1
|
Zhao Y, Tian M, Tong X, Yang X, Gan L, Yong T. Emerging strategies in lymph node-targeted nano-delivery systems for tumor immunotherapy. Essays Biochem 2025; 69:EBC20253008. [PMID: 40159756 DOI: 10.1042/ebc20253008] [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: 01/13/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
The emergence of immunotherapy has led to the clinical approval of several related drugs. However, their efficacy against solid tumors remains limited. As the hub of immune activation, lymph nodes (LNs) play a critical role in tumor immunotherapy by initiating and amplifying immune responses. Nevertheless, the intricate physiological structure and barriers within LNs, combined with the immunosuppressive microenvironment induced by tumor cells, significantly impede the therapeutic efficacy of immunotherapy. Engineered nanoparticles (NPs) have shown great potential in overcoming these challenges by facilitating targeted drug transport to LNs and directly or indirectly activating T cells. This review systematically examines the structural features of LNs, key factors influencing the targeting efficiency of NPs, and current strategies for remodeling the immunosuppressive microenvironment of LNs. Additionally, it discusses future opportunities for optimizing NPs to enhance tumor immunotherapy, addressing challenges in clinical translation and safety evaluation.
Collapse
Affiliation(s)
- Yaoli Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Muzi Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Tong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
2
|
Cai Q, Guo R, Chen D, Deng Z, Gao J. SynBioNanoDesign: pioneering targeted drug delivery with engineered nanomaterials. J Nanobiotechnology 2025; 23:178. [PMID: 40050980 PMCID: PMC11884119 DOI: 10.1186/s12951-025-03254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/19/2025] [Indexed: 03/10/2025] Open
Abstract
Synthetic biology and nanotechnology fusion represent a transformative approach promoting fundamental and clinical biomedical science development. In SynBioNanoDesign, biological systems are reimagined as dynamic and programmable materials to yield engineered nanomaterials with emerging and specific functionalities. This review elucidates a comprehensive examination of synthetic biology's pivotal role in advancing engineered nanomaterials for targeted drug delivery systems. It begins with exploring the fundamental synergy between synthetic biology and nanotechnology, then highlights the current landscape of nanomaterials in targeted drug delivery applications. Subsequently, the review discusses the design of novel nanomaterials informed by biological principles, focusing on expounding the synthetic biology tools and the potential for developing advanced nanomaterials. Afterward, the research advances of innovative materials design through synthetic biology were systematically summarized, emphasizing the integration of genetic circuitry to program nanomaterial responses. Furthermore, the challenges, current weaknesses and opportunities, prospective directions, and ethical and societal implications of SynBioNanoDesign in drug delivery are elucidated. Finally, the review summarizes the transformative impact that synthetic biology may have on drug-delivery technologies in the future.
Collapse
Affiliation(s)
- Qian Cai
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, Fujian, China
| | - Rui Guo
- National and Local United Engineering Laboratory of Natural Biotoxin, College of Bee and Biomedical Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Dafu Chen
- National and Local United Engineering Laboratory of Natural Biotoxin, College of Bee and Biomedical Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, Joint International Laboratory on Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jiangtao Gao
- National and Local United Engineering Laboratory of Natural Biotoxin, College of Bee and Biomedical Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
3
|
Pandey A, Karmous I. Exploring the Potential of Plant-Based Nanotechnology in Cancer Immunotherapy: Benefits, Limitations, and Future Perspectives. Biol Trace Elem Res 2025; 203:1746-1763. [PMID: 38862749 DOI: 10.1007/s12011-024-04266-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Reconceptualizing cancer immunotherapy can be improved if combined with plant production systems and nanotechnology. This review aims to contribute to the knowledge of plant use in nanomedicine and cancer immunotherapy. In the foreground, we outlined each of these approaches; nanomedicine, green synthesis, and immunotherapy. The benefits of plant-based nanoparticles in mending the immune systems were subsequently analyzed, with reference to the literature. The combining effects of biological and therapeutic properties of some phytochemicals and their derivatives, with targeted nanoparticles and selective immunotherapy, can enhance the delivery of drugs and antibodies, and induce antitumor immune responses, via activation of functions of neutrophils, lymphocyte cells, and natural killer cells, and macrophages, resulting in induced apoptosis and phagocytosis of tumor cells, which can improve designing immunotherapeutic strategies targeting cancer, with a larger spectrum compared to the current cytotoxic anticancer drugs commonly used in clinics. This study uncovers the mechanistic drivers of cancer immunoengineering in cancer therapy using plant-based nanomaterials, enhancing therapeutic benefits while minimizing toxic and side effects.
Collapse
Affiliation(s)
- Ashish Pandey
- Department of Radiology, Tech4Health Institute, NYU Langone Health, New York, NY, USA
| | - Ines Karmous
- Biology and Environmental Department, Institute of Applied Biology of Medenine (ISBAM), University of Gabes, Gabes, Tunisia.
- Plant Toxicology and Molecular Biology of Microorganisms, Faculty of Sciences of Bizerta, University of Carthage, Carthage, Tunisia.
| |
Collapse
|
4
|
Castro A, Pittini Á, Berois N, Faccio R, Miranda P, Mombrú ÁW, Osinaga E, Pardo H. Development, Characterization, and Evaluation of Chi-Tn mAb-Functionalized DOTAP-PLGA Hybrid Nanoparticles Loaded with Docetaxel for Lung Cancer Therapy. Pharmaceutics 2025; 17:164. [PMID: 40006531 PMCID: PMC11859453 DOI: 10.3390/pharmaceutics17020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The focus of this study was to prepare and characterize docetaxel (DCX)-loaded lipid/polymer hybrid nanoparticles (LPHNps) functionalized with the monoclonal antibody (mAb) Chi-Tn for a potential active targeting approach in lung cancer treatment. Methods: We synthesized DOTAP-PLGA hybrid nanoparticles loaded with DCX and functionalized them with Chi-Tn mAb through a biotin-avidin approach. The physicochemical characterization involved dynamic light scattering, transmission electron microscopy, Raman spectroscopy, and atomic force microscopy. The in vitro and in vivo evaluations encompassed uptake studies, cell viability tests, and the assessment of tumor growth control in a lung cancer model. Results: The nanoparticles featured a hydrophobic PLGA core with 99.9% DCX encapsulation efficiency, surrounded by a DOTAP lipid shell ensuring colloidal stability with a high positive surface charge. The incorporation of PEGylated lipids on their surface helps evade the immune system and facilitate Chi-Tn mAb attachment. The resulting nanoparticles exhibit a spherical shape with monodisperse particle sizes averaging 250 nm, and demonstrate sustained drug release. In vitro uptake studies and viability assays conducted in A549 cancer cells show that the Chi-Tn mAb enhances nanoparticle internalization and significantly reduces cell viability. In vivo studies demonstrate a notable reduction in tumor volume and an increased survival rate in the A549 tumor xenograft mice model when DCX was encapsulated in nanoparticles and targeted with Chi-Tn mAb in comparison to the free drug. Conclusions: Therefore, Chi-Tn-functionalized LPHNps hold promise as carriers for actively targeting DCX to Tn-expressing carcinomas.
Collapse
Affiliation(s)
- Analía Castro
- Centro NanoMat, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Canelones 91000, Uruguay; (A.C.); (R.F.); (P.M.); (Á.W.M.)
| | - Álvaro Pittini
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; (Á.P.); (N.B.); (E.O.)
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Nora Berois
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; (Á.P.); (N.B.); (E.O.)
| | - Ricardo Faccio
- Centro NanoMat, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Canelones 91000, Uruguay; (A.C.); (R.F.); (P.M.); (Á.W.M.)
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República, Montevideo 11800, Uruguay
| | - Pablo Miranda
- Centro NanoMat, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Canelones 91000, Uruguay; (A.C.); (R.F.); (P.M.); (Á.W.M.)
| | - Álvaro W. Mombrú
- Centro NanoMat, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Canelones 91000, Uruguay; (A.C.); (R.F.); (P.M.); (Á.W.M.)
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República, Montevideo 11800, Uruguay
| | - Eduardo Osinaga
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; (Á.P.); (N.B.); (E.O.)
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Helena Pardo
- Centro NanoMat, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Canelones 91000, Uruguay; (A.C.); (R.F.); (P.M.); (Á.W.M.)
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República, Montevideo 11800, Uruguay
| |
Collapse
|
5
|
Ci Z, Wang H, Luo J, Wei C, Chen J, Wang D, Zhou Y. Application of Nanomaterials Targeting Immune Cells in the Treatment of Chronic Inflammation. Int J Nanomedicine 2024; 19:13925-13946. [PMID: 39735324 PMCID: PMC11682674 DOI: 10.2147/ijn.s497590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024] Open
Abstract
Chronic inflammation is a common characteristic of all kinds of diseases, including autoimmune diseases, metabolic diseases, and tumors. It is distinguished by the presence of low concentrations of inflammatory factors stimulating the body for an extended period, resulting in a persistent state of infection. This condition is manifested by the aggregation and infiltration of mononuclear cells, lymphocytes, and other immune cells, leading to tissue hyperplasia and lesions. Although various anti-inflammatory medications, including glucocorticoids and non-steroidal anti-inflammatory drugs (NSAIDs), have shown strong therapeutic effects, they lack specificity and targeting ability, and require high dosages, which can lead to severe adverse reactions. Nanoparticle drug delivery mechanisms possess the capacity to minimize the effect on healthy cells or tissues due to their targeting capabilities and sustained drug release properties. However, most nanosystems can only target the inflammatory sites rather than specific types of immune cells, leaving room for further improvement in the therapeutic effects of nanomaterials. This article reviews the current research progress on the role of diverse immune cells in inflammation, focusing on the functions of neutrophils and macrophages during inflammation. It provides an overview of the domestic and international applications of nanomaterials in targeted therapy for inflammation, aiming to establish a conceptual foundation for the utilization of nanomaterials targeting immune cells in the treatment of chronic inflammation and offer new perspectives for the avoidance and management of inflammation.
Collapse
Affiliation(s)
- Zhen Ci
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Hanchi Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiaxin Luo
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Chuqiao Wei
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jingxia Chen
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Dongyang Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Biology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
6
|
Ouyang B, Shan C, Shen S, Dai X, Chen Q, Su X, Cao Y, Qin X, He Y, Wang S, Xu R, Hu R, Shi L, Lu T, Yang W, Peng S, Zhang J, Wang J, Li D, Pang Z. AI-powered omics-based drug pair discovery for pyroptosis therapy targeting triple-negative breast cancer. Nat Commun 2024; 15:7560. [PMID: 39215014 PMCID: PMC11364624 DOI: 10.1038/s41467-024-51980-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Due to low success rates and long cycles of traditional drug development, the clinical tendency is to apply omics techniques to reveal patient-level disease characteristics and individualized responses to treatment. However, the heterogeneous form of data and uneven distribution of targets make drug discovery and precision medicine a non-trivial task. This study takes pyroptosis therapy for triple-negative breast cancer (TNBC) as a paradigm and uses data mining of a large TNBC cohort and drug databases to establish a biofactor-regulated neural network for rapidly screening and optimizing compound pyroptosis drug pairs. Subsequently, biomimetic nanococrystals are prepared using the preferred combination of mitoxantrone and gambogic acid for rational drug delivery. The unique mechanism of obtained nanococrystals regulating pyroptosis genes through ribosomal stress and triggering pyroptosis cascade immune effects are revealed in TNBC models. In this work, a target omics-based intelligent compound drug discovery framework explores an innovative drug development paradigm, which repurposes existing drugs and enables precise treatment of refractory diseases.
