1
|
Chen ZA, Wu CH, Wu SH, Huang CY, Mou CY, Wei KC, Yen Y, Chien IT, Runa S, Chen YP, Chen P. Receptor Ligand-Free Mesoporous Silica Nanoparticles: A Streamlined Strategy for Targeted Drug Delivery across the Blood-Brain Barrier. ACS NANO 2024; 18:12716-12736. [PMID: 38718220 PMCID: PMC11112986 DOI: 10.1021/acsnano.3c08993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024]
Abstract
Mesoporous silica nanoparticles (MSNs) represent a promising avenue for targeted brain tumor therapy. However, the blood-brain barrier (BBB) often presents a formidable obstacle to efficient drug delivery. This study introduces a ligand-free PEGylated MSN variant (RMSN25-PEG-TA) with a 25 nm size and a slight positive charge, which exhibits superior BBB penetration. Utilizing two-photon imaging, RMSN25-PEG-TA particles remained in circulation for over 24 h, indicating significant traversal beyond the cerebrovascular realm. Importantly, DOX@RMSN25-PEG-TA, our MSN loaded with doxorubicin (DOX), harnessed the enhanced permeability and retention (EPR) effect to achieve a 6-fold increase in brain accumulation compared to free DOX. In vivo evaluations confirmed the potent inhibition of orthotopic glioma growth by DOX@RMSN25-PEG-TA, extending survival rates in spontaneous brain tumor models by over 28% and offering an improved biosafety profile. Advanced LC-MS/MS investigations unveiled a distinctive protein corona surrounding RMSN25-PEG-TA, suggesting proteins such as apolipoprotein E and albumin could play pivotal roles in enabling its BBB penetration. Our results underscore the potential of ligand-free MSNs in treating brain tumors, which supports the development of future drug-nanoparticle design paradigms.
Collapse
Affiliation(s)
- Zih-An Chen
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Research
Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Cheng-Hsun Wu
- Nano
Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Si-Han Wu
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International
Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chiung-Yin Huang
- Neuroscience
Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chung-Yuan Mou
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Nano
Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Kuo-Chen Wei
- Neuroscience
Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department
of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- School
of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department
of Neurosurgery, New Taipei Municipal TuCheng
Hospital, New Taipei City 23652, Taiwan
| | - Yun Yen
- Center
for Cancer Translational Research, Tzu Chi
University, Hualien 970374, Taiwan
- Cancer
Center, Taipei Municipal WanFang Hospital, Taipei 116081, Taiwan
| | - I-Ting Chien
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Sabiha Runa
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- SRS Medical Communications,
LLC, Cleveland, Ohio 44124, United States
| | - Yi-Ping Chen
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International
Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Peilin Chen
- Research
Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
2
|
Mironiuk-Puchalska E, Karatsai O, Żuchowska A, Wróblewski W, Borys F, Lehka L, Rędowicz MJ, Koszytkowska-Stawińska M. Development of 5-fluorouracil-dichloroacetate mutual prodrugs as anticancer agents. Bioorg Chem 2023; 140:106784. [PMID: 37639758 DOI: 10.1016/j.bioorg.2023.106784] [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: 06/02/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
5-Fluorouracil (5-FU) is one of the most widely applied chemotherapeutic agents with a broad spectrum of activity. However, despite this versatile activity, its use poses many limitations. Herein, novel derivatives of 5-FU and dichloroacetic acid have been designed and synthesized as a new type of codrugs, also known as mutual prodrugs, to overcome the drawbacks of 5-FU and enhance its therapeutic efficiency. The stability of the obtained compounds has been tested at various pH values using different analytical techniques, namely HPLC and potentiometry. The antiproliferative activity of the new 5-FU derivatives was assessed in vitro on SK-MEL-28 and WM793 human melanoma cell lines in 2D culture as well as on A549 human lung carcinoma, MDA-MB-231 breast adenocarcinoma, LL24 normal lung tissue, and HMF normal breast tissue as a multicellular 3D spheroid model cultured in standard (static) conditions and with the use of microfluidic systems, which to a great extent resembles the in vivo environment. In all cases, new mutual prodrugs showed a higher cytotoxic activity toward cancer models and lower to normal cell models than the parent 5-FU itself.
Collapse
Affiliation(s)
- Ewa Mironiuk-Puchalska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland.
