1
|
Zhou Q, Jin M, Cui Y, Jiang S, Shang P, Li L. Advances in pharmacological activity and drug delivery systems of vinca alkaloids. Nat Prod Res 2025:1-21. [PMID: 40276897 DOI: 10.1080/14786419.2025.2494625] [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: 09/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
Vinca alkaloids (VAs), derived from the Catharanthus roseus, are naturally occurring or semi-synthetic alkaloids primarily used in the treatment approach for diverse types of cancer. They have shown significant efficacy in treating leukaemia, Hodgkin's lymphoma. Nevertheless, their clinical application is considerably limited owing to the severe side effects, low bioavailability, and multidrug resistance (MDR). Over the past few years, drug delivery systems such as nanoparticles, liposomes, and solid lipid nanoparticles (SLN) have been shown to improve the pharmacokinetic properties and tumour targeting of VAs. The use of multiple drugs in combination can also reduce the adverse reactions of VAs and significantly enhance their efficacy, thereby broadening their application. This review introduces the main pharmacologically active components of VAs, summarises their chemotherapeutic effects, and provides a statistical overview and analysis of recent research progress in VAs drug delivery technologies, offering a reference for further research and clinical application of VAs in cancer treatment.
Collapse
Affiliation(s)
- Quanying Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Min Jin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yulong Cui
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siqi Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peizhao Shang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lingjun Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
2
|
Zhang Z, Li M, Zhang X, Zhou F. Novel Strategies for Tumor Treatment: Harnessing ROS-Inducing Active Ingredients from Traditional Chinese Medicine Through Multifunctional Nanoformulations. Int J Nanomedicine 2024; 19:9659-9688. [PMID: 39309188 PMCID: PMC11416109 DOI: 10.2147/ijn.s479212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Reactive oxygen species (ROS) encompass a diverse group of chemically reactive molecules or ions distinguished by their substantial oxidative potential. Empirical studies have shown that the targeted administration of high toxic concentrations of ROS can effectively induce tumor cell death in various types. Numerous bioactive ingredients derived from traditional Chinese medicine (TCM), recognized for their ROS-inducing properties, have demonstrated significant anti-tumor activity. Nonetheless, their clinical application has been hindered by challenges such as low solubility, limited bioavailability, and poor selectivity. Multifunctional nanoformulations possess the potential to overcome these challenges and enhance the anticancer efficacy of ROS-inducing active compounds. Through extensive searches of various academic databases and a thorough review and screening of relevant literature, this study aims to systematically summarize and generalize multiple active ingredients in TCM that induce ROS generation, along with their multifunctional nanoformulations, from various perspectives. The objective is to provide new insights and references for fundamental cancer research and clinical treatments. Furthermore, we acknowledge that although numerous active ingredients and their nanoformulations in TCM have demonstrated ROS-inducing and anti-tumor potentials, potentially offering novel strategies for tumor therapy, the underlying mechanisms require further comprehensive investigation.
Collapse
Affiliation(s)
- Zhengguang Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Xiaolong Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| |
Collapse
|
3
|
Nguyen-Huu AM, Le NTT, Vo Do MH, Dong Yen PN, Nguyen-Dinh TD, Nguyen NH, Nguyen DH. Development and Characterization of Quercetin-Loaded Polymeric Liposomes with Gelatin-Poly(ethylene glycol)-Folic Acid Coating to Increase Their Long-Circulating and Anticancer Activity. ACS APPLIED BIO MATERIALS 2024; 7:4454-4470. [PMID: 38857443 DOI: 10.1021/acsabm.4c00334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Liposomes as drug-delivery systems have been researched and applied in multiple scientific reports and introduced as patented products with interesting therapeutic properties. Despite various advantages, this drug carrier faces major difficulties in its innate stability, cancer cell specificity, and control over the release of hydrophobic drugs, particularly quercetin, a naturally derived drug that carries many desirable characteristics for anticancer treatment. To improve the effectiveness of liposomes to deliver quercetin by tackling and mitigating the mentioned hurdles, we developed a strategy to establish the ability to passively target cancerous cells, as well as to increase the bioavailability of loaded drugs by incorporating poly(ethylene glycol), gelatin, and folic acid moieties to modify the liposomal system's surface. This research developed a chemically synthesized gelatin, poly(ethylene glycol), and folic acid as a single polymer to coat drug-loaded liposome systems. Liposomes were coated with gelatin-poly(ethylene glycol)-folic acid by electrostatic interaction, characterized by their size, morphology, ζ potential, drug loading efficiency, infrared structures, differential scanning calorimetry spectra, and drug-releasing profiles, and then evaluated for their cytotoxicity to MCF-7 breast cancer cells, as well as cellular uptake, analyzed by confocal imaging to further elaborate on the in vitro behavior of the coated liposome. The results indicated an unusual change in size with increased coating materials, followed by increased colloidal stability, ζ potential, and improved cytotoxicity to cancer cells, as shown by the cellular viability test with MCF-7. Cellular uptake also confirmed these results, providing data for the effects of biopolymer coating, while confirming that folic acid can increase the uptake of liposome by cancer cells. In consideration of such results, the modified gelatin-poly(ethylene glycol)-folic acid-coated liposome can be a potential system in delivering the assigned anticancer compound. This modified biopolymer showed excellent properties as a coating material and should be considered for further practical applications in the future.