Collapse
Affiliation(s)
- Boshu Ouyang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
- Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, P. R. China
| | - Caihua Shan
- Microsoft Research Asia, Shanghai, 200232, P. R. China
| | - Shun Shen
- Pharmacy Department & Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, P. R. China
| | - Xinnan Dai
- Microsoft Research Asia, Shanghai, 200232, P. R. China
| | - Qingwang Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Xiaomin Su
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Yongbin Cao
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Xifeng Qin
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ying He
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Siyu Wang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ruizhe Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ruining Hu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Tun Lu
- School of Computer Science, Fudan University, Shanghai, 200438, P. R. China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| | - Shaojun Peng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University); Zhuhai, Guangdong, 519000, P. R. China.
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China.
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China.
| | - Dongsheng Li
- Microsoft Research Asia, Shanghai, 200232, P. R. China.
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China.
| |
Collapse
|
7
|
Wu D, Ji W, Xu S, Li Y, Ji Y, Fu K, Yang G. Near-infrared Light-Triggered Size-Shrinkable theranostic nanomicelles for effective tumor targeting and regression. Int J Pharm 2024; 658:124203. [PMID: 38705249 DOI: 10.1016/j.ijpharm.2024.124203] [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: 01/04/2024] [Revised: 04/03/2024] [Accepted: 05/03/2024] [Indexed: 05/07/2024]
Abstract
Most nanomedicines with suitable sizes (normally 100-200 nm) exhibit favorable accumulation in the periphery of tumors but hardly penetrate into deep tumors. Effective penetration of nanomedicines requires smaller sizes (less than 30 nm) to overcome the elevated tumor interstitial fluid pressure. Moreover, integrating an efficient diagnostic agent in the nanomedicines is in high demand for precision theranostics of tumors. To this end, a near-infrared light (NIR) -triggered size-shrinkable micelle system (Fe3O4@AuNFs/DOX-M) coloaded antitumor drug doxorubicin (DOX) and biomodal imaging agent magnetic gold nanoflower (Fe3O4@AuNFs) was developed to achieve efficient theranostic of tumors. Upon the accumulation of Fe3O4@AuNFs/DOX-M in the tumor periphery, a NIR laser was irradiated near the tumor sites, and the loaded Fe3O4@Au NFs could convert the light energy to heat, which triggered the cleavage of DOX-M to the ultra-small micelles (∼5 nm), thus realizing the deep penetration of micelles and on-demand drug release. Moreover, Fe3O4@AuNFs in the micelles could also be used as CT/MRI dual-modal contrast agent to "visualize" the tumor. Up to 92.6 % of tumor inhibition was achieved for the developed Fe3O4@AuNFs/DOX-M under NIR irradiation. This versatile micelle system provided a promising drug carrier platform realizing efficient tumor dual-modal diagnosis and photothermal-chemotherapy integration.
Collapse
Affiliation(s)
- Danjun Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Weili Ji
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shumin Xu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yazhen Li
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yaning Ji
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Kaili Fu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
8
|
Mosleh AM, El-Sherif AA, El-Sayed AA, Fahmy HM. Characterization and Cytotoxicity Assessment of Synthesized Palladium (II) Complex-Encapsulated Zinc Oxide Nanoparticles for Cancer Treatment. Cell Biochem Biophys 2024; 82:1225-1234. [PMID: 38744782 DOI: 10.1007/s12013-024-01273-1] [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] [Accepted: 04/08/2024] [Indexed: 05/16/2024]
Abstract
The treatment of cancer often leads to a range of adverse effects. Encapsulating drugs can mitigate these effects and enhance drug efficacy by enabling a controlled release at the site of interest. This study details the successful synthesis of zinc oxide nanoparticles (ZnONPs) through the precipitation of Zn(NO3)2·6H2O with KOH. A Pd(II) complex drug was synthesized from a Schiff base ligand derived from 2-hydroxybenzohydrazide and (E)-1-(2-(p-tolyl)hydrazono)propan-2-one using potassium tetrachloropalladate(II). This complex was subsequently incorporated into ZnONPs. Characterization of the resulting compounds was performed using Transmission Electron Microscopy (TEM), Dynamic Light Scattering (DLS), Zeta Potential, Fourier Transform Infrared (FTIR) Spectroscopy, and UV-visible spectroscopy. TEM imaging revealed particle sizes of 160.69 ± 4.74 nm for ZnONPs and 185.28 ± 2.3 nm for the Pd(II) complex-encapsulated ZnONPs. The Zeta potential values were 6.53 mV for ZnONPs and 7.36 mV for Pd(II) complex-encapsulated ZnONPs. UV-visible spectroscopy showed an absorption peak at 360 nm for ZnONPs, while the Pd(II) complex-encapsulated ZnONPs exhibited a peak at 410 nm. FTIR analysis indicated the presence of the Pd(II) complex within the ZnONPs, as evidenced by a consistent Zn-O vibrational band at 832 cm-1 and a shift in another peak from 460 to 413 cm-1. Additionally, the detection of a C = N stretching vibration at 1548 cm-1 and a carbonyl stretch at 1626 cm-1 was observed. The Encapsulation Efficiency (E.E.) of the Pd(II) complex was 97.2%. A drug release experiment conducted at pH 7 showed a steady-state release pattern after 16 h, with a cumulative release of 44.3%. The cytotoxic effects of the Pd(II) complex and its encapsulated form in ZnONPs on the MCF-7 cell line were assessed via MTT test. The Pd(II) complex encapsulated within ZnONPs exhibited decreased toxicity relative to the unencapsulated drug, as evidenced by a higher IC50 value of 418.5 μg/ml. This suggests that the encapsulation facilitates a sustained release, which allows for targeted accumulation within cells. The elevated IC50 value indicates that the drug delivery system may be engineered to modulate the release of the drug in a more controlled manner, potentially resulting in a prolonged release profile rather than an immediate therapeutic impact.
Collapse
Affiliation(s)
- Ayaat M Mosleh
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Ahmed A El-Sherif
- Chemistry Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Anwar A El-Sayed
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt.
| |
Collapse
|
9
|
Tunç T, Hepokur C, Kari̇per A. Synthesis and Characterization of Paclitaxel-Loaded Silver Nanoparticles: Evaluation of Cytotoxic Effects and Antimicrobial Activity. Bioinorg Chem Appl 2024; 2024:9916187. [PMID: 38380152 PMCID: PMC10878759 DOI: 10.1155/2024/9916187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/06/2024] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
Carrier system therapies based on combining cancer drugs with nanoparticles have been reported to control tumor growth and significantly reduce the side effects of cancer drugs. We thought that paclitaxel-loaded silver nanoparticles (AgNPs-PTX) were the right carrier to target cancer cells. We also carried out antimicrobial activity experiments as systems formed with nanoparticles have been shown to have antimicrobial activity. In our study, we used easy-to-synthesize and low-cost silver nanoparticles (AgNPs) with biocatalytic and photocatalytic advantages as drug carriers. We investigated the antiproliferative activities of silver nanoparticles synthesized by adding paclitaxel on MCF-7 (breast adenocarcinoma cell line), A549 (lung carcinoma cell line), C6 (brain glioma cell line) cells, and healthy WI-38 (fibroblast normal cell line) cell lines and their antimicrobial activities on 10 different microorganisms. The synthesized AgNPs and AgNPs-PTX were characterized by dynamic light scattering (DLS), scanning transmission electron microscopy, UV-visible spectroscopy, Fourier transform infrared spectroscopy, and X-ray spectroscopy. The nanoparticles were spherical in shape, with AgNPs ranging in size from 2.32 to 5.6 nm and AgNPs-PTXs from 24.36 to 58.77 nm. AgNPs demonstrated well stability of -47.3 mV, and AgNPs-PTX showed good stability of -25.4 mV. The antiproliferative effects of the synthesized nanoparticles were determined by XTT (tetrazolium dye; 2,3-bis-(2-methoxy-4-nitro-5-sulfenyl)-(2H)-tetrazolium-5-carboxanilide), and the proapoptotic effects were determined by annexin V/propidium iodide (PI) staining. The effect of AgNPs-PTX was more effective, and anticancer activity was higher than PTX in all cell lines. When selectivity indices were calculated, AgNPs-PTX was more selective in the A549 cell line (SI value 6.53 μg/mL). AgNPs-PTX was determined to increase apoptosis cells by inducing DNA fragmentation. To determine the antimicrobial activity, the MIC (minimum inhibitory concentration) test was performed using 8 different bacteria and 2 different fungi. Seven of the 10 microorganisms tested exhibited high antimicrobial activity according to the MIC ≤100 μg/mL standard, reaching MIC values below 100 μg/mL and 100 μg/mL for both AgNPs and AgNPs-PTX compared to reference sources. Compared to standard antibiotics, AgNPs-PTX was highly effective against 4 microorganisms.
Collapse
Affiliation(s)
- Tutku Tunç
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Ceylan Hepokur
- Department of Biochemistry, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Afşin Kari̇per
- Department Mathematics and Science Education, Faculty of Education, Erciyes University, Kayseri, Türkiye
| |
Collapse
|
10
|
Zhang N, Zhou J, Li S, Cai W, Ru B, Hu J, Liu W, Liu X, Tong X, Zheng X. Advances in Nanoplatforms for Immunotherapy Applications Targeting the Tumor Microenvironment. Mol Pharm 2024; 21:410-426. [PMID: 38170627 DOI: 10.1021/acs.molpharmaceut.3c00846] [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: 01/05/2024]
Abstract
Cancer immunotherapy is a treatment method that activates or enhances the autoimmune response of the body to fight tumor growth and metastasis, has fewer toxic side effects and a longer-lasting efficacy than radiotherapy and chemotherapy, and has become an important means for the clinical treatment of cancer. However, clinical results from immunotherapy have shown that most patients lack responsiveness to immunotherapy and cannot benefit from this treatment strategy. The tumor microenvironment (TME) plays a critical role in the response to immunotherapy. The TME typically prevents effective lymphocyte activation, reducing their infiltration, and inhibiting the infiltration of effector T cells. According to the characteristic differences between the TME and normal tissues, various nanoplatforms with TME targeting and regulation properties have been developed for more precise regulation of the TME and have the ability to codeliver a variety of active pharmaceutical ingredients, thereby reducing systemic toxicity and improving the therapeutic effect of antitumor. In addition, the precise structural design of the nanoplatform can integrate specific functional motifs, such as surface-targeted ligands, degradable backbones, and TME stimulus-responsive components, into nanomedicines, thereby reshaping the tumor microenvironment, improving the body's immunosuppressive state, and enhancing the permeability of drugs in tumor tissues, in order to achieve controlled and stimulus-triggered release of load cargo. In this review, the physiological characteristics of the TME and the latest research regarding the application of TME-regulated nanoplatforms in improving antitumor immunotherapy will be described. Furthermore, the existing problems and further applications perspectives of TME-regulated platforms for cancer immunotherapy will also be discussed.
