1
|
Gupta S, Gupta A, Mukherjee M, Bose S, Sinha S. Chemical Insights into Oligonucleotide-Protein Binding for Therapeutic Applications. J Med Chem 2025; 68:9848-9863. [PMID: 40332202 DOI: 10.1021/acs.jmedchem.5c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Plasma protein binding is an important determinant in the clinical success of oligonucleotide-based drugs. Optimal protein binding of the oligonucleotide is critical to its tissue distribution and retention by preventing renal excretion. This property can be modulated through suitable chemical modifications depending on the oligonucleotide backbone to achieve a balanced pharmacokinetic profile and minimize off-target effects. The macromolecular structure of the oligonucleotide leads to dynamic protein binding characteristics as compared to small-molecule-based drugs, which are not associated with additional barriers such as intracellular delivery. This perspective provides insight into the diverse plasma protein interactions of various classes of oligonucleotides and explores chemical strategies for modulating these interactions. Furthermore, we have discussed different methods for the quantification of plasma protein binding along with the correlation of chemistry and therapeutic outcomes of FDA-approved oligonucleotides.
Collapse
Affiliation(s)
- Shalini Gupta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Abhishek Gupta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Maria Mukherjee
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Sritama Bose
- Medical Research Council, Nucleic Acid Therapy Accelerator, (UKRI) Research Complex at Harwell (RCaH), Rutherford Appleton Laboratory, Harwell OX11 0FA, U.K
| | - Surajit Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
2
|
Yang C, Ma H, Liang Z, Zhuang Y, Hu L, Zhang K, Huang L, Li M, Zhang S, Zhen Y. Cyclic RGD modified dextran-quercetin polymer micelles for targeted therapy of breast cancer. Int J Biol Macromol 2025; 308:142272. [PMID: 40118409 DOI: 10.1016/j.ijbiomac.2025.142272] [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: 08/02/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Quercetin is a natural flavonoid found in many plants which has various pharmacological activities including antitumor effect. However, the poor water solubility and bioavailability limit the potential benefits of quercetin for patients. Thus, modifying quercetin structure and developing actively targeted drug delivery systems are extremely important for tumor precision therapy. Herein, polymer-drug conjugates dextran-quercetin (D-Q) and cRGD-dextran (R-D) were synthesized by grafting quercetin and polypeptide cRGDfk (Arg-Gly-Asp-(D-Phe)-Lys) to dextran. Then cRGD-modified dextran-quercetin polymer micelles (R-D-Q) were constructed by self-assembling of D-Q and R-D. R-D-Q micelles possessed appropriate particle size (133.4 nm), nearly neutral potential (8.14 mV) and excellent drug-loading efficiency (13.1 %) and achieved higher cytotoxicity, apoptosis induction and penetration to human breast cancer MCF-7 cells than the micelles unmodified with cRGD, which were ascribed to cRGD-integrin mediated transcytosis. R-D-Q micelles effectively suppressed tumor growth in tumor-bearing mice by delivering more quercetin throughout the tumor tissue. And R-D-Q micelles could promote the apoptosis of tumor cells by activating p38 and JNK signal pathways and suppressing ERK signal pathway. In addition, R-D-Q micelles had no damage to normal tissues of mice at therapeutic dose. These results indicate promising prospects for R-D-Q micelles as an effective drug delivery system against tumor.
Collapse
Affiliation(s)
- Chunpeng Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Huiling Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ze Liang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ying Zhuang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Litao Hu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Kexin Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Leixiao Huang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Min Li
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, China; Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
3
|
Lei D, Wang W, Zhao J, Zhou Y, Chen Y, Dai J, Qiu Y, Qi H, Li C, Liang B, Liu B, Wang Q, Li R. An injectable gambogic acid loaded nanocomposite hydrogel enhances antitumor effect by reshaping immunosuppressive tumor microenvironment. Mater Today Bio 2025; 31:101611. [PMID: 40104652 PMCID: PMC11919334 DOI: 10.1016/j.mtbio.2025.101611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Gambogic acid(GA)is a natural compound that exhibits strong antitumor activity against a variety of tumors. However, its poor water solubility, low specificity, and high toxicity lead to inevitable systemic adverse effects. To minimize side effects, combining gambogic acid (GA) with delivery systems such as nanohydrogels to develop an in situ vaccine system (ISV) shows great promise. In this study, we loaded GA into a novel in situ nanocomposite hydrogel vaccine system (Gel-NPs@GA) along with a near-infrared (NIR) fluorescent dye, IR-1061. The Gel-NPs@GA system allowed for temperature-triggered gelation, simplifying injection and the in vivo formation of a drug-releasing gel, with near-infrared monitoring for drug metabolism. Slow, continuous release of gelatinase-targeted GA nanoparticles from the hydrogel occurs, followed by cleavage of mPEG-peptide-PCL conjugates by gelatinase, causing particle aggregation for endocytosis by tumor cells. This approach tackled solubility issues and curbs excessive GA release, boosting therapeutic drug levels. The sustained GA release induces tumor cell apoptosis, releasing tumor antigens and reprogramming the immune-suppressive tumor microenvironment. In the CT26 colorectal cancer mice model, this in situ vaccine system significantly inhibited tumor growth. By integrating information about immune cell clusters within the tumor microenvironment with RNA sequencing results, we hypothesized that Gel-NPs@GA could synergistically stimulate the immune response through various pathways, promote the maturation of dendritic cells (DCs), increase the infiltration of T cells, and thereby remodel the tumor's immune microenvironment.
Collapse
Affiliation(s)
- Dan Lei
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wanru Wang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianhang Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Yingling Zhou
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Chen
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Juanjuan Dai
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuling Qiu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haoyue Qi
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chunhua Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Baorui Liu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| | - Qin Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| | - Rutian Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| |
Collapse
|
4
|
Xiao TG, Witek L, Bundy RA, Moses A, Obermiller CS, Schreiner AD, Dharod A, Russo MW, Rudnick SR. Identifying and Linking Patients At Risk for MASLD with Advanced Fibrosis to Care in Primary Care. J Gen Intern Med 2025; 40:629-636. [PMID: 39060786 PMCID: PMC11861828 DOI: 10.1007/s11606-024-08955-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND AND AIMS Severity of fibrosis is the driver of liver-related outcomes in metabolic dysfunction-associated steatotic liver disease (MASLD), and non-invasive testing such as fibrosis-4 (FIB-4) score is utilized for risk stratification. We aimed to determine if primary care patients at risk for MASLD and advanced fibrosis were evaluated with subsequent testing. A secondary aim was to determine if at-risk patients with normal aminotransferases had advanced fibrosis. METHODS Primary care patients at increased risk for MASLD with advanced fibrosis (n = 91,914) were identified using previously established criteria. Patients with known alternative/concomitant etiology of liver disease or cirrhosis were excluded. The study cohort included patients with calculated FIB-4 score in 2020 (n = 52,006), and stratified into low, indeterminate, and high likelihood of advanced fibrosis. Among those at indeterminate/high risk, rates of subsequent testing were measured. RESULTS Risk stratification with FIB-4 characterized 77% (n = 40,026) as low risk, 17% (n = 8847) as indeterminate, and 6% (n = 3133) as high risk. Among indeterminate/high-risk patients (n = 11,980), 78.7% (n = 9433) had aminotransferases within normal limits, 0.95% (n = 114) had elastography, and 8.2% (n = 984) were referred for subspecialty evaluation. CONCLUSION In this cohort of primary care patients at risk for MASLD with fibrosis, the FIB-4 score identified a substantial proportion of indeterminate/high-risk patients, the majority of which had normal aminotransferase levels. Low rates of subsequent testing were observed. These data suggest that a majority of patients at increased risk for liver-related outcomes remain unrecognized and highlight opportunities to facilitate their identification.
Collapse
Affiliation(s)
- Ted G Xiao
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lauren Witek
- Informatics and Analytics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Richa A Bundy
- Informatics and Analytics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Adam Moses
- Informatics and Analytics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Corey S Obermiller
- Informatics and Analytics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Andrew D Schreiner
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ajay Dharod
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Informatics and Analytics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Implementation Science, Division of Public Health Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Wake Forest Center for Healthcare Innovation, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Wake Forest Center for Biomedical Informatics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Mark W Russo
- Division of Liver Diseases and Transplant, Atrium Health Carolina Medical Center, Charlotte, NC, USA
| | - Sean R Rudnick
- Section of Gastroenterology and Hepatology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
5
|
Zhang Y, Lin X, Chen X, Fang W, Yu K, Gu W, Wei Y, Zheng H, Piao J, Li F. Strategies to Regulate the Degradation and Clearance of Mesoporous Silica Nanoparticles: A Review. Int J Nanomedicine 2024; 19:5859-5878. [PMID: 38887691 PMCID: PMC11182361 DOI: 10.2147/ijn.s451919] [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: 11/27/2023] [Accepted: 04/26/2024] [Indexed: 06/20/2024] Open
Abstract
Mesoporous silica nanoparticles (MSNs) have attracted extensive attention as drug delivery systems because of their unique meso-structural features (high specific surface area, large pore volume, and tunable pore structure), easily modified surface, high drug-loading capacity, and sustained-release profiles. However, the enduring and non-specific enrichment of MSNs in healthy tissues may lead to toxicity due to their slow degradability and hinder their clinical application. The emergence of degradable MSNs provided a solution to this problem. The understanding of strategies to regulate degradation and clearance of these MSNs for promoting clinical trials and expanding their biological applications is essential. Here, a diverse variety of degradable MSNs regarding considerations of physiochemical properties and doping strategies of degradation, the biodistribution of MSNs in vivo, internal clearance mechanism, and adjusting physical parameters of clearance are highlighted. Finally, an overview of these degradable and clearable MSNs strategies for biosafety is provided along with an outlook of the encountered challenges.