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Agnieszka Żuchowska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Wojciech Wróblewski
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Filip Borys
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | | |
Collapse
|
3
|
Hossain M, Roth S, Dimmock JR, Das U. Cytotoxic derivatives of dichloroacetic acid and some metal complexes. Arch Pharm (Weinheim) 2022; 355:e2200236. [DOI: 10.1002/ardp.202200236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/27/2022] [Accepted: 07/30/2022] [Indexed: 11/12/2022]
Affiliation(s)
| | - Shayne Roth
- School of Sciences Indiana University Kokomo Kokomo Indiana USA
| | - Jonathan R. Dimmock
- Drug Discovery and Development Research Cluster University of Saskatchewan Saskatoon Saskatchewan Canada
| | - Umashankar Das
- Drug Discovery and Development Research Cluster University of Saskatchewan Saskatoon Saskatchewan Canada
| |
Collapse
|
4
|
Chen H, Fang Z, Song M, Liu K. Mitochondrial targeted hierarchical drug delivery system based on HA-modified liposomes for cancer therapy. Eur J Med Chem 2022; 241:114648. [DOI: 10.1016/j.ejmech.2022.114648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022]
|
5
|
Hossain M, Roayapalley PK, Sakagami H, Satoh K, Bandow K, Das U, Dimmock JR. Dichloroacetyl Amides of 3,5-Bis(benzylidene)-4-piperidones Displaying Greater Toxicity to Neoplasms than to Non-Malignant Cells. MEDICINES 2022; 9:medicines9060035. [PMID: 35736248 PMCID: PMC9228592 DOI: 10.3390/medicines9060035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/21/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022]
Abstract
A series of 3,5-bis(benzylidene)-1-dichloroacetyl-4-piperidones 1a–l was evaluated against Ca9-22, HSC-2, HSC-3, and HSC-4 squamous cell carcinomas. Virtually all of the compounds displayed potent cytotoxicity, with 83% of the CC50 values being submicromolar and several CC50 values being in the double digit nanomolar range. The compounds were appreciably less toxic to human HGF, HPLF, and HPC non-malignant cells, which led to some noteworthy selectivity index (SI) figures. From these studies, 1d,g,k emerged as the lead molecules in terms of their potencies and SI values. A Quantitative Structure-Activity Relationship (QSAR) study revealed that cytotoxic potencies and potency–selectivity expression figures increased when the magnitude of the sigma values in the aryl rings was elevated. The modes of action of the representative cytotoxins in Ca9-22 cells were found to include G2/M arrest and stimulation of the cells to undergo mitosis and cause poly(ADP-ribose) polymerase (PARP) and procaspase 3 cleavage.
Collapse
Affiliation(s)
- Mohammad Hossain
- School of Sciences, Indiana University Kokomo, Kokomo, IN 46904, USA;
| | - Praveen K. Roayapalley
- Drug Discovery and Development Research Cluster, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (U.D.); (J.R.D.)
- Correspondence: ; Tel.: +1-306-715-4217
| | - Hiroshi Sakagami
- Meikai University School of Dentistry, Sakado 350-0283, Japan; (H.S.); (K.S.); (K.B.)
| | - Keitaro Satoh
- Meikai University School of Dentistry, Sakado 350-0283, Japan; (H.S.); (K.S.); (K.B.)
| | - Kenjiro Bandow
- Meikai University School of Dentistry, Sakado 350-0283, Japan; (H.S.); (K.S.); (K.B.)
| | - Umashankar Das
- Drug Discovery and Development Research Cluster, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (U.D.); (J.R.D.)
| | - Jonathan R. Dimmock
- Drug Discovery and Development Research Cluster, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (U.D.); (J.R.D.)
| |
Collapse
|
6
|
Thabet NM, Abdel-Rafei MK, El-Sayyad GS, Elkodous MA, Shaaban A, Du YC, Rashed LA, Askar MA. Multifunctional nanocomposites DDMplusAF inhibit the proliferation and enhance the radiotherapy of breast cancer cells via modulating tumor-promoting factors and metabolic reprogramming. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Tumor-promoting factors (TPF) and metabolic reprogramming are hallmarks of cancer cell growth. This study is designed to combine the newly synthesized two nanocomposites DDM (HA-FA-2DG@DCA@MgO) and AF (HA-FA-Amygdaline@Fe2O3) with fractionated doses of radiotherapy (6 Gy-FDR; fractionated dose radiotherapy) to improve the efficiency of chemo-radiotherapy against breast cancer cell lines (BCCs; MCF-7 and MDA-MB-231). The physicochemical properties of each nanocomposite were confirmed using energy dispersive XRD, FTIR, HR-TEM, and SEM. The stability of DDMPlusAF was also examined, as well as its release and selective cellular uptake in response to acidic pH. A multiple-MTT assay was performed to evaluate the radiosensitivity of BCCs to DDMPlusAF at 3 Gy (single dose radiotherapy; SDR) and 6 Gy-FDR after 24, 48, and 72 h. Finally, the anti-cancer activity of DDMPlusAF with 6 Gy-FDR was investigated via assessing the cell cycle distribution and cell apoptosis by flow cytometry, the biochemical mediators (HIF-1α, TNF-α, IL-10, P53, PPAR-α, and PRMT-1), along with glycolytic pathway (glucose, HK, PDH, lactate, and ATP) as well as the signaling effectors (protein expression of AKT, AMPK, SIRT-1, TGF-β, PGC-1α, and gene expression of ERR-α) were determined in this study.
Results
The stability of DDMPlusAF was verified over 6 days without nanoparticle aggregation. DDMPlusAF release and selectivity data revealed that their release was amenable to the acidic pH of the cancer environment, and their selectivity was enhanced towards BCCs owing to CD44 and FR-α receptors-mediated uptake. After 24 h, DDMPlusAF boosted the BCC radiosensitivity to 6 Gy-FDR. Cell cycle arrest (G2/M and pre-G1), apoptosis induction, modulation of TPF mediators and signaling effectors, and suppression of aerobic glycolysis, all confirmed DDMPlusAF + 6 Gy’s anti-cancer activity.