Collapse
Affiliation(s)
- Nannan Zhang
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Junyu Zhou
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Shun Li
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Wenjun Cai
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Bin Ru
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Jiaqi Hu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Wenlong Liu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xuanxi Liu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiangmin Tong
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaoyan Zheng
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China
| |
Collapse
|
11
|
Li X, Peng X, Zoulikha M, Boafo GF, Magar KT, Ju Y, He W. Multifunctional nanoparticle-mediated combining therapy for human diseases. Signal Transduct Target Ther 2024; 9:1. [PMID: 38161204 PMCID: PMC10758001 DOI: 10.1038/s41392-023-01668-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 09/14/2023] [Accepted: 10/10/2023] [Indexed: 01/03/2024] Open
Abstract
Combining existing drug therapy is essential in developing new therapeutic agents in disease prevention and treatment. In preclinical investigations, combined effect of certain known drugs has been well established in treating extensive human diseases. Attributed to synergistic effects by targeting various disease pathways and advantages, such as reduced administration dose, decreased toxicity, and alleviated drug resistance, combinatorial treatment is now being pursued by delivering therapeutic agents to combat major clinical illnesses, such as cancer, atherosclerosis, pulmonary hypertension, myocarditis, rheumatoid arthritis, inflammatory bowel disease, metabolic disorders and neurodegenerative diseases. Combinatorial therapy involves combining or co-delivering two or more drugs for treating a specific disease. Nanoparticle (NP)-mediated drug delivery systems, i.e., liposomal NPs, polymeric NPs and nanocrystals, are of great interest in combinatorial therapy for a wide range of disorders due to targeted drug delivery, extended drug release, and higher drug stability to avoid rapid clearance at infected areas. This review summarizes various targets of diseases, preclinical or clinically approved drug combinations and the development of multifunctional NPs for combining therapy and emphasizes combinatorial therapeutic strategies based on drug delivery for treating severe clinical diseases. Ultimately, we discuss the challenging of developing NP-codelivery and translation and provide potential approaches to address the limitations. This review offers a comprehensive overview for recent cutting-edge and challenging in developing NP-mediated combination therapy for human diseases.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Xiuju Peng
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Makhloufi Zoulikha
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - George Frimpong Boafo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, PR China
| | - Kosheli Thapa Magar
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Yanmin Ju
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
| |
Collapse
|
12
|
Zang J, Zhang J, Mei Y, Xiong Y, Ci T, Feng N. Immunogenic dead cells engineered by the sequential treatment of ultraviolet irradiation/cryo-shocking for lung-targeting delivery and tumor vaccination. Biomater Sci 2023; 12:164-175. [PMID: 37947455 DOI: 10.1039/d3bm00854a] [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: 11/12/2023]
Abstract
Chemoimmunotherapy is a promising strategy in tumor treatments. In this study, immunogenic dead cells were engineered by the sequential treatment of live tumor cells with ultraviolet (UV) irradiation and cryo-shocking. The dead cells could serve as a lung-targeting vehicle and tumor vaccine after differential loading of the chemo-drug 10-hydroxycamptothecin (HCPT) and immune adjuvant Quillaja saponin-21 (QS-21) via physical absorption and chemical conjugation, respectively. After intravenous administration, the dead cells could be trapped in pulmonary capillaries and could fast release HCPT to enhance the drug accumulation in local tissues. Further, the immunogenic dead cells elicited antitumor immune responses together with the conjugated adjuvant QS-21 to achieve the elimination and long-term surveillance of tumor cells. In a lung tumor-bearing mice model, this drug-delivery system achieved synergistic antitumor efficacy and prolonged the survival of mice.
Collapse
Affiliation(s)
- Jing Zang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jinniu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yijun Mei
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu province, 210009, China
| | - Yaoxuan Xiong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Tianyuan Ci
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Nianping Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
13
|
Flont M, Jastrzębska E. A Multi-Layer Breast Cancer Model to Study the Synergistic Effect of Photochemotherapy. MICROMACHINES 2023; 14:1806. [PMID: 37763969 PMCID: PMC10535669 DOI: 10.3390/mi14091806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Breast cancer is one of the most common cancers among women. The development of new and effective therapeutic approaches in the treatment of breast cancer is an important challenge in modern oncology. Two-dimensional (2D) cell cultures are most often used in the study of compounds with potential anti-tumor nature. However, it is necessary to develop advanced three-dimensional (3D) cell models that can, to some extent, reflect the physiological conditions. The use of miniature cancer-on-a-chip microfluidic systems can help to mimic the complex cancer microenvironment. In this report, we developed a 3D breast cancer model in the form of a cell multilayer, composed of stromal cells (HMF) and breast cancer parenchyma (MCF-7). The developed cell model was successfully used to analyze the effectiveness of combined sequential photochemotherapy, based on doxorubicin and meso-tetraphenylporphyrin. We proved that the key factor that allows achieving the synergistic effect of combination therapy are the order of drug administration to the cells and the sequence of therapeutic procedures. To the best of our knowledge, studies on the effectiveness of combination photochemotherapy depending on the sequence of the component drugs were performed for the first time under microfluidic conditions on a 3D multilayered model of breast cancer tissue.
Collapse
Affiliation(s)
- Magdalena Flont
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Elżbieta Jastrzębska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| |
Collapse
|
14
|
Jiang S, Fu W, Wang S, Zhu G, Wang J, Ma Y. Bacterial Outer Membrane Vesicles Loaded with Perhexiline Suppress Tumor Development by Regulating Tumor-Associated Macrophages Repolarization in a Synergistic Way. Int J Mol Sci 2023; 24:11222. [PMID: 37446401 DOI: 10.3390/ijms241311222] [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: 06/09/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Tumor-associated macrophages (TAMs) promote tumor development and metastasis and are categorized into M1-like macrophages, suppressing tumor cells, and M2-like macrophages. M2-like macrophages, occupying a major role in TAMs, can be repolarized into anti-tumoral phenotypes. In this study, outer membrane vesicles (OMVs) secreted by Escherichia coli Nissle 1917 carry perhexiline (OMV@Perhx) to explore the influence of OMVs and perhexiline on TAM repolarization. OMV@Perhx was internalized by macrophages and regulated the phenotype of TAMs from M2-like to M1-like efficiently to increase the level of tumor suppressor accordingly. Re-polarized macrophages promoted apoptosis and inhibited the mobility of tumor, cells including invasion and migration. The results indicate that OMVs improve the efficacy of perhexiline and also represent a promising natural immunomodulator. Combining OMVs with perhexiline treatments shows powerfully synergistic anti-tumor effects through co-culturing with re-polarized macrophages. This work is promising to exploit the extensive applications of OMVs and chemical drugs, therefore developing a meaningful drug carrier and immunomodulator as well as expanding the purposes of traditional chemical drugs.
Collapse
Affiliation(s)
- Shoujin Jiang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Wei Fu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Sijia Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Guanshu Zhu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jufang Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yi Ma
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
15
|
Ouyang B, Deng L, Yang F, Shi H, Wang N, Tang W, Huang X, Zhou Y, Yu H, Wei Y, Dong J. Albumin-based formononetin nanomedicines for lung injury and fibrosis therapy via blocking macrophage pyroptosis. Mater Today Bio 2023; 20:100643. [PMID: 37214555 PMCID: PMC10193015 DOI: 10.1016/j.mtbio.2023.100643] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/23/2023] [Accepted: 04/23/2023] [Indexed: 05/24/2023] Open
Abstract
Pulmonary fibrosis that occurs following lung injury is a progressive and fatal disease since continual damage to lung tissue triggers the dysregulated inflammation response and accompanying abnormal healing process. Pyroptosis of alveolar macrophages has been found to play an essential role in the deterioration of lung injury and fibrosis. However, the lack of inhibitors against this inflammatory cell death in macrophages and the dense stroma pose major barriers to lung injury and fibrosis treatment. Herein, we developed an albumin-based nanoformulation to realize active delivery of formononetin (FMN) to improve the treatment of lung injury and fibrosis. The obtained nanoparticle, FMN@BSA NPs, could efficiently accumulate at the impaired lesion benefiting from the leaky vasculatures and the affinity between albumin and the overexpressed SPARC protein. Through blocking the NLRP3 inflammasome-involved pyroptosis process of macrophages, FMN@BSA NPs remarkably improved lung function and prolonged animal survival in the bleomycin (BLM)-induced lung injury and fibrosis model without noticeable side effects. Meanwhile, we proved FMN as a pyroptosis inhibitor and the corresponding lipid metabolism-related mechanisms through multi-omics analysis. This study first employed an albumin-based nanoparticle to deliver the pyroptosis inhibitor to the impaired lung tissue actively, providing a promising strategy for lung injury and fibrosis treatment.
Collapse
Affiliation(s)
- Boshu Ouyang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Lingling Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Fangyong Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Na Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| |
Collapse
|
16
|
Hu J, Zheng Y, Wen Z, Fu H, Yang X, Ye X, Zhu S, Kang L, Li X, Yang X, Hu Y. Construction of redox-sensitive liposomes modified by glycyrrhetinic acid and evaluation of anti-hepatocellular carcinoma activity. Chem Phys Lipids 2023; 252:105292. [PMID: 36931583 DOI: 10.1016/j.chemphyslip.2023.105292] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
The aim of this study was to construct a bifunctional liposome with hepatic-targeting capacity by modifying with a targeting ligand and an intracellular tumor reduction response functional group to deliver drugs precisely to focal liver tissues and release them in large quantities in hepatocellular carcinoma cells. This could improve drug efficacy and reduce toxic side effects at the same time. First, the bifunctional ligand for liposome was successfully obtained by chemically synthesizing it from the hepatic-targeting glycyrrhetinic acid (GA) molecule, cystamine, and the membrane component cholesterol. Then the ligand was used to modify the liposomes. The particle size, PDI and zeta potential of the liposomes were determined with a nanoparticle sizer, and the morphology was observed by transmission electron microscopy. The encapsulation efficiency and drug release behavior were also determined. Further, the stability in vitro of the liposomes and the changes in the simulated reducing environment were determined. Finally, the antitumor activity in vitro and cellular uptake efficiency of the drug-loaded liposomes were investigated by performing cellular assays. The results showed that the prepared liposomes had a uniform particle size of 143.6 ± 2.86 nm with good stability and an encapsulation rate of 84.3 ± 2.1 %. Moreover, the particle size of the liposomes significantly increased and the structure was destroyed in a DTT reducing environment. Cellular experiments showed that the modified liposoes had better cytotoxic effects on hepatocarcinoma cells than both normal liposomes and free drugs. This study has great potential for tumor therapy and provides novel ideas for the clinical use of oncology drugs in dosage forms.
Collapse
Affiliation(s)
- Jie Hu
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Yongsheng Zheng
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Zhijie Wen
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Hudie Fu
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Xuedan Yang
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Xuexin Ye
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Shengpeng Zhu
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Li Kang
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Xiaojun Li
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Xinzhou Yang
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Yan Hu
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, PR China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China.
| |
Collapse
|
17
|
Akkın S, Varan G, Işık A, Gökşen S, Karakoç E, Malanga M, Esendağlı G, Korkusuz P, Bilensoy E. Synergistic Antitumor Potency of a Self-Assembling Cyclodextrin Nanoplex for the Co-Delivery of 5-Fluorouracil and Interleukin-2 in the Treatment of Colorectal Cancer. Pharmaceutics 2023; 15:pharmaceutics15020314. [PMID: 36839637 PMCID: PMC9963231 DOI: 10.3390/pharmaceutics15020314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Chemotherapy is the most used method after surgery in the treatment of colon cancer. Cancer cells escape the recognition mechanism of immune system cells to survive and develop chemoresistance. Therefore, the use of immunotherapy in combination with chemotherapy can increase the effectiveness of the treatment. Nanoparticles have been used clinically to increase the accumulation of therapeutics in target tissues and reduce toxicity. In this paper, nanoplexes were formed via cationic cyclodextrin polymer, 5-Fluorouracil, and Interleukin-2 based on the opposite charge interaction of macromolecules without undergoing any structural changes or losing the biological activity of Interleukin-2. Anticancer activities of nanoplexes were determined in two-dimensional and three-dimensional cell culture setups. The dual drug-loaded cyclodextrin nanoplexes diffused deeper into the spheroids and accelerated apoptosis when compared with 5-FU solutions. In the colorectal tumor-bearing animal model, survival rate, antitumor activity, metastasis, and immune response parameters were assessed using a cyclodextrin derivative, which was found to be safe based on the ALT/AST levels in healthy mice. Histomorphometric analysis showed that the groups treated with the nanoplex formulation had significantly fewer initial tumors and lung foci when compared with the control. The dual drug-loaded nanoplex could be a promising drug delivery technique in the immunochemotherapy of colorectal cancer.
