1
|
Martin JD, Toh K, Martin MR, Chen P, Wang C, Igarashi K, Mpekris F, Stylianopoulos T, Stuber MD, Kataoka K, Cabral H. Bone marrow vessels are hyperpermeable to macromolecules and nanoscale medicine in a size-dependent manner. J Control Release 2025; 382:113669. [PMID: 40158811 DOI: 10.1016/j.jconrel.2025.113669] [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: 06/30/2024] [Revised: 01/22/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Bone marrow (BM) has roles in health and disease, so systemically administered nanocarriers (NCs) targeting or avoiding BM are desirable. While the hydrodynamic diameter of NCs can be tuned to target or avoid various organs, the size dependence of extravasation from BM vessels is unknown. To clarify the size dependence of passive transvascular transport in the BM, we performed vessel permeability measurements in murine calvaria using confocal fluorescent microscopy with fluorescently labeled dextrans, albumin, and polymeric micelles as model probes. Unexpectedly, we found the permeability of BM vessels to macromolecules decreases with increasing hydrodynamic diameter between 4 nm and 32 nm. We modeled this permeability data with mathematical models to predict an effective pore size for sinusoids of 47 nm and non-sinusoids of 37 nm, with estimated maximum pore sizes of 61 nm and 53 nm, respectively. Finally, we tested these model predictions by demonstrating that the extravasation of 70 nm polymeric micelles, which are larger than the estimated maximum pore size, is hindered relative to 30 nm polymeric micelles. These results establish design criteria for controlling NC hydrodynamic diameter towards modulating delivery to BM.
Collapse
Affiliation(s)
- John D Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, Japan
| | - Margaret R Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Chenyu Wang
- Process Systems and Operations Research Laboratory, Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA
| | - Kazunori Igarashi
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Matthew D Stuber
- Process Systems and Operations Research Laboratory, Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA.
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan.
| |
Collapse
|
2
|
Xue Q, Wang W, Liu J, Wang D, Zhang T, Shen T, Liu X, Wang X, Shao X, Zhou W, Fang X. LRPPRC confers enhanced oxidative phosphorylation metabolism in triple-negative breast cancer and represents a therapeutic target. J Transl Med 2025; 23:372. [PMID: 40133967 PMCID: PMC11938637 DOI: 10.1186/s12967-024-05946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/06/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a highly malignant tumor that requires effective therapeutic targets and drugs. Oxidative phosphorylation (OXPHOS) is a metabolic vulnerability of TNBC, but the molecular mechanism responsible for the enhanced OXPHOS remains unclear. The current strategies that target the electronic transfer function of OXPHOS cannot distinguish tumor cells from normal cells. Investigating the mechanism underlying OXPHOS regulation and developing corresponding therapy strategies for TNBC is of great significance. METHODS Immunohistochemistry and sequencing data reanalysis were used to investigate LRPPRC expression in TNBC. In vitro and in vivo assays were applied to investigate the roles of LRPPRC in TNBC progression. RT-qPCR, immunoblotting, and Seahorse XF assay were used to examine LRPPRC's functions in the expression of OXPHOS subunits and energy metabolism. In vitro and in vivo functional assays were used to test the therapeutic effect of gossypol acetate (GAA), a traditional gynecological drug, on LRPPRC suppression and OXPOHS inhibition. RESULTS LRPPRC was specifically overexpressed in TNBC. LRPPRC knockdown suppressed the proliferation, metastasis, and tumor formation of TNBC cells. LRPPRC enhanced OXPHOS metabolism by increasing the expression of OXPHOS complex subunits encoded by the mitochondrial genome. GAA inhibited OXPHOS metabolism by directly binding LRPPRC, causing LRPPRC degradation, and downregulating the expression of OXPHOS complex subunits encoded by the mitochondrial genome. GAA administration suppressed TNBC cell proliferation, metastasis in vitro, and tumor formation in vivo. CONCLUSIONS This work demonstrated a new regulatory pathway of TNBC to promote the expression of mitochondrial genes by upregulating the nuclear gene LRPPRC, resulting in increased OXPHOS. We also suggested a promising therapeutic target LRPPRC for TNBC, and its inhibitor, the traditional gynecological medicine GAA, presented significant antitumor activity.
Collapse
Affiliation(s)
- Qiqi Xue
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wenxi Wang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Jie Liu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Dachi Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Tianyu Zhang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Tingting Shen
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Xiangsheng Liu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Xiaojia Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xiying Shao
- Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | - Wei Zhou
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China.
| | - Xiaohong Fang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China.
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
3
|
Chen Y, Dai L, Shi K, Pan M, Yuan L, Qian Z. Cabazitaxel-Loaded Thermosensitive Hydrogel System for Suppressed Orthotopic Colorectal Cancer and Liver Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404800. [PMID: 38934894 PMCID: PMC11434046 DOI: 10.1002/advs.202404800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
The treatment of colorectal cancer is always a major challenge in the field of cancer research. The number of estimated new cases of colorectal cancer worldwide in 2020 is 1 148 515, and the estimated number of deaths is 576 858, revealing that mortality accounted for approximately half of the disease incidence. The development of new drugs and strategies for colorectal cancer treatment is urgently needed. Thermosensitive injectable hydrogel PDLLA-PEG-PDLLA (PLEL) loaded with cabazitaxel (CTX) is used to explore its anti-tumor effect on mice with orthotopic colorectal cancer. CTX/PLEL is characterized by a solution state at room temperature and a hydrogel state at physiologic temperature. The excipients MPEG-PCL and PDLLA-PEG-PDLLA have good biocompatibility and biodegradability. The simple material synthesis and preparation process renders this system cost-effective and more conducive to clinical transformation. An orthotopic colorectal cancer model is established by transplantation subcutaneous tumors onto the cecum of mice. According to the results of experiments in vivo, CTX/PLEL significantly inhibits orthotopic colorectal cancer and liver metastasis in mice. The results indicate that CTX/PLEL nanoparticle preparations have high security and excellent anti-tumor effects, and have great application potential in colorectal cancer therapy.
Collapse
Affiliation(s)
- Yu Chen
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Liqun Dai
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Kun Shi
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Meng Pan
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Liping Yuan
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Zhiyong Qian
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
4
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
5
|
Meng J, Wang ZG, Zhao X, Wang Y, Chen DY, Liu DL, Ji CC, Wang TF, Zhang LM, Bai HX, Li BY, Liu Y, Wang L, Yu WG, Yin ZT. Silica nanoparticle design for colorectal cancer treatment: Recent progress and clinical potential. World J Clin Oncol 2024; 15:667-673. [PMID: 38946830 PMCID: PMC11212613 DOI: 10.5306/wjco.v15.i6.667] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 06/24/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and the second most common cause of cancer death. Nanotherapies are able to selectively target the delivery of cancer therapeutics, thus improving overall antitumor efficiency and reducing conventional chemotherapy side effects. Mesoporous silica nanoparticles (MSNs) have attracted the attention of many researchers due to their remarkable advantages and biosafety. We offer insights into the recent advances of MSNs in CRC treatment and their potential clinical application value.
Collapse
Affiliation(s)
- Jin Meng
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Zhi-Gang Wang
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Xiu Zhao
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Ying Wang
- Acupuncture and Tuina College, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning Province, China
| | - De-Yu Chen
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - De-Long Liu
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Cheng-Chun Ji
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Tian-Fu Wang
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Li-Mei Zhang
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian 116001, Liaoning Province, China
| | - Hai-Xia Bai
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Bo-Yang Li
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Yuan Liu
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Lei Wang
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Wei-Gang Yu
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Zhi-Tao Yin
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| |
Collapse
|
6
|
Shaw I, Boafo GF, Ali YS, Liu Y, Mlambo R, Tan S, Chen C. Advancements and prospects of lipid-based nanoparticles: dual frontiers in cancer treatment and vaccine development. J Microencapsul 2024; 41:226-254. [PMID: 38560994 DOI: 10.1080/02652048.2024.2326091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024]
Abstract
Cancer is a complex heterogeneous disease that poses a significant public health challenge. In recent years, lipid-based nanoparticles (LBNPs) have expanded drug delivery and vaccine development options owing to their adaptable, non-toxic, tuneable physicochemical properties, versatile surface functionalisation, and biocompatibility. LBNPs are tiny artificial structures composed of lipid-like materials that can be engineered to encapsulate and deliver therapeutic agents with pinpoint accuracy. They have been widely explored in oncology; however, our understanding of their pharmacological mechanisms, effects of their composition, charge, and size on cellular uptake, tumour penetration, and how they can be utilised to develop cancer vaccines is still limited. Hence, we reviewed LBNPs' unique characteristics, biochemical features, and tumour-targeting mechanisms. Furthermore, we examined their ability to enhance cancer therapies and their potential contribution in developing anticancer vaccines. We critically analysed their advantages and challenges impeding swift advancements in oncology and highlighted promising avenues for future research.
Collapse
Affiliation(s)
- Ibrahim Shaw
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - George Frimpong Boafo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Yimer Seid Ali
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
- Department of Pharmacy, College of Medicine and Health Science, Wollo University, Dessie, Ethiopia
| | - Yang Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Ronald Mlambo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Chuanpin Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
7
|
Nele V, Campani V, Alia Moosavian S, De Rosa G. Lipid nanoparticles for RNA delivery: Self-assembling vs driven-assembling strategies. Adv Drug Deliv Rev 2024; 208:115291. [PMID: 38514018 DOI: 10.1016/j.addr.2024.115291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
Among non-viral vectors, lipid nanovectors are considered the gold standard for the delivery of RNA therapeutics. The success of lipid nanoparticles for RNA delivery, with three products approved for human use, has stimulated further investigation into RNA therapeutics for different pathologies. This requires decoding the pathological intracellular processes and tailoring the delivery system to the target tissue and cells. The complexity of the lipid nanovectors morphology originates from the assembling of the lipidic components, which can be elicited by various methods able to drive the formation of nanoparticles with the desired organization. In other cases, pre-formed nanoparticles can be mixed with RNA to induce self-assembly and structural reorganization into RNA-loaded nanoparticles. In this review, the most relevant lipid nanovectors and their potentialities for RNA delivery are described on the basis of the assembling mechanism and of the particle architecture.
Collapse
Affiliation(s)
- Valeria Nele
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Virginia Campani
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Seyedeh Alia Moosavian
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy.
| |
Collapse
|
8
|
Garbati P, Picco C, Magrassi R, Signorello P, Cacopardo L, Dalla Serra M, Faticato MG, De Luca M, Balestra F, Scavo MP, Viti F. Targeting the Gut: A Systematic Review of Specific Drug Nanocarriers. Pharmaceutics 2024; 16:431. [PMID: 38543324 PMCID: PMC10974668 DOI: 10.3390/pharmaceutics16030431] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 01/05/2025] Open
Abstract
The intestine is essential for the modulation of nutrient absorption and the removal of waste. Gut pathologies, such as cancer, inflammatory bowel diseases (IBD), irritable bowel syndrome (IBS), and celiac disease, which extensively impact gut functions, are thus critical for human health. Targeted drug delivery is essential to tackle these diseases, improve therapy efficacy, and minimize side effects. Recent strategies have taken advantage of both active and passive nanocarriers, which are designed to protect the drug until it reaches the correct delivery site and to modulate drug release via the use of different physical-chemical strategies. In this systematic review, we present a literature overview of the different nanocarriers used for drug delivery in a set of chronic intestinal pathologies, highlighting the rationale behind the controlled release of intestinal therapies. The overall aim is to provide the reader with useful information on the current approaches for gut targeting in novel therapeutic strategies.
