1
|
Jiang Y, Zhou H, Liu J, Ha W, Xia X, Li J, Chao T, Xiong H. Progress and Innovative Combination Therapies in Trop-2-Targeted ADCs. Pharmaceuticals (Basel) 2024; 17:652. [PMID: 38794221 PMCID: PMC11125602 DOI: 10.3390/ph17050652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Precise targeting has become the main direction of anti-cancer drug development. Trophoblast cell surface antigen 2 (Trop-2) is highly expressed in different solid tumors but rarely in normal tissues, rendering it an attractive target. Trop-2-targeted antibody-drug conjugates (ADCs) have displayed promising efficacy in treating diverse solid tumors, especially breast cancer and urothelial carcinoma. However, their clinical application is still limited by insufficient efficacy, excessive toxicity, and the lack of biological markers related to effectiveness. This review summarizes the clinical trials and combination therapy strategies for Trop-2-targeted ADCs, discusses the current challenges, and provides new insights for future advancements.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.J.); (H.Z.); (J.L.); (W.H.); (X.X.); (J.L.)
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.J.); (H.Z.); (J.L.); (W.H.); (X.X.); (J.L.)
| |
Collapse
|
2
|
Kabil MF, Badary OA, Bier F, Mousa SA, El-Sherbiny IM. A comprehensive review on lipid nanocarrier systems for cancer treatment: fabrication, future prospects and clinical trials. J Liposome Res 2024; 34:135-177. [PMID: 37144339 DOI: 10.1080/08982104.2023.2204372] [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: 05/03/2022] [Accepted: 04/02/2023] [Indexed: 05/06/2023]
Abstract
Over the last few decades, cancer has been considered a clinical challenge, being among the leading causes of mortality all over the world. Although many treatment approaches have been developed for cancer, chemotherapy is still the most utilized in the clinical setting. However, the available chemotherapeutics-based treatments have several caveats including their lack of specificity, adverse effects as well as cancer relapse and metastasis which mainly explains the low survival rate of patients. Lipid nanoparticles (LNPs) have been utilized as promising nanocarrier systems for chemotherapeutics to overcome the challenges of the currently applied therapeutic strategies for cancer treatment. Loading chemotherapeutic agent(s) into LNPs improves drug delivery at different aspects including specific targeting of tumours, and enhancing the bioavailability of drugs at the tumour site through selective release of their payload, thus reducing their undesired side effects on healthy cells. This review article delineates an overview of the clinical challenges in many cancer treatments as well as depicts the role of LNPs in achieving optimal therapeutic outcomes. Moreover, the review contains a comprehensive description of the many LNPs categories used as nanocarriers in cancer treatment to date, as well as the potential of LNPs for future applications in other areas of medicine and research.
Collapse
Affiliation(s)
- Mohamed Fawzi Kabil
- Nanomedicine Research Labs, Center for Materials Science (CMS), Zewail City of Science and Technology, Giza, Egypt
| | - Osama A Badary
- Clinical Pharmacy Department, Faculty of Pharmacy, The British University in Egypt, El-Shorouk City, Egypt
| | - Frank Bier
- AG Molekulare Bioanalytik und Bioelektronik, Institut für Biochemie und Biologie, Universität Potsdam Karl-Liebknecht-Straße 24/25, Potsdam (OT Golm), Germany
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science (CMS), Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
3
|
Bareth D, Jain S, Kumawat J, Kishore D, Dwivedi J, Hashmi SZ. Synthetic and pharmacological developments in the hybrid s-triazine moiety: A review. Bioorg Chem 2024; 143:106971. [PMID: 38016395 DOI: 10.1016/j.bioorg.2023.106971] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/03/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023]
Abstract
This article summarizes the most recent advancements in the synthetic and pharmacological approaches along with the structure activity relationship towards the s-triazine and its derivatives. Much attention has been given to s-triazine core due to its facile synthesis, interesting pharmacology, high reactivity, and binding characteristics towards various enzymes. An array of biological applications has been demonstrated by s-triazines including antimalarial, anti-HIV, anti-viral, antimicrobial, anti-tuberculosis to name a few. In the present investigation s-triazine based molecular structures have been assembled in respect to their synthesis and medicinal properties. Further, the competence of s-triazine has been correlated and compared with the other heterocyclic moieties to substantiates-triazine a privileged scaffold. From the literature it is revealed that nucleophilic substitution at 2, 4, and 6 positions is significant for various biological applications. This article would help in assisting the chemists in designing novel molecular entities with high medicinal value.
Collapse
Affiliation(s)
- Diksha Bareth
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Sonika Jain
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Jyoti Kumawat
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Dharma Kishore
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Sonia Zeba Hashmi
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India.
| |
Collapse
|
4
|
Atteia HH. MicroRNAs in Anticancer Drugs Hepatotoxicity: From Pathogenic Mechanism and Early Diagnosis to Therapeutic Targeting by Natural Products. Curr Pharm Biotechnol 2024; 25:1791-1806. [PMID: 38178678 DOI: 10.2174/0113892010282155231222071903] [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: 09/13/2023] [Revised: 11/11/2023] [Accepted: 11/24/2023] [Indexed: 01/06/2024]
Abstract
Patients receiving cancer therapies experience severe adverse effects, including hepatotoxicity, even at therapeutic doses. Consequently, monitoring patients on cancer therapy for hepatic functioning is necessary to avoid permanent liver damage. Several pathways of anticancer drug-induced hepatotoxicity involve microRNAs (miRNAs) via targeting mRNAs. These short and non-coding RNAs undergo rapid modulation in non-targeted organs due to cancer therapy insults. Recently, there has been an interest for miRNAs as useful and promising biomarkers for monitoring toxicity since they have conserved sequences across species and are cellular-specific, stable, released during injury, and simple to analyze. Herein, we tried to review the literature handling miRNAs as mediators and biomarkers of anticancer drug-induced hepatotoxicity. Natural products and phytochemicals are suggested as safe and effective candidates in treating cancer. There is also an attempt to combine anticancer drugs with natural compounds to enhance their efficiencies and reduce systemic toxicities. We also discussed natural products protecting against chemotherapy hepatotoxicity via modulating miRNAs, given that miRNAs have pathogenic and diagnostic roles in chemotherapy-induced hepatotoxicity and that many natural products can potentially regulate their expression. Future studies should integrate these findings into clinical trials by formulating suitable therapeutic dosages of natural products to target miRNAs involved in anticancer drug hepatotoxicity.
Collapse
Affiliation(s)
- Hebatallah Husseini Atteia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkia, 44519, Egypt
| |
Collapse
|
5
|
Jiang X, Lee MJ, Luo T, Tillman L, Lin W. Co-delivery of three synergistic chemotherapeutics in a core-shell nanoscale coordination polymer for the treatment of pancreatic cancer. Biomaterials 2023; 301:122235. [PMID: 37441902 PMCID: PMC10528488 DOI: 10.1016/j.biomaterials.2023.122235] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/08/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023]
Abstract
The combination chemotherapy regimen FOLFIRINOX comprising folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin is the first-line treatment for patients with advanced pancreatic cancer, but its use remains prohibitive for the majority of patients due to severe side effects. Here, we report a core-shell nanoscale coordination polymer (NCP) nanoparticle co-delivering a potent and synergistic combination of oxaliplatin, gemcitabine, and SN38 (OGS), for the treatment of pancreatic cancer in mouse models. OGS contains key synergistic components of FOLFIRINOX in a controllable drug ratio., It exhibited particle stability in blood circulation and enhanced deposition of the drugs in acidic tumor environments. In vitro, OGS showed superior cytotoxicity over free drug combinations and robust cytotoxic synergism among its three components. In vivo, OGS improved drug circulation, increased tumor deposition, and exhibited superior antitumor efficacy over the free drug combination in both subcutaneous and orthotopic pancreatic tumor models. OGS treatment achieved 75-91% tumor growth inhibition and prolonged mouse survival by 1.6- to 2.8-folds while minimizing systemic toxicities such as neutropenia, hepatotoxicity, and renal toxicity. This work uncovers a novel and clinically relevant nanomedicine strategy to co-deliver synergistic combination chemotherapies for difficult-to-treat cancers.
Collapse
Affiliation(s)
- Xiaomin Jiang
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL, 60637, USA
| | - Morten J Lee
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL, 60637, USA
| | - Taokun Luo
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL, 60637, USA
| | - Langston Tillman
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL, 60637, USA; Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758, S Maryland Ave, Chicago, IL, 60637, USA.
| |
Collapse
|
6
|
Zhao W, Zheng XD, Tang PYZ, Li HM, Liu X, Zhong JJ, Tang YJ. Advances of antitumor drug discovery in traditional Chinese medicine and natural active products by using multi-active components combination. Med Res Rev 2023; 43:1778-1808. [PMID: 37183170 DOI: 10.1002/med.21963] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023]
Abstract
The antitumor efficacy of Chinese herbal medicines has been widely recognized. Leading compounds such as sterols, glycosides, flavonoids, alkaloids, terpenoids, phenylpropanoids, and polyketides constitute their complex active components. The antitumor monomers derived from Chinese medicine possess an attractive anticancer activity. However, their use was limited by low bioavailability, significant toxicity, and side effects, hindering their clinical applications. Recently, new chemical entities have been designed and synthesized by combining natural drugs with other small drug molecules or active moieties to improve the antitumor activity and selectivity, and reduce side effects. Such a novel conjugated drug that can interact with several vital biological targets in cells may have a more significant or synergistic anticancer activity than a single-molecule drug. In addition, antitumor conjugates could be obtained by combining pharmacophores containing two or more known drugs or leading compounds. Based on these studies, the new drug research and development could be greatly shortened. This study reviews the research progress of conjugates with antitumor activity based on Chinese herbal medicine. It is expected to serve as a valuable reference to antitumor drug research and clinical application of traditional Chinese medicine.
Collapse
Affiliation(s)
- Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiao-Di Zheng
- Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | | | - Hong-Mei Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xue Liu
- Jinan Intellectual Property Protection Center, Jinan, China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
7
|
Gong J, Shi T, Liu J, Pei Z, Liu J, Ren X, Li F, Qiu F. Dual-drug codelivery nanosystems: An emerging approach for overcoming cancer multidrug resistance. Biomed Pharmacother 2023; 161:114505. [PMID: 36921532 DOI: 10.1016/j.biopha.2023.114505] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Multidrug resistance (MDR) promotes tumor recurrence and metastasis and heavily reduces anticancer efficiency, which has become a primary reason for the failure of clinical chemotherapy. The mechanisms of MDR are so complex that conventional chemotherapy usually fails to achieve an ideal therapeutic effect and even accelerates the occurrence of MDR. In contrast, the combination of chemotherapy with dual-drug has significant advantages in tumor therapy. A novel dual-drug codelivery nanosystem, which combines dual-drug administration with nanotechnology, can overcome the application limitation of free drugs. Both the characteristics of nanoparticles and the synergistic effect of dual drugs contribute to circumventing various drug-resistant mechanisms in tumor cells. Therefore, developing dual-drug codelivery nanosystems with different multidrug-resistant mechanisms has an important reference value for reversing MDR and enhancing the clinical antitumor effect. In this review, the advantages, principles, and common codelivery nanocarriers in the application of dual-drug codelivery systems are summarized. The molecular mechanisms of MDR and the dual-drug codelivery nanosystems designed based on different mechanisms are mainly introduced. Meanwhile, the development prospects and challenges of codelivery nanosystems are also discussed, which provide guidelines to exploit optimized combined chemotherapy strategies in the future.