Collapse
Affiliation(s)
- Yuelin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Xue Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Xinxin Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Weixiang Fang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Kailing Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Wenting Gu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Hangsheng Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Jigang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Fanzhu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| |
Collapse
|
6
|
Yamashita S, Azuma K, Tanaka Y, Kimura S, Kiriyama A. Variations in the inner core affect the pharmacokinetics of indomethacin-encapsulated polymeric micelles. Int J Pharm 2024; 654:123933. [PMID: 38403090 DOI: 10.1016/j.ijpharm.2024.123933] [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: 11/09/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Hydrophobic ion pairing (HIP) is a drug encapsulation technology that uses electrostatic interactions between a drug and an additive. However, although polymeric micelles can encapsulate hydrophobic drugs in the core, the encapsulated drug often leaks. Therefore, we designed polymeric micelles with HIP functionalized in a hydrophobic inner core using three diblock copolymers comprising polypeptides with different ratios of polar and hydrophobic amino acids and polyethylene glycol (PEG) to encapsulate indomethacin (IND). The three IND-encapsulated HIP micelles showed different area under the curve (AUC) values as an index of blood retention after intravenous injection in mice. Despite having the same PEG shell, IND-PEG-poly(H/F)n showed a 1.56-fold higher AUC than IND-PEG-poly(D/F)n. PEG interface morphologies were evaluated to determine the differences in pharmacokinetic parameters caused by changes in inner core HIP patterns. The micellarized diblock copolymer was desorbed from IND-PEG-poly(D/F)n due to electrostatic repulsion between IND and the diblock copolymer comprising aspartic acid. Our results suggest that changes in the HIP patterns of the micelle inner core affected the PEG interface morphologies, such as PEG density and diblock copolymer desorption from micelles. These phenomena might lead to changes in the interaction of plasma proteins and drug dispositions.
Collapse
Affiliation(s)
- Shugo Yamashita
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan.
| | - Karen Azuma
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Yuka Tanaka
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Shunsuke Kimura
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Akiko Kiriyama
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| |
Collapse
|
7
|
Koeppe H, Horn D, Scholz J, Quaas E, Schötz S, Reisbeck F, Achazi K, Mohammadifar E, Dernedde J, Haag R. Shell-Sheddable Dendritic Polyglycerol Sulfates Loaded with Sunitinib for Inhibition of Tumor Angiogenesis. Int J Pharm 2023:123158. [PMID: 37336299 DOI: 10.1016/j.ijpharm.2023.123158] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Induced angiogenesis, a specific hallmark of cancer, plays a vital role in tumor progression and can be targeted by inhibitors like sunitinib. Sunitinib is a small hydrophobic molecule suffering from low bioavailability and a short half-life in the bloodstream. To overcome these drawbacks, suitable drug delivery systems need to be developed. In this work dendritic polyglycerol (dPG), a well-known polymer, was functionalized with a sheddable shell. Therefore, aliphatic chains of different lengths (C5, C9, C11) were coupled to dPG through a cleavable ester bond. To restore water solubility and improve tumor targeting, the surface was decorated with sulfate groups. The resulting shell-sheddable dPG sulfates were characterized and evaluated regarding their loading capacity and biocompatibility in cell culture. The nine-carbon chain derivative (dPG-TNS) was selected as the best candidate for further experiments due to its high drug loading capacity (20wt%), and a sustained release in vitro. The cellular biocompatibility of the blank carrier up to 1mg/mL was confirmed after 24h incubation on HeLa cells. Furthermore, the shell-cleavability of dPG-TNS under different physiological conditions was shown in a degradation study over four weeks. The activity of sunitinib-loaded dPG-TNS was demonstrated in a tube formation assay on Human umbilical vein endothelial cells (HUVECs). Our results suggest that the drug-loaded nanocarrier is a promising candidate to be further investigated in tumor treatments, as it shows similar efficacy to free sunitinib while overcoming its limitations.
Collapse
Affiliation(s)
- Hanna Koeppe
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Daniel Horn
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Johanna Scholz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Elisa Quaas
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Sebastian Schötz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Felix Reisbeck
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Ehsan Mohammadifar
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| | - Jens Dernedde
- Institute for Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
8
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [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: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
9
|
Tinku, Prajapati AK, Choudhary S. Physicochemical insights into the micellar delivery of doxycycline and minocycline to the carrier protein in aqueous environment. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
10
|
Lamanna L, Cataldi P, Friuli M, Demitri C, Caironi M. Monitoring of Drug Release via Intra Body Communication with an Edible Pill. ADVANCED MATERIALS TECHNOLOGIES 2023; 8. [DOI: 10.1002/admt.202200731] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Indexed: 01/03/2025]
Abstract
AbstractOral drug administration provides a convenient and patient‐compliant way for drug delivery, especially for chronic diseases and prolonged pharmacological treatments. However, due to the repetitiveness of such therapeutic approach, the patients are led to neglect/forget the therapy affecting the healthcare delivery. Indeed, the non‐adherence to pharmacological prescriptions and the unknown amount of real‐time drug release result in a non‐compliant therapeutic drug level over the protracted therapies. The proposed technology will enable the monitoring of both pharmacological adherence and real‐time drug release. The approach exploits a passive intrabody communication (IBC) activation in order to enable an edible pill, realized starting from food additives and food‐grade materials, to monitor pharmacological adherence. Following activation, the signal is modulated by IBC coupling switching triggered by pill degradation in a gastrointestinal tract, resulting in a monitored drug release. The proof‐of‐concept is designed for a targeted release and monitoring of Metformin in the intestine. The system shows an in vitro limit of cumulative drug release detection of 18 µg mL−1 and a limit of real‐time drug release detection of 2 µg mL−1 min−1. This platform represents the first solution to monitor passive drug release in real‐time, from intake to complete absorption, enabling unique and long‐sought healthcare therapy and treatment opportunity.
Collapse
Affiliation(s)
- Leonardo Lamanna
- Center for Nano Science and Technology @PoliMi Istituto Italiano di Tecnologia Via G. Pascoli, 70/3 Milano 20133 Italy
- Department of Engineering for Innovation Campus Ecotekne University of Salento Via per Monteroni Lecce 73100 Italy
| | - Pietro Cataldi
- Center for Nano Science and Technology @PoliMi Istituto Italiano di Tecnologia Via G. Pascoli, 70/3 Milano 20133 Italy
| | - Marco Friuli
- Department of Engineering for Innovation Campus Ecotekne University of Salento Via per Monteroni Lecce 73100 Italy
| | - Christian Demitri
- Department of Engineering for Innovation Campus Ecotekne University of Salento Via per Monteroni Lecce 73100 Italy
| | - Mario Caironi
- Center for Nano Science and Technology @PoliMi Istituto Italiano di Tecnologia Via G. Pascoli, 70/3 Milano 20133 Italy
| |
Collapse
|
11
|
Rastogi V, Yadav P, Porwal M, Sur S, Verma A. Dendrimer as nanocarrier for drug delivery and drug targeting therapeutics: a fundamental to advanced systematic review. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2158334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Vaibhav Rastogi
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, India
| | - Pragya Yadav
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, India
| | - Mayur Porwal
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, India
| | - Souvik Sur
- Research and Development Center, Teerthanker Mahaveer University, Moradabad, India
| | - Anurag Verma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, India
| |
Collapse
|
12
|
Biasizzo M, Javoršek U, Vidak E, Zarić M, Turk B. Cysteine cathepsins: A long and winding road towards clinics. Mol Aspects Med 2022; 88:101150. [PMID: 36283280 DOI: 10.1016/j.mam.2022.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022]
Abstract
Biomedical research often focuses on properties that differentiate between diseased and healthy tissue; one of the current focuses is elevated expression and altered localisation of proteases. Among these proteases, dysregulation of cysteine cathepsins can frequently be observed in inflammation-associated diseases, which tips the functional balance from normal physiological to pathological manifestations. Their overexpression and secretion regularly exhibit a strong correlation with the development and progression of such diseases, making them attractive pharmacological targets. But beyond their mostly detrimental role in inflammation-associated diseases, cysteine cathepsins are physiologically highly important enzymes involved in various biological processes crucial for maintaining homeostasis and responding to different stimuli. Consequently, several challenges have emerged during the efforts made to translate basic research data into clinical applications. In this review, we present both physiological and pathological roles of cysteine cathepsins and discuss the clinical potential of cysteine cathepsin-targeting strategies for disease management and diagnosis.
Collapse
Affiliation(s)
- Monika Biasizzo
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Miki Zarić
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
13
|
Eljack S, David S, Chourpa I, Faggad A, Allard-Vannier E. Formulation of Lipid-Based Nanoparticles for Simultaneous Delivery of Lapatinib and Anti-Survivin siRNA for HER2+ Breast Cancer Treatment. Pharmaceuticals (Basel) 2022; 15:ph15121452. [PMID: 36558904 PMCID: PMC9784347 DOI: 10.3390/ph15121452] [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: 09/16/2022] [Revised: 11/13/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
In this work, lipid-based nanoparticles (LBNP) were designed to combine tyrosine kinase inhibitor (TKI) Lapatinib (LAPA) with siRNA directed against apoptosis inhibitor protein Survivin (siSurvivin) in an injectable form. This nanosystem is based on lipid nanocapsules (LNCs) coated with a cationic polymeric shell composed of chitosan grafted through a transacylation reaction. The hydrophobic LAPA is solubilized in the inner oily core, while hydrophilic siRNA is associated electrostatically onto the nanocarrier’s surface. The co-loaded LBNP showed a narrow size distribution (polydispersity index (PDI) < 0.3), a size of 130 nm, and a slightly positive zeta potential (+21 mV). LAPA and siRNA were loaded in LBNP at a high rate of >90% (10.6 mM) and 100% (4.6 µM), respectively. The siRNA-LAPA_LBNP was readily uptaken by the human epidermal growth factor receptor 2 overexpressed (HER2+) breast cancer cell line SK-BR-3. Moreover, the cytotoxicity studies confirmed that the blank chitosan decorated LBNP is not toxic to the cells with the tested concentrations, which correspond to LAPA concentrations from 1 to 10 µM, at different incubation times up to 96 h. Furthermore, siCtrl.-LAPA_LBNP had a more cytotoxic effect than Lapatinib salt, while siSurvivin-LAPA_LBNP had a significant synergistic cytotoxic effect compared to siCtrl.-LAPA_LBNP. All these findings suggested that the developed modified LBNP could potentiate anti-Survivin siRNA and LAPA anti-cancer activity.