Collapse
Affiliation(s)
- Patrizia Garbati
- Institute of Biophysics, National Research Council, Via De Marini 16, 16149 Genova, Italy; (P.G.); (C.P.); (R.M.); (M.D.S.)
| | - Cristiana Picco
- Institute of Biophysics, National Research Council, Via De Marini 16, 16149 Genova, Italy; (P.G.); (C.P.); (R.M.); (M.D.S.)
| | - Raffaella Magrassi
- Institute of Biophysics, National Research Council, Via De Marini 16, 16149 Genova, Italy; (P.G.); (C.P.); (R.M.); (M.D.S.)
| | - Paolo Signorello
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122 Pisa, Italy; (P.S.); (L.C.)
- Research Center ‘E. Piaggio’, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Centro 3R: Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 56122 Pisa, Italy
| | - Ludovica Cacopardo
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122 Pisa, Italy; (P.S.); (L.C.)
- Research Center ‘E. Piaggio’, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Centro 3R: Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 56122 Pisa, Italy
| | - Mauro Dalla Serra
- Institute of Biophysics, National Research Council, Via De Marini 16, 16149 Genova, Italy; (P.G.); (C.P.); (R.M.); (M.D.S.)
| | - Maria Grazia Faticato
- Pediatric Surgery, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Maria De Luca
- National Institute of Gastroenterology, IRCCS de Bellis, Via Turi 27, 70013 Castellana Grotte, Bari, Italy; (M.D.L.); (F.B.); (M.P.S.)
| | - Francesco Balestra
- National Institute of Gastroenterology, IRCCS de Bellis, Via Turi 27, 70013 Castellana Grotte, Bari, Italy; (M.D.L.); (F.B.); (M.P.S.)
| | - Maria Principia Scavo
- National Institute of Gastroenterology, IRCCS de Bellis, Via Turi 27, 70013 Castellana Grotte, Bari, Italy; (M.D.L.); (F.B.); (M.P.S.)
| | - Federica Viti
- Institute of Biophysics, National Research Council, Via De Marini 16, 16149 Genova, Italy; (P.G.); (C.P.); (R.M.); (M.D.S.)
| |
Collapse
|
9
|
Sobhanan J, Ono K, Okamoto T, Sawada M, Weiss PS, Biju V. Photosensitizer-singlet oxygen sensor conjugated silica nanoparticles for photodynamic therapy and bioimaging. Chem Sci 2024; 15:2007-2018. [PMID: 38332815 PMCID: PMC10848760 DOI: 10.1039/d3sc03877g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/10/2023] [Indexed: 02/10/2024] Open
Abstract
Intracellular singlet oxygen (1O2) generation and detection help optimize the outcome of photodynamic therapy (PDT). Theranostics programmed for on-demand phototriggered 1O2 release and bioimaging have great potential to transform PDT. We demonstrate an ultrasensitive fluorescence turn-on sensor-sensitizer-RGD peptide-silica nanoarchitecture and its 1O2 generation-releasing-storing-sensing properties at the single-particle level or in living cells. The sensor and sensitizer in the nanoarchitecture are an aminomethyl anthracene (AMA)-coumarin dyad and a porphyrin or CdSe/ZnS quantum dots (QDs), respectively. The AMA in the dyad quantitatively quenches the fluorescence of coumarin by intramolecular electron transfer, the porphyrin or QD moiety generates 1O2, and the RGD peptide facilitates intracellular delivery. The small size, below 200 nm, as verified by scanning electron microscopy and differential light scattering measurements, of the architecture within the 1O2 diffusion length enables fast and efficient intracellular fluorescence switching by the tandem ultraviolet (UV)-visible or visible-near-infrared (NIR) photo-triggering. While the red emission and 1O2 generation by the porphyrin are continually turned on, the blue emission of coumarin is uncaged into 230-fold intensity enhancement by on-demand photo-triggering. The 1O2 production and release by the nanoarchitecture enable spectro-temporally controlled cell imaging and apoptotic cell death; the latter is verified from cytotoxic data under dark and phototriggering conditions. Furthermore, the bioimaging potential of the TCPP-based nanoarchitecture is examined in vivo in B6 mice.
Collapse
Affiliation(s)
- Jeladhara Sobhanan
- Graduate School of Environmental Science, Hokkaido University Sapporo Hokkaido 060-0810 Japan
- Department of Chemistry, Rice University Houston Texas 77005 USA
| | - Kenji Ono
- Research Institute of Environmental Medicine, Nagoya University Nagoya 464-8601 Japan
| | - Takuya Okamoto
- Graduate School of Environmental Science, Hokkaido University Sapporo Hokkaido 060-0810 Japan
- Research Institute for Electronic Science, Hokkaido University Sapporo Hokkaido 001-0020 Japan
| | - Makoto Sawada
- Research Institute of Environmental Medicine, Nagoya University Nagoya 464-8601 Japan
| | - Paul S Weiss
- California NanoSystems Institute and the Departments of Chemistry and Biochemistry, Bioengineering, and Materials Science and Engineering, University of California Los Angeles CA 90095-1487 USA
| | - Vasudevanpillai Biju
- Graduate School of Environmental Science, Hokkaido University Sapporo Hokkaido 060-0810 Japan
- Research Institute for Electronic Science, Hokkaido University Sapporo Hokkaido 001-0020 Japan
| |
Collapse
|
10
|
Luo L, Wang X, Liao YP, Xu X, Chang CH, Nel AE. Reprogramming the pancreatic cancer stroma and immune landscape by a silicasome nanocarrier delivering nintedanib, a protein tyrosine kinase inhibitor. NANO TODAY 2024; 54:102058. [PMID: 38681872 PMCID: PMC11044875 DOI: 10.1016/j.nantod.2023.102058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
The prevailing desmoplastic stroma and immunosuppressive microenvironment within pancreatic ductal adenocarcinoma (PDAC) pose substantial challenges to therapeutic intervention. Despite the potential of protein tyrosine kinase (PTK) inhibitors in mitigating the desmoplastic stromal response and enhancing the immune milieu, their efficacy is curtailed by suboptimal pharmacokinetics (PK) and insufficient tumor penetration. To surmount these hurdles, we have pioneered a novel strategy, employing lipid bilayer-coated mesoporous silica nanoparticles (termed "silicasomes") as a carrier for the delivery of Nintedanib. Nintedanib, a triple PTK inhibitor that targets vascular endothelial growth factor, platelet-derived growth factor and fibroblast growth factor receptors, was encapsulated in the pores of silicasomes via a remote loading mechanism for weak bases. This innovative approach not only enhanced pharmacokinetics and intratumor drug concentrations but also orchestrated a transformative shift in the desmoplastic and immune landscape in a robust orthotopic KRAS-mediated pancreatic carcinoma (KPC) model. Our results demonstrate attenuation of vascular density and collagen content through encapsulated Nintedanib treatment, concomitant with significant augmentation of the CD8+/FoxP3+ T-cell ratio. This remodeling was notably correlated with tumor regression in the KPC model. Strikingly, the synergy between encapsulated Nintedanib and anti-PD-1 immunotherapy further potentiated the antitumor effect. Both free and encapsulated Nintedanib induced a transcriptional upregulation of PD-L1 via the extracellular signal-regulated kinase (ERK) pathway. In summary, our pioneering approach involving the silicasome carrier not only improved antitumor angiogenesis but also profoundly reshaped the desmoplastic stromal and immune landscape within PDAC. These insights hold excellent promise for the development of innovative combinatorial strategies in PDAC therapy.
Collapse
Affiliation(s)
- Lijia Luo
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Xiang Wang
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Yu-Pei Liao
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Xiao Xu
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Chong Hyun Chang
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Andre E. Nel
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Chen M, Wang S, Chen Y, Shen H, Chen L, Ding L, Tang Q, Yang Z, Chen W, Shen Z. Precision cardiac targeting: empowering curcumin therapy through smart exosome-mediated drug delivery in myocardial infarction. Regen Biomater 2023; 11:rbad108. [PMID: 38223291 PMCID: PMC10786676 DOI: 10.1093/rb/rbad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 01/16/2024] Open
Abstract
Nanoparticle-mediated drug delivery has emerged as a highly promising and effective therapeutic approach for addressing myocardial infarction. However, clinical translation tends to be a failure due to low cardiac retention as well as liver and spleen entrapment in previous therapies. Herein, we report a two-step exosome delivery system, which precludes internalization by the mononuclear phagocyte system before the delivery of therapeutic cardiac targeting exosomes (ExoCTP). Importantly, curcumin released by ExoCTP diminishes reactive oxygen species over-accumulation in ischemic myocardium, as well as serum levels of lactate dehydrogenase, malonyldialdehyde, superoxide dismutase and glutathione, indicating better antioxidant capacity than free curcumin. Finally, our strategy was proven to greatly potentiate the delivery and therapeutic efficacy of curcumin without systemic toxicity. Taken together, our smart exosome-mediated drug delivery strategy can serve either as therapeutics alone or in combination with other drugs for effective heart targeting and subsequent wound healing.
Collapse
Affiliation(s)
- Ming Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Shengnan Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Yihuan Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Han Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Lei Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Liang Ding
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Qingsong Tang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Ziying Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Weiqian Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215006, China
| |
Collapse
|
12
|
Wang L, Zhang T, Zheng Y, Li Y, Tang X, Chen Q, Mao W, Li W, Liu X, Zhu J. Combination of irinotecan silicasome nanoparticles with radiation therapy sensitizes immunotherapy by modulating the activation of the cGAS/STING pathway for colorectal cancer. Mater Today Bio 2023; 23:100809. [PMID: 37779919 PMCID: PMC10540048 DOI: 10.1016/j.mtbio.2023.100809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
Our previous clinical trial (Identifier: NCT02605265) revealed that addition of irinotecan (IRIN) to neoadjuvant chemoradiotherapy for rectal cancer could improve the curative effect. However, the adverse effects caused by IRIN limited the wide application of IRIN chemoradiotherapy. This study aimed to explore the mechanism under the synergistic effects of IRIN plus radiation therapy in colorectal cancer (CRC) cells and optimization of IRIN delivery via a silicasome nanocarrier in vivo. Our results revealed that compared with single IRIN or radiation treatment, IRIN combined with radiation therapy remarkably activated the intracellular cGAS/STING pathway, and promoted the expression levels of major histocompatibility complex class I (MHC-I) and programmed death ligand 1 (PD-L1). Further, a silicasome (mesoporous silica nanoparticle coated with lipid bilayer) nanocarrier was utilized to improve the delivery of IRIN with enhanced efficacy and reduced side effects. In the MC38 CRC syngeneic tumor model, IRIN silicasome combined with radiation therapy demonstrated a greater antitumor efficacy than free IRIN plus radiation therapy. Flow cytometry showed the increased number of CD4+ T cells, CD8+ T cells, and dendritic cells (DCs) in tumor in the IRIN silicasome plus radiation group. The immunofluorescence staining further confirmed the activated immune microenvironment with the elevated interferon-γ (IFN-γ) deposition. Besides, the antitumor effect of IRIN silicasome plus radiation therapy was synergistically enhanced by anti-PD-1 immunotherapy. These findings indicated that the combination of IRIN silicasome with radiation therapy could sensitize immunotherapy by manipulating the cGAS/STING pathway serving as a new strategy for CRC treatment.
