1
|
Yang D, Liu B, Sha H. Advances and prospects of cell-penetrating peptides in tumor immunotherapy. Sci Rep 2025; 15:3392. [PMID: 39870681 PMCID: PMC11772771 DOI: 10.1038/s41598-025-86130-8] [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: 10/25/2024] [Accepted: 01/08/2025] [Indexed: 01/29/2025] Open
Abstract
Cell-penetrating peptides (CPPs) have been shown to have superior material transport ability because poor infiltration of activated lymphocytes into tumors is one of the crucial factors limiting the therapeutic effect of tumor immunotherapy. Numerous studies have investigated the potential application of CPPs in tumor immunotherapy. This review delves into the crucial role that CPPs play in enhancing tumor immunotherapy, emphasizing their impact on various immunotherapy strategies, such as cytokine therapy, adoptive cell therapy, cancer vaccines, and immune checkpoint inhibitors. We also discuss the practical application challenges associated with enhancing the efficiency of CPPs in terms of their stability and targeting ability. In conclusion, the combination of CPPs with tumor immunotherapy is a promising strategy that has potential for precision administration and requires further research for optimal implementation.
Collapse
Affiliation(s)
- Di Yang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210023, China
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210023, China.
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China.
| | - Huizi Sha
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
2
|
Kumar A, Vaiphei KK, Gulbake A. A nanotechnology driven effectual localized lung cancer targeting approaches using tyrosine kinases inhibitors: Recent progress, preclinical assessment, challenges, and future perspectives. Int J Pharm 2024; 666:124745. [PMID: 39321904 DOI: 10.1016/j.ijpharm.2024.124745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
The higher incidence and mortality rate among all populations worldwide explains the unmet solutions in the treatment of lung cancer. The evolution of targeted therapies using tyrosine kinase inhibitors (TKI) has encouraged anticancer therapies. However, on-target and off-target effects and the development of drug resistance limited the anticancer potential of such targeted biologics. The advances in nanotechnology-driven-TKI embedded carriers that offered a new path toward lung cancer treatment. It is the inhalation route of administration known for its specific, precise, and efficient drug delivery to the lungs. The development of numerous TKI-nanocarriers through inhalation is proof of TKI growth. The future scopes involve using potential lung cancer biomarkers to achieve localized active cancer-targeting strategies. The adequate knowledge of in vitro absorption models usually helps establish better in vitro - in vivo correlation/extrapolation (IVIVC/E) to successfully evaluate inhalable drugs and drug products. The advanced in vitro and ex vivo lung tissue/ organ models offered better tumor heterogeneity, etiology, and microenvironment heterogeneity. The involvement of lung cancer organoids (LCOs), human organ chip models, and genetically modified mouse models (GEMMs) has resolved the challenges associated with conventional in vitro and in vivo models. To access potential inhalation-based drugtherapies, biological barriers, drug delivery, device-based challenges, and regulatory challenges must be encountered associated with their development. A proper understanding of material toxicity, size-based particle deposition at active disease sites, mucociliary clearance, phagocytosis, and the presence of enzymes and surfactants are required to achieve successful inhalational drug delivery (IDD). This article summarizes the future of lung cancer therapy using targeted drug-mediated inhalation using TKI.
Collapse
Affiliation(s)
- Ankaj Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Klaudi K Vaiphei
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India.
| |
Collapse
|
3
|
Jourdain MA, Eyer J. Recent advances in liposomes and peptide-based therapeutics for glioblastoma treatment. J Control Release 2024; 376:732-752. [PMID: 39437968 DOI: 10.1016/j.jconrel.2024.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
In the context of glioblastoma treatment, the penetration of drugs is drastically limited by the blood-brain-barrier (BBB). Emerging therapies have focused on the field of therapeutic peptides for their excellent BBB targeting properties that promote a deep tumor penetration. Peptide-based strategies are also renowned for their abilities of driving cargo such as liposomal system allowing an active targeting of receptors overexpressed on GBM cells. This review provides a detailed description of the internalization mechanisms of specific GBM homing and penetrating peptides as well as the latest in vitro/in vivo studies of liposomes functionalized with them. The purpose of this review is to summarize a selection of promising pre-clinical results that demonstrate the advantages of this nanosystem, including an increase of tumor cell targeting, triggering drug accumulation and thus a strong antitumor effect. Aware of the early stage of these studies, many challenges need to be overcome to promote peptide-directed liposome at clinical level. In particular, the lack of suitable production, the difficulty to characterize the nanosystem and therapeutic competition leaded by antibodies.
Collapse
Affiliation(s)
- M-A Jourdain
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France.
| | - J Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
4
|
Wang C, Shen Z, Chen Y, Wang Y, Zhou X, Chen X, Li Y, Zhang P, Zhang Q. Research Progress on Cyclic-Peptide Functionalized Nanoparticles for Tumor-Penetrating Delivery. Int J Nanomedicine 2024; 19:12633-12652. [PMID: 39624118 PMCID: PMC11609414 DOI: 10.2147/ijn.s487303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/14/2024] [Indexed: 01/03/2025] Open
Abstract
A key challenge in cancer treatment is the effective delivery of drugs into deep regions of tumor tissues, which are impermeable due to abnormal vascular network, increased interstitial fluid pressure (IFP), abundant extra cellular matrix (ECM), and heterogeneity of tumor cells. Cyclic peptides have been used for the surface engineering of nanoparticles to enhance the tumor-penetrating efficacy of drugs. Compared with other surface ligands, cyclic peptides are more easily produced by automated chemical synthesis, and they are featured by their higher binding affinity with their targets, tumor selectivity, stability against degradation, and low toxicity. In this review, different types of cyclic peptides, their physicochemical properties and their in vivo pharmacokinetics are introduced. Next, the progress of cyclic peptide-functionalized drug delivery nanodevices is updated, and the mechanism underlying the tumor-penetrating properties of cyclic peptide-functionalized drug delivery nanodevices is discussed.
Collapse
Affiliation(s)
- Chenkai Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Zefan Shen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yiyang Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yifan Wang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Xuanyi Zhou
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Xinyi Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Yuhang Li
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Pu Zhang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Qi Zhang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| |
Collapse
|
5
|
Nikitovic D, Kukovyakina E, Berdiaki A, Tzanakakis A, Luss A, Vlaskina E, Yagolovich A, Tsatsakis A, Kuskov A. Enhancing Tumor Targeted Therapy: The Role of iRGD Peptide in Advanced Drug Delivery Systems. Cancers (Basel) 2024; 16:3768. [PMID: 39594723 PMCID: PMC11592346 DOI: 10.3390/cancers16223768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Chemotherapy remains the primary therapeutic approach in treating cancer. The tumor microenvironment (TME) is the complex network surrounding tumor cells, comprising various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as ECM components, blood vessels, and signaling molecules. The often stiff and dense network of the TME interacts dynamically with tumor cells, influencing cancer growth, immune response, metastasis, and resistance to therapy. The effectiveness of the treatment of solid tumors is frequently reduced due to the poor penetration of the drug, which leads to attaining concentrations below the therapeutic levels at the site. Cell-penetrating peptides (CPPs) present a promising approach that improves the internalization of therapeutic agents. CPPs, which are short amino acid sequences, exhibit a high ability to pass cell membranes, enabling them to deliver drugs efficiently with minimal toxicity. Specifically, the iRGD peptide, a member of CPPs, is notable for its capacity to deeply penetrate tumor tissues by binding simultaneously integrins ανβ3/ανβ5 and neuropilin receptors. Indeed, ανβ3/ανβ5 integrins are characteristically expressed by tumor cells, which allows the iRGD peptide to home onto tumor cells. Notably, the respective dual-receptor targeting mechanism considerably increases the permeability of blood vessels in tumors, enabling an efficient delivery of co-administered drugs or nanoparticles into the tumor mass. Therefore, the iRGD peptide facilitates deeper drug penetration and improves the efficacy of co-administered therapies. Distinctively, we will focus on the iRGD mechanism of action, drug delivery systems and their application, and deliberate future perspectives in developing iRGD-conjugated therapeutics. In summary, this review discusses the potential of iRGD in overcoming barriers to drug delivery in cancer to maximize treatment efficiency while minimizing side effects.
Collapse
Affiliation(s)
- Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Ekaterina Kukovyakina
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| | - Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Alexandros Tzanakakis
- School of Electrical and Computer Engineering, National Technical University of Athens, 15780 Athens, Greece;
| | - Anna Luss
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| | - Elizaveta Vlaskina
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| | - Anne Yagolovich
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Aristides Tsatsakis
- Forensic Medicine Department, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Andrey Kuskov
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| |
Collapse
|
6
|
Hou X, Chen Q, Fang Y, Zhang L, Huang S, Xu M, Ren Y, Shi Z, Wei Y, Li L. iRGD-Guided Silica/Gold Nanoparticles for Efficient Tumor-Targeting and Enhancing Antitumor Efficacy Against Breast Cancer. Int J Nanomedicine 2024; 19:8237-8251. [PMID: 39157735 PMCID: PMC11329605 DOI: 10.2147/ijn.s474135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024] Open
Abstract
Background Breast cancer presents significant challenges due to the limited effectiveness of available treatments and the high likelihood of recurrence. iRGD possesses both RGD sequence and C-terminal sequence and has dual functions of targeting and membrane penetration. iRGD-modified nanocarriers can enhance drug targeting of tumor vascular endothelial cells and penetration of new microvessels, increasing drug concentration in tumor tissues. Methods The amidation reaction was carried out between SiO2/AuNCs and iRGD/PTX, yielding a conjugated drug delivery system (SiO2/AuNCs-iRGD/PTX, SAIP@NPs). The assessment encompassed the characterization of the morphology, particle size distribution, physicochemical properties, in vitro release profile, cytotoxicity, and cellular uptake of SAIP@NPs. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed using a small animal in vivo imaging system and a tumor-bearing nude mice model, respectively. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed utilizing a small animal in vivo imaging system and an in situ nude mice breast cancer xenograft model, respectively. Results The prepared SAIP@NPs exhibited decent stability and a certain slow-release effect in phosphate buffer (PBS, pH 7.4). In vitro studies had shown that, due to the dual functions of transmembrane and targeting of iRGD peptide, SAIP@NPs exhibited strong binding to integrin αvβ3, which was highly expressed on the membrane of MDA-MB-231 cells, improving the uptake capacity of tumor cells, inhibiting the rapid growth of tumor cells, and promoting tumor cell apoptosis. The results of animal experiments further proved that SAIP@NPs had longer residence time in tumor sites, stronger anti-tumor effect, and no obvious toxicity to major organs of experimental animals. Conclusion The engineered SAIP@NPs exhibited superior functionalities including efficient membrane permeability, precise tumor targeting, and imaging, thereby significantly augmenting the therapeutic efficacy against breast cancer with a favorable safety profile.