Collapse
Affiliation(s)
- Sahar Eljack
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Medani 21111, Sudan
| | - Stephanie David
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
| | - Igor Chourpa
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
| | - Areeg Faggad
- Department of Molecular Biology, National Cancer Institute, University of Gezira (NCI-UG), Wad Medani 21111, Sudan
| | - Emilie Allard-Vannier
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
- Correspondence:
| |
Collapse
|
14
|
Huang S, Gao Y, Lv Y, Wang Y, Cao Y, Zhao W, Zuo D, Mu H, Hua Y. Applications of Nano/Micromotors for Treatment and Diagnosis in Biological Lumens. MICROMACHINES 2022; 13:mi13101780. [PMID: 36296133 PMCID: PMC9610721 DOI: 10.3390/mi13101780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 06/01/2023]
Abstract
Natural biological lumens in the human body, such as blood vessels and the gastrointestinal tract, are important to the delivery of materials. Depending on the anatomic features of these biological lumens, the invention of nano/micromotors could automatically locomote targeted sites for disease treatment and diagnosis. These nano/micromotors are designed to utilize chemical, physical, or even hybrid power in self-propulsion or propulsion by external forces. In this review, the research progress of nano/micromotors is summarized with regard to treatment and diagnosis in different biological lumens. Challenges to the development of nano/micromotors more suitable for specific biological lumens are discussed, and the overlooked biological lumens are indicated for further studies.
Collapse
Affiliation(s)
- Shandeng Huang
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yinghua Gao
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yu Lv
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yun Wang
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yinghao Cao
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Weisong Zhao
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Dongqing Zuo
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| |
Collapse
|
15
|
Daskhan GC, Motyka B, Bascom R, Tran HT, Tao K, West LJ, Cairo CW. Extending the in vivo persistence of synthetic glycoconjugates using a serum-protein binder. RSC Chem Biol 2022; 3:1260-1275. [PMID: 36320887 PMCID: PMC9533409 DOI: 10.1039/d2cb00126h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/22/2022] [Indexed: 12/08/2023] Open
Abstract
Synthetic glycoconjugates are used in the development of vaccines and the design of inhibitors for glycan-protein interactions. The in vivo persistence of synthetic glycoconjugates is an important factor in their efficacy, especially when prolonged interactions with specific cell types may be required. In this study, we applied a strategy for non-covalent association of an active compound with serum proteins for extension of glycoconjugate half-life in serum. The small molecule, AG10, has previously been used to extend the half-life of small molecules through its high affinity for transthyretin (TTR), a serum protein. Using a tetravalent polyethylene glycol (PEG)-based scaffold we developed a synthetic strategy for glycoconjugates that allowed for controlled addition of multiple tags, such as a TTR affinity tag or fluorophore. We designed a version of AG10 modified at the pyrazole core, named GD10, amenable to our conjugation strategy and introduced to glycoconjugates using a tri-functional linker. This approach allowed for attachment of GD10 and fluorophore tags, as well as carbohydrate antigens. We then tested the influence of the GD10 tag on glycoconjugate half-life in vivo using a mouse model. Our results suggest that the combination of the GD10 tag and the PEG scaffold extended the half-life of glycoconjugates by as much as 10-fold when compared to proteins of similar molecular weight. The GD10 tag was able to extend the half-life of similar glycoconjugates by as much as 2-fold. We observed a role for the terminal saccharide residue of the carbohydrate antigen and confirmed that conjugates were able to penetrate multiple compartments in vivo including bone marrow, lymph nodes, and other organs. The introduction of the GD10 tag did not obstruct the ability of conjugates to interact with lectin receptors. We conclude that serum protein binders can be used to extend the persistence of glycoconjugates in vivo.
Collapse
Affiliation(s)
- Gour Chand Daskhan
- Department of Chemistry, University of Alberta Edmonton Alberta T6G 2G2 Canada +1 780 492 8231 +1 780 492 0377
| | - Bruce Motyka
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Roger Bascom
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Hanh Thuc Tran
- Department of Chemistry, University of Alberta Edmonton Alberta T6G 2G2 Canada +1 780 492 8231 +1 780 492 0377
| | - Kesheng Tao
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Lori J West
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Departments of Surgery, Medical Microbiology & Immunology, and Laboratory Medicine & Pathology, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Christopher W Cairo
- Department of Chemistry, University of Alberta Edmonton Alberta T6G 2G2 Canada +1 780 492 8231 +1 780 492 0377
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| |
Collapse
|
16
|
Kalave S, Hegde N, Juvale K. Applications of Nanotechnology-based Approaches to Overcome Multi-drug Resistance in Cancer. Curr Pharm Des 2022; 28:3140-3157. [PMID: 35366765 DOI: 10.2174/1381612828666220401142300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/27/2022] [Indexed: 01/28/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Chemotherapy and radiation therapy are the major treatments used for the management of cancer. Multidrug resistance (MDR) is a major hindrance faced in the treatment of cancer and is also responsible for cancer relapse. To date, several studies have been carried out on strategies to overcome or reverse MDR in cancer. Unfortunately, the MDR reversing agents have been proven to have minimal clinical benefits, and eventually, no improvement has been made in therapeutic efficacy to date. Thus, several investigational studies have also focused on overcoming drug resistance rather than reversing the MDR. In this review, we focus primarily on nanoformulations regarded as a novel approach to overcome or bypass the MDR in cancer. The nanoformulation systems serve as an attractive strategy as these nanosized materials selectively get accumulated in tumor tissues, thereby improving the clinical outcomes of patients suffering from MDR cancer. In the current work, we present an overview of recent trends in the application of various nano-formulations, belonging to different mechanistic classes and functionalization like carbon nanotubes, carbon nanohorns, carbon nanospheres, liposomes, dendrimers, etc., to overcome MDR in cancer. A detailed overview of these techniques will help researchers in exploring the applicability of nanotechnologybased approaches to treat MDR.
Collapse
Affiliation(s)
- Sana Kalave
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle [W], Mumbai, India
| | - Namita Hegde
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle [W], Mumbai, India
| | - Kapil Juvale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle [W], Mumbai, India
| |
Collapse
|
17
|
Jahanban-Esfahlan A, Davaran S, Dastmalchi S. Preparation and Antiproliferative Activity Evaluation of Juglone-Loaded BSA Nanoparticles. Adv Pharm Bull 2022; 12:818-827. [PMID: 36415643 PMCID: PMC9675913 DOI: 10.34172/apb.2022.087] [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: 02/24/2020] [Revised: 12/11/2021] [Accepted: 12/31/2021] [Indexed: 06/16/2024] Open
Abstract
Purpose: Today, the discovery of novel and effective chemotherapeutic compounds is the main challenge in cancer therapy. In recent years, the anti-tumoral activity of natural naphthoquinone juglone (JUG), present in different parts of walnut trees, has received considerable interest. The purpose of the current study was to prepare and evaluate the in vitro antiproliferative activity of JUG-loaded bovine serum albumin nanoparticles (JUG-BSA NPs). Methods: BSA NPs and JUG-BSA NPs were prepared using the desolvation technique. The NPs were characterized for their particle size (PS), zeta potential (ZP), drug loading (DL) capacity and encapsulation efficiency (EE). The anti-proliferative activity of JUG-BSA NPs was evaluated on A431 and HT29 cancer cell lines using cellular uptake and MTT assays. Results: The PS and ZP values of JUG-BSA NPs were 85 ± 6.55 nm and -29.6 mV, respectively. The DL capacity and EE were 3.7% to 5% and 50.4% to 94.6%, respectively. The cytotoxicity of JUG-BSA NPs was significantly less on both cultured A431 and HT29 cells at the studied concentrations when compared to free JUG. However, the effect was not very substantial, particularly at high levels. Conclusion: In conclusion, BSA NPs can be used as a suitable and safe carrier for the delivery of JUG, a cytotoxic hydrophobic natural compound.
Collapse
Affiliation(s)
| | - Soodabeh Davaran
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Near East University, POBOX: 99138, Nicosia, North Cyprus, Mersin 10, Turkey
| |
Collapse
|
18
|
Tong S, Zhao W, Zhao D, Zhang W, Zhang Z. Biomaterials-Mediated Tumor Infarction Therapy. Front Bioeng Biotechnol 2022; 10:916926. [PMID: 35757801 PMCID: PMC9218593 DOI: 10.3389/fbioe.2022.916926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
Agents for tumor vascular infarction are recently developed therapeutic agents for the vascular destruction of tumors. They can suppress the progression of the tumor by preventing the flow of nutrition and oxygen to its tissues. Agents of tumor vascular infarction can be divided into three categories according to the differences in their pathways of action: those that use the thrombin-activating pathway, fibrin-activating pathway, and platelet-activating pathway. However, poor targeting ability, low permeation, and potential side-effects restrict the development of the corresponding drugs. Biomaterials can subtly avoid these drawbacks to suppress the tumor. In this article, the authors summarize currently used biomaterials for tumor infarction therapy with the goal of identifying its mechanism, and discuss outstanding deficiencies in methods of this kind.
