1
|
van den Bemt PMLA, Blijham MY, Ten Broek L, Hugtenburg JG, Pouls BPH, van Boven JFM, Bekker CL, van den Bemt B, van Dijk L. Patient reported medication-related problems, adherence and waste of oral anticancer medication over time. Eur J Hosp Pharm 2024:ejhpharm-2024-004205. [PMID: 39019578 DOI: 10.1136/ejhpharm-2024-004205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
OBJECTIVES Patients on oral anticancer therapy regularly experience medication-related problems (MRPs), potentially leading to non-adherence and medication waste. Most studies reporting these experiences have cross-sectional designs. The aim of our study was to explore patient reported MRPs, adherence and waste of oral anticancer medication over time. METHODS A prospective longitudinal quantitative interview study with 4 months follow-up was performed among patients on oral anticancer medication (mainly tyrosine kinase inhibitors, (anti)hormonal therapy, pyrimidine antagonists) using a semi-structured questionnaire. Patients from two Dutch university medical centres were included from March to December 2022 after informed consent was given. Four interviews were performed with 1 month in between. All interviews were audiotaped, after which the data were entered into an electronic case report form. The primary outcome was the mean number of MRPs per patient per interview round. Secondary outcomes were the proportion of patients with at least one MRP, types of MRPs, perceived non-adherence, medication waste (both in general and specifically for anticancer medication), costs of anticancer medication waste, and factors associated with medication waste as mentioned by the patient. Descriptive statistics were used to analyse the data. RESULTS Forty patients were included with a mean (SD) age of 64 (9) years; 43% were male. The mean number of MRPs per patient was 2.1 in the first interview and 1.2, 1.0 and 0.9 in the second, third and fourth interviews, respectively. Adverse drug reactions were the most frequently reported type of MRPs (30 (75%) patients in the first interview and 19 (65%) in the last interview). Unintentional non-adherence was regularly reported, especially in the first interview. Medication changes were frequent and associated medication waste was mentioned in all interviews. CONCLUSIONS Many patients using oral anticancer treatment report MRPs and this number remains substantial over time.
Collapse
Affiliation(s)
- Patricia M L A van den Bemt
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Margriet Y Blijham
- Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Laura Ten Broek
- Hospital Pharmacy, Reinier de Graaf Gasthuis, Delft, The Netherlands
| | - Jacqueline G Hugtenburg
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
| | - Bart P H Pouls
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Job F M van Boven
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Charlotte L Bekker
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Liset van Dijk
- Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Netherlands Institute for Health Services Research, Utrecht, The Netherlands
| |
Collapse
|
2
|
Surwase AJ, Thakur NL. Production of marine-derived bioactive peptide molecules for industrial applications: A reverse engineering approach. Biotechnol Adv 2024; 77:108449. [PMID: 39260778 DOI: 10.1016/j.biotechadv.2024.108449] [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: 07/13/2023] [Revised: 06/28/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
This review examines a wide range of marine microbial-derived bioactive peptide molecules, emphasizing the significance of reverse engineering in their production. The discussion encompasses the advancements in Marine Natural Products (MNPs) bio-manufacturing through the integration of omics-driven microbial engineering and bioinformatics. The distinctive features of non-ribosomally synthesised peptides (NRPs), and ribosomally synthesised precursor peptides (RiPP) biosynthesis is elucidated and presented. Additionally, the article delves into the origins of common peptide modifications. It highlights various genome mining approaches for the targeted identification of Biosynthetic Gene Clusters (BGCs) and novel RiPP and NRPs-derived peptides. The review aims to demonstrate the advancements, prospects, and obstacles in engineering both RiPP and NRP biosynthetic pathways.