Collapse
Affiliation(s)
- Jianing Gong
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Taoran Shi
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinfeng Liu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zerong Pei
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
8
|
Cheng TM, Chu HY, Huang HM, Li ZL, Chen CY, Shih YJ, Whang-Peng J, Cheng RH, Mo JK, Lin HY, Wang K. Toxicologic Concerns with Current Medical Nanoparticles. Int J Mol Sci 2022; 23:7597. [PMID: 35886945 PMCID: PMC9322368 DOI: 10.3390/ijms23147597] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
Nanotechnology is one of the scientific advances in technology. Nanoparticles (NPs) are small materials ranging from 1 to 100 nm. When the shape of the supplied nanoparticles changes, the physiological response of the cells can be very different. Several characteristics of NPs such as the composition, surface chemistry, surface charge, and shape are also important parameters affecting the toxicity of nanomaterials. This review covered specific topics that address the effects of NPs on nanomedicine. Furthermore, mechanisms of different types of nanomaterial-induced cytotoxicities were described. The distributions of different NPs in organs and their adverse effects were also emphasized. This review provides insight into the scientific community interested in nano(bio)technology, nanomedicine, and nanotoxicology. The content may also be of interest to a broad range of scientists.
Collapse
Affiliation(s)
- Tsai-Mu Cheng
- Graduate Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (T.-M.C.); (H.-Y.C.)
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Yi Chu
- Graduate Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (T.-M.C.); (H.-Y.C.)
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Zi-Lin Li
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Chiang-Ying Chen
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Ya-Jung Shih
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | | | - R. Holland Cheng
- Department of Molecular & Cellular Biology, University of California, Davis, CA 95616, USA;
| | - Ju-Ku Mo
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Hung-Yun Lin
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Molecular & Cellular Biology, University of California, Davis, CA 95616, USA;
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
| |
Collapse
|
9
|
Behl A, Sarwalia P, Kumar S, Behera C, Mintoo MJ, Datta TK, Gupta PN, Chhillar AK. Codelivery of Gemcitabine and MUC1 Inhibitor Using PEG-PCL Nanoparticles for Breast Cancer Therapy. Mol Pharm 2022; 19:2429-2440. [PMID: 35639628 DOI: 10.1021/acs.molpharmaceut.2c00175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In breast cancer therapy, Gemcitabine (Gem) is an antineoplastic antimetabolite with greater anticancer efficacy and tolerability. However, effectiveness of Gem is limited by its off-target effects. The synergistic potential of MUC1 (mucin 1) inhibitors and Gem-loaded polymeric nanoparticles (NPs) was discussed in this work in order to reduce dose-related toxicities and enhance the therapeutic efficacy. The double emulsion solvent evaporation method was used to prepare poly(ethylene glycol) methyl ether-block-poly-caprolactone (PEG-PCL)-loaded Gem and MUC 1 inhibitor NPs. The average size of Gem and MUC 1 inhibitor-loaded NPs was 128 nm, with a spherical shape. Twin-loaded NPs containing Gem and the MUC1 inhibitor decreased IC50 and behaved synergistically. Furthermore, in vitro mechanistic studies, that is, loss of MMP, clonogenic assay, Annexin V FITC assay, and Western blotting to confirm apoptosis with simultaneous induction of autophagy using acridine orange (AO) staining were performed in this study. Furthermore, the investigated NPs upon combination exhibited greater loss of MMP and decreased clonogenic potential with simultaneous induction of autophagy in MCF-7 cells. Annexin V FITC clearly showed the percentage of apoptosis while Western blotting protein expression analysis revealed an increase in caspase-3 activity with simultaneous decrease in Bcl-2 protein expression, a hallmark of apoptosis. The effectiveness of the Ehrlich ascites solid (EAT) mice treated with Gem-MUC1 inhibitor NPs was higher than that of the animals treated alone. Overall, the combined administration of Gem and MUC1 inhibitor-loaded NPs was found to be more efficacious than Gem and MUC1 inhibitor delivered separately.
Collapse
Affiliation(s)
- Akanksha Behl
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124 001, India
| | - Parul Sarwalia
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Sushil Kumar
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Chittaranjan Behera
- PK-PD Tox and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Mubashir Javed Mintoo
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Tirtha Kumar Datta
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Prem N Gupta
- PK-PD Tox and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Anil K Chhillar
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124 001, India
| |
Collapse
|
10
|
Chemotherapeutic and Safety Profile of a Fraction from Mimosa caesalpiniifolia Stem Bark. JOURNAL OF ONCOLOGY 2021; 2021:9031975. [PMID: 34917149 PMCID: PMC8670915 DOI: 10.1155/2021/9031975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/22/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022]
Abstract
Mimosa caesalpiniifolia (Fabaceae) is used by Brazilian people to treat hypertension, bronchitis, and skin infections. Herein, we evaluated the antiproliferative action of the dichloromethane fraction from M. caesalpiniifolia (DFMC) stem bark on murine tumor cells and the in vivo toxicogenetic profile. Initially, the cytotoxic activity of DFMC on primary cultures of Sarcoma 180 (S180) cells by Alamar Blue, trypan, and cytokinesis block micronucleus (CBMN) assays was assessed after 72 h of exposure, followed by the treatment of S180-bearing Swiss mice for 7 days, physiological investigations, and DNA/chromosomal damage. DFMC and betulinic acid revealed similar in vitro antiproliferative action on S180 cells and induced a reduction in viable cells, induced a reduction in viable cells and caused the emergence of bridges, buds, and morphological features of apoptosis and necrosis. S180-transplanted mice treated with DFMC (50 and 100 mg/kg/day), a betulinic acid-rich dichloromethane, showed for the first time in vivo tumor growth reduction (64.8 and 80.0%) and poorer peri- and intratumor quantities of vessels. Such antiproliferative action was associated with detectible side effects (loss of weight, reduction of spleen, lymphocytopenia, and neutrophilia and increasing of GOT and micronucleus in bone marrow), but preclinical general anticancer properties of the DFMC were not threatened by toxicological effects, and these biomedical discoveries validate the ethnopharmacological reputation of Mimosa species as emerging phytotherapy sources of lead molecules.
Collapse
|
11
|
A review on epidermal growth factor receptor's role in breast and non-small cell lung cancer. Chem Biol Interact 2021; 351:109735. [PMID: 34742684 DOI: 10.1016/j.cbi.2021.109735] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/28/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022]
Abstract
Epithelial growth factor receptor (EGFR) is a cell surface transmembrane receptor that mediates the tyrosine signaling pathway to carry the extracellular messages inside the cell and thereby alter the function of nucleus. This leads to the generation of various protein products to up or downregulate the cellular function. It is encoded by cell erythroblastosis virus oncogene B1, so called C-erb B1/ERBB2/HER-2 gene that acts as a proto-oncogene. It belongs to the HER-2 receptor-family in breast cancer and responds best with anti-Herceptin therapy (anti-tyrosine kinase monoclonal antibody). HER-2 positive breast cancer patient exhibits worse prognosis without Herceptin therapy. Similar incidence and prognosis are reported in other epithelial neoplasms like EGFR + lung non-small cell carcinoma and glioblastoma (grade IV brain glial tumor). Present study highlights the role and connectivity of EGF with various cancers via signaling pathways, cell surface receptors mechanism, macromolecules, mitochondrial genes and neoplasm. Present study describes the EGFR associated gene expression profiling (in breast cancer and NSCLC), relation between mitrochondrial genes and carcinoma, and several in vitro and in vivo models to screen the synergistic effect of various combination treatments. According to this study, although clinical studies including targeted treatments, immunotherapies, radiotherapy, TKi-EGFR combined targeted therapy have been carried out to investigate the synergism of combination therapy; however still there is a gap to apply the scenarios of experimental and clinical studies for further developments. This review will give an idea about the transition from experimental to most advanced clinical studies with different combination drug strategies to treat cancer.
Collapse
|
12
|
Nano-Strategies Targeting the Integrin αvβ3 Network for Cancer Therapy. Cells 2021; 10:cells10071684. [PMID: 34359854 PMCID: PMC8307885 DOI: 10.3390/cells10071684] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Integrin αvβ3, a cell surface receptor, participates in signaling transduction pathways in cancer cell proliferation and metastasis. Several ligands bind to integrin αvβ3 to regulate proliferation and metastasis in cancer cells. Crosstalk between the integrin and other signal transduction pathways also plays an important role in modulating cancer proliferation. Carcinoembryonic antigen cell adhesion molecule 6 (CEACAM6) activates the downstream integrin FAK to stimulate biological activities including cancer proliferation and metastasis. Blockage of signals related to integrin αvβ3 was shown to be a promising target for cancer therapies. 3,3′,5,5′-tetraiodothyroacetic acid (tetrac) completely binds to the integrin with the thyroid hormone to suppress cancer proliferation. The (E)-stilbene analog, resveratrol, also binds to integrin αvβ3 to inhibit cancer growth. Recently, nanotechnologies have been used in the biomedical field for detection and therapeutic purposes. In the current review, we show and evaluate the potentiation of the nanomaterial carrier RGD peptide, derivatives of PLGA-tetrac (NDAT), and nanoresveratrol targeting integrin αvβ3 in cancer therapies.
