systemic concentration for these compounds. This will enable further evaluation of the biological response upon a compound-target interaction, providing deeper insight into the postulated biological pathways. Our study elucidates alternative drug delivery paradigms by comparing formulation strategies across oral (PO), intraperitoneal (IP), subcutaneous (SC), and intravenous (IV) routes. While each modality boasts its own set of merits and constraints, it is the drug's formulation that crucially influences its pharmacokinetic (PK) trajectory and the maintenance of its therapeutic levels. Our examination of model compounds G7883 and G6893 highlighted their distinct physio-chemical attributes. By harnessing varied formulation methods, we sought to fine-tune their PK profiles. PK studies showcased G7883's extended half-life using an SC oil formulation, resulting in a 4.5-fold and 2.5-fold enhancement compared with the IP and PO routes, respectively. In contrast, with G6893, we achieved a prolonged systemic coverage time above the desired target concentration through a different approach using an IV infusion pump. These outcomes underscore the need for tailored formulation strategies, which are dictated by the compound's innate properties, to reach the optimal in vivo systemic concentrations. Prioritizing formulation and delivery optimization early on is pivotal for effective systemic uptake, thereby facilitating a deeper understanding of biological pathways and expediting the overall clinical drug development timeline.
Collapse
Affiliation(s)
- Le An
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA;
| | - Tom De Bruyn
- Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (T.D.B.); (J.P.); (S.U.); (L.S.); (X.Z.); (L.L.); (R.L.)
| | - Jodie Pang
- Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (T.D.B.); (J.P.); (S.U.); (L.S.); (X.Z.); (L.L.); (R.L.)
| | - Savita Ubhayakar
- Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (T.D.B.); (J.P.); (S.U.); (L.S.); (X.Z.); (L.L.); (R.L.)
| | - Laurent Salphati
- Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (T.D.B.); (J.P.); (S.U.); (L.S.); (X.Z.); (L.L.); (R.L.)
| | - Xing Zhang
- Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (T.D.B.); (J.P.); (S.U.); (L.S.); (X.Z.); (L.L.); (R.L.)
| | - Liling Liu
- Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (T.D.B.); (J.P.); (S.U.); (L.S.); (X.Z.); (L.L.); (R.L.)
| | - Ruina Li
- Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (T.D.B.); (J.P.); (S.U.); (L.S.); (X.Z.); (L.L.); (R.L.)
| | - Bryan Chan
- Discovery Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA;
| | - Anwesha Dey
- Discovery Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA;
| | - Elizabeth S. Levy
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA;
| |
Collapse
|
9
|
Heck AG, Stickdorn J, Rosenberger LJ, Scherger M, Woller J, Eigen K, Bros M, Grabbe S, Nuhn L. Polymerizable 2-Propionic-3-methylmaleic Anhydrides as a Macromolecular Carrier Platform for pH-Responsive Immunodrug Delivery. J Am Chem Soc 2023; 145:27424-27436. [PMID: 38054646 DOI: 10.1021/jacs.3c08511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
The design of functional polymers coupled with stimuli-triggered drug release mechanisms is a promising achievement to overcome various biological barriers. pH trigger methods yield significant potential for controlled targeting and release of therapeutics due to their simplicity and relevance, especially upon cell internalization. Here, we introduce reactive polymers that conjugate primary or secondary amines and release potential drugs under acidic conditions. For that purpose, we introduced methacrylamide-based monomers with pendant 2-propionic-3-methylmaleic anhydride groups. Such groups allow the conjugation of primary and secondary amines but are resistant to radical polymerization conditions. We, therefore, polymerized 2-propionic-3-methylmaleic anhydride amide-based methacrylates via reversible addition-fragmentation chain transfer (RAFT) polymerization. Their amine-reactive anhydrides could sequentially be derivatized by primary or secondary amines into hydrophilic polymers. Acidic pH-triggered drug release from the polymeric systems was fine-tuned by comparing different amines. Thereby, the conjugation of primary amines led to the formation of irreversible imide bonds in dimethyl sulfoxide, while secondary amines could quantitatively be released upon acidification. In vitro, this installed pH-responsiveness can contribute to an effective release of conjugated immune stimulatory drugs under endosomal pH conditions. Interestingly, the amine-modified polymers generally showed no toxicity and a high cellular uptake. Furthermore, secondary amine-modified immune stimulatory drugs conjugated to the polymers yielded better receptor activity and immune cell maturation than their primary amine derivatives due to their pH-sensitive drug release mechanism. Consequently, 2-propionic-3-methylmaleic anhydride-based polymers can be considered as a versatile platform for pH-triggered delivery of various (immuno)drugs, thus enabling new strategies in macromolecule-assisted immunotherapy.