Collapse
Affiliation(s)
- Lu Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Tianyu Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yile Zheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yuting Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xiyuan Tang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Qianping Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Wei Mao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Weiwei Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Xiangsheng Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Ji Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
13
|
Chen Y, Hu Z, Jiang J, Liu C, Gao S, Song M, Hang T. Evaluation of pharmacological and pharmacokinetic herb-drug interaction between irinotecan hydrochloride injection and Kangai injection in colorectal tumor-bearing mice and healthy rats. Front Pharmacol 2023; 14:1282062. [PMID: 38094890 PMCID: PMC10716275 DOI: 10.3389/fphar.2023.1282062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/15/2023] [Indexed: 07/17/2024] Open
Abstract
Introduction: Kangai (KA) injection, a Chinese herbal injection, is often used in combination with irinotecan (CPT-11) to enhance the effectiveness of anti-colorectal cancer treatment and alleviate side effects. However, the combined administration of this herb-drug pair remains controversial due to limited pre-clinical evidence and safety concerns. This study aimed to determine the pre-clinical herb-drug interactions between CPT-11 and KA injection to provide a reference for their clinical co-administration. Methods: In the pharmacological study, BALB/c mice with CT26 colorectal tumors were divided into four groups and treated with vehicle alone (0.9% saline), CPT-11 injection (100 mg/kg), KA injection (10 mL/kg), or a combination of CPT-11 and KA injection, respectively. The tumor volume of mice was monitored daily to evaluate the therapeutic effect. Daily body weight, survival rate, hematopoietic toxicity, immune organ indices, and gut toxicity were analyzed to study the adverse effects. Healthy Sprague-Dawley rats in the pharmacokinetic study were administered KA injection only (4 mL/kg), or a combination of CPT-11 injection (20 mg/kg) and KA injection, respectively. Six key components of KA injection (oxymatrine, matrine, ginsenoside Rb1, Rg1, Re, and astragaloside IV) in rat plasma samples collected within 24 h after administration were determined by LC-MS/MS. Results: The pharmacological study indicated that KA injection has the potential to enhance the anti-colorectal cancer efficacy of CPT-11 injection and alleviate the severe weight loss induced by CPT-11 injection in tumor-bearing mice. The pharmacokinetic study revealed that co-administration resulted in inhibition of oxymatrine metabolism in rats, evidenced by the significantly reduced Cmax and AUC0-t of its metabolite, matrine (p < 0.05), from 2.23 ± 0.24 to 1.38 ± 0.12 μg/mL and 8.29 ± 1.34 to 5.30 ± 0.79 μg h/mL, respectively. However, due to the similar efficacy of oxymatrine and matrine, this may not compromise the anti-cancer effect of this herb-drug pair. Discussion: This study clarified the pre-clinical pharmacology and pharmacokinetic benefits and risks of the CPT-11-KA combination and provided a reference for their clinical co-administration.
Collapse
Affiliation(s)
- Yanfei Chen
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
| | - Zhaoliang Hu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
| | - Jing Jiang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
| | - Chenxi Liu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
| | - Shuxiao Gao
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
| | - Min Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
| | - Taijun Hang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
| |
Collapse
|
14
|
Pan Q, Fan X, Xie L, Wu D, Liu R, Gao W, Luo K, He B, Pu Y. Nano-enabled colorectal cancer therapy. J Control Release 2023; 362:548-564. [PMID: 37683732 DOI: 10.1016/j.jconrel.2023.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Colorectal cancer (CRC), one of the most common and deadliest diseases worldwide, poses a great health threat and social burden. The clinical treatments of CRC encompassing surgery, chemotherapy, and radiotherapy are challenged with toxicity, therapy resistance, and recurrence. In the past two decades, targeted therapy and immunotherapy have greatly improved the therapeutic benefits of CRC patients but they still suffer from drug resistance and low response rates. Very recently, gut microbiota regulation has exhibited a great potential in preventing and treating CRC, as well as in modulating the efficacy and toxicity of chemotherapy and immunotherapy. In this review, we provide a cutting-edge summary of nanomedicine-based treatment in colorectal cancer, highlighting the recent progress of oral and systemic tumor-targeting and/or tumor-activatable drug delivery systems as well as novel therapeutic strategies against CRC, including nano-sensitizing immunotherapy, anti-inflammation, gut microbiota modulation therapy, etc. Finally, the recent endeavors to address therapy resistance, metastasis, and recurrence in CRC were discussed. We hope this review could offer insight into the design and development of nanomedicines for CRC and beyond.
Collapse
Affiliation(s)
- Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Li Xie
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Di Wu
- Meat Processing Key Laboratory of Sichuan Province, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Rong Liu
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China.
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
15
|
Hu S, Xia K, Huang X, Zhao Y, Zhang Q, Huang D, Xu W, Chen Z, Wang C, Zhang Z. Multifunctional CaCO 3@Cur@QTX125@HA nanoparticles for effectively inhibiting growth of colorectal cancer cells. J Nanobiotechnology 2023; 21:353. [PMID: 37773145 PMCID: PMC10543835 DOI: 10.1186/s12951-023-02104-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related deaths in humans, and effective treatments are still needed in clinical practice. Despite significant developments in anticancer drugs and inhibitors, their poor stability, water solubility, and cellular membrane permeability limit their therapeutic efficacy. To address these issues, multifunctional CaCO3 nanoparticles loaded with Curcumin (Cur) and protein deacetylase (HDAC) inhibitor QTX125, and coated with hyaluronic acid (HA) (CaCO3@Cur@QTX125@HA), were prepared through a one-step gas diffusion strategy. Dynamic light scattering (DLS), transmission electron microscopy (TEM), and scanning electron microscopy (SEM) showed that CaCO3@Cur@QTX125@HA nanoparticles have uniform spherical morphology and elemental distribution, with diameters around 450 nm and a Zeta potential of - 8.11 mV. The controlled release of Cur from the nanoparticles was observed over time periods of 48 h. Cellular uptake showed that CaCO3@Cur@QTX125@HA nanoparticles were efficiently taken up by cancer cells and significantly inhibited their growth. Importantly, CaCO3@Cur@QTX125@HA nanoparticles showed specific inhibitory effects on CRC cell growth. Encouragingly, CaCO3@Cur@QTX125@HA nanoparticles successfully internalized into CRC patient-derived organoid (PDO) models and induced apoptosis of tumor cells. The multifunctional CaCO3@Cur@QTX125@HA nanoparticles hold promise for the treatment of CRC.
Collapse
Affiliation(s)
- Shengyun Hu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kunkun Xia
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaobei Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ye Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qingqing Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dongdong Huang
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China
| | - Weiyi Xu
- Department of Dermatology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China.
| | - Chenfei Wang
- Department of Dermatology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Zhiyong Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
16
|
Noureddine A, Maestas-Olguin A, Tang L, Corman-Hijar JI, Olewine M, Krawchuck JA, Tsala Ebode J, Edeh C, Dang C, Negrete OA, Watt J, Howard T, Coker EN, Guo J, Brinker CJ. Future of Mesoporous Silica Nanoparticles in Nanomedicine: Protocol for Reproducible Synthesis, Characterization, Lipid Coating, and Loading of Therapeutics (Chemotherapeutic, Proteins, siRNA and mRNA). ACS NANO 2023; 17:16308-16325. [PMID: 37643407 DOI: 10.1021/acsnano.3c07621] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Owing to their uniform and tunable particle size, pore size, and shape, along with their modular surface chemistry and biocompatibility, mesoporous silica nanoparticles (MSNs) have found extensive applications as nanocarriers to deliver therapeutic, diagnostic and combined "theranostic" cargos to cells and tissues. Although thoroughly investigated, MSN have garnered FDA approval for only one MSN system via oral administration. One possible reason is that there is no recognized, reproducible, and widely adopted MSN synthetic protocol, meaning not all MSNs are created equal in the laboratory nor in the eyes of the FDA. This manuscript provides the sol-gel and MSN research communities a reproducible, fully characterized synthetic protocol to synthesize MSNs and corresponding lipid-coated MSN delivery vehicles with predetermined particle size, pore size, and drug loading and release characteristics. By carefully articulating the step-by-step synthetic procedures and highlighting critical points and troubleshooting, augmented with videos and schematics, this Article will help researchers entering this rapidly expanding field to yield reliable results.
Collapse
Affiliation(s)
- Achraf Noureddine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Angelea Maestas-Olguin
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Lien Tang
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, United States
| | - Jim I Corman-Hijar
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- Biomedical Engineering Department, Pontifical Catholic University of Peru, San Miguel 15088, Peru
| | - Marian Olewine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Jacob A Krawchuck
- Center for Integrated Nanotechnologies, Sandia National Laboratories, Albuquerque, New Mexico 87123, United States
| | - Johanna Tsala Ebode
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Chuzube Edeh
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Caleb Dang
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Oscar A Negrete
- Systems Biology Department, Sandia National Laboratories, Livermore, California 94550 United States
| | - John Watt
- Center for Integrated Nanotechnologies, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Tamara Howard
- Department of Cell Biology & Physiology, UNM School of Medicine, Albuquerque, New Mexico 87131, United States
| | - Eric N Coker
- Electronic, Optical, and Nano Materials, Sandia National Laboratories, Albuquerque, New Mexico 87185, United States
| | - Jimin Guo
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - C Jeffrey Brinker
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
17
|
Hicks MR, Liu X, Young CS, Saleh K, Ji Y, Jiang J, Emami MR, Mokhonova E, Spencer MJ, Meng H, Pyle AD. Nanoparticles systemically biodistribute to regenerating skeletal muscle in DMD. J Nanobiotechnology 2023; 21:303. [PMID: 37641124 PMCID: PMC10463982 DOI: 10.1186/s12951-023-01994-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 07/09/2023] [Indexed: 08/31/2023] Open
Abstract
Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle.
Collapse
Affiliation(s)
- Michael R Hicks
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Xiangsheng Liu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Courtney S Young
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- MyoGene Bio, San Diego, CA, USA
| | - Kholoud Saleh
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ying Ji
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jinhong Jiang
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Michael R Emami
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ekaterina Mokhonova
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Melissa J Spencer
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA.
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Huan Meng
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China.
| | - April D Pyle
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Song S, Sun D, Wang H, Wang J, Yan H, Zhao X, Fawcett JP, Xu X, Cai D, Gu J. Full-profile pharmacokinetics, anticancer activity and toxicity of an extended release trivalent PEGylated irinotecan prodrug. Acta Pharm Sin B 2023; 13:3444-3453. [PMID: 37655324 PMCID: PMC10466002 DOI: 10.1016/j.apsb.2023.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/22/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Irinotecan is an anticancer topoisomerase I inhibitor that acts as a prodrug of the active metabolite, SN-38. Unfortunately, the limited utility of irinotecan is attributed to its pH-dependent stability, short half-life and dose-limiting toxicity. To address this problem, a novel trivalent PEGylated prodrug (PEG-[Irinotecan]3) has been synthesized and its full-profile pharmacokinetics, antitumor activity and toxicity compared with those of irinotecan. The results show that after intravenous administration to rats, PEG-[Irinotecan]3 undergoes stepwise loss of irinotecan to form PEG-[Irinotecan]3‒x (x = 1,2) and PEG-[linker] during which time the released irinotecan undergoes conversion to SN-38. As compared with conventional irinotecan, PEG-[Irinotecan]3 displays extended release of irinotecan and efficient formation of SN-38 with significantly improved AUC and half-life. In a colorectal cancer-bearing model in nude mice, the tumor concentrations of irinotecan and SN-38 produced by PEG-[Irinotecan]3 were respectively 86.2 and 2293 times higher at 48 h than produced by irinotecan. In summary, PEG-[Irinotecan]3 displays superior pharmacokinetic characteristics and antitumor activity with lower toxicity than irinotecan. This supports the view that PEG-[Irinotecan]3 is a superior anticancer drug to irinotecan and it has entered the phase II trial stage.