Collapse
Affiliation(s)
- Anh-Minh Nguyen-Huu
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Department of Chemical and Material Engineering, Vlab, New Jersey Institute of Technology, Newark, New Jersey 07001, United States
| | - Ngoc Thuy Trang Le
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| | - Minh Hoang Vo Do
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
| | - Pham Nguyen Dong Yen
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
| | - Tien-Dung Nguyen-Dinh
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| | - Ngoc Hoi Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| | - Dai Hai Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| |
Collapse
|
4
|
André AS, Dias JNR, Aguiar SI, Leonardo A, Nogueira S, Amaral JD, Fernandes C, Gano L, Correia JDG, Cavaco M, Neves V, Correia J, Castanho M, Rodrigues CMP, Gaspar MM, Tavares L, Aires-da-Silva F. Panobinostat-loaded folate targeted liposomes as a promising drug delivery system for treatment of canine B-cell lymphoma. Front Vet Sci 2023; 10:1236136. [PMID: 37711439 PMCID: PMC10498770 DOI: 10.3389/fvets.2023.1236136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Cancer is a major public health problem with over 19 million cases reported in 2020. Similarly to humans, dogs are also largely affected by cancer, with non-Hodgkin's lymphoma (NHL) among the most common cancers in both species. Comparative medicine has the potential to accelerate the development of new therapeutic options in oncology by leveraging commonalities between diseases affecting both humans and animals. Within this context, in the present study, we investigated the potential of panobinostat (Pan)-loaded folate-targeted PEGylated liposomes (FA-PEG-Pan-Lip) for the treatment of canine B-cell lymphoma, while contributing to new perspectives in comparative oncology. Methods and results Two formulations were developed, namely: PEG-Pan-Lip and FA-PEG-Pan-Lip. Firstly, folate receptor expression in the CLBL-1 canine B-cell lymphoma cell line was assessed. After confirming receptor expression, both Pan-loaded formulations (PEG-Pan-Lip, FA-PEG-Pan-Lip) demonstrated dose-dependent inhibitory effects on CLBL-1 cell proliferation. The FA-PEG-Pan-Lip formulation (IC50 = 10.9 ± 0.03 nM) showed higher cytotoxicity than the non-targeted PEG-Pan-Lip formulation (IC50 = 12.9 ± 0.03 nM) and the free panobinostat (Pan) compound (IC50 = 18.32±0.03 nM). Moreover, mechanistically, both Pan-containing formulations induced acetylation of H3 histone and apoptosis. Flow cytometry and immunofluorescence analysis of intracellular uptake of rhodamine-labeled liposome formulations in CLBL-1 cells confirmed cellular internalization of PEG-Lip and FA-PEG-Lip formulations and higher uptake profile for the latter. Biodistribution studies of both radiolabeled formulations in CD1 and SCID mice revealed a rapid clearance from the major organs and a 1.6-fold enhancement of tumor uptake at 24 h for 111In-FA-PEG-Pan-Lip (2.2 ± 0.1 %ID/g of tumor) compared to 111In-PEG-Pan-Lip formulation (1.2±0.2 %ID/g of tumor). Discussion In summary, our results provide new data validating Pan-loaded folate liposomes as a promising targeted drug delivery system for the treatment of canine B-cell lymphoma and open innovative perspectives for comparative oncology.
