1
|
Tao S, Song Y, Liu L, Ma W, Zhou B, Hu F. Herbal polysaccharide-based carrier enhances immunogenic cell death in cancer chemotherapy. Int J Biol Macromol 2025; 302:140501. [PMID: 39892542 DOI: 10.1016/j.ijbiomac.2025.140501] [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: 10/09/2024] [Revised: 01/13/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Breast cancer is a significant global health burden, with growth and metastasis contributing to its high mortality rate, highlighting the need for innovative treatment strategies. This study aims to develop breast cancer-targeted nanoparticles loaded with Doxorubicin (Dox) to combat both primary tumors and metastasis. By utilizing Dendrobium officinale polysaccharide (DOP), an herb polysaccharide with immunomodulatory activity, as a carrier, and incorporating folic acid (FA) as an active targeting ligand, we've achieved advancements in chemoimmunotherapy. After delivery of resulting FA@Dox nanoparticles to solid tumors by passive and active targeting, the tumor cells were killed by released Dox, and the immunogenic cell death was enhanced by the DOP, which promotes the proliferation of natural killer (NK) cells, facilitates dendritic cell maturation, and synergistically modulates various immune cells, preventing lung metastasis. The comprehensive assessments included material characterizations, in vitro and in vivo efficacy evaluations, biosafety analysis, and immune response studies. Given that DOP is derived from the medicinal and edible Dendrobium officinale, the developed DOP-based carrier holds great promise as an advanced nanomedicine platform. This offers promising prospects for synergistic modulation of multiple immune cells, particularly through NK cell proliferation, amplifying the efficacy of chemoimmunotherapy across diverse tumor types.
Collapse
Affiliation(s)
- Shengchang Tao
- Department of Pharmacy, Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; Biomaterials Research Center, School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medial Image Processing, Southern Medical University, Guangzhou 510515, China
| | - Yuchen Song
- Biomaterials Research Center, School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medial Image Processing, Southern Medical University, Guangzhou 510515, China
| | - Liuyi Liu
- Department of Pharmacy, Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Wenwen Ma
- Biomaterials Research Center, School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medial Image Processing, Southern Medical University, Guangzhou 510515, China
| | - Benjie Zhou
- Department of Pharmacy, Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Fang Hu
- Biomaterials Research Center, School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medial Image Processing, Southern Medical University, Guangzhou 510515, China; Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
2
|
Du L, Min H, Meng Y, Zhang K, Wang L, Zhang Y, Sun P, Zhang W, Qi Y, Wu G. LRG1-Targeted Camptothecin Nanomicelles with Simultaneous Delivery of Olaparib for Enhanced Colorectal Cancer Chemotherapy. NANO LETTERS 2025; 25:3130-3140. [PMID: 39947227 DOI: 10.1021/acs.nanolett.4c05354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Camptothecin (CPT), an effective topoisomerase I inhibitor used in colorectal cancer chemotherapy, often faces limitations due to severe toxicities. Addressing this, we developed OCENM, a nanomedicine featuring the CPT-ET conjugate─comprising a cathepsin B-sensitive linker and CPT linked to the ET peptide targeting leucine-rich alpha-2-glycoprotein 1 (LRG1) and encapsulating Olaparib, a potent poly ADP-ribose polymerase (PARP) inhibitor. OCENM aims for precise CPT delivery to colorectal cancer sites overexpressing LRG1, while Olaparib disrupts compromised DNA repair pathways, enhancing DNA damage and promoting increased tumor cell apoptosis. Our results show OCENM accumulates preferentially in colorectal cancer models through LRG1 targeting and triggers synergistic tumor cell apoptosis through the dual action of enhanced DNA damage and inhibited repair mechanisms. This study not only highlights the potential of LRG1 as a strategic target for nanomedicine delivery but also underscores the therapeutic promise of combining CPT with Olaparib for colorectal cancer treatment.