Collapse
Affiliation(s)
| | | | | | | | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
19
|
Hatami E, Nagesh PKB, Chauhan N, Jaggi M, Chauhan SC, Yallapu MM. In Situ Nanoparticle Self-Assembly for Combination Delivery of Therapeutics to Non-Small Cell Lung Cancer. ACS APPLIED BIO MATERIALS 2022; 5:1104-1119. [PMID: 35179871 DOI: 10.1021/acsabm.1c01158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chemotherapy often experiences several challenges including severe systemic toxicity and adverse effects. The combination chemotherapy arose as an effective clinical practice aimed at reducing doses of drugs to achieve synergistic actions with low toxicity. Our recent efforts demonstrated a synergistic therapeutic benefit of gambogic acid (GA) and gemcitabine (Gem) against lung cancer. However, simultaneous delivery of these two drugs at the tumor site is highly challenging. Therefore, the development of an injectable formulation that can effectively deliver both hydrophobic (GA) and hydrophilic (Gem) drugs in one formulation is a clinically unmet need. Herein, this study reports an in situ human serum albumin (HSA)- and tannic acid (TA)-mediated complexed GA and Gem nanoparticles (G-G@HTA NPs). G-G@HTA NP formation was confirmed by the particle size, Fourier transform infrared spectroscopy, and 1H NMR spectroscopy. The superior therapeutic activity of G-G@HTA NPs was demonstrated by multiple in vitro functional assays. Additionally, G-G@HTA NPs revealed an obvious and precise targeting of tumors in vivo. The promoted and more synergistic anti-tumor efficacy of G-G@HTA NPs was attained than that of combined treatments and single drug treatments. These events have resulted in no apparent systemic and organ toxicities. Together, this study suggests that in situ HSA-TA-based combinatorial treatment strategy is a suitable approach for application in lung cancer treatment.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Neeraj Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| |
Collapse
|
20
|
Lim HM, Park SH. Regulation of reactive oxygen species by phytochemicals for the management of cancer and diabetes. Crit Rev Food Sci Nutr 2022; 63:5911-5936. [PMID: 34996316 DOI: 10.1080/10408398.2022.2025574] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer and diabetes mellitus are served as typical life-threatening diseases with common risk factors. Developing therapeutic measures in cancers and diabetes have aroused attention for a long time. However, the problems with conventional treatments are in challenge, including side effects, economic burdens, and patient compliance. It is essential to secure safe and efficient therapeutic methods to overcome these issues. As an alternative method, antioxidant and pro-oxidant properties of phytochemicals from edible plants have come to the fore. Phytochemicals are naturally occurring compounds, considered promising agent applicable in treatment of various diseases with beneficial effects. Either antioxidative or pro-oxidative activity of various phytochemicals were found to contribute to regulation of cell proliferation, differentiation, cell cycle arrest, and apoptosis, which can exert preventive and therapeutic effects against cancer and diabetes. In this article, the antioxidant or pro-oxidant effects and underlying mechanisms of flavonoids, alkaloids, and saponins in cancer or diabetic models demonstrated by the recent studies are summarized.
Collapse
Affiliation(s)
- Heui Min Lim
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| |
Collapse
|
21
|
Judy E, Lopus M, Kishore N. Mechanistic insights into encapsulation and release of drugs in colloidal niosomal systems: biophysical aspects. RSC Adv 2021; 11:35110-35126. [PMID: 35493162 PMCID: PMC9042874 DOI: 10.1039/d1ra06057k] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Vesicular systems such as niosomes provide an alternative to improve drug delivery systems. The efficiency of a drug delivery vehicle is strongly dependent on its components which decide its interaction with partitioned drug(s) and locus of site of partitioning. A quantitative understanding of the physical chemistry underlying partitioning of drugs in complex systems of self-assemblies such as niosomes is scarcely available. In order to obtain quantitative mechanistic insights into partitioning and release of drugs [mitoxantrone (MTX) and ketoprofen (KTP)] in systems of niosomes, we have employed ultrasensitive calorimetry, spectroscopy and microscopy to establish correlations between functionality and energetics which could provide guidance towards rational drug design and choice of suitable non-ionic surfactant-based drug delivery vehicles. Electron microscopy and dynamic light scattering (DLS) methods were used for characterization and assessing the morphology of niosomes. We present here a calorimetry-based approach in assessing the partitioning of the anticancer drugs mitoxantrone and ketoprofen in niosomes and their release to human serum albumin (HSA) employing isothermal titration calorimetry (ITC), differential scanning calorimetry (DSC) and comparison with equilibrium dialysis. The thermodynamic signatures and kinetics of release were analyzed to obtain insights into the role of the functional groups on the drugs in the partitioning process. The assessment of thermal and conformational stability of proteins during drug binding and the effect of drug delivery vehicles on proteins is also crucial. To assess these effects, DSC studies on HSA in the presence and absence of drugs and niosomes were also performed. Finally, the efficacy of the system to impact the cell viability of the MDA-MB-231 triple-negative breast carcinoma cell line was analysed using MTT assay.
Collapse
Affiliation(s)
- Eva Judy
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400 076 India
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai Vidyanagari Mumbai 400 098 India
| | - Nand Kishore
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400 076 India
| |
Collapse
|
22
|
Mohamed Sofian Z, Harun N, Mahat MM, Nor Hashim NA, Jones SA. Investigating how amine structure influences drug-amine ion-pair formation and uptake via the polyamine transporter in A549 lung cells. Eur J Pharm Biopharm 2021; 168:53-61. [PMID: 34455038 DOI: 10.1016/j.ejpb.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 11/24/2022]
Abstract
Transiently associating amines with therapeutic agents through the formation of ion-pairs has been established both in vitro and in vivo as an effective means to systemically direct drug delivery to the lung via the polyamine transport system (PTS). However, there remains a need to better understand the structural traits required for effective PTS uptake of drug ion-pairs. This study aimed to use a structurally related series of amine counterions to investigate how they influenced the stability of theophylline ion-pairs and their active uptake in A549 cells. Using ethylamine (mono-amine), ethylenediamine (di-amine), spermidine (tri-amine) and spermine (tetra-amine) as counterions the ion-pair affinity was shown to increase as the number of protonated amine groups in the counterion structure increased. The mono and diamines generated a single hydrogen bond and the weakest ion-pair affinities (pKFTIR: 1.32 ± 0.04 and 1.43 ± 0.02) whereas the polyamines produced two hydrogen bonds and thus the strongest ion-pair affinities (pKFTIR: 1.93 ± 0.05 and 1.96 ± 0.04). In A549 cells depleted of endogenous polyamines using α-difluoromethylornithine (DFMO), the spermine-theophylline uptake was significantly increased (p < 0.05) compared to non-amine depleted cells and this evidenced the active PTS sequestering of the ion-pair. The mono-amine and di-amine failed to enhance theophylline uptake in these A549 cells, but the tri-amine and tetra-amine both almost doubled the theophylline uptake into the cells when compared to the uptake of free drug. As the data indicated that polyamines with at least 3 amines were required to form ion-pairs that could enhance A549 cell uptake, it suggested that at least two amines were required to physically stabilise the ion-pair and one to interact with the PTS.
Collapse
Affiliation(s)
- Zarif Mohamed Sofian
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Insitute of Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| | - Norsyifa Harun
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Mohd Muzamir Mahat
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40000 Shah Alam, Selangor, Malaysia
| | - Nikman Adli Nor Hashim
- Centre for Drug Research in Systems Biology, Structural Bioinformatics and Human Digital Imaging (CRYSTAL), Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Stuart A Jones
- Insitute of Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
23
|
Nanotechnological interventions for treatment of trypanosomiasis in humans and animals. Drug Deliv Transl Res 2021; 10:945-961. [PMID: 32383004 DOI: 10.1007/s13346-020-00764-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Trypanosomiasis is a parasitic infection caused by Trypanosoma. It is one of the major causes of deaths in underprivileged, rural areas of Africa, America and Asia. Depending on the parasite species responsible for the disease, it can take two forms namely African trypanosomiasis (sleeping sickness) and American trypanosomiasis (Chagas disease). The complete life-cycle stages of trypanosomes span between insect vector (tsetse fly, triatomine bug) and mammalian host (humans, animals). Only few drugs have been approved for the treatment of trypanosomiasis. Moreover, current trypanocidal therapy has major limitations of poor efficacy, serious side effects and drug resistance. Due to the lack of economic gains from tropical parasitic infection, it has always been neglected by the researchers and drug manufacturers. There is an immense need of more effective innovative strategies to decrease the deaths associated with this diseases. Nanotechnological approaches for delivery of existing drugs have shown significant improvement in efficacy with many-fold decrease in their dose. The review emphasizes on nanotechnological interventions in the treatment of trypanosomiasis in both humans and animals. Current trypanocidal therapy and their limitations have also been discussed briefly. Graphical abstract.
Collapse
|
24
|
Chavas TEJ, Su FY, Srinivasan S, Roy D, Lee B, Lovelace-Macon L, Rerolle GF, Limqueco E, Skerrett SJ, Ratner DM, West TE, Stayton PS. A macrophage-targeted platform for extending drug dosing with polymer prodrugs for pulmonary infection prophylaxis. J Control Release 2021; 330:284-292. [PMID: 33221351 PMCID: PMC7909327 DOI: 10.1016/j.jconrel.2020.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/03/2020] [Accepted: 11/15/2020] [Indexed: 01/03/2023]
Abstract
Pulmonary melioidosis is a bacterial disease with high morbidity and a mortality rate that can be as high as 40% in resource-poor regions of South Asia. This disease burden is linked to the pathogen's intrinsic antibiotic resistance and protected intracellular localization in alveolar macrophages. Current treatment regimens require several antibiotics with multi-month oral and intravenous administrations that are difficult to implement in under-resourced settings. Herein, we report that a macrophage-targeted polyciprofloxacin prodrug acts as a surprisingly effective pre-exposure prophylactic in highly lethal murine models of aerosolized human pulmonary melioidosis. A single dose of the polymeric prodrug maintained high lung drug levels and targeted an intracellular depot of ciprofloxacin to the alveolar macrophage compartment that was sustained over a period of 7 days above minimal inhibitory concentrations. This intracellular pharmacokinetic profile provided complete pre-exposure protection in a BSL-3 model with an aerosolized clinical isolate of Burkholderia pseudomallei from Thailand. This total protection was achieved despite the bacteria's relative resistance to ciprofloxacin and where an equivalent dose of pulmonary-administered ciprofloxacin was ineffective. For the first time, we demonstrate that targeting the intracellular macrophage compartment with extended antibiotic dosing can achieve pre-exposure prophylaxis in a model of pulmonary melioidosis. This fully synthetic and modular therapeutic platform could be an important therapeutic approach with new or re-purposed antibiotics for melioidosis prevention and treatment, especially as portable inhalation devices in high-risk, resource-poor settings.