Collapse
Affiliation(s)
- Safiye Akkın
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey
| | - Gamze Varan
- Department of Vaccine Technology, Vaccine Institute, Hacettepe University, 06100 Ankara, Turkey
| | - Anıl Işık
- Department of Basic Oncology, Cancer Institute, Hacettepe University, 06100 Ankara, Turkey
| | - Sibel Gökşen
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, 06100 Ankara, Turkey
| | - Elif Karakoç
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, 06100 Ankara, Turkey
| | - Milo Malanga
- CycloLab-Cyclodextrin Research & Development Laboratory, Organic Synthesis Laboratory, 1097 Budapest, Hungary
| | - Güneş Esendağlı
- Department of Basic Oncology, Cancer Institute, Hacettepe University, 06100 Ankara, Turkey
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, 06100 Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, 06100 Ankara, Turkey
| | - Erem Bilensoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey
- Correspondence:
| |
Collapse
|
18
|
Gu Z, Da Silva CG, Hao Y, Schomann T, Camps MGM, van der Maaden K, Liu Q, Ossendorp F, Cruz LJ. Effective combination of liposome-targeted chemotherapy and PD-L1 blockade of murine colon cancer. J Control Release 2023; 353:490-506. [PMID: 36460179 DOI: 10.1016/j.jconrel.2022.11.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
Therapeutic cancer drug efficacy can be limited by insufficient tumor penetration, rapid clearance, systemic toxicity and (acquired) drug resistance. The poor therapeutic index due to inefficient drug penetration and rapid drug clearance and toxicity can be improved by using a liposomal platform. Drug resistance for instance against pemetrexed, can be reduced by combination with docetaxel. Here, we developed a specific liposomal formulation to simultaneously deliver docetaxel and pemetrexed to enhance efficacy and safety. Hydrophobic docetaxel and hydrophilic pemetrexed were co-encapsulated into pH-sensitive liposomes using a thin-film hydration method with high efficiency. The physicochemical properties, toxicity, and immunological effects of liposomes were examined in vitro. Biodistribution, anti-tumor efficacy, and systemic immune response were evaluated in vivo in combination with PD-L1 immune checkpoint therapy using two murine colon cancer models. In cellular experiments, the liposomes exhibited strong cytotoxicity and induced immunogenic cell death. In vivo, the treatment with the liposome-based drug combination inhibited tumor development and stimulated immune responses. Liposomal encapsulation significantly reduced systemic toxicity compared to the delivery of the free drug. Tumor control was strongly enhanced when combined with anti-PDL1 immunotherapy in immunocompetent mice carrying syngeneic MC38 or CT26 colon tumors. We showed that treatment with liposome-mediated chemotherapy of docetaxel and pemetrexed combined with anti-PD-L1 immunotherapy is a promising strategy for the treatment of colon cancers.
Collapse
Affiliation(s)
- Zili Gu
- Department of Radiology, Leiden University Medical Center, the Netherlands
| | - Candido G Da Silva
- Department of Radiology, Leiden University Medical Center, the Netherlands
| | - Yang Hao
- Department of Radiology, Leiden University Medical Center, the Netherlands
| | - Timo Schomann
- Department of Radiology, Leiden University Medical Center, the Netherlands; Percuros B.V., Leiden, the Netherlands
| | - Marcel G M Camps
- Department of Immunology, Leiden University Medical Center, the Netherlands
| | - K van der Maaden
- Department of Immunology, Leiden University Medical Center, the Netherlands
| | - Qi Liu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ferry Ossendorp
- Department of Immunology, Leiden University Medical Center, the Netherlands.
| | - Luis J Cruz
- Department of Radiology, Leiden University Medical Center, the Netherlands.
| |
Collapse
|
19
|
Ghafelehbashi R, Farshbafnadi M, Aghdam NS, Amiri S, Salehi M, Razi S. Nanoimmunoengineering strategies in cancer diagnosis and therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:78-90. [PMID: 36076122 DOI: 10.1007/s12094-022-02935-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
Cancer immunotherapy strategies in combination with engineered nanosystems have yielded beneficial results in the treatment of cancer and their application is increasing day by day. The pivotal role of stimuli-responsive nanosystems and nanomedicine-based cancer immunotherapy, as a subsidiary discipline in the field of immunology, cannot be ignored. Today, rapid advances in nanomedicine are used as a platform for exploring new therapeutic applications and modern smart healthcare management strategies. The progress of nanomedicine in cancer treatment has confirmed the findings of immunotherapy in the medical research phase. This study concentrates on approaches connected to the efficacy of nanoimmunoengineering strategies for cancer immunotherapies and their applications. By assessing improved approaches, different aspects of the nanoimmunoengineering strategies for cancer therapies are discussed in this study.
Collapse
Affiliation(s)
- Robabehbeygom Ghafelehbashi
- Department of Materials and Textile Engineering, College of Engineering, Razi University, Kermanshah, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Melina Farshbafnadi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shahin Amiri
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Mitra Salehi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Iqubal MK, Kaur H, Md S, Alhakamy NA, Iqubal A, Ali J, Baboota S. A technical note on emerging combination approach involved in the onconanotherapeutics. Drug Deliv 2022; 29:3197-3212. [PMID: 36226570 PMCID: PMC9578464 DOI: 10.1080/10717544.2022.2132018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cancer is the second cause of mortality worldwide, and the currently available conventional treatment approach is associated with serious side effects and poor clinical outcomes. Based on the outcome of the exploratory preclinical and clinical studies, it was found that therapeutic response increases multiple folds when anticancer drugs are used in combination. However, the conventional combination of anticancer drugs was associated with various limitations such as increased cost of treatment, systemic toxicity, drug resistance, and reduced pharmacokinetic attributes. Hence, attempts were made to formulate nanocarrier fabricated combinatorial drugs (NFCDs) to effectively manage and treat cancer. This approach offers several advantages, such as improved stability, lower drug exposure, targeted drug delivery, low side effects, and improved clinical outcome. Hence, in this review, first time, we have discussed the recent advancement and various types of nano carrier-based combinatorial drug delivery systems in a different type of cancer and highlighted the personalized combinatorial theranostic medicine as a futuristic anticancer treatment approach.
Collapse
Affiliation(s)
- Mohammad Kashif Iqubal
- Product Development Department, Sentiss Research Centre, Sentiss Pharma Pvt Ltd, Gurugram, India.,Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Harsimran Kaur
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
21
|
Ravi R, Zeyaullah M, Ghosh S, Khan Warsi M, Baweja R, AlShahrani AM, Mishra A, Ahmad R. Use of gold nanoparticle-silibinin conjugates: A novel approach against lung cancer cells. Front Chem 2022; 10:1018759. [PMID: 36311430 PMCID: PMC9606463 DOI: 10.3389/fchem.2022.1018759] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/23/2022] [Indexed: 08/11/2023] Open
Abstract
Lung cancer presents one of the most challenging carcinomas with meager 5-year survival rates (less than 20%), high metastasis and high recurrence due to chemo- and radio- resistance. An alternative or complementation to existing prognosis modalities is the use of phytochemicals such as silibinin, which targets essential cytokines, angiogenic factors and transcription factors for a profound anti-tumor effect. However, the problems of low solubility in an aqueous physiological environment, poor penetration, high metabolism and rapid systemic clearance limit the therapeutic use of silibinin. Conjugation of gold nanoparticles (GNPs) with silibinin may overcome the above challenges along with distinct advantages of biocompatibility, optical properties for monitoring and causation of cytotoxicity in cancer cells. The current study thus aims to develop silibinin conjugated gold nanoparticles (Sb-GNPs) with pH responsive release in the cancer microenvironment, optimizing several parameters for its higher activity and further evaluate the nanoplatform for their efficacy in inducing cell death in vitro against A549 lung cancer cells. GNPs was synthesized using trisodium citrate dihydrate as the reducing agent and further used for the conjugation of silibinin. The synthesized GNPs were found to be monodispersed and spherical in shape. The silibinin was successfully conjugated with gold nanoparticles and long-term stability of GNPs and Sb-GNPs nanoconjugates in suspension phase was confirmed by FTIR and DLS. Anticancer properties of Sb-GNPs were confirmed by different assay using MTT, Trypan blue dye exclusion assay and cell cycle analysis assay. After conjugation of silibinin with GNPs, the efficacy of silibinin increased 4-5 times in killing the cancer cells. This is the first report on using silibinin gold nanoconjugate system for lung cancer therapy with promising future applications.
Collapse
Affiliation(s)
- Rangnath Ravi
- Department of Chemistry, Shivaji College, University of Delhi, New Delhi, India
| | - Md. Zeyaullah
- Department of Basic Medical Science, College of Applied Medical Sciences, King Khalid University (KKU), Khamis Mushayt Campus, Abha, Saudi Arabia
| | - Shubhrima Ghosh
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Mohiuddin Khan Warsi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Renu Baweja
- Department of Biochemistry, Shivaji College, University of Delhi, New Delhi, India
| | - Abdullah M. AlShahrani
- Department of Basic Medical Science, College of Applied Medical Sciences, King Khalid University (KKU), Khamis Mushayt Campus, Abha, Saudi Arabia
| | - Abhijeet Mishra
- Department of Biochemistry, Shivaji College, University of Delhi, New Delhi, India
| | - Razi Ahmad
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi, India
- Quality and Research Department, Anantaa GSK Innovations Pvt Ltd., DLF Industrial Area, Faridabad, India
| |
Collapse
|
22
|
Recent progress in two-dimensional nanomaterials for cancer theranostics. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Effects of Cancer Cell-Derived Nanovesicle Vaccines Produced by the Oxidative Stress-Induced Expression of DAMP and Spontaneous Release/Filter Extrusion in the Interplay of Cancer Cells and Macrophages. Biomedicines 2022; 10:biomedicines10081977. [PMID: 36009524 PMCID: PMC9405549 DOI: 10.3390/biomedicines10081977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022] Open
Abstract
Photodynamic therapy (PDT)-based cancer vaccines are shown to be more effective modalities for treating cancer in animal models compared to other methods used to generate cancer cell-derived vaccines. The higher efficacy seems to stem from the generation of cell membrane nanovesicles or fragments that carry both cancer cell-specific antigens and high surface content of damage-associated molecular pattern (DAMP) molecules induced by oxidative stress. To develop more effective cancer vaccines in this direction, we explored the generation of cancer vaccines by applying different sources of oxidative stress on cancer cell cultures followed by spontaneous release or filter extrusions to produce cancer cell-derived DAMP-expressing nanovesicles. Through an in-vitro test based on the co-culture of cancer cells and macrophages, it was found that the nanovesicle vaccines generated by H2O2 are as effective as those generated by PDT in diminishing cancer cell culture masses, providing a simpler way to manufacture vaccines. In addition, the nanovesicle vaccines produced by filter extrusion are as potent as those produced by spontaneous release, rendering a more stable way for vaccine production.
Collapse
|
24
|
Zhao Z, Fang L, Xiao P, Sun X, Zhou L, Liu X, Wang J, Wang G, Cao H, Zhang P, Jiang Y, Wang D, Li Y. Walking Dead Tumor Cells for Targeted Drug Delivery Against Lung Metastasis of Triple-Negative Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2205462. [PMID: 35759925 DOI: 10.1002/adma.202205462] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Indexed: 06/15/2023]
Abstract
Lung metastasis is challenging in patients with triple-negative breast cancer (TNBC). Surgery is always not available due to the dissemination of metastatic foci and most drugs are powerless because of poor retention at metastatic sites. TNBC cells generate an inflamed microenvironment and overexpress adhesive molecules to promote invasion and colonization. Herein, "walking dead" TNBC cells are developed through conjugating anti-PD-1 (programmed death protein 1 inhibitor) and doxorubicin (DOX)-loaded liposomes onto cell corpses for temporal chemo-immunotherapy against lung metastasis. The walking dead TNBC cells maintain plenary tumor antigens to conduct vaccination effects. Anti-PD-1 antibodies are conjugated to cell corpses via reduction-activated linker, and DOX-loaded liposomes are attached by maleimide-thiol coupling. This anchor strategy enables rapid release of anti-PD-1 upon reduction conditions while long-lasting release of DOX at inflamed metastatic sites. The walking dead TNBC cells improve pulmonary accumulation and local retention of drugs, reprogram the lung microenvironment through damage-associated molecular patterns (DAMPs) and PD-1 blockade, and prolong overall survival of lung metastatic 4T1 and EMT6-bearing mice. Taking advantage of the walking dead TNBC cells for pulmonary preferred delivery of chemotherapeutics and checkpoint inhibitors, this study suggests an alternative treatment option of chemo-immunotherapy to augment the efficacy against lung metastasis.