Collapse
Affiliation(s)
- Lin Du
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Huan Min
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yongkang Meng
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Kejuan Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Longdi Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Yana Zhang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Peichun Sun
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Wei Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Yingqiu Qi
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Gang Wu
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| |
Collapse
|
3
|
Yang JB, Xu DZ, Zhang ZH, Zhang X, Ren ZX, Lu ZL, Liu R, Liu Y. Multifunctional System with Camptothecin and [12]aneN 3 Units for Effective In Vivo Anti Pancreatic Cancer through Synergistic Chemotherapy, Gene Therapy, and Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67203-67215. [PMID: 39585759 DOI: 10.1021/acsami.4c12936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Maximizing drug cargo carrying capacity in blood circulation, controlling the in vivo fate of nanoparticles, and precisely drug release to tumor targets are the main aims of multifunctional nanomedicine-based antitumor therapy. Here we combined macrocyclic polyamine di(triazole-[12]aneN3) (M) and chemical drug camptothecin (CPT, C) through photosensitizer 1,1-dicyano-2-phenyl-2-(4-diphenylamino) phenyl-ethylene (DT) containing the cleavable disulfide (S) linkage as an all-in-one theranostic nanoprodrug, MDTSC. The corresponding compound with carbon chain (C) linkage, MDTCC, was also prepared for a comparison study. MDTSC showed the ability to carry plasmids, including the p53 tumor suppressor gene, to form lipoplexes with a size of ∼150 nm. Further addition of DOPE-PEG2k resulted in the hybrid lipoplexes MDTSC/DOPE-PEG2k@DNA, which showed good stability in blood circulation and good biocompatibility to normal cell lines. Experiments demonstrated that the hybrid lipoplexes were able to realize the successful cellular uptake and endosomal escape, to generate ROS under visible light irradiation as well as to trigger the localized release of CPT and the plasmid encoding p53 in tumor cells. In vitro, the hybrid lipoplexes showed better EGFP expression than the commercial Lipo2000, and markedly reduced tumor cell proliferation and migration rate irrespective of whether the BxPC-3 cell lines were grown on plates or 3D tumor spheroids. In vivo, the hybrid lipoplexes showed effective anticancer activity by reducing the BxPC-3 pancreatic tumor growth by 99% through the synergetic combination of chemotherapy, photodynamic therapy, and gene therapy. This research represented the first example of using a cocktail of three therapeutic approaches to achieve cooperative and effective anti pancreatic cancer treatment in vivo and in vitro.
Collapse
Affiliation(s)
- Jing-Bo Yang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- China National Institute for Food and Drug Control, Institute of Chemical Drug Control, HuaTuo Road 29, Beijing 100050, China
| | - De-Zhong Xu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zi-Han Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Xi Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zhi-Xuan Ren
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zhong-Lin Lu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Rui Liu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Yang Liu
- China National Institute for Food and Drug Control, Institute of Chemical Drug Control, HuaTuo Road 29, Beijing 100050, China
| |
Collapse
|
4
|
Zhang R, Yu J, Guo Z, Jiang H, Wang C. Camptothecin-based prodrug nanomedicines for cancer therapy. NANOSCALE 2023; 15:17658-17697. [PMID: 37909755 DOI: 10.1039/d3nr04147f] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Camptothecin (CPT) is a cytotoxic alkaloid that attenuates the replication of cancer cells via blocking DNA topoisomerase 1. Despite its encouraging and wide-spectrum antitumour activity, its application is significantly restricted owing to its instability, low solubility, significant toxicity, and acquired tumour cell resistance. This has resulted in the development of many CPT-based therapeutic agents, especially CPT-based nanomedicines, with improved pharmacokinetic and pharmacodynamic profiles. Specifically, smart CPT-based prodrug nanomedicines with stimuli-responsive release capacity have been extensively explored owing to the advantages such as high drug loading, improved stability, and decreased potential toxicity caused by the carrier materials in comparison with normal nanodrugs and traditional delivery systems. In this review, the potential strategies and applications of CPT-based nanoprodrugs for enhanced CPT delivery toward cancer cells are summarized. We appraise in detail the chemical structures and release mechanisms of these nanoprodrugs and guide materials chemists to develop more powerful nanomedicines that have real clinical therapeutic capacities.