Collapse
Affiliation(s)
- Shiwen Song
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Dong Sun
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Hong Wang
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- Center for Food and Drug Inspection of NMPA, Changchun 130012, China
| | | | - Huijing Yan
- JenKem Technology Co., Ltd., Tianjin 300450, China
| | - Xuan Zhao
- JenKem Technology Co., Ltd., Tianjin 300450, China
| | - John Paul Fawcett
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Xu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Deqi Cai
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
- Beijing Institute of Drug Metabolism, Beijing 102209, China
| |
Collapse
|
19
|
Freire N, Barbosa RDM, García-Villén F, Viseras C, Perioli L, Fialho R, Albuquerque E. Environmentally Friendly Strategies for Formulating Vegetable Oil-Based Nanoparticles for Anticancer Medicine. Pharmaceutics 2023; 15:1908. [PMID: 37514094 PMCID: PMC10386571 DOI: 10.3390/pharmaceutics15071908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
The development of green synthesized polymeric nanoparticles with anticancer studies has been an emerging field in academia and the pharmaceutical and chemical industries. Vegetable oils are potential substitutes for petroleum derivatives, as they present a clean and environmentally friendly alternative and are available in abundance at relatively low prices. Biomass-derived chemicals can be converted into monomers with a unique structure, generating materials with new properties for the synthesis of sustainable monomers and polymers. The production of bio-based polymeric nanoparticles is a promising application of green chemistry for biomedical uses. There is an increasing demand for biocompatible and biodegradable materials for specific applications in the biomedical area, such as cancer therapy. This is encouraging scientists to work on research toward designing polymers with enhanced properties and clean processes, containing oncology active pharmaceutical ingredients (APIs). The nanoencapsulation of these APIs in bio-based polymeric nanoparticles can control the release of the substances, increase bioavailability, reduce problems of volatility and degradation, reduce side effects, and increase treatment efficiency. This review discusses the use of green chemistry for bio-based nanoparticle production and its application in anticancer medicine. The use of castor oil for the production of renewable monomers and polymers is proposed as an ideal candidate for such applications, as well as more suitable methods for the production of bio-based nanoparticles and some oncology APIs available for anticancer application.
Collapse
Affiliation(s)
- Nathália Freire
- Graduate Program in Industrial Engineering, Polytechnic School, Federal University of Bahia, Salvador 40210-630, Brazil
| | - Raquel de Melo Barbosa
- Laboratory of Drug Development, Department of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil
| | - Fátima García-Villén
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Granada, Campus of Cartuja, 18071 Granada, Spain
| | - César Viseras
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Granada, Campus of Cartuja, 18071 Granada, Spain
- Andalusian Institute of Earth Sciences, CSIC-University of Granada, Av. de las Palmeras 4, Armilla, 18100 Granada, Spain
| | - Luana Perioli
- Department of Pharmaceutic Science, University of Perugia, 06123 Perugia, Italy
| | - Rosana Fialho
- Graduate Program in Industrial Engineering, Polytechnic School, Federal University of Bahia, Salvador 40210-630, Brazil
| | - Elaine Albuquerque
- Graduate Program in Industrial Engineering, Polytechnic School, Federal University of Bahia, Salvador 40210-630, Brazil
| |
Collapse
|
20
|
Liu S, Wang H, Shao X, Chen H, Chao S, Zhang Y, Gao Z, Yao Q, Zhang P. Advances in PD-1 signaling inhibition-based nano-delivery systems for tumor therapy. J Nanobiotechnology 2023; 21:207. [PMID: 37403095 PMCID: PMC10318732 DOI: 10.1186/s12951-023-01966-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
In recent years, cancer immunotherapy has emerged as an exciting cancer treatment. Immune checkpoint blockade brings new opportunities for more researchers and clinicians. Programmed cell death receptor-1 (PD-1) is a widely studied immune checkpoint, and PD-1 blockade therapy has shown promising results in a variety of tumors, including melanoma, non-small cell lung cancer and renal cell carcinoma, which greatly improves patient overall survival and becomes a promising tool for the eradication of metastatic or inoperable tumors. However, low responsiveness and immune-related adverse effects currently limit its clinical application. Overcoming these difficulties is a major challenge to improve PD-1 blockade therapies. Nanomaterials have unique properties that enable targeted drug delivery, combination therapy through multidrug co-delivery strategies, and controlled drug release through sensitive bonds construction. In recent years, combining nanomaterials with PD-1 blockade therapy to construct novel single-drug-based or combination therapy-based nano-delivery systems has become an effective mean to address the limitations of PD-1 blockade therapy. In this study, the application of nanomaterial carriers in individual delivery of PD-1 inhibitors, combined delivery of PD-1 inhibitors and other immunomodulators, chemotherapeutic drugs, photothermal reagents were reviewed, which provides effective references for designing new PD-1 blockade therapeutic strategies.
Collapse
Affiliation(s)
- Songlin Liu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Haiyang Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
- Qingdao University of Science and Technology, Qingdao, 266042, People's Republic of China
| | - Xinzhe Shao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Haonan Chen
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Shushu Chao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Yanyan Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Zhaoju Gao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Qingqiang Yao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Pingping Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China.
| |
Collapse
|
21
|
Kim J, Cho H, Lim DK, Joo MK, Kim K. Perspectives for Improving the Tumor Targeting of Nanomedicine via the EPR Effect in Clinical Tumors. Int J Mol Sci 2023; 24:10082. [PMID: 37373227 DOI: 10.3390/ijms241210082] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Over the past few decades, the enhanced permeability and retention (EPR) effect of nanomedicine has been a crucial phenomenon in targeted cancer therapy. Specifically, understanding the EPR effect has been a significant aspect of delivering anticancer agents efficiently to targeted tumors. Although the therapeutic effect has been demonstrated in experimental models using mouse xenografts, the clinical translation of the EPR effect of nanomedicine faces several challenges due to dense extracellular matrix (ECM), high interstitial fluid pressure (IFP) levels, and other factors that arise from tumor heterogeneity and complexity. Therefore, understanding the mechanism of the EPR effect of nanomedicine in clinics is essential to overcome the hurdles of the clinical translation of nanomedicine. This paper introduces the basic mechanism of the EPR effect of nanomedicine, the recently discussed challenges of the EPR effect of nanomedicine, and various strategies of recent nanomedicine to overcome the limitations expected from the patients' tumor microenvironments.
Collapse
Affiliation(s)
- Jinseong Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman's University, Seoul 03760, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman's University, Seoul 03760, Republic of Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Min Kyung Joo
- Noxpharm Co., Ltd., #518, 150, Bugahyeon-ro, Seodaemun-gu, Seoul 03759, Republic of Korea
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman's University, Seoul 03760, Republic of Korea
| |
Collapse
|
22
|
Dong S, Feng Z, Ma R, Zhang T, Jiang J, Li Y, Zhang Y, Li S, Liu X, Liu X, Meng H. Engineered Design of a Mesoporous Silica Nanoparticle-Based Nanocarrier for Efficient mRNA Delivery in Vivo. NANO LETTERS 2023; 23:2137-2147. [PMID: 36881967 DOI: 10.1021/acs.nanolett.2c04486] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
We have developed tailor-designed mesoporous silica nanoparticles (MSNPs) specifically for delivering mRNA. Our unique assembly protocol involves premixing mRNA with a cationic polymer and then electrostatically binding it to the MSNP surface. Since the key physicochemical parameters of MSNPs could influence the biological outcome, we also investigated the roles of size, porosity, surface topology, and aspect ratio on the mRNA delivery. These efforts allow us to identify the best-performing carrier, which was able to achieve efficient cellular uptake and intracellular escape while delivering a luciferase mRNA in mice. The optimized carrier remained stable and active for at least 7 days after being stored at 4 °C and was able to enable tissue-specific mRNA expression, particularly in the pancreas and mesentery after intraperitoneal injection. The optimized carrier was further manufactured in a larger batch size and found to be equally efficient in delivering mRNA in mice and rats, without any obvious toxicity.
Collapse
Affiliation(s)
- Shuwen Dong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhenhan Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Runpu Ma
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- College of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Tianyu Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jinhong Jiang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yibo Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yumo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
| | - Silu Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiao Liu
- Department of Gastroenterology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, National Center of Gerontology, Beijing 100730, China
| | - Xiangsheng Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Yin L, Li H, Shi L, Chen K, Pan H, Han W. Research advances in nanomedicine applied to the systemic treatment of colorectal cancer. Int J Cancer 2023; 152:807-821. [PMID: 35984398 DOI: 10.1002/ijc.34256] [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: 10/14/2021] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 01/06/2023]
Abstract
The systematic treatment of colorectal cancer (CRC) still has room for improvement. The efficacy of chemotherapy, that of anti-vascular therapy, and that of immunotherapy have been unsatisfactory. In recent years, nanomaterials have been used as carriers to improve the bioavailability of anticancer drugs. For the treatment of colorectal cancer, nanodrugs increase the possibility of more precise targeted delivery. However, the actual benefits may cover more aspects. Nanocarriers can produce synergistic effects with anticancer drugs, including the scavenging of reactive oxygen species and co-delivery of a variety of drugs. Currently, immunotherapy has very limited clinical applications in CRC. Modified nanocarriers can activate the immune microenvironment, which can be used for staging antigen recognition or the immune response. Cancer vaccines based on nanomaterials and modified immune checkpoint inhibitors have shown therapeutic potential in animal models. Considering the direct or indirect relationship between the intestinal microflora and CRC, a variety of nanodrugs that regulate microbial function have been explored as an anticancer strategy, and the special structure of microorganisms can also be used as a basis for improving the delivery of traditional nanoparticles (NPs). This review summarizes recent research performed on nanocarriers in in vivo and in vitro models and the synergistic anticancer effects of nanocarriers, focusing on the interaction between NPs and the body, resulting in enhanced efficacy and immune activation. Furthermore, this review describes the current trend of NPs used in the treatment of CRC.
