1
|
Poursani E, Cirillo G, Curcio M, Vittorio O, De Luca M, Leggio A, Nicoletta FP, Iemma F. Dual-responsive chondroitin sulfate self-assembling nanoparticles for combination therapy in metastatic cancer cells. Int J Pharm X 2024; 7:100235. [PMID: 38486882 PMCID: PMC10937311 DOI: 10.1016/j.ijpx.2024.100235] [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: 05/23/2023] [Revised: 01/19/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
In this study, we developed self-assembling nanoparticles (LCPs) able to trigger the release of Chlorambucil (Chl) and Doxorubicin (DOX) to MDA-MB-231 cells by exploiting the enzyme and redox signals. The DOX loaded LCPs was prepared by the self-assembly of two chondroitin sulphate (CS) derivatives, obtained by the covalent conjugation of Lipoic Acid (LA) and Chlorambucil (Chl) to the CS backbone. After the physic-chemical characterization of the conjugates by FT-IR, 1H NMR, and determination of the critical aggregation concentration, spherical nanoparticles with mean hydrodynamic diameter of 45 nm (P.D.I. 0.24) and Z-potential of - 44 mV were obtained by water addition/solvent evaporation method. In vitro experiments for the release of Chl and DOX were performed in healthy and cancer cells, using a cell culture media to maintain the physiological intracellular conditions (pH 7.4) (and concentration of esterase and GSH. The results allowed the selective release of the payloads to be detected: Chl release of 0 and 41% were obtained after 2 h incubation in normal and in cancer cells respectively, while values of 35 (in healthy cells) and 60% (in cancer cells) were recorded for DOX release after 96 h. Finally, viability studies proved the ability of the newly proposed nanosystem to enhance the cytotoxic activity of the two drugs against cancer cells.
Collapse
Affiliation(s)
- Ensieh Poursani
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Randwick, NSW 2052, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, Rende 87036, Italy
| | - Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, Rende 87036, Italy
| | - Orazio Vittorio
- School of Biomedical Science, University of New South Wales, Randwick, NSW 2052, Australia
| | - Michele De Luca
- Department of Pharmacy Health and Nutritional Science, University of Calabria, Rende 87036, Italy
| | - Antonella Leggio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, Rende 87036, Italy
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria, Rende 87036, Italy
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria, Rende 87036, Italy
| |
Collapse
|
2
|
Wang X, Lu H, Luo F, Wang D, Wang A, Wang X, Feng W, Wang X, Su J, Liu M, Xia G. Lipid-like gemcitabine diester-loaded liposomes for improved chemotherapy of pancreatic cancer. J Control Release 2024; 365:112-131. [PMID: 37981050 DOI: 10.1016/j.jconrel.2023.11.028] [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: 06/24/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023]
Abstract
Gemcitabine (GEM) is a non-selective chemotherapeutic agent used in the treatment of pancreatic cancer. Its antitumor efficacy is limited by a short plasma half-life and severe adverse reactions. To overcome these shortcomings, four novel lipid-like GEM diesters were synthesized and encapsulated into liposomes. Through optimization, dimyristoyl GEM (dmGEM)-loaded liposomes (LipodmGEM) were successfully obtained with an almost complete encapsulation efficiency. Compared to free GEM, LipodmGEM showed enhanced cellular uptake and cell apoptosis, improved inhibition of cell migration on AsPC-1 cells and a greatly extended half-life (7.22 vs. 1.78 h). LipodmGEM succeeded in enriching the drug in the tumor (5.28 vs. 0.03 μmol/g at 8 h), overcoming a major shortcoming of GEM, showed excellent anticancer efficacy in vivo and negligible systemic toxicity, superior to GEM. Attractive as well, suspensions of LipodmGEM remained stable at 2-10 °C away from light for no <2 years. Our results suggest that LipodmGEM might become of high interest for treating pancreatic cancer while the simple strategy we reported might be explored as well for converting other antitumor drugs with high water-solubility and short plasma half-life into attractive nanomedicines.
Collapse
Affiliation(s)
- Xiaowei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Hongwei Lu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Fang Luo
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Dan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Apeng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Xuelei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Wenkai Feng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Xiaobo Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Jiayi Su
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Mingliang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
3
|
Liang W, Fan Y, Liu Y, Fang T, Zhang J, Xu Y, Li J, Wang D. ROS/pH dual-sensitive emodin-chlorambucil co-loaded micelles enhance anti-tumor effect through combining oxidative damage and chemotherapy. Int J Pharm 2023; 647:123537. [PMID: 37866554 DOI: 10.1016/j.ijpharm.2023.123537] [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: 06/27/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
The high level of reactive oxygen species (ROS) at the tumor site has been widely used in the tumor targeted delivery. However, the ROS stimulus-responsive vector itself is also a ROS consumer, and the consumption of endogenous ROS may not be sufficient to maintain sustained drug release. In this study, we designed and synthesized ROS/pH dual-sensitive polymer micelles for the co-delivery of emodin (EMD) and chlorambucil (CLB). The release of quinone methides (QM) can consume glutathione (GSH), on the one hand, it can enhance the chemotoxicity of phenylbutyrate nitrogen mustard, on the other hand, emodin can induce oxidative damage of tumor cells and maintain the sustained targeted release of drugs.
Collapse
Affiliation(s)
- Wendi Liang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Yingzhen Fan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Yinghui Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Ting Fang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Jian Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Yuyi Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Ji Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| | - Dongkai Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
4
|
Tian Y, Huang Y, Zhang X, Tang G, Gao Y, Zhou Z, Li Y, Wang H, Yu X, Li X, Liu Y, Yan G, Wang J, Cao Y. Self-Assembled Nanoparticles of a Prodrug Conjugate Based on Pyrimethanil for Efficient Plant Disease Management. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11901-11910. [PMID: 36111893 DOI: 10.1021/acs.jafc.2c04489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Self-assembled nanotechnology is a promising strategy for improving the effective utilization of pesticides due to its distinct advantages. Herein, an amide-bonded prodrug conjugate based on pyrimethanil (PYR) and butyric acid (BA) was successfully synthesized by the nucleophilic substitution reaction and subsequently self-assembled into spherical nanoparticles (PB NPs) with an average size of 85 nm through the solvent exchange method without using any toxic adjuvant. The results showed that PB NPs based on PYR and BA had a synergistic antimicrobial activity against S. sclerotiorum on plant leaves due to good photostability, low volatilization, good surface activity, and improved retention. Additionally, PB NPs could be used by plant cells as nutrients to promote the growth of plants and thus reduced the toxicity of PYR to plant. Therefore, this prodrug conjugate self-assembly nanotechnology would provide a promising strategy for improving the effective utilization rates of pesticides and reducing their toxicities to plants.