Collapse
Affiliation(s)
- Alina G Heck
- Max Planck Institute for Polymer Research, Mainz 55128, Germany
| | | | - Laura J Rosenberger
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | | | - Jonas Woller
- Max Planck Institute for Polymer Research, Mainz 55128, Germany
| | - Katharina Eigen
- Institute of Functional Materials and Biofabrication, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Würzburg 97070, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research, Mainz 55128, Germany
- Institute of Functional Materials and Biofabrication, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Würzburg 97070, Germany
| |
Collapse
|
10
|
Qu X, Zhou D, Lu J, Qin D, Zhou J, Liu HJ. Cancer nanomedicine in preoperative therapeutics: Nanotechnology-enabled neoadjuvant chemotherapy, radiotherapy, immunotherapy, and phototherapy. Bioact Mater 2023; 24:136-152. [PMID: 36606253 PMCID: PMC9792706 DOI: 10.1016/j.bioactmat.2022.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Surgical resection remains a mainstay in the treatment of malignant solid tumors. However, the use of neoadjuvant treatments, including chemotherapy, radiotherapy, phototherapy, and immunotherapy, either alone or in combination, as a preoperative intervention regimen, have attracted increasing attention in the last decade. Early randomized, controlled trials in some tumor settings have not shown a significant difference between the survival rates in long-term neoadjuvant therapy and adjuvant therapy. However, this has not hampered the increasing use of neoadjuvant treatments in clinical practice, due to its evident downstaging of primary tumors to delineate the surgical margin, tailoring systemic therapy response as a clinical tool to optimize subsequent therapeutic regimens, and decreasing the need for surgery, with its potential for increased morbidity. The recent expansion of nanotechnology-based nanomedicine and related medical technologies provides a new approach to address the current challenges of neoadjuvant therapy for preoperative therapeutics. This review not only summarizes how nanomedicine plays an important role in a range of neoadjuvant therapeutic modalities, but also highlights the potential use of nanomedicine as neoadjuvant therapy in preclinical and clinic settings for tumor management.
Collapse
Affiliation(s)
- Xiaogang Qu
- Department of General Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, 215500, China
| | - Dong Zhou
- Department of General Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, 215500, China
| | - Jianpu Lu
- Department of General Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, 215500, China
| | - Duotian Qin
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jun Zhou
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hai-Jun Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
11
|
Manna S, Maiti S, Shen J, Weiss A, Mulder E, Du W, Esser-Kahn AP. Nanovaccine that activates the NLRP3 inflammasome enhances tumor specific activation of anti-cancer immunity. Biomaterials 2023; 296:122062. [PMID: 36863071 PMCID: PMC10085859 DOI: 10.1016/j.biomaterials.2023.122062] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Neoantigen cancer vaccines that target tumor specific mutations are emerging as a promising modality for cancer immunotherapy. To date, various approaches have been adopted to enhance efficacy of these therapies, but the low immunogenicity of neoantigens has hindered clinical application. To address this challenge, we developed a polymeric nanovaccine platform that activates the NLRP3 inflammasome, a key immunological signaling pathway in pathogen recognition and clearance. The nanovaccine is comprised of a poly (orthoester) scaffold engrafted with a small-molecule TLR7/8 agonist and an endosomal escape peptide that facilitates lysosomal rupture and NLRP3 inflammasome activation. Upon solvent transfer, the polymer self-assembles with neoantigens to form ∼50 nm nanoparticles that facilitate co-delivery to antigen-presenting cells. This polymeric activator of the inflammasome (PAI) was found to induce potent antigen-specific CD8+ T cell responses characterized by IFN-γ and GranzymeB secretion. Moreover, in combination with immune checkpoint blockade therapy, the nanovaccine stimulated robust anti-tumor immune responses against established tumors in EG.7-OVA, B16·F10, and CT-26 models. Results from our studies indicate that NLRP3 inflammasome activating nanovaccines demonstrate promise for development as a robust platform to enhance immunogenicity of neoantigen therapies.