Collapse
Affiliation(s)
- Ana S. André
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Joana N. R. Dias
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Sandra I. Aguiar
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Ana Leonardo
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Sara Nogueira
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Joana D. Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Célia Fernandes
- Departamento de Engenharia e Ciências Nucleares, Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Bobadela, Portugal
| | - Lurdes Gano
- Departamento de Engenharia e Ciências Nucleares, Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Bobadela, Portugal
| | - João D. G. Correia
- Departamento de Engenharia e Ciências Nucleares, Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Bobadela, Portugal
| | - Marco Cavaco
- Faculdade de Medicina, Instituto de Medicina Molecular-João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Vera Neves
- Faculdade de Medicina, Instituto de Medicina Molecular-João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Jorge Correia
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Miguel Castanho
- Faculdade de Medicina, Instituto de Medicina Molecular-João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Luís Tavares
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Frederico Aires-da-Silva
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| |
Collapse
|
5
|
Li J, Wang Q, Han Y, Jiang L, Lu S, Wang B, Qian W, Zhu M, Huang H, Qian P. Development and application of nanomaterials, nanotechnology and nanomedicine for treating hematological malignancies. J Hematol Oncol 2023; 16:65. [PMID: 37353849 PMCID: PMC10290401 DOI: 10.1186/s13045-023-01460-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/30/2023] [Indexed: 06/25/2023] Open
Abstract
Hematologic malignancies (HMs) pose a serious threat to patients' health and life, and the five-year overall survival of HMs remains low. The lack of understanding of the pathogenesis and the complex clinical symptoms brings immense challenges to the diagnosis and treatment of HMs. Traditional therapeutic strategies for HMs include radiotherapy, chemotherapy, targeted therapy and hematopoietic stem cell transplantation. Although immunotherapy and cell therapy have made considerable progress in the last decade, nearly half of patients still relapse or suffer from drug resistance. Recently, studies have emerged that nanomaterials, nanotechnology and nanomedicine show great promise in cancer therapy by enhancing drug targeting, reducing toxicity and side effects and boosting the immune response to promote durable immunological memory. In this review, we summarized the strategies of recently developed nanomaterials, nanotechnology and nanomedicines against HMs and then proposed emerging strategies for the future designment of nanomedicines to treat HMs based on urgent clinical needs and technological progress.
Collapse
Affiliation(s)
- Jinxin Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Qiwei Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Yingli Han
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Lingli Jiang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Siqi Lu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Beini Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Wenchang Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Meng Zhu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - He Huang
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
| |
Collapse
|
6
|
Capolla S, Argenziano M, Bozzer S, D’Agaro T, Bittolo T, De Leo L, Not T, Busato D, Dal Bo M, Toffoli G, Cavalli R, Gattei V, Bomben R, Macor P. Targeted chitosan nanobubbles as a strategy to down-regulate microRNA-17 into B-cell lymphoma models. Front Immunol 2023; 14:1200310. [PMID: 37359561 PMCID: PMC10285521 DOI: 10.3389/fimmu.2023.1200310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction MicroRNAs represent interesting targets for new therapies because their altered expression influences tumor development and progression. miR-17 is a prototype of onco-miRNA, known to be overexpressed in B-cell non-Hodgkin lymphoma (B-NHL) with peculiar clinic-biological features. AntagomiR molecules have been largely studied to repress the regulatory functions of up-regulated onco-miRNAs, but their clinical use is mainly limited by their rapid degradation, kidney elimination and poor cellular uptake when injected as naked oligonucleotides. Methods To overcome these problems, we exploited CD20 targeted chitosan nanobubbles (NBs) for a preferential and safe delivery of antagomiR17 to B-NHL cells. Results Positively charged 400 nm-sized nanobubbles (NBs) represent a stable and effective nanoplatform for antagomiR encapsulation and specific release into B-NHL cells. NBs rapidly accumulated in tumor microenvironment, but only those conjugated with a targeting system (antiCD20 antibodies) were internalized into B-NHL cells, releasing antagomiR17 in the cytoplasm, both in vitro and in vivo. The result is the down-regulation of miR-17 level and the reduction in tumor burden in a human-mouse B-NHL model, without any documented side effects. Discussion Anti-CD20 targeted NBs investigated in this study showed physico-chemical and stability properties suitable for antagomiR17 delivery in vivo and represent a useful nanoplatform to address B-cell malignancies or other cancers through the modification of their surface with specific targeting antibodies.