Collapse
|
13
|
Majeed Ganai A, Khan Pathan T, Hampannavar GA, Pawar C, Obakachi VA, Kushwaha B, Deshwar Kushwaha N, Karpoormath R. Recent Advances on the s‐Triazine Scaffold with Emphasis on Synthesis, Structure‐Activity and Pharmacological Aspects: A Concise Review. ChemistrySelect 2021. [DOI: 10.1002/slct.202004591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Ab Majeed Ganai
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Tabasum Khan Pathan
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Girish A. Hampannavar
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
- Department of Pharmaceutical Chemistry K.L.E.U's College of Pharmacy Vidyanagar, Hubli 580031, Karnataka India
| | - Chandrakant Pawar
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Vincent A. Obakachi
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Babita Kushwaha
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Narva Deshwar Kushwaha
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| |
Collapse
|
14
|
Mohareb RM, Wardakhan WW, Abbas NS. Synthesis of Tetrahydrobenzo[ b]thiophene-3-carbohydrazide Derivatives as Potential Anti-cancer Agents and Pim-1 Kinase Inhibitors. Anticancer Agents Med Chem 2020; 19:1737-1753. [PMID: 30947678 DOI: 10.2174/1871520619666190402153429] [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: 09/15/2018] [Revised: 12/22/2018] [Accepted: 03/15/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Tetrahydrobenzo[b]thiophene derivatives are well known to be biologically active compounds and many of them occupy a wide range of anticancer agent drugs. OBJECTIVE One of the main aim of this work was to synthesize target molecules not only possessing anti-tumor activities but also kinase inhibitors. To achieve this goal, our strategy was to synthesize a series of 4,5,6,7- tetrahydrobenzo[b]thiophene-3-carbohydrazide derivatives using cyclohexan-1,4-dione and cyanoacetylhydrazine to give the 2-amino-6-oxo-4,5,6,7-tetrahydrobenzo[b]thiophene-3-carbohydrazide (3) as the key starting material for many heterocyclization reactions. METHODS Compound 3 was reacted with some aryldiazonium salts and the products were cyclised when reacted with either malononitrile or ethyl cyanoacetate. Thiazole derivatives were also obtained through the reaction of compound 3 with phenylisothiocyanate followed by heterocyclization with α-halocarbonyl derivatives. Pyrazole, triazole and pyran derivatives were also obtained. RESULTS The compounds obtained in this work were evaluated for their in-vitro cytotoxic activity against c-Met kinase, and the six typical cancer cell lines (A549, H460, HT-29, MKN-45, U87MG, and SMMC-7721). The results of anti-proliferative evaluations and c-Met kinase, Pim-1 kinse inhibitions revealed that some compounds showed high activities. CONCLUSION The most promising compounds 5b, 5c, 7c, 7d, 11b, 14a, 16b, 18b, 19, 21a, 23c, 23d and 23i against c-Met kinase were further investigated against the five tyrosin kinases (c-Kit, Flt-3, VEGFR-2, EGFR, and PDGFR). Compounds 5b, 5c, 7d, 7e, 11b, 11c, 16c, 16d, 18c, 19, 23e, 23k and 23m were selected to examine their Pim-1 kinase inhibitions activity where compounds 7d, 7e, 11b, 11c, 16d, 18c and 23e showed high activities. All of the synthesized compounds have no impaired effect toward the VERO normal cell line.
Collapse
Affiliation(s)
- Rafat M Mohareb
- Chemistry Department, Faculty of Science Cairo University, New Cairo, A.R, Egypt
| | - Wagnat W Wardakhan
- National Organization for Drug Control & Research, P.O. Box 29, Cairo, A.R, Egypt
| | - Nermeen S Abbas
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, A.R, Egypt.,Department of Chemistry, Faculty of Science, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
15
|
Qu Y, Tan HY, Chan YT, Jiang H, Wang N, Wang D. The functional role of long noncoding RNA in resistance to anticancer treatment. Ther Adv Med Oncol 2020; 12:1758835920927850. [PMID: 32536982 PMCID: PMC7268113 DOI: 10.1177/1758835920927850] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy is one of the fundamental methods of cancer treatment. However, drug resistance remains the main cause of clinical treatment failure. We comprehensively review the newly identified roles of long noncoding RNAs (lncRNAs) in oncobiology that are associated with drug resistance. The expression of lncRNAs is tissue-specific and often dysregulated in human cancers. Accumulating evidence suggests that lncRNAs are involved in chemoresistance of cancer cells. The main lncRNA-driven mechanisms of chemoresistance include regulation of drug efflux, DNA damage repair, cell cycle, apoptosis, epithelial-mesenchymal transition (EMT), induction of signaling pathways, and angiogenesis. LncRNA-driven mechanisms of resistance to various antineoplastic agents have been studied extensively. There are unique mechanisms of resistance against different types of drugs, and each mechanism may have more than one contributing factor. We summarize the emerging strategies that can be used to overcome the technical challenges in studying and addressing lncRNA-mediated drug resistance.
Collapse
Affiliation(s)
- Yidi Qu
- School of Life Sciences, Jilin University, Changchun, China
| | - Hor-Yue Tan
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., P.R. China
| | - Yau-Tuen Chan
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., P.R. China
| | - Hongbo Jiang
- School of Life Sciences, Jilin University, Changchun, China
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., P.R. China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, 130012, China
| |
Collapse
|
16
|
Syntheses and Structure-Activity Relationships in Growth Inhibition Activity against Human Cancer Cell Lines of 12 Substituted Berberine Derivatives. Molecules 2020; 25:molecules25081871. [PMID: 32325679 PMCID: PMC7221678 DOI: 10.3390/molecules25081871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/02/2020] [Accepted: 04/05/2020] [Indexed: 01/29/2023] Open
Abstract
In this study, quaternary berberine chloride is used as a lead compound to design and synthesize a series of berberine-12-amine derivatives to evaluate the growth inhibition activity against human cancer cell lines. Forty-two compounds of several series were obtained. The quaternary berberine-12-N,N-di-n-alkylamine chlorides showed the targeted activities with the IC50 values of most active compounds being dozens of times those of the positive control. A significant structure–activity relationship (SAR) was observed. The activities of quaternary berberine-12-N,N-di-n-alkylamine chlorides are significantly stronger than those of the reduced counterparts. In the range of about 6-8 carbon atoms, the activities increase with the elongation of n-alkyl carbon chain of 12-N,N-di-n-alkylamino, and when the carbon atom numbers are more than 6-8, the activities decrease with the elongation of n-alkyl carbon chain. The activities of the tertiary amine structure are significantly higher than that of the secondary amine structure.
Collapse
|
17
|
Therapeutic efficacy of nanoparticles and routes of administration. Biomater Res 2019; 23:20. [PMID: 31832232 PMCID: PMC6869321 DOI: 10.1186/s40824-019-0166-x] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022] Open
Abstract
In modern-day medicine, nanotechnology and nanoparticles are some of the indispensable tools in disease monitoring and therapy. The term “nanomaterials” describes materials with nanoscale dimensions (< 100 nm) and are broadly classified into natural and synthetic nanomaterials. However, “engineered” nanomaterials have received significant attention due to their versatility. Although enormous strides have been made in research and development in the field of nanotechnology, it is often confusing for beginners to make an informed choice regarding the nanocarrier system and its potential applications. Hence, in this review, we have endeavored to briefly explain the most commonly used nanomaterials, their core properties and how surface functionalization would facilitate competent delivery of drugs or therapeutic molecules. Similarly, the suitability of carbon-based nanomaterials like CNT and QD has been discussed for targeted drug delivery and siRNA therapy. One of the biggest challenges in the formulation of drug delivery systems is fulfilling targeted/specific drug delivery, controlling drug release and preventing opsonization. Thus, a different mechanism of drug targeting, the role of suitable drug-laden nanocarrier fabrication and methods to augment drug solubility and bioavailability are discussed. Additionally, different routes of nanocarrier administration are discussed to provide greater understanding of the biological and other barriers and their impact on drug transport. The overall aim of this article is to facilitate straightforward perception of nanocarrier design, routes of various nanoparticle administration and the challenges associated with each drug delivery method.
Collapse
|
18
|
Turnbull A, Bermejo-Rodríguez C, Preston MA, Garrido-Barros M, Pimentel B, de la Cueva-Méndez G. Targeted Cancer Cell Killing by Highly Selective miRNA-Triggered Activation of a Prokaryotic Toxin-Antitoxin System. ACS Synth Biol 2019; 8:1730-1736. [PMID: 31348648 DOI: 10.1021/acssynbio.9b00172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Although not evolved to function in eukaryotes, prokaryotic toxin Kid induces apoptosis in human cells, and this is avoided by coexpression of its neutralizing antitoxin, Kis. Inspired by the way Kid becomes active in bacterial cells we had previously engineered a synthetic toxin-antitoxin system bearing a Kis protein variant that is selectively degraded in cells expressing viral oncoprotein E6, thus achieving highly selective killing of cancer cells transformed by human papillomavirus. Here we aimed to broaden the type of oncogenic insults, and therefore of cancer cells, that can be targeted using this approach. We show that appropriate linkage of the kis gene to a single, fully complementary, target site for an oncogenic human microRNA enables the construction of a synthetic toxin-antitoxin pair that selectively kills cancer cells overexpressing that particular microRNA. Importantly, the resulting system spares nontargeted cells from collateral damage, even when they overexpress highly homologous, though nontargeted, microRNAs.
Collapse
Affiliation(s)
- Alice Turnbull
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
| | | | - Mark A. Preston
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
| | - María Garrido-Barros
- Synthetic Biology and Smart Therapeutic Nanosystems Laboratory, Andalusian Centre for Nanomedicine and Biotechnology−BIONAND, 29590 Málaga, Spain
- Nanobioengineering of Smart Therapeutic and Diagnostic Systems Laboratory, Area of Oncology and Oncohematology, Instituto de Investigación Biomédica de Málaga−IBIMA, 29010 Málaga, Spain
| | - Belén Pimentel
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
- Synthetic Biology and Smart Therapeutic Nanosystems Laboratory, Andalusian Centre for Nanomedicine and Biotechnology−BIONAND, 29590 Málaga, Spain
- Nanobioengineering of Smart Therapeutic and Diagnostic Systems Laboratory, Area of Oncology and Oncohematology, Instituto de Investigación Biomédica de Málaga−IBIMA, 29010 Málaga, Spain
| | - Guillermo de la Cueva-Méndez
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
- Synthetic Biology and Smart Therapeutic Nanosystems Laboratory, Andalusian Centre for Nanomedicine and Biotechnology−BIONAND, 29590 Málaga, Spain
- Nanobioengineering of Smart Therapeutic and Diagnostic Systems Laboratory, Area of Oncology and Oncohematology, Instituto de Investigación Biomédica de Málaga−IBIMA, 29010 Málaga, Spain
| |
Collapse
|
19
|
Abdo NYM, Mohareb RM, Al-Darkazali WN. Heterocyclization of 2-Arylidenecyclohexan-1,3-dione: Synthesis of Thiophene, Thiazole, and Isoxazole Derivatives with Potential Antitumor Activities. Anticancer Agents Med Chem 2019; 20:335-345. [PMID: 31362693 DOI: 10.2174/1871520619666190730103425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/06/2019] [Accepted: 07/01/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Thiophene, thiazole, and isoxazole derivatives are present in a wide range of natural and synthetic compounds with heterogeneous pharmacological activity. Due to their structural diversity, they are some of the most versatile classes of compounds for anticancer drug design and discovery. OBJECTIVE Thiophene, thiazole, and isoxazole derivatives were herein designed with a dual purpose: as antiproliferative agents and kinase inhibitors. METHODS The test compounds were synthesized in moderate to high yields through a simple methodology. Tetrahydrobenzo[b]thiophen-5-one derivatives 5a-f were prepared from the reaction of 2-arylidencyclohexan- 1,3-dione 3a-c with elemental sulfur and either of malononitrile (4a) or ethyl cyanoacetate (4b) in 1,4-dioxan in the presence of triethylamine. Compounds 5a,b were used for the synthesis of thiophene, thiazole, and isoxazole derivatives through their reactions with different chemical reagents. RESULTS Antiproliferative evaluations, c-Met kinase, and Pim-1 kinase inhibitions were performed where some compounds revealed high activities. In all cases, antiproliferative activity and the kinase inhibitions were performed against six cancer cell lines and five tyrosine kinases, respectively. Where the most cytotoxic compounds were 3c, 5d, and 16c with IC50's 0.29, 0.68, and 0.42μM, respectively, against the A549 cell line. CONCLUSION The anti-proliferative activities of the newly synthesized compounds were evaluated against the six cancer cell lines (A549, HT-29, MKN-45, U87MG, SMMC-7721, and H460). The most potent compounds toward the cancer cell lines (3a, 3c, 5d, 7c, 11c, 16a, and 16c) were further investigated towards the five tyrosine kinases (c-kit, FIT-3, VEGFR-2, EGFR, and PDGFR). Compounds 3c, 5d, and 16c were selected for testing of their inhibition for the Pim-1 kinase due to their anti-proliferation activities against the cancer cell lines and their high activities against the tyrosine kinases.