Conclusions
It could be concluded that DDMPlusAF exerted a selective cancer radiosensitizing efficacy with targeted properties for TPF and metabolic reprogramming in BCCs therapy.
Collapse
|
7
|
Dai Z, Wang Q, Tang J, Qu R, Wu M, Li H, Yang Y, Zhen X, Yu C. A Sub-6 nm MnFe2O4-dichloroacetic acid nanocomposite modulates tumor metabolism and catabolism for reversing tumor immunosuppressive microenvironment and boosting immunotherapy. Biomaterials 2022; 284:121533. [DOI: 10.1016/j.biomaterials.2022.121533] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 12/19/2022]
|
8
|
Shang L, Jiang X, Yang T, Xu H, Xie Q, Hu M, Yang C, Kong L, Zhang Z. Enhancing cancer chemo-immunotherapy by biomimetic nanogel with tumor targeting capacity and rapid drug-releasing in tumor microenvironment. Acta Pharm Sin B 2022; 12:2550-2567. [PMID: 35646526 PMCID: PMC9136611 DOI: 10.1016/j.apsb.2021.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/15/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
In the development of chemo-immunotherapy, many efforts have been focusing on designing suitable carriers to realize the co-delivery of chemotherapeutic and immunotherapeutic with different physicochemical properties and mechanisms of action. Besides, rapid drug release at the tumor site with minimal drug degradation is also essential to facilitate the antitumor effect in a short time. Here, we reported a cancer cell membrane-coated pH-responsive nanogel (NG@M) to co-deliver chemotherapeutic paclitaxel (PTX) and immunotherapeutic agent interleukin-2 (IL-2) under mild conditions for combinational treatment of triple-negative breast cancer. In the designed nanogels, the synthetic copolymer PDEA-co-HP-β-cyclodextrin-co-Pluronic F127 and charge reversible polymer dimethylmaleic anhydride-modified polyethyleneimine endowed nanogels with excellent drug-loading capacity and rapid responsive drug-releasing behavior under acidic tumor microenvironment. Benefited from tumor homologous targeting capacity, NG@M exhibited 4.59-fold higher accumulation at the homologous tumor site than heterologous cancer cell membrane-coated NG. Rapidly released PTX and IL-2 enhanced the maturation of dendritic cells and quickly activated the antitumor immune response in situ, followed by prompted infiltration of immune effector cells. By the combined chemo-immunotherapy, enhanced antitumor effect and efficient pulmonary metastasis inhibition were achieved with a prolonged median survival rate (39 days).
Collapse
Affiliation(s)
- Lihuan Shang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ting Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbo Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Xie
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mei Hu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- Corresponding authors. Tel./fax: +86 27 83692762.
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
- Corresponding authors. Tel./fax: +86 27 83692762.
| |
Collapse
|
9
|
Baghbanbashi M, Pazuki G, Khoee S. One Pot Silica Nanoparticle Modification and Doxorubicin Encapsulation as pH-Responsive Nanocarriers, Applying PEG/Lysine Aqueous Two Phase System. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.118472] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
10
|
Khalil M, Haq EA, Dwiranti A, Prasedya ES, Kitamoto Y. Bifunctional folic-conjugated aspartic-modified Fe 3O 4 nanocarriers for efficient targeted anticancer drug delivery. RSC Adv 2022; 12:4961-4971. [PMID: 35425526 PMCID: PMC8981417 DOI: 10.1039/d1ra08776b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/03/2022] [Indexed: 01/01/2023] Open
Abstract
Functionalization of nanocarriers has been considered the most promising way of ensuring an accurate and targeted drug delivery system. This study reports the synthesis of bifunctional folic-conjugated aspartic-modified Fe3O4 nanocarriers with an excellent ability to deliver doxorubicin (DOX), an anticancer drug, into the intercellular matrix. Here, the presence of amine and carboxylate groups enables aspartic acid (AA) to be used as an efficient anchoring molecule for the conjugation of folic acid (FA) (EDC-NHS coupling) and DOX (electrostatic interaction). Based on the results, surface functionalization showed little effect on the physicochemical properties of the nanoparticles but significantly influenced both the loading and release efficiency of DOX. This is primarily caused by the steric hindrance effect due to large and bulky FA molecules. Furthermore, in vitro MTT assay of B16-F1 cell lines revealed that FA conjugation was responsible for a significant increase in the cytotoxicity of DOX-loaded nanocarriers, which was also found to be proportional to AA concentration. This high cytotoxicity resulted from an efficient cellular uptake induced by the over-expressed folate receptors and fast pH triggered DOX release inside the target cell. Here, the lowest IC50 value of DOX-loaded nanocarriers was achieved at 2.814 ± 0.449 μg mL-1. Besides, further investigation also showed that the drug-loaded nanocarriers exhibited less or no toxicity against normal cells.