Collapse
Affiliation(s)
- Saikat Manna
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA
| | - Sampa Maiti
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA; Department of Chemistry and Biochemistry, Science of Advanced Material, Central Michigan University, Mount Pleasant, MI 48858, United States
| | - Jingjing Shen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA
| | - Adam Weiss
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA; Department of Chemistry, University of Chicago, 5735 S Ellis Ave., Chicago, IL 60637, USA
| | - Elizabeth Mulder
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA
| | - Wenjun Du
- Department of Chemistry and Biochemistry, Science of Advanced Material, Central Michigan University, Mount Pleasant, MI 48858, United States
| | - Aaron P Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA.
| |
Collapse
|
12
|
Son H, Shin J, Park J. Recent progress in nanomedicine-mediated cytosolic delivery. RSC Adv 2023; 13:9788-9799. [PMID: 36998521 PMCID: PMC10043881 DOI: 10.1039/d2ra07111h] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Cytosolic delivery of bioactive agents has exhibited great potential to cure undruggable targets and diseases. Because biological cell membranes are a natural barrier for living cells, efficient delivery methods are required to transfer bioactive and therapeutic agents into the cytosol. Various strategies that do not require cell invasive and harmful processes, such as endosomal escape, cell-penetrating peptides, stimuli-sensitive delivery, and fusogenic liposomes, have been developed for cytosolic delivery. Nanoparticles can easily display functionalization ligands on their surfaces, enabling many bio-applications for cytosolic delivery of various cargo, including genes, proteins, and small-molecule drugs. Cytosolic delivery uses nanoparticle-based delivery systems to avoid degradation of proteins and keep the functionality of other bioactive molecules, and functionalization of nanoparticle-based delivery vehicles imparts a specific targeting ability. With these advantages, nanomedicines have been used for organelle-specific tagging, vaccine delivery for enhanced immunotherapy, and intracellular delivery of proteins and genes. Optimization of the size, surface charges, specific targeting ability, and composition of nanoparticles is needed for various cargos and target cells. Toxicity issues with the nanoparticle material must be managed to enable clinical use.
Collapse
Affiliation(s)
- Hangyu Son
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea 222 Banpo-daero, Seocho-gu Seoul 06591 Republic of Korea
| | - Jeongsu Shin
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea 222 Banpo-daero, Seocho-gu Seoul 06591 Republic of Korea
| | - Joonhyuck Park
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea 222 Banpo-daero, Seocho-gu Seoul 06591 Republic of Korea
| |
Collapse
|
13
|
Song W, Zhang X, Song Y, Fan K, Shao F, Long Y, Gao Y, Cai W, Lan X. Enhancing Photothermal Therapy Efficacy by In Situ Self-Assembly in Glioma. ACS APPLIED MATERIALS & INTERFACES 2023; 15:57-66. [PMID: 36206382 PMCID: PMC9839507 DOI: 10.1021/acsami.2c14413] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The residence time of some small molecular imaging and therapeutic agents in tumor tissue is short and the molecules can be easily dispersed, which decreases treatment efficacy. Therefore, methods that enhance oncotherapy performance are of significant importance. Here, we report an in situ self-assembly strategy aimed at enhancing the photothermal therapy of glioblastomas. The probe, ICG-PEP-c(RGD)fk, consisted of a glutathione-reactive self-assembling polypeptide as the skeleton, indocyanine green (ICG) as a theranostic agent, and cyclic Arg-Gly-Asp [c(RGD)fk] peptides as the targeting group. ICG-PEP-c(RGD)fk was synthesized and found to be assembled in the glutathione environment at 9.446 μM in vitro. Human glioblastoma cell line U87MG-luc with high integrin αvβ3 expression was applied to invivo experiments. ICG-PEP-c(RGD)fk provided clearer tumor imaging and had a tumor retention time of 6.12 times longer than that of ICG-c(RGD)fk. In therapeutic experiments, ICG-PEP-c(RGD)fk significantly suppressed glioblastoma growth and the tumor volume was 2.61 times smaller than in the ICG-c(RGD)fk group at the end of the observation period. Moreover, the median survival time of ICG-PEP-c(RGD)fk group was significantly improved by 2.78 times compared with that of the control group. In conclusion, glutathione-reactive self-assembling peptides are capable of increasing the tumor retention time and improving the photothermal therapeutic effect. The in situ self-assembly strategy is a potential and feasible method to enhance oncotherapy.