Collapse
Affiliation(s)
- Thomas E J Chavas
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Fang-Yi Su
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Selvi Srinivasan
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Debashish Roy
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Brian Lee
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington 98104, United States
| | - Lara Lovelace-Macon
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington 98104, United States; Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Guilhem F Rerolle
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington 98104, United States; Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Elaine Limqueco
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Shawn J Skerrett
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington 98104, United States.
| | - Daniel M Ratner
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States.
| | - T Eoin West
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington 98104, United States; Department of Global Health, University of Washington, Seattle, Washington 98195, United States.
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States.
| |
Collapse
|
25
|
Younis MA, Khalil IA, Elewa YHA, Kon Y, Harashima H. Ultra-small lipid nanoparticles encapsulating sorafenib and midkine-siRNA selectively-eradicate sorafenib-resistant hepatocellular carcinoma in vivo. J Control Release 2021; 331:335-349. [PMID: 33484779 DOI: 10.1016/j.jconrel.2021.01.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/24/2020] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a fatal disease with limited therapeutic choices. The stroma-rich tumor microenvironment hinders the in vivo delivery of most nanomedicines. Ultra-small lipid nanoparticles (usLNPs) were designed for the selective co-delivery of the cytotoxic drug, sorafenib (SOR), and siRNA against the Midkine gene (MK-siRNA) to HCC in mice. The usLNPs composed of a novel pH-sensitive lipid, a diversity of phospholipids and a highly-selective targeting peptide. A microfluidic device, iLiNP, was used and a variety of factors were controlled to tune particle size aiming at maximizing tumor penetration efficiency. Optimizing the composition and physico-chemical properties of the usLNPs resulted in an enhanced tumor accumulation, selectivity and in vivo gene silencing. The optimized usLNPs exerted potent gene silencing in the tumor (median effective dose, ED50~0.1 mg/Kg) with limited effect on the healthy liver. The novel combination synergistically-eradicated HCC in mice (~85%) at a surprisingly-low dose of SOR (2.5 mg/Kg) which could not be achieved via individual monotherapy. Toxicity studies revealed the biosafety of the usLNPs upon either acute or chronic treatment. Furthermore, the SOR-resistant HCC established in mice was eradicated by 70% using this approach. We conclude that our strategy is promising for potential clinical applications in HCC treatment.
Collapse
Affiliation(s)
- Mahmoud A Younis
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Ikramy A Khalil
- Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Yaser H A Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo 060-0818, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo 060-0818, Japan
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
26
|
Sofian ZM, Benaouda F, Wang JT, Lu Y, Barlow DJ, Royall PG, Farag DB, Rahman KM, Al‐Jamal KT, Forbes B, Jones SA. A Cyclodextrin-Stabilized Spermine-Tagged Drug Triplex that Targets Theophylline to the Lungs Selectively in Respiratory Emergency. ADVANCED THERAPEUTICS 2020; 3:2000153. [PMID: 33043128 PMCID: PMC7536984 DOI: 10.1002/adtp.202000153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/04/2020] [Indexed: 12/21/2022]
Abstract
Ion-pairing a lifesaving drug such as theophylline with a targeting moiety could have a significant impact on medical emergencies such as status asthmaticus or COVID-19 induced pneumomediastinum. However, to achieve rapid drug targeting in vivo the ion-pair must be protected against breakdown before the entry into the target tissue. This study aims to investigate if inserting theophylline, when ion-paired to the polyamine transporter substrate spermine, into a cyclodextrin (CD), to form a triplex, could direct the bronchodilator to the lungs selectively after intravenous administration. NMR demonstrates that upon the formation of the triplex spermine protruded from the CD cavity and this results in energy-dependent uptake in A549 cells (1.8-fold enhancement), which persists for more than 20 min. In vivo, the triplex produces a 2.4-fold and 2.2-fold increase in theophylline in the lungs 20 min after injection in rats and mice, respectively (p < 0.05). The lung targeting is selective with no increase in uptake into the brain or the heart where the side-effects of theophylline are treatment-limiting. Selectively doubling the concentration of theophylline in the lungs could improve the benefit-risk ratio of this narrow therapeutic index medicine, which continues to be important in critical care.
Collapse
Affiliation(s)
- Zarif M. Sofian
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversiti MalayaKuala Lumpur50603Malaysia
| | - Faiza Benaouda
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - Julie Tzu‐Wen Wang
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - Yuan Lu
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - David J. Barlow
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - Paul G. Royall
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - Doaa B. Farag
- Faculty of PharmacyMisr International UniversityCairo11431Egypt
| | - Khondaker Miraz Rahman
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - Khuloud T. Al‐Jamal
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - Ben Forbes
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| | - Stuart A. Jones
- School of Cancer and Pharmaceutical SciencesFaculty of Life Sciences & MedicineKing's College LondonFranklin‐Wilkins Building, 150 Stamford StreetLondonSE1 9NHUK
| |
Collapse
|
27
|
Yamashita S, Katsumi H, Shimizu E, Nakao Y, Yoshioka A, Fukui M, Kimura H, Sakane T, Yamamoto A. Dendrimer-based micelles with highly potent targeting to sites of active bone turnover for the treatment of bone metastasis. Eur J Pharm Biopharm 2020; 157:85-96. [PMID: 33039547 DOI: 10.1016/j.ejpb.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 01/29/2023]
Abstract
Bone-drug targeting therapies using nanoparticles based on targeting ligands remain challenging due to their uptake clearance at non-target sites such as the liver, kidney, and spleen. Furthermore, the distribution sites of nanoparticles in bones have not been fully investigated, thus halting the development of more effective bone metastasis treatment strategies. In this study, we developed nanoparticles self-assembled from cholesterol-terminated, polyethylene glycol-conjugated, aspartic acid (Asp)-modified polyamidoamine dendrimer (Asp-PAMAM-Micelles) with targeting to active bone turnover sites associated with bone metastasis pathogenesis. On analysis through whole-body single photon emission computed tomography/computed tomography (SPECT/CT) imaging, 111In-Asp-PAMAM-Micelles showed high specificity to active bone turnover sites (especially the joints in the lower limbs, shoulder, and pelvis) after intravenous injection in mice. The lower limb bone uptake clearance for 111In-Asp-PAMAM-Micelles encapsulating paclitaxel (PTX) was 3.5-fold higher than that for 111In-unmodified PAMAM-Micelles (PTX). 3H-PTX encapsulated Asp-PAMAM-Micelles effectively accumulated in the lower limb bones in a similar manner as the 111In-Asp-PAMAM-Micelles (PTX). In a bone metastatic tumor mouse model, the tumor growth in the lower limb bones was significantly inhibited by injection of Asp-PAMAM-Micelles (PTX) compared to unmodified PAMAM-Micelles (PTX). Our results demonstrate that Asp-PAMAM-Micelles are sophisticated drug delivery systems for highly potent targeting to active bone turnover sites.
Collapse
Affiliation(s)
- Shugo Yamashita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Erika Shimizu
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Yuto Nakao
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Ayane Yoshioka
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Minako Fukui
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Toshiyasu Sakane
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan; Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| |
Collapse
|
28
|
Liu HJ, Wang M, Hu X, Shi S, Xu P. Enhanced Photothermal Therapy through the In Situ Activation of a Temperature and Redox Dual-Sensitive Nanoreservoir of Triptolide. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003398. [PMID: 32797711 PMCID: PMC7983299 DOI: 10.1002/smll.202003398] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/29/2020] [Indexed: 05/30/2023]
Abstract
Photothermal therapy (PTT) has attracted tremendous attention due to its noninvasiveness and localized treatment advantages. However, heat shock proteins (HSPs) associated self-preservation mechanisms bestow cancer cells thermoresistance to protect them from the damage of PTT. To minimize the thermoresistance of cancer cells and improve the efficacy of PTT, an integrated on-demand nanoplatform composed of a photothermal conversion core (gold nanorod, GNR), a cargo of a HSPs inhibitor (triptolide, TPL), a mesoporous silica based nanoreservoir, and a photothermal and redox di-responsive polymer shell is developed. The nanoplatform can be enriched in the tumor site, and internalized into cancer cells, releasing the encapsulated TPL under the trigger of intracellular elevated glutathione and near-infrared laser irradiation. Ultimately, the liberated TPL could diminish thermoresistance of cancer cells by antagonizing the PTT induced heat shock response via multiple mechanisms to maximize the PTT effect for cancer treatment.