Collapse
Affiliation(s)
- Zitong Zhao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Lei Fang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ping Xiao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiangshi Sun
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lei Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaochen Liu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jue Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guanru Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haiqiang Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China
| | - Yanyan Jiang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Dangge Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shangdong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Shandong, 264000, China
| |
Collapse
|
25
|
Shinde VR, Revi N, Murugappan S, Singh SP, Rengan AK. Enhanced Permeability and Retention Effect: A key facilitator for solid tumor targeting by nanoparticles. Photodiagnosis Photodyn Ther 2022; 39:102915. [PMID: 35597441 DOI: 10.1016/j.pdpdt.2022.102915] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 12/14/2022]
Abstract
Exploring the enhanced permeability and retention (EPR) effect through therapeutic nanoparticles has been a subject of considerable interest in tumor biology. This passive targeting based phenomenon exploits the leaky blood vasculature and the defective lymphatic drainage system of the heterogeneous tumor microenvironment resulting in enhanced preferential accumulation of the nanoparticles within the tumor tissues. This article reviews the fundamental studies to assess how the EPR effect plays an essential role in passive targeting. Further, it summarizes various therapeutic modalities of nanoformulation including chemo-photodynamic therapy, intravascular drug release, and photothermal immunotherapy to combat cancer using enhanced EPR effect in neoplasia region.
Collapse
Affiliation(s)
- Vinod Ravasaheb Shinde
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Neeraja Revi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | | | - Surya Prakash Singh
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India.
| |
Collapse
|
26
|
Boone CE, Wang L, Gautam A, Newton IG, Steinmetz NF. Combining nanomedicine and immune checkpoint therapy for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1739. [PMID: 34296535 PMCID: PMC8906799 DOI: 10.1002/wnan.1739] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/07/2021] [Accepted: 06/24/2021] [Indexed: 01/03/2023]
Abstract
Cancer immunotherapy has emerged as a pillar of the cancer therapy armamentarium. Immune checkpoint therapy (ICT) is a mainstay of modern immunotherapy. Although ICT monotherapy has demonstrated remarkable clinical efficacy in some patients, the majority do not respond to treatment. In addition, many patients eventually develop resistance to ICT, disease recurrence, and toxicity from off-target effects. Combination therapy is a keystone strategy to overcome the limitations of monotherapy. With the integration of ICT and any therapy that induces tumor cell lysis and release of tumor-associated antigens (TAAs), ICT is expected to strengthen the coordinated innate and adaptive immune responses to TAA release and promote systemic, cellular antitumor immunity. Nanomedicine is well poised to facilitate combination ICT. Nanoparticles with delivery and/or immunomodulation capacities have been successfully combined with ICT in preclinical applications. Delivery nanoparticles protect and control the targeted release of their cargo. Inherently immunomodulatory nanoparticles can facilitate immunogenic cell death, modification of the tumor microenvironment, immune cell mimicry and modulation, and/or in situ vaccination. Nanoparticles are frequently multifunctional, combining multiple treatment strategies into a single platform with ICT. Nanomedicine and ICT combinations have great potential to yield novel, powerful treatments for patients with cancer. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Lu Wang
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92039, USA
| | - Aayushma Gautam
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92039, USA
| | - Isabel G. Newton
- Department of Radiology, University of California, San Diego, La Jolla CA 92039, USA,Veterans Administration San Diego Healthcare System, 3350 La Jolla Village Drive San Diego, CA 92161
| | | |
Collapse
|
27
|
Garofalo C, De Marco C, Cristiani CM. NK Cells in the Tumor Microenvironment as New Potential Players Mediating Chemotherapy Effects in Metastatic Melanoma. Front Oncol 2021; 11:754541. [PMID: 34712615 PMCID: PMC8547654 DOI: 10.3389/fonc.2021.754541] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Until the last decade, chemotherapy was the standard treatment for metastatic cutaneous melanoma, even with poor results. The introduction of immune checkpoints inhibitors (ICIs) radically changed the outcome, increasing 5-year survival from 5% to 60%. However, there is still a large portion of unresponsive patients that would need further therapies. NK cells are skin-resident innate cytotoxic lymphocytes that recognize and kill virus-infected as well as cancer cells thanks to a balance between inhibitory and activating signals delivered by surface molecules expressed by the target. Since NK cells are equipped with cytotoxic machinery but lack of antigen restriction and needing to be primed, they are nowadays gaining attention as an alternative to T cells to be exploited in immunotherapy. However, their usage suffers of the same limitations reported for T cells, that is the loss of immunogenicity by target cells and the difficulty to penetrate and be activated in the suppressive tumor microenvironment (TME). Several evidence showed that chemotherapy used in metastatic melanoma therapy possess immunomodulatory properties that may restore NK cells functions within TME. Here, we will discuss the capability of such chemotherapeutics to: i) up-regulate melanoma cells susceptibility to NK cell-mediated killing, ii) promote NK cells infiltration within TME, iii) target other immune cell subsets that affect NK cells activities. Alongside traditional systemic melanoma chemotherapy, a new pharmacological strategy based on nanocarriers loaded with chemotherapeutics is developing. The use of nanotechnologies represents a very promising approach to improve drug tolerability and effectiveness thanks to the targeted delivery of the therapeutic molecules. Here, we will also discuss the recent developments in using nanocarriers to deliver anti-cancer drugs within the melanoma microenvironment in order to improve chemotherapeutics effects. Overall, we highlight the possibility to use standard chemotherapeutics, possibly delivered by nanosystems, to enhance NK cells anti-tumor cytotoxicity. Combined with immunotherapies targeting NK cells, this may represent a valuable alternative approach to treat those patients that do not respond to current ICIs.
Collapse
Affiliation(s)
- Cinzia Garofalo
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Carmela De Marco
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Costanza Maria Cristiani
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
28
|
Zhang L, Li Q, Chen J, Tang C, Yin C. Enhanced antitumor efficacy of glutathione-responsive chitosan based nanoparticles through co-delivery of chemotherapeutics, genes, and immune agents. Carbohydr Polym 2021; 270:118384. [PMID: 34364626 DOI: 10.1016/j.carbpol.2021.118384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/15/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022]
Abstract
To achieve the co-delivery of chemotherapeutic drugs, genes, and immune agents in a single nanoparticulate system, p-mercaptobenzoic acid-grafted N, N, N-trimethyl chitosan nanoparticles (MT NPs) were successfully synthesized. Paclitaxel (PTX) was encapsulated into the hydrophobic core of the MT NPs, and meanwhile, survivin shRNA-expressing plasmid (iSur-pDNA) and recombinant human interleukin-2 (rhIL-2) were loaded onto the hydrophilic shell of the MT NPs. Owing to the redox-sensitiveness of MT NPs, a rapid release of PTX was triggered by the high concentration of glutathione. The synergistic effects of PTX (1.5 mg/kg), iSur-pDNA (1.875 mg/kg), and rhIL-2 (6 × 105 IU/kg) at a low dose endowed the MT/PTX/pDNA/rhIL-2 NPs with enhanced antitumor efficacies and improved tumor-induced immunosuppression. These results demonstrated that the co-delivery of PTX, iSur-pDNA, and rhIL-2 by the amphiphilic chitosan based NPs with redox-sensitiveness could be a promising strategy in the treatment of tumors.
Collapse
Affiliation(s)
- Linlin Zhang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Qiuping Li
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jinxuan Chen
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
29
|
Liu Z, Liu C, Yao W, Gao S, Wang J, Zhang P, Ge H. Efficacy and safety of toripalimab combined with doxorubicin as first-line treatment for metastatic soft tissue sarcomas: an observational study. Anticancer Drugs 2021; 32:962-968. [PMID: 34001702 PMCID: PMC8448405 DOI: 10.1097/cad.0000000000001088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 04/20/2021] [Indexed: 11/25/2022]
Abstract
Programmed cell death protein 1 (PD-1) inhibitors have demonstrated promising activity among patients with advanced soft tissue sarcomas (STS) in phase II trials. The purpose of this study was to assess the efficacy and safety of toripalimab (a novel PD-1 inhibitor) combined with doxorubicin as first-line treatment in patients with metastatic STS between December 2018 and September 2019. A total of 30 patients with metastatic STS were included and followed up retrospectively. One patient had complete response (CR), 10 patients obtained partial response, and 13 patients achieved stable disease. The objective response rate was 36.7% and the disease control rate was 80%. The median progression-free survival (PFS) was 8 months (95% CI: 6.30-10.64). The most frequent any grade adverse events were nausea (66.7%), fatigue (60%), and vomiting (40%). Neutropenia (20%) was the most common grade 3/4 adverse events, followed by leucopenia (13.3%) and febrile neutropenia (6.7%). No death related to treatment was observed during the drugs administration. Toripalimab combined with doxorubicin is effective in patients with metastatic STS as first-line treatment with manageable adverse events.
Collapse
Affiliation(s)
- Zhiyong Liu
- Department of Orthopedics, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital
| | | | - Weitao Yao
- Department of Orthopedics, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital
| | - Songtao Gao
- Department of Orthopedics, Henan Provincial People’s Hospital and People’s Hospital of Zhengzhou University
| | - Jiaqiang Wang
- Department of Orthopedics, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital
| | - Peng Zhang
- Department of Orthopedics, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, China
| |
Collapse
|
30
|
Combining Photodynamic Therapy with Immunostimulatory Nanoparticles Elicits Effective Anti-Tumor Immune Responses in Preclinical Murine Models. Pharmaceutics 2021; 13:pharmaceutics13091470. [PMID: 34575546 PMCID: PMC8465537 DOI: 10.3390/pharmaceutics13091470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/03/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) has shown encouraging but limited clinical efficacy when used as a standalone treatment against solid tumors. Conversely, a limitation for immunotherapeutic efficacy is related to the immunosuppressive state observed in large, advanced tumors. In the present study, we employ a strategy, in which we use a combination of PDT and immunostimulatory nanoparticles (NPs), consisting of poly(lactic-co-glycolic) acid (PLGA)-polyethylene glycol (PEG) particles, loaded with the Toll-like receptor 3 (TLR3) agonist poly(I:C), the TLR7/8 agonist R848, the lymphocyte-attracting chemokine, and macrophage inflammatory protein 3α (MIP3α). The combination provoked strong anti-tumor responses, including an abscopal effects, in three clinically relevant murine models of cancer: MC38 (colorectal), CT26 (colorectal), and TC-1 (human papillomavirus 16-induced). We show that the local and distal anti-tumor effects depended on the presence of CD8+ T cells. The combination elicited tumor-specific oncoviral- or neoepitope-directed CD8+ T cells immune responses against the respective tumors, providing evidence that PDT can be used as an in situ vaccination strategy against cancer (neo)epitopes. Finally, we show that the treatment alters the tumor microenvironment in tumor-bearing mice, from cold (immunosuppressed) to hot (pro-inflammatory), based on greater neutrophil infiltration and higher levels of inflammatory myeloid and CD8+ T cells, compared to untreated mice. Together, our results provide a rationale for combining PDT with immunostimulatory NPs for the treatment of solid tumors.