Collapse
Affiliation(s)
- Renshuai Zhang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Zhu Guo
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
- The Affiliated Hospital of Qingdao University, Qingdao 266061, China
| | - Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Chao Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| |
Collapse
|
5
|
Chopra H, Verma R, Kaushik S, Parashar J, Madan K, Bano A, Bhardwaj R, Pandey P, Kumari B, Purohit D, Kumar M, Bhatia S, Rahman MH, Mittal V, Singh I, Kaushik D. Cyclodextrin-Based Arsenal for Anti-Cancer Treatments. Crit Rev Ther Drug Carrier Syst 2023; 40:1-41. [PMID: 36734912 DOI: 10.1615/critrevtherdrugcarriersyst.2022038398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Anti-cancer drugs are mostly limited in their use due to poor physicochemical and biopharmaceutical properties. Their lower solubility is the most common hurdle limiting their use upto their potential. In the recent years, the cyclodextrin (CD) complexation have emerged as existing approach to overcome the problem of poor solubility. CD-based nano-technological approaches are safe, stable and showed well in vivo tolerance and greater payload for encapsulation of hydrophobic drugs for the targeted delivery. They are generally chosen due to their ability to get self-assembled to form liposomes, nanoparticles, micelles and nano-sponges etc. This review paper describes a birds-eye view of the various CD-based nano-technological approaches applied for the delivery of anti-cancer moieties to the desired target such as CD based liposomes, niosomes, niosoponges, micelles, nanoparticles, monoclonal antibody, magnetic nanoparticles, small interfering RNA, nanorods, miscellaneous formulation of anti-cancer drugs containing CD. Moreover, the author also summarizes the various shortcomings of such a system and their way ahead.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Ravinder Verma
- Department of Pharmacy, G.D. Goenka University, Sohna Road, Gurugram 122103, India
| | - Sakshi Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Jatin Parashar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Kumud Madan
- Lloyd Institute of Management and Technology (Pharm), Knowledge Park, Greater Noida, U.P., India
| | - Afsareen Bano
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak 124001, India
| | - Rashmi Bhardwaj
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak 124001, India
| | - Parijat Pandey
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram 122413, India
| | - Beena Kumari
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, India
| | - Deepika Purohit
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, India
| | - Manish Kumar
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India
| | - Saurabh Bhatia
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| |
Collapse
|
6
|
Zhang L, Zhu L, Tang L, Xie J, Gao Y, Yu C, Shang K, Han H, Liu C, Lu Y. Glutathione-Responsive Nanoparticles of Camptothecin Prodrug for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205246. [PMID: 36442854 PMCID: PMC9875659 DOI: 10.1002/advs.202205246] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/17/2022] [Indexed: 05/28/2023]
Abstract
Camptothecin (CPT) is a potent chemotherapeutic agent for various cancers, but the broader application of CPT is still hindered by its poor bioavailability and systemic toxicity. Here, a prodrug that releases CPT in response to glutathione (GSH), which is commonly overexpressed by cancer cells is reported. Through assembling with PEGylated lipids, the prodrug is incorporated within as-assembled nanoparticles, affording CPT with a prolonged half-life in blood circulation, enhanced tumor targetingability, and improved therapeutic efficacy. Furthermore, such prodrug nanoparticles can also promote dendritic cell maturation and tumor infiltration of CD8+ T cells, providing a novel strategy to improve the therapeutic efficacy of CPT.