Collapse
Affiliation(s)
- Hai-Jun Liu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Mingming Wang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Xiangxiang Hu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Shanshan Shi
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Peisheng Xu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| |
Collapse
|
29
|
Zou H, Banerjee P, Leung SSY, Yan X. Application of Pharmacokinetic-Pharmacodynamic Modeling in Drug Delivery: Development and Challenges. Front Pharmacol 2020; 11:997. [PMID: 32719604 PMCID: PMC7348046 DOI: 10.3389/fphar.2020.00997] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022] Open
Abstract
With the advancement of technology, drug delivery systems and molecules with more complex architecture are developed. As a result, the drug absorption and disposition processes after administration of these drug delivery systems and engineered molecules become exceedingly complex. As the pharmacokinetic and pharmacodynamic (PK-PD) modeling allows for the separation of the drug-, carrier- and pharmacological system-specific parameters, it has been widely used to improve understanding of the in vivo behavior of these complex delivery systems and help their development. In this review, we summarized the basic PK-PD modeling theory in drug delivery and demonstrated how it had been applied to help the development of new delivery systems and modified large molecules. The linkage between PK and PD was highlighted. In particular, we exemplified the application of PK-PD modeling in the development of extended-release formulations, liposomal drugs, modified proteins, and antibody-drug conjugates. Furthermore, the model-based simulation using primary PD models for direct and indirect PD responses was conducted to explain the assertion of hypothetical minimal effective concentration or threshold in the exposure-response relationship of many drugs and its misconception. The limitations and challenges of the mechanism-based PK-PD model were also discussed.
Collapse
Affiliation(s)
- Huixi Zou
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Parikshit Banerjee
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Sharon Shui Yee Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Xiaoyu Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
30
|
Katsumi H, Yamashita S, Morishita M, Yamamoto A. Bone-Targeted Drug Delivery Systems and Strategies for Treatment of Bone Metastasis. Chem Pharm Bull (Tokyo) 2020; 68:560-566. [DOI: 10.1248/cpb.c20-00017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Shugo Yamashita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | | | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| |
Collapse
|
31
|
Hatami E, Jaggi M, Chauhan SC, Yallapu MM. Gambogic acid: A shining natural compound to nanomedicine for cancer therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874:188381. [PMID: 32492470 DOI: 10.1016/j.bbcan.2020.188381] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023]
Abstract
The United States Food and Drug Administration has permitted number of therapeutic agents for cancer treatment. Most of them are expensive and have some degree of systemic toxicity which makes overbearing in clinical settings. Although advanced research continuously applied in cancer therapeutics, but drug resistance, metastasis, and recurrence remain unanswerable. These accounts to an urgent clinical need to discover natural compounds with precisely safe and highly efficient for the cancer prevention and cancer therapy. Gambogic acid (GA) is the principle bioactive and caged xanthone component, a brownish gamboge resin secreted from the of Garcinia hanburyi tree. This molecule showed a spectrum of biological and clinical benefits against various cancers. In this review, we document distinct biological characteristics of GA as a novel anti-cancer agent. This review also delineates specific molecular mechanism(s) of GA that are involved in anti-cancer, anti-metastasis, anti-angiogenesis, and chemo-/radiation sensitizer activities. Furthermore, recent evidence, development, and implementation of various nanoformulations of gambogic acid (nanomedicine) have been described.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.
| |
Collapse
|
32
|
Xu C, Zhang T, Lu G, Chen K, Tao J, Zhang Y, Teng Z, Yang B. Disulfiram-gold-nanorod integrate for effective tumor targeting and photothermal-chemical synergistic therapy. Biomater Sci 2020; 8:3310-3319. [PMID: 32400782 DOI: 10.1039/d0bm00062k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein, we successfully constructed a combination therapeutic nanoplatform with high tumor targeting for cancer treatment by integrating gold nanorods with disulfiram (denoted Au-DSF). The Au-DSF integrates possess a uniform length (70 nm), excellent photothermal conversion ability and a high DSF loading content (23.2%), and the loaded DSFs show glutathione-, acid-, and laser-responsive release properties. The Au-DSF integrates show significantly enhanced cellular uptake efficiency in breast cancer cells due to the ability of DSF to chelate to the intracellular copper (Cu) which is present at high concentrations. Furthermore, the Au-DSF exhibits improved circulation time (mean residence time = 28.4 h) and increased tumor accumulation (12.0%), due to the targeting of DSF to the abundant Cu ions at the tumor site. Moreover, the DSF/Cu complexes potently elevate reactive oxygen species, which effectively induce cancer cell apoptosis. In vivo experiments show that the Au-DSF integrates dramatically decrease tumor size via photothermal therapy and chemotherapy. Hematoxylin-eosin and TUNEL staining show that the Au-DSF integrates induce necrosis and apoptosis in cancer cells. The high therapeutic efficiency of the Au-DSF integrates for breast cancer is further demonstrated by the reduced elasticity seen in ultrasound elastography, and the absence of perfusion of the contrast agent in contrast-enhanced ultrasound imaging in tumors.
Collapse
Affiliation(s)
- Chaoli Xu
- Department of Ultrasound Diagnostic, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Da Silva FLO, Marques MBDF, Kato KC, Carneiro G. Nanonization techniques to overcome poor water-solubility with drugs. Expert Opin Drug Discov 2020; 15:853-864. [DOI: 10.1080/17460441.2020.1750591] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Flávia Lidiane Oliveira Da Silva
- Department of Pharmacy, Faculty of Biological and Health Sciences, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Maria Betânia De Freitas Marques
- Department of Pharmacy, Faculty of Biological and Health Sciences, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
- Department of Chemistry, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Kelly Cristina Kato
- Department of Pharmacy, Faculty of Biological and Health Sciences, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Guilherme Carneiro
- Department of Pharmacy, Faculty of Biological and Health Sciences, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| |
Collapse
|
34
|
Biomimetic cellular vectors for enhancing drug delivery to the lungs. Sci Rep 2020; 10:172. [PMID: 31932600 PMCID: PMC6957529 DOI: 10.1038/s41598-019-55909-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/10/2019] [Indexed: 02/01/2023] Open
Abstract
Despite recent advances in drug delivery, the targeted treatment of unhealthy cells or tissues continues to remain a priority. In cancer (much like other pathologies), delivery vectors are designed to exploit physical and biological features of unhealthy tissues that are not always homogenous across the disease. In some cases, shifting the target from unhealthy tissues to the whole organ can represent an advantage. Specifically, the natural organ-specific retention of nanotherapeutics following intravenous administration as seen in the lung, liver, and spleen can be strategically exploited to enhance drug delivery. Herein, we outline the development of a cell-based delivery system using macrophages as a delivery vehicle. When loaded with a chemotherapeutic payload (i.e., doxorubicin), these cellular vectors (CELVEC) were shown to provide continued release within the lung. This study provides proof-of-concept evidence of an alternative class of biomimetic delivery vectors that capitalize on cell size to provide therapeutic advantages for pulmonary treatments.
Collapse
|
35
|
Hashida M. Role of pharmacokinetic consideration for the development of drug delivery systems: A historical overview. Adv Drug Deliv Rev 2020; 157:71-82. [PMID: 32565225 DOI: 10.1016/j.addr.2020.06.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/09/2020] [Accepted: 06/13/2020] [Indexed: 12/13/2022]
Abstract
Drug delivery system is defined as a system or technology to achieve optimum therapeutic effects of drugs through precise control of their movements in the body. In order to optimize function of drug delivery systems aiming at targeting, their whole-body distribution profiles should be systematically evaluated and analyzed, where pharmacokinetic analysis based on the clearance concepts plays important role. Organ perfusion experiments combined with statistical moment analysis further supply detailed information on drug disposition at organ and cellular levels. Based on general relationship between physicochemical properties and distribution profile, macromolecular prodrugs or polymer conjugates of proteins are rationally designed and further introduction of ligand structure brings cell-specific delivery for them. These approaches are also applicable for particulate carriers such as liposomes and offer various opportunities for biological drugs such as nucleic acid drugs for their delivery. Mechanistic approach for dermal absorption analysis based on physiological skin model offers another opportunity in rational design of drug delivery. Potential of drug delivery technology in future medicines such as cell therapy and nanomaterial platform application is further discussed in relation to pharmacokinetic consideration.
Collapse
|
36
|
|
37
|
El-Sayed NS, Shirazi AN, Sajid MI, Park SE, Parang K, Tiwari RK. Synthesis and Antiproliferative Activities of Conjugates of Paclitaxel and Camptothecin with a Cyclic Cell-Penetrating Peptide. Molecules 2019; 24:1427. [PMID: 30978971 PMCID: PMC6480016 DOI: 10.3390/molecules24071427] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/04/2019] [Accepted: 04/06/2019] [Indexed: 12/20/2022] Open
Abstract
Cell-penetrating peptide [WR]₅ has been previously shown to be an efficient molecular transporter for various hydrophilic and hydrophobic molecules. The peptide was synthesized using Fmoc/tBu solid-phase chemistry, and one arginine was replaced with one lysine to enable the conjugation with the anticancer drugs. Paclitaxel (PTX) was functionalized with an esterification reaction at the C2' hydroxyl group of PTX with glutaric anhydride and conjugated with the cyclic peptide [W(WR)₄K(βAla)] in DMF to obtain the peptide-drug conjugate PTX1. Furthermore, camptothecin (CPT) was modified at the C(20)-hydroxyl group through the reaction with triphosgene. Then, it was conjugated with two functionalized cyclic peptides through a formyl linker affording two different conjugates, namely CPT1 and CPT2. All the conjugates showed better water solubility as compared to the parent drug. The cytotoxicity assay of the drugs and their conjugates with the peptides were evaluated in the human breast cancer MCF-7 cell line. PTX inhibited cell proliferation by 39% while the PTX-peptide conjugate inhibited the proliferation by ~18% after 72 h incubation. On the other hand, CPT, CPT1, and CPT2 reduced the cell proliferation by 68%, 39%, and 62%, respectively, in the MCF-7 cell lines at 5 µM concentration after 72 h incubation.