Collapse
|
31
|
Huis In 't Veld RV, Da Silva CG, Jager MJ, Cruz LJ, Ossendorp F. Combining Photodynamic Therapy with Immunostimulatory Nanoparticles Elicits Effective Anti-Tumor Immune Responses in Preclinical Murine Models. Pharmaceutics 2021. [PMID: 34575546 DOI: 10.3390/pharmaceutics1309147010.3390/pharmaceutics13091470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023] Open
Abstract
Photodynamic therapy (PDT) has shown encouraging but limited clinical efficacy when used as a standalone treatment against solid tumors. Conversely, a limitation for immunotherapeutic efficacy is related to the immunosuppressive state observed in large, advanced tumors. In the present study, we employ a strategy, in which we use a combination of PDT and immunostimulatory nanoparticles (NPs), consisting of poly(lactic-co-glycolic) acid (PLGA)-polyethylene glycol (PEG) particles, loaded with the Toll-like receptor 3 (TLR3) agonist poly(I:C), the TLR7/8 agonist R848, the lymphocyte-attracting chemokine, and macrophage inflammatory protein 3α (MIP3α). The combination provoked strong anti-tumor responses, including an abscopal effects, in three clinically relevant murine models of cancer: MC38 (colorectal), CT26 (colorectal), and TC-1 (human papillomavirus 16-induced). We show that the local and distal anti-tumor effects depended on the presence of CD8+ T cells. The combination elicited tumor-specific oncoviral- or neoepitope-directed CD8+ T cells immune responses against the respective tumors, providing evidence that PDT can be used as an in situ vaccination strategy against cancer (neo)epitopes. Finally, we show that the treatment alters the tumor microenvironment in tumor-bearing mice, from cold (immunosuppressed) to hot (pro-inflammatory), based on greater neutrophil infiltration and higher levels of inflammatory myeloid and CD8+ T cells, compared to untreated mice. Together, our results provide a rationale for combining PDT with immunostimulatory NPs for the treatment of solid tumors.
Collapse
Affiliation(s)
- Ruben Victor Huis In 't Veld
- Department of Radiology, Leiden University Medical Centre (LUMC), Room C2-187h, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Candido G Da Silva
- Department of Radiology, Leiden University Medical Centre (LUMC), Room C2-187h, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Luis J Cruz
- Department of Radiology, Leiden University Medical Centre (LUMC), Room C2-187h, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Ferry Ossendorp
- Department of Immunology, Leiden University Medical Centre (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
32
|
Zou X, Jiang Z, Li L, Huang Z. Selenium nanoparticles coated with pH responsive silk fibroin complex for fingolimod release and enhanced targeting in thyroid cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 49:83-95. [PMID: 33438446 DOI: 10.1080/21691401.2021.1871620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cancer-targeted drug delivery systems based on nanoparticles (NPs) have been considered promising therapies. In this study, we developed a pH-responsive smart NPs drug delivery system using silk fibroin (SF), selenium nanoparticles (Se NPs), fingolimod (FTY720), and heptapeptide (T7). The prepared FTY720@T7-SF-Se NPs were spheres with an average diameter of 120 nm, which would contribute to the enhanced permeability and retention effects in tumour regions. The encapsulation efficiency (EE) of the FTY720@T7-SF-Se NPs was 71.95 ± 3.81%. The release of FTY720 from the nanocarriers was pH-dependent, and the release of FTY720 was accelerated in an acidic environment. Both in vitro and in vivo studies showed that FTY720@T7-SF-Se NPs had an enhanced cellular uptake selectivity and antitumor activity for thyroid cancer. The bio-distribution study in vivo further demonstrated that FTY720@T7-SF-Se NPs could effectively accumulate in the tumour region, thereby enhancing the ability to kill cancer cells in vivo. In addition, studies of histology and immunohistochemistry showed that FTY720@T7-SF-Se NPs had low toxicity to the major organs of tumour-bearing mice, indicating the prepared NPs has good biocompatibility in vivo. These results suggest that the tumour-targeted NPs delivery system (FTY720@T7-SF-Se NPs) has great potential as a new tool for thyroid cancer therapy.
Collapse
Affiliation(s)
- Xiangcai Zou
- Department of General Surgery, Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of General Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhipeng Jiang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Li
- Digestive Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Recent advances in immunotherapy, immunoadjuvant, and nanomaterial-based combination immunotherapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Correction to: DNA Nanotechnology for Multimodal Synergistic Theranostics. JOURNAL OF ANALYSIS AND TESTING 2021. [DOI: 10.1007/s41664-021-00190-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
|
36
|
Bagherifar R, Kiaie SH, Hatami Z, Ahmadi A, Sadeghnejad A, Baradaran B, Jafari R, Javadzadeh Y. Nanoparticle-mediated synergistic chemoimmunotherapy for tailoring cancer therapy: recent advances and perspectives. J Nanobiotechnology 2021; 19:110. [PMID: 33865432 PMCID: PMC8052859 DOI: 10.1186/s12951-021-00861-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
Nowadays, a potent challenge in cancer treatment is considered the lack of efficacious strategy, which has not been able to significantly reduce mortality. Chemoimmunotherapy (CIT) as a promising approach in both for the first-line and relapsed therapy demonstrated particular benefit from two key gating strategies, including chemotherapy and immunotherapy to cancer therapy; therefore, the discernment of their participation and role of potential synergies in CIT approach is determinant. In this study, in addition to balancing the pros and cons of CIT with the challenges of each of two main strategies, the recent advances in the cancer CIT have been discussed. Additionally, immunotherapeutic strategies and the immunomodulation effect induced by chemotherapy, which boosts CIT have been brought up. Finally, harnessing and development of the nanoparticles, which mediated CIT have expatiated in detail.
Collapse
Affiliation(s)
- Rafieh Bagherifar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Kiaie
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Hatami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Armin Ahmadi
- Department of Chemical & Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St, Ershad Blvd., P.O. BoX: 1138, 57147, Urmia, Iran.
- Department of Immunology and Genetics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Yousef Javadzadeh
- Biotechnology Research Center, and Faculty of Pharmacy, Tabriz University of Medical Science, 5166-15731, Tabriz, Iran.
| |
Collapse
|
37
|
Neha Desai, Momin M, Khan T, Gharat S, Ningthoujam RS, Omri A. Metallic nanoparticles as drug delivery system for the treatment of cancer. Expert Opin Drug Deliv 2021; 18:1261-1290. [PMID: 33793359 DOI: 10.1080/17425247.2021.1912008] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The targeted delivery of anticancer agents to tumor is a major challenge because most of the drugs show off-target effect resulting in nonspecific cell death. Multifunctionalized metallic nanoparticles (NPs) are explored as new carrier system in the era of cancer therapeutics. Researchers investigated the potential of metallic NPs to target tumor cells by active and passive mechanisms, thereby reducing off-target effects of anticancer agents. Moreover, photocatalytic activity of upconversion nanoparticles (UCNPs) and the enhanced permeation and retention (EPR) effect have also gained wide potential in cancer treatment. Recent advancement in the field of nanotechnology highlights their potency for cancer therapy. AREAS COVERED This review summarizes the types of gold and silver metallic NPs with targeting mechanisms and their potentiality in cancer therapy. EXPERT OPINION Recent advances in the field of nanotechnology for cancer therapy offer high specificity and targeting efficiency. Targeting tumor cells through mechanistic pathways using metallic NPs for the disruption/alteration of molecular profile and survival rate of the tumor cells has led to an effective approach for cancer therapeutics. This alteration in the survival rate of the tumor cells might decrease the proliferation thereby resulting in more efficient management in the treatment of cancer.
Collapse
Affiliation(s)
- Neha Desai
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Sankalp Gharat
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | | | - Abdelwahab Omri
- The Novel Drug and Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada
| |
Collapse
|
38
|
Castro A, Berois N, Malanga A, Ortega C, Oppezzo P, Pristch O, Mombrú AW, Osinaga E, Pardo H. Docetaxel in chitosan-based nanocapsules conjugated with an anti-Tn antigen mouse/human chimeric antibody as a promising targeting strategy of lung tumors. Int J Biol Macromol 2021; 182:806-814. [PMID: 33857513 DOI: 10.1016/j.ijbiomac.2021.04.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 11/17/2022]
Abstract
The aim of this work was to evaluate the physicochemical and biological properties of docetaxel (DCX) loaded chitosan nanocapsules (CS Nc) functionalized with the monoclonal antibody Chi-Tn (CS-PEG-ChiTn mAb Nc) as a potential improvement treatment for cancer therapy. The Tn antigen is highly specific for carcinomas, and this is the first time that such structure is targeted for drug delivery. The nanocapsules (Ncs), formed as a polymeric shell around an oily core, allowed a 99.9% encapsulation efficiency of DCX with a monodispersity particle size in the range of 200 nm and a high positive surface charge that provide substantial stability to the nanosystems. Release profile of DCX from Ncs showed a sustained and pH dependent behavior with a faster release at acidic pH, which could be favorable in the intracellular drug delivery. We have designed PEGylated CS Nc modified with a monoclonal antibody which recognize Tn antigen, one of the most specific tumor associated antigen. A biotin-avidin approach achieved the successful attachment of the antibody to the nanocapsules. Uptake studies and viability assay conducted in A549 human lung cancer cell line in vitro demonstrate that ChiTn mAb enhance nanoparticles internalization and cell viability reduction. Consequently, these ChiTn functionalized nanocapsules are promising carriers for the active targeting of DCX to Tn expressing carcinomas.
Collapse
Affiliation(s)
- Analía Castro
- Centro NanoMat, DETEMA, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Nora Berois
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo, CP 11400, Uruguay
| | - Antonio Malanga
- Laboratorio de Biofarmacia y Tecnología Farmacéutica, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Canelones, Uruguay
| | - Claudia Ortega
- Unidad de Proteínas Recombinantes, Institut Pasteur de Montevideo, Uruguay
| | - Pablo Oppezzo
- Unidad de Proteínas Recombinantes, Institut Pasteur de Montevideo, Uruguay
| | - Otto Pristch
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Laboratorio de Inmuno-Virología, Institut Pasteur de Montevideo, Uruguay
| | - Alvaro W Mombrú
- Centro NanoMat, DETEMA, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Eduardo Osinaga
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo, CP 11400, Uruguay; Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Helena Pardo
- Centro NanoMat, DETEMA, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
39
|
Xiao Q, Li X, Li Y, Wu Z, Xu C, Chen Z, He W. Biological drug and drug delivery-mediated immunotherapy. Acta Pharm Sin B 2021; 11:941-960. [PMID: 33996408 PMCID: PMC8105778 DOI: 10.1016/j.apsb.2020.12.018] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/03/2020] [Accepted: 11/15/2020] [Indexed: 12/11/2022] Open
Abstract
The initiation and development of major inflammatory diseases, i.e., cancer, vascular inflammation, and some autoimmune diseases are closely linked to the immune system. Biologics-based immunotherapy is exerting a critical role against these diseases, whereas the usage of the immunomodulators is always limited by various factors such as susceptibility to digestion by enzymes in vivo, poor penetration across biological barriers, and rapid clearance by the reticuloendothelial system. Drug delivery strategies are potent to promote their delivery. Herein, we reviewed the potential targets for immunotherapy against the major inflammatory diseases, discussed the biologics and drug delivery systems involved in the immunotherapy, particularly highlighted the approved therapy tactics, and finally offer perspectives in this field.