Collapse
Affiliation(s)
- Lingpu Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Lin Zhu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Lin Tang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Jiayi Xie
- Department of AutomaticTsinghua UniversityPeking University Third HospitalBeijing Key Laboratory of Magnetic Resonance Imaging Devices and TechnologyBeijing100191P. R. China
| | - Yajuan Gao
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Changyuan Yu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Kun Shang
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Hongbin Han
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Yunfeng Lu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| |
Collapse
|
7
|
Sabin C, Sam S, Hrishikes A, Salin B, Vigneshkumar PN, George J, John F. Supramolecular Drug Delivery Systems Based on Host‐Guest Interactions for Nucleic Acid Delivery. ChemistrySelect 2022. [DOI: 10.1002/slct.202203644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Christeena Sabin
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous), Thevara Kochi Kerala India 682013
| | - Samanta Sam
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous), Thevara Kochi Kerala India 682013
| | - A. Hrishikes
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous), Thevara Kochi Kerala India 682013
| | - Biyatris Salin
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous), Thevara Kochi Kerala India 682013
| | - P. N. Vigneshkumar
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous), Thevara Kochi Kerala India 682013
- Department of Chemistry The University of British Columbia Okanagan Vancouver BC V6T 1Z4 Canada
| | - Jinu George
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous), Thevara Kochi Kerala India 682013
| | - Franklin John
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous), Thevara Kochi Kerala India 682013
| |
Collapse
|
8
|
Murugan D, Murugesan V, Panchapakesan B, Rangasamy L. Nanoparticle Enhancement of Natural Killer (NK) Cell-Based Immunotherapy. Cancers (Basel) 2022; 14:cancers14215438. [PMID: 36358857 PMCID: PMC9653801 DOI: 10.3390/cancers14215438] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Simple Summary Natural killer cells are a part of the native immune response to cancer. NK cell-based immunotherapies are an emerging strategy to kill tumor cells. This paper reviews the role of NK cells, their mechanism of action for killing tumor cells, and the receptors which could serve as potential targets for signaling. In this review, the role of nanoparticles in NK cell activation and increased cytotoxicity of NK cells against cancer are highlighted. Abstract Natural killer (NK) cells are one of the first lines of defense against infections and malignancies. NK cell-based immunotherapies are emerging as an alternative to T cell-based immunotherapies. Preclinical and clinical studies of NK cell-based immunotherapies have given promising results in the past few decades for hematologic malignancies. Despite these achievements, NK cell-based immunotherapies have limitations, such as limited performance/low therapeutic efficiency in solid tumors, the short lifespan of NK cells, limited specificity of adoptive transfer and genetic modification, NK cell rejection by the patient’s immune system, insignificant infiltration of NK cells into the tumor microenvironment (TME), and the expensive nature of the treatment. Nanotechnology could potentially assist with the activation, proliferation, near-real time imaging, and enhancement of NK cell cytotoxic activity by guiding their function, analyzing their performance in near-real time, and improving immunotherapeutic efficiency. This paper reviews the role of NK cells, their mechanism of action in killing tumor cells, and the receptors which could serve as potential targets for signaling. Specifically, we have reviewed five different areas of nanotechnology that could enhance immunotherapy efficiency: nanoparticle-assisted immunomodulation to enhance NK cell activity, nanoparticles enhancing homing of NK cells, nanoparticle delivery of RNAi to enhance NK cell activity, genetic modulation of NK cells based on nanoparticles, and nanoparticle activation of NKG2D, which is the master regulator of all NK cell responses.
Collapse
Affiliation(s)
- Dhanashree Murugan
- School of Biosciences & Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Vasanth Murugesan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
- School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Balaji Panchapakesan
- Small Systems Laboratory, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
- Correspondence: (B.P.); (L.R.)
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
- Correspondence: (B.P.); (L.R.)
| |
Collapse
|
9
|
An Updated Overview of Cyclodextrin-Based Drug Delivery Systems for Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14081748. [PMID: 36015374 PMCID: PMC9412332 DOI: 10.3390/pharmaceutics14081748] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/19/2022] [Indexed: 12/18/2022] Open
Abstract
Encompassing a group of complex and heterogeneous diseases, cancer continues to be a challenge for patients and healthcare systems worldwide. Thus, it is of vital importance to develop advanced treatment strategies that could reduce the trends of cancer-associated morbidity and mortality rates. Scientists have focused on creating performant delivery vehicles for anti-cancer agents. Among the possible materials, cyclodextrins (CDs) attracted increasing interest over the past few years, leading to the emergence of promising anti-tumor nanomedicines. Tackling their advantageous chemical structure, ease of modification, natural origin, biocompatibility, low immunogenicity, and commercial availability, researchers investigated CD-based therapeutical formulations against many types of cancer. In this respect, in this paper, we briefly present the properties of interest of CDs for designing performant nanocarriers, further reviewing some of the most recent potential applications of CD-based delivery systems in cancer management.