Collapse
Affiliation(s)
- Yuyang Tian
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Yuqi Huang
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Xiaohong Zhang
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Gang Tang
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Yunhao Gao
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Zhiyuan Zhou
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Yan Li
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Huachen Wang
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Xueyang Yu
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Xuan Li
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Yulu Liu
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Guangyao Yan
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Jialu Wang
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| | - Yongsong Cao
- College of Plant Protection, China Agricultural University, Beijing 100093, China
| |
Collapse
|
5
|
Shibata A, Koseki Y, Tanita K, Suzuki R, Dao ATN, Kasai H. Development of camptothecin nano-prodrugs based on trimethyl lock groups toward selective drug release in cancer cells. Tetrahedron Lett 2022. [DOI: 10.1016/j.tetlet.2022.153989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
6
|
Cancer nanomedicine: A step towards improving the drug delivery and enhanced efficacy of chemotherapeutic drugs. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
7
|
Ovejero-Paredes K, Díaz-García D, Mena-Palomo I, Marciello M, Lozano-Chamizo L, Morato YL, Prashar S, Gómez-Ruiz S, Filice M. Synthesis of a theranostic platform based on fibrous silica nanoparticles for the enhanced treatment of triple-negative breast cancer promoted by a combination of chemotherapeutic agents. BIOMATERIALS ADVANCES 2022; 137:212823. [PMID: 35929238 DOI: 10.1016/j.bioadv.2022.212823] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/03/2022] [Accepted: 04/20/2022] [Indexed: 06/15/2023]
Abstract
A new series of theranostic silica materials based on fibrous silica particles acting as nanocarriers of two different cytotoxic agents, namely, chlorambucil and an organotin metallodrug have been prepared and structurally characterized. Besides the combined therapeutic activity, these platforms have been decorated with a targeting molecule (folic acid, to selectively target triple negative breast cancer) and a molecular imaging agent (Alexa Fluor 647, to enable their tracking both in vitro and in vivo). The in vitro behaviour of the multifunctional silica systems showed a synergistic activity of the two chemotherapeutic agents in the form of an enhanced cytotoxicity against MDA-MB-231 cells (triple negative breast cancer) as well as by a higher cell migration inhibition. Subsequently, the in vivo applicability of the siliceous nanotheranostics was successfully assessed by observing with in vivo optical imaging techniques a selective tumour accumulation (targeting ability), a marked inhibition of tumour growth paired to a marked antiangiogenic ability after 13 days of systemic administration, thus, confirming the enhanced theranostic activity. The systemic nanotoxicity was also evaluated by analyzing specific biochemical markers. The results showed a positive effect in form of reduced cytotoxicity when both chemotherapeutics are administered in combination thanks to the fibrous silica nanoparticles. Overall, our results confirm the promising applicability of these novel silica-based nanoplatforms as advanced drug-delivery systems for the synergistic theranosis of triple negative breast cancer.
Collapse
Affiliation(s)
- Karina Ovejero-Paredes
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, E-28040 Madrid, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernandez Almagro 3, E-28029 Madrid, Spain
| | - Diana Díaz-García
- COMET-NANO Group, Department of Biology and Geology, Physics and Inorganic Chemistry, ESCET, Universidad Rey Juan Carlos, Calle Tulipán s/n, E-28933 Móstoles, Madrid, Spain
| | - Irene Mena-Palomo
- COMET-NANO Group, Department of Biology and Geology, Physics and Inorganic Chemistry, ESCET, Universidad Rey Juan Carlos, Calle Tulipán s/n, E-28933 Móstoles, Madrid, Spain
| | - Marzia Marciello
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, E-28040 Madrid, Spain
| | - Laura Lozano-Chamizo
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, E-28040 Madrid, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernandez Almagro 3, E-28029 Madrid, Spain
| | - Yurena Luengo Morato
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, E-28040 Madrid, Spain
| | - Sanjiv Prashar
- COMET-NANO Group, Department of Biology and Geology, Physics and Inorganic Chemistry, ESCET, Universidad Rey Juan Carlos, Calle Tulipán s/n, E-28933 Móstoles, Madrid, Spain
| | - Santiago Gómez-Ruiz
- COMET-NANO Group, Department of Biology and Geology, Physics and Inorganic Chemistry, ESCET, Universidad Rey Juan Carlos, Calle Tulipán s/n, E-28933 Móstoles, Madrid, Spain.
| | - Marco Filice
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, E-28040 Madrid, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernandez Almagro 3, E-28029 Madrid, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
| |
Collapse
|
8
|
Xiang J, Liu X, Yuan G, Zhang R, Zhou Q, Xie T, Shen Y. Nanomedicine from amphiphilizedprodrugs: Concept and clinical translation. Adv Drug Deliv Rev 2021; 179:114027. [PMID: 34732344 DOI: 10.1016/j.addr.2021.114027] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/30/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
Nanomedicines generally consisting of carrier materials with small fractions of active pharmaceutical ingredients (API) have long been used to improve the pharmacokinetics and biodistributions, augment the therapeutic efficacies and mitigate the side effects. Amphiphilizing hydrophobic/hydrophilic drugs to prodrugs capable of self-assembly into well-defined nanostructures has emerged as a facile approach to fabricating nanomedicines because this amphiphilized prodrug (APD) strategy presents many advantages, including minimized use of inert carrier materials, well-characterized prodrug structures, fixed and high drug loading contents, 100% loading efficiency, and burst-free but controlled drug release. This review comprehensively summarizes recent advances in APDs and their nanomedicines, from the rationale and the stimuli-responsive linker chemistry for on-demand drug release to their progress to the clinics, clinical performance of APDs, as well as the challenges and perspective on future development.