Collapse
Affiliation(s)
- Xuefeng Hou
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Qi Chen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Ying Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Li Zhang
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Shuoheng Huang
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Minjie Xu
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Yaning Ren
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Zhansen Shi
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Yan Wei
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Lihua Li
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| |
Collapse
|
7
|
Milewska S, Sadowska A, Stefaniuk N, Misztalewska-Turkowicz I, Wilczewska AZ, Car H, Niemirowicz-Laskowska K. Tumor-Homing Peptides as Crucial Component of Magnetic-Based Delivery Systems: Recent Developments and Pharmacoeconomical Perspective. Int J Mol Sci 2024; 25:6219. [PMID: 38892406 PMCID: PMC11172452 DOI: 10.3390/ijms25116219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
According to data from the World Health Organization (WHO), cancer is considered to be one of the leading causes of death worldwide, and new therapeutic approaches, especially improved novel cancer treatment regimens, are in high demand. Considering that many chemotherapeutic drugs tend to have poor pharmacokinetic profiles, including rapid clearance and limited on-site accumulation, a combined approach with tumor-homing peptide (THP)-functionalized magnetic nanoparticles could lead to remarkable improvements. This is confirmed by an increasing number of papers in this field, showing that the on-target peptide functionalization of magnetic nanoparticles improves their penetration properties and ensures tumor-specific binding, which results in an increased clinical response. This review aims to highlight the potential applications of THPs in combination with magnetic carriers across various fields, including a pharmacoeconomic perspective.
Collapse
Affiliation(s)
- Sylwia Milewska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | - Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | - Natalia Stefaniuk
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | | | - Agnieszka Z. Wilczewska
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland; (I.M.-T.); (A.Z.W.)
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | - Katarzyna Niemirowicz-Laskowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| |
Collapse
|
8
|
Creeden JF, Sevier J, Zhang JT, Lapitsky Y, Brunicardi FC, Jin G, Nemunaitis J, Liu JY, Kalinoski A, Rao D, Liu SH. Smart exosomes enhance PDAC targeted therapy. J Control Release 2024; 368:413-429. [PMID: 38431093 DOI: 10.1016/j.jconrel.2024.02.037] [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: 11/02/2023] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Exosomes continue to attract interest as a promising nanocarrier drug delivery technology. They are naturally derived nanoscale extracellular vesicles with innate properties well suited to shuttle proteins, lipids, and nucleic acids between cells. Nonetheless, their clinical utility is currently limited by several major challenges, such as their inability to target tumor cells and a high proportion of clearance by the mononuclear phagocyte system (MPS) of the liver and spleen. To overcome these limitations, we developed "Smart Exosomes" that co-display RGD and CD47p110-130 through CD9 engineering (ExoSmart). The resultant ExoSmart demonstrates enhanced binding capacity to αvβ3 on pancreatic ductal adenocarcinoma (PDAC) cells, resulting in amplified cellular uptake in in vitro and in vivo models and increased chemotherapeutic efficacies. Simultaneously, ExoSmart significantly reduced liver and spleen clearance of exosomes by inhibiting macrophage phagocytosis via CD47p110-130 interaction with signal regulatory proteins (SIRPα) on macrophages. These studies demonstrate that an engineered exosome drug delivery system increases PDAC therapeutic efficacy by enhancing active PDAC targeting and prolonging circulation times, and their findings hold tremendous translational potential for cancer therapy while providing a concrete foundation for future work utilizing novel peptide-engineered exosome strategies.
Collapse
Affiliation(s)
- Justin F Creeden
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Jonathan Sevier
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Yakov Lapitsky
- Department of Chemical Engineering, University of Toledo, Toledo, OH, USA
| | - F Charles Brunicardi
- Department of Surgery, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Ge Jin
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | - Jing-Yuan Liu
- Department of Medicine, University of Toledo, Toledo, OH, USA
| | | | | | - Shi-He Liu
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
9
|
Wang Y, Zhang L, Liu C, Luo Y, Chen D. Peptide-Mediated Nanocarriers for Targeted Drug Delivery: Developments and Strategies. Pharmaceutics 2024; 16:240. [PMID: 38399294 PMCID: PMC10893007 DOI: 10.3390/pharmaceutics16020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Effective drug delivery is essential for cancer treatment. Drug delivery systems, which can be tailored to targeted transport and integrated tumor therapy, are vital in improving the efficiency of cancer treatment. Peptides play a significant role in various biological and physiological functions and offer high design flexibility, excellent biocompatibility, adjustable morphology, and biodegradability, making them promising candidates for drug delivery. This paper reviews peptide-mediated drug delivery systems, focusing on self-assembled peptides and peptide-drug conjugates. It discusses the mechanisms and structural control of self-assembled peptides, the varieties and roles of peptide-drug conjugates, and strategies to augment peptide stability. The review concludes by addressing challenges and future directions.
Collapse
Affiliation(s)
- Yubo Wang
- Medical College, Guangxi University, Da-Xue-Dong Road No. 100, Nanning 530004, China;
| | - Lu Zhang
- School of Life Sciences, Xiamen University, Xiamen 361005, China;
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| | - Chen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| | - Yiming Luo
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen 361003, China
- The School of Clinical Medicine, Fujian Medical University, Fuzhou 351002, China
| | - Dengyue Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| |
Collapse
|
10
|
Dailey KM, Small JM, Pullan JE, Winfree S, Vance KE, Orr M, Mallik S, Bayles KW, Hollingsworth MA, Brooks AE. An intravenous pancreatic cancer therapeutic: Characterization of CRISPR/Cas9n-modified Clostridium novyi-Non Toxic. PLoS One 2023; 18:e0289183. [PMID: 37963142 PMCID: PMC10645340 DOI: 10.1371/journal.pone.0289183] [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: 03/17/2023] [Accepted: 07/12/2023] [Indexed: 11/16/2023] Open
Abstract
Clostridium novyi has demonstrated selective efficacy against solid tumors largely due to the microenvironment contained within dense tumor cores. The core of a solid tumor is typically hypoxic, acidic, and necrotic-impeding the penetration of current therapeutics. C. novyi is attracted to the tumor microenvironment and once there, can both lyse and proliferate while simultaneously re-activating the suppressed immune system. C. novyi systemic toxicity is easily mitigated by knocking out the phage DNA plasmid encoded alpha toxin resulting in C. novyi-NT; but, after intravenous injection spores are quickly cleared by phagocytosis before accomplishing significant tumor localization. C. novyi-NT could be designed to accomplish intravenous delivery with the potential to target all solid tumors and their metastases in a single dose. This study characterizes CRISPR/Cas9 modified C. novyi-NT to insert the gene for RGD, a tumor targeting peptide, expressed within the promoter region of a spore coat protein. Expression of the RGD peptide on the outer spore coat of C. novyi-NT indicates an increased capacity for tumor localization of C. novyi upon intravenous introduction based on the natural binding of RGD with the αvβ3 integrin commonly overexpressed on the epithelial tissue surrounding a tumor, and lead to immune stimulation.
Collapse
Affiliation(s)
- Kaitlin M. Dailey
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, United States of America
- Cell and Molecular Biology Program, North Dakota State University, Fargo, ND, United States of America
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States of America
| | - James M. Small
- Department of Pathology and Microbiology, Rocky Vista University, Parker, CO, United States of America
| | - Jessica E. Pullan
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States of America
- Department of Physical Science, Southern Utah University, Cedar City, UT, United States of America
| | - Seth Winfree
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, United States of America
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Krysten E. Vance
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Megan Orr
- Department of Statistics, North Dakota State University, Fargo, ND, United States of America
- Center for Diagnostics and Therapeutic Strategies in Pancreatic Cancer Biostatistics Core Facility, North Dakota State University, Fargo, ND, United States of America
| | - Sanku Mallik
- Cell and Molecular Biology Program, North Dakota State University, Fargo, ND, United States of America
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States of America
| | - Kenneth W. Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Amanda E. Brooks
- Cell and Molecular Biology Program, North Dakota State University, Fargo, ND, United States of America
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States of America
- Department of Research and Scholarly Activity, Rocky Vista University, Ivins, UT, United States of America
| |
Collapse
|
11
|
D’Amore V, Donati G, Lenci E, Ludwig BS, Kossatz S, Baiula M, Trabocchi A, Kessler H, Di Leva FS, Marinelli L. Molecular View on the iRGD Peptide Binding Mechanism: Implications for Integrin Activity and Selectivity Profiles. J Chem Inf Model 2023; 63:6302-6315. [PMID: 37788340 PMCID: PMC10598797 DOI: 10.1021/acs.jcim.3c01071] [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: 07/14/2023] [Indexed: 10/05/2023]
Abstract
Receptor-selective peptides are widely used as smart carriers for specific tumor-targeted delivery. A remarkable example is the cyclic nonapeptide iRGD (CRGDKPGDC, 1) that couples intrinsic cytotoxic effects with striking tumor-homing properties. These peculiar features are based on a rather complex multistep mechanism of action, where the primary event is the recognition of RGD integrins. Despite the high number of preclinical studies and the recent success of a phase I trial for the treatment of pancreatic ductal adenocarcinoma (PDAC), there is little information available about the iRGD three-dimensional (3D) structure and integrin binding properties. Here, we re-evaluate the peptide's affinity for cancer-related integrins including not only the previously known targets αvβ3 and αvβ5 but also the αvβ6 isoform, which is known to drive cell growth, migration, and invasion in many malignancies including PDAC. Furthermore, we use parallel tempering in the well-tempered ensemble (PT-WTE) metadynamics simulations to characterize the in-solution conformation of iRGD and extensive molecular dynamics calculations to fully investigate its binding mechanism to integrin partners. Finally, we provide clues for fine-tuning the peptide's potency and selectivity profile, which, in turn, may further improve its tumor-homing properties.
Collapse
Affiliation(s)
- Vincenzo
Maria D’Amore
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Greta Donati
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Elena Lenci
- Department
of Chemistry “Ugo Schiff″, University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Florence, Italy
| | - Beatrice Stefanie Ludwig
- Department
of Nuclear Medicine, University Hospital Klinikum Rechts der Isar
and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, Munich 81675, Germany
| | - Susanne Kossatz
- Department
of Nuclear Medicine, University Hospital Klinikum Rechts der Isar
and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, Munich 81675, Germany
- Department
of Chemistry, Institute for Advanced Study, Technical University Munich, Garching 85748, Germany
| | - Monica Baiula
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Andrea Trabocchi
- Department
of Chemistry “Ugo Schiff″, University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Florence, Italy
| | - Horst Kessler
- Department
of Chemistry, Institute for Advanced Study, Technical University Munich, Garching 85748, Germany
| | - Francesco Saverio Di Leva
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Luciana Marinelli
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| |
Collapse
|
12
|
Yang X, Xie X, Liu S, Ma W, Zheng Z, Wei H, Yu CY. Engineered Exosomes as Theranostic Platforms for Cancer Treatment. ACS Biomater Sci Eng 2023; 9:5479-5503. [PMID: 37695590 DOI: 10.1021/acsbiomaterials.3c00745] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Tremendous progress in nanotechnology and nanomedicine has made a significant positive effect on cancer treatment by integrating multicomponents into a single multifunctional nanosized delivery system for combinatorial therapies. Although numerous nanocarriers developed so far have achieved excellent therapeutic performance in mouse models via elegant integration of chemotherapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, their synthetic origin may still cause systemic toxicity, immunogenicity, and preferential detection or elimination by the immune system. Exosomes, endogenous nanosized particles secreted by multiple biological cells, could be absorbed by recipient cells to facilitate intercellular communication and content delivery. Therefore, exosomes have emerged as novel cargo delivery tools and attracted considerable attention for cancer diagnosis and treatment due to their innate stability, biological compatibility, and biomembrane penetration capacity. Exosome-related properties and functions have been well-documented; however, there are few reviews, to our knowledge, with a focus on the combination of exosomes and nanotechnology for the development of exosome-based theranostic platforms. To make a timely review on this hot subject of research, we summarize the basic information, isolation and functionalization methodologies, diagnostic and therapeutic potential of exosomes in various cancers with an emphasis on the description of exosome-related nanomedicine for cancer theranostics. The existing appealing challenges and outlook in exosome clinical translation are finally introduced. Advanced biotechnology and nanotechnology will definitely not only promote the integration of intrinsic advantages of natural nanosized exosomes with traditional synthetic nanomaterials for modulated precise cancer treatment but also contribute to the clinical translations of exosome-based nanomedicine as theranostic nanoplatforms.