Collapse
Affiliation(s)
- Sara Capolla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Monica Argenziano
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Sara Bozzer
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Tiziana D’Agaro
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Tamara Bittolo
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Luigina De Leo
- Department of Pediatrics, Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Tarcisio Not
- Department of Pediatrics, Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Roberta Cavalli
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
7
|
Rostami N, Gomari MM, Abdouss M, Moeinzadeh A, Choupani E, Davarnejad R, Heidari R, Bencherif SA. Synthesis and Characterization of Folic Acid-Functionalized DPLA-co-PEG Nanomicelles for the Targeted Delivery of Letrozole. ACS APPLIED BIO MATERIALS 2023; 6:1806-1815. [PMID: 37093754 PMCID: PMC10629236 DOI: 10.1021/acsabm.3c00041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/07/2023] [Indexed: 04/25/2023]
Abstract
An effective treatment for hormone-dependent breast cancer is chemotherapy using cytotoxic agents such as letrozole (LTZ). However, most anticancer drugs, including LTZ, are classified as class IV biopharmaceuticals, which are associated with low water solubility, poor bioavailability, and significant toxicity. As a result, developing a targeted delivery system for LTZ is critical for overcoming these challenges and limitations. Here, biodegradable LTZ-loaded nanocarriers were synthesized by solvent emulsification evaporation using nanomicelles prepared with dodecanol-polylactic acid-co-polyethylene glycol (DPLA-co-PEG). Furthermore, cancer cell-targeting folic acid (FA) was conjugated into the nanomicelles to achieve a more effective and safer cancer treatment. During our investigation, DPLA-co-PEG and DPLA-co-PEG-FA displayed a uniform and spherical morphology. The average diameters of DPLA-co-PEG and DPLA-co-PEG-FA nanomicelles were 86.5 and 241.3 nm, respectively. Our preliminary data suggest that both nanoformulations were cytocompatible, with ≥90% cell viability across all concentrations tested. In addition, the amphiphilic nature of the nanomicelles led to high drug loading and dispersion in water, resulting in the extended release of LTZ for up to 50 h. According to the Higuchi model, nanomicelles functionalized with FA have a greater potential for the controlled delivery of LTZ into target cells. This model was confirmed experimentally, as LTZ-containing DPLA-co-PEG-FA was significantly and specifically more cytotoxic (up to 90% cell death) toward MCF-7 cells, a hormone-dependent human breast cancer cell line, when compared to free LTZ and LTZ-containing DPLA-co-PEG. Furthermore, a half-maximal inhibitory concentration (IC50) of 87 ± 1 nM was achieved when MCF-7 cells were exposed to LTZ-containing DPLA-co-PEG-FA, whereas higher doses of 125 ± 2 and 100 ± 2 nM were required for free LTZ and LTZ-containing DPLA-co-PEG, respectively. Collectively, DPLA-co-PEG-FA represents a promising nanosized drug delivery system to target controllably the delivery of drugs such as chemotherapeutics.
Collapse
Affiliation(s)
- Neda Rostami
- Department
of Chemistry, Amirkabir University of Technology, Tehran 1591634311, Iran
| | - Mohammad Mahmoudi Gomari
- Department
of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Majid Abdouss
- Department
of Chemistry, Amirkabir University of Technology, Tehran 1591634311, Iran
| | - Alaa Moeinzadeh
- Department
of Tissue Engineering and Regenerative Medicine, Faculty of Advanced
Technologies in Medicine, Iran University
of Medical Sciences, Tehran 1449614535, Iran
| | - Edris Choupani
- Department
of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Reza Davarnejad
- Department
of Chemical Engineering, Faculty of Engineering, Arak University, Arak 3848177584, Iran
| | - Reza Heidari
- Research
Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran 1411718541, Iran
| | - Sidi A. Bencherif
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
- Harvard
John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
- Sorbonne
University, UTC CNRS UMR 7338, Biomechanics and Bioengineering (BMBI),
University of Technology of Compiègne, Compiègne 60203, France
| |
Collapse
|
8
|
Li H, Dai W, Liu Z, He L. Renal Proximal Tubular Cells: A New Site for Targeted Delivery Therapy of Diabetic Kidney Disease. Pharmaceuticals (Basel) 2022; 15:ph15121494. [PMID: 36558944 PMCID: PMC9786989 DOI: 10.3390/ph15121494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Diabetic kidney disease (DKD) is a major complication of diabetes mellitus (DM) and the leading cause of end-stage kidney disease (ESKD) worldwide. A significant number of drugs have been clinically investigated for the treatment of DKD. However, a large proportion of patients still develop end-stage kidney disease unstoppably. As a result, new effective therapies are urgently needed to slow down the progression of DKD. Recently, there is increasing evidence that targeted drug delivery strategies such as large molecule carriers, small molecule prodrugs, and nanoparticles can improve drug efficacy and reduce adverse side effects. There is no doubt that targeted drug delivery strategies have epoch-making significance and great application prospects for the treatment of DKD. In addition, the proximal tubule plays a very critical role in the progression of DKD. Consequently, the purpose of this paper is to summarize the current understanding of proximal tubule cell-targeted therapy, screen for optimal targeting strategies, and find new therapeutic approaches for the treatment of DKD.