Collapse
|
9
|
Preparation, characterization, and cytotoxicity evaluation of self-assembled nanoparticles of diosgenin-cytarabine conjugate. Food Chem Toxicol 2021; 151:112101. [PMID: 33684518 DOI: 10.1016/j.fct.2021.112101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022]
Abstract
Diosgenin (DG) isolated from yam roots revealed various bioactivities and applications as drug carrier. In the present study, a conjugate of DG with cytarabine (Ara-C) was used to prepare the self-assembled nanoparticles (NPs) of DG-Ara-C by a nanoprecipitation method. Dynamic light scattering (DLS) as well as transmission electron microscopy (TEM) were employed to analyze the size and the morphology of NPs, respectively. The stability and absorption of DG-Ara-C NPs were measured. Additionally, the cytotoxicity of the NPs was determined via MTT assay. The results indicated that the average particle size of DG-Ara-C NPs was around 190 nm with a narrow size distribution (PDI = 0.1). TEM showed that DG-Ara-C NPs had a spherical morphology. Compared to free DG or Ara-C, the self-assembled DG-Ara-C NPs exhibited a better anti-tumor activity against solid tumor cells as well as leukemia cells. In conclusion, DG possesses dual role in the self-assembled NPs of DG-Ara-C conjugate, being as a promising anticancer drug and drug carrier.
Collapse
|
10
|
Fan M, Li J. A Novel Combinational Nanodrug Delivery System Induces Synergistic Inhibition of Lung Adenocarcinoma Cells In vitro. LETT DRUG DES DISCOV 2021. [DOI: 10.2174/1570180817999200719152426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
The combination of two or more therapeutic drugs is an attractive approach
to improve the treatment of experimental tumors. Leveraging nanocarriers for combinational drug
delivery can allow control over drug biological fate and promote co-localization in the same area of
the body. However, there are certain concerns regarding the biodegradability and potential longterm
toxicity arising from these synthetic nanoscale carriers.
Objective:
Our aim was to develop a combinational nanodrug delivery system formed by selfassembling
of amphiphilic drug molecules.minimizing potential toxicities associated with using
additional synthetic nanocarriers.
Methods:
A novel prodrug chlorambucil gemcitabine conjugate was synthesized, this prodrug was
used for the encapsulation of an additional hydrophobic anticancer drug paclitaxel, taking the form
of combinational nanodrugs. Particle size and zeta potential were evaluated, cytotoxicity assay and
apoptosis/cell cycle analysis were also performed to validate the anticancer efficacy of the combinational
nanodrugs.
Results:
The combinational nanodrugs were acquired by means of nanoprecipitation. In A549 lung
adenocarcinoma cell line, cellular assays revealed that co-delivery of low dosage paclitaxel with
chlorambucil gemcitabine conjugate can act synergistically to inhibit cell growth and induce accumulation
of cells in the G2/M phase with a concomitant decrease in G0/G1 compartment.
Conclusion:
Chlorambucil gemcitabine conjugate and paclitaxel can co-assemble into composite
nanoparticles by a nanoprecipitation process and the resulting combinational nanodrugs showed a
synergistic anticancer effect. This synthetic nanocarrier-free approach might broaden the nanodrug
concept and have potential in cancer therapy.
Collapse
Affiliation(s)
- Mingliang Fan
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jiping Li
- Department of Otolaryngology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200032, China
| |
Collapse
|
11
|
Upadhyay T, Ansari VA, Ahmad U, Sultana N, Akhtar J. Exploring Nanoemulsion for Liver Cancer Therapy. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394716666200302123336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer is a leading cause of mortality worldwide, accounting for 8.8 million deaths in
2015. Among these, at least 0.78 million people died of liver cancer alone. The recognized risk
factors for liver cancer include chronic hepatitis B virus (HBV) and hepatitis C virus (HCV) infection,
exposure to dietary aflatoxin, fatty liver disease, alcohol-induced cirrhosis, obesity, smoking,
diabetes, and iron overload. The treatment plan for early diagnosed patients includes radiation
therapy, tumour ablation, surgery, immunotherapy, and chemotherapy. Some sort of drug delivery
vehicles has to be used when the treatment plan is targeted chemotherapy. Nanoemulsions are a
class of biphasic liquid dosage form which are mixtures of oil and water stabilized by a surfactant.
They are either transparent or bluish in hue and serve as a wonderful carrier system for chemotherapeutic
drugs. These vehicles have a particle size in the range of 20-200 nm allowing them
to be delivered successfully in the deepest of tissues. Recent publications on nanoemulsions
reveal their acceptance and a popular choice for delivering both synthetic and herbal drugs to the
liver. This work focuses on some anti-cancer agents that utilized the advantages of nanoemulsion
for liver cancer therapy.
Collapse
Affiliation(s)
- Tanmay Upadhyay
- Faculty of Pharmacy, Integral University, Lucknow-226026, India
| | | | - Usama Ahmad
- Faculty of Pharmacy, Integral University, Lucknow-226026, India
| | - Nazneen Sultana
- Faculty of Pharmacy, Integral University, Lucknow-226026, India
| | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow-226026, India
| |
Collapse
|
12
|
Exploring the action of RGDV-gemcitabine on tumor metastasis, tumor growth and possible action pathway. Sci Rep 2020; 10:15729. [PMID: 32978501 PMCID: PMC7519057 DOI: 10.1038/s41598-020-72824-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 09/07/2020] [Indexed: 11/09/2022] Open
Abstract
The coupling of Arg-Gly-Asp-Val (RGDV) and gemcitabine led to a hypothesis that the conjugate (RGDV-gemcitabine) could inhibit tumor metastasis. To confirm this hypothesis the activities of RGDV-gemcitabine inhibiting tumor metastasis in vitro and in vivo were presented for the first time. AFM (atomic force microscopy) imaged that RGDV-gemcitabine was able to adhere onto the surface of serum-starved A549 cells, to block the extending of the pseudopodia. Thereby RGDV-gemcitabine was able to inhibit the invasion, migration and adhesion of serum-starved A549 cells in vitro. On C57BL/6 mouse model RGDV-gemcitabine dose dependently inhibited the metastasis of planted tumor towards the lung and the minimal dose was 0.084 µmol/kg/3 days. The decrease of serum TNF-α (tumor necrosis factor), IL-8 (interleukin-8), MMP-2 (matrix metalloprotein-2) and MMP-9 (matrix metalloprotein-9) of the treated C57BL/6 mice was correlated with the action pathway of RGDV-gemcitabine inhibiting the metastasis of the planted tumor towards lung.