Collapse
Affiliation(s)
- Xu Yang
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Xiangyu Xie
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Songbin Liu
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Wei Ma
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Zhi Zheng
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Hua Wei
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Cui-Yun Yu
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
13
|
Gong L, Zhao H, Liu Y, Wu H, Liu C, Chang S, Chen L, Jin M, Wang Q, Gao Z, Huang W. Research advances in peptide‒drug conjugates. Acta Pharm Sin B 2023; 13:3659-3677. [PMID: 37719380 PMCID: PMC10501876 DOI: 10.1016/j.apsb.2023.02.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/05/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
Peptide‒drug conjugates (PDCs) are drug delivery systems consisting of a drug covalently coupled to a multifunctional peptide via a cleavable linker. As an emerging prodrug strategy, PDCs not only preserve the function and bioactivity of the peptides but also release the drugs responsively with the cleavable property of the linkers. Given the ability to significantly improve the circulation stability and targeting of drugs in vivo and reduce the toxic side effects of drugs, PDCs have already been extensively applied in drug delivery. Herein, we review the types and mechanisms of peptides, linkers and drugs used to construct PDCs, and summarize the clinical applications and challenges of PDC drugs.
Collapse
Affiliation(s)
- Liming Gong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Heming Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao Wu
- Department of Pharmacy, Yanbian University, Yanji 133000, China
| | - Chao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuangyan Chang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qiming Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
14
|
Wei D, Zhang N, Qu S, Wang H, Li J. Advances in nanotechnology for the treatment of GBM. Front Neurosci 2023; 17:1180943. [PMID: 37214394 PMCID: PMC10196029 DOI: 10.3389/fnins.2023.1180943] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/05/2023] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM), a highly malignant glioma of the central nervous system, is the most dread and common brain tumor with a high rate of therapeutic resistance and recurrence. Currently, the clinical treatment methods are surgery, radiotherapy, and chemotherapy. However, owning to the highly invasive nature of GBM, it is difficult to completely resect them due to the unclear boundary between the edges of GBM and normal brain tissue. Traditional radiotherapy and the combination of alkylating agents and radiotherapy have significant side effects, therapeutic drugs are difficult to penetrate the blood brain barrier. Patients receiving treatment have a high postoperative recurrence rate and a median survival of less than 2 years, Less than 5% of patients live longer than 5 years. Therefore, it is urgent to achieve precise treatment through the blood brain barrier and reduce toxic and side effects. Nanotechnology exhibit great potential in this area. This article summarizes the current treatment methods and shortcomings of GBM, and summarizes the research progress in the diagnosis and treatment of GBM using nanotechnology.
Collapse
Affiliation(s)
- Dongyan Wei
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, China
- College of Life Sciences, Tarim University, Alar, China
| | - Ni Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shuang Qu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Hao Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jin Li
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Liu Y, Chang R, Xing R, Yan X. Bioactive Peptide Nanodrugs Based on Supramolecular Assembly for Boosting Immunogenic Cell Death-Induced Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201708. [PMID: 36720041 DOI: 10.1002/smtd.202201708] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/12/2023] [Indexed: 05/17/2023]
Abstract
Immunogenic cell death (ICD)-induced immunotherapy holds promise for complete elimination and long-term protective immune responses against cancer by combining direct tumor cell killing and antitumor immune response. Some therapeutic approaches (such as hyperthermia, photodynamic therapy, or radiotherapy) and inducers (certain chemotherapy drugs, oncolytic viruses) have been devoted to initiating and/or boosting ICD, leading to the activation of tumor-specific immune responses. Recently, supramolecular assembled bioactive peptide nanodrugs have been employed to improve the efficacy of ICD-induced cancer immunotherapy by increasing tumor targeted accumulation as well as responsive release of ICD inducers, directly inducing high levels of ICD and realizing the simultaneous enhancement of immune response through the immune function of the active peptide itself. Here, the authors review bioactive peptide nanodrugs based on supramolecular assembly, mainly as an intelligent delivery system, a direct ICD inducer and an immune response enhancer, for boosting ICD induced cancer immunotherapy. The functions of diverse bioactive peptides used in the construction of nanodrugs are described. The design of a supramolecular assembly, the mechanism of boosting ICD, and synergetic effects of bioactive peptides combined immunotherapy are critically emphasized.
Collapse
Affiliation(s)
- Yamei Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Rui Chang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ruirui Xing
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- Center for Mesoscience, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
16
|
Pan R, He T, Zhang K, Zhu L, Lin J, Chen P, Liu X, Huang H, Zhou D, Li W, Yang S, Ye G. Tumor-Targeting Extracellular Vesicles Loaded with siS100A4 for Suppressing Postoperative Breast Cancer Metastasis. Cell Mol Bioeng 2023; 16:117-125. [PMID: 37096069 PMCID: PMC10121989 DOI: 10.1007/s12195-022-00757-5] [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: 07/25/2022] [Accepted: 12/14/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction S100A4 promotes the establishment of tumor microenvironment for malignant cancer cells, and knockdown of S100A4 can inhibit tumorigenesis. However, there is no efficient way to target S100A4 in metastatic tumor tissues. Here, we investigated the role of siS100A4-loaded iRGD-modified extracellular vesicles (siS100A4-iRGD-EVs) in postoperative breast cancer metastasis. Methods siS100A4-iRGD-EVs nanoparticles were engineered and analyzed using TEM and DLS. siRNA protection, cellular uptake, and cytotoxicity of EV nanoparticles were examined in vitro. Postoperative lung metastasis mouse model was created to investigate the tissue distribution and anti-metastasis roles of nanoparticles in vivo. Results siS100A4-iRGD-EVs protected siRNA from RNase degradation, enhanced the cellular uptake and compatibility in vitro. Strikingly, iRGD-modified EVs significantly increased tumor organotropism and siRNA accumulation in lung PMNs compared to siS100A4-EVs in vivo. Moreover, siS100A4-iRGD-EVs treatment remarkedly attenuated lung metastases from breast cancer and increased survival rate of mice through suppressing S100A4 expression in lung. Conclusions siS100A4-iRGD-EVs nanoparticles show more potent anti-metastasis effect in postoperative breast cancer metastasis mouse model. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00757-5.
Collapse
Affiliation(s)
- Ruiling Pan
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Tiancheng He
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Kun Zhang
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Lewei Zhu
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Jiawei Lin
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Peixian Chen
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Xiangwei Liu
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Huiqi Huang
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Dan Zhou
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Wei Li
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Shuqing Yang
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Guolin Ye
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| |
Collapse
|
17
|
Gorachinov F, Mraiche F, Moustafa DA, Hishari O, Ismail Y, Joseph J, Crcarevska MS, Dodov MG, Geskovski N, Goracinova K. Nanotechnology - a robust tool for fighting the challenges of drug resistance in non-small cell lung cancer. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:240-261. [PMID: 36865093 PMCID: PMC9972888 DOI: 10.3762/bjnano.14.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Genomic and proteomic mutation analysis is the standard of care for selecting candidates for therapies with tyrosine kinase inhibitors against the human epidermal growth factor receptor (EGFR TKI therapies) and further monitoring cancer treatment efficacy and cancer development. Acquired resistance due to various genetic aberrations is an unavoidable problem during EGFR TKI therapy, leading to the rapid exhaustion of standard molecularly targeted therapeutic options against mutant variants. Attacking multiple molecular targets within one or several signaling pathways by co-delivery of multiple agents is a viable strategy for overcoming and preventing resistance to EGFR TKIs. However, because of the difference in pharmacokinetics among agents, combined therapies may not effectively reach their targets. The obstacles regarding the simultaneous co-delivery of therapeutic agents at the site of action can be overcome using nanomedicine as a platform and nanotools as delivery agents. Precision oncology research to identify targetable biomarkers and optimize tumor homing agents, hand in hand with designing multifunctional and multistage nanocarriers that respond to the inherent heterogeneity of the tumors, may resolve the challenges of inadequate tumor localization, improve intracellular internalization, and bring advantages over conventional nanocarriers.
Collapse
Affiliation(s)
- Filip Gorachinov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, T6G 2R3 Edmonton, Canada
| | | | - Ola Hishari
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Yomna Ismail
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Jensa Joseph
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Maja Simonoska Crcarevska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Marija Glavas Dodov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Katerina Goracinova
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| |
Collapse
|
18
|
Chavda VP, Solanki HK, Davidson M, Apostolopoulos V, Bojarska J. Peptide-Drug Conjugates: A New Hope for Cancer Management. Molecules 2022; 27:7232. [PMID: 36364057 PMCID: PMC9658517 DOI: 10.3390/molecules27217232] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/29/2022] [Accepted: 10/18/2022] [Indexed: 08/07/2023] Open
Abstract
Cancer remains the leading cause of death worldwide despite advances in treatment options for patients. As such, safe and effective therapeutics are required. Short peptides provide advantages to be used in cancer management due to their unique properties, amazing versatility, and progress in biotechnology to overcome peptide limitations. Several appealing peptide-based therapeutic strategies have been developed. Here, we provide an overview of peptide conjugates, the better equivalents of antibody-drug conjugates, as the next generation of drugs for required precise targeting, enhanced cellular permeability, improved drug selectivity, and reduced toxicity for the efficient treatment of cancers. We discuss the basic components of drug conjugates and their release action, including the release of cytotoxins from the linker. We also present peptide-drug conjugates under different stages of clinical development as well as regulatory and other challenges.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Hetvi K. Solanki
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Majid Davidson
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Joanna Bojarska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 116 Zeromskiego Street, 90-924 Lodz, Poland
| |
Collapse
|
19
|
Yagolovich AV, Isakova AA, Artykov AA, Vorontsova YV, Mazur DV, Antipova NV, Pavlyukov MS, Shakhparonov MI, Gileva AM, Markvicheva EA, Plotnikova EA, Pankratov AA, Kirpichnikov MP, Gasparian ME, Dolgikh DA. DR5-Selective TRAIL Variant DR5-B Functionalized with Tumor-Penetrating iRGD Peptide for Enhanced Antitumor Activity against Glioblastoma. Int J Mol Sci 2022; 23:12687. [PMID: 36293545 PMCID: PMC9604365 DOI: 10.3390/ijms232012687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
TRAIL (TNF-related apoptosis-inducing ligand) and its derivatives are potentials for anticancer therapy due to the selective induction of apoptosis in tumor cells upon binding to death receptors DR4 or DR5. Previously, we generated a DR5-selective TRAIL mutant variant DR5-B overcoming receptor-dependent resistance of tumor cells to TRAIL. In the current study, we improved the antitumor activity of DR5-B by fusion with a tumor-homing iRGD peptide, which is known to enhance the drug penetration into tumor tissues. The obtained bispecific fusion protein DR5-B-iRGD exhibited dual affinity for DR5 and integrin αvβ3 receptors. DR5-B-iRGD penetrated into U-87 tumor spheroids faster than DR5-B and demonstrated an enhanced antitumor effect in human glioblastoma cell lines T98G and U-87, as well as in primary patient-derived glioblastoma neurospheres in vitro. Additionally, DR5-B-iRGD was highly effective in a xenograft mouse model of the U-87 human glioblastoma cell line in vivo. We suggest that DR5-B-iRGD may become a promising candidate for targeted therapy for glioblastoma.