Collapse
Affiliation(s)
- Luxi Yin
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haozhe Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Linlin Shi
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Tumor vasculature VS tumor cell targeting: Understanding the latest trends in using functional nanoparticles for cancer treatment. OPENNANO 2023. [DOI: 10.1016/j.onano.2023.100136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
|
25
|
Nguyen NT, Kim J, Le XT, Lee WT, Lee ES, Oh KT, Choi HG, Youn YS. Amplified Fenton-Based Oxidative Stress Utilizing Ultraviolet Upconversion Luminescence-Fueled Nanoreactors for Apoptosis-Strengthened Ferroptosis Anticancer Therapy. ACS NANO 2023; 17:382-401. [PMID: 36579941 DOI: 10.1021/acsnano.2c08706] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
As an emerging anticancer strategy, ferroptosis has recently been developed in combination with current therapeutic modalities to overcome the existing limitations of conventional therapies. Herein, an ultraviolet (UV) upconversion luminescence-fueled nanoreactor is explored to combine ferroptosis and apoptosis through the UV-catalyzed Fenton reaction of an iron supplement (ferric ammonium citrate) loaded in a mesoporous silica layer in addition to the support of a chemotherapeutic agent (cisplatin) attached on the functionalized silica surface for the treatment of triple negative breast cancer (TNBC). The nanoplatform can circumvent the low penetration depth typical of UV light by upconverting near-infrared irradiation and emitting UV photons that convert Fe3+ to Fe2+ to boost the generation of hydroxyl radicals (·OH), causing devastating lipid peroxidation. Apart from DNA damage-induced apoptosis, cisplatin can also catalyze Fenton-based therapy by its abundant production of hydrogen peroxide (H2O2). As a bioinspired lipid membrane, the folate receptor-targeted liposome as the coating layer offers high biocompatibility and colloidal stability for the upconversion nanoparticles, in addition to prevention of the premature release of encapsulated hydrophilic compounds, before driving the nanoformulation to the target tumor site. As a result, superior antitumor efficacy has been observed in a 4T1 tumor-bearing mouse model with negligible side effects, suggesting that such a nanoformulation could play a pivotal role in effective apoptosis-strengthened ferroptosis TNBC therapy.
Collapse
Affiliation(s)
- Nguyen Thi Nguyen
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Juho Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Xuan Thien Le
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Woo Tak Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Eun Seong Lee
- Department of Biotechnology and Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| |
Collapse
|
26
|
Shi H, Wan Y, Tian X, Wang L, Shan L, Zhang C, Wu MY, Feng S. Synergistically Enhancing Tumor Chemotherapy Using an Aggregation-Induced Emission Photosensitizer on Covalently Conjugated Molecularly Imprinted Polymer Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2022; 14:56585-56596. [PMID: 36513426 DOI: 10.1021/acsami.2c17731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Due to the polygenic and heterogeneous nature of the tumorigenesis process, traditional chemotherapy is far from desirable. Fabricating multifunctional nanoplatforms integrating photodynamic effect can synergistically enhance chemotherapy because they can make the cancer cells much sensitive to chemotherapeutics. However, how to assemble different units in nanoplatforms and minimize side effects caused by chemodrugs and photosensitizers (PSs) still needs to be explored. Herein, a nanoplatform CPP/PS-MIP@DOX is developed using a simultaneously covalently conjugated new aggregation-induced emission (AIE) PS and a cell-penetrating peptide (CPP) on the surface of silica-based molecularly imprinted polymer (MIP) nanoparticles, prepared with doxorubicin (DOX) as the template in the water system via a sol-gel technique. CPP/PS-MIP@DOX has good biocompatibility, high DOX-loading ability, promoted cellular uptake, and sustained and pH-sensitive drug release capability. Furthermore, it can efficiently penetrate into tumor tissue, accurately home to, and accumulate at the tumor site. As a result, a better efficacy with lower cytotoxicity is achieved with a smaller dosage of DOX by utilizing either the photodynamic effect or unique characteristics of the MIP. It is the first nanoplatform fabricated by chemically conjugating AIE PSs directly on the surface of the scaffold via the surface-decorated strategy and successfully applied in cancer therapy. This work provides an effective strategy by constructing AIE PS-based cancer nanomedicines with MIPs as scaffolds.
Collapse
Affiliation(s)
- Haizhu Shi
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yu Wan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiao Tian
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lijuan Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lianhai Shan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Chungu Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Ming-Yu Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
27
|
Ezati N, Abdouss M, Rouhani M, Kerr PG, Kowsari E. Novel serotonin decorated molecularly imprinted polymer nanoparticles based on biodegradable materials; A potential self-targeted delivery system for Irinotecan. REACT FUNCT POLYM 2022. [DOI: 10.1016/j.reactfunctpolym.2022.105437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
28
|
Panagi M, Mpekris F, Chen P, Voutouri C, Nakagawa Y, Martin JD, Hiroi T, Hashimoto H, Demetriou P, Pierides C, Samuel R, Stylianou A, Michael C, Fukushima S, Georgiou P, Papageorgis P, Papaphilippou PC, Koumas L, Costeas P, Ishii G, Kojima M, Kataoka K, Cabral H, Stylianopoulos T. Polymeric micelles effectively reprogram the tumor microenvironment to potentiate nano-immunotherapy in mouse breast cancer models. Nat Commun 2022; 13:7165. [PMID: 36418896 PMCID: PMC9684407 DOI: 10.1038/s41467-022-34744-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
Nano-immunotherapy improves breast cancer outcomes but not all patients respond and none are cured. To improve efficacy, research focuses on drugs that reprogram cancer-associated fibroblasts (CAFs) to improve therapeutic delivery and immunostimulation. These drugs, however, have a narrow therapeutic window and cause adverse effects. Developing strategies that increase CAF-reprogramming while limiting adverse effects is urgent. Here, taking advantage of the CAF-reprogramming capabilities of tranilast, we developed tranilast-loaded micelles. Strikingly, a 100-fold reduced dose of tranilast-micelles induces superior reprogramming compared to free drug owing to enhanced intratumoral accumulation and cancer-associated fibroblast uptake. Combination of tranilast-micelles and epirubicin-micelles or Doxil with immunotherapy increases T-cell infiltration, resulting in cures and immunological memory in mice bearing immunotherapy-resistant breast cancer. Furthermore, shear wave elastography (SWE) is able to monitor reduced tumor stiffness caused by tranilast-micelles and predict response to nano-immunotherapy. Micellar encapsulation is a promising strategy for TME-reprogramming and SWE is a potential biomarker of response.
Collapse
Affiliation(s)
- Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Yasuhiro Nakagawa
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - John D Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Tetsuro Hiroi
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwanoha, Kashiwa, Chiba, Japan
- Department of Integrated Biosciences, Laboratory of Cancer Biology, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwanoha, Kashiwa, Chiba, Japan
| | - Hiroko Hashimoto
- Division of Innovative Pathology and Laboratory Medicine, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwanoha, Kashiwa, Chiba, Japan
| | - Philippos Demetriou
- The Center for the Study of Hematological and other Malignancies, Nicosia, Cyprus
| | - Chryso Pierides
- The Center for the Study of Hematological and other Malignancies, Nicosia, Cyprus
| | - Rekha Samuel
- Center for Stem Cell Research (a unit of inStem Bengaluru), Christian Medical College Campus Bagayam, Vellore, Tamil Nadu, India
| | - Andreas Stylianou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
- Basic and Translational Cancer Research Center, School of Sciences, European University of Cyprus, Nicosia, Cyprus
| | - Christina Michael
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Shigeto Fukushima
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Paraskevi Georgiou
- Basic and Translational Cancer Research Center, School of Sciences, European University of Cyprus, Nicosia, Cyprus
| | - Panagiotis Papageorgis
- Basic and Translational Cancer Research Center, School of Sciences, European University of Cyprus, Nicosia, Cyprus
| | - Petri Ch Papaphilippou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Laura Koumas
- The Center for the Study of Hematological and other Malignancies, Nicosia, Cyprus
- Karaiskakio Foundation, Nicosia, Cyprus
| | - Paul Costeas
- The Center for the Study of Hematological and other Malignancies, Nicosia, Cyprus
- Karaiskakio Foundation, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
| | - Genichiro Ishii
- Division of Innovative Pathology and Laboratory Medicine, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwanoha, Kashiwa, Chiba, Japan
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwanoha, Kashiwa, Chiba, Japan
| | - Motohiro Kojima
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwanoha, Kashiwa, Chiba, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
- Institute for Future Initiatives, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan.
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
29
|
Saad MA, Hasan T. Spotlight on Photoactivatable Liposomes beyond Drug Delivery: An Enabler of Multitargeting of Molecular Pathways. Bioconjug Chem 2022; 33:2041-2064. [PMID: 36197738 DOI: 10.1021/acs.bioconjchem.2c00376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potential of photoactivating certain molecules, photosensitizers (PS), resulting in photochemical processes, has long been realized in the form of photodynamic therapy (PDT) for the management of several cancerous and noncancerous pathologies. With an improved understanding of the photoactivation process and its broader implications, efforts are being made to exploit the various facets of photoactivation, PDT, and the associated phenomenon of photodynamic priming in enhancing treatment outcomes, specifically in cancer therapeutics. The parallel emergence of nanomedicine, specifically liposome-based nanoformulations, and the convergence of the two fields of liposome-based drug delivery and PDT have led to the development of unique hybrid systems, which combine the exciting features of liposomes with adequate complementation through the photoactivation process. While initially liposomes carrying photosensitizers (PSs) were developed for enhancing the pharmacokinetics and the general applicability of PSs, more recently, PS-loaded liposomes, apart from their utility in PDT, have found several applications including enhanced targeting of drugs, coloading multiple therapeutic agents to enhance synergistic effects, imaging, priming, triggering drug release, and facilitating the escape of therapeutic agents from the endolysosomal complex. This review discusses the design strategies, potential, and unique attributes of these hybrid systems, with not only photoactivation as an attribute but also the ability to encapsulate multiple agents for imaging, biomodulation, priming, and therapy referred to as photoactivatable multiagent/inhibitor liposomes (PMILS) and their targeted versions─targeted PMILS (TPMILS). While liposomes have formed their own niche in nanotechnology and nanomedicine with several clinically approved formulations, we try to highlight how using PS-loaded liposomes could address some of the limitations and concerns usually associated with liposomes to overcome them and enhance their preclinical and clinical utility in the future.
Collapse
Affiliation(s)
- Mohammad A Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States.,Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
30
|
Buyana B, Naki T, Alven S, Aderibigbe BA. Nanoparticles Loaded with Platinum Drugs for Colorectal Cancer Therapy. Int J Mol Sci 2022; 23:11261. [PMID: 36232561 PMCID: PMC9569963 DOI: 10.3390/ijms231911261] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is a common cancer in both men and women. Numerous studies on the therapeutic effectiveness of nanoparticles against colorectal cancer have been reported. Platinum treatments as well as other medications comprising of nanoparticles have been utilized. Drug resistance restricts the use of platinum medicines, despite their considerable efficacy against a variety of cancers. This review reports clinically licensed platinum medicines (cisplatin, carboplatin, and oxaliplatin) combined with various nanoparticles that have been evaluated for their therapeutic efficacy in the treatment of colorectal cancer, including their mechanism of action, resistance, and limitations.
Collapse
Affiliation(s)
| | | | | | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice 5700, Eastern Cape Province, South Africa
| |
Collapse
|
31
|
Miclea LC, Mihailescu M, Tarba N, Brezoiu AM, Sandu AM, Mitran RA, Berger D, Matei C, Moisescu MG, Savopol T. Evaluation of intracellular distribution of folate functionalized silica nanoparticles using fluorescence and hyperspectral enhanced dark field microscopy. NANOSCALE 2022; 14:12744-12756. [PMID: 36000453 DOI: 10.1039/d2nr01821g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Using nanoparticles as carriers for drug delivery systems has become a widely applied strategy in therapeutics and diagnostics. However, the pattern of their intracellular distribution is yet to be clarified. Here we present an in vitro study on the incorporation of mesoporous silica nanoparticles conjugated with folate and loaded with a cytotoxic drug, Irinotecan. The nanoparticles count and distribution within the cell frame were evaluated by means of enhanced dark field microscopy combined with hyperspectral imagery and 3D reconstructions from double-labeled fluorescent samples. An original post-processing procedure was developed to emphasize the nanoparticles' localization in 3D reconstruction of cellular compartments. By these means, it has been shown that the conjugation of mesoporous silica nanoparticles with folate increases the efficiency of nanoparticles entering the cell and their preferential localization in the close vicinity of the nucleus. As revealed by metabolic viability assays, the nanoparticles functionalized with folate enhance the cytotoxic efficiency of Irinotecan.