Collapse
|
13
|
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
|
14
|
Wang Z, Chen J, Little N, Lu J. Self-assembling prodrug nanotherapeutics for synergistic tumor targeted drug delivery. Acta Biomater 2020; 111:20-28. [PMID: 32454086 PMCID: PMC7245299 DOI: 10.1016/j.actbio.2020.05.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/27/2020] [Accepted: 05/18/2020] [Indexed: 01/08/2023]
Abstract
Self-assembling prodrugs represents a robust and effective nanotherapeutic approach for delivering poorly soluble anticancer drugs. With numerous intrinsic advantages, self-assembling prodrugs possess the maximum drug loading capacity, controlled drug release kinetics, prolonged blood circulation, and preferential tumor accumulation based on the enhanced permeability and retention (EPR) effect. These prodrug conjugates allow for efficient self-assembly into nanodrugs with the potential of encapsulating other therapeutic agents that have different molecular targets, enabling simultaneous temporal-spatial release of drugs for synergistic antitumor efficacy with reduced systemic side effects. The aim of this review is to summarize the recent progress of self-assembling prodrug cancer nanotherapeutics that are made through conjugating therapeutically active agents to Polyethylene glycol, Vitamin E, or drugs with different physicochemical properties via rational design, for synergistic tumor targeted drug delivery. Statement of Significance All current FDA-approved nanomedicines use inert biomaterials as drug delivery carriers. These biomaterials lack any therapeutic potential, contributing not only to the cost, but may also elicit severe unfavorable adverse effects. Despite the reduction in toxicity associated with the payload, these nanotherapeutics have been met with limited clinical success, likely due to the monotherapy regimen. The self-assembling prodrug (SAP) has been emerging as a powerful platform for enhancing efficacy through co-delivering other therapeutic modalities with distinct molecular targets. Herein, we opportunely present a comprehensive review article summarizing three unique approaches of making SAP for synergistic drug delivery: pegylation, vitamin E-derivatization, and drug-drug conjugation. These SAPs may inevitably pave the way for developing more efficacious, clinically translatable, combination cancer nanotherapies.
Collapse
|
15
|
Yan G, Chen R, Xiong N, Song J, Wang X, Tang R. pH-sensitive small molecule nanodrug self-assembled from amphiphilic vitamin B6-E analogue conjugate for targeted synergistic cancer therapy. Colloids Surf B Biointerfaces 2020; 191:111000. [PMID: 32247946 DOI: 10.1016/j.colsurfb.2020.111000] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023]
Abstract
To promote the targeted cancer therapy, the pH-sensitive small molecule nanodrug self-assembled from amphiphilic vitamin B6-E analogue conjugate was successfully constructed. Herein, water-soluble vitamin B6 with pKa (5.6) was chemically conjugated to lipid-soluble vitamin E succinate (α-TOS), which showed selective cancer cell killing ability and this amphiphilic small molecule vitamin conjugate could self-assemble to be free nanoparticles (NPs) and doxorubicin-loaded NPs (α-TOS-B6-NPs-DOX). The small molecule nanodrugs could perform the following characteristic: (i) stability in the sodium dodecyl sulfonate (SDS) solution and long-term storage stability in PBS via surface negative charge; (ii) tumor accumulation by enhanced penetration and retention (EPR) effect; (iii) improved cellular internalization by means of vitamin B6 transporting membrane carrier (VTC); and (iv) facilitating endosomal escape and rapid drug release for synergistic toxicity to tumor cells via charge reversal and ester hydrolysis at intracellular pH and/or esterase. Moreover, α-TOS-B6-NPs-DOX exhibited long blood circulation stability and significant tumor accumulation and inhibition with the decreased side effects in vivo. Thus, the pH-sensitive small molecule nanodrug self-assembled from amphiphilic vitamin B6-E analogue conjugate could be the potential drug carriers in targeted synergistic cancer therapy.
Collapse
Affiliation(s)
- Guoqing Yan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Ran Chen
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Nanchi Xiong
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Jiayu Song
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China.
| |
Collapse
|
16
|
Liu W, Mao Y, Zhang X, Wang Y, Wu J, Zhao S, Peng S, Zhao M. RGDV-modified gemcitabine: a nano-medicine capable of prolonging half-life, overcoming resistance and eliminating bone marrow toxicity of gemcitabine. Int J Nanomedicine 2019; 14:7263-7279. [PMID: 31686807 PMCID: PMC6737205 DOI: 10.2147/ijn.s212978] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Gemcitabine has been widely used as a chemotherapeutic drug. However, drug resistance, short half-life and side effects seriously decrease its chemotherapeutic efficacy. PURPOSE The object of preparing RGDV-gemcitabine was to prolong the half-life, to overcome drug resistance and to eliminate bone marrow toxicity of gemcitabine. METHODS Arg-Gly-Asp-Val was coupled with gemcitabine, forming 4-(Arg-Gly-Asp-Val-amino)-1-[3,3-difluoro-4-hydroxy-5-(hydroxylmethyl)oxo-lan-2-yl]pyrimidin-2-one (RGDV-gemcitabine) involving 9-step reactions. The advantages of RGDV-gemcitabine to gemcitabine were demonstrated by a series of assays, such as in vitro half-life assay, in vitro drug resistance assay, in vivo anti-tumor assay, in vivo kidney toxicity assay, in vivo liver toxicity assay and in vivo marrow toxicity assay. The nano-features of RGDV-gemcitabine were visualized by TEM, SEM and AFM images. The tumor-targeting action and release of RGDV-gemcitabine were evidenced by FT-MS spectra. RESULTS Half-life and anti-tumor activity of RGDV-gemcitabine were 17-fold longer and 10-fold higher than that of gemcitabine, respectively. RGDV-gemcitabine, but not gemcitabine, showed no kidney toxicity, no liver toxicity, no marrow toxicity and no drug resistance. The advantages attributed to the nanofeatures of RGDV-gemcitabine were targeting tumor tissue and releasing gemcitabine in tumor tissue. CONCLUSION RGDV-gemcitabine successively overcame the defects of gemcitabine and provided a practical strategy of nano-medicine.