Collapse
|
10
|
Păduraru DN, Niculescu AG, Bolocan A, Andronic O, Grumezescu AM, Bîrlă R. An Updated Overview of Cyclodextrin-Based Drug Delivery Systems for Cancer Therapy. Pharmaceutics 2022. [DOI: https://doi.org/10.3390/pharmaceutics14081748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Encompassing a group of complex and heterogeneous diseases, cancer continues to be a challenge for patients and healthcare systems worldwide. Thus, it is of vital importance to develop advanced treatment strategies that could reduce the trends of cancer-associated morbidity and mortality rates. Scientists have focused on creating performant delivery vehicles for anti-cancer agents. Among the possible materials, cyclodextrins (CDs) attracted increasing interest over the past few years, leading to the emergence of promising anti-tumor nanomedicines. Tackling their advantageous chemical structure, ease of modification, natural origin, biocompatibility, low immunogenicity, and commercial availability, researchers investigated CD-based therapeutical formulations against many types of cancer. In this respect, in this paper, we briefly present the properties of interest of CDs for designing performant nanocarriers, further reviewing some of the most recent potential applications of CD-based delivery systems in cancer management.
Collapse
|
11
|
Topuz F, Uyar T. Advances in the development of cyclodextrin-based nanogels/microgels for biomedical applications: Drug delivery and beyond. Carbohydr Polym 2022; 297:120033. [DOI: 10.1016/j.carbpol.2022.120033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 12/20/2022]
|
12
|
Fernández-Caro H, Méndez-Ardoy A, Montenegro J. Dynamic nanosurface reconfiguration by host-guest supramolecular interactions. NANOSCALE 2022; 14:3599-3608. [PMID: 35188162 DOI: 10.1039/d1nr05315a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The dynamic functionalization of the nanoparticle surface with biocompatible coatings is a critical step towards the development of functional nano-sized systems. While covalent approaches have been broadly exploited in the stabilization of nanoparticle colloidal systems, these strategies hinder the dynamic nanosurface chemical reconfiguration. Supramolecular strategies based on specific host-guest interactions hold promise due to their intrinsic reversibility, self-healing capabilities and modularity. Host/guest couples have recently been implemented in nanoparticle platforms for the exchange and release of effector molecules. However, the direct exchange of biocompatible hydrophilic oligomers (e.g. peptides) for the modulation of the surface charge and chemical properties of nanoparticles still remains a challenge. Here, we show the intracellular reconfiguration of nanoparticles by a host/guest mechanism with biocompatible oligomeric competitors. The surface of gold nanoparticles was functionalized with cyclodextrin hosts and the guest exchange was studied with biocompatible mono and divalent adamantyl competitors. The systematic characterization of the size and surface potential of the host/guest nanoparticles allowed the optimization of the binding and the stabilization properties of these supramolecular systems. The in cellulo host/guest-mediated direct reconfiguration of the peptide layer at the surface of nanoparticles is achieved by controlling the valence of adamantane-equipped peptides. This work demonstrates that host/guest supramolecular systems can be exploited for the direct exchange of pendants at the surface of nanoparticles and the intracellular dynamic chemical reconfiguration of biocompatible colloidal systems.