Collapse
Affiliation(s)
| | | | | | - Liyu He
- Correspondence: ; Tel.: +86-731-8529-2064
| |
Collapse
|
9
|
Polysialylated nanoinducer for precisely enhancing apoptosis and anti-tumor immune response in B-cell lymphoma. Acta Biomater 2022; 149:321-333. [PMID: 35779772 DOI: 10.1016/j.actbio.2022.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/12/2022] [Accepted: 06/19/2022] [Indexed: 11/24/2022]
Abstract
B-cell lymphoma is one of the most common types of lymphoma, and chemotherapy is still the current first-line treatment. However, due to the systemic side effects caused by chemotherapy drugs, traditional regimens have limitations and are difficult to achieve ideal efficacy. Recent studies have found that CD22 (also known as Siglec-2), as a specific marker of B-cells, is significantly up-regulated on B-cell lymphomas. Inspired by the specific recognition and binding of sialic acid residues by CD22, a polysialic acid (PSA)-modified PLGA nanocarrier (SAPC NP) designed to target B-cell lymphoma was fabricated. Mitoxantrone (MTO) was further loaded into SAPC NP through hydrophobic interactions to obtain polysialylated immunogenic cell death (ICD) nanoinducer (MTO@SAPC NP). Cellular experiments confirmed that MTO@SAPC NP could be specifically taken up by two types of CD22+ B lymphoma cells including Raji and Ramos cells, unlike the poor endocytic performance in other lymphocytes or macrophages. MTO@SAPC NP was determined to enhance the ICD and show better apoptotic effect on CD22+ cells. In the mouse model of B-cell lymphoma, MTO@SAPC NP significantly reduced the systemic side effects of MTO through lymphoma targeting, then achieved enhanced anti-tumor immune response, better tumor suppressive effect, and improved survival rate. Therefore, the polysialylated ICD nanoinducer provides a new strategy for precise therapy of B-cell lymphoma. STATEMENT OF SIGNIFICANCE: • Polysialic acid functionalized nanocarrier (SAPC NP) was designed and prepared. • SAPC NP is specifically endocytosed by two CD22+ B lymphoma cells. • Mitoxantrone-loaded nanoinducer (MTO@SAPC NP) promote immunogenic cell death and anti-tumor immune response. • "Polysialylation" is a potential new approach for precision treatment of B-cell lymphoma.
Collapse
|
10
|
Mirzaghavami PS, Khoei S, Khoee S, Shirvalilou S. Folic acid-conjugated magnetic triblock copolymer nanoparticles for dual targeted delivery of 5-fluorouracil to colon cancer cells. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00120-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
In the current study, folic acid-conjugated PEG-PCL-PEG triblock copolymer were synthesized and loaded with 5-fluorouracil and magnetite nanoparticles (5-FU-SPION-PEG-PCL-PEG-FA) for targeted delivery of drug to HT29 human colon cancer cells and CT26 mouse colon cancer model. The nanoparticles were synthesized and characterized by nuclear magnetic resonance spectroscopy (NMR) and transmission electron microscopy (TEM). The cellular uptake of nanoparticles was assessed in vitro (on HUVEC and HT29) and in vivo (on CT26 colon tumor tissues). The cytotoxic effect of nanoparticles was assessed on human colon cell lines (HT29, Caco-2, HTC116, and SW480) and normal HUVEC cells. In addition, antitumor effects of nanoparticles were investigated based on tumor volume, survival time and protein expression of Bax and Bcl-2 on CT26 tumor-bearing BALB/c mice.
Results
Characterization of nanoparticles showed 5-FU-SPION-PEG-PCL-PEG-FA (5-FU-NPs-FA) nanoparticles had spherical shape with hydrodynamic diameter of 85 nm. The drug-release profile exhibited sustained pH-responsive release with cumulative release reaching approximately 23% after 24 h. Cellular uptake studies revealed that HT29 cancer cells absorb higher amount of 5-FU-NPs-FA as compared to HUVEC normal cells (P < 0.05). In addition, 5-FU-NPs-FA was found to be more antitumor efficient in comparison to free 5-FU based on Bax/Bcl2 ratio, survival rate of tumoral mouse and inhibitory tumor volume (P < 0.05).
Conclusions
The results suggested that 5-FU-NPs-FA could be considered as promising sustained drug delivery platform for in vitro and in vivo conditions, which may provide selective treatment of tumor cancer cells.