Collapse
Affiliation(s)
- Anne V. Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
- Manebio LLC, 115280 Moscow, Russia
| | - Alina A. Isakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Artem A. Artykov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Manebio LLC, 115280 Moscow, Russia
| | | | - Diana V. Mazur
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Nadezhda V. Antipova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Marat S. Pavlyukov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | | | - Anastasia M. Gileva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Elena A. Markvicheva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Ekaterina A. Plotnikova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow Oncology Research Institute, 125284 Moscow, Russia
| | - Andrey A. Pankratov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow Oncology Research Institute, 125284 Moscow, Russia
| | - Mikhail P. Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Marine E. Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Dmitry A. Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| |
Collapse
|
20
|
Zhang X, Chen Y, Sun D, Zhu X, Ying X, Yao Y, Fei W, Zheng C. Emerging pharmacologic interventions for pre-eclampsia treatment. Expert Opin Ther Targets 2022; 26:739-759. [PMID: 36223503 DOI: 10.1080/14728222.2022.2134779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Pre-eclampsia is a serious pregnancy complication and a major global concern for the mortality of both mother and fetus. Existing symptomatic treatments do not delay disease progression; thus, timely delivery of the baby is the most effective measure. However, the risk of various maternal and fetal injuries remains. AREAS COVERED In this review, we summarize the potential strategies for pharmacologic interventions in pre-eclamptic therapy. Specifically, we discuss the pathophysiological process of various effective candidate therapeutics that act on potential pathways and molecular targets to inhibit key stages of the disease. We refer to this pathogenesis-focused drug discovery model as a pathogenesis-target-drug (P-T-D) strategy. Finally, we discuss the introduction of nanotechnologies to improve the safety and efficacy of therapeutics via their specific placental targeting ability and placental retention effects. EXPERT OPINION Despite the active development of novel pharmacological treatments based on our current knowledge of pre-eclamptic pathogenesis, investigations are still in the early phase. Thus, further exploration of the pathological mechanisms, integrated with the P-T-D strategy and novel nanosystems, could encourage the development of more effective and safer strategies. Such advances could lead to a shift from expectant management to mechanistic-based therapy for pre-eclampsia.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Dongli Sun
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xia Ying
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
21
|
Chen Y, Wang Z, Wang X, Su M, Xu F, Yang L, Jia L, Zhang Z. Advances in Antitumor Nano-Drug Delivery Systems of 10-Hydroxycamptothecin. Int J Nanomedicine 2022; 17:4227-4259. [PMID: 36134205 PMCID: PMC9482956 DOI: 10.2147/ijn.s377149] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/25/2022] [Indexed: 01/10/2023] Open
Abstract
10-Hydroxycamptothecin (HCPT) is a natural plant alkaloid from Camptotheca that shows potent antitumor activity by targeting intracellular topoisomerase I. However, factors such as instability of the lactone ring and insolubility in water have limited the clinical application of this drug. In recent years, unprecedented advances in biomedical nanotechnology have facilitated the development of nano drug delivery systems. It has been found that nanomedicine can significantly improve the stability and water solubility of HCPT. NanoMedicines with different diagnostic and therapeutic functions have been developed to significantly improve the anticancer effect of HCPT. In this paper, we collected reports on HCPT nanomedicines against tumors in the past decade. Based on current research advances, we dissected the current status and limitations of HCPT nanomedicines development and looked forward to future research directions.
Collapse
Affiliation(s)
- Yukun Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Zhenzhi Wang
- Shaanxi University of Chinese Medicine, Xianyang, 712046, People's Republic of China
| | - Xiaofan Wang
- Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People's Republic of China
| | - Mingliang Su
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Fan Xu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Lian Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| |
Collapse
|
22
|
Mitochondrial targeting theranostic nanomedicine and molecular biomarkers for efficient cancer diagnosis and therapy. Biomed Pharmacother 2022; 153:113451. [DOI: 10.1016/j.biopha.2022.113451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
|
23
|
Aliouat H, Peng Y, Waseem Z, Wang S, Zhou W. Pure DNA scaffolded drug delivery systems for cancer therapy. Biomaterials 2022; 285:121532. [DOI: 10.1016/j.biomaterials.2022.121532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/04/2022] [Accepted: 04/15/2022] [Indexed: 02/07/2023]
|
24
|
Al-Mansoori L, Elsinga P, Goda SK. Bio-vehicles of cytotoxic drugs for delivery to tumor specific targets for cancer precision therapy. Biomed Pharmacother 2021; 144:112260. [PMID: 34607105 DOI: 10.1016/j.biopha.2021.112260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/09/2023] Open
Abstract
Abnormal structural and molecular changes in malignant tissues were thoroughly investigated and utilized to target tumor cells, hence rescuing normal healthy tissues and lowering the unwanted side effects as non-specific cytotoxicity. Various ligands for cancer cell specific markers have been uncovered and inspected for directional delivery of the anti-cancer drug to the tumor site, in addition to diagnostic applications. Over the past few decades research related to the ligand targeted therapy (LTT) increased tremendously aiming to treat various pathologies, mainly cancers with well exclusive markers. Malignant tumors are known to induce elevated levels of a variety of proteins and peptides known as cancer "markers" as certain antigens (e.g., Prostate specific membrane antigen "PSMA", carcinoembryonic antigen "CEA"), receptors (folate receptor, somatostatin receptor), integrins (Integrin αvβ3) and cluster of differentiation molecules (CD13). The choice of an appropriate marker to be targeted and the design of effective ligand-drug conjugate all has to be carefully selected to generate the required therapeutic effect. Moreover, since some tumors express aberrantly high levels of more than one marker, some approaches investigated targeting cancer cells with more than one ligand (dual or multi targeting). We aim in this review to report an update on the cancer-specific receptors and the vehicles to deliver cytotoxic drugs, including recent advancements on nano delivery systems and their implementation in targeted cancer therapy. We will discuss the advantages and limitations facing this approach and possible solutions to mitigate these obstacles. To achieve the said aim a literature search in electronic data bases (PubMed and others) using keywords "Cancer specific receptors, cancer specific antibody, tumor specific peptide carriers, cancer overexpressed proteins, gold nanotechnology and gold nanoparticles in cancer treatment" was carried out.
Collapse
Affiliation(s)
- Layla Al-Mansoori
- Qatar University, Biomedical Research Centre, Qatar University, Doha 2713, Qatar.
| | - Philip Elsinga
- University of Groningen, University Medical Center Groningen (UMCG), Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sayed K Goda
- Cairo University, Faculty of Science, Giza, Egypt; University of Derby, College of Science and Engineering, Derby, UK.
| |
Collapse
|
25
|
Liang H, Wu X, Zhao G, Feng K, Ni K, Sun X. Renal Clearable Ultrasmall Single-Crystal Fe Nanoparticles for Highly Selective and Effective Ferroptosis Therapy and Immunotherapy. J Am Chem Soc 2021; 143:15812-15823. [PMID: 34473493 DOI: 10.1021/jacs.1c07471] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Iron-based nanoparticles have attracted much attention because of their ability to induce ferroptosis via a catalyzing Fenton reaction and to further potentiate immunotherapy. However, current iron-based nanoparticles need to be used in cooperation with other treatments or be applied in a high dose for effective therapy because of their low reactive oxygen species production efficacy. Here, we synthesized ultrasmall single-crystal Fe nanoparticles (bcc-USINPs) that stayed stable in a normal physiological environment but were highly active in a tumor microenvironment because of the selective acidic etching of an Fe3O4 shell and the exposure of the Fe(0) core. The bcc-USINPs could efficiently induce tumor cell ferroptosis and immunogenetic cell death at a very low concentration. Intravenous injection of iRGD-bcc-USINPs at three doses of 1 mg/kg could effectively suppress the tumor growth, promote the maturation of dendritic cells, and trigger the adaptive T cell response. Combined with programmed death-ligand 1 (PD-L1) immune checkpoint blockade immunotherapy, the iRGD-bcc-USINP-mediated ferroptosis therapy greatly potentiated the immune response and developed strong immune memory. In addition, these USINPs were quickly renal excreted with no side effects in normal tissues. These iRGD-bcc-USINPs provide a simple, safe, effective, and selectively tumor-responsive Fe(0) delivery system for ferroptosis-based immunotherapy.
Collapse
Affiliation(s)
- Huan Liang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xiyao Wu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Guizhen Zhao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Kai Feng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Kaiyuan Ni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Building 76, Cambridge, Massachusetts 02142, United States
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| |
Collapse
|
26
|
Ilangala AB, Lechanteur A, Fillet M, Piel G. Therapeutic peptides for chemotherapy: Trends and challenges for advanced delivery systems. Eur J Pharm Biopharm 2021; 167:140-158. [PMID: 34311093 DOI: 10.1016/j.ejpb.2021.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/26/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023]
Abstract
The past decades witnessed an increasing interest in peptides as clinical therapeutics. Rightfully considered as a potential alternative for small molecule therapy, these remarkable pharmaceuticals can be structurally fine-tuned to impact properties such as high target affinity, selectivity, low immunogenicity along with satisfactory tissue penetration. Although physicochemical and pharmacokinetic challenges have mitigated, to some extent, the clinical applications of therapeutic peptides, their potential impact on modern healthcare remains encouraging. According to recent reports, there are more than 400 peptides under clinical trials and 60 were already approved for clinical use. As the demand for efficient and safer therapy became high, especially for cancers, peptides have shown some exciting developments not only due to their potent antiproliferative action but also when used as adjuvant therapies, either to decrease side effects with tumor-targeted therapy or to enhance the activity of anticancer drugs via transbarrier delivery. The first part of the present review gives an insight into challenges related to peptide product development. Both molecular and formulation approaches intended to optimize peptide's pharmaceutical properties are covered, and some of their current issues are highlighted. The second part offers a comprehensive overview of the emerging applications of therapeutic peptides in chemotherapy from bioconjugates to nanovectorized therapeutics.
Collapse
Affiliation(s)
- Ange B Ilangala
- Laboratory for the Analysis of Medicines, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium; Laboratory of Pharmaceutical Technology and Biopharmacy, Nanomedicine Development, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium.
| | - Anna Lechanteur
- Laboratory of Pharmaceutical Technology and Biopharmacy, Nanomedicine Development, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, Nanomedicine Development, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium
| |
Collapse
|
27
|
Hawryłkiewicz A, Ptaszyńska N. Gemcitabine Peptide-Based Conjugates and Their Application in Targeted Tumor Therapy. Molecules 2021; 26:E364. [PMID: 33445797 PMCID: PMC7828243 DOI: 10.3390/molecules26020364] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/29/2020] [Accepted: 01/10/2021] [Indexed: 12/25/2022] Open
Abstract
A major obstacle in tumor treatment is associated with the poor penetration of a therapeutic agent into the tumor tissue and with their adverse influence on healthy cells, which limits the dose of drug that can be safely administered to cancer patients. Gemcitabine is an anticancer drug used to treat a wide range of solid tumors and is a first-line treatment for pancreatic cancer. The effect of gemcitabine is significantly weakened by its rapid plasma degradation. In addition, the systemic toxicity and drug resistance significantly reduce its chemotherapeutic efficacy. Up to now, many approaches have been made to improve the therapeutic index of gemcitabine. One of the recently developed approaches to improve conventional chemotherapy is based on the direct targeting of chemotherapeutics to cancer cells using the drug-peptide conjugates. In this work, we summarize recently published gemcitabine peptide-based conjugates and their efficacy in anticancer therapy.