Collapse
Affiliation(s)
- Naglaa Salem El-Sayed
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
- Cellulose and Paper Department, National Research Center, Dokki 12622, Cairo, Egypt.
| | - Amir Nasrolahi Shirazi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, Marshall B. Ketchum University, Fullerton, CA 92831, USA.
| | - Muhammad Imran Sajid
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
- Faculty of Pharmacy, University of Central Punjab, Lahore 54000, Pakistan.
| | - Shang Eun Park
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Rakesh Kumar Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| |
Collapse
|
38
|
Raja MAG, Katas H, Amjad MW. Design, mechanism, delivery and therapeutics of canonical and Dicer-substrate siRNA. Asian J Pharm Sci 2019; 14:497-510. [PMID: 32104477 PMCID: PMC7032099 DOI: 10.1016/j.ajps.2018.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 12/07/2018] [Accepted: 12/24/2018] [Indexed: 12/12/2022] Open
Abstract
Upon the discovery of RNA interference (RNAi), canonical small interfering RNA (siRNA) has been recognized to trigger sequence-specific gene silencing. Despite the benefits of siRNAs as potential new drugs, there are obstacles still to be overcome, including off-target effects and immune stimulation. More recently, Dicer substrate siRNA (DsiRNA) has been introduced as an alternative to siRNA. Similarly, it also is proving to be potent and target-specific, while rendering less immune stimulation. DsiRNA is 25–30 nucleotides in length, and is further cleaved and processed by the Dicer enzyme. As with siRNA, it is crucial to design and develop a stable, safe, and efficient system for the delivery of DsiRNA into the cytoplasm of targeted cells. Several polymeric nanoparticle systems have been well established to load DsiRNA for in vitro and in vivo delivery, thereby overcoming a major hurdle in the therapeutic uses of DsiRNA. The present review focuses on a comparison of siRNA and DsiRNA on the basis of their design, mechanism, in vitro and in vivo delivery, and therapeutics.
Collapse
Affiliation(s)
- Maria Abdul Ghafoor Raja
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 73211, Saudi Arabia
| | - Haliza Katas
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Muhammad Wahab Amjad
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 73211, Saudi Arabia
| |
Collapse
|
39
|
Chen J, Yang X, Huang L, Lai H, Gan C, Luo X. Development of dual-drug-loaded stealth nanocarriers for targeted and synergistic anti-lung cancer efficacy. Drug Deliv 2018; 25:1932-1942. [PMID: 30472899 PMCID: PMC6263111 DOI: 10.1080/10717544.2018.1477856] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023] Open
Abstract
Combination chemotherapy is widely exploited for suppressing drug resistance and achieving synergistic anticancer efficacy in the clinic. In this paper, the nanostructured targeting methotrexate (MTX) plus pemetrexed (PMX) chitosan nanoparticles (CNPs) were developed by modifying methoxy polye (thylene glycol) (mPEG), in which PEGylation CNPs was used as stealth nanocarriers (PCNPs) and MTX was employed as a targeting ligand and chemotherapeutic agent as well. Studies were undertaken on human lung adenocarcinoma epithelial (A549) and Lewis lung carcinoma (LLC) cell lines, revealing the anti-tumor efficacy of nanoparticle drug delivery system. The co-delivery nanoparticles (MTX-PMX-PCNPs) had well-dispersed with sustained release behavior. Cell counting kit-8 (CCK8) has been used to measure A549 cell viability and the research showed that MTX-PMX-PCNPs were much more effective than free drugs when it came to the inhibition of growth and proliferation. Cell cycle assay by flow cytometry manifested that the MTX-PMX-PCNPs exhibited stronger intracellular taken up ability than free drugs at the same concentration. In vivo anticancer effect results indicated that MTX-PMX-PCNPs exhibited a significantly prolong blood circulation, more tumoral location accumulation, and resulted in a robust synergistic anticancer efficacy in lung cancer in mice. The results clearly demonstrated that such unique synergistic anticancer efficacy of co-delivery of MTX and PMX via stealth nanocarriers, providing a prospective strategy for lung cancer treatment.
Collapse
Affiliation(s)
- Juan Chen
- Department of Pharmacy, Zhongshan Hospital Xiamen University, Xiamen, P. R. China
| | - Xiaobing Yang
- College of Ecology and Resource Engineering, Wuyi University, Wuyishan, P. R. China
| | - Liuqing Huang
- Department of Materials Science and Engineering College of Materials, Xiamen University, Xiamen, P. R. China
| | - Huixian Lai
- Department of Materials Science and Engineering College of Materials, Xiamen University, Xiamen, P. R. China
| | - Chuanhai Gan
- Department of Materials Science and Engineering College of Materials, Xiamen University, Xiamen, P. R. China
| | - Xuetao Luo
- Department of Materials Science and Engineering College of Materials, Xiamen University, Xiamen, P. R. China
| |
Collapse
|
40
|
Roles of alcohol desolvating agents on the size control of bovine serum albumin nanoparticles in drug delivery system. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.07.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
41
|
Kleynhans J, Grobler AF, Ebenhan T, Sathekge MM, Zeevaart JR. Radiopharmaceutical enhancement by drug delivery systems: A review. J Control Release 2018; 287:177-193. [DOI: 10.1016/j.jconrel.2018.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
|
42
|
Biodistribution and Tumors MRI Contrast Enhancement of Magnetic Nanocubes, Nanoclusters, and Nanorods in Multiple Mice Models. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:8264208. [PMID: 30344459 PMCID: PMC6174815 DOI: 10.1155/2018/8264208] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/19/2018] [Indexed: 01/26/2023]
Abstract
Magnetic resonance imaging (MRI) is a powerful technique for tumor diagnostics. Iron oxide nanoparticles (IONPs) are safe and biocompatible tools that can be used for further enhancing MR tumor contrasting. Although numerous IONPs have been proposed as MRI contrast agents, low delivery rates to tumor site limit its application. IONPs accumulation in malignancies depends on both IONPs characteristics and tumor properties. In the current paper, three differently shaped Pluronic F-127-modified IONPs (nanocubes, nanoclusters, and nanorods) were compared side by side in three murine tumor models (4T1 breast cancer, B16 melanoma, and CT26 colon cancer). Orthotopic B16 tumors demonstrated more efficient IONPs uptake than heterotopic implants. Magnetic nanocubes (MNCb) had the highest r2-relaxivity in vitro (300 mM−1·s−1) compared with magnetic nanoclusters (MNCl, 104 mM−1·s−1) and magnetic nanorods (MNRd, 51 mM−1·s−1). As measured by atomic emission spectroscopy, MNCb also demonstrated better delivery efficiency to tumors (3.79% ID) than MNCl (2.94% ID) and MNRd (1.21% ID). Nevertheless, MNCl overperformed its counterparts in tumor imaging, providing contrast enhancement in 96% of studied malignancies, whereas MNCb and MNRd were detected by MRI in 73% and 63% of tumors, respectively. Maximum MR contrasting efficiency for MNCb and MNCl was around 6-24 hours after systemic administration, whereas for MNRd maximum contrast enhancement was found within first 30 minutes upon treatment. Presumably, MNRd poor MRI performance was due to low r2-relaxivity and rapid clearance by lungs (17.3% ID) immediately after injection. MNCb and MNCl were mainly captured by the liver and spleen without significant accumulation in the lungs, kidneys, and heart. High biocompatibility and profound accumulation in tumor tissues make MNCb and MNCl the promising platforms for MRI-based tumor diagnostics and drug delivery.
Collapse
|
43
|
Wei Y, Quan L, Zhou C, Zhan Q. Factors relating to the biodistribution & clearance of nanoparticles & their effects on in vivo application. Nanomedicine (Lond) 2018; 13:1495-1512. [DOI: 10.2217/nnm-2018-0040] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles have promising biomedical applications for drug delivery, tumor imaging and tumor treatment. Pharmacokinetics are important for the in vivo application of nanoparticles. Biodistribution and clearance are largely defined as the key points of pharmacokinetics to maximize therapeutic efficacy and to minimize side effects. Different engineered nanoparticles have different biodistribution and clearance processes. The interactions of organs with nanoparticles, which are determined by the characteristics of the organs and the biochemical/physical properties of the nanoparticles, are a major factor influencing biodistribution and clearance. In this review, the clearance functions of organs and the properties related to pharmacokinetics, including nanoparticle size, shape, biodegradation and surface modifications are discussed.