Collapse
Affiliation(s)
- Héctor Fernández-Caro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | | | - Javier Montenegro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
13
|
Wang Z, Little N, Chen J, Lambesis KT, Le KT, Han W, Scott AJ, Lu J. Immunogenic camptothesome nanovesicles comprising sphingomyelin-derived camptothecin bilayers for safe and synergistic cancer immunochemotherapy. NATURE NANOTECHNOLOGY 2021; 16:1130-1140. [PMID: 34385682 PMCID: PMC8855709 DOI: 10.1038/s41565-021-00950-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 06/28/2021] [Indexed: 05/02/2023]
Abstract
Despite the enormous therapeutic potential of immune checkpoint blockade (ICB), it benefits only a small subset of patients. Some chemotherapeutics can switch 'immune-cold' tumours to 'immune-hot' to synergize with ICB. However, safe and universal therapeutic platforms implementing such immune effects remain scarce. We demonstrate that sphingomyelin-derived camptothecin nanovesicles (camptothesomes) elicit potent granzyme-B- and perforin-mediated cytotoxic T lymphocyte (CTL) responses, potentiating PD-L1/PD-1 co-blockade to eradicate subcutaneous MC38 adenocarcinoma with developed memory immunity. In addition, camptothesomes improve the pharmacokinetics and lactone stability of camptothecin, avoid systemic toxicities, penetrate deeply into the tumour and outperform the antitumour efficacy of Onivyde. Camptothesome co-load the indoleamine 2,3-dioxygenase inhibitor indoximod into its interior using the lipid-bilayer-crossing capability of the immunogenic cell death inducer doxorubicin, eliminating clinically relevant advanced orthotopic CT26-Luc tumours and late-stage B16-F10-Luc2 melanoma, and achieving complete metastasis remission when combined with ICB and folate targeting. The sphingomyelin-derived nanotherapeutic platform and doxorubicin-enabled transmembrane transporting technology are generalizable to various therapeutics, paving the way for transformation of the cancer immunochemotherapy paradigm.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Nicholas Little
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Jiawei Chen
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Kevin Tyler Lambesis
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Kimberly Thi Le
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Weiguo Han
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Aaron James Scott
- NCI-Designated University of Arizona Comprehensive Cancer Center, Tucson, AZ, USA
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA.
- NCI-Designated University of Arizona Comprehensive Cancer Center, Tucson, AZ, USA.
- BIO5 Institute, The University of Arizona, Tucson, AZ, USA.
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
14
|
Lu N, Xi L, Zha Z, Wang Y, Han X, Ge Z. Acid-responsive endosomolytic polymeric nanoparticles with amplification of intracellular oxidative stress for prodrug delivery and activation. Biomater Sci 2021; 9:4613-4629. [PMID: 34190224 DOI: 10.1039/d1bm00159k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prodrug strategy especially in the field of chemotherapy of cancers possesses significant advantages reducing the side toxicity of anticancer drugs. However, high-efficiency delivery and in situ activation of prodrugs for tumor growth suppression are still a great challenge. Herein, we report rationally engineered pH-responsive endosomolytic polymeric micelles for the delivery of an oxidation-activable prodrug into the cytoplasm of cancer cells and amplification of intracellular oxidative stress for further prodrug activation. The prepared block copolymers consist of a poly(ethylene glycol) (PEG) block and a segment grafted by endosomolytic moieties and acetal linkage-connected cinnamaldehyde groups. The amphiphilic diblock copolymers can self-assemble to form micelles in water for loading the oxidation-activable phenylboronic pinacol ester-caged camptothecin prodrug (ProCPT). The obtained micelles can release free cinnamaldehyde under acidic conditions in tumor tissues and endo/lysosomes followed by efficient endosomal escape, which further induces enhancement of intracellular reactive oxygen species (ROS) to activate the prodrugs. Simultaneously, intracellular glutathione (GSH) can be reduced by quinone methide that was produced during prodrug activation. The ProCPT-loaded micelles can finally achieve efficient tumor accumulation and retention as well as effective tumor growth inhibition. More importantly, hematological and pathological analysis of toxicity reveals that the ProCPT-loaded micelles do not cause obvious toxic side effects toward important organs of mice. A positive immunomodulatory microenvironment in tumor tissue and serum can be detected after treatment with ProCPT-loaded micelles. Therefore, the endosomolytic ProCPT-loaded micelles exert synergistic therapeutic effects toward tumors through amplification of intracellular oxidative stress and activation of the prodrugs.
Collapse
Affiliation(s)
- Nannan Lu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
| | - Longchang Xi
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China.
| | - Zengshi Zha
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China.
| | - Yuheng Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China.
| | - Xinghua Han
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China.
| |
Collapse
|
15
|
Carrasco-Esteban E, Domínguez-Rullán JA, Barrionuevo-Castillo P, Pelari-Mici L, Leaman O, Sastre-Gallego S, López-Campos F. Current role of nanoparticles in the treatment of lung cancer. J Clin Transl Res 2021; 7:140-155. [PMID: 34104817 PMCID: PMC8177846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/20/2020] [Accepted: 01/27/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Worldwide, lung cancer is one of the leading causes of cancer death. Nevertheless, new therapeutic agents have been developed to treat lung cancer that could change this mortality-rate. Interestingly, incredible advances have occurred in recent years in the development and application of nanotechnology in the detection, diagnosis, and treatment of lung cancer. AIM Nanoparticles (NPs) have the ability to incorporate multiple drugs and targeting agents and therefore lead to an improved bioavailability, sustained delivery, solubility, and intestinal absorption. RELEVANCE FOR PATIENTS This review briefly summarizes the latest innovations in therapeutic nanomedicine in lung cancer with examples on magnetic, lipid, and polymer NP. Emphasis will be placed on future studies and ongoing clinical trials in this field.