Collapse
Affiliation(s)
- Wenchao Liu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yujia Mao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Department of Biomaterials, Beijing Laboratory of Biomedical Materials and Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing100026, People’s Republic of China
| |
Collapse
|
17
|
Dong S, He J, Sun Y, Li D, Li L, Zhang M, Ni P. Efficient Click Synthesis of a Protonized and Reduction-Sensitive Amphiphilic Small-Molecule Prodrug Containing Camptothecin and Gemcitabine for a Drug Self-Delivery System. Mol Pharm 2019; 16:3770-3779. [PMID: 31348660 DOI: 10.1021/acs.molpharmaceut.9b00349] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Drug self-delivery systems consisting of small-molecule active drugs with nanoscale features for intracellular delivery without the need for additional polymeric carriers have drawn much attention recently. In this work, we proposed a highly efficient strategy to fabricate protonized and reduction-responsive self-assembled drug nanoparticles from an amphiphilic small-molecule camptothecin-ss-1,2,3-triazole-gemcitabine conjugate (abbreviated as CPT-ss-triazole-GEM) for combination chemotherapy, which was prepared via a Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) "click" reaction. To obtain this drug-triazole-drug conjugate, we first prepared a CPT derivate containing a propargyl group linked with a disulfide group and a GEM derivate attached to an azide group. Subsequently, the two kinds of modified drugs were connected together through a CuAAC reaction between the alkynyl and azide groups to yield the CPT-ss-triazole-GEM prodrug. The characterizations of chemical structures of these intermediates and the final product were performed by 1H NMR, Fourier transform infrared, and liquid chromatography/mass spectrometry measurements. This amphiphilic small-molecule drug-triazole-drug conjugate displayed a high drug loading content, that is, 36.0% of CPT and 27.2% of GEM. This kind of amphiphilic small-molecule prodrugs could form spherical nanoparticles in an aqueous solution in the absence of any other polymeric carriers, in which the hydrophobic CPT formed the core of the nanoparticles, whereas the hydrophilic GEM and protonated 1,2,3-triazole group yielded the shell. In the tumor microenvironment, the prodrug nanoparticles could release both pristine drugs simultaneously. Under the conditions of pH 7.4, and pH 7.4 and 2 μM glutathione (GSH), the prodrug nanoparticles could maintain stability and only 7% of CPT was leaked. However, in a high-GSH environment (pH 7.4 and 10 mM GSH) with the same incubation time, the disulfide linkage would be dissociated and lead to about 34% of CPT release. The results of the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test demonstrated that these prodrug nanoparticles showed a higher cytotoxicity toward HepG2 cells than free CPT and free GEM on both 48 and 72 h of incubation. Both in vitro cellular uptake and flow cytometry results implied that these prodrug nanoparticles could be internalized by HepG2 cells with efficient drug release inside cells. The pharmacokinetics and tissue distribution of the prodrug showed a moderate half-life in vivo, and the prodrug peak concentration in most of the collected tissues appeared at 0.25 h after administration. In addition, the CPT-ss-triazole-GEM prodrug could not cross the blood-brain barrier. Even more important is the fact that there is no accumulation in tissues and a rapid elimination of this small-molecule prodrug could be achieved. In brief, this protonized and reduction-sensitive prodrug simultaneously binds both antitumor drugs and has good self-delivery behavior through the donor-acceptor interaction of the H-bonding ligand, that is, the 1,2,3-triazole group. It provides a new method for combined drug therapy.
Collapse
Affiliation(s)
- Shuxiang Dong
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis , Soochow University , Suzhou 215123 , P. R. China
| | - Jinlin He
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis , Soochow University , Suzhou 215123 , P. R. China
| | - Yue Sun
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis , Soochow University , Suzhou 215123 , P. R. China
| | - Dian Li
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis , Soochow University , Suzhou 215123 , P. R. China
| | - Lei Li
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis , Soochow University , Suzhou 215123 , P. R. China
| | - Mingzu Zhang
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis , Soochow University , Suzhou 215123 , P. R. China
| | - Peihong Ni
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis , Soochow University , Suzhou 215123 , P. R. China
| |
Collapse
|
18
|
Dorababu A. Recent Advances in Nanoformulated Chemotherapeutic Drug Delivery (2015‐2019). ChemistrySelect 2019. [DOI: 10.1002/slct.201901064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Atukuri Dorababu
- Department of ChemistrySRMPP Govt. First Grade College, Huvinahadagali, Ballari (Dt), Karnataka India – 583219
| |
Collapse
|
19
|
Samie HAA, Saeed M, Faisal SM, Kausar MA, Kamal MA. Recent Findings on Nanotechnology-based Therapeutic Strategies Against Hepatocellular Carcinoma. Curr Drug Metab 2019; 20:283-291. [DOI: 10.2174/1389200220666190308134351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 12/14/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023]
Abstract
Background:
Nanotechnology-based therapies are emerging as a promising new anticancer approach.
Early clinical studies suggest that nanoparticle-based therapeutics can show enhanced efficacy while reducing side
effects minimal, owing to targeted delivery and active intracellular uptake.
Methods:
To overcome the problems of gene and drug delivery, nanotechnology based delivery system gained interest
in the last two decades. Encouraging results from Nano formulation based drug delivery systems revealed that
these emerging restoratives can efficiently lead to more effective, targeted, selective and efficacious delivery of chemotherapeutic
agents to the affected target cells.
Results:
Nanotechnology not only inhibits targeted gene products in patients with cancer, but also taught us valuable
lessons regarding appropriate dosages and route of administrations. Besides, nanotechnology based therapeutics
holds remarkable potential as an effective drug delivery system. We critically highlight the recent findings on
nanotechnology mediated therapeutics strategies to combat hepatocellular carcinoma and discuss how nanotechnology
platform can have enhanced anticancer effects compared with the parent therapeutic agents they contain.