Collapse
Affiliation(s)
- Munawar Khalil
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia 16424 Depok West Java Indonesia
| | - Ely Arina Haq
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia 16424 Depok West Java Indonesia
| | - Astari Dwiranti
- Department of Biology, Cellular and Molecular Mechanism in Biological System (CEMBIOS) Research Group, Faculty of Mathematics and Natural Sciences, Universitas Indonesia 16424 Depok West Java Indonesia
| | - Eka Sunarwidhi Prasedya
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Mataram 83125 Lombok West Nusa Tenggara Indonesia
- Bioscience and Biotechnology Research Center, Faculty of Mathematics and Natural Sciences, University of Mataram 83125 Lombok West Nusa Tenggara Indonesia
| | - Yoshitaka Kitamoto
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology Yokohama 226-8503 Japan
| |
Collapse
|
11
|
Askar MA, Thabet NM, El-Sayyad GS, El-Batal AI, Abd Elkodous M, El Shawi OE, Helal H, Abdel-Rafei MK. Dual Hyaluronic Acid and Folic Acid Targeting pH-Sensitive Multifunctional 2DG@DCA@MgO-Nano-Core-Shell-Radiosensitizer for Breast Cancer Therapy. Cancers (Basel) 2021; 13:cancers13215571. [PMID: 34771733 PMCID: PMC8583154 DOI: 10.3390/cancers13215571] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary In this study, we have developed CD44 and folate receptor-targeting multi-functional dual drug-loaded nanoparticles. This comprises hyaluronic acid (HA) and folic acid (FA) conjugated to 2-deoxy glucose (2DG) and a shell linked to a dichloroacetate (DCA) and magnesium oxide (MgO) core (2DG@DCA@MgO; DDM) to enhance the localized chemo-radiotherapy for effective breast cancer (BC) treatment. The physicochemical properties of nanoparticles including stability, selectivity, responsive release to pH, cellular uptake, and anticancer efficacy were comprehensively examined. Mechanistically, we identified multiple component signal pathways as important regulators of BC metabolism and mediators for the inhibitory effects exerted by DDM. Nanoparticles exhibited sustained DDM release properties in bio-relevant media, which was responsive to acidic pH providing edibility to the control of drug release from nanoparticles. DDM-loaded and HA–FA-functionalized nanoparticles exhibited increased selectivity and uptake by BC cells. Cell-based assays indicated that the functionalized DDM significantly suppressed cancer cell growth and boosted radiotherapy (RT) efficacy via inducing cell cycle arrest, enhancing apoptosis, and modulating glycolytic and OXPHOS pathways. Accordingly, the inhibition of glycolysis/OXPHOS by DDM and RT treatment may result in cancer metabolic reprogramming via a novel PI3K/AKT/mTOR/P53NF-κB/VEGF pathway in BC cells. Therefore, the dual targeting of glycolysis/OXPHOS pathways is suggested as a promising antitumor strategy. Abstract Globally, breast cancer (BC) poses a serious public health risk. The disease exhibits a complex heterogeneous etiology and is associated with a glycolytic and oxidative phosphorylation (OXPHOS) metabolic reprogramming phenotype, which fuels proliferation and progression. Due to the late manifestation of symptoms, rigorous treatment regimens are required following diagnosis. Existing treatments are limited by a lack of specificity, systemic toxicity, temporary remission, and radio-resistance in BC. In this study, we have developed CD44 and folate receptor-targeting multi-functional dual drug-loaded nanoparticles. This composed of hyaluronic acid (HA) and folic acid (FA) conjugated to a 2-deoxy glucose (2DG) shell linked to a layer of dichloroacetate (DCA) and a magnesium oxide (MgO) core (2DG@DCA@MgO; DDM) to enhance the localized chemo-radiotherapy for effective BC treatment. The physicochemical properties of nanoparticles including stability, selectivity, responsive release to pH, cellular uptake, and anticancer efficacy were thoroughly examined. Mechanistically, we identified multiple component signaling pathways as important regulators of BC metabolism and mediators for the inhibitory effects elicited by DDM. Nanoparticles exhibited sustained DDM release properties in a bio-relevant media, which was responsive to the acidic pH enabling eligibility to the control of drug release from nanoparticles. DDM-loaded and HA–FA-functionalized nanoparticles exhibited increased selectivity and uptake by BC cells. Cell-based assays revealed that the functionalized DDM significantly suppressed cancer cell growth and improved radiotherapy (RT) through inducing cell cycle arrest, enhancing apoptosis, and modulating glycolytic and OXPHOS pathways. By highlighting DDM mechanisms as an antitumor and radio-sensitizing reagent, our data suggest that glycolytic and OXPHOS pathway modulation occurs via the PI3K/AKT/mTOR/NF-κB/VEGFlow and P53high signaling pathway. In conclusion, the multi-functionalized DDM opposed tumor-associated metabolic reprogramming via multiple signaling pathways in BC cells as a promising targeted metabolic approach.