Collapse
Key Words
- AAs, amino acids
- ACT, adoptive T cell therapy
- AHC, Chlamydia pneumonia
- ALL, acute lymphoblastic leukemia
- AP, ascorbyl palmitate
- APCs, antigen-presenting cells
- AS, atherosclerosis
- ASIT, antigen-specific immunotherapy
- Adoptive cell transfer
- ApoA–I, apolipoprotein A–I
- ApoB LPs, apolipoprotein-B-containing lipoproteins
- Atherosclerosis
- BMPR-II, bone morphogenetic protein type II receptor
- Biologics
- Bregs, regulatory B lymphocytes
- CAR, chimeric antigen receptor
- CCR9–CCL25, CC receptor 9–CC chemokine ligand 25
- CD, Crohn's disease
- CETP, cholesterol ester transfer protein
- CTLA-4, cytotoxic T-lymphocyte-associated protein-4
- CX3CL1, CXXXC-chemokine ligand 1
- CXCL 16, CXC-chemokine ligand 16
- CXCR 2, CXC-chemokine receptor 2
- Cancer immunotherapy
- CpG ODNs, CpG oligodeoxynucleotides
- DAMPs, danger-associated molecular patterns
- DCs, dendritic cells
- DDS, drug delivery system
- DMARDs, disease-modifying antirheumatic drugs
- DMPC, 1,2-dimyristoyl-sn-glycero-3-phosphatidylcholine
- DSS, dextran sulfate sodium
- Dex, dexamethasone
- Drug delivery
- ECM, extracellular matrix
- ECs, endothelial cells
- EGFR, epidermal growth factor receptor
- EPR, enhanced permeability and retention effect
- ET-1, endothelin-1
- ETAR, endothelin-1 receptor type A
- FAO, fatty acid oxidation
- GM-CSF, granulocyte–macrophage colony-stimulating factor
- HA, hyaluronic acid
- HDL, high density lipoprotein
- HER2, human epidermal growth factor-2
- IBD, inflammatory bowel diseases
- ICOS, inducible co-stimulator
- ICP, immune checkpoint
- IFN, interferon
- IL, interleukin
- IT-hydrogel, inflammation-targeting hydrogel
- Immune targets
- Inflammatory diseases
- JAK, Janus kinase
- LAG-3, lymphocyte-activation gene 3
- LDL, low density lipoprotein
- LPS, lipopolysaccharide
- LTB4, leukotriene B4
- MCP-1, monocyte chemotactic protein-1
- MCT, monocrotaline
- MDSC, myeloid-derived suppressor cell
- MHCs, major histocompatibility complexes
- MHPC, 1-myristoyl-2-hydroxy-sn-glycero-phosphocholine
- MIF, migration inhibitory factor
- MM, multiple myeloma
- MMP, matrix metalloproteinase
- MOF, metal–organic framework
- MPO, myeloperoxidase
- MSCs, mesenchymal stem cells
- NF-κB, nuclear factor κ-B
- NK, natural killer
- NPs, nanoparticles
- NSAIDs, nonsteroidal anti-inflammatory drugs
- PAECs, pulmonary artery endothelial cells
- PAH, pulmonary arterial hypertension
- PASMCs, pulmonary arterial smooth muscle cells
- PBMCs, peripheral blood mononuclear cells
- PCSK9, proprotein convertase subtilisin kexin type 9
- PD-1, programmed death protein-1
- PD-L1, programmed cell death-ligand 1
- PLGA, poly lactic-co-glycolic acid
- Pulmonary artery hypertension
- RA, rheumatoid arthritis
- ROS, reactive oxygen species
- SHP-2, Src homology 2 domain–containing tyrosine phosphatase 2
- SLE, systemic lupus erythematosus
- SMCs, smooth muscle cells
- Src, sarcoma gene
- TCR, T cell receptor
- TGF-β, transforming growth factor β
- TILs, tumor-infiltrating lymphocytes
- TIM-3, T-cell immunoglobulin mucin 3
- TLR, Toll-like receptor
- TNF, tumor necrosis factor
- TRAF6, tumor necrosis factor receptor-associated factor 6
- Teff, effector T cell
- Th17, T helper 17
- Tph, T peripheral helper
- Tregs, regulatory T cells
- UC, ulcerative colitis
- VEC, vascular endothelial cadherin
- VEGF, vascular endothelial growth factor
- VISTA, V-domain immunoglobulin-containing suppressor of T-cell activation
- YCs, yeast-derived microcapsules
- bDMARDs, biological DMARDs
- hsCRP, high-sensitivity C-reactive protein
- mAbs, monoclonal antibodies
- mPAP, mean pulmonary artery pressure
- nCmP, nanocomposite microparticle
- rHDL, recombinant HDL
- rhTNFRFc, recombinant human TNF-α receptor II-IgG Fc fusion protein
- scFv, single-chain variable fragment
- α1D-AR, α1D-adrenergic receptor
Collapse
Affiliation(s)
- Qingqing Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong 999077, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
40
|
Beg S, Almalki WH, Khatoon F, Alharbi KS, Alghamdi S, Akhter MH, Khalilullah H, Baothman AA, Hafeez A, Rahman M, Akhter S, Choudhry H. Lipid/polymer-based nanocomplexes in nucleic acid delivery as cancer vaccines. Drug Discov Today 2021; 26:1891-1903. [PMID: 33610757 DOI: 10.1016/j.drudis.2021.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/13/2020] [Accepted: 02/15/2021] [Indexed: 12/24/2022]
Abstract
Cancer vaccines consist of nucleic acid derivatives such as plasmid DNA, small interfering RNA and mRNA, and can be customized according to the patient's needs. Nanomedicines have proven to be exceptionally good as miniaturized drug carriers, and thus they offer great advantages for delivering cancer vaccines. This review provides an overview of the literature on cancer vaccines, from their inception to current developments in the field.
Collapse
Affiliation(s)
- Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Fahmida Khatoon
- Department of Biochemistry, College of Medicine, University of Hail, Saudi Arabia
| | - Khalid S Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Saad Alghamdi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Habibullah Khalilullah
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Saudi Arabia
| | - Abdullah A Baothman
- Ministry of National Guard-Health Affairs, King Saud Bin Abdulaziz University for Health Science (KSAU-HS), King Abdullah International Medical Research Center (KAIMARC), Saudi Arabia
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Mirzapur Pole, Sahranpur, Uttar Pradesh, India
| | - Mahfoozur Rahman
- Department of Pharmaceutical Sciences, SIHAS, Faculty of Health Science, Sam Higginbottom University of Agriculture, Technology and Sciences, Allahabad, India.
| | - Sohail Akhter
- New Product Development, Global R&D, Sterile ops, TEVA Pharmaceutical Industries Ltd., Aston Ln N, Halton, Preston Brook, Runcorn WA7 3FA, UK; Centre de Biophysique Moléculaire, CNRS UPR4301, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| | - Hani Choudhry
- Department of Biochemistry, Cancer Metabolism & Epigenetic Unit, Faculty of Science, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
41
|
Jeon IS, Yoo JD, Gurung S, Kim M, Lee C, Park EJ, Park RW, Lee B, Kim S. Anticancer nanocage platforms for combined immunotherapy designed to harness immune checkpoints and deliver anticancer drugs. Biomaterials 2021; 270:120685. [PMID: 33524811 DOI: 10.1016/j.biomaterials.2021.120685] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/27/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022]
Abstract
The interaction of programmed cell death 1 ligand 1 (PD-L1) with its receptor, programmed cell death 1 (PD-1), inhibits T cell responses. Monoclonal antibodies that block this interaction have been shown effective as immunotherapy. However, only a subset of cancers exhibits a durable response to PD-1/PD-L1 blockade. Moreover, antibody-based immune checkpoint blockade is costly and is occasionally accompanied by systemic side effects. To overcome these limitations of antibody-based immune checkpoint blockade, an immune checkpoint-blocking ferritin nanocage displaying 24 PD-L1 binding peptides (PD-L1pep1) on its surface was designed and constructed. These ferritin nanocages displaying PD-L1pep1 (PpNF) specifically bind to PD-L1 expressed on cancer cells or to purified PD-L1 with a ~30 nM binding affinity. The addition of PpNF to co-cultures of T cells and cancer cells inhibited PD-1/PD-L1 interactions and restored T cell activities. In a mouse model of syngeneic colon cancer, PpNF specifically targeted tumors and showed antitumor activity. Moreover, PpNF nanocages encapsulating the chemotherapeutic drug doxorubicin had more potent antitumor activity than a monoclonal antibody against PD-L1. These results demonstrate that ferritin nanocages displaying surface PD-L1pep1 can be efficiently applied for immunotherapy, especially when encapsulating small chemotherapeutic drugs. These nanocages may have promise as an immunotherapeutic nanomedicine against various solid tumors.
Collapse
Affiliation(s)
- In Seon Jeon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Jae Do Yoo
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Smriti Gurung
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Minseong Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Chanju Lee
- Cancer Immunology Branch, Division of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Eun Jung Park
- Cancer Immunology Branch, Division of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea.
| |
Collapse
|
42
|
Zinc oxide nanoparticles (ZnO NPs) combined with cisplatin and gemcitabine inhibits tumor activity of NSCLC cells. Aging (Albany NY) 2020; 12:25767-25777. [PMID: 33232271 PMCID: PMC7803530 DOI: 10.18632/aging.104187] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/18/2020] [Indexed: 01/06/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common malignancies worldwide. The use of a combination of chemotherapy drugs and zinc oxide nanoparticles (ZnO-NPs), which have proven to induce tumor-selective cell death, reduce the drug resistance and reduce the side effects in vitro. In the present study, we developed ZnO-NPs loaded with both cisplatin (Cp) and gemcitabine (Gem) (ZnO-NPs(Cp/Gem)), then the morphologies and the size distribution of ZnO-NPs(Cp/Gem) particles were observed by transmission electron microscopy (TEM) and dynamic light scattering (DLS). Also, MTT, western blot and Annexin V-PI were used to assess the anti-tumor role of ZnO-NPs(Cp/Gem) in A549 cells. The viability for A549 cells showed a significant decrease in the ZnO NPs(Cp/Gem) group, respectively relative to Cp, Gem, the combination of Cp and Gem (Cp+Gem), and ZnO-NPs loaded with Cp (ZnO-NPs(Cp)) or Gem (ZnO-NPs(Gem)). Furthermore, ZnO-NPs(Cp/Gem) remarkably enhanced the apoptosis-promoting effect of Cp and Gem in A549 cells. The xenograft model showed that Zno-NPS (Cp/Gem) significantly enhanced the inhibition of Cp and Gem on tumor formation. The above results suggested that therapy of NSCLC with ZnO-NPs(Cp/Gem) could enhance the cytotoxic action of chemotherapeutic agents synergistically, indicating a promising potential for ZnO-NPs in antitumor applications.
Collapse
|
43
|
Siamof CM, Goel S, Cai W. Moving Beyond the Pillars of Cancer Treatment: Perspectives From Nanotechnology. Front Chem 2020; 8:598100. [PMID: 33240859 PMCID: PMC7683771 DOI: 10.3389/fchem.2020.598100] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Nanotechnology has made a significant impact on basic and clinical cancer research over the past two decades. Owing to multidisciplinary advances, cancer nanotechnology aims to address the problems in current cancer treatment paradigms, with the ultimate goal to improve treatment efficacy, increase patient survival, and decrease toxic side-effects. The potential for use of nanomedicine in cancer targeting and therapy has grown, and is now used to advance the four traditional pillars of cancer treatment: surgery, chemotherapy, radiation therapy and the newest pillar, immunotherapy. In this review we provide an overview of notable advances of nanomedicine in improving drug delivery, radiation therapy and immunotherapy. Potential barriers in the translation of nanomedicine from bench to bedside as well as strategies to overcome these barriers are also discussed. Promising preclinical findings highlight the translational and clinical potential of integrating nanotechnology approaches into cancer care.