Collapse
Affiliation(s)
| | | | | | - Lira Pelari-Mici
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Olwen Leaman
- Department of Radiation Oncology, Hospital Universitario Gregorio Marañon, Madrid, Spain
| | - Sara Sastre-Gallego
- Department of Radiation Oncology, Hospital Universitario Rey Juan Carlos, Madrid, Spain
| | - Fernando López-Campos
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| |
Collapse
|
16
|
Soni SS, Alsasa A, Rodell CB. Applications of Macrocyclic Host Molecules in Immune Modulation and Therapeutic Delivery. Front Chem 2021; 9:658548. [PMID: 33889565 PMCID: PMC8055865 DOI: 10.3389/fchem.2021.658548] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/12/2021] [Indexed: 12/17/2022] Open
Abstract
The immune system plays a central role in the development and progression of human disease. Modulation of the immune response is therefore a critical therapeutic target that enables us to approach some of the most vexing problems in medicine today such as obesity, cancer, viral infection, and autoimmunity. Methods of manipulating the immune system through therapeutic delivery centralize around two common themes: the local delivery of biomaterials to affect the surrounding tissue or the systemic delivery of soluble material systems, often aided by context-specific cell or tissue targeting strategies. In either case, supramolecular interactions enable control of biomaterial composition, structure, and behavior at the molecular-scale; through rational biomaterial design, the realization of next-generation immunotherapeutics and immunotheranostics is therefore made possible. This brief review highlights methods of harnessing macromolecular interaction for immunotherapeutic applications, with an emphasis on modes of drug delivery.
Collapse
Affiliation(s)
| | | | - Christopher B. Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
17
|
Li W, Peng A, Wu H, Quan Y, Li Y, Lu L, Cui M. Anti-Cancer Nanomedicines: A Revolution of Tumor Immunotherapy. Front Immunol 2020; 11:601497. [PMID: 33408716 PMCID: PMC7779686 DOI: 10.3389/fimmu.2020.601497] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Immunotherapies have been accelerating the development of anti-cancer clinical treatment, but its low objective responses and severe off-target immune-related adverse events (irAEs) limit the range of application. Strategies to remove these obstacles primarily focus on the combination of different therapies and the exploitation of new immunotherapeutic agents. Nanomedicine potentiates the effects of activating immune cells selectively and reversing tumor induced immune deficiency microenvironment through multiple mechanisms. In the last decade, a variety of nano-enabled tumor immunotherapies was under clinical investigation. As time goes by, the advantages of nanomedicine are increasingly prominent. With the continuous development of nanotechnology, nanomedicine will offer more distinctive perspectives in imaging diagnosis and treatment of tumors. In this Review, we wish to provide an overview of tumor immunotherapy and the mechanisms of nanomaterials that aim to enhance the efficacy of tumor immunotherapy under development or in clinic treatment.