Conclusion:
In this review, we discussed the key challenges, recent findings and future perspective in the development
of effective nanotechnology-based cancer therapeutics. The emphasis here is focused on nanotechnology-based
therapies that are likely to affect clinical investigations and their implications for advancing the treatment of patients
with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hany A. Abdel Samie
- Department of Zoology, Faculty of Science, Menoufia University, Al Minufya, Egypt
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Syed Mohd Faisal
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh-202002, India
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Saudi Arabia
| | - Mohammad A. Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
20
|
Enhanced anti-tumor efficiency of gemcitabine prodrug by FAPα-mediated activation. Int J Pharm 2019; 559:48-57. [DOI: 10.1016/j.ijpharm.2019.01.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/28/2018] [Accepted: 01/10/2019] [Indexed: 12/22/2022]
|
21
|
Wang Q, Zhang P, Li Z, Feng X, Lv C, Zhang H, Xiao H, Ding J, Chen X. Evaluation of Polymer Nanoformulations in Hepatoma Therapy by Established Rodent Models. Theranostics 2019; 9:1426-1452. [PMID: 30867842 PMCID: PMC6401493 DOI: 10.7150/thno.31683] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 01/10/2023] Open
Abstract
Hepatoma is one of the most severe malignancies usually with poor prognosis, and many patients are insensitive to the existing therapeutic agents, including the drugs for chemotherapy and molecular targeted therapy. Currently, researchers are committed to developing the advanced formulations with controlled drug delivery to improve the efficacy of hepatoma therapy. Numerous inoculated, induced, and genetically engineered hepatoma rodent models are now available for formulation screening. However, animal models of hepatoma cannot accurately represent human hepatoma in terms of histological characteristics, metastatic pathways, and post-treatment responses. Therefore, advanced animal hepatoma models with comparable pathogenesis and pathological features are in urgent need in the further studies. Moreover, the development of nanomedicines has renewed hope for chemotherapy and molecular targeted therapy of advanced hepatoma. As one kind of advanced formulations, the polymer-based nanoformulated drugs have many advantages over the traditional ones, such as improved tumor selectivity and treatment efficacy, and reduced systemic side effects. In this article, the construction of rodent hepatoma model and much information about the current development of polymer nanomedicines were reviewed in order to provide a basis for the development of advanced formulations with clinical therapeutic potential for hepatoma.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Chengyue Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Huaiyu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| |
Collapse
|
22
|
Zhang D, Zhang J, Li Q, Tian H, Zhang N, Li Z, Luan Y. pH- and Enzyme-Sensitive IR820-Paclitaxel Conjugate Self-Assembled Nanovehicles for Near-Infrared Fluorescence Imaging-Guided Chemo-Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:30092-30102. [PMID: 30118198 DOI: 10.1021/acsami.8b09098] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The short lifetime and easy quick elimination of the near-infrared (NIR) dye new indocyanine green (IR820) in the body restrict its practical application as a photothermal agent in cancer therapy. Meanwhile, the drawback of poor water solubility of the chemotherapeutic drug paclitaxel (PTX) largely restricts its clinical applications. Herein, we, for the first time, combined IR820 and PTX in an "all-in-one" fluorescence imaging-guided chemo-photothermal therapy (PTT) platform by a rational design of a novel pH- and enzyme-sensitive IR820-PTX conjugate assembly. Specifically, the IR820-PTX conjugate nanoparticles exhibit an extremely high therapeutic agent content (IR820 and PTX, 95.7%). Besides the good stability in bloodstream, the IR820-PTX nanoparticles can target tumors for high accumulation via the enhanced permeation and retention effect. Particularly, our IR820-PTX nanoparticles simultaneously solve the obstacles of PTX poor solubility and the short lifetime of IR820 for cancer therapy. The simultaneous release of the free drug and dye can efficiently kill tumor cells by the combination of PTT and chemotherapy via NIR irradiation. Furthermore, the combined therapy can be imaging-guided by measuring the NIR fluorescence imaging resulting from the IR820 component. Therefore, our rationally designed pH- and enzyme-sensitive IR820-PTX conjugate nanoparticles provide an alternative "all-in-one" option for an efficient combinational dual-therapy and imaging.
Collapse
Affiliation(s)
- Di Zhang
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology (Ministry of Education) , Shandong University , 44 West Wenhua Road , Jinan , Shandong Province 250012 , China
| | - Jing Zhang
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology (Ministry of Education) , Shandong University , 44 West Wenhua Road , Jinan , Shandong Province 250012 , China
| | - Qian Li
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology (Ministry of Education) , Shandong University , 44 West Wenhua Road , Jinan , Shandong Province 250012 , China
| | - Hailong Tian
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology (Ministry of Education) , Shandong University , 44 West Wenhua Road , Jinan , Shandong Province 250012 , China
| | - Na Zhang
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology (Ministry of Education) , Shandong University , 44 West Wenhua Road , Jinan , Shandong Province 250012 , China
| | - Zhonghao Li
- Key Laboratory of Colloid & Interface Chemistry, Ministry of Education , Shandong University , Jinan , Shandong Province 250100 China
| | - Yuxia Luan
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology (Ministry of Education) , Shandong University , 44 West Wenhua Road , Jinan , Shandong Province 250012 , China
| |
Collapse
|
23
|
Ma ZY, Wang DB, Song XQ, Wu YG, Chen Q, Zhao CL, Li JY, Cheng SH, Xu JY. Chlorambucil-conjugated platinum(IV) prodrugs to treat triple-negative breast cancer in vitro and in vivo. Eur J Med Chem 2018; 157:1292-1299. [PMID: 30195239 DOI: 10.1016/j.ejmech.2018.08.065] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/01/2018] [Accepted: 08/24/2018] [Indexed: 10/28/2022]
Abstract
Modification of platinum (II) into lipophilic platinum (IV) compounds by introducing biologically active molecules were widely employed to develop new platinum-based prodrugs in the past decade. In this paper, two chlorambucil platinum (IV) complexes, CLB-Pt and CLB-Pt-CLB, were synthesized and displayed very potent antiproliferative activity against all the tested cancer cell lines, such as A549, HeLa and MCF-7, especially to treat the well-known refractory triple-negative breast cancer. CLB-Pt-CLB significantly improved cell-killing effect in triple-negative subtype MDA-MB-231 cells, and showed much stronger cytotoxicity than either monotherapy or combination of cisplatin and chlorambucil. CLB-Pt-CLB prodrug entered cells in dramatically increased amount compared with cisplatin and enhanced DNA damage, inducing cancer cell apoptosis. It exhibited high anticancer activity and no observable toxicity in BALB/c nude mice bearing MDA-MB-231 tumors. The chlorambucil moiety not only greatly assisted the passive diffusion of CLB-Pt-CLB into cells, but also produced the synergism with cisplatin in targeting DNA.