Collapse
Affiliation(s)
- Cerise M. Siamof
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, United States
| | - Shreya Goel
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
44
|
Gu Z, Da Silva CG, Van der Maaden K, Ossendorp F, Cruz LJ. Liposome-Based Drug Delivery Systems in Cancer Immunotherapy. Pharmaceutics 2020; 12:E1054. [PMID: 33158166 PMCID: PMC7694212 DOI: 10.3390/pharmaceutics12111054] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy has shown remarkable progress in recent years. Nanocarriers, such as liposomes, have favorable advantages with the potential to further improve cancer immunotherapy and even stronger immune responses by improving cell type-specific delivery and enhancing drug efficacy. Liposomes can offer solutions to common problems faced by several cancer immunotherapies, including the following: (1) Vaccination: Liposomes can improve the delivery of antigens and other stimulatory molecules to antigen-presenting cells or T cells; (2) Tumor normalization: Liposomes can deliver drugs selectively to the tumor microenvironment to overcome the immune-suppressive state; (3) Rewiring of tumor signaling: Liposomes can be used for the delivery of specific drugs to specific cell types to correct or modulate pathways to facilitate better anti-tumor immune responses; (4) Combinational therapy: Liposomes are ideal vehicles for the simultaneous delivery of drugs to be combined with other therapies, including chemotherapy, radiotherapy, and phototherapy. In this review, different liposomal systems specifically developed for immunomodulation in cancer are summarized and discussed.
Collapse
Affiliation(s)
- Zili Gu
- Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (Z.G.); (C.G.D.S.)
| | - Candido G. Da Silva
- Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (Z.G.); (C.G.D.S.)
| | - Koen Van der Maaden
- Tumor Immunology Group, Department of Immunology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (K.v.d.M.); (F.O.)
- TECOdevelopment GmbH, 53359 Rheinbach, Germany
| | - Ferry Ossendorp
- Tumor Immunology Group, Department of Immunology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (K.v.d.M.); (F.O.)
| | - Luis J. Cruz
- Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (Z.G.); (C.G.D.S.)
| |
Collapse
|
45
|
Yang T, Yu S, Liu L, Sun Y, Lan Y, Ma X, Zhu R, Li L, Hou Y, Liu Y. Dual polymeric prodrug co-assembled nanoparticles with precise ratiometric co-delivery of cisplatin and metformin for lung cancer chemoimmunotherapy. Biomater Sci 2020; 8:5698-5714. [PMID: 32930254 DOI: 10.1039/d0bm01191f] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The combination therapy of cisplatin (CDDP) and metformin (MET) is a clinical strategy to enhance therapeutic outcomes in lung cancer. However, the efficacy of this combination is limited due to the asynchronous pharmacokinetic behavior of CDDP and MET, used as free drugs. Therefore, in this work, hyaluronic acid-cisplatin/polystyrene-polymetformin (HA-CDDP/PMet) dual-prodrug co-assembled nanoparticles were developed, with precise ratiometric co-delivery of CDDP and MET for chemo-immunotherapy against lung cancer. The HA-CDDP/PMet NPs showed a spherical morphology with an average particle size of 166.5 nm and a zeta potential of -17.4 mV at an HA-CDDP and PMet mass ratio of 1/1. The content of CDDP and MET in HA-CDDP/PMet NPs was 3.7% and 15.2%, respectively. In vitro antitumor effects of CDDP and MET resulted in an improved synergistic action on proliferation inhibition and apoptosis induction on Lewis lung cancer cells. Moreover, in vivo by co-delivered HA-CDDP/PMet NPs into tumor cells, with an excellent intracellular CDDP and MET cleavage. These nanoparticles exhibited significantly increased tumor accumulation and tumor growth inhibition and prolonged animal overall survival in Lewis lung cancer bearing mice without nephrotoxicity, excess of free drugs and homo-prodrugs. The synergistic effect of MET and CDDP in HA-CDDP/PMet NPs resulted in up-regulation of the cleaved poly(ADP)-ribose polymerase (PARP) protein to induce tumor cell apoptosis, and down-regulation of the excision repair cross-complementation group 1 (ERCC1) protein level to decrease the resistance to CDDP. The synergistic effect of MET and CDDP in HA-CDDP/PMet NPs also resulted in induction of the adenosine monophosphate (AMP)-activated protein kinase-α (AMPK-α) pathway and inhibition of the mammalian target of rapamycin (mTOR), finally exerting a chemotherapeutic effect and modulating a potent immunotherapeutic function with an increase in CD4+ and CD8+ T cells, a concomitant decrease in regulatory T (Treg) cells, and an increased expression of the cytokines IFN-γ and TNF-α. Therefore, the immunochemotherapy using CDDP and MET mediated by this dual prodrug co-assembled nano-platform might provide a promising treatment strategy against lung cancer.
Collapse
Affiliation(s)
- Tong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan, 750004, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Banstola A, Jeong JH, Yook S. Immunoadjuvants for cancer immunotherapy: A review of recent developments. Acta Biomater 2020; 114:16-30. [PMID: 32777293 DOI: 10.1016/j.actbio.2020.07.063] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/14/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy evolved as a new treatment modality to eradicate tumor cells and has gained in popularity after its successful clinical transition. By activating antigen-presenting cells (APCs), and thus, inducing innate or adaptive immune responses, immunoadjuvants have become promising tools for cancer immunotherapy. Different types of immunoadjuvants such as toll-like receptor (TLR) agonists, exosomes, and metallic and plant-derived immunoadjuvants have been studied for their immunological effects. However, the clinical use of immunoadjuvants is limited by short response rates and various side-effects. The rapid progress made in the development of nanoparticle systems as immunoadjuvant carrier vehicles has provided potential carriers for cancer immunotherapy. In this review article, we describe different types of immunoadjuvants, their limitations, modes of action, and the reasons for their clinical adoption. In addition, we review recent progress made in the nanoparticle-based immunoadjuvant field and on the combined use of nanoparticle-based immunoadjuvants and chemotherapy, phototherapy, radiation therapy, and immune checkpoint inhibitor-based therapy. STATEMENT OF SIGNIFICANCE: Cancer immunotherapy emerged as a new hope for treating malignant tumors. Different types of immunoadjuvants serve as an important tool for cancer immunotherapy by activating an innate or adaptive immune response. Limitation of free immunoadjuvant has paved the path for the development of nanoparticle-based immunoadjuvant therapy with the hope of prolonging the therapeutic efficacy. This review highlights the recent advancement made in nanoparticle-based immunoadjuvant therapy in modulating the adaptive and innate immune system. The application of the combinatorial approach of chemotherapy, phototherapy, radiation therapy adds synergy in nanoparticle-based immunoadjuvant therapy. It will broaden the reader's understanding on the recent progress made in immunotherapy with the aid of immunoadjuvant-based nanosystem.
Collapse
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
47
|
Yang R, Zhang Z, Fu S, Hou T, Mu W, Liang S, Gao T, Guan L, Fang Y, Liu Y, Zhang N. Charge and Size Dual Switchable Nanocage for Novel Triple-Interlocked Combination Therapy Pattern. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000906. [PMID: 32999836 PMCID: PMC7509747 DOI: 10.1002/advs.202000906] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/15/2020] [Indexed: 05/28/2023]
Abstract
Combination therapy is a current hot topic in cancer treatment. Multiple synergistic effects elicited by combined drugs are essential in improving antitumor activity. Herein, a pH-triggered charge and size dual switchable nanocage co-loaded with abemaciclib and IMD-0354 (PA/PI-ND) is reported, exhibiting a novel triple-interlocked combination of chemotherapy, immunotherapy, and chemoimmunotherapy. The charge reversal polymer NGR-poly(ethylene glycol)-poly(l-lysine)-dimethylmaleic anhydride (NGR-PEG-PLL-DMA, ND) in PA/PI-ND promotes the pH-triggered charge reversal from negative to positive and size reduction from about 180 to 10 nm in an acidic tumor microenvironment, which greatly enhances cellular uptake and tumor tissue deep penetration. With the PA/PI-ND triple-interlocked combination therapy, the chemotherapeutic effect is enhanced by the action of abemaciclib to induce cell cycle arrest in the G1 phase, together with the reduction in cyclin D levels caused by IMD-0354. The dual anti-tumor promoting immunotherapy is achieved by abemaciclib selectively inhibiting the proliferation of regulatory T cells (Tregs) and by IMD-0354 promoting tumor-associated macrophage (TAM) repolarization from an M2 to M1 phenotype. Furthermore, PA/PI-ND has improved anti-tumor efficiency resulting from the third synergistic effect provided by chemoimmunotherapy. Taken together, PA/PI-ND is a promising strategy to guide the design of future drug delivery carriers and cancer combination therapy.
Collapse
Affiliation(s)
- Rui Yang
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Zipeng Zhang
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Shunli Fu
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Teng Hou
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Weiwei Mu
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Shuang Liang
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Tong Gao
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Li Guan
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Yuxiao Fang
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Yongjun Liu
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| | - Na Zhang
- Department of PharmaceuticsKey Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical Sciences Shandong University44 Wenhuaxi RoadJinanShandong250012China
| |
Collapse
|
48
|
Mu W, Chu Q, Liu Y, Zhang N. A Review on Nano-Based Drug Delivery System for Cancer Chemoimmunotherapy. NANO-MICRO LETTERS 2020; 12:142. [PMID: 34138136 PMCID: PMC7770879 DOI: 10.1007/s40820-020-00482-6] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/11/2020] [Indexed: 05/11/2023]
Abstract
Although notable progress has been made on novel cancer treatments, the overall survival rate and therapeutic effects are still unsatisfactory for cancer patients. Chemoimmunotherapy, combining chemotherapeutics and immunotherapeutic drugs, has emerged as a promising approach for cancer treatment, with the advantages of cooperating two kinds of treatment mechanism, reducing the dosage of the drug and enhancing therapeutic effect. Moreover, nano-based drug delivery system (NDDS) was applied to encapsulate chemotherapeutic agents and exhibited outstanding properties such as targeted delivery, tumor microenvironment response and site-specific release. Several nanocarriers have been approved in clinical cancer chemotherapy and showed significant improvement in therapeutic efficiency compared with traditional formulations, such as liposomes (Doxil®, Lipusu®), nanoparticles (Abraxane®) and micelles (Genexol-PM®). The applications of NDDS to chemoimmunotherapy would be a powerful strategy for future cancer treatment, which could greatly enhance the therapeutic efficacy, reduce the side effects and optimize the clinical outcomes of cancer patients. Herein, the current approaches of cancer immunotherapy and chemoimmunotherapy were discussed, and recent advances of NDDS applied for chemoimmunotherapy were further reviewed.
Collapse
Affiliation(s)
- Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Qihui Chu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
49
|
Qian C, Yang LJ, Cui H. Recent Advances in Nanotechnology for Dendritic Cell-Based Immunotherapy. Front Pharmacol 2020; 11:960. [PMID: 32694998 PMCID: PMC7338589 DOI: 10.3389/fphar.2020.00960] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) are the most important antigen-presenting cells that determine cancer immune responses by regulating immune activation and tolerance, especially in the initiation stage of specific responses. Manipulation of DCs to enhance specific antitumor immune response is considered to be a powerful tool for tumor eradication. Nanotechnology, which can incorporate multifunction components and show spatiotemporal control properties, is of great interest and is widely investigated for its ability to improve immune response activity against cancer and even for prevention and avoiding recurrence. In this mini-review, we aim to provide a general view of DC-based immunotherapy, including that involving the promising nanotechnology. Particularly we discuss: (1) manipulation or engineering of DCs for adoptive vaccination, (2) employing DCs as a combination to more existing therapeutics in tumor treatment, and (3) direct modulation of DCs in vivo to enhance antigen presentation efficacy and priming T cells subsequently. We comprehensively discuss the updates on the application of nanotechnology in DC-based immunotherapy and provide some insights on the challenges and opportunities of DC-based immunotherapeutics, including the potential of nanotechnology, against cancers.
Collapse
Affiliation(s)
| | | | - Hong Cui
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
50
|
Aggarwal R, Targhotra M, Sahoo P, Chauhan MK. Onychomycosis: Novel strategies for treatment. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|