Collapse
Affiliation(s)
| | - Natalia Ptaszyńska
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland;
| |
Collapse
|
28
|
Abstract
Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.
Collapse
|
29
|
Ye H, Yang Y, Chen R, Shi X, Fang Y, Yang J, Dong Y, Chen L, Xia J, Wang C, Yang C, Feng J, Wang Y, Feng X, Lü C. Recognition of Invasive Prostate Cancer Using a GHRL Polypeptide Probe Targeting GHSR in a Mouse Model In Vivo. Curr Pharm Des 2020; 26:1614-1621. [PMID: 31880242 DOI: 10.2174/1381612826666191227160001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ghrelin (GHRL) is a polypeptide that can specifically bind to the growth hormone secretagogue receptor (GHSR). The expression of GHSR is significantly different in normal and prostate cancer (PC) tissues in humans. It is important to find an effective diagnostic method for the diagnosis and prognosis of invasive PC/neuroendocrine prostate cancer (NEPC). METHODS GHRL and GHSR mRNA levels were determined by a quantitative real-time polymerase chain reaction in PC tissues. The expression of GHRL and GHSR proteins was assessed by Western blot assay and immunohistochemistry. A GHRL polypeptide probe was synthesized by standard solid-phase polypeptide synthesis, and labeled with Alexa Fluor 660. Confocal microscopy was used to capture fluorescence images. Living imaging analysis showed tumor areas of different invasiveness in mice models. RESULTS The levels of GHRL and GHSR copy number amplification and mRNA expression were increased in invasive PC/NEPC, and the protein expression levels of GHRL and GHSR were similarly increased in NEPC. The GHRL polypeptide probe could effectively bind to GHSR. In PC3 cells, it was found that the GHRL probe specifically binds to GHSR on the cell membrane and accumulates in the cells through internalization after binding. Live imaging in mice models showed that there were different signal intensities in tumor areas with different invasiveness. CONCLUSION GHSR and GHRL might be used in molecular imaging diagnosis for invasive PC/NEPC in the future.
Collapse
Affiliation(s)
- Huamao Ye
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Yue Yang
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Rui Chen
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Xiaolei Shi
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Yu Fang
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Jun Yang
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Yuanzhen Dong
- State Key Laboratory of Innovative Drugs and Pharmaceutical Technology, Shanghai Institute of Pharmaceutical Industry, Shanghai, China
| | - Lili Chen
- State Key Laboratory of Innovative Drugs and Pharmaceutical Technology, Shanghai Institute of Pharmaceutical Industry, Shanghai, China
| | - Jianghua Xia
- State Key Laboratory of Innovative Drugs and Pharmaceutical Technology, Shanghai Institute of Pharmaceutical Industry, Shanghai, China
| | - Chao Wang
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Chenghua Yang
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Jun Feng
- State Key Laboratory of Innovative Drugs and Pharmaceutical Technology, Shanghai Institute of Pharmaceutical Industry, Shanghai, China
| | - Yang Wang
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Xiang Feng
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Chen Lü
- Department of Urology, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China.,Department of Urology, Shanghai East Hospital Affiliated to Shanghai Tongji University, Shanghai, China
| |
Collapse
|
30
|
Liang H, Guo J, Shi Y, Zhao G, Sun S, Sun X. Porous yolk-shell Fe/Fe 3O 4 nanoparticles with controlled exposure of highly active Fe(0) for cancer therapy. Biomaterials 2020; 268:120530. [PMID: 33296795 DOI: 10.1016/j.biomaterials.2020.120530] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/23/2022]
Abstract
The iron-based Fenton-type reaction has drawn tremendous attention in cancer therapy. Compared with oxidized iron, Fe(0) possesses high catalytic activity but unstable for biomedical application. Here, we report a new strategy to stabilize Fe(0) via a porous yolk shell nanostructure of Fe/Fe3O4 (PYSNPs) in normal physiological condition, and to control the release of Fe(0) in tumor microenvironment for enhanced cancer therapy. These PYSNPs display superior tumor inhibition with the IC50 down to 20 μg/mL (over 1 mg/mL for iron oxide nanoparticles as control) for HepG2 cell. A single intravenous injection of as low as 1 mg/kg dosage is effective to suppress tumor growth in vivo. Moreover, the disintegration of PYSNPs in the acidic tumor microenvironment could cause significant change in MRI signal for contrast-enhanced diagnosis. Of note, the resulting Fe3O4 fragments are renal clearable with minimized side effect. In all, this work represented a nanoplatform to stabilize and selectively deliver Fe(0) for highly effective cancer therapy.
Collapse
Affiliation(s)
- Huan Liang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Jingru Guo
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Yiyue Shi
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Guizhen Zhao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Shouheng Sun
- Department of Chemistry, Brown University, Providence, RI, 02912, USA.
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
31
|
Sarkhosh-Inanlou R, Imani M, Sam MR. The response of PIK3CA/KRAS-mutant colorectal cancer stem-like cells to RGD-peptide FraC produced by the strawberry anemone: A promising water-soluble peptide-based inhibitor of metastasis-driver gene CXCR4, stem cell regulatory genes and self-renewal. Biomed Pharmacother 2020; 132:110807. [PMID: 33068939 DOI: 10.1016/j.biopha.2020.110807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/20/2020] [Accepted: 09/25/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a stem cell-based disease. PIK3CA/KRAS-mutant CRC stem cells (CRCSCs) display high self-renewal, metastatic properties, high activity of PI3K and KRAS signaling pathways with chemoresistant phenotypes. Recently, RGD peptide (containing Arg-Gly-Asp motif)-based therapy of solid tumor cells has attracted much attention. However, little is known whether this method can target self-renewal capacity, key effectors of PI3K and KRAS signaling pathways such as metastasis-driver gene CXCR4 and stem cell regulatory genes with caspase-3 reactivation in CRCSCs overexpressing RGD-dependent integrins. The sea anemone Actinia fragacea produces a water-soluble RGD-peptide fragacea toxin C (FraC) suggesting the possible activity of FraC against PIK3CA/KRAS-mutant CRCSCs. Recombinant FraC was expressed via pET-28a(+)-FraC in E. coli and purified through affinity chromatography followed by performing SDS-PAGE and hemolytic activity assay. Next, PIK3CA/KRAS-mutant HCT-116 cells that serve as an attractive model for CRCSCs were treated with FraC. Thereafter, cell numbers, viability, proliferation, LDH activity, cytotoxicity index, CXCR4 and pluripotency network genes expression, self-renewal capacity, caspase-3 activity with apoptosis were evaluated. Caspase-1, -2, -3,…, -9 sequences were analyzed for RGD-binding motifs. FraC sequence and structure were also evaluated by bioinformatics software. FraC altered cellular morphology to round shapes and disrupted cell connections. 48 h post-treatment with 0.056- to 7.2 μM FraC resulted in 12 %-99 % and 8 %-97.6 % decreases in cell numbers and viabilities respectively and increased LDH activity by 0.2 %-66.7 % in a dose-dependent manner. The results of the cytotoxicity index showed that FraC induces significant toxicity on HCT-116 cells compared to PBMCs and Huvec cells. FraC dramatically decreased the expression of CXCR4 and pluripotency network genes Bmi-1, Sox-2, Oct-4 and Nanog followed by remarkable decreases in self-renewal capacity ranged from 91- to 0 colonies per well for 0.056- to 3.6 μM FraC after 2 weeks. Caspase-3 was found to contain an RGD-binding motif and its activity increased with increasing FraC concentrations followed by apoptosis induction. Potential RGD-binding motifs for FraC were also found in caspase-1, -7, -8 and -9. Unique advantages of FraC peptide, such as low molecular weight, water solubility, high sensitivity of CRC stem-like cells with more selective toxicity to this compound, targeting tumor cell membrane and self-renewal capacity along with the modulation of CXCR4 and stem cell regulatory genes as upstream and downstream effectors of undruggable PI3K and KRAS signaling pathways may open up avenues for FraC peptide-based therapy of PIK3CA/KRAS-mutant CRCSCs with lower toxicity on healthy cells.
Collapse
Affiliation(s)
- Roya Sarkhosh-Inanlou
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran.
| | - Mehdi Imani
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran; Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Mohammad Reza Sam
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran; Department of Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran; Department of Biotechnology, Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran.
| |
Collapse
|
32
|
Kang S, Lee S, Park S. iRGD Peptide as a Tumor-Penetrating Enhancer for Tumor-Targeted Drug Delivery. Polymers (Basel) 2020; 12:E1906. [PMID: 32847045 PMCID: PMC7563641 DOI: 10.3390/polym12091906] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
The unique structure and physiology of a tumor microenvironment impede intra-tumoral penetration of chemotherapeutic agents. A novel iRGD peptide that exploits the tumor microenvironment can activate integrin-dependent binding to tumor vasculatures and neuropilin-1 (NRP-1)-dependent transport to tumor tissues. Recent studies have focused on its dual-targeting ability to achieve enhanced penetration of chemotherapeutics for the efficient eradication of cancer cells. Both the covalent conjugation and the co-administration of iRGD with chemotherapeutic agents and engineered delivery vehicles have been explored. Interestingly, the iRGD-mediated drug delivery also enhances penetration through the blood-brain barrier (BBB). Recent studies have shown its synergistic effect with BBB disruptive techniques. The efficacy of immunotherapy involving immune checkpoint blockades has also been amplified by using iRGD as a targeting moiety. In this review, we presented the recent advances in iRGD technology, focusing on cancer treatment modalities, including the current clinical trials using iRGD. The iRGD-mediated nano-carrier system could serve as a promising strategy in drug delivery to the deeper tumor regions, and be combined with various therapeutic interventions due to its novel targeting ability.
Collapse
Affiliation(s)
| | | | - Soyeun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea; (S.K.); (S.L.)
| |
Collapse
|
33
|
Confeld MI, Mamnoon B, Feng L, Jensen-Smith H, Ray P, Froberg J, Kim J, Hollingsworth MA, Quadir M, Choi Y, Mallik S. Targeting the Tumor Core: Hypoxia-Responsive Nanoparticles for the Delivery of Chemotherapy to Pancreatic Tumors. Mol Pharm 2020; 17:2849-2863. [PMID: 32521162 DOI: 10.1021/acs.molpharmaceut.0c00247] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), early onset of hypoxia triggers remodeling of the extracellular matrix, epithelial-to-mesenchymal transition, increased cell survival, the formation of cancer stem cells, and drug resistance. Hypoxia in PDAC is also associated with the development of collagen-rich, fibrous extracellular stroma (desmoplasia), resulting in severely impaired drug penetration. To overcome these daunting challenges, we created polymer nanoparticles (polymersomes) that target and penetrate pancreatic tumors, reach the hypoxic niches, undergo rapid structural destabilization, and release the encapsulated drugs. In vitro studies indicated a high cellular uptake of the polymersomes and increased cytotoxicity of the drugs under hypoxia compared to unencapsulated drugs. The polymersomes decreased tumor growth by nearly 250% and significantly increased necrosis within the tumors by 60% in mice compared to untreated controls. We anticipate that these polymer nanoparticles possess a considerable translational potential for delivering drugs to solid hypoxic tumors.