Collapse
Affiliation(s)
- Zhong-Ying Ma
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Dong-Bo Wang
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xue-Qing Song
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yi-Gang Wu
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Qian Chen
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Chun-Lai Zhao
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Jing-Yi Li
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Shi-Hao Cheng
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Jing-Yuan Xu
- Department of Chemical Biology, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
24
|
Therapeutic journery of nitrogen mustard as alkylating anticancer agents: Historic to future perspectives. Eur J Med Chem 2018; 151:401-433. [DOI: 10.1016/j.ejmech.2018.04.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/30/2018] [Accepted: 04/01/2018] [Indexed: 12/17/2022]
|
25
|
Hu X, Liu R, Zhang D, Zhang J, Li Z, Luan Y. Rational Design of an Amphiphilic Chlorambucil Prodrug Realizing Self-Assembled Micelles for Efficient Anticancer Therapy. ACS Biomater Sci Eng 2018; 4:973-980. [PMID: 33418779 DOI: 10.1021/acsbiomaterials.7b00892] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The application of anticancer drug chlorambucil (CLB) in chemotherapy is severely restricted by its insolubility, lability, and toxic side effects; therefore, it is challenging to realize a highly efficient anticancer therapy of chlorambucil. To solve the above drawbacks encountered by chlorambucil, herein we proposed an amphiphilic chlorambucil prodrug-based self-assembled micelle strategy to realize the highly efficient anticancer therapy of chlorambucil. 1,6-Hexanediamine hydrochloride (HDH) serving as the hydrophilic segment was covalently bound to hydrophobic CLB to prepare an amphiphilic prodrug CLB-HDH which could self-assemble into micelles in aqueous solution. These micelles can passively target tumor tissues via the enhanced permeability and retention (EPR) effect, leading to enhanced cellular internalization. Both the cytotoxicity assay in vitro and anticancer study in vivo confirmed the excellent therapeutic activity of CLB-HDH micelles in comparison with free chlorambucil. Moreover, the hemolysis examination and histological analysis demonstrated the designed CLB-HDH micelles are safe in drug delivery. Therefore, our designed amphiphilic prodrug CLB-HDH micelles bring new opportunity for chlorambucil clinical application to combat cancers.
Collapse
Affiliation(s)
- Xu Hu
- School of Pharmaceutical Science, Shandong University, 44 West Wenhua Road, Jinan, Shandong Province 250012, P. R. China
| | - Ruiling Liu
- School of Pharmaceutical Science, Shandong University, 44 West Wenhua Road, Jinan, Shandong Province 250012, P. R. China
| | - Di Zhang
- School of Pharmaceutical Science, Shandong University, 44 West Wenhua Road, Jinan, Shandong Province 250012, P. R. China
| | - Jing Zhang
- School of Pharmaceutical Science, Shandong University, 44 West Wenhua Road, Jinan, Shandong Province 250012, P. R. China
| | - Zhonghao Li
- Key Laboratory of Colloid & Interface Chemistry, Shandong University, Ministry of Education, Jinan, Shandong Province 250100, P. R. China
| | - Yuxia Luan
- School of Pharmaceutical Science, Shandong University, 44 West Wenhua Road, Jinan, Shandong Province 250012, P. R. China
| |
Collapse
|
26
|
Lin Y, Jiang XF, Duan X, Zeng F, Wu B, Wu S. Therapeutic Nanosystem Consisting of Singlet-Oxygen-Responsive Prodrug and Photosensitizer Excited by Two-Photon Light. ACS Med Chem Lett 2018; 9:23-27. [PMID: 29348806 PMCID: PMC5767894 DOI: 10.1021/acsmedchemlett.7b00394] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/22/2017] [Indexed: 12/23/2022] Open
Abstract
Using light as the sole stimulus and employing the generated singlet oxygen as a therapeutic agent and the trigger to activate chemo-drug release could serve as an elegant way to bring into full play the advantageous features of light and enhance therapeutic efficacy through a combination of chemotherapy and photodynamic therapy. Herein a liposomal drug system has been developed by embedding a fluorescent photosensitizer and a prodrug into phospholipid vesicles. Upon one- or two-photon light irradiation, the photosensitizer generates singlet oxygen, which removes the protecting group of the prodrug and subsequently causes the release of the active drug chlorambucil. With the combined action of O21 and chlorambucil, highly controllable cytotoxicity toward cancer cells was achieved. In addition, the fluorescent photosensitizer gives out fluorescent signal acting as the drug monitoring agent. This strategy may provide an efficient approach for cancer treatment and some useful insights for designing light-stimulated on-demand therapeutic systems.
Collapse
Affiliation(s)
- Yi Lin
- State Key Laboratory of Luminescent Materials
and Devices, College of Materials Science and Engineering, South China University of Technology (SCUT), Guangzhou 510640, China
| | - Xiao-fang Jiang
- State Key Laboratory of Luminescent Materials
and Devices, College of Materials Science and Engineering, South China University of Technology (SCUT), Guangzhou 510640, China
| | - Xiangyan Duan
- State Key Laboratory of Luminescent Materials
and Devices, College of Materials Science and Engineering, South China University of Technology (SCUT), Guangzhou 510640, China
| | - Fang Zeng
- State Key Laboratory of Luminescent Materials
and Devices, College of Materials Science and Engineering, South China University of Technology (SCUT), Guangzhou 510640, China
| | - Bo Wu
- State Key Laboratory of Luminescent Materials
and Devices, College of Materials Science and Engineering, South China University of Technology (SCUT), Guangzhou 510640, China
| | - Shuizhu Wu
- State Key Laboratory of Luminescent Materials
and Devices, College of Materials Science and Engineering, South China University of Technology (SCUT), Guangzhou 510640, China
| |
Collapse
|
27
|
Zhang X, Huang B, Shen Y, Yang C, Huang Z, Huang Y, Xu X, Jiang Y, Sun X, Li X, Yan M, Zhao C. Near infrared light triggered reactive oxygen species responsive nanoparticles for chemo-photodynamic combined therapy. J Mater Chem B 2018; 6:2347-2357. [DOI: 10.1039/c8tb00308d] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanoparticles with ROS-responsive properties could realize spatial and temporal drug release under NIR irradiation and the excess ROS could be used for PDT.