Graphical Abstarct
Collapse
|
11
|
Mahajan S, Aalhate M, Guru SK, Singh PK. Nanomedicine as a magic bullet for combating lymphoma. J Control Release 2022; 347:211-236. [PMID: 35533946 DOI: 10.1016/j.jconrel.2022.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Hematological malignancy like lymphoma originates in lymph tissues and has a propensity to spread across other organs. Managing such tumors is challenging as conventional strategies like surgery and local treatment are not plausible options and there are high chances of relapse. The advent of novel targeted therapies and antibody-mediated treatments has proven revolutionary in the management of these tumors. Although these therapies have an added advantage of specificity in comparison to the traditional chemotherapy approach, such treatment alternatives suffer from the occurrence of drug resistance and dose-related toxicities. In past decades, nanomedicine has emerged as an excellent surrogate to increase the bioavailability of therapeutic moieties along with a reduction in toxicities of highly cytotoxic drugs. Nanotherapeutics achieve targeted delivery of the therapeutic agents into the malignant cells and also have the ability to carry genes and therapeutic proteins to the desired sites. Furthermore, nanomedicine has an edge in rendering personalized medicine as one type of lymphoma is pathologically different from others. In this review, we have highlighted various applications of nanotechnology-based delivery systems based on lipidic, polymeric and inorganic nanomaterials that address different targets for effectively tackling lymphomas. Moreover, we have discussed recent advances and therapies available exclusively for managing this malignancy.
Collapse
Affiliation(s)
- Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Santosh Kumar Guru
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
12
|
Vincristine-doxorubicin co-loaded artificial low-density lipoproteins towards solid tumours. Eur J Med Chem 2021; 226:113802. [PMID: 34543934 DOI: 10.1016/j.ejmech.2021.113802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/16/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022]
Abstract
To construct an artificial low-density lipoprotein (aLDL) that highly mimics low-density lipoprotein (LDL) in vivo, and deliver vincristine (VCR) - doxorubicin (DOX) simultaneously, the 100 nm and 35 nm DOX-VCR-aLDLs (DV-aLDLs) were constructed, then the physicochemical characteristics were evaluated. Through in vitro inverse gravity diffusion experiment, the tumour cake and sphere model experiment, draw a conclusion that the diffusion of 35 nm DV-aLDLs was stronger than 100 nm DV-aLDLs, and the tumour retention of 35 nm DV-aLDLs was better than the DV-solution. In addition, the three-dimension (3D) in vivo distribution imaging of aLDLs was performed on HepG-2 tumour-bearing nude mice, followed by the biodistribution and therapeutic efficacy on these xenograft models. Taking advantage of better diffusion capacity in tumour tissue, as well as the synergistic effect of VCR and DOX, the 35 nm DV-aLDL had the strongest efficacy and the lowest toxicity. High entrapment efficiency and stability, both active and passive targeting, making aLDL a potential carrier for tumour-targeted therapy at the same time.
Collapse
|
13
|
Ebrahimnejad P, Sodagar Taleghani A, Asare-Addo K, Nokhodchi A. An updated review of folate-functionalized nanocarriers: A promising ligand in cancer. Drug Discov Today 2021; 27:471-489. [PMID: 34781032 DOI: 10.1016/j.drudis.2021.11.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/27/2021] [Accepted: 11/09/2021] [Indexed: 12/18/2022]
Abstract
The uncontrolled release of drugs in conventional drug delivery systems has led to the introduction of new nanotechnology-based drug delivery systems and the use of targeted nanocarriers for cancer treatment. These targeted nanocarriers, which consist of intelligent nanoparticles modified with targeting ligands, can deliver drugs to specified locations at the right time and reduce drug doses to prevent side effects. Folate is a suitable targeting ligand for folate receptors overexpressed on cancer cells and has shown promising results in the diagnosis and treatment of cancer. In this review, we highlight the latest developments on the use of folate-conjugated nanoparticles in cancer diagnosis and treatment. Moreover, the toxicity, biocompatibility and efficacy of these nanocarriers are discussed.
Collapse
Affiliation(s)
- Pedram Ebrahimnejad
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Arezoo Sodagar Taleghani
- Department of Petroleum and Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kofi Asare-Addo
- Department of Pharmacy, University of Huddersfield, Huddersfield, UK
| | - Ali Nokhodchi
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Brighton, UK.