Collapse
Affiliation(s)
- Matthew I Confeld
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Babak Mamnoon
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Li Feng
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Heather Jensen-Smith
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Priyanka Ray
- Coatings and Polymeric Materials Department, North Dakota State University, Fargo, North Dakota 58108, United States
| | - James Froberg
- Physics Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Michael A Hollingsworth
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Mohiuddin Quadir
- Coatings and Polymeric Materials Department, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Yongki Choi
- Physics Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Sanku Mallik
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
34
|
De Angelis B, Depalo N, Petronella F, Quintarelli C, Curri ML, Pani R, Calogero A, Locatelli F, De Sio L. Stimuli-responsive nanoparticle-assisted immunotherapy: a new weapon against solid tumours. J Mater Chem B 2020; 8:1823-1840. [DOI: 10.1039/c9tb02246e] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The interplay between photo-thermal therapy and immunotherapy allows the realization of new nanotechnology-based cancer treatments for solid tumors.
Collapse
Affiliation(s)
- Biagio De Angelis
- Department of Onco-Haematology and Cell and Gene Therapy
- Bambino Gesù Children's Hospital
- IRCCS
- Rome
- Italy
| | - Nicoletta Depalo
- CNR-IPCF
- National Research Council of Italy
- Institute for Physical and Chemical Processes-Bari Division
- I-70126 Bari
- Italy
| | - Francesca Petronella
- CNR-IC
- National Research Council of Italy
- Institute Crystallography
- 00015 Monterotondo – Rome
- Italy
| | - Concetta Quintarelli
- Department of Onco-Haematology and Cell and Gene Therapy
- Bambino Gesù Children's Hospital
- IRCCS
- Rome
- Italy
| | - M. Lucia Curri
- CNR-IPCF
- National Research Council of Italy
- Institute for Physical and Chemical Processes-Bari Division
- I-70126 Bari
- Italy
| | - Roberto Pani
- Center for Biophotonics and Department of Medico-surgical Sciences and Biotechnologies
- Sapienza University of Rome
- Latina
- Italy
| | - Antonella Calogero
- Center for Biophotonics and Department of Medico-surgical Sciences and Biotechnologies
- Sapienza University of Rome
- Latina
- Italy
| | - Franco Locatelli
- Department of Onco-Haematology and Cell and Gene Therapy
- Bambino Gesù Children's Hospital
- IRCCS
- Rome
- Italy
| | - Luciano De Sio
- Center for Biophotonics and Department of Medico-surgical Sciences and Biotechnologies
- Sapienza University of Rome
- Latina
- Italy
| |
Collapse
|
35
|
Arrighetti N, Corbo C, Evangelopoulos M, Pastò A, Zuco V, Tasciotti E. Exosome-like Nanovectors for Drug Delivery in Cancer. Curr Med Chem 2019; 26:6132-6148. [PMID: 30182846 DOI: 10.2174/0929867325666180831150259] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/20/2018] [Accepted: 07/04/2018] [Indexed: 02/07/2023]
Abstract
Cancer treatment still represents a formidable challenge, despite substantial advancements in available therapies being made over the past decade. One major issue is poor therapeutic efficacy due to lack of specificity and low bioavailability. The progress of nanotechnology and the development of a variety of nanoplatforms have had a significant impact in improving the therapeutic outcome of chemotherapeutics. Nanoparticles can overcome various biological barriers and localize at tumor site, while simultaneously protecting a therapeutic cargo and increasing its circulation time. Despite this, due to their synthetic origin, nanoparticles are often detected by the immune system and preferentially sequestered by filtering organs. Exosomes have recently been investigated as suitable substitutes for the shortcomings of nanoparticles due to their biological compatibility and particularly small size (i.e., 30-150 nm). In addition, exosomes have been found to play important roles in cell communication, acting as natural carriers of biological cargoes throughout the body. This review aims to highlight the use of exosomes as drug delivery vehicles for cancer and showcases the various attempts used to exploit exosomes with a focus on the delivery of chemotherapeutics and nucleic acids.
Collapse
Affiliation(s)
- Noemi Arrighetti
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Claudia Corbo
- Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, United States
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, United States
| | - Anna Pastò
- Istituto Oncologico Veneto-IRCCS, Padova, Italy
| | - Valentina Zuco
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, United States.,Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, United States
| |
Collapse
|
36
|
Xiao W, Zhang W, Huang H, Xie Y, Zhang Y, Guo X, Jin C, Liao X, Yao S, Chen G, Song X. Cancer Targeted Gene Therapy for Inhibition of Melanoma Lung Metastasis with eIF3i shRNA Loaded Liposomes. Mol Pharm 2019; 17:229-238. [PMID: 31765158 DOI: 10.1021/acs.molpharmaceut.9b00943] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Eukaryotic translation initiation factors 3i (eIF3i) is a proto-oncogene that is overexpressed in various tumors, reducing its expression by eIF3i shRNA is a promising strategy to inhibit tumor growth or metastasis. Tumor cell is the target of eIF3i shRNA so that tumor-site accumulation could be important for fulfilling its therapeutic effect. Thus, the iRGD modified liposome (R-LP) was rationally synthesized to enhance the antitumor effect by active targeted delivery of eIF3i shRNA to B16F10 melanoma cells. R-LP encapsulating eIF3i shRNA gene (R-LP/sheIF3i) were prepared by a film dispersion method. The transfection experiment proves that R-LP could effectively transfect B16F10 cells. R-LP/sheIF3i notably restrained the migration, invasion, and adhesion of melanoma cells in vitro. In a mouse model of lung metastasis, R-LP/sheIF3i administered by intravenous injection suppressed pulmonary metastasis of melanoma by dramatically downregulated eIF3i expression and subsequently inhibiting tumor neovascularization and tumor cells proliferation in vivo. Our results provide a basis for tumor cells targeting strategies to reduce the expression of eIF3i by RNAi in the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Wen Xiao
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Weiyi Zhang
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Hai Huang
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Yafei Xie
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Yi Zhang
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Xia Guo
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Chaohui Jin
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Xuelian Liao
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Shaohua Yao
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Guo Chen
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Xiangrong Song
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| |
Collapse
|
37
|
Shreffler JW, Pullan JE, Dailey KM, Mallik S, Brooks AE. Overcoming Hurdles in Nanoparticle Clinical Translation: The Influence of Experimental Design and Surface Modification. Int J Mol Sci 2019; 20:E6056. [PMID: 31801303 PMCID: PMC6928924 DOI: 10.3390/ijms20236056] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/23/2019] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles are becoming an increasingly popular tool for biomedical imaging and drug delivery. While the prevalence of nanoparticle drug-delivery systems reported in the literature increases yearly, relatively little translation from the bench to the bedside has occurred. It is crucial for the scientific community to recognize this shortcoming and re-evaluate standard practices in the field, to increase clinical translatability. Currently, nanoparticle drug-delivery systems are designed to increase circulation, target disease states, enhance retention in diseased tissues, and provide targeted payload release. To manage these demands, the surface of the particle is often modified with a variety of chemical and biological moieties, including PEG, tumor targeting peptides, and environmentally responsive linkers. Regardless of the surface modifications, the nano-bio interface, which is mediated by opsonization and the protein corona, often remains problematic. While fabrication and assessment techniques for nanoparticles have seen continued advances, a thorough evaluation of the particle's interaction with the immune system has lagged behind, seemingly taking a backseat to particle characterization. This review explores current limitations in the evaluation of surface-modified nanoparticle biocompatibility and in vivo model selection, suggesting a promising standardized pathway to clinical translation.
Collapse
Affiliation(s)
| | | | | | | | - Amanda E. Brooks
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (J.W.S.); (J.E.P.); (K.M.D.); (S.M.)
| |
Collapse
|
38
|
Prajapati SK, Jain A, Jain A, Jain S. Biodegradable polymers and constructs: A novel approach in drug delivery. Eur Polym J 2019; 120:109191. [DOI: 10.1016/j.eurpolymj.2019.08.018] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Blum AP, Nelles DA, Hidalgo FJ, Touve MA, Sim DS, Madrigal AA, Yeo GW, Gianneschi NC. Peptide Brush Polymers for Efficient Delivery of a Gene Editing Protein to Stem Cells. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201904894] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Angela P. Blum
- Department of Chemistry and Biochemistry University of California, San Diego La Jolla CA USA
- Departments of Chemistry Hamilton College Clinton NY USA
| | - David A. Nelles
- Department of Cellular and Molecular Medicine, Stem Cell Program Institute of Genomic Medicine University of California, San Diego La Jolla CA USA
| | - Francisco J. Hidalgo
- Department of Chemistry and Biochemistry University of California, San Diego La Jolla CA USA
| | - Mollie A. Touve
- Departments of Chemistry Materials Science & Engineering Biomedical Engineering International Institute for Nanotechnology Northwestern University Evanston IL USA
| | - Deborah S. Sim
- Departments of Chemistry Hamilton College Clinton NY USA
| | - Assael A. Madrigal
- Department of Cellular and Molecular Medicine, Stem Cell Program Institute of Genomic Medicine University of California, San Diego La Jolla CA USA
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, Stem Cell Program Institute of Genomic Medicine University of California, San Diego La Jolla CA USA
- Molecular Engineering Laboratory A*STAR Singapore Singapore
| | - Nathan C. Gianneschi
- Department of Chemistry and Biochemistry University of California, San Diego La Jolla CA USA
- Departments of Chemistry Materials Science & Engineering Biomedical Engineering International Institute for Nanotechnology Northwestern University Evanston IL USA
| |
Collapse
|
40
|
Blum AP, Nelles DA, Hidalgo FJ, Touve MA, Sim DS, Madrigal AA, Yeo GW, Gianneschi NC. Peptide Brush Polymers for Efficient Delivery of a Gene Editing Protein to Stem Cells. Angew Chem Int Ed Engl 2019; 58:15646-15649. [DOI: 10.1002/anie.201904894] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Angela P. Blum
- Department of Chemistry and Biochemistry University of California, San Diego La Jolla CA USA
- Departments of Chemistry Hamilton College Clinton NY USA
| | - David A. Nelles
- Department of Cellular and Molecular Medicine, Stem Cell Program Institute of Genomic Medicine University of California, San Diego La Jolla CA USA
| | - Francisco J. Hidalgo
- Department of Chemistry and Biochemistry University of California, San Diego La Jolla CA USA
| | - Mollie A. Touve
- Departments of Chemistry Materials Science & Engineering Biomedical Engineering International Institute for Nanotechnology Northwestern University Evanston IL USA
| | - Deborah S. Sim
- Departments of Chemistry Hamilton College Clinton NY USA
| | - Assael A. Madrigal
- Department of Cellular and Molecular Medicine, Stem Cell Program Institute of Genomic Medicine University of California, San Diego La Jolla CA USA
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, Stem Cell Program Institute of Genomic Medicine University of California, San Diego La Jolla CA USA
- Molecular Engineering Laboratory A*STAR Singapore Singapore
| | - Nathan C. Gianneschi
- Department of Chemistry and Biochemistry University of California, San Diego La Jolla CA USA
- Departments of Chemistry Materials Science & Engineering Biomedical Engineering International Institute for Nanotechnology Northwestern University Evanston IL USA
| |
Collapse
|
41
|
Wu D, Daly HC, Grossi M, Conroy E, Li B, Gallagher WM, Elmes R, O'Shea DF. RGD conjugated cell uptake off to on responsive NIR-AZA fluorophores: applications toward intraoperative fluorescence guided surgery. Chem Sci 2019; 10:6944-6956. [PMID: 31588261 PMCID: PMC6686729 DOI: 10.1039/c9sc02197c] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
The use of NIR-fluorescence imaging to demarcate tumour boundaries for real-time guidance of their surgical resection has a huge untapped potential. However, fluorescence imaging using molecular fluorophores, even with a targeting biomolecule attached, has a major shortcoming of signal interference from non-specific background fluorescence outside the region of interest. This poor selectivity necessitates prolonged time delays to allow clearance of background fluorophore and retention within the tumour prior to image acquisition. In this report, an innovative approach to overcome this issue is described in which cancer targeted off to on bio-responsive NIR-fluorophores are utilised to switch-on first within the tumour. Bio-responsive cRGD, iRGD and PEG conjugates have been synthesised using activated ester/amine or maleimide/thiol couplings to link targeting and fluorophore components. Their off to on emission responses were measured and compared with an always-on non-responsive control with each bio-responsive derivative showing large fluorescence enhancement values. Live cell imaging experiments using metastatic breast cancer cells confirmed in vitro bio-responsive capabilities. An in vivo assessment of MDA-MB 231 tumour imaging performance for bio-responsive and always-on fluorophores was conducted with monitoring of fluorescence distributions over 96 h. As anticipated, the always-on fluorophore gave an immediate, non-specific and very strong emission throughout whereas the bio-responsive derivatives initially displayed very low fluorescence. All three bio-responsive derivatives switched on within tumours at time points consistent with their conjugated targeting groups. cRGD and iRGD conjugates both had effective tumour turn-on in the first hour, though the cRGD derivative had superior specificity for tumour over the iRGD conjugate. The pegylated derivative had similar switch-on characteristics but over a much longer period, taking 9 h before a significant emission was observable from the tumour. Evidence for in vivo active tumour targeting was obtained for the best performing cRGD bio-responsive NIR-AZA derivative from competitive binding studies. Overall, this cRGD-conjugate has the potential to overcome the inherent drawback of targeted always-on fluorophores requiring prolonged clearance times and shows excellent potential for clinical translation for intraoperative use in fluorescence guided tumour resections.