Collapse
Affiliation(s)
- Mostafa A. Askar
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt; (N.M.T.); (M.K.A.-R.)
- Correspondence: (M.A.A.); (G.S.E.-S.); Tel.: +20-010-1704-8253 (M.A.A.)
| | - Noura M. Thabet
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt; (N.M.T.); (M.K.A.-R.)
| | - Gharieb S. El-Sayyad
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt;
- Correspondence: (M.A.A.); (G.S.E.-S.); Tel.: +20-010-1704-8253 (M.A.A.)
| | - Ahmed I. El-Batal
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt;
| | - Mohamed Abd Elkodous
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Toyohashi 441-8580, Japan;
| | - Omama E. El Shawi
- Health and Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt;
| | - Hamed Helal
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo 11651, Egypt;
| | - Mohamed K. Abdel-Rafei
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt; (N.M.T.); (M.K.A.-R.)
| |
Collapse
|
12
|
Sivanesan I, Gopal J, Muthu M, Shin J, Oh JW. Reviewing Chitin/Chitosan Nanofibers and Associated Nanocomposites and Their Attained Medical Milestones. Polymers (Basel) 2021; 13:2330. [PMID: 34301087 PMCID: PMC8309474 DOI: 10.3390/polym13142330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 11/25/2022] Open
Abstract
Chitin/chitosan research is an expanding field with wide scope within polymer research. This topic is highly inviting as chitin/chitosan's are natural biopolymers that can be recovered from food waste and hold high potentials for medical applications. This review gives a brief overview of the chitin/chitosan based nanomaterials, their preparation methods and their biomedical applications. Chitin nanofibers and Chitosan nanofibers have been reviewed, their fabrication methods presented and their biomedical applications summarized. The chitin/chitosan based nanocomposites have also been discussed. Chitin and chitosan nanofibers and their binary and ternary composites are represented by scattered superficial reports. Delving deep into synergistic approaches, bringing up novel chitin/chitosan nanocomposites, could help diligently deliver medical expectations. This review highlights such lacunae and further lapses in chitin related inputs towards medical applications. The grey areas and future outlook for aligning chitin/chitosan nanofiber research are outlined as research directions for the future.
Collapse
Affiliation(s)
- Iyyakkannu Sivanesan
- Department of Bioresources and Food Science, Konkuk University, Seoul 143-701, Korea;
| | - Judy Gopal
- Laboratory of Neo Natural Farming, Chunnampet 603 401, Tamil Nadu, India; (J.G.); (M.M.)
| | - Manikandan Muthu
- Laboratory of Neo Natural Farming, Chunnampet 603 401, Tamil Nadu, India; (J.G.); (M.M.)
| | - Juhyun Shin
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea;
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea;
| |
Collapse
|
13
|
Jiang X, Zheng YW, Bao S, Zhang H, Chen R, Yao Q, Kou L. Drug discovery and formulation development for acute pancreatitis. Drug Deliv 2020; 27:1562-1580. [PMID: 33118404 PMCID: PMC7598990 DOI: 10.1080/10717544.2020.1840665] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acute pancreatitis is a sudden inflammation and only last for a short time, but might lead to a life-threatening emergency. Traditional drug therapy is an essential supportive method for acute pancreatitis treatment, yet, failed to achieve satisfactory therapeutic outcomes. To date, it is still challenging to develop therapeutic medicine to redress the intricate microenvironment promptly in the inflamed pancreas, and more importantly, avoid multi-organ failure. The understanding of the acute pancreatitis, including the causes, mechanism, and severity judgment, could help the scientists bring up more effective intervention and treatment strategies. New formulation approaches have been investigated to precisely deliver therapeutics to inflammatory lesions in the pancreas, and some even could directly attenuate the pancreatic damages. In this review, we will briefly introduce the involved pathogenesis and underlying mechanisms of acute pancreatitis, as well as the traditional Chinese medicine and the new drug option. Most of all, we will summarize the drug delivery strategies to reduce inflammation and potentially prevent the further development of pancreatitis, with an emphasis on the bifunctional nanoparticles that act as both drug delivery carriers and therapeutics.