Collapse
Affiliation(s)
- Yanchun Wei
- Provincial Key Laboratory for Interventional Medical Devices, Huaiyin Institute of Technology, Huai'an, Jiangsu 223001, PR China
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
| | - Li Quan
- Provincial Key Laboratory for Interventional Medical Devices, Huaiyin Institute of Technology, Huai'an, Jiangsu 223001, PR China
| | - Chao Zhou
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
| | - Qiuqiang Zhan
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
- Key Laboratory of Optoelectronic Devices & Systems of Ministry of Education & Guangdong Province, Shenzhen University, Shenzhen 518052, PR China
| |
Collapse
|
44
|
Watanabe H, Morigaki A, Yuba M, Yamada K, Miyake M, Tobori N, Aramaki K. Structural Analyses of Hydrated Crystals in Mixed Green Surfactant Systems: α-Sulfonated Fatty Acid Methyl Ester Salt and Fatty Acid Soap Mixtures. J SURFACTANTS DETERG 2018. [DOI: 10.1002/jsde.12010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Hideaki Watanabe
- Functional Materials Science Research Laboratories, R&D; LION CORPORATION, 7-2-1 Hirai, Edogawa-ku; Tokyo 132-0035 Japan
- Graduate School of Environment and Information Sciences; Yokohama National University, 79-7 Tokiwadai, Hodogaya-ku; Yokohama 240-8501 Japan
| | - Atsunori Morigaki
- Functional Materials Science Research Laboratories, R&D; LION CORPORATION, 7-2-1 Hirai, Edogawa-ku; Tokyo 132-0035 Japan
| | - Misako Yuba
- Graduate School of Environment and Information Sciences; Yokohama National University, 79-7 Tokiwadai, Hodogaya-ku; Yokohama 240-8501 Japan
| | - Ken Yamada
- Packaging and Container Technology Research Laboratories, R&D; LION CORPORATION, 7-2-1 Hirai, Edogawa-ku; Tokyo 132-0035 Japan
| | - Miyuki Miyake
- Functional Materials Science Research Laboratories, R&D; LION CORPORATION, 7-2-1 Hirai, Edogawa-ku; Tokyo 132-0035 Japan
| | - Norio Tobori
- Functional Materials Science Research Laboratories, R&D; LION CORPORATION, 7-2-1 Hirai, Edogawa-ku; Tokyo 132-0035 Japan
| | - Kenji Aramaki
- Graduate School of Environment and Information Sciences; Yokohama National University, 79-7 Tokiwadai, Hodogaya-ku; Yokohama 240-8501 Japan
| |
Collapse
|
45
|
Nigam S, Bahadur D. Doxorubicin-loaded dendritic-Fe 3O 4 supramolecular nanoparticles for magnetic drug targeting and tumor regression in spheroid murine melanoma model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:759-768. [PMID: 29339187 DOI: 10.1016/j.nano.2018.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/15/2017] [Accepted: 01/05/2018] [Indexed: 01/22/2023]
Abstract
This work evaluates the magnetically-guided delivery of DOX-loaded dendritic-Fe3O4 nanoparticles and their tumor regression efficacy in subcutaneous melanoma in C57BL/6 mice. The hematological, biochemical and histopathological parameters were minimally affected. The nanoparticles localized in lungs, liver and spleen suggesting non-specific uptake. However, in tumor-bearing mice, substantially higher localization in magnetically-targeted tumor was observed when compared to passive localization in non-targeted tumor. The animals of treated group showed significantly high iron levels (161 μg of Fe/mg dry organ weight) in the tumor against the control (<25 μg of Fe/mg dry organ weight). This high localization led to high concentrations of DOX in the tumor which not only induced significant tumor regression but also arrested further growth. Within 14 days, the average tumor volume was reduced to 55±8.3 mm3 (treated) as compared to 4794±844 mm3 (control), i.e. ~88-fold decrease. The tumor disappeared by the end of 20th day post-treatment and ~100% survival rate was observed.
Collapse
Affiliation(s)
- Saumya Nigam
- IITB-Monash Research Academy, IIT Bombay, Mumbai, India
| | - D Bahadur
- Department of Metallurgical Engineering and Materials Science, IIT Bombay, Mumbai, India.
| |
Collapse
|
46
|
Yazdani S, Bansal R, Prakash J. Drug targeting to myofibroblasts: Implications for fibrosis and cancer. Adv Drug Deliv Rev 2017; 121:101-116. [PMID: 28720422 DOI: 10.1016/j.addr.2017.07.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/20/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
Myofibroblasts are the key players in extracellular matrix remodeling, a core phenomenon in numerous devastating fibrotic diseases. Not only in organ fibrosis, but also the pivotal role of myofibroblasts in tumor progression, invasion and metastasis has recently been highlighted. Myofibroblast targeting has gained tremendous attention in order to inhibit the progression of incurable fibrotic diseases, or to limit the myofibroblast-induced tumor progression and metastasis. In this review, we outline the origin of myofibroblasts, their general characteristics and functions during fibrosis progression in three major organs: liver, kidneys and lungs as well as in cancer. We will then discuss the state-of-the art drug targeting technologies to myofibroblasts in context of the above-mentioned organs and tumor microenvironment. The overall objective of this review is therefore to advance our understanding in drug targeting to myofibroblasts, and concurrently identify opportunities and challenges for designing new strategies to develop novel diagnostics and therapeutics against fibrosis and cancer.
Collapse
Affiliation(s)
- Saleh Yazdani
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; ScarTec Therapeutics BV, Enschede, The Netherlands.
| |
Collapse
|
47
|
Pang L, Zhang C, Qin J, Han L, Li R, Hong C, He H, Wang J. A novel strategy to achieve effective drug delivery: exploit cells as carrier combined with nanoparticles. Drug Deliv 2017; 24:83-91. [PMID: 28155538 PMCID: PMC8241159 DOI: 10.1080/10717544.2016.1230903] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/23/2016] [Accepted: 08/28/2016] [Indexed: 12/21/2022] Open
Abstract
Cell-mediated drug delivery systems employ specific cells as drug vehicles to deliver drugs to targeted sites. Therapeutics or imaging agents are loaded into these cells and then released in diseased sites. These specific cells mainly include red blood cells, leukocytes, stem cells and so on. The cell acts as a Trojan horse to transfer the drug from circulating blood to the diseased tissue. In such a system, these cells keep their original properties, which allow them to mimic the migration behavior of specific cells to carry drug to the targeted site after in vivo administration. This strategy elegantly combines the advantages of both carriers, i.e. the adjustability of nanoparticles (NPs) and the natural functions of active cells, which therefore provides a new perspective to challenge current obstacles in drug delivery. This review will describe a fundamental understanding of these cell-based drug delivery systems, and discuss the great potential of combinational application of cell carrier and NPs.
Collapse
Affiliation(s)
- Liang Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Chun Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Jing Qin
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Limei Han
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Ruixiang Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Chao Hong
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jianxin Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| |
Collapse
|
48
|
Wang L, Huo M, Chen Y, Shi J. Coordination-Accelerated "Iron Extraction" Enables Fast Biodegradation of Mesoporous Silica-Based Hollow Nanoparticles. Adv Healthc Mater 2017; 6. [PMID: 28941142 DOI: 10.1002/adhm.201700720] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/04/2017] [Indexed: 11/10/2022]
Abstract
Biodegradation behavior of inorganic silica-based nanoplatforms is of critical importance in their clinical translations, but still remains a great challenge in achieving this goal by composition regulation of biocompatible silica framework. In the present work, a chemical coordination-accelerated biodegradation strategy to endow hollow mesoporous silica nanoparticles (HMSNs) with unique coordination-responsive biodegradability, on-demand coordination-responsive drug releasing behavior, and significantly enhanced chemotherapeutic efficacy by directly doping iron (Fe) ions into the framework of mesoporous silica is reported. A simple but versatile dissolution-regrowth strategy has been developed to enable the framework Fe doping via chemical bonding. The deferiprone-mediated biodegradation of Fe-doped HMSNs (Fe-HMSNs) has been comprehensively evaluated both in simulated body fluid and intracellular level, which have exhibited a specific coordination-accelerated biodegradation behavior. In addition to high biocompatibility of Fe-HMSNs, the anticancer drug doxorubicin (DOX)-loaded Fe-HMSNs show enhanced tumor-suppressing effect on 4T1 mammary cancer xenograft. This work paves a new way for tuning the biodegradation performance of mesoporous silica-based nanoplatforms simply by biocompatible Fe-ion doping into silica framework based on the specific coordination property between introduced metal Fe ions with Fe-coordination proteins.
Collapse
Affiliation(s)
- Liying Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Shanghai Institute of Ceramics; Chinese Academy of Sciences; Shanghai 200050 P. R. China
- School of Physical Science and Technology; ShanghaiTech University; Shanghai 201210 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Minfeng Huo
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Shanghai Institute of Ceramics; Chinese Academy of Sciences; Shanghai 200050 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Shanghai Institute of Ceramics; Chinese Academy of Sciences; Shanghai 200050 P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Shanghai Institute of Ceramics; Chinese Academy of Sciences; Shanghai 200050 P. R. China
| |
Collapse
|
49
|
Rastegari B, Karbalaei-Heidari HR, Zeinali S, Sheardown H. The enzyme-sensitive release of prodigiosin grafted β-cyclodextrin and chitosan magnetic nanoparticles as an anticancer drug delivery system: Synthesis, characterization and cytotoxicity studies. Colloids Surf B Biointerfaces 2017; 158:589-601. [DOI: 10.1016/j.colsurfb.2017.07.044] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 07/14/2017] [Accepted: 07/16/2017] [Indexed: 12/14/2022]
|
50
|
Naz S, Shahzad H, Ali A, Zia M. Nanomaterials as nanocarriers: a critical assessment why these are multi-chore vanquisher in breast cancer treatment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:899-916. [PMID: 28914553 DOI: 10.1080/21691401.2017.1375937] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Breast cancer is a group of diseases with various subtypes and leads to high mortality throughout the globe. Various conventional techniques are in practice to cure breast cancer but these techniques are linked with various shortcomings. Mostly these treatments are not site directed and cause toxicity towards normal cells. In order to overcome these issues, we need smart system that can deliver anticancer drugs to specific sites. Targeted drug delivery can be achieved via passive or active drug delivery using nanocarriers. This mode of drug delivery is more effective against breast cancer and may help in the reduction of mortality rate. Potentially used nanocarriers for targeted drug delivery belong to organic and inorganic molecules. Various FDA approved nano products are in use to cure breast cancer. However, body's defense system is main limitation for potential use of nano systems. However, this can be overcome by surface modification of nanocarriers. In this review, breast cancer and its types, targeted drug delivery and nanocarriers used to cure breast cancer are discussed. By progressing nanotechnology, we will be able to fight against this life threatening issue and serve the humanity, which is the basic aim of scientific knowledge.
Collapse
Affiliation(s)
- Sania Naz
- a Department of Biotechnology , Quaid-i-Azam University , Islamabad , Pakistan
| | - Hira Shahzad
- b Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture , Rawalpindi , Pakistan
| | - Attarad Ali
- a Department of Biotechnology , Quaid-i-Azam University , Islamabad , Pakistan
| | - Muhammad Zia
- a Department of Biotechnology , Quaid-i-Azam University , Islamabad , Pakistan
| |
Collapse
|