Collapse
Affiliation(s)
- Nadia Y Megally Abdo
- Department of Chemistry, Faculty of Education, Alexandria University, Alexandria 21526, Egypt
| | - Rafat M Mohareb
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | | |
Collapse
|
20
|
Abdallah HMI, Abdel-Rahman RF, El Awdan SA, Allam RM, El-Mosallamy AEMK, Selim MS, Mohamed SS, Arbid MS, Farrag ARH. Protective effect of some natural products against chemotherapy-induced toxicity in rats. Heliyon 2019; 5:e01590. [PMID: 31080906 PMCID: PMC6507045 DOI: 10.1016/j.heliyon.2019.e01590] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 03/01/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
Aim There is a great interest in combining anticancer drugs with natural products aiming at maximizing their efficacy while minimizing systemic toxicity. Hence, the present study was constructed aiming to investigate the protective potential of three natural products, 1,8-cineole an essential oil from Artemisia herba alba, exopolysaccharide (EPS) from locally identified marine streptomycete, and ellagic acid (EA), against chemotherapy-induced organ toxicity. Methods Isolation, production and characterization of EPS from marine streptomycete was done. Animals were allocated into five groups, GP1: normal control, GP2: cyclophosphamide (CYC), GP3: 1,8-cineole + CYC, GP4: EPS + CYC, GP4: EA + CYC. All drugs were administered orally 1 week before and concomitantly with CYC. Electrocardiography (ECG) analysis, liver enzymes (ALT and AST), cardiac serum markers (LDH and CK), oxidative stress biomarkers in hepatic and cardiac tissues (GSH and MDA), TGF-β1 and histopathological examination of hepatic and cardiac tissues were executed. Results The isolated stain produced EPS was identified as Streptomyces xiamenensis. EPS contains uronic, sulphate groups and different monosugars with Mw 4.65 × 104 g/mol and showed antioxidant activity against DPPH. Pretreatment of rats with 1,8-cineole, EPS and EA improved ECG abnormalities, decrease serum markers of hepato- and cardiotoxicity, prevent oxidative stress and decrease TGF-β1 in liver and heart tissues. Conclusion The present results demonstrate the hepatoprotective and cardioprotective effects of the above-mentioned natural products against CYC organ toxicity.
Collapse
Affiliation(s)
- Heba M I Abdallah
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Rehab F Abdel-Rahman
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Sally A El Awdan
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Rasha M Allam
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | | | - Manal S Selim
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Division, National Research Centre, Giza, Egypt
| | - Sahar S Mohamed
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Division, National Research Centre, Giza, Egypt
| | - Mahmoud S Arbid
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Abdel Razik H Farrag
- Department of Pathology, Medical Division, National Research Centre, Giza, Egypt
| |
Collapse
|
21
|
Solomon VR, Pundir S, Lee H. Examination of novel 4-aminoquinoline derivatives designed and synthesized by a hybrid pharmacophore approach to enhance their anticancer activities. Sci Rep 2019; 9:6315. [PMID: 31004122 PMCID: PMC6474902 DOI: 10.1038/s41598-019-42816-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 04/09/2019] [Indexed: 12/13/2022] Open
Abstract
In an attempt to develop effective and potentially safe anticancer agents, thirty-six 4-aminoquinoline derived sulfonyl analogs were designed and synthesized using a hybrid pharmacophore approach. The cytotoxicity of these compounds was determined using three breast tumor cell lines (MDA-MB231, MDA-MB468 and MCF7) and two matching non-cancer breast epithelial cell lines (184B5 and MCF10A). Although most of the compounds were quite effective on the breast cancer cells, the compound 7-chloro-4-(4-(2,4-dinitrophenylsulfonyl)piperazin-1-yl)quinoline (13; VR23) emerged as potentially the most desirable one in this series of compounds. Data from the NCI-60 cancer panel screening show that compound 13 is effective on a wide range of different cancers. Importantly, compound 13 is needed up to 17.6-fold less doses to achieve the same IC50 against cancer than non-cancer cells (MDA-MB468 vs MCF10A), suggesting that it can potentially be less toxic to normal cells. Cancer cells formed multiple centrosomes in the presence of compound 13, resulting in the cell cycle arrest at prometa-meta phase. This abnormality leads to eventual cell demise with sub-G1 DNA content typically shown with apoptotic cells. In addition, compound 13 also causes an increase in lysosomal volume in cancer but not in non-cancer cells, which may contribute at least in part to its preferential cancer cell-killing. The cancer cell-killing effect of compound 13 is highly potentiated when combined with either bortezomib or monastrol.
Collapse
Affiliation(s)
- V Raja Solomon
- Health Sciences North Research Institute, 56 Walfor Road, Sudbury, Ontario, P3E 2H3, Canada.,Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, Ontario, P3E 2C6, Canada
| | - Sheetal Pundir
- Health Sciences North Research Institute, 56 Walfor Road, Sudbury, Ontario, P3E 2H3, Canada.,Departments of Medicine, the Faculty of Medicine, the University of Ottawa, Ottawa, Ontario, K1H 5M8, Canada
| | - Hoyun Lee
- Health Sciences North Research Institute, 56 Walfor Road, Sudbury, Ontario, P3E 2H3, Canada. .,Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, Ontario, P3E 2C6, Canada. .,Departments of Medicine, the Faculty of Medicine, the University of Ottawa, Ottawa, Ontario, K1H 5M8, Canada.
| |
Collapse
|
22
|
Chang SM, Jain V, Chen TL, Patel AS, Pidugu HB, Lin YW, Wu MH, Huang JR, Wu HC, Shah A, Su TL, Lee TC. Design and Synthesis of 1,2-Bis(hydroxymethyl)pyrrolo[2,1-a]phthalazine Hybrids as Potent Anticancer Agents that Inhibit Angiogenesis and Induce DNA Interstrand Cross-links. J Med Chem 2019; 62:2404-2418. [DOI: 10.1021/acs.jmedchem.8b01689] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Anamik Shah
- Center of Excellence in Drug Discovery, Saurashtra University, Rajkot 360005, India
| | | | | |
Collapse
|
23
|
Fang L, Qin X, Zhao J, Gou S. Construction of Dual Stimuli-Responsive Platinum(IV) Hybrids with NQO1 Targeting Ability and Overcoming Cisplatin Resistance. Inorg Chem 2019; 58:2191-2200. [PMID: 30657321 DOI: 10.1021/acs.inorgchem.8b03386] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Quinone oxidoreductase isozyme I (NQO1) is a cytoprotective two-electron-specific reductase that highly expresses in various cancer cells. Taking NQO1 as the target, we herein report three hybrid compounds from Pt(IV) complexes and a quinone propionic acid unit. The mechanism studies showed that the hybrids could be activated by both NQO1 and ascorbic acid to release the cytotoxic Pt(II) unit, exhibiting a dual stimuli-responsive character. In the pharmacological studies, complexes 2 and 3 presented higher antitumor activity than cisplatin. More importantly, the hybrid could also overcome cisplatin resistance due to the NQO1 targeting ability, improved cellular uptake, and/or different action mechanism. Significantly, complex 3 containing a coumarin moiety could be effectively activated in NQO1-overexpressed cancer cells to "turn on" fluorescence, showing a promising visual effect in cancer cells. In vivo study revealed that both 2 and 3 exhibited higher antitumor efficacy than cisplatin in the A549 xenograft mouse model at an equimolar dose to cisplatin. In all, the hybrids may serve as promising NQO1-targeting anticancer agents.
Collapse
Affiliation(s)
- Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, School of Chemistry and Chemical Engineering , Southeast University , Nanjing 211189 , China.,State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources , Guangxi Normal University , Guilin 541004 , China
| | - Xiaodong Qin
- Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, School of Chemistry and Chemical Engineering , Southeast University , Nanjing 211189 , China
| | - Jian Zhao
- Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, School of Chemistry and Chemical Engineering , Southeast University , Nanjing 211189 , China
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, School of Chemistry and Chemical Engineering , Southeast University , Nanjing 211189 , China
| |
Collapse
|
24
|
Lei M, Sha S, Wang X, Wang J, Du X, Miao H, Zhou H, Bai E, Shi J, Zhu Y. Co-delivery of paclitaxel and gemcitabine via a self-assembling nanoparticle for targeted treatment of breast cancer. RSC Adv 2019; 9:5512-5520. [PMID: 35515924 PMCID: PMC9060788 DOI: 10.1039/c9ra00276f] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/07/2019] [Indexed: 11/27/2022] Open
Abstract
Multi-functional nanoparticles can be used to improve the treatment index and reduce side effects of anti-tumor drugs. Herein, we developed a kind of multi-functional and highly biocompatible nanoparticle (NP) loaded with folic acid (FA), paclitaxel (PTX) and gemcitabine (GEM) via self-assembly to target cancer cells. The transmission electron microscopy (TEM) results showed that multi-functional FA targeting nanoparticles (MF-FA NPs) exhibited spherical morphology and favorable structural stability in aqueous solution. In addition, NPs (MF-FA NPs and MF NPs) exhibited comparable proliferation inhibition to breast cancer cell 4T1 compared with the pure drug. In in vivo antitumor studies, NPs showed an obviously enhanced anti-tumor efficacy compared with the pure drug. Furthermore, MF-FA NPs displayed higher tumor growth inhibition than MF NPs due to the specific targeting of FA to cancer cells. Consequently, the novel MF-FA NPs could be used as a potential chemotherapeutic formulation for breast cancer therapy. Preparation of MF-FA nanoparticles and the release behavior of drugs in tumor cells.![]()
Collapse
Affiliation(s)
- Meng Lei
- College of Science
- Nanjing Forestry University
- Nanjing 210037
- PR China
| | - Sijia Sha
- College of Science
- Nanjing Forestry University
- Nanjing 210037
- PR China
| | - Xueyuan Wang
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| | - Jia Wang
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd
- Nanjing 210046
- PR China
| | - Xiao Du
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing 210009
- PR China
| | - Hang Miao
- College of Science
- Nanjing Forestry University
- Nanjing 210037
- PR China
| | - Hui Zhou
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| | - Enhe Bai
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| | - Jingmiao Shi
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd
- Nanjing 210046
- PR China
| | - Yongqiang Zhu
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| |
Collapse
|
25
|
Ferreira EB, da Silva Júnior WF, de Oliveira Pinheiro JG, da Fonseca AG, Moura Lemos TMA, de Oliveira Rocha HA, de Azevedo EP, Mendonça Junior FJB, Neves de Lima ÁA. Characterization and Antiproliferative Activity of a Novel 2-Aminothiophene Derivative-β-Cyclodextrin Binary System. Molecules 2018; 23:E3130. [PMID: 30501061 PMCID: PMC6320897 DOI: 10.3390/molecules23123130] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/08/2018] [Accepted: 11/10/2018] [Indexed: 12/11/2022] Open
Abstract
The novel 2-aminothiophene derivative 2-amino-4,5,6,7-tetrahydrobenzo[b]thiophene-3-carbonitrile (6CN) has shown potential anti-proliferative activity in human cancer cell lines. However, the poor aqueous solubility of 6CN impairs its clinical use. This work aimed to develop binary 6CN-β-cyclodextrin (βCD) systems with the purpose of increasing 6CN solubility in water and therefore, to improve its pharmacological activity. The 6CN-βCD binary systems were prepared by physical mixing, kneading and rotary evaporation methods and further characterized by FTIR, XRD, DSC, TG and SEM. In addition, molecular modeling and phase solubility studies were performed. Finally, MTT assays were performed to investigate the cytostatic and anti-proliferative effects of 6CN-βCD binary systems. The characterization results show evident changes in the physicochemical properties of 6CN after the formation of the binary systems with βCD. In addition, 6CN was associated with βCD in aqueous solution and the solid state, which was confirmed by molecular modeling and the aforementioned characterization techniques. Phase solubility studies indicated that βCD forms stable 1:1 complexes with 6CN. The MTT assay demonstrated the cytostatic and anti-proliferative activities of 6CN-βCD binary systems and therefore, these might be considered as promising candidates for new anticancer drugs.