Collapse
Affiliation(s)
- Xue Jiang
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ya-Wen Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shihui Bao
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hailin Zhang
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Children's Respiration Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Ma Y, Mou Q, Yan D, Zhu X. Engineering small molecule nanodrugs to overcome barriers for cancer therapy. VIEW 2020. [DOI: 10.1002/viw.20200062] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Yuan Ma
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Quanbing Mou
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
15
|
Chu C, Bao Z, Sun M, Wang X, Zhang H, Chen W, Sui Y, Li J, Zhuang Y, Wang D. NIR Stimulus-Responsive PdPt Bimetallic Nanoparticles for Drug Delivery and Chemo-Photothermal Therapy. Pharmaceutics 2020; 12:E675. [PMID: 32709022 PMCID: PMC7408621 DOI: 10.3390/pharmaceutics12070675] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 11/17/2022] Open
Abstract
The combination of chemotherapy and phototherapy has attracted increasing attention for cancer treatment in recent years. In the current study, porous PdPt bimetallic nanoparticles (NPs) were synthesized and used as delivery carriers for the anti-cancer drug doxorubicin (DOX). DOX@PdPt NPs were modified with thiol functionalized hyaluronic acid (HA-SH) to generate DOX@PdPt@HA NPs with an average size of 105.2 ± 6.7 nm. Characterization and in vivo and in vitro assessment of anti-tumor effects of DOX@PdPt@HA NPs were further performed. The prepared DOX@PdPt@HA NPs presented a high photothermal conversion efficiency of 49.1% under the irradiation of a single 808 nm near-infrared (NIR) laser. Moreover, NIR laser irradiation-induced photothermal effect triggered the release of DOX from DOX@PdPt@HA NPs. The combined chemo-photothermal treatment of NIR-irradiated DOX@PdPt@HA NPs exerted a stronger inhibitory effect on cell viability than that of DOX or NIR-irradiated PdPt@HA NPs in mouse mammary carcinoma 4T1 cells in vitro. Further, the in vivo combination therapy, which used NIR-irradiated DOX@PdPt@HA NPs in a mouse tumor model established by subcutaneous inoculation of 4T1 cells, was demonstrated to achieve a remarkable tumor-growth inhibition in comparison with chemotherapy or photothermal therapy alone. Results of immunohistochemical staining for caspase-3 and Ki-67 indicated the increased apoptosis and decreased proliferation of tumor cells contributed to the anti-tumor effect of chemo-photothermal treatment. In addition, DOX@PdPt@HA NPs induced negligible toxicity in vivo. Hence, the developed nanoplatform demonstrates great potential for applications in photothermal therapy, drug delivery and controlled release.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Dongkai Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.C.); (Z.B.); (M.S.); (X.W.); (H.Z.); (W.C.); (Y.S.); (J.L.); (Y.Z.)
| |
Collapse
|
16
|
Geng S, Pan T, Zhou W, Cui H, Wu L, Li Z, Chu PK, Yu XF. Bioactive phospho-therapy with black phosphorus for in vivo tumor suppression. Theranostics 2020; 10:4720-4736. [PMID: 32308745 PMCID: PMC7163432 DOI: 10.7150/thno.43092] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/18/2020] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose: Although inorganic nanomaterials have been widely used in multimodal cancer therapies, the intrinsic contributions of the materials are not well understood and sometimes underestimated. In this work, bioactive phospho-therapy with black phosphorus nanosheets (BPs) for in vivo tumor suppression is studied. Methods: Orthotopic liver tumor and acute myeloid leukemia are chosen as the models for the solid tumor and hematological tumor, respectively. BPs are injected into mice through the tail vein and tumor growth is monitored by IVIS bioluminescence imaging. Tumor tissues and serum samples are collected to determine the suppression effect and biosafety of BPs after treatment. Results: The in vitro studies show that BPs with high intracellular uptake produce apoptosis- and autophagy-mediated programmed cell death of human liver carcinoma cells but do not affect normal cells. BPs passively accumulate in the tumor site at a high concentration and inhibit tumor growth. The tumor weight is much less than that observed from the doxorubicin (DOX)-treated group. The average survival time is extended by at least two months and the survival rate is 100% after 120 days. Western bolt analysis confirms that BPs suppress carcinoma growth via the apoptosis and autophagy pathways. In addition, administration of BPs into mice suffering from leukemia results in tumor suppression and long survival. Conclusions: This study reveals that BPs constitute a type of bioactive anti-cancer agents and provides insights into the application of inorganic nanomaterials to cancer therapy.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Doxorubicin/pharmacology
- Female
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Liver Neoplasms/chemistry
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Nude
- Mice, SCID
- Nanostructures/administration & dosage
- Nanostructures/chemistry
- Phosphorus/administration & dosage
- Phosphorus/pharmacokinetics
- Tissue Distribution
- Topoisomerase II Inhibitors/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Shengyong Geng
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Medical Oncology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen 518055, China
| | - Ting Pan
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenhua Zhou
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Haodong Cui
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lie Wu
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhibin Li
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Paul K. Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Xue-Feng Yu
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| |
Collapse
|
17
|
Zhu C, Zhu Y, Pan H, Chen Z, Zhu Q. Current Progresses of Functional Nanomaterials for Imaging Diagnosis and Treatment of Melanoma. Curr Top Med Chem 2019; 19:2494-2506. [PMID: 31642783 DOI: 10.2174/1568026619666191023130524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Abstract
Melanoma is a malignant skin tumor that results in poor disease prognosis due to unsuccessful
treatment options. During the early stages of tumor progression, surgery is the primary approach
that assures a good outcome. However, in the presence of metastasis, melanoma hasbecome almost
immedicable, since the tumors can not be removed and the disease recurs easily in a short period of
time. However, in recent years, the combination of nanomedicine and chemotherapeutic drugs has offered
promising solutions to the treatment of late-stage melanoma. Extensive studies have demonstrated
that nanomaterials and their advanced applications can improve the efficacy of traditional chemotherapeutic
drugs in order to overcome the disadvantages, such as drug resistance, low drug delivery rate and
reduced targeting to the tumor tissue. In the present review, we summarized the latest progress in imaging
diagnosis and treatment of melanoma using functional nanomaterials, including polymers,
liposomes, metal nanoparticles, magnetic nanoparticles and carbon-based nanoparticles. These
nanoparticles are reported widely in melanoma chemotherapy, gene therapy, immunotherapy, photodynamic
therapy, and hyperthermia.