Collapse
Affiliation(s)
- Akash J Surwase
- CSIR-National Institute of Oceanography, Dona Paula 403004, Goa, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Narsinh L Thakur
- CSIR-National Institute of Oceanography, Dona Paula 403004, Goa, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
3
|
Qu M, Chen J, Xu B, Shi Q, Zhao S, Wang Z, Li Z, Ma B, Xu H, Ye Q, Xie J. Assessing genotoxic effects of chemotherapy agents by a robust in vitro assay based on mass spectrometric quantification of γ-H2AX in HepG2 cells. Front Pharmacol 2024; 15:1356753. [PMID: 38962306 PMCID: PMC11219945 DOI: 10.3389/fphar.2024.1356753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Chemotherapy has already proven widely effective in treating cancer. Chemotherapeutic agents usually include DNA damaging agents and non-DNA damaging agents. Assessing genotoxic effect is significant during chemotherapy drug development, since the ability to attack DNA is the major concern for DNA damaging agents which relates to the therapeutic effect, meanwhile genotoxicity should also be evaluated for chemotherapy agents' safety especially for non-DNA damaging agents. However, currently applicability of in vitro genotoxicity assays is hampered by the fact that genotoxicity results have comparatively high false positive rates. γ-H2AX has been shown to be a bifunctional biomarker reflecting both DNA damage response and repair. Previously, we developed an in vitro genotoxicity assay based on γ-H2AX quantification using mass spectrometry. Here, we employed the assay to quantitatively assess the genotoxic effects of 34 classic chemotherapy agents in HepG2 cells. Results demonstrated that the evaluation of cellular γ-H2AX could be an effective approach to screen and distinguish types of action of different classes of chemotherapy agents. In addition, two crucial indexes of DNA repair kinetic curve, i.e., k (speed of γ-H2AX descending) and t50 (time required for γ-H2AX to drop to half of the maximum value) estimated by our developed online tools were employed to further evaluate nine representative chemotherapy agents, which showed a close association with therapeutic index or carcinogenic level. The present study demonstrated that mass spectrometric quantification of γ-H2AX may be an appropriate tool to preliminarily evaluate genotoxic effects of chemotherapy agents.
Collapse
Affiliation(s)
- Minmin Qu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jia Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Bin Xu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Qinyun Shi
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Shujing Zhao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhaoxia Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhi Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Bo Ma
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hua Xu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
| | - Jianwei Xie
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
4
|
Abrahams B, Gerber A, Hiss DC. Combination Treatment with EGFR Inhibitor and Doxorubicin Synergistically Inhibits Proliferation of MCF-7 Cells and MDA-MB-231 Triple-Negative Breast Cancer Cells In Vitro. Int J Mol Sci 2024; 25:3066. [PMID: 38474312 DOI: 10.3390/ijms25053066] [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: 02/19/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
The role of the epidermal growth factor receptor (EGFR) in tumor progression and survival is often underplayed. Its expression and/or dysregulation is associated with disease advancement and poor patient outcome as well as drug resistance in breast cancer. EGFR is often overexpressed in breast cancer and particularly triple-negative breast cancer (TNBC), which currently lacks molecular targets. We examined the synergistic potential of an EGFR inhibitor (EGFRi) in combination with doxorubicin (Dox) in estrogen-positive (ER+) MCF-7 and MDA-MB-231 TNBC cell lines. The exposure of MDA-MB-231 and MCF-7 to EGFRi produced an IC50s of 6.03 µM and 3.96 µM, respectively. Dox induced MDA-MB-231 (IC50 9.67 µM) and MCF-7 (IC50 1.4 µM) cytotoxicity. Combinations of EGFRi-Dox significantly reduced the IC50 in MCF-7 (0.46 µM) and MBA-MB 231 (0.01 µM). Synergistic drug interactions in both cell lines were confirmed using the Bliss independence model. Pro-apoptotic Caspase-3/7 activation occurred in MCF-7 at 0.1-10 µM of EGFRi and Dox single treatments, whilst 1 μM Dox yielded a more potent effect on MDA-MB-231. EGFRi and Dox individually and in combination downregulated the EGFR gene expression in MCF-7 and MDA-MB-231 (p < 0.001). This study demonstrates EGFRi's potential for eliciting synergistic interactions with Dox, causing enhanced growth inhibition, apoptosis induction, and downregulation of EGFR in both cell lines.