| |
Collapse
|
14
|
Choi JS, Park JW, Seu YB, Doh KO. Enhanced efficacy of folate-incorporated cholesteryl doxorubicin liposome in folate receptor abundant cancer cell. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
15
|
Jurczyk M, Jelonek K, Musiał-Kulik M, Beberok A, Wrześniok D, Kasperczyk J. Single- versus Dual-Targeted Nanoparticles with Folic Acid and Biotin for Anticancer Drug Delivery. Pharmaceutics 2021; 13:326. [PMID: 33802531 PMCID: PMC8001342 DOI: 10.3390/pharmaceutics13030326] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer is one of the major causes of death worldwide and its treatment remains very challenging. The effectiveness of cancer therapy significantly depends upon tumour-specific delivery of the drug. Nanoparticle drug delivery systems have been developed to avoid the side effects of the conventional chemotherapy. However, according to the most recent recommendations, future nanomedicine should be focused mainly on active targeting of nanocarriers based on ligand-receptor recognition, which may show better efficacy than passive targeting in human cancer therapy. Nevertheless, the efficacy of single-ligand nanomedicines is still limited due to the complexity of the tumour microenvironment. Thus, the NPs are improved toward an additional functionality, e.g., pH-sensitivity (advanced single-targeted NPs). Moreover, dual-targeted nanoparticles which contain two different types of targeting agents on the same drug delivery system are developed. The advanced single-targeted NPs and dual-targeted nanocarriers present superior properties related to cell selectivity, cellular uptake and cytotoxicity toward cancer cells than conventional drug, non-targeted systems and single-targeted systems without additional functionality. Folic acid and biotin are used as targeting ligands for cancer chemotherapy, since they are available, inexpensive, nontoxic, nonimmunogenic and easy to modify. These ligands are used in both, single- and dual-targeted systems although the latter are still a novel approach. This review presents the recent achievements in the development of single- or dual-targeted nanoparticles for anticancer drug delivery.
Collapse
Affiliation(s)
- Magdalena Jurczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Katarzyna Jelonek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
| | - Monika Musiał-Kulik
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Janusz Kasperczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland
| |
Collapse
|
16
|
Shukla S, Roe AJ, Liu R, Veliz FA, Commandeur U, Wald DN, Steinmetz NF. Affinity of plant viral nanoparticle potato virus X (PVX) towards malignant B cells enables cancer drug delivery. Biomater Sci 2020; 8:3935-3943. [PMID: 32662788 DOI: 10.1039/d0bm00683a] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Non-Hodgkin's B cell lymphomas (NHL) include a diverse set of neoplasms that constitute ∼90% of all lymphomas and the largest subset of blood cancers. While chemotherapy is the first line of treatment, the efficacy of contemporary chemotherapies is hampered by dose-limiting toxicities. Partly due to suboptimal dosing, ∼40% of patients exhibit relapsed or refractory disease. Therefore more efficacious drug delivery systems are urgently needed to improve survival of NHL patients. In this study we demonstrate a new drug delivery platform for NHL based on the plant virus Potato virus X (PVX). We observed a binding affinity of PVX towards malignant B cells. In a metastatic mouse model of NHL, we show that systemically administered PVX home to tissues harboring malignant B cells. When loaded with the chemotherapy monomethyl auristatin (MMAE), the PVX nanocarrier enables effective delivery of MMAE to human B lymphoma cells in a NHL mouse model leading to inhibition of lymphoma growth in vivo and improved survival. Thus, PVX nanoparticle is a promising drug delivery platform for B cell malignancies.
Collapse
Affiliation(s)
- Sourabh Shukla
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Li Y, Wang T, Liu Y, Xu Y, Sun Z, Yang G. NIR-laser-triggered drug release from folate decorated albumin nanoparticles for synergistic chemo-photothermal tumor therapy. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
18
|
Kumar P, Huo P, Liu B. Formulation Strategies for Folate-Targeted Liposomes and Their Biomedical Applications. Pharmaceutics 2019; 11:E381. [PMID: 31382369 PMCID: PMC6722551 DOI: 10.3390/pharmaceutics11080381] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/22/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
The folate receptor (FR) is a tumor-associated antigen that can bind with folic acid (FA) and its conjugates with high affinity and ingests the bound molecules inside the cell via the endocytic mechanism. A wide variety of payloads can be delivered to FR-overexpressed cells using folate as the ligand, ranging from small drug molecules to large DNA-containing macromolecules. A broad range of folate attached liposomes have been proven to be highly effective as the targeted delivery system. For the rational design of folate-targeted liposomes, an intense conceptual understanding combining chemical and biomedical points of view is necessary because of the interdisciplinary nature of the field. The fabrication of the folate-conjugated liposomes basically involves the attachment of FA with phospholipids, cholesterol or peptides before liposomal formulation. The present review aims to provide detailed information about the design and fabrication of folate-conjugated liposomes using FA attached uncleavable/cleavable phospholipids, cholesterol or peptides. Advances in the area of folate-targeted liposomes and their biomedical applications have also been discussed.