Collapse
Affiliation(s)
- Luminita Claudia Miclea
- Biophysics and Cellular Biotechnology Department, Excellence Center for Research in Biophysics and Cellular Biotechnology, Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Bucharest, 050474, Romania.
| | - Mona Mihailescu
- Digital Holography Imaging and Processing Laboratory, Fundamental Sciences Applied in Engineering Research Center, Faculty of Applied Sciences, University "Politehnica" of Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania.
| | - Nicolae Tarba
- Physics Department, Faculty of Applied Sciences, Doctoral School of Automatic Control and Computers, University "Politehnica" of Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
| | - Ana-Maria Brezoiu
- Department of Inorganic Chemistry, Physical-Chemistry & Electrochemistry, Faculty of Chemical Engineering and Biotechnologies, University "Politehnica" of Bucharest, 1-7 Polizu st., 11061, Bucharest, Romania
| | - Ana Maria Sandu
- CAMPUS Research Center, University "Politehnica" of Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
| | - Raul-Augustin Mitran
- "Ilie Murgulescu" Institute of Physical-Chemistry, Romanian Academy, 202 Splaiul Indepedenţei, Bucharest, 060021, Romania
| | - Daniela Berger
- Department of Inorganic Chemistry, Physical-Chemistry & Electrochemistry, Faculty of Chemical Engineering and Biotechnologies, University "Politehnica" of Bucharest, 1-7 Polizu st., 11061, Bucharest, Romania
| | - Cristian Matei
- Department of Inorganic Chemistry, Physical-Chemistry & Electrochemistry, Faculty of Chemical Engineering and Biotechnologies, University "Politehnica" of Bucharest, 1-7 Polizu st., 11061, Bucharest, Romania
| | - Mihaela Georgeta Moisescu
- Biophysics and Cellular Biotechnology Department, Excellence Center for Research in Biophysics and Cellular Biotechnology, Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Bucharest, 050474, Romania.
| | - Tudor Savopol
- Biophysics and Cellular Biotechnology Department, Excellence Center for Research in Biophysics and Cellular Biotechnology, Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Bucharest, 050474, Romania.
| |
Collapse
|
32
|
Osouli-Bostanabad K, Puliga S, Serrano DR, Bucchi A, Halbert G, Lalatsa A. Microfluidic Manufacture of Lipid-Based Nanomedicines. Pharmaceutics 2022; 14:pharmaceutics14091940. [PMID: 36145688 PMCID: PMC9506151 DOI: 10.3390/pharmaceutics14091940] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Nanoparticulate technologies have revolutionized drug delivery allowing for passive and active targeting, altered biodistribution, controlled drug release (temporospatial or triggered), enhanced stability, improved solubilization capacity, and a reduction in dose and adverse effects. However, their manufacture remains immature, and challenges exist on an industrial scale due to high batch-to-batch variability hindering their clinical translation. Lipid-based nanomedicines remain the most widely approved nanomedicines, and their current manufacturing methods remain discontinuous and face several problems such as high batch-to-batch variability affecting the critical quality attributes (CQAs) of the product, laborious multistep processes, need for an expert workforce, and not being easily amenable to industrial scale-up involving typically a complex process control. Several techniques have emerged in recent years for nanomedicine manufacture, but a paradigm shift occurred when microfluidic strategies able to mix fluids in channels with dimensions of tens of micrometers and small volumes of liquid reagents in a highly controlled manner to form nanoparticles with tunable and reproducible structure were employed. In this review, we summarize the recent advancements in the manufacturing of lipid-based nanomedicines using microfluidics with particular emphasis on the parameters that govern the control of CQAs of final nanomedicines. The impact of microfluidic environments on formation dynamics of nanomaterials, and the application of microdevices as platforms for nanomaterial screening are also discussed.
Collapse
Affiliation(s)
- Karim Osouli-Bostanabad
- Biomaterials, Bio-Engineering and Nanomedicine (BioN) Lab, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, UK
- School of Pharmacy and Biomedical Sciences, Robertson Wing, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
| | - Sara Puliga
- Biomaterials, Bio-Engineering and Nanomedicine (BioN) Lab, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, UK
| | - Dolores R. Serrano
- Pharmaceutics and Food Technology Department, School of Pharmacy, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Facultad de Farmacia, Instituto Universitario de Farmacia Industrial, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Correspondence: (D.R.S.); (A.L.); Tel.: +44-141-548-2675 (A.L.)
| | - Andrea Bucchi
- School of Mechanical and Design Engineering, Faculty of Technology, University of Portsmouth, Portsmouth PO1 3DJ, UK
| | - Gavin Halbert
- CRUK Formulation Unit, School of Pharmacy and Biomedical Sciences, Robertson Wing, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
| | - Aikaterini Lalatsa
- Biomaterials, Bio-Engineering and Nanomedicine (BioN) Lab, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, UK
- School of Pharmacy and Biomedical Sciences, Robertson Wing, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
- CRUK Formulation Unit, School of Pharmacy and Biomedical Sciences, Robertson Wing, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
- Correspondence: (D.R.S.); (A.L.); Tel.: +44-141-548-2675 (A.L.)
| |
Collapse
|
33
|
Ghosh S, He X, Huang WC, Lovell JF. Immune checkpoint blockade enhances chemophototherapy in a syngeneic pancreatic tumor model. APL Bioeng 2022; 6:036105. [PMID: 36164594 PMCID: PMC9509203 DOI: 10.1063/5.0099811] [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: 05/19/2022] [Accepted: 08/29/2022] [Indexed: 11/14/2022] Open
Abstract
Pancreatic cancer (PaCa) suffers from poor treatment options for locally advanced cases. Chemophototherapy (CPT) is an emerging anti-tumor modality, and porphyrin–phospholipid liposomes have been shown to be versatile drug carriers for CPT in preclinical rodent models. Here we show that in the syngeneic subcutaneous KPC PaCa tumor model, exhausted CD8+ T cells are localized in the tumor, and that CPT is enhanced in combination with immune checkpoint blockade (ICB). Addition of ICB using anti-programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies resulted in ablation of medium-sized, established KPC tumors (∼200 mm3) without recurrence for over 100 days. Mice rejected subsequent tumor re-challenge. Flow cytometry and tumor slice analysis following injection of a fluorescently labeled anti-PD-1 antibody showed that CPT improved antibody delivery to the tumor microenvironment. Treatment of large established tumors (∼400 mm3) using with CPT and ICB induced appreciable tumor regression and delay in regrowth. Taken together, these data demonstrate the utility of combining CPT with immunotherapies.
Collapse
Affiliation(s)
- Sanjana Ghosh
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Xuedan He
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| |
Collapse
|
34
|
Luo L, Wang X, Liao YP, Chang CH, Nel AE. Nanocarrier Co-formulation for Delivery of a TLR7 Agonist plus an Immunogenic Cell Death Stimulus Triggers Effective Pancreatic Cancer Chemo-immunotherapy. ACS NANO 2022; 16:13168-13182. [PMID: 35920660 PMCID: PMC10117630 DOI: 10.1021/acsnano.2c06300] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Although toll-like receptor (TLR) agonists hold great promise as immune modulators for reprogramming the suppressive immune landscape in pancreatic ductal adenocarcinoma (PDAC), their use is limited by poor pharmacokinetics (PK) and off-target systemic inflammatory effects. To overcome these challenges as well as to attain drug synergy, we developed a lipid bilayer (LB)-coated mesoporous silica nanoparticle (silicasome) platform for co-delivery of the TLR7/8 agonist 3M-052 with the immunogenic chemotherapeutic agent irinotecan. This was accomplished by incorporating the C18 lipid tail of 3M-052 in the coated LB, also useful for irinotecan remote loading in the porous interior. Not only did the co-formulated carrier improve PK, but it strengthened the irinotecan-induced immunogenic cell death response by 3M-052-mediated dendritic cell activation at the tumor site as well as participating lymph nodes. The accompanying increase in CD8+ T-cell infiltration along with a reduced number of regulatory T-cells was associated with tumor shrinkage and metastasis disappearance in subcutaneous and orthotopic KRAS-mediated pancreatic carcinoma tumor models. Moreover, this therapeutic outcome was accomplished without drug or nanocarrier toxicity. All considered, dual-delivery strategies that combine chemo-immunotherapy with co-formulated TLR agonists or other lipid-soluble immune modulators predict successful intervention in heterogeneous PDAC immune landscapes.
Collapse
Affiliation(s)
- Lijia Luo
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiang Wang
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Chong Hyun Chang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Andre E. Nel
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
35
|
Wang Z, Li W, Park J, Gonzalez KM, Scott AJ, Lu J. Camptothesome elicits immunogenic cell death to boost colorectal cancer immune checkpoint blockade. J Control Release 2022; 349:929-939. [PMID: 35926754 DOI: 10.1016/j.jconrel.2022.07.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/22/2022]
Abstract
Camptothesome is an innovative nanovesicle therapeutic comprising the sphingomyelin-derived camptothecin (CPT) lipid bilayer. In this work, we deciphered that Camptothesome was taken up by colorectal cancer (CRC) cells through primarily the clathrin-mediated endocytotic pathway and displayed the potential of eliciting robust immunogenic cancer cell death (ICD) via upregulating calreticulin, high mobility group box 1 protein (HMGB-1), and adenosine triphosphate (ATP), three hallmarks involved in the induction of ICD. In addition, use of dying MC38 tumor cells treated with Camptothesome as vaccine prevented tumor growth in 60% mice that received subsequent injection of live MC38 cells on the contralateral flank, validating Camptothesome was a legitimate ICD inducer in vivo. Camptothesome markedly reduced the acute bone marrow toxicity and gastrointestinal mucositis associated with free CPT and beat free CPT and Onivyde on anti-CRC efficacy and immune responses in a partially interferon gamma (IFN-γ)-dependent manner. Furthermore, Camptothesome enhanced the efficacy of immune checkpoint inhibitors to shrink late-stage orthotopic MC38 CRC tumors with diminished tumor metastasis and markedly prolonged mice survival.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Jonghan Park
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Aaron James Scott
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ 85721, United States; Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, United States; Southwest Environmental Health Sciences Center, The University of Arizona, Tucson 85721, United States.
| |
Collapse
|
36
|
Younis NK, Roumieh R, Bassil EP, Ghoubaira JA, Kobeissy F, Eid AH. Nanoparticles: attractive tools to treat colorectal cancer. Semin Cancer Biol 2022; 86:1-13. [DOI: 10.1016/j.semcancer.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 10/31/2022]
|
37
|
li B, Zhang X, Wu Z, Chu T, Yang Z, Xu S, Wu S, Qie Y, Lu Z, Qi F, Hu M, Zhao G, Wei J, Zhao Y, Nie G, Meng H, Liu R, Li S. Reducing Postoperative Recurrence of Early-Stage Hepatocellular Carcinoma by a Wound-Targeted Nanodrug. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200477. [PMID: 35524631 PMCID: PMC9284141 DOI: 10.1002/advs.202200477] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/18/2022] [Indexed: 05/06/2023]
Abstract
New strategies to decrease risk of relapse after surgery are needed for improving 5-year survival rate of hepatocellular carcinoma (HCC). To address this need, a wound-targeted nanodrug is developed, that contains an immune checkpoint inhibitor (anti-PD-L1)and an angiogenesis inhibitor (sorafenib)). These nanoparticles consist of highly biocompatible mesoporous silica (MSNP) that is surface-coated with platelet membrane (PM) to achieve surgical site targeting in a self-amplified accumulation manner. Sorafenib is introduced into the MSNP pores while covalently attaching anti-PD-L1 antibody on the PM surface. The resulting nano-formulation, abbreviated as a-PM-S-MSNP, can effectively target the surgical margin when intraperitoneally (IP) administered into an immune competent murine orthotopic HCC model. Multiple administrations of a-PM-S-MSNP generate potent anti-HCC effect and significantly prolong overall mice survival. Immunophenotyping and immunochemistry staining reveal the signatures of favorable anti-HCC immunity and anti-angiogenesis effect at tumor sites. More importantly, microscopic inspection of a-PM-S-MSNP treated mice shows that 2 out 6 are histologically tumor-free, which is in sharp contrast to the control mice where tumor foci can be easily identified. The data suggest that a-PM-S-MSNP can efficiently inhibit post-surgical HCC relapse without obvious side effects and holds considerable promise for clinical translation as a novel nanodrug.