Collapse
Affiliation(s)
- Beynon Abrahams
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Anthonie Gerber
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Donavon Charles Hiss
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Bellville 7535, South Africa
| |
Collapse
|
5
|
Brandão SR, Reis-Mendes A, Araújo MD, Neuparth MJ, Rocha H, Carvalho F, Ferreira R, Costa VM. Cardiac Molecular Remodeling by Anticancer Drugs: Doxorubicin Affects More Metabolism While Mitoxantrone Impacts More Autophagy in Adult CD-1 Male Mice. Biomolecules 2023; 13:921. [PMID: 37371499 PMCID: PMC10296231 DOI: 10.3390/biom13060921] [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: 04/17/2023] [Revised: 05/14/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Doxorubicin (DOX) and mitoxantrone (MTX) are classical chemotherapeutic agents used in cancer that induce similar clinical cardiotoxic effects, although it is not clear if they share similar underlying molecular mechanisms. We aimed to assess the effects of DOX and MTX on the cardiac remodeling, focusing mainly on metabolism and autophagy. Adult male CD-1 mice received pharmacologically relevant cumulative doses of DOX (18 mg/kg) and MTX (6 mg/kg). Both DOX and MTX disturbed cardiac metabolism, decreasing glycolysis, and increasing the dependency on fatty acids (FA) oxidation, namely, through decreased AMP-activated protein kinase (AMPK) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) content and decreased free carnitine (C0) and increased acetylcarnitine (C2) concentration. Additionally, DOX heavily influenced glycolysis, oxidative metabolism, and amino acids turnover by exclusively decreasing phosphofructokinase (PFKM) and electron transfer flavoprotein-ubiquinone oxidoreductase (ETFDH) content, and the concentration of several amino acids. Conversely, both drugs downregulated autophagy given by the decreased content of autophagy protein 5 (ATG5) and microtubule-associated protein light chain 3 (LC3B), with MTX having also an impact on Beclin1. These results emphasize that DOX and MTX modulate cardiac remodeling differently, despite their clinical similarities, which is of paramount importance for future treatments.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Reis-Mendes
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Margarida Duarte Araújo
- LAQV-REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Department of Imuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Maria João Neuparth
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
- TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, 4585-116 Gandra, Portugal
| | - Hugo Rocha
- Newborn Screening, Metabolism and Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, 4000-053 Porto, Portugal
- Department of Pathological, Cytological and Thanatological Anatomy, School of Health, Polytechnic Institute of Porto, 4200-072 Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
6
|
Brandão SR, Carvalho F, Amado F, Ferreira R, Costa VM. Insights on the molecular targets of cardiotoxicity induced by anticancer drugs: A systematic review based on proteomic findings. Metabolism 2022; 134:155250. [PMID: 35809654 DOI: 10.1016/j.metabol.2022.155250] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/26/2022] [Indexed: 11/27/2022]
Abstract
Several anticancer agents have been associated with cardiac toxic effects. The currently proposed mechanisms to explain cardiotoxicity differ among anticancer agents, but in fact, the specific modulation is not completely elucidated. Thus, this systematic review aims to provide an integrative perspective of the molecular mechanisms underlying the toxicity of anticancer agents on heart muscle while using a high-throughput technology, mass spectrometry (MS)-based proteomics. A literature search using PubMed database led to the selection of 27 studies, of which 13 reported results exclusively on animal models, 13 on cardiomyocyte-derived cell lines and only one included both animal and a cardiomyocyte line. The reported anticancer agents were the proteasome inhibitor carfilzomib, the anthracyclines daunorubicin, doxorubicin, epirubicin and idarubicin, the antimicrotubule agent docetaxel, the alkylating agent melphalan, the anthracenedione mitoxantrone, the tyrosine kinase inhibitors (TKIs) erlotinib, lapatinib, sorafenib and sunitinib, and the monoclonal antibody trastuzumab. Regarding the MS-based proteomic approaches, electrophoretic separation using two-dimensional (2D) gels coupled with tandem MS (MS/MS) and liquid chromatography-MS/MS (LC-MS/MS) were the most common. Overall, the studies highlighted 1826 differentially expressed proteins across 116 biological processes. Most of them were grouped in larger processes and critically analyzed in the present review. The selection of studies using proteomics on heart muscle allowed to obtain information about the anticancer therapy-induced modulation of numerous proteins in this tissue and to establish connections that have been disregarded in other studies. This systematic review provides interesting points for a comprehensive understanding of the cellular cardiotoxicity mechanisms of different anticancer drugs.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 28, 4050-313 Porto, Portugal; LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 28, 4050-313 Porto, Portugal
| | - Francisco Amado
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 28, 4050-313 Porto, Portugal.