Collapse
Affiliation(s)
- Dan Wu
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| | - Harrison C Daly
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| | - Marco Grossi
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| | - Emer Conroy
- School of Biomolecular and Biomedical Science , Conway Institute, University College Dublin , Belfield , Dublin 4 , Ireland
| | - Bo Li
- School of Biomolecular and Biomedical Science , Conway Institute, University College Dublin , Belfield , Dublin 4 , Ireland
| | - William M Gallagher
- School of Biomolecular and Biomedical Science , Conway Institute, University College Dublin , Belfield , Dublin 4 , Ireland
| | - Robert Elmes
- Department of Chemistry , Maynooth University Human Health Institute , Maynooth University , Maynooth , Ireland
| | - Donal F O'Shea
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| |
Collapse
|
42
|
E M Eid E, S Alanazi A, Koosha S, A Alrasheedy A, Azam F, M Taban I, Khalilullah H, Sadiq Al-Qubaisi M, A Alshawsh M. Zerumbone Induces Apoptosis in Breast Cancer Cells by Targeting αvβ3 Integrin upon Co-Administration with TP5-iRGD Peptide. Molecules 2019; 24:molecules24142554. [PMID: 31337024 PMCID: PMC6680663 DOI: 10.3390/molecules24142554] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 12/24/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are highly promising tools to deliver therapeutic molecules into tumours. αVβ3 integrins are cell-matrix adhesion receptors, and are considered as an attractive target for anticancer therapies owing to their roles in the process of metastasis and angiogenesis. Therefore, this study aims to assess the effect of co-administration of zerumbone (ZER) and ZERencapsulated in hydroxypropyl-β-cyclodextrin with TP5-iRGD peptide towards cell cytotoxicity, apoptosis induction, and proliferation of normal and cancerous breast cells utilizing in vitro assays, as well as to study the molecular docking of ZER in complex with TP5-iRGD peptide. Cell viability assay findings indicated that ZER and ZERencapsulated in hydroxypropyl-β-cyclodextrin (ZER-HPβCD) inhibited the growth of estrogen receptor positivebreast cancer cells (ER+ MCF-7) at 72 h treatment with an inhibitory concentration (IC)50 of 7.51 ± 0.2 and 5.08 ± 0.2 µg/mL, respectively, and inhibited the growth of triple negative breast cancer cells (MDA-MB-231) with an IC50 of 14.96 ± 1.52 µg/mL and 12.18 ± 0.7 µg/mL, respectively. On the other hand, TP5-iRGD peptide showed no significant cytotoxicity on both cancer and normal cells. Interestingly, co-administration of TP5-iRGD peptide in MCF-7 cells reduced the IC50 of ZER from 7.51 ± 0.2 µg/mL to 3.13 ± 0.7 µg/mL and reduced the IC50 of ZER-HPβCD from 5.08 ± 0.2 µg/mL to 0.49 ± 0.004 µg/mL, indicating that the co-administration enhances the potency and increases the efficacy of ZER and ZER-HPβCD compounds. Acridine orange (AO)/propidium iodide (PI) staining under fluorescence microscopy showed evidence of early apoptosis after 72 h from the co-administration of ZER or ZER-HPβCD with TP5-iRGD peptide in MCF-7 breast cancer cells. The findings of the computational modelling experiment provide novel insights into the ZER interaction with integrin αvβ3 in the presence of TP5-iRGD, and this could explain why ZER has better antitumor activities when co-administered with TP5-iRGD peptide.
Collapse
Affiliation(s)
- Eltayeb E M Eid
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia.
| | | | - Sanaz Koosha
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Alian A Alrasheedy
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia
| | - Ismail M Taban
- School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK
| | - Habibullah Khalilullah
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia
| | | | - Mohammed A Alshawsh
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
43
|
Zhou M, Huang H, Wang D, Lu H, Chen J, Chai Z, Yao SQ, Hu Y. Light-Triggered PEGylation/dePEGylation of the Nanocarriers for Enhanced Tumor Penetration. NANO LETTERS 2019; 19:3671-3675. [PMID: 31062980 DOI: 10.1021/acs.nanolett.9b00737] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Nanocarriers-derived anticancer therapeutics typically suffers from poor tumor penetration and suboptimal antitumor efficacy. Although PEGylation improves the stability of nanoparticles and prolongs drug circulation, it further increases the size of nanoparticles and adversely affects the tumor penetration. Here, we developed a light-triggered PEGylation/dePEGylation strategy, whereby near-infrared (NIR)-/pH- dual responsive dePEGylation activates iRGD for tumor targeting. The embedded up-conversion nanoparticles (UCNPs) could efficiently convert NIR to UV-vis which cleaved the linker to remove PEG. NIR-induced dePEGylation remarkably improved vascular extravasation of drugs and deep tumor penetration. Therefore, the stimuli-responsive nanocarriers facilitated the tumor-targeted delivery of drugs through blood circulation and enhanced the antitumor effects.
Collapse
Affiliation(s)
- Mengxue Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS) , Chinese Academy of Sciences (CAS) , Beijing 100049 , P.R. China
| | - Hui Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS) , Chinese Academy of Sciences (CAS) , Beijing 100049 , P.R. China
| | - Dongqing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS) , Chinese Academy of Sciences (CAS) , Beijing 100049 , P.R. China
| | - Huiru Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS) , Chinese Academy of Sciences (CAS) , Beijing 100049 , P.R. China
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS) , Chinese Academy of Sciences (CAS) , Beijing 100049 , P.R. China
| | - Zhifang Chai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS) , Chinese Academy of Sciences (CAS) , Beijing 100049 , P.R. China
| | - Shao Q Yao
- Department of Chemistry , National University of Singapore , Singapore 117543
| | - Yi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS) , Chinese Academy of Sciences (CAS) , Beijing 100049 , P.R. China
| |
Collapse
|
44
|
Napolitano V, Tamagnone L. Neuropilins Controlling Cancer Therapy Responsiveness. Int J Mol Sci 2019; 20:ijms20082049. [PMID: 31027288 PMCID: PMC6515012 DOI: 10.3390/ijms20082049] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/17/2022] Open
Abstract
Neuropilins (NRPs) are cell surface glycoproteins, acting as co-receptors for secreted Semaphorins (SEMAs) and for members of the vascular endothelial growth factor (VEGF) family; they have been initially implicated in axon guidance and angiogenesis regulation, and more recently in cancer progression. In addition, NRPs have been shown to control many other fundamental signaling pathways, especially mediated by tyrosine kinase receptors (RTKs) of growth factors, such as HGF (hepatocyte growth factor), PDGF (platelet derived growth factor) and EGF (epidermal growth factor). This enables NRPs to control a range of pivotal mechanisms in the cancer context, from tumor cell proliferation and metastatic dissemination, to tumor angiogenesis and immune escape. Moreover, cancer treatment failures due to resistance to innovative oncogene-targeted drugs is typically associated with the activity of alternative RTK-dependent pathways; and neuropilins’ capacity to control oncogenic signaling cascades supports the hypothesis that they could elicit such mechanisms in cancer cells, in order to escape cytotoxic stress and therapeutic attacks. Intriguingly, several studies have recently assayed the impact of NRPs inhibition in combination with diverse anti-cancer drugs. In this minireview, we will discuss the state-of-art about the relevance of NRPs as potential predictive biomarkers of drug response, and the rationale to target these proteins in combination with other anticancer therapies.
Collapse
Affiliation(s)
- Virginia Napolitano
- Cancer Cell Biology Laboratory, Candiolo Cancer Institute-FPO, IRCCS, 10060 Candiolo, Italy.
| | - Luca Tamagnone
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, 10168 Rome, Italy.
- Fondazione Policlinico Universitario Agostino Gemelli, 10168 Rome, Italy.
| |
Collapse
|
45
|
Cho HJ, Park SJ, Lee YS, Kim S. Theranostic iRGD peptide containing cisplatin prodrug: Dual-cargo tumor penetration for improved imaging and therapy. J Control Release 2019; 300:73-80. [PMID: 30831135 DOI: 10.1016/j.jconrel.2019.02.043] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/07/2019] [Accepted: 02/28/2019] [Indexed: 01/30/2023]
Abstract
In theranostics, peptide-based platforms have widely been exploited owing to their unique biological functions and chemical versatilities. As a tumor-homing ligand, internalizing RGD peptide (iRGD), composed of a tumor-targeting sequence (RGD) and a cell-penetrating C-end Rule (CendR) motif, is known to facilitate the tumor-specific delivery of payloads that are covalently conjugated on its N-terminal fragment or co-administered without any covalent linkages. However, theranostic uses of the iRGD-based platform remain in its infancy with its full potential unexplored; for instance, detailed mechanism of iRGD fragmentation during internalization, strategies for the tumor-specific release of payloads from iRGD and the role of the C-terminal iRGD fragment in delivery have yet to be revealed. In this study, we designed a dual-channel fluorescent cyclic iRGD (TAMRA-iRGDC-Cy5.5) to track each of the N- and C-terminal fragments separately during the tumor internalization process. It turned out that both fragments undergo translocation into cancer cells together and are localized within endosomal-lysosomal compartments. The resulting co-internalization of both iRGD fragments allowed us to develop a new theranostic peptide platform (Cy5.5-iRGDC-Pt(IV)) by conjugating a fluorescent dye and a cisplatin prodrug on each terminus of cyclic iRGD for simultaneous cancer-targeted imaging and therapy. Compared to a control peptide having a non-iRGD sequence, the Cy5.5-iRGDC-Pt(IV) did not only provide a better tumor imaging contrast but also induced tumor-specific apoptosis leading to efficacious tumor suppression. Besides the outstanding cancer imaging and therapeutic performance, the Cy5.5-iRGDC-Pt(IV) revealed negligible systemic toxicity, holding potential to be applied for theranostic uses.