Collapse
Affiliation(s)
- Parveen Kumar
- Laboratory of Functional Molecules and Materials, School of Physics and Optoelectronic Engineering, Shandong University of Technology, Xincun West Road 266, Zibo 255000, China
| | - Peipei Huo
- Laboratory of Functional Molecules and Materials, School of Physics and Optoelectronic Engineering, Shandong University of Technology, Xincun West Road 266, Zibo 255000, China
| | - Bo Liu
- Laboratory of Functional Molecules and Materials, School of Physics and Optoelectronic Engineering, Shandong University of Technology, Xincun West Road 266, Zibo 255000, China.
| |
Collapse
|
19
|
Limongi T, Susa F, Cauda V. Nanoparticles for hematologic diseases detection and treatment. HEMATOLOGY & MEDICAL ONCOLOGY 2019; 4:1000183. [PMID: 33860108 PMCID: PMC7610588 DOI: 10.15761/hmo.1000183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanotechnology, as an interdisciplinary science, combines engineering, physics, material sciences, and chemistry with the biomedicine knowhow, trying the management of a wide range of diseases. Nanoparticle-based devices holding tumor imaging, targeting and therapy capabilities are formerly under study. Since conventional hematological therapies are sometimes defined by reduced selectivity, low therapeutic efficacy and many side effects, in this review we discuss the potential advantages of the NPs' use in alternative/combined strategies. In the introduction the basic notion of nanomedicine and nanoparticles' classification are described, while in the main text nanodiagnostics, nanotherapeutics and theranostics solutions coming out from the use of a wide-ranging NPs availability are listed and discussed.
Collapse
Affiliation(s)
- Tania Limongi
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| |
Collapse
|
20
|
Monteiro LOF, Fernandes RS, Castro L, Reis D, Cassali GD, Evangelista F, Loures C, Sabino AP, Cardoso V, Oliveira MC, Branco de Barros A, Leite EA. Paclitaxel-Loaded Folate-Coated pH-Sensitive Liposomes Enhance Cellular Uptake and Antitumor Activity. Mol Pharm 2019; 16:3477-3488. [DOI: 10.1021/acs.molpharmaceut.9b00329] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
21
|
Biffi S, Voltan R, Bortot B, Zauli G, Secchiero P. Actively targeted nanocarriers for drug delivery to cancer cells. Expert Opin Drug Deliv 2019; 16:481-496. [DOI: 10.1080/17425247.2019.1604679] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Stefania Biffi
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Rebecca Voltan
- Department of Morphology, Surgery, Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Barbara Bortot
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery, Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery, Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
22
|
Unnam S, Panduragaiah VM, Sidramappa MA, Muddana Eswara BR. Gemcitabine-loaded Folic Acid Tagged Liposomes: Improved Pharmacokinetic and Biodistribution Profile. Curr Drug Deliv 2019; 16:111-122. [PMID: 30360740 DOI: 10.2174/1567201815666181024112252] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/05/2018] [Accepted: 10/18/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND Gemcitabine (GEM) is found effective in the treatment of many solid tumors. However, its use is restricted due to its small circulation half-life, fast metabolism and low capacity for selective tumor uptake. Folate receptors (FRs) have been recognized as cellular surface markers, which can be used for cancer targeting. PEGylated liposomes decorated with folic acid have been investigated for several anticancer agents not only to extend plasma half-life but also for tumor targeting via folic acid receptors which overexpressed on tumor cell surface. OBJECTIVE Therefore, the objective of the present study was to prepare GEM-loaded folic acid tagged liposomes to improve the pharmacokinetics and tumor distribution of GEM. METHODS The blank folate-targeted liposomes composed of HSPC/DSPE-mPEG2000/DSPE-mPEG-Folic acid were prepared first by thin film hydration technique. GEM was then loaded into liposomes by remote loading technique. The optimized liposomal formulations were evaluated in vitro for GEM release using dialysis technique, HeLa cell uptake using FACS technique, and cytotoxicity using MTT dye reduction assay. The comparative in vivo pharmacokinetic and biodistribution characteristics of radiolabeled (99mTc-labeled) plain GEM solution, and all liposomal formulations (conventional:CLs; stealth: SLs; folate targeted: FTLs) were evaluated in mice model. RESULTS GEM-loaded FTLs showed sustained release profile, efficient uptake by HeLa cells and greater cytotoxicity. Further, FTLs displayed significantly improved pharmacokinetics, and biodistribution profile of loaded GEM. CONCLUSION In conclusion, the developed GEM-loaded folic acid receptor-targeted liposomal formulation could be a promising and potential alternative formulation for further development.
Collapse
Affiliation(s)
- Sambamoorthy Unnam
- Department of Pharmacy, Biju Patnaik University of Technology, Rourkela, Odisha, India.,NRI College of Pharmacy, Pothavarappadu, Agiripalli, Krishna District, Andhrapradesh, India
| | | | | | | |
Collapse
|