| |
Collapse
|
7
|
Conde I, Ribeiro AS, Paredes J. Breast Cancer Stem Cell Membrane Biomarkers: Therapy Targeting and Clinical Implications. Cells 2022; 11:934. [PMID: 35326385 PMCID: PMC8946706 DOI: 10.3390/cells11060934] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common malignancy affecting women worldwide. Importantly, there have been significant improvements in prevention, early diagnosis, and treatment options, which resulted in a significant decrease in breast cancer mortality rates. Nevertheless, the high rates of incidence combined with therapy resistance result in cancer relapse and metastasis, which still contributes to unacceptably high mortality of breast cancer patients. In this context, a small subpopulation of highly tumourigenic cancer cells within the tumour bulk, commonly designated as breast cancer stem cells (BCSCs), have been suggested as key elements in therapy resistance, which are responsible for breast cancer relapses and distant metastasis. Thus, improvements in BCSC-targeting therapies are crucial to tackling the metastatic progression and might allow therapy resistance to be overcome. However, the design of effective and specific BCSC-targeting therapies has been challenging since there is a lack of specific biomarkers for BCSCs, and the most common clinical approaches are designed for commonly altered BCSCs signalling pathways. Therefore, the search for a new class of BCSC biomarkers, such as the expression of membrane proteins with cancer stem cell potential, is an area of clinical relevance, once membrane proteins are accessible on the cell surface and easily recognized by specific antibodies. Here, we discuss the significance of BCSC membrane biomarkers as potential prognostic and therapeutic targets, reviewing the CSC-targeting therapies under clinical trials for breast cancer.
Collapse
Affiliation(s)
- Inês Conde
- i3S, Institute of Investigation and Innovation in Health, 4200-135 Porto, Portugal; (I.C.); (A.S.R.)
- Ipatimup, Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Ana Sofia Ribeiro
- i3S, Institute of Investigation and Innovation in Health, 4200-135 Porto, Portugal; (I.C.); (A.S.R.)
- Ipatimup, Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Joana Paredes
- i3S, Institute of Investigation and Innovation in Health, 4200-135 Porto, Portugal; (I.C.); (A.S.R.)
- Ipatimup, Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
8
|
Álvarez-Ortiz P, Ascacio-Valdés J, Vera-Reyes I, Esparza-González C, Rodríguez-Herrera R, Salinas-Santander M, del Ángel-Martínez M, Morlett-Chávez A. Purshia plicata Triggers and Regulates Proteins Related to Apoptosis in HeLa Cancer Cells. PLANTS (BASEL, SWITZERLAND) 2021; 10:2559. [PMID: 34961030 PMCID: PMC8707402 DOI: 10.3390/plants10122559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 05/12/2023]
Abstract
Cervical cancer represents a public health problem, develops resistance to traditional therapies and cost-of-treatment is high. These disadvantages have led to the search for alternative bioactive-compound-based therapies. Said bioactive compounds include phenolic compounds, flavonoids, and tannins. The present study aimed to evaluate the therapeutic effect of a P. plicata extract on the HeLa cell line. Viability and apoptosis assays were run on the two cell lines treated with the extract. The peptides, up- and down-expressed in both cell lines, were identified by PDQuest analysis software and high-performance liquid chromatography/mass spectrometry/mass spectrometry (HPLC/MS/MS). Our results show that a 500 mg/L treatment deregulated cell viability, with different apoptotic morphologies observed which are associated with the presence of bio-compounds, which up- and down-regulated the peptides. In conclusion, P. plicata regulates proteins associated with apoptosis in HeLa cancer cells.