Collapse
Affiliation(s)
- Elayne Barros Ferreira
- Department of Pharmacy, Federal University of Rio Grande do Norte, Av. General Cordeiro de Farias, s/n, Petrópolis, Natal, Rio Grande do Norte 59012-570, Brazil.
| | - Walter Ferreira da Silva Júnior
- Department of Pharmacy, Federal University of Rio Grande do Norte, Av. General Cordeiro de Farias, s/n, Petrópolis, Natal, Rio Grande do Norte 59012-570, Brazil.
| | - Jonas Gabriel de Oliveira Pinheiro
- Department of Pharmacy, Federal University of Rio Grande do Norte, Av. General Cordeiro de Farias, s/n, Petrópolis, Natal, Rio Grande do Norte 59012-570, Brazil.
| | - Aldilane Gonçalves da Fonseca
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Av. General Cordeiro de Faria, s/n, Petrópolis, Natal, Rio Grande do Norte 59012-570, Brazil.
| | - Telma Maria Araújo Moura Lemos
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Av. General Cordeiro de Faria, s/n, Petrópolis, Natal, Rio Grande do Norte 59012-570, Brazil.
| | - Hugo Alexandre de Oliveira Rocha
- Department of Biochemistry, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Lagoa Nova, Natal, Rio Grande do Norte 59078-970, Brazil.
| | - Eduardo Pereira de Azevedo
- Graduate Program of Biotechnology, Laureate International Universities-Universidade Potiguar (UnP), Av. Sen. Salgado Filho, 1610, Lagoa Nova, Natal, Rio Grande do Norte 59056-000, Brazil.
| | | | - Ádley Antonini Neves de Lima
- Department of Pharmacy, Federal University of Rio Grande do Norte, Av. General Cordeiro de Farias, s/n, Petrópolis, Natal, Rio Grande do Norte 59012-570, Brazil.
| |
Collapse
|
26
|
C-phycocyanin to overcome the multidrug resistance phenotype in human erythroleukemias with or without interaction with ABC transporters. Biomed Pharmacother 2018; 106:532-542. [DOI: 10.1016/j.biopha.2018.06.145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/26/2018] [Indexed: 01/12/2023] Open
|
27
|
Yang Z, Zhang SL, Hu X, Tam KY. Inhibition of pyruvate dehydrogenase kinase 1 enhances the anti-cancer effect of EGFR tyrosine kinase inhibitors in non-small cell lung cancer. Eur J Pharmacol 2018; 838:41-52. [PMID: 30213498 DOI: 10.1016/j.ejphar.2018.09.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022]
Abstract
Although epidermal growth factor receptor (EGFR) inhibitors have been used to treat non-small cell lung cancer (NSCLC) for decades with great success in patients with EGFR mutations, acquired-resistance inevitably occurs after long-term exposure to the treatment of EGFR inhibitors. Glycolysis is a predominant process for most cancer cells to utilize glucose, which referred to as the Warburg Effect. Targeting critical enzymes, such as pyruvate dehydrogenase kinase 1 (PDK1) that inversely regulating the process of glycolysis could be a promising approach to work alone or in combination with other treatments for cancer therapy. The purpose of this study is to evaluate whether PDK1 inhibition could enhance the anti-cancer effects of EGFR-TKi. Herein, we utilized a recently reported PDK1 inhibitor 2,2-Dichloro-1-(4-isopropoxy-3-nitrophenyl)ethan-1-one (Cpd64), which was more potent and selective than dichloroacetate (DCA) and/or dichloroacetophenone (DAP), to study the mechanism of PDK1 inhibition in TKi-mediated anti-cancer activity. We found that the introduction of Cpd64 in EGFR-TKi therapy enhanced the anti-proliferative effects in EGFR-mutant NSCLC cells under hypoxia. In particular, Cpd64 was shown to increase the activity of pyruvate dehydrogenase (PDH) and improved XPHOS, such as elevated mitochondrial respiration, and increased ATP generation, which effectively modulated the upregulation of PDK1 by EGFR-TKi treatment. We have observed that Cpd64 effectively enhanced the tumor growth inhibition induced by erlotinib in a NCI-H1975 xenograft mouse model. Collectively, our results suggested that combined use of selective PDK inhibitor and EGFR-TKi could be a potential strategy for NSCLC therapy.
Collapse
Affiliation(s)
- Zheng Yang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Shao-Lin Zhang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xiaohui Hu
- Drug Development Core, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kin Yip Tam
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
28
|
Suraweera A, O’Byrne KJ, Richard DJ. Combination Therapy With Histone Deacetylase Inhibitors (HDACi) for the Treatment of Cancer: Achieving the Full Therapeutic Potential of HDACi. Front Oncol 2018; 8:92. [PMID: 29651407 PMCID: PMC5884928 DOI: 10.3389/fonc.2018.00092] [Citation(s) in RCA: 459] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/16/2018] [Indexed: 01/10/2023] Open
Abstract
Genetic and epigenetic changes in DNA are involved in cancer development and tumor progression. Histone deacetylases (HDACs) are key regulators of gene expression that act as transcriptional repressors by removing acetyl groups from histones. HDACs are dysregulated in many cancers, making them a therapeutic target for the treatment of cancer. Histone deacetylase inhibitors (HDACi), a novel class of small-molecular therapeutics, are now approved by the Food and Drug Administration as anticancer agents. While they have shown great promise, resistance to HDACi is often observed and furthermore, HDACi have shown limited success in treating solid tumors. The combination of HDACi with standard chemotherapeutic drugs has demonstrated promising anticancer effects in both preclinical and clinical studies. In this review, we summarize the research thus far on HDACi in combination therapy, with other anticancer agents and their translation into preclinical and clinical studies. We additionally highlight the side effects associated with HDACi in cancer therapy and discuss potential biomarkers to either select or predict a patient's response to these agents, in order to limit the off-target toxicity associated with HDACi.
Collapse
Affiliation(s)
- Amila Suraweera
- School of Biomedical Research, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kenneth J. O’Byrne
- School of Biomedical Research, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
- Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Derek J. Richard
- School of Biomedical Research, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
29
|
Yang Z, Tam KY. Combination Strategies Using EGFR-TKi in NSCLC Therapy: Learning from the Gap between Pre-Clinical Results and Clinical Outcomes. Int J Biol Sci 2018; 14:204-216. [PMID: 29483838 PMCID: PMC5821041 DOI: 10.7150/ijbs.22955] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/09/2017] [Indexed: 01/04/2023] Open
Abstract
Although epidermal growth factor receptor (EGFR) inhibitors have been used to treat non-small cell lung cancer (NSCLC) for decades with great success in patients with EGFR mutations, acquired resistance inevitably occurs after long-term exposure. More recently, combination therapy has emerged as a promising strategy to overcome this issue. Several experiments have been carried out to evaluate the synergism of combination therapy both in vitro and in vivo. Additionally, many clinical studies have been carried out to investigate the feasibility of treatment with EGFR-tyrosine kinase inhibitors (TKi) combined with other NSCLC treatments, including radiotherapy, cytotoxic chemotherapies, targeted therapies, and emerging immunotherapies. However, a significant gap still exists when applying pre-clinical results to clinical scenarios, which hinders the development and use of these strategies. This article is a literature review analysing the rationalities and controversies in the transition from pre-clinical investigation to clinical practice associated with various combination strategies. It also highlights clues and challenges regarding future combination therapeutic options in NSCLC treatment.
Collapse
Affiliation(s)
| | - Kin Yip Tam
- Faculty of Health Science, University of Macau, Taipa, Macau, China
| |
Collapse
|
30
|
Maleki Vareki S, Salim KY, Danter WR, Koropatnick J. Novel anti-cancer drug COTI-2 synergizes with therapeutic agents and does not induce resistance or exhibit cross-resistance in human cancer cell lines. PLoS One 2018; 13:e0191766. [PMID: 29364966 PMCID: PMC5783418 DOI: 10.1371/journal.pone.0191766] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 01/10/2018] [Indexed: 01/05/2023] Open
Abstract
Emerging drug-resistance and drug-associated toxicities are two major factors limiting successful cancer therapy. Combinations of chemotherapeutic drugs have been used in the clinic to improve patient outcome. However, cancer cells can acquire resistance to drugs, alone or in combination. Resistant tumors can also exhibit cross-resistance to other chemotherapeutic agents, resulting in sub-optimal treatment and/or treatment failure. Therefore, developing novel oncology drugs that induce no or little acquired resistance and with a favorable safety profile is essential. We show here that combining COTI-2, a novel clinical stage agent, with multiple chemotherapeutic and targeted agents enhances the activity of these drugs in vitro and in vivo. Importantly, no overt toxicity was observed in the combination treatment groups in vivo. Furthermore, unlike the tested chemotherapeutic drugs, cancer cells did not develop resistance to COTI-2. Finally, some chemo-resistant tumor cell lines only showed mild cross-resistance to COTI-2 while most remained sensitive to it.