Collapse
Affiliation(s)
- Congcong Zhu
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Yunjie Zhu
- Cellular Biomedicine Group Inc., Shanghai 201210, China
| | - Huijun Pan
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Zhongjian Chen
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Quangang Zhu
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
18
|
Rizeq BR, Younes NN, Rasool K, Nasrallah GK. Synthesis, Bioapplications, and Toxicity Evaluation of Chitosan-Based Nanoparticles. Int J Mol Sci 2019; 20:E5776. [PMID: 31744157 PMCID: PMC6888098 DOI: 10.3390/ijms20225776] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/23/2019] [Accepted: 10/02/2019] [Indexed: 01/06/2023] Open
Abstract
The development of advanced nanomaterials and technologies is essential in biomedical engineering to improve the quality of life. Chitosan-based nanomaterials are on the forefront and attract wide interest due to their versatile physicochemical characteristics such as biodegradability, biocompatibility, and non-toxicity, which play a promising role in biological applications. Chitosan and its derivatives are employed in several applications including pharmaceuticals and biomedical engineering. This article presents a comprehensive overview of recent advances in chitosan derivatives and nanoparticle synthesis, as well as emerging applications in medicine, tissue engineering, drug delivery, gene therapy, and cancer therapy. In addition to the applications, we critically review the main concerns and mitigation strategies related to chitosan bactericidal properties, toxicity/safety using tissue cultures and animal models, and also their potential environmental impact. At the end of this review, we also provide some of future directions and conclusions that are important for expanding the field of biomedical applications of the chitosan nanoparticles.
Collapse
Affiliation(s)
- Balsam R. Rizeq
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha, Qatar;
- Biomedical Research Center, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Nadin N. Younes
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar;
| | - Kashif Rasool
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University (HBKU), P.O. Box 5825, Doha, Qatar
| | - Gheyath K. Nasrallah
- Biomedical Research Center, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar;
| |
Collapse
|
19
|
Dichloroacetate (DCA) and Cancer: An Overview towards Clinical Applications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8201079. [PMID: 31827705 PMCID: PMC6885244 DOI: 10.1155/2019/8201079] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/12/2019] [Accepted: 10/11/2019] [Indexed: 12/19/2022]
Abstract
An extensive body of literature describes anticancer property of dichloroacetate (DCA), but its effective clinical administration in cancer therapy is still limited to clinical trials. The occurrence of side effects such as neurotoxicity as well as the suspicion of DCA carcinogenicity still restricts the clinical use of DCA. However, in the last years, the number of reports supporting DCA employment against cancer increased also because of the great interest in targeting metabolism of tumour cells. Dissecting DCA mechanism of action helped to understand the bases of its selective efficacy against cancer cells. A successful coadministration of DCA with conventional chemotherapy, radiotherapy, other drugs, or natural compounds has been tested in several cancer models. New drug delivery systems and multiaction compounds containing DCA and other drugs seem to ameliorate bioavailability and appear more efficient thanks to a synergistic action of multiple agents. The spread of reports supporting the efficiency of DCA in cancer therapy has prompted additional studies that let to find other potential molecular targets of DCA. Interestingly, DCA could significantly affect cancer stem cell fraction and contribute to cancer eradication. Collectively, these findings provide a strong rationale towards novel clinical translational studies of DCA in cancer therapy.
Collapse
|
20
|
Tataranni T, Agriesti F, Pacelli C, Ruggieri V, Laurenzana I, Mazzoccoli C, Sala GD, Panebianco C, Pazienza V, Capitanio N, Piccoli C. Dichloroacetate Affects Mitochondrial Function and Stemness-Associated Properties in Pancreatic Cancer Cell Lines. Cells 2019; 8:cells8050478. [PMID: 31109089 PMCID: PMC6562462 DOI: 10.3390/cells8050478] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Targeting metabolism represents a possible successful approach to treat cancer. Dichloroacetate (DCA) is a drug known to divert metabolism from anaerobic glycolysis to mitochondrial oxidative phosphorylation by stimulation of PDH. In this study, we investigated the response of two pancreatic cancer cell lines to DCA, in two-dimensional and three-dimension cell cultures, as well as in a mouse model. PANC-1 and BXPC-3 treated with DCA showed a marked decrease in cell proliferation and migration which did not correlate with enhanced apoptosis indicating a cytostatic rather than a cytotoxic effect. Despite PDH activation, DCA treatment resulted in reduced mitochondrial oxygen consumption without affecting glycolysis. Moreover, DCA caused enhancement of ROS production, mtDNA, and of the mitophagy-marker LC3B-II in both cell lines but reduced mitochondrial fusion markers only in BXPC-3. Notably, DCA downregulated the expression of the cancer stem cells markers CD24/CD44/EPCAM only in PANC-1 but inhibited spheroid formation/viability in both cell lines. In a xenograft pancreatic cancer mouse-model DCA treatment resulted in retarding cancer progression. Collectively, our results clearly indicate that the efficacy of DCA in inhibiting cancer growth mechanistically depends on the cell phenotype and on multiple off-target pathways. In this context, the novelty that DCA might affect the cancer stem cell compartment is therapeutically relevant.