Collapse
Affiliation(s)
- Bozhao li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- College of Pharmaceutical ScienceJilin UniversityChangchun130021China
| | - Xiuping Zhang
- Faculty of Hepato‐Biliary‐Pancreatic SurgeryChinese People's Liberation Army (PLA) General HospitalInstitute of Hepatobiliary Surgery of Chinese PLAKey Laboratory of Digital Hepatobiliary SurgeryPLABeijing100853China
| | - Zhouliang Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Tianjiao Chu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- College of Pharmaceutical ScienceJilin UniversityChangchun130021China
| | - Zhenlin Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Shuai Xu
- Faculty of Hepato‐Biliary‐Pancreatic SurgeryChinese People's Liberation Army (PLA) General HospitalInstitute of Hepatobiliary Surgery of Chinese PLAKey Laboratory of Digital Hepatobiliary SurgeryPLABeijing100853China
| | - Suying Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yunkai Qie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Zefang Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Feilong Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Minggen Hu
- Faculty of Hepato‐Biliary‐Pancreatic SurgeryChinese People's Liberation Army (PLA) General HospitalInstitute of Hepatobiliary Surgery of Chinese PLAKey Laboratory of Digital Hepatobiliary SurgeryPLABeijing100853China
| | - Guodong Zhao
- Faculty of Hepato‐Biliary‐Pancreatic SurgeryChinese People's Liberation Army (PLA) General HospitalInstitute of Hepatobiliary Surgery of Chinese PLAKey Laboratory of Digital Hepatobiliary SurgeryPLABeijing100853China
| | - Jingyan Wei
- College of Pharmaceutical ScienceJilin UniversityChangchun130021China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049China
- GBA Research Innovation Institute for NanotechnologyGuangzhou510530China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049China
- GBA Research Innovation Institute for NanotechnologyGuangzhou510530China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Rong Liu
- Faculty of Hepato‐Biliary‐Pancreatic SurgeryChinese People's Liberation Army (PLA) General HospitalInstitute of Hepatobiliary Surgery of Chinese PLAKey Laboratory of Digital Hepatobiliary SurgeryPLABeijing100853China
| | - Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049China
- GBA Research Innovation Institute for NanotechnologyGuangzhou510530China
| |
Collapse
|
38
|
Irinotecan-loaded ROS-responsive liposomes containing thioether phosphatidylcholine for improving anticancer activity. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103321] [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]
|
39
|
Nel AE, Mei KC, Liao YP, Lu X. Multifunctional Lipid Bilayer Nanocarriers for Cancer Immunotherapy in Heterogeneous Tumor Microenvironments, Combining Immunogenic Cell Death Stimuli with Immune Modulatory Drugs. ACS NANO 2022; 16:5184-5232. [PMID: 35348320 PMCID: PMC9519818 DOI: 10.1021/acsnano.2c01252] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In addition to the contribution of cancer cells, the solid tumor microenvironment (TME) has a critical role in determining tumor expansion, antitumor immunity, and the response to immunotherapy. Understanding the details of the complex interplay between cancer cells and components of the TME provides an unprecedented opportunity to explore combination therapy for intervening in the immune landscape to improve immunotherapy outcome. One approach is the introduction of multifunctional nanocarriers, capable of delivering drug combinations that provide immunogenic stimuli for improvement of tumor antigen presentation, contemporaneous with the delivery of coformulated drug or synthetic molecules that provide immune danger signals or interfere in immune-escape, immune-suppressive, and T-cell exclusion pathways. This forward-looking review will discuss the use of lipid-bilayer-encapsulated liposomes and mesoporous silica nanoparticles for combination immunotherapy of the heterogeneous immune landscapes in pancreatic ductal adenocarcinoma and triple-negative breast cancer. We describe how the combination of remote drug loading and lipid bilayer encapsulation is used for the synthesis of synergistic drug combinations that induce immunogenic cell death, interfere in the PD-1/PD-L1 axis, inhibit the indoleamine-pyrrole 2,3-dioxygenase (IDO-1) immune metabolic pathway, restore spatial access to activated T-cells to the cancer site, or reduce the impact of immunosuppressive stromal components. We show how an integration of current knowledge and future discovery can be used for a rational approach to nanoenabled cancer immunotherapy.
Collapse
Affiliation(s)
- André E. Nel
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, United States
| | - Kuo-Ching Mei
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiangsheng Lu
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
40
|
Kankala RK, Han YH, Xia HY, Wang SB, Chen AZ. Nanoarchitectured prototypes of mesoporous silica nanoparticles for innovative biomedical applications. J Nanobiotechnology 2022; 20:126. [PMID: 35279150 PMCID: PMC8917689 DOI: 10.1186/s12951-022-01315-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
Despite exceptional morphological and physicochemical attributes, mesoporous silica nanoparticles (MSNs) are often employed as carriers or vectors. Moreover, these conventional MSNs often suffer from various limitations in biomedicine, such as reduced drug encapsulation efficacy, deprived compatibility, and poor degradability, resulting in poor therapeutic outcomes. To address these limitations, several modifications have been corroborated to fabricating hierarchically-engineered MSNs in terms of tuning the pore sizes, modifying the surfaces, and engineering of siliceous networks. Interestingly, the further advancements of engineered MSNs lead to the generation of highly complex and nature-mimicking structures, such as Janus-type, multi-podal, and flower-like architectures, as well as streamlined tadpole-like nanomotors. In this review, we present explicit discussions relevant to these advanced hierarchical architectures in different fields of biomedicine, including drug delivery, bioimaging, tissue engineering, and miscellaneous applications, such as photoluminescence, artificial enzymes, peptide enrichment, DNA detection, and biosensing, among others. Initially, we give a brief overview of diverse, innovative stimuli-responsive (pH, light, ultrasound, and thermos)- and targeted drug delivery strategies, along with discussions on recent advancements in cancer immune therapy and applicability of advanced MSNs in other ailments related to cardiac, vascular, and nervous systems, as well as diabetes. Then, we provide initiatives taken so far in clinical translation of various silica-based materials and their scope towards clinical translation. Finally, we summarize the review with interesting perspectives on lessons learned in exploring the biomedical applications of advanced MSNs and further requirements to be explored.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China.
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People's Republic of China.
| | - Ya-Hui Han
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
| | - Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People's Republic of China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People's Republic of China
| |
Collapse
|
41
|
Slapak EJ, el Mandili M, Bijlsma MF, Spek CA. Mesoporous Silica Nanoparticle-Based Drug Delivery Systems for the Treatment of Pancreatic Cancer: A Systematic Literature Overview. Pharmaceutics 2022; 14:390. [PMID: 35214121 PMCID: PMC8876630 DOI: 10.3390/pharmaceutics14020390] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 12/23/2022] Open
Abstract
Pancreatic cancer is a devastating disease with the worst outcome of any human cancer. Despite significant improvements in cancer treatment in general, little progress has been made in pancreatic cancer (PDAC), resulting in an overall 5-year survival rate of less than 10%. This dismal prognosis can be attributed to the limited clinical efficacy of systemic chemotherapy due to its high toxicity and consequent dose reductions. Targeted delivery of chemotherapeutic drugs to PDAC cells without affecting healthy non-tumor cells will largely reduce collateral toxicity leading to reduced morbidity and an increased number of PDAC patients eligible for chemotherapy treatment. To achieve targeted delivery in PDAC, several strategies have been explored over the last years, and especially the use of mesoporous silica nanoparticles (MSNs) seem an attractive approach. MSNs show high biocompatibility, are relatively easy to surface modify, and the porous structure of MSNs enables high drug-loading capacity. In the current systematic review, we explore the suitability of MSN-based targeted therapies in the setting of PDAC. We provide an extensive overview of MSN-formulations employed in preclinical PDAC models and conclude that MSN-based tumor-targeting strategies may indeed hold therapeutic potential for PDAC, although true clinical translation has lagged behind.
Collapse
Affiliation(s)
- Etienne J. Slapak
- Center of Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.e.M.); (C.A.S.)
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
- Oncode Institute, 1105 AZ Amsterdam, The Netherlands
| | - Mouad el Mandili
- Center of Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.e.M.); (C.A.S.)
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
| | - Maarten F. Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
- Oncode Institute, 1105 AZ Amsterdam, The Netherlands
| | - C. Arnold Spek
- Center of Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.e.M.); (C.A.S.)
| |
Collapse
|
42
|
Turchin I, Bano S, Kirillin M, Orlova A, Perekatova V, Plekhanov V, Sergeeva E, Kurakina D, Khilov A, Kurnikov A, Subochev P, Shirmanova M, Komarova A, Yuzhakova D, Gavrina A, Mallidi S, Hasan T. Combined Fluorescence and Optoacoustic Imaging for Monitoring Treatments against CT26 Tumors with Photoactivatable Liposomes. Cancers (Basel) 2021; 14:197. [PMID: 35008362 PMCID: PMC8750546 DOI: 10.3390/cancers14010197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
The newly developed multimodal imaging system combining raster-scan optoacoustic (OA) microscopy and fluorescence (FL) wide-field imaging was used for characterizing the tumor vascular structure with 38/50 μm axial/transverse resolution and assessment of photosensitizer fluorescence kinetics during treatment with novel theranostic agents. A multifunctional photoactivatable multi-inhibitor liposomal (PMILs) nano platform was engineered here, containing a clinically approved photosensitizer, Benzoporphyrin derivative (BPD) in the bilayer, and topoisomerase I inhibitor, Irinotecan (IRI) in its inner core, for a synergetic therapeutic impact. The optimized PMIL was anionic, with the hydrodynamic diameter of 131.6 ± 2.1 nm and polydispersity index (PDI) of 0.05 ± 0.01, and the zeta potential between -14.9 ± 1.04 to -16.9 ± 0.92 mV. In the in vivo studies on BALB/c mice with CT26 tumors were performed to evaluate PMILs' therapeutic efficacy. PMILs demonstrated the best inhibitory effect of 97% on tumor growth compared to the treatment with BPD-PC containing liposomes (PALs), 81%, or IRI containing liposomes (L-[IRI]) alone, 50%. This confirms the release of IRI within the tumor cells upon PMILs triggering by NIR light, which is additionally illustrated by FL monitoring demonstrating enhancement of drug accumulation in tumor initiated by PDT in 24 h after the treatment. OA monitoring revealed the largest alterations of the tumor vascular structure in the PMILs treated mice as compared to BPD-PC or IRI treated mice. The results were further corroborated with histological data that also showed a 5-fold higher percentage of hemorrhages in PMIL treated mice compared to the control groups. Overall, these results suggest that multifunctional PMILs simultaneously delivering PDT and chemotherapy agents along with OA and FL multi-modal imaging offers an efficient and personalized image-guided platform to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Ilya Turchin
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (S.B.); (S.M.); (T.H.)
| | - Mikhail Kirillin
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Anna Orlova
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Valeriya Perekatova
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Vladimir Plekhanov
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Ekaterina Sergeeva
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Daria Kurakina
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Aleksandr Khilov
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Alexey Kurnikov
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Pavel Subochev
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Marina Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Anastasiya Komarova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Diana Yuzhakova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Alena Gavrina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (S.B.); (S.M.); (T.H.)