Collapse
Affiliation(s)
- Wei Li
- Department of General Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Anghui Peng
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Huajun Wu
- Department of General Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yingyao Quan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Yong Li
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Min Cui
- Department of General Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| |
Collapse
|
18
|
Schmidt KT, Huitema ADR, Dorlo TPC, Peer CJ, Cordes LM, Sciuto L, Wroblewski S, Pommier Y, Madan RA, Thomas A, Figg WD. Population pharmacokinetic analysis of nanoparticle-bound and free camptothecin after administration of NLG207 in adults with advanced solid tumors. Cancer Chemother Pharmacol 2020; 86:475-486. [PMID: 32897402 PMCID: PMC7515962 DOI: 10.1007/s00280-020-04134-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/22/2020] [Indexed: 12/27/2022]
Abstract
PURPOSE NLG207 (formerly CRLX101) is a nanoparticle-drug conjugate (NDC) of the potent topoisomerase I inhibitor, camptothecin (CPT). The present study sought to characterize the complex pharmacokinetics (PK) of NLG207 and better describe CPT release from nanoparticles using a population PK (popPK) model. METHODS From 27 patients enrolled on two phase II clinical trials (NCT02769962 and NCT03531827), dense sampling was performed up to 48 h post-administration of NLG207 during cycle one and six of treatment; samples were also collected at ~ 360 h post-dose. Conjugated and free CPT concentrations were quantified from each sample, resulting in 477 observations to build a popPK model using non-linear mixed-effects modeling. RESULTS The PK of NLG207 was characterized by combining two linear two-compartment models with first-order kinetics each to describe nanoparticle-bound (conjugated) and free CPT. Allometric scaling based on body weight provided the best body-size descriptor for all PK parameters. The typical volumes of distribution of the conjugated CPT central and free CPT central compartments were 3.16 L (BSV CV%; 18.1%) and 21.1 L (CV%; 79.8%), respectively. CPT release from the nanoparticle formulation was characterized via an initial rapid clearance of 5.71 L/h (CV%; 62.6%), which decreased via first-order decay (estimated half-life of 0.307 h) to the steady-state value of 0.0988 L/h (CV%; 33.5%) by ~ 4 h after end of infusion. Renal clearance of free CPT was 0.874 L/h (CV%; 42.2%). CONCLUSION The popPK model confirmed nanoparticle behavior of conjugated CPT and mechanistically characterized CPT release from NLG207. The current analysis provides a strong foundation for future study as a potential predictive tool in ongoing NLG207 clinical trials.
Collapse
Affiliation(s)
- Keith T Schmidt
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10/Room 5A03, Bethesda, MD, 20892, USA
| | - Alwin D R Huitema
- Department Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Thomas P C Dorlo
- Department Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cody J Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10/Room 5A03, Bethesda, MD, 20892, USA
| | - Lisa M Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Linda Sciuto
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan Wroblewski
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William D Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10/Room 5A03, Bethesda, MD, 20892, USA.
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Kopeček J, Yang J. Polymer nanomedicines. Adv Drug Deliv Rev 2020; 156:40-64. [PMID: 32735811 PMCID: PMC7736172 DOI: 10.1016/j.addr.2020.07.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
Polymer nanomedicines (macromolecular therapeutics, polymer-drug conjugates, drug-free macromolecular therapeutics) are a group of biologically active compounds that are characterized by their large molecular weight. This review focuses on bioconjugates of water-soluble macromolecules with low molecular weight drugs and selected proteins. After analyzing the design principles, different structures of polymer carriers are discussed followed by the examination of the efficacy of the conjugates in animal models and challenges for their translation into the clinic. Two innovative directions in macromolecular therapeutics that depend on receptor crosslinking are highlighted: a) Combination chemotherapy of backbone degradable polymer-drug conjugates with immune checkpoint blockade by multivalent polymer peptide antagonists; and b) Drug-free macromolecular therapeutics, a new paradigm in drug delivery.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
20
|
Cyclodextrin as a magic switch in covalent and non-covalent anticancer drug release systems. Carbohydr Polym 2020; 242:116401. [PMID: 32564836 DOI: 10.1016/j.carbpol.2020.116401] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Cancer has been a threat to human health, so its treatment is a huge challenge to the present medical field. One of commonly used methods is the controlled release of anticancer drug to reduce the dose for patients, increase the stability of drug treatment and minimize side effects. Cyclodextrin is a kind of cyclic oligosaccharide produced by amylase hydrolysis. Because cyclodextrin contains a cavity structure and active hydroxyl groups, it has a positive effect on the study of the controlled release of anticancer drugs. This article reviews the controlled release of current anticancer drugs based on cyclodextrins as a "flexible switch", and discusses the classification of different types of release systems, highlighting their role in cancer treatment. Moreover, the opportunities and challenges of cyclodextrin as a magic switch in the controlled release of anticancer drugs are discussed.
Collapse
|