Collapse
Affiliation(s)
- Tiziana Tataranni
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Francesca Agriesti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| | - Vitalba Ruggieri
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Gerardo Della Sala
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Concetta Panebianco
- Division of Gastroenterology, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy.
| | - Valerio Pazienza
- Division of Gastroenterology, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy.
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| | - Claudia Piccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| |
Collapse
|
21
|
Ye J, Zhang R, Chai W, Du X. Low-density lipoprotein decorated silica nanoparticles co-delivering sorafenib and doxorubicin for effective treatment of hepatocellular carcinoma. Drug Deliv 2018; 25:2007-2014. [PMID: 30799656 PMCID: PMC6319454 DOI: 10.1080/10717544.2018.1531953] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023] Open
Abstract
Combinational therapy is usually considered as a preferable approach for effective cancer therapy. Especially, combinational chemotherapies targeting different molecular targets are of particular interest due to its high flexibility as well as efficiency. In our study, the surface of silica nanoparticles (SLN) was modified with low-density lipoprotein (LDL) to construct platform (LDL-SLN) capable of specifically targeting low-density lipoprotein receptors (LDLRs) that overexpressing in hepatocellular carcinoma (HCC). In addition, the versatile drug loading capacity of LDL-SLN was employed to fabricate a preferable drug delivery system to co-deliver sorafenib (Sor) and doxorubicin (Dox) for combinational chemotherapy of HCC. Our results revealed that the LDL-SLN/Sor/Dox nanoparticles with size around 100 nm showed preferable stability in physiological environments. Moreover, the LDL-SLN/Sor/Dox could target LDLR overexpressed HepG2 cells. More importantly, both in vitro and in vivo experiments demonstrated that the LDL-SLN/Sor/Dox exerted elevated antitumor efficacy compared to Sor or Dox alone, which indicated that LDL-SLN/Sor/Dox could be a powerful tool for effective combinational chemotherapy of HCC.
Collapse
Affiliation(s)
- Junfeng Ye
- Department of Hepato-Biliary-Pancreatic Surgery, First Hospital of Jilin University, Changchun, PR China
| | - Ruoyan Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, First Hospital of Jilin University, Changchun, PR China
| | - Wengang Chai
- Department of Hepato-Biliary-Pancreatic Surgery, First Hospital of Jilin University, Changchun, PR China
| | - Xiaohong Du
- Department of Hepato-Biliary-Pancreatic Surgery, First Hospital of Jilin University, Changchun, PR China
| |
Collapse
|
22
|
Jin X, Yang Q, Cai N. Preparation of ginsenoside compound-K mixed micelles with improved retention and antitumor efficacy. Int J Nanomedicine 2018; 13:3827-3838. [PMID: 30013338 PMCID: PMC6039058 DOI: 10.2147/ijn.s167529] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Introduction Ginsenoside compound K (CK) has effects on cell-cycle regulation, tumor growth inhibition, and apoptosis induction. However, it has limited applications in clinical settings because of its low solubility and poor absorption. Methods To overcome these limitations, we aimed to develop a mixed micellar system composed of phosphatidylcholine (PC) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine polyethylene glycol 2000 (DSPE PEG 2000; DP). CK encapsulated in PC/DP mixed micelles had enhanced solubility, permeability, and retention effects. Results Compared to free CK, the CK PC/DP micellar system exhibited improved anticancer effects in vitro, including cell-cycle arrest, apoptosis, and anti-invasion in human lung carcinoma A549 cells. The significant proapoptotic effect was reflected by increased chromosomal condensation, annexin V/propidium iodide staining, and related protein expression. In vitro cellular uptake and optical mouse imaging in vivo suggested that the improved antitumor effect was caused primarily by enhanced uptake and tumor targeting. Furthermore, an in vivo antitumor efficacy study indicated that the CK mixed micelles significantly inhibited tumor growth, thereby decreasing tumor volume at the end of the experiment as compared with that in the control mice. Histological analysis confirmed the antitumor effect with low toxicity. Conclusion The PC/DP micellar system was an effective drug delivery system for CK in tumor therapy.
Collapse
Affiliation(s)
- Xin Jin
- Department of Hospital Pharmacy, Suqian Branch Jiangsu Province Hospital, Suqian 223800, China,
| | - Qing Yang
- Department of Hospital Pharmacy, Suqian Branch Jiangsu Province Hospital, Suqian 223800, China,
| | - Ning Cai
- Department of Hospital Pharmacy, Suqian Branch Jiangsu Province Hospital, Suqian 223800, China,
| |
Collapse
|