Collapse
Affiliation(s)
- Wenyu Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022 Hubei Province, China
| | - Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022 Hubei Province, China
| | - Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022 Hubei Province, China
| | - Kevin Fan
- Department of Radiology and Department of Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin 53705, United States
| | - Fuqiang Shao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022 Hubei Province, China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022 Hubei Province, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022 Hubei Province, China
| | - Weibo Cai
- Department of Radiology and Department of Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin 53705, United States
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022 Hubei Province, China
| |
Collapse
|
14
|
Li JH, Huang LJ, Zhou HL, Shan YM, Chen FM, Lehto VP, Xu WJ, Luo LQ, Yu HJ. Engineered nanomedicines block the PD-1/PD-L1 axis for potentiated cancer immunotherapy. Acta Pharmacol Sin 2022; 43:2749-2758. [PMID: 35484402 PMCID: PMC9622913 DOI: 10.1038/s41401-022-00910-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Immunotherapy, in particular immune checkpoint blockade (ICB) therapy targeting the programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis, has remarkably revolutionized cancer treatment in the clinic. Anti-PD-1/PD-L1 therapy is designed to restore the antitumor response of cytotoxic T cells (CTLs) by blocking the interaction between PD-L1 on tumour cells and PD-1 on CTLs. Nevertheless, current anti-PD-1/PD-L1 therapy suffers from poor therapeutic outcomes in a large variety of solid tumours due to insufficient tumour specificity, severe cytotoxic effects, and the occurrence of immune resistance. In recent years, nanosized drug delivery systems (NDDSs), endowed with highly efficient tumour targeting and versatility for combination therapy, have paved a new avenue for cancer immunotherapy. In this review article, we summarized the recent advances in NDDSs for anti-PD-1/PD-L1 therapy. We then discussed the challenges and further provided perspectives to promote the clinical application of NDDS-based anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Jun-Hao Li
- College of Sciences, Shanghai University, Shanghai, 200444, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu-Jia Huang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Hui-Ling Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi-Ming Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang-Min Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Vesa-Pekka Lehto
- Department of Applied Physics, University of Eastern Finland, 70211, Kuopio, Finland
| | - Wu-Jun Xu
- Department of Applied Physics, University of Eastern Finland, 70211, Kuopio, Finland.
| | - Li-Qiang Luo
- College of Sciences, Shanghai University, Shanghai, 200444, China
| | - Hai-Jun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
15
|
Yang Y, Zhao T, Chen Q, Li Y, Xiao Z, Xiang Y, Wang B, Qiu Y, Tu S, Jiang Y, Nan Y, Huang Q, Ai K. Nanomedicine Strategies for Heating "Cold" Ovarian Cancer (OC): Next Evolution in Immunotherapy of OC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202797. [PMID: 35869032 PMCID: PMC9534959 DOI: 10.1002/advs.202202797] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/17/2022] [Indexed: 05/08/2023]
Abstract
Immunotherapy has revolutionized cancer treatment, dramatically improving survival rates of melanoma and lung cancer patients. Nevertheless, immunotherapy is almost ineffective against ovarian cancer (OC) due to its cold tumor immune microenvironment (TIM). Many traditional medications aimed at remodeling TIM are often associated with severe systemic toxicity, require frequent dosing, and show only modest clinical efficacy. In recent years, emerging nanomedicines have demonstrated extraordinary immunotherapeutic effects for OC by reversing the TIM because the physical and biochemical features of nanomedicines can all be harnessed to obtain optimal and expected tissue distribution and cellular uptake. However, nanomedicines are far from being widely explored in the field of OC immunotherapy due to the lack of appreciation for the professional barriers of nanomedicine and pathology, limiting the horizons of biomedical researchers and materials scientists. Herein, a typical cold tumor-OC is adopted as a paradigm to introduce the classification of TIM, the TIM characteristics of OC, and the advantages of nanomedicines for immunotherapy. Subsequently, current nanomedicines are comprehensively summarized through five general strategies to substantially enhance the efficacy of immunotherapy by heating the cold OC. Finally, the challenges and perspectives of this expanding field for improved development of clinical applications are also discussed.