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (S.B.); (S.M.); (T.H.)
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
43
|
Efficient nano-enabled therapy for gastrointestinal cancer using silicasome delivery technology. Sci China Chem 2021. [DOI: 10.1007/s11426-021-1126-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
44
|
Zhang G, Wang N, Sun H, Fu X, Zhai S, Cui J. Self-adjuvanting photosensitizer nanoparticles for combination photodynamic immunotherapy. Biomater Sci 2021; 9:6940-6949. [PMID: 34528658 DOI: 10.1039/d1bm01139a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Combination cancer immunotherapy that synergizes the advantages of multiple therapeutic agents has shown great potential in tumor treatment. Herein, we report the one-step assembly of therapeutic nanoparticles (NPs) to co-deliver photosensitizers and adjuvants for combination photodynamic therapy (PDT) and immunotherapy. The NPs are obtained via self-assembly of chlorin e6 (Ce6) and imidazoquinoline-based TLR7 agonists (IMDQ), which results in a high loading efficacy of 72.2% and 27.8% for Ce6 and IMDQ, respectively. Upon laser irradiation, the resulting NPs could not only effectively induce photodynamic immunogenic cancer cell death, but also elicit robust antitumor immunity, leading to significant inhibition of both primary and distant tumors in a bilateral tumor model. This study demonstrates the potential of self-assembled NPs in co-delivering multiple therapeutics for potential immunotherapy to enhance the antitumor efficacy.
Collapse
Affiliation(s)
- Guiqiang Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Ning Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Haifeng Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Xiao Fu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China. .,State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.,Advanced Medical Research Institute, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
45
|
Živojević K, Mladenović M, Djisalov M, Mundzic M, Ruiz-Hernandez E, Gadjanski I, Knežević NŽ. Advanced mesoporous silica nanocarriers in cancer theranostics and gene editing applications. J Control Release 2021; 337:193-211. [PMID: 34293320 DOI: 10.1016/j.jconrel.2021.07.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022]
Abstract
Targeted nanomaterials for cancer theranostics have been the subject of an expanding volume of research studies in recent years. Mesoporous silica nanoparticles (MSNs) are particularly attractive for such applications due to possibilities to synthesize nanoparticles (NPs) of different morphologies, pore diameters and pore arrangements, large surface areas and various options for surface functionalization. Functionalization of MSNs with different organic and inorganic molecules, polymers, surface-attachment of other NPs, loading and entrapping cargo molecules with on-desire release capabilities, lead to seemingly endless prospects for designing advanced nanoconstructs exerting multiple functions, such as simultaneous cancer-targeting, imaging and therapy. Describing composition and multifunctional capabilities of these advanced nanoassemblies for targeted therapy (passive, ligand-functionalized MSNs, stimuli-responsive therapy), including one or more modalities for imaging of tumors, is the subject of this review article, along with an overview of developments within a novel and attractive research trend, comprising the use of MSNs for CRISPR/Cas9 systems delivery and gene editing in cancer. Such advanced nanconstructs exhibit high potential for applications in image-guided therapies and the development of personalized cancer treatment.
Collapse
Affiliation(s)
- Kristina Živojević
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Minja Mladenović
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Mila Djisalov
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Mirjana Mundzic
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | | | - Ivana Gadjanski
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Nikola Ž Knežević
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia.
| |
Collapse
|
46
|
Liu X, Meng H. Consideration for the scale‐up manufacture of nanotherapeutics—A critical step for technology transfer. VIEW 2021. [DOI: 10.1002/viw.20200190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Xiangsheng Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beijing P. R. China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences Hangzhou Zhejiang P. R. China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beijing P. R. China
| |
Collapse
|
47
|
Ying K, Bai B, Gao X, Xu Y, Wang H, Xie B. Orally Administrable Therapeutic Nanoparticles for the Treatment of Colorectal Cancer. Front Bioeng Biotechnol 2021; 9:670124. [PMID: 34307319 PMCID: PMC8293278 DOI: 10.3389/fbioe.2021.670124] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/14/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common and lethal human malignancies worldwide; however, the therapeutic outcomes in the clinic still are unsatisfactory due to the lack of effective and safe therapeutic regimens. Orally administrable and CRC-targetable drug delivery is an attractive approach for CRC therapy as it improves the efficacy by local drug delivery and reduces systemic toxicity. Currently, chemotherapy remains the mainstay modality for CRC therapy; however, most of chemo drugs have low water solubility and are unstable in the gastrointestinal tract (GIT), poor intestinal permeability, and are susceptible to P-glycoprotein (P-gp) efflux, resulting in limited therapeutic outcomes. Orally administrable nanoformulations hold the great potential for improving the bioavailability of poorly permeable and poorly soluble therapeutics, but there are still limitations associated with these regimes. This review focuses on the barriers for oral drug delivery and various oral therapeutic nanoparticles for the management of CRC.
Collapse
Affiliation(s)
- Kangkang Ying
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC), Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingjun Bai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xing Gao
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuzi Xu
- Department of Oral Implantology and Prosthodontics, The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Hangxiang Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC), Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
| | - Binbin Xie
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC), Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
48
|
Wang T, He W, Du Y, Wang J, Li X. Redox-sensitive irinotecan liposomes with active ultra-high loading and enhanced intracellular drug release. Colloids Surf B Biointerfaces 2021; 206:111967. [PMID: 34256270 DOI: 10.1016/j.colsurfb.2021.111967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 01/21/2023]
Abstract
In this report, a novel irinotecan (IR) encapsulated redox-responsive liposome was developed. The redox-responsive liposomes were prepared based on disulfide phosphatidylcholine (SS-PC), DSPC, DSPE-PEG2000 and cholesterol by ethanol injection method. IR was actively loaded by triethylammonium sucrose octasulfate (TEA8-SOS) gradient method to generate IR/SS-PC liposomes (IR/SS-LP). The particle size of IR/SS-PC was characterized by using dynamic light scattering (DLS) and transmission electron microscopy (TEM). It was found that IR/SS-LP with 30 % content of SS-PC (IR/SS30-LP) had an average size of 125.5 ± 5.8 nm with a negative zeta potential of -19.5 ± 0.1. The encapsulation efficiency (EE) was further determined to be 98.1 ± 0.8 % and drug loading (DL) was 31.8 ± 0.1 %. The redox-responsiveness of IR/SS-LP was investigated by observing the change of particle size and morphology as well as the release behavior of IR triggered by glutathione (GSH). The data indicated GSH breaks the disulfide bonds in SS-PC and leads to the controlled release of IR. At 1 mM GSH, 60.2 % irinotecan was released from IR/SS30-LP within 24 h. Finally, the effects of IR/SS-LP in cell and animal experiments were evaluated in detail. The results showed that IR/SS30-LP had superior pharmacokinetic and antitumor efficacy compared to free irinotecan and traditional irinotecan liposome (ONIVYDE®-like). Taken together, IR/SS30-LP displayed redox-responsive release of IR, ultra-high loading and enhanced anti-tumor activity, which has great potential for clinical application as a new generation of IR liposomal formulation.
Collapse
Affiliation(s)
- Tao Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Wei He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China.
| |
Collapse
|
49
|
Kashapov R, Ibragimova A, Pavlov R, Gabdrakhmanov D, Kashapova N, Burilova E, Zakharova L, Sinyashin O. Nanocarriers for Biomedicine: From Lipid Formulations to Inorganic and Hybrid Nanoparticles. Int J Mol Sci 2021; 22:7055. [PMID: 34209023 PMCID: PMC8269010 DOI: 10.3390/ijms22137055] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Encapsulation of cargoes in nanocontainers is widely used in different fields to solve the problems of their solubility, homogeneity, stability, protection from unwanted chemical and biological destructive effects, and functional activity improvement. This approach is of special importance in biomedicine, since this makes it possible to reduce the limitations of drug delivery related to the toxicity and side effects of therapeutics, their low bioavailability and biocompatibility. This review highlights current progress in the use of lipid systems to deliver active substances to the human body. Various lipid compositions modified with amphiphilic open-chain and macrocyclic compounds, peptide molecules and alternative target ligands are discussed. Liposome modification also evolves by creating new hybrid structures consisting of organic and inorganic parts. Such nanohybrid platforms include cerasomes, which are considered as alternative nanocarriers allowing to reduce inherent limitations of lipid nanoparticles. Compositions based on mesoporous silica are beginning to acquire no less relevance due to their unique features, such as advanced porous properties, well-proven drug delivery efficiency and their versatility for creating highly efficient nanomaterials. The types of silica nanoparticles, their efficacy in biomedical applications and hybrid inorganic-polymer platforms are the subject of discussion in this review, with current challenges emphasized.
Collapse
Affiliation(s)
- Ruslan Kashapov
- A.E. Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Street 8, 420088 Kazan, Russia; (A.I.); (R.P.); (D.G.); (N.K.); (E.B.); (L.Z.); (O.S.)
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Song S, Xia H, Guo M, Wang S, Zhang S, Ma P, Jin Y. Role of macrophage in nanomedicine-based disease treatment. Drug Deliv 2021; 28:752-766. [PMID: 33860719 PMCID: PMC8079019 DOI: 10.1080/10717544.2021.1909175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Macrophages are a major component of the immunoresponse. Diversity and plasticity are two of the hallmarks of macrophages, which allow them to act as proinflammatory, anti-inflammatory, and homeostatic agents. Research has found that cancer and many inflammatory or autoimmune disorders are correlated with activation and tissue infiltration of macrophages. Recent developments in macrophage nanomedicine-based disease treatment are proving to be timely owing to the increasing inadequacy of traditional treatment. Here, we review the role of macrophages in nanomedicine-based disease treatment. First, we present a brief background on macrophages and nanomedicine. Then, we delve into applications of macrophages as a target for disease treatment and delivery systems and summarize the applications of macrophage-derived extracellular vesicles. Finally, we provide an outlook on the clinical utility of macrophages in nanomedicine-based disease treatment.
Collapse
Affiliation(s)
- Siwei Song
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujing Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|