Collapse
Affiliation(s)
- Saman Maleki Vareki
- Cancer Research Laboratory Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Kowthar Y. Salim
- Cotinga Pharmaceuticals (formerly Critical Outcome Technologies Inc.), London, Ontario, Canada
| | - Wayne R. Danter
- Cotinga Pharmaceuticals (formerly Critical Outcome Technologies Inc.), London, Ontario, Canada
| | - James Koropatnick
- Cancer Research Laboratory Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Department of Pathology, Western University, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
31
|
Patel HH, Maniar M, Ren C, Dave RH. Determination of Degradation Kinetics and Effect of Anion Exchange Resin on Dissolution of Novel Anticancer Drug Rigosertib in Acidic Conditions. AAPS PharmSciTech 2018; 19:93-100. [PMID: 28600665 DOI: 10.1208/s12249-017-0820-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023] Open
Abstract
Rigosertib is a novel anticancer drug in clinical development by Onconova therapeutics, Inc. Currently, it is in pivotal phase III clinical trials for myelodysplastic syndrome (MDS) patients. Chemically, it is a sodium salt of weak acid with low solubility in lower pH solutions. In the preliminary studies, it was found that rigosertib is unstable in acidic conditions and forms multiple degradation products. In this research, drug degradation kinetics of rigosertib were studied in acidic conditions. Rigosertib follows pseudo-first-order general acid catalysis reaction. Cholestyramine, which is a strong anion exchange resin, was used to form complex with drug to improve stability and dissolution in acidic conditions. Drug complex with cholestyramine showed better dissolution profile compared to drug alone. Effect of polyethylene glycol was investigated on the release of drug from the drug resin complex. Polyethylene glycol further improved dissolution profile by improving drug solubility in acidic medium.
Collapse
|
32
|
Mohareb RM, Abbas NS, Ibrahim RA. Uses of Cyclohexan-1,4-dione for the Synthesis of 2-Amino-4,5-dihydrobenzo[b]thiophen-6(7H)-one Derivatives with Anti-proliferative and Pim-1 Kinase Activities. Chem Pharm Bull (Tokyo) 2017; 65:1117-1131. [PMID: 29199218 DOI: 10.1248/cpb.c17-00582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The reaction of cyclohexan-1,4-dione with elemental sulfur and any of the 2-cyano-N-arylacetamide derivatives 2a-c gave the 2-amino-4,5-dihydrobenzo[b]thiophen-6(7H)-one derivatives 3a-c to be used in some heterocyclization reactions. The multicomponent reactions of any of compounds 3a-c with aromatic aldehydes 6a-c and either of malononitrile or ethylcyanoacetate gave the 5,9-dihydro-4H-thieno[2,3-f]chromene derivatives 9a-r, respectively. The anti-proliferative evaluation of the newly synthesized compounds against the six cancer cell lines A549, HT-29, MKN-45, U87MG, SMMC-7721 and H460 showed that the nine compounds 3c, 5c, 9e, 9h, 9i, 9j, 9l, 9q, 11e and 13e with highest cytotoxcity. Toxicity of these compounds against shrimp larvae revealed that compounds 3c, 9j, 9q, and 13e showed no toxicity against the tested organisms. The c-Met kinase inhibition of the most potent compounds showed that compounds 9j, 9q, 10e, 12e and 13e have the highest activities. Compounds 9j, 9l, 9q and 11e showed high activity towards tyrosine kinases. Moreover, compounds 9j, 9q and 13e showed the highest inhibitor activity towards Pim-1 kinase.
Collapse
Affiliation(s)
| | - Nermeen Saeed Abbas
- Department of Chemistry, Faculty of Science, Helwan University.,Department of Chemistry, Faculty of Science, Taibah University
| | | |
Collapse
|
33
|
Wu YP, Deng J, Ouyang SH, Mao ZF, Wang GE, Kurihara H, He RR, Li YF. Immune regulation effect of lienal polypeptides extract in Lewis lung carcinoma-bearing mice treated with cyclophosphamide. Exp Biol Med (Maywood) 2017; 243:66-77. [PMID: 29078731 DOI: 10.1177/1535370217737982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Polypeptides extracted from animal immune organs have been proved to exert immunomodulatory activities in previous reports. However, relative experimental data regarding the influence of a polypeptide mixture extracted from healthy calf spleen (lienal polypeptide [LP]) on the immune function in tumor therapy are limited, and the components in LP remain unclear. In the present study, the immune regulatory effect of LP was investigated in normal mice and Lewis lung carcinoma (LLC)-bearing mice treated with cyclophosphamide (CTX). The components of LP were identified by liquid chromatography-electrospray ionization-coupled with tandem mass spectrometry (LC-MS/MS) analysis and bioinformatic analysis. In LLC-bearing mice, LP showed a synergic antitumor effect with CTX, whereas LP alone did not present direct antitumor activity. Further, LP was found to enhance immune organ indexes, splenocyte number, and T lymphocyte subsets in normal mice and LLC-bearing mice treated with CTX. The decline of white blood cell and platelet counts, splenocyte proliferation activity, and peritoneal macrophage phagocytic function caused by CTX were also significantly suppressed by LP treatment in LLC-bearing mice. Notably, LP treatment significantly decreased the expression of phagocytosis-related proteins including CD47/signal regulatory protein α/Src homology phosphatase-1 in the tumor tissue of LLC-bearing mice treated with CTX. LC-MS/MS-based peptidomics unraveled the main polypeptides in LP with a length from 8 to 25 amino acids. Bioinformatics analysis further confirmed the possibility of LP to regulate immunity, especially in phagocytosis-related pathway. Our above findings indicated that LP can relieve the immunosuppression induced by chemotherapy and is a beneficial supplement in cancer therapy. Impact statement The immunomodulatory activities of polypeptides extracted from animal immune organs have incurred people's interests since a long time ago. In this study, we investigated the immune regulation effects of a polypeptide mixture extracted from health calf spleen (lienal polypeptide [LP]) in Lewis lung carcinoma-bearing mice treated with cyclophosphamide (CTX). Liquid chromatography-electrospray ionization-coupled with tandem mass spectrometry-based peptidomics and bioinformatics analysis unraveled the main polypeptides in LP and further confirmed that LP is mainly associated with immune regulating pathway, especially in tumor cell phagocytosis-related pathway. Our study for the first time revealed that polypeptides from spleen can relieve the immunosuppression induced by CTX and is a beneficial supplement in cancer therapy.
Collapse
Affiliation(s)
- Yan-Ping Wu
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jie Deng
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Shu-Hua Ouyang
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhong-Fu Mao
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Guo-En Wang
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Hiroshi Kurihara
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yi-Fang Li
- 1 Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China.,2 Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China
| |
Collapse
|
34
|
Mohareb RM, Al-Omran F, Ibrahim RA. The uses of cyclohexan-1,4-dione for the synthesis of thiophene derivatives as new anti-proliferative, prostate anticancer, c-Met and tyrosine kinase inhibitors. Med Chem Res 2017. [DOI: 10.1007/s00044-017-2087-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
35
|
Yang Z, Hu X, Zhang S, Zhang W, Tam KY. Pharmacological synergism of 2,2-dichloroacetophenone and EGFR-TKi to overcome TKi-induced resistance in NSCLC cells. Eur J Pharmacol 2017; 815:80-87. [PMID: 28870456 DOI: 10.1016/j.ejphar.2017.08.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 01/28/2023]
Abstract
Combination treatment has been used as one of the therapeutic approaches for patients suffering from lung cancer, either to cope with the issue of acquired drug resistance due to prolong the use of a particular EGFR-TKi treatment, or to decrease the doses of each compound in order to reduce potential toxicity. 2,2-dichloroacetophenone (DAP) was reported as a PDK inhibitor recently, which is much more potent than dichloroacetate (DCA) in anti-cancer therapy. In this study, we applied DAP in combined with EGFR-TKis, erlotinib or gefitinib in NSCLC cell lines and NSCLC xenograft model. Synergistic anti-cancer effects in two NSCLC cell lines with EGFR mutation, NCI-H1975 and NCI-H1650, as well as in NCI-H1975 xenograft model were observed. In comparison with either DAP or EGFR-TKi applied alone, the combination treatment not only further suppressed the EGFR signaling in vitro and in vivo, but also significantly promoted cell apoptosis. Interestingly, this synergistic anti-cancer effect was also observed in NCI-H1975 gefitinib induced-resistant cell line. Taken together, our results suggested that the combined use of DAP and EGFR-TKi exhibited anti-cancer synergy which may offer an additional treatment option for patients with EGFR-TKi induced-resistance.
Collapse
Affiliation(s)
- Zheng Yang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xiaohui Hu
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Shaolin Zhang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wen Zhang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
36
|
Zhang D, Kong YY, Sun JH, Huo SJ, Zhou M, Gui YL, Mu X, Chen H, Yu SQ, Xu Q. Co-delivery nanoparticles with characteristics of intracellular precision release drugs for overcoming multidrug resistance. Int J Nanomedicine 2017; 12:2081-2108. [PMID: 28356731 PMCID: PMC5360411 DOI: 10.2147/ijn.s128790] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Combination chemotherapy in clinical practice has been generally accepted as a feasible strategy for overcoming multidrug resistance (MDR). Here, we designed and successfully prepared a co-delivery system named S-D1@L-D2 NPs, where denoted some smaller nanoparticles (NPs) carrying a drug doxorubicin (DOX) were loaded into a larger NP containing another drug (vincristine [VCR]) via water-in-oil-in-water double-emulsion solvent diffusion-evaporation method. Chitosan-alginate nanoparticles carrying DOX (CS-ALG-DOX NPs) with a smaller diameter of about 20 nm formed S-D1 NPs; vitamin E D-α-tocopheryl polyethylene glycol 1000 succinate-modified poly(lactic-co-glycolic acid) nanoparticles carrying VCR (TPGS-PLGA-VCR NPs) with a larger diameter of about 200 nm constituted L-D2 NPs. Some CS-ALG-DOX NPs loaded into TPGS-PLGA-VCR NPs formed CS-ALG-DOX@TPGS-PLGA-VCR NPs. Under the acidic environment of cytosol and endosome or lysosome in MDR cell, CS-ALG-DOX@TPGS-PLGA-VCR NPs released VCR and CS-ALG-DOX NPs. VCR could arrest cell cycles at metaphase by inhibiting microtubule polymerization in the cytoplasm. After CS-ALG-DOX NPs escaped from endosome, they entered the nucleus through the nuclear pore and released DOX in the intra-nuclear alkaline environment, which interacted with DNA to stop the replication of MDR cells. These results indicated that S-D1@L-D2 NPs was a co-delivery system of intracellular precision release loaded drugs with pH-sensitive characteristics. S-D1@L-D2 NPs could obviously enhance the in vitro cytotoxicity and the in vivo anticancer efficiency of co-delivery drugs, while reducing their adverse effects. Overall, S-D1@L-D2 NPs can be considered an innovative platform for the co-delivery drugs of clinical combination chemotherapy for the treatment of MDR tumor.