Collapse
Affiliation(s)
- Yuqi Yang
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yumei Li
- Department of Assisted ReproductionXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yuting Xiang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Boyu Wang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yige Qiu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Shiqi Tu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yitian Jiang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yayun Nan
- Geriatric Medical CenterPeople's Hospital of Ningxia Hui Autonomous RegionYinchuanNingxia750002P. R. China
| | - Qiong Huang
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| |
Collapse
|
16
|
Liu Y, Li H, Zhao H, Hao Y, Van Herck S, Xu Z, Wang G, Wang X, Zhang X, Ge X, Li X, Yang A, Chen H, Zou J, Wang W, De Geest BG, Zhang Z. In Situ Tumor Vaccination with Calcium-Linked Degradable Coacervate Nanocomplex Co-Delivering Photosensitizer and TLR7/8 Agonist to Trigger Effective Anti-Tumor Immune Responses. Adv Healthc Mater 2022; 11:e2102781. [PMID: 35285581 DOI: 10.1002/adhm.202102781] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/24/2022] [Indexed: 12/19/2022]
Abstract
In situ anti-tumor vaccination is an attractive type of cancer immunotherapy which relies on the effectiveness of dendritic cells (DCs) to engulf tumor antigens, become activated, and present antigens to T cells in lymphoid tissue. Here, a multifunctional nanocomplex based on calcium crosslinked polyaspartic acid conjugated to either a toll-like receptor (TLR)7/8 agonist or a photosensitizer is reported. Intratumoral administration of the nanocomplex followed by laser irradiation induces cell killing and hence generation of a pool of tumor-associated antigens, with concomitant promotion of DCs maturation and expansion of T cells in tumor-draining lymph nodes. Suppression of tumor growth is observed both at the primary site and at the distal site, thereby hinting at successful induction of an adaptive anti-tumor response. This strategy holds promise for therapeutic application in a pre-operative and post-operative setting to leverage to mutanome of the patient's own tumor to mount immunological memory to clear residual tumor cells and metastasis.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Hui Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, P. R. China
| | - Yanyun Hao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Simon Van Herck
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent, 9000, Belgium
| | - Zejun Xu
- Department of Natural Products Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, P. R. China
| | - Guan Wang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, P. R. China
| | - Xiao Wang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, P. R. China
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, P. R. China
| | - Xiaoyan Ge
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Xia Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Ailu Yang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, P. R. China
| | - Hongfei Chen
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Jing Zou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Wentao Wang
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, P. R. China
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent, 9000, Belgium
| | - Zhiyue Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| |
Collapse
|
17
|
Polyketal-based nanocarriers: A new class of stimuli-responsive delivery systems for therapeutic applications. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
18
|
Van Herck S, Feng B, Tang L. Delivery of STING agonists for adjuvanting subunit vaccines. Adv Drug Deliv Rev 2021; 179:114020. [PMID: 34756942 DOI: 10.1016/j.addr.2021.114020] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/16/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023]
Abstract
Adjuvant is an essential component in subunit vaccines. Many agonists of pathogen recognition receptors have been developed as potent adjuvants to optimize the immunogenicity and efficacy of vaccines. Recently discovered cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has attracted much attention as it is a key mediator for modulating immune responses. Vaccines adjuvanted with STING agonists are found to mediate a robust immune defense against infections and cancer. In this review, we first discuss the mechanisms of STING agonists in the context of vaccination. Next, we present recent progress in novel STING agonist discovery and the delivery strategies. We next highlight recent work in optimizing the efficacy while minimizing toxicity of STING agonist-assisted subunit vaccines for protection against infectious diseases or treatment of cancer. Finally, we share our perspectives of current issues and future directions in further developing STING agonists for adjuvanting subunit vaccines.