Collapse
Affiliation(s)
- Hong-Jun Cho
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sung-Jun Park
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea; School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon-Sik Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehoon Kim
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
46
|
Feng S, Wu ZX, Zhao Z, Liu J, Sun K, Guo C, Wang H, Wu Z. Engineering of Bone- and CD44-Dual-Targeting Redox-Sensitive Liposomes for the Treatment of Orthotopic Osteosarcoma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:7357-7368. [PMID: 30682240 DOI: 10.1021/acsami.8b18820] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This study aimed to develop an efficient step-by-step osteosarcoma (OS)-targeting liposome system functionalized with a redox-cleavable, bone- and cluster of differentiation 44 (CD44)-dual-targeting polymer. Furthermore, the effect of coadministration of a tumor-penetrating peptide, internalizing RGD (iRGD), was investigated. First, a bone-targeting moiety, alendronate (ALN), was conjugated with hyaluronic acid (HA), a ligand for CD44. This ALN-HA conjugate was coupled with DSPE-PEG2000-COOH through a bioreducible disulfide linker (-SS-) to obtain a functionalized lipid, ALN-HA-SS-L, to be postinserted into preformed liposomes loaded with doxorubicin (DOX). The roles of ALN, HA, and the redox sensitivity of the ALN-HA-SS-L liposomes (ALN-HA-SS-L-L) in the anti-OS effect were critically evaluated against various reference liposomal formulations (with only ALN, HA, or redox sensitivity). ALN-HA-SS-L-L displayed a zeta potential of -26.07 ± 0.32 mV and selectively disassembled in the presence of a reducing agent, 10 mM glutathione, which can be found in cancer cells. Compared to various reference liposomes, ALN-HA-SS-L-L/DOX had significantly higher cytotoxicity to human OS MG-63 cells alongside high and rapid cellular uptake. In the orthotopic OS nude mouse models, ALN-HA-SS-L-L/DOX showed remarkable tumor growth suppression and prolonged survival time. This result was further improved by the coadministration of iRGD. The antitumor effects of various liposomes were ranked in the same order as the degree of tumor biodistribution shown by in vivo/ex vivo imaging: ALN-HA-SS-L-L coadministered with iRGD > ALN-HA-SS-L-L > HA-SS-L-L > HA-L-L > PEG-L> free drug. ALN-HA-SS-L-L/DOX also reduced the cardiotoxicity of DOX and lung metastases. Overall, this study demonstrated that ALN-HA-SS-L-L/DOX, equipped with bone- and CD44-dual-targeting abilities and redox sensitivity, could be a promising OS-targeted therapy. The efficacy could also be augmented by coadministration of iRGD.
Collapse
Affiliation(s)
- Shuaishuai Feng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in University of Shandong , Yantai University , Yantai 264005 , PR China
| | - Zi-Xin Wu
- Qingdao Municipal Hospital , Qingdao 266071 Shandong Province , PR China
| | - Ziyan Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in University of Shandong , Yantai University , Yantai 264005 , PR China
| | - Jinhu Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in University of Shandong , Yantai University , Yantai 264005 , PR China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in University of Shandong , Yantai University , Yantai 264005 , PR China
| | - Chuanyou Guo
- Qingdao Municipal Hospital , Qingdao 266071 Shandong Province , PR China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in University of Shandong , Yantai University , Yantai 264005 , PR China
| | - Zimei Wu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in University of Shandong , Yantai University , Yantai 264005 , PR China
- School of Pharmacy , University of Auckland , Auckland 1142 , New Zealand
| |
Collapse
|
47
|
Yang J, Yin H, Yang J, Wei Y, Fang L, Chai D, Zhang Q, Zheng J. Tumor-Penetrating Peptide Enhances Antitumor Effects of IL-24 Against Prostate Cancer. Transl Oncol 2018; 12:453-461. [PMID: 30580153 PMCID: PMC6302246 DOI: 10.1016/j.tranon.2018.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022] Open
Abstract
The interleukin-24 (IL-24), a member of the IL-10-related cytokine gene family, is well known for its tumor suppressor activity in a broad spectrum of human tumors without damaging normal cells. However, poor tumor penetration remains a key problem for the efficacy of IL-24 as a treatment. iRGD is a novel tumor-specific peptide with unique tumor-penetrating and cell-internalizing properties. To enhance the tumor-penetrating and antitumor effects of IL-24, we engineered a recombinant protein consisting of the IL-24 fused to iRGD, which was named IL-24-iRGD. The aim of the present study was to investigate the antitumor effects of IL-24-iRGD in prostate cancer cells in vitro and in vivo. It was observed that IL-24-iRGD induced cell apoptosis, suppressed cell growth of PC-3 in vitro, and promoted protein penetration into tumors in vivo, whereas it had no effect on normal cell line RWPE-1. Then, PC-3 cells were subcutaneously injected into nude mice, and these tumor-bearing mice were administered with IL-24, IL-24-iRGD, or PBS via the tail vein. The IL-24- and IL-24-iRGD-treated groups exhibited tumor growth inhibition rates of 38.6% and 65.6%, respectively, when compared with the PBS-treated group. Besides, cell apoptosis was examined by TdT-mediated dUTP nick end labeling, and the expression of cleaved caspase-3 was analyzed by immunohistochemical staining. The results demonstrated that IL-24-iRGD induced apoptosis and inhibited the growth of PC-3 cells to a significantly greater extent when compared with IL-24 treatment alone. It may provide an improved strategy for antitumor therapy and the clinical treatment of prostate cancer.
Collapse
Affiliation(s)
- Jie Yang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Hong Yin
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; Center of Radiotherapy of The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Jie Yang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; Center of Radiotherapy of The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Yanhong Wei
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; Center of Cancer of The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China.
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; Center of Radiotherapy of The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
48
|
Umeshappa CS, Shao K. Comment on "Coadministration of iRGD with Multistage Responsive Nanoparticles Enhanced Tumor Targeting and Penetration Abilities for Breast Cancer Therapy". ACS APPLIED MATERIALS & INTERFACES 2018; 10:38659-38662. [PMID: 30360098 DOI: 10.1021/acsami.8b11346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanomedicine is at the forefront of targeted drug delivery for cancer therapy. An improved combinatorial approach is highlighted for breast cancer treatment by Hu et al. in this issue of ACS Applied Materials and Interfaces. The authors demonstrated that, by combining multistage-responsive nanoparticles carrying a therapeutic drug, doxorubicin, a photothermal agent, indocyanine green, and a nitric oxide donor with photothermal therapy and intravenous injection of a tumor-homing iRGD peptide, one could achieve efficient therapeutics distribution deep inside the tumor and nearly eradicate primary tumor growth. An in-depth understanding of this approach in combination with other strategies such as the use of immunomodulators would facilitate treating metastasis in distant organs, and clinical translation of this platform, benefiting cancer patients by providing long-lasting efficacy.
Collapse
Affiliation(s)
- Channakeshava Sokke Umeshappa
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Kun Shao
- State Key Laboratory of Fine Chemicals , Dalian University of Technology , No 2 Linggong Road , Dalian 116024 , China
| |
Collapse
|
49
|
Zhang F, Stephan SB, Ene CI, Smith TT, Holland EC, Stephan MT. Nanoparticles That Reshape the Tumor Milieu Create a Therapeutic Window for Effective T-cell Therapy in Solid Malignancies. Cancer Res 2018; 78:3718-3730. [PMID: 29760047 DOI: 10.1158/0008-5472.can-18-0306] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/28/2018] [Accepted: 05/09/2018] [Indexed: 12/31/2022]
Abstract
A major obstacle to the success rate of chimeric antigen receptor (CAR-) T-cell therapy against solid tumors is the microenvironment antagonistic to T cells that solid tumors create. Conventional checkpoint blockade can silence lymphocyte antisurvival pathways activated by tumors, but because they are systemic, these treatments disrupt immune homeostasis and induce autoimmune side effects. Thus, new technologies are required to remodel the tumor milieu without causing systemic toxicities. Here, we demonstrate that targeted nanocarriers that deliver a combination of immune-modulatory agents can remove protumor cell populations and simultaneously stimulate antitumor effector cells. We administered repeated infusions of lipid nanoparticles coated with the tumor-targeting peptide iRGD and loaded with a combination of a PI3K inhibitor to inhibit immune-suppressive tumor cells and an α-GalCer agonist of therapeutic T cells to synergistically sway the tumor microenvironment of solid tumors from suppressive to stimulatory. This treatment created a therapeutic window of 2 weeks, enabling tumor-specific CAR-T cells to home to the lesion, undergo robust expansion, and trigger tumor regression. CAR-T cells administered outside this therapeutic window had no curative effect. The lipid nanoparticles we used are easy to manufacture in substantial amounts, and we demonstrate that repeated infusions of them are safe. Our technology may therefore provide a practical and low-cost strategy to potentiate many cancer immunotherapies used to treat solid tumors, including T-cell therapy, vaccines, and BITE platforms.Significance: A new nanotechnology approach can promote T-cell therapy for solid tumors. Cancer Res; 78(13); 3718-30. ©2018 AACR.
Collapse
Affiliation(s)
- Fan Zhang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sirkka B Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Chibawanye I Ene
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington
| | - Tyrel T Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Eric C Holland
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington.,Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Alvord Brain Tumor Center, University of Washington, Seattle, Washington
| | - Matthias T Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington.,Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, Washington
| |
Collapse
|
50
|
Amreddy N, Babu A, Muralidharan R, Panneerselvam J, Srivastava A, Ahmed R, Mehta M, Munshi A, Ramesh R. Recent Advances in Nanoparticle-Based Cancer Drug and Gene Delivery. Adv Cancer Res 2017; 137:115-170. [PMID: 29405974 PMCID: PMC6550462 DOI: 10.1016/bs.acr.2017.11.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Effective and safe delivery of anticancer agents is among the major challenges in cancer therapy. The majority of anticancer agents are toxic to normal cells, have poor bioavailability, and lack in vivo stability. Recent advancements in nanotechnology provide safe and efficient drug delivery systems for successful delivery of anticancer agents via nanoparticles. The physicochemical and functional properties of the nanoparticle vary for each of these anticancer agents, including chemotherapeutics, nucleic acid-based therapeutics, small molecule inhibitors, and photodynamic agents. The characteristics of the anticancer agents influence the design and development of nanoparticle carriers. This review focuses on strategies of nanoparticle-based drug delivery for various anticancer agents. Recent advancements in the field are also highlighted, with suitable examples from our own research efforts and from the literature.
Collapse
Affiliation(s)
- Narsireddy Amreddy
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Anish Babu
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Ranganayaki Muralidharan
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Janani Panneerselvam
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Akhil Srivastava
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rebaz Ahmed
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Meghna Mehta
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Anupama Munshi
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rajagopal Ramesh
- The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.
| |
Collapse
|