Collapse
Affiliation(s)
- DanDan Zhang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Yan Yan Kong
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Jia Hui Sun
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Shao Jie Huo
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Min Zhou
- School of Pharmacy, Jiangsu Food and Pharmaceutical Science College, Huai’an
| | - Yi Ling Gui
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Xu Mu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Huan Chen
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Shu Qin Yu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing
| | - Qian Xu
- Ministry of Education Key Laboratory of Environmental Medicine and Engineering, School of Public Health, Southeast University, Nanjing, People’s Republic of China
| |
Collapse
|
37
|
Véras of Aguiar AC, of Moura RO, Bezerra Mendonça JF, de Oliveira Rocha HA, Gomes Câmara RB, dos Santos Carvalho Schiavon M. Evaluation of the antiproliferative activity of 2-amino thiophene derivatives against human cancer cells lines. Biomed Pharmacother 2016; 84:403-414. [DOI: 10.1016/j.biopha.2016.09.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 11/15/2022] Open
|
38
|
Zhang L, Tan L, Chen L, Chen X, Long C, Peng J, Qian Z. A simple method to improve the stability of docetaxel micelles. Sci Rep 2016; 6:36957. [PMID: 27833135 PMCID: PMC5105067 DOI: 10.1038/srep36957] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/24/2016] [Indexed: 02/05/2023] Open
Abstract
Self-assembled polymeric micelles have been widely applied in drug delivery systems. In this study, we found that pH value of micellar system solution was the decisive factor of physical stability. Furthermore, the weak basic solution could maintain the solution clarification for a relative long time. To investigate the stability of polymeric micelles in different pH solutions, the micellar particle size and the docetaxel content remaining in solution were detected at predetermined time points. The crystallographic assay of freeze-drying powder was characterized by an X-ray diffractometer. In vitro release results indicated that the PBS had little influence on the sustained-release effect of docetaxel-loaded polymeric micelles (DPM). Besides, the safety of micellar formulation was determined by an MTT assay on HEK293 cells, and the anti-tumor activity was tested on MCF-7 cells. The results demonstrated that DPM adjusted with PBS (DPM (PBS)) was of low toxicity and maintained the effectiveness of docetaxel. In vivo antitumor results indicated that DPM (PBS) had better antitumor efficacy than common docetaxel injection (DTX). Thus it was concluded that regulation of micellar solution PH by PBS is a safe and effective method to improve the physical stability of DPM. It might promote the application of micellar formulation in clinical applications.
Collapse
Affiliation(s)
- Lan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.,Research and Development Department, Guangdong Zhongsheng Pharmacy, Dongguan, 523325, China
| | - LiWei Tan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - LiJuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - XiaoXin Chen
- Research and Development Department, Guangdong Zhongsheng Pharmacy, Dongguan, 523325, China
| | - ChaoFeng Long
- Research and Development Department, Guangdong Zhongsheng Pharmacy, Dongguan, 523325, China
| | - JinRong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - ZhiYong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
39
|
Preston MA, Pimentel B, Bermejo-Rodríguez C, Dionne I, Turnbull A, de la Cueva-Méndez G. Repurposing a Prokaryotic Toxin-Antitoxin System for the Selective Killing of Oncogenically Stressed Human Cells. ACS Synth Biol 2016; 5:540-6. [PMID: 26230535 DOI: 10.1021/acssynbio.5b00096] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prokaryotes express intracellular toxins that pass unnoticed to carrying cells until coexpressed antitoxin partners are degraded in response to stress. Although not evolved to function in eukaryotes, one of these toxins, Kid, induces apoptosis in mammalian cells, an effect that is neutralized by its cognate antitoxin, Kis. Here we engineered this toxin-antitoxin pair to create a synthetic system that becomes active in human cells suffering a specific oncogenic stress. Inspired by the way Kid becomes active in bacterial cells, we produced a Kis variant that is selectively degraded in human cells expressing oncoprotein E6. The resulting toxin-antitoxin system functions autonomously in human cells, distinguishing those that suffer the oncogenic insult, which are killed by Kid, from those that do not, which remain protected by Kis. Our results provide a framework for developing personalized anticancer strategies avoiding off-target effects, a challenge that has been hardly tractable by other means thus far.
Collapse
Affiliation(s)
- Mark A. Preston
- MRC Cancer Cell Unit. Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
| | - Belén Pimentel
- MRC Cancer Cell Unit. Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
| | | | - Isabelle Dionne
- MRC Cancer Cell Unit. Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
| | - Alice Turnbull
- MRC Cancer Cell Unit. Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
| | - Guillermo de la Cueva-Méndez
- MRC Cancer Cell Unit. Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, U.K
- Andalusian Centre for Nanomedicine and Biotechnology (BIONAND), Parque Tecnológico de Andalucı́a, C/Severo Ochoa 35, 29590 Campanillas, Málaga, Spain
| |
Collapse
|
40
|
Kupcewicz B, Małecka M, Zapadka M, Krajewska U, Rozalski M, Budzisz E. Quantitative relationships between structure and cytotoxic activity of flavonoid derivatives. An application of Hirshfeld surface derived descriptors. Bioorg Med Chem Lett 2016; 26:3336-3341. [DOI: 10.1016/j.bmcl.2016.05.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/09/2016] [Accepted: 05/12/2016] [Indexed: 11/30/2022]
|
41
|
Suzumura DN, Schleder JC, Appel MH, Naliwaiko K, Tanhoffer R, Fernandes LC. Fish Oil Supplementation Enhances Pulmonary Strength and Endurance in Women Undergoing Chemotherapy. Nutr Cancer 2016; 68:935-42. [DOI: 10.1080/01635581.2016.1187282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
42
|
Ferreira PMP, Bezerra DP, Silva JDN, da Costa MP, Ferreira JRDO, Alencar NMN, Figueiredo ISTD, Cavalheiro AJ, Machado CML, Chammas R, Alves APNN, Moraes MOD, Pessoa C. Preclinical anticancer effectiveness of a fraction from Casearia sylvestris and its component Casearin X: in vivo and ex vivo methods and microscopy examinations. JOURNAL OF ETHNOPHARMACOLOGY 2016; 186:270-279. [PMID: 27067367 DOI: 10.1016/j.jep.2016.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Casearia sylvestris (Salicaceae) is found in South America and presents antiulcerogenic, cytotoxic, antimicrobial, anti-inflammatory and antihypertensive activities. AIM OF THE STUDY To assess the in vivo and ex vivo antitumor action of a fraction with casearins (FC) and its main component - Casearin X-isolated from C. sylvestris leaves. MATERIALS AND METHODS Firstly, Sarcoma 180 bearing Swiss mice were treated with FC and Cas X for 7 days. Secondly, BALB/c nude animals received hollow fibers with colon carcinoma (HCT-116) or glioblastoma (SF-295) cells and were treated with FC for 4 days. On 5th day, proliferation was determined by MTT assay. RESULTS FC 10 and 25mg/kg/day i.p. and 50mg/kg/day oral and Cas X 25mg/kg/day i.p. and 50mg/kg/day oral revealed tumor growth inhibition rates of 35.8, 86.2, 53.7, 90.0 and 65.5% and such tumors demonstrated rare mitoses and coagulation necrosis areas. Similarly, FC reduced multiplying of HCT-116 and SF-295 cells when evaluated by the Hollow Fiber Assay (2.5 and 5mg/kg/day i.p. and 25 and 50mg/kg/day oral), with cell growth inhibition rates ranging from 33.3 to 67.4% (p<0.05). Flow cytometry experiments revealed that FC reduced membrane integrity and induced DNA fragmentation and mitochondrial depolarization (p<0.05). CONCLUSIONS FC and Cas X were efficient antitumor substances against murine and human cancer cells and caused reversible morphological changes in liver, kidneys and spleens, emphasizing clerodane diterpenes as an emerging class of anticancer molecules.
Collapse
Affiliation(s)
- Paulo Michel Pinheiro Ferreira
- Laboratory in Experimental Cancerology, Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil; Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil; Postgraduate Program in Biotechnology, Federal University of Piauí, Teresina, Brazil.
| | | | | | - Marcília Pinheiro da Costa
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil; Department of Pharmacy, Federal University of Piauí, Teresina, Brazil
| | | | - Nylane Maria Nunes Alencar
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | | | | | - Camila Maria Longo Machado
- Radioisotopes Research Laboratory, Center for Medical Nuclear, State University of São Paulo, São Paulo, Brazil
| | - Roger Chammas
- Department of Radiology, Faculty of Medicine, State University of São Paulo, São Paulo, Brazil
| | | | - Manoel Odorico de Moraes
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Claudia Pessoa
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil; Oswaldo Cruz Foundation, Fortaleza, Brazil
| |
Collapse
|
43
|
Shin WS, Lee MG, Verwilst P, Lee JH, Chi SG, Kim JS. Mitochondria-targeted aggregation induced emission theranostics: crucial importance of in situ activation. Chem Sci 2016; 7:6050-6059. [PMID: 30034745 PMCID: PMC6022148 DOI: 10.1039/c6sc02236g] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/07/2016] [Indexed: 01/05/2023] Open
Abstract
A mitochondria targeted AIE fluorophore was further decorated with an NQO1 cleavable masking unit and showed selective targeting to and activation in cancer cells resulting in bright AIE fluorescence and apoptosis triggered by mitochondrial dysfunction.
Tissue selective targeting and specific suborganellular localization combined with an efficient pathology associated enzymatic activation of drugs in drug delivery systems may exhibit a clear advantage over conventional cancer treatment. Here, a mitochondria targeted aggregation induced emission (AIE) fluorophore further conjugated with an NAD(P)H:quinone oxidoreductase-1 (NQO1) cleavable masking unit showed preferential uptake in cancer cells and was selectively activated, resulting in bright AIE fluorescence and apoptosis via the caspase pathway, triggered by mitochondrial dysfunction. In vivo experimental data further support the conclusions from in vitro experiments, clearly showing the dependence of the therapy's success on both the suborganelle localization and specific in situ activation. And the site specific and enzyme dependent activation and aggregation was further supported by in vivo and ex vivo imaging. As a whole, the data comprised in this work represent a strong argument for the further development of this type of novel anticancer drugs.
Collapse
Affiliation(s)
- Weon Sup Shin
- Department of Chemistry , Korea University , Seoul , 136-701 , Korea .
| | - Min-Goo Lee
- School of Life Sciences and Biotechnology , Korea University , Seoul , 136-701 , Korea .
| | - Peter Verwilst
- Department of Chemistry , Korea University , Seoul , 136-701 , Korea .
| | - Joung Hae Lee
- Korea Research Institute of Standards and Science , Daejeon 305-600 , Korea
| | - Sung-Gil Chi
- School of Life Sciences and Biotechnology , Korea University , Seoul , 136-701 , Korea .
| | - Jong Seung Kim
- Department of Chemistry , Korea University , Seoul , 136-701 , Korea .
| |
Collapse
|
44
|
Nanoparticle-based combination drug delivery systems for synergistic cancer treatment. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2016. [DOI: 10.1007/s40005-016-0252-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
45
|
Hong WG, Cho JH, Hwang SG, Lee E, Lee J, Kim JI, Um HD, Park JK. Chemosensitizing effect of podophyllotoxin acetate on topoisomerase inhibitors leads to synergistic enhancement of lung cancer cell apoptosis. Int J Oncol 2016; 48:2265-76. [PMID: 27035096 PMCID: PMC4863929 DOI: 10.3892/ijo.2016.3471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/04/2016] [Indexed: 12/26/2022] Open
Abstract
Podophyllotoxin acetate (PA) acts as a radiosensitizer against non-small cell lung cancer (NSCLC) in vitro and in vivo. In this study, we examined its potential role as a chemosensitizer in conjunction with the topoisomerase inhibitors etoposide (Eto) and camptothecin (Cpt). The effects of combinations of PA and Eto/Cpt were examined with CompuSyn software in two NSCLC cell lines, A549 and NCI-H1299. Combination index (CI) values indicated synergistic effects of PA and the topoisomerase inhibitors. The intracellular mechanism underlying synergism was further determined using propidium iodide uptake, immunoblotting and electrophoretic mobility shift assay (EMSA). Combination of PA with Eto/Cpt promoted disruption of the dynamics of actin filaments, leading to subsequent enhancement of apoptotic cell death via induction of caspase-3, -8, and -9, accompanied by increased phosphorylation of p38. Conversely, suppression of p38 phosphorylation blocked the apoptotic effect of the drug combinations. Notably, CREB-1, a transcription factor, was constitutively activated in both cell types, and synergistically inhibited upon combination treatment. Our results collectively indicate that PA functions as a chemosensitizer by enhancing apoptosis through activation of the p38/caspase axis and suppression of CREB-1.