Collapse
|
28
|
Abstract
Covalent modification of therapeutic compounds is a clinically proven strategy to devise prodrugs with enhanced treatment efficacies. This prodrug strategy relies on the modified drugs that possess advantageous pharmacokinetic properties and administration routes over their parent drug. Self-assembling prodrugs represent an emerging class of therapeutic agents capable of spontaneously associating into well-defined supramolecular nanostructures in aqueous solutions. The self-assembly of prodrugs expands the functional space of conventional prodrug design, affording a possible pathway to more effective therapies as the assembled nanostructure possesses distinct physicochemical properties and interaction potentials that can be tailored to specific administration routes and disease treatment. In this review, we will discuss the various types of self-assembling prodrugs in development, providing an overview of the methods used to control their structure and function and, ultimately, our perspective on their current and future potential.
Collapse
Affiliation(s)
- Andrew G Cheetham
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Eastern Road, Zhengzhou 450052, Henan, China
| | | | | | | |
Collapse
|
29
|
Ling L, Yao C, Du Y, Ismail M, He R, Hou Y, Zhang Y, Li X. Assembled liposomes of dual podophyllotoxin phospholipid: preparation, characterization and in vivo anticancer activity. Nanomedicine (Lond) 2017; 12:657-672. [DOI: 10.2217/nnm-2016-0396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: A novel amphiphilic prodrug dual podophyllotoxin (PPT) succinate glycerophosphorylcholine (Di-PPT-GPC) assembled liposomes was developed to improve efficiency of PPT. Materials & methods: Di-PPT-GPC liposomes were prepared by thin film technique and characterized by dynamic light scattering and cryo-electron microscopy. Results: In vitro release studies showed that Di-PPT-GPC liposomes could significantly release PPT in weakly acidic environment but had good stability under biological conditions. Methyl tetrazolium assay data revealed that the liposomes have comparable cytotoxicities to free PPT against MCF-7, HeLa and U87 cells. More importantly, in vivo antitumor evaluation indicated that Di-PPT-GPC liposomes exhibited favorable tumor growth inhibition without side effects. Conclusion: Di-PPT-GPC liposomes might have potential to promote the therapeutic effect of PPT for cancer therapy.
Collapse
Affiliation(s)
- Longbing Ling
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Chen Yao
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yawei Du
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Muhammad Ismail
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ruiyu He
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yongpeng Hou
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ying Zhang
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinsong Li
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| |
Collapse
|
30
|
Drug self-delivery systems for cancer therapy. Biomaterials 2017; 112:234-247. [DOI: 10.1016/j.biomaterials.2016.10.016] [Citation(s) in RCA: 351] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/03/2016] [Accepted: 10/11/2016] [Indexed: 12/26/2022]
|
31
|
Fumagalli G, Marucci C, Christodoulou MS, Stella B, Dosio F, Passarella D. Self-assembly drug conjugates for anticancer treatment. Drug Discov Today 2016; 21:1321-9. [DOI: 10.1016/j.drudis.2016.06.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/10/2016] [Accepted: 06/15/2016] [Indexed: 12/28/2022]
|
32
|
Zhang W, Zhu W, He R, Fang S, Zhang Y, Yao C, Ismail M, Li X. Improvement of Stability and Anticancer Activity of Chlorambucil-Tetrapeptide Conjugate Vesicles. CHINESE J CHEM 2016. [DOI: 10.1002/cjoc.201500908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Nowak-Sliwinska P, Weiss A, Păunescu E, Clavel CM, Griffioen AW, Dyson PJ. Anti-angiogenic properties of chlorambucil derivatives with fluorous and hydrocarbon appendages. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00271d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chlorambucil (CLB) derivatives with long fluorous (referred to as 1 and 2) or hydrocarbon (3) chains have been evaluated in a series of in vitro and in vivo assays.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Angiogenesis Laboratory
- Department of Medical Oncology
- VU University Medical Center
- Amsterdam
- The Netherlands
| | - Andrea Weiss
- School of Pharmaceutical Sciences
- University of Geneva
- CH-1211 Geneva 4
- Switzerland
| | - Emilia Păunescu
- School of Pharmaceutical Sciences
- University of Geneva
- CH-1211 Geneva 4
- Switzerland
| | - Catherine M. Clavel
- School of Pharmaceutical Sciences
- University of Geneva
- CH-1211 Geneva 4
- Switzerland
| | - Arjan W. Griffioen
- Angiogenesis Laboratory
- Department of Medical Oncology
- VU University Medical Center
- Amsterdam
- The Netherlands
| | - Paul J. Dyson
- Institute of Chemical Sciences and Engineering
- Swiss Federal Institute of Technology (EPFL)
- Lausanne
- Switzerland
| |
Collapse
|
34
|
Mondal G, Kumar V, Shukla SK, Singh PK, Mahato RI. EGFR-Targeted Polymeric Mixed Micelles Carrying Gemcitabine for Treating Pancreatic Cancer. Biomacromolecules 2015; 17:301-13. [PMID: 26626700 DOI: 10.1021/acs.biomac.5b01419] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The objective of this study was to design GE11 peptide (YHWYGYTPQNVI) linked micelles of poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate-graft-gemcitabine-graft-dodecanol (PEG-b-PCC-g-GEM-g-DC) for enhanced stability and target specificity of gemcitabine (GEM) to EGFR-positive pancreatic cancer cells. GE11-PEG-PCD/mPEG-b-PCC-g-GEM-g-DC mixed micelles showed EGFR-dependent enhanced cellular uptake, and cytotoxicity as compared to scrambled peptide HW12-PEG-PCD/mPEG-b-PCC-g-GEM-g-DC mixed micelles and unmodified mPEG-b-PCC-g-GEM-g-DC micelles. Importantly, GE11-linked mixed micelles preferentially accumulated in orthotopic pancreatic tumor and tumor vasculature at 24 h post systemic administration. GE11-linked mixed micelles inhibited orthotopic pancreatic tumor growth compared to HW12-linked mixed micelles, unmodified mPEG-b-PCC-g-GEM-g-DC micelles, and free GEM formulations. Tumor growth inhibition was mediated by apoptosis of tumor cells and endothelial cells as determined by immunohistochemical staining. In summary, GE11-linked mixed micelles is a promising approach to treat EGFR overexpressing cancers.
Collapse
Affiliation(s)
- Goutam Mondal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Surendra K Shukla
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center , Omaha, Nebraska United States
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center , Omaha, Nebraska United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| |
Collapse
|