Collapse
Affiliation(s)
- Simon Van Herck
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Department of Pharmaceutics, Ghent University, 9000 Ghent, Belgium
| | - Bing Feng
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland.
| |
Collapse
|
19
|
Jeong S, Choi Y, Kim K. Engineering Therapeutic Strategies in Cancer Immunotherapy via Exogenous Delivery of Toll-like Receptor Agonists. Pharmaceutics 2021; 13:1374. [PMID: 34575449 PMCID: PMC8466827 DOI: 10.3390/pharmaceutics13091374] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 12/11/2022] Open
Abstract
As a currently spotlighted method for cancer treatment, cancer immunotherapy has made a lot of progress in recent years. Among tremendous cancer immunotherapy boosters available nowadays, Toll-like receptor (TLR) agonists were specifically selected, because of their effective activation of innate and adaptive immune cells, such as dendritic cells (DCs), T cells, and macrophages. TLR agonists can activate signaling pathways of DCs to express CD80 and CD86 molecules, and secrete various cytokines and chemokines. The maturation of DCs stimulates naïve T cells to differentiate into functional cells, and induces B cell activation. Although TLR agonists have anti-tumor ability by activating the immune system of the host, their drawbacks, which include poor efficiency and remarkably short retention time in the body, must be overcome. In this review, we classify and summarize the recently reported delivery strategies using (1) exogenous TLR agonists to maintain the biological and physiological signaling activities of cargo agonists, (2) usage of multiple TLR agonists for synergistic immune responses, and (3) co-delivery using the combination with other immunomodulators or stimulants. In contrast to naked TLR agonists, these exogenous TLR delivery strategies successfully facilitated immune responses and subsequently mediated anti-tumor efficacy.
Collapse
Affiliation(s)
| | | | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul 22012, Korea; (S.J.); (Y.C.)
| |
Collapse
|
20
|
Manna S, Maiti S, Shen J, Du W, Esser-Kahn AP. Pathogen-like Nanoassemblies of Covalently Linked TLR Agonists Enhance CD8 and NK Cell-Mediated Antitumor Immunity. ACS CENTRAL SCIENCE 2020; 6:2071-2078. [PMID: 33274283 PMCID: PMC7706081 DOI: 10.1021/acscentsci.0c01001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Indexed: 05/03/2023]
Abstract
Therapies based on Toll Like Receptor agonists (TLRa) are emerging as a promising modality for cancer immunotherapy to recruit antitumor T-cells in unresponsive immunologically "cold" tumors. Often, combinations of agonists are employed to synergistically enhance efficacy. However, low efficacy and severe toxicities deter these TLR-based therapeutics from further clinical applications. Studies have suggested that the rapid systemic diffusion of agonists to nontarget tissues is the primary cause. To address this challenge, we developed supramolecular nanotherapeutics of covalently linked TLRas for multivalent, synergistic interactions by drawing inspiration from immune recognition of pathogens. This new nanotherapeutic increased stimulation of key pro-inflammatory cytokines and remarkably enhanced CD8 and NK cell-mediated antitumor response while exhibiting ultralow off-target toxicity in an aggressive B16.F10 tumor model. Results from our studies thereby indicate that such supramolecular immune-agonist therapeutics may be further developed as a viable treatment modality for cancer immunotherapy.
Collapse
Affiliation(s)
- Saikat Manna
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Sampa Maiti
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Department
of Chemistry and Biochemistry, Science of Advanced Material, Central Michigan University, Mount Pleasant, Michigan 48858, United States
| | - Jingjing Shen
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Wenjun Du
- Department
of Chemistry and Biochemistry, Science of Advanced Material, Central Michigan University, Mount Pleasant, Michigan 48858, United States
- (W.D.)
| | - Aaron P. Esser-Kahn
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- (A.P.E-K.)
| |
Collapse
|
21
|
|