Collapse
Affiliation(s)
- Wan Gi Hong
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Jeong Hyun Cho
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Sang-Gu Hwang
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Eunah Lee
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si, Geonggi-do, Republic of Korea
| | - Jaeseok Lee
- Department of Biological Science, College of Biological Science, Konkuk University, Seoul, Republic of Korea
| | - Jong-Il Kim
- Department of Food and Microbial Technology, College of Natural Sciences, Seoul Women's University, Seoul, Republic of Korea
| | - Hong-Duck Um
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Jong Kuk Park
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| |
Collapse
|
46
|
Glasgow MDK, Chougule MB. Recent Developments in Active Tumor Targeted Multifunctional Nanoparticles for Combination Chemotherapy in Cancer Treatment and Imaging. J Biomed Nanotechnol 2016; 11:1859-98. [PMID: 26554150 DOI: 10.1166/jbn.2015.2145] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nanotechnology and combination therapy are two major fields that show great promise in the treatment of cancer. The delivery of drugs via nanoparticles helps to improve drug's therapeutic effectiveness while reducing adverse side effects associated wifh high dosage by improving their pharmacokinetics. Taking advantage of molecular markers over-expressing on tumor tissues compared to normal cells, an "active" molecular marker targeted approach would be-beneficial for cancer therapy. These actively targeted nanoparticles would increase drug concentration at the tumor site, improving efficacy while further reducing chemo-resistance. The multidisciplinary approach may help to improve the overall efficacy in cancer therapy. This review article summarizes recent developments of targeted multifunctional nanoparticles in the delivery, of various drugs for a combinational chemotherapy approach to cancer treatment and imaging.
Collapse
|
47
|
Liu Q, Zhang C, Ding X, Deng H, Zhang D, Cui W, Xu H, Wang Y, Xu W, Lv L, Zhang H, He Y, Wu Q, Szyf M, Ho CM, Zhu J. Preclinical optimization of a broad-spectrum anti-bladder cancer tri-drug regimen via the Feedback System Control (FSC) platform. Sci Rep 2015; 5:11464. [PMID: 26088171 PMCID: PMC5155572 DOI: 10.1038/srep11464] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022] Open
Abstract
Therapeutic outcomes of combination chemotherapy have not significantly advanced during the past decades. This has been attributed to the formidable challenges of optimizing drug combinations. Testing a matrix of all possible combinations of doses and agents in a single cell line is unfeasible due to the virtually infinite number of possibilities. We utilized the Feedback System Control (FSC) platform, a phenotype oriented approach to test 100 options among 15,625 possible combinations in four rounds of assaying to identify an optimal tri-drug combination in eight distinct chemoresistant bladder cancer cell lines. This combination killed between 82.86% and 99.52% of BCa cells, but only 47.47% of the immortalized benign bladder epithelial cells. Preclinical in vivo verification revealed its markedly enhanced anti-tumor efficacy as compared to its bi- or mono-drug components in cell line-derived tumor xenografts. The collective response of these pathways to component drugs was both cell type- and drug type specific. However, the entire spectrum of pathways triggered by the tri-drug regimen was similar in all four cancer cell lines, explaining its broad spectrum killing of BCa lines, which did not occur with its component drugs. Our findings here suggest that the FSC platform holdspromise for optimization of anti-cancer combination chemotherapy.
Collapse
Affiliation(s)
- Qi Liu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China, and Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xianting Ding
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Hui Deng
- Cancer Epigenetics Program, Anhui Cancer Hospital, Hefei, Anhui 230031, China
| | - Daming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wei Cui
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China, and Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Hongwei Xu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yingwei Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wanhai Xu
- Department of Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lei Lv
- Cancer Epigenetics Program, Anhui Cancer Hospital, Hefei, Anhui 230031, China
| | - Hongyu Zhang
- Cancer Epigenetics Program, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University, Shanghai 200032, China
| | - Yinghua He
- Cancer Epigenetics Program, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University, Shanghai 200032, China
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China, and Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics McGill University Medical School 3655 Sir William Osler Promenade #1309, Montreal, Quebec Canada
| | - Chih-Ming Ho
- Mechanical and Aerospace Engineering Department, Biomedical Engineering Department, University of California, Los Angeles, CA 90095-1597, USA
| | - Jingde Zhu
- 1] Cancer Epigenetics Program, Anhui Cancer Hospital, Hefei, Anhui 230031, China [2] Cancer Epigenetics Program, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University, Shanghai 200032, China
| |
Collapse
|
48
|
Self-assembled nanoscale coordination polymers carrying oxaliplatin and gemcitabine for synergistic combination therapy of pancreatic cancer. J Control Release 2015; 201:90-9. [PMID: 25620067 DOI: 10.1016/j.jconrel.2015.01.026] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/12/2015] [Accepted: 01/22/2015] [Indexed: 01/05/2023]
Abstract
Gemcitabine has long been the standard of care for treating pancreatic ductal adenocarcinoma (PDAC), despite its poor pharmacokinetics/dynamics and rapid development of drug resistance. In this study, we have developed a novel nanoparticle platform based on nanoscale coordination polymer-1 (NCP-1) for simultaneous delivery of two chemotherapeutics, oxaliplatin and gemcitabine monophosphate (GMP), at 30 wt.% and 12 wt.% drug loadings, respectively. A strong synergistic therapeutic effect of oxaliplatin and GMP was observed in vitro against AsPc-1 and BxPc-3 pancreatic cancer cells. NCP-1 particles effectively avoid uptake by the mononuclear phagocyte system (MPS) in vivo with a long blood circulation half-life of 10.1 ± 3.3h, and potently inhibit tumor growth when compared to NCP particles carrying oxaliplatin or GMP alone. Our findings demonstrate NCP-1 as a novel nanocarrier for the co-delivery of two chemotherapeutics that have distinctive mechanisms of action to simultaneously disrupt multiple anticancer pathways with maximal therapeutic efficacy and minimal side effects.
Collapse
|
49
|
Song J, Zhang Y, Zhang W, Chen J, Yang X, Ma P, Zhang B, Liu B, Ni J, Wang R. Cell penetrating peptide TAT can kill cancer cells via membrane disruption after attachment of camptothecin. Peptides 2015; 63:143-9. [PMID: 25496911 DOI: 10.1016/j.peptides.2014.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Abstract
Attachment of traditional anticancer drugs to cell penetrating peptides is an effective strategy to improve their application in cancer treatment. In this study, we designed and synthesized the conjugates TAT-CPT and TAT-2CPT by attaching camptothecin (CPT) to the N-terminus of the cell penetrating peptide TAT. Interestingly, we found that TAT-CPT and especially TAT-2CPT could kill cancer cells via membrane disruption, which is similar to antimicrobial peptides. This might be because that CPT could perform as a hydrophobic residue to increase the extent of membrane insertion of TAT and the stability of the pores. In addition, TAT-CPT and TAT-2CPT could also kill cancer cells by the released CPT after they entered cells. Taken together, attachment of CPT could turn cell penetrating peptide TAT into an antimicrobial peptide with a dual mechanism of anticancer action, which presents a new strategy to develop anticancer peptides based on cell penetrating peptides.
Collapse
Affiliation(s)
- Jingjing Song
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yun Zhang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Wei Zhang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jianbo Chen
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiaoli Yang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Panpan Ma
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Bangzhi Zhang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Beijun Liu
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jingman Ni
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Rui Wang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
50
|
Liu MY, Xiao L, Chen GH, Wang YX, Xiong WX, Li F, Liu Y, Huang XL, Deng YF, Zhang Z, Sun HY, Liu QH, Yin M. Oral JS-38, a metabolite from Xenorhabdus sp., has both anti-tumor activity and the ability to elevate peripheral neutrophils. Chin J Nat Med 2014; 12:768-76. [PMID: 25443370 DOI: 10.1016/s1875-5364(14)60117-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Indexed: 01/01/2023]
Abstract
AIM JS-38 (mitothiolore), a synthetic version of a metabolite isolated from Xenorhabdus sp., was evaluated for its anti-tumor and white blood cell (WBC) elevating activities. METHOD These anti-proliferative activities were assessed in vitro using a panel of ten cell lines. The anti-tumor activities were tested in vivo using B16 allograft mouse models and xenograft models of A549 human lung carcinoma and QGY human hepatoma in nude mice. The anti-tumor interactions of JS-38 and cyclophosphamide (CTX) or 5-fluorouracil (5-Fu) were studied in a S180 sarcoma model in ICR mice. Specific stimulatory effects were determined on peripheral neutrophils in normal and CTX- and 5-Fu-induced neutropenic mice. RESULTS The IC50 values ranged from 0.1 to 2.0 μmol·L(-1). JS-38 (1 μmol·L(-1)) caused an increase in A549 tumor cell apoptosis. Multi-daily gavage of JS-38 (15, 30, and 60 mg·kg(-1)·d(-1)) inhibited in vivo tumor progression without a significant effect on body weight. JS-38 additively enhanced the in vivo anti-tumor effects of CTX or 5-Fu. JS-38 increased peripheral neutrophil counts and neutrophil rates in normal BALB/c mice almost as effectively as granulocyte colony-stimulating factor (G-CSF). In mice with neutropenia induced by CTX or 5-Fu, JS-38 rapidly restored neutrophil counts. CONCLUSION These results suggest that JS-38 has anti-tumor activity, and also has the ability to increase peripheral blood neutrophils.
Collapse
Affiliation(s)
- Min-Yu Liu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Lin Xiao
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Geng-Hui Chen
- Beijing Wenfeng Tianji Pharmaceuticals Ltd., Beijing 100027, China
| | - Yong-Xiang Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei-Xia Xiong
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Fei Li
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Ying Liu
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Xiao-Ling Huang
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Yi-Fang Deng
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Zhen Zhang
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Hai-Yan Sun
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China
| | - Quan-Hai Liu
- Department of Pharmacology, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China.
| | - Ming Yin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|