Collapse
Affiliation(s)
- Patricia Álvarez-Ortiz
- Laboratory of Molecular Biology, Chemistry School, Autonomous University of Coahuila, Saltillo 25280, Mexico;
| | - Juan Ascacio-Valdés
- Bioprocesses and Bioproducts Research Group and Laboratory of Molecular Biology, Food Research Department, Chemistry School, Autonomous University of Coahuila, Saltillo 25280, Mexico; (J.A.-V.); (R.R.-H.)
| | - Ileana Vera-Reyes
- Proteomics Laboratory, Agricultural Plant Science and Biotechnology, Research Center for Applied Chemistry, Blvd. Enrique Reyna 140, Saltillo 25294, Mexico;
| | - Cecilia Esparza-González
- Laboratory of Histology, Dentistry School, Autonomous University of Coahuila, Saltillo 25125, Mexico;
| | - Raúl Rodríguez-Herrera
- Bioprocesses and Bioproducts Research Group and Laboratory of Molecular Biology, Food Research Department, Chemistry School, Autonomous University of Coahuila, Saltillo 25280, Mexico; (J.A.-V.); (R.R.-H.)
| | - Mauricio Salinas-Santander
- Laboratory of Molecular Biology, Health Research Department, Medicine School, Autonomous University of Coahuila, Saltillo 25000, Mexico; (M.S.-S.); (M.d.Á.-M.)
| | - Mayela del Ángel-Martínez
- Laboratory of Molecular Biology, Health Research Department, Medicine School, Autonomous University of Coahuila, Saltillo 25000, Mexico; (M.S.-S.); (M.d.Á.-M.)
| | - Antonio Morlett-Chávez
- Laboratory of Molecular Biology, Chemistry School, Autonomous University of Coahuila, Saltillo 25280, Mexico;
- Laboratory of Molecular Biology, Health Research Department, Medicine School, Autonomous University of Coahuila, Saltillo 25000, Mexico; (M.S.-S.); (M.d.Á.-M.)
- Clinical Laboratory Department, General Hospital No. 2, Mexican Institute of Social Security, Saltillo 25017, Mexico
| |
Collapse
|
9
|
Limam I, Ben Aissa-Fennira F, Essid R, Chahbi A, Kefi S, Mkadmini K, Elkahoui S, Abdelkarim M. Hydromethanolic root and aerial part extracts from Echium arenarium Guss suppress proliferation and induce apoptosis of multiple myeloma cells through mitochondrial pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:874-886. [PMID: 33393729 DOI: 10.1002/tox.23090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
Echium arenarium Guss is a Mediterranean plant traditionally used in healing skin wound and it was reported exhibiting potent antioxidant, antibacterial, and antiparasitic activities. However, antitumoral activities of this plant have not yet been explored. Here we investigated for the first time, root (EARE) and aerial part (EAAPE) extracts of E. arenarium Guss to examine cytotoxicity and apoptosis activation pathway on U266 human multiple myeloma (MM) cell line. We demonstrated that EARE and EAAPE decreased U266 cell viability in a dose dependent manner. Based on 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, EARE was significantly two times more efficient (IC50 value 41 μg/ml) than EAAPE (IC50 value 82 μg/ml) considering 48 h of treatment. Furthermore, after 24 h of exposure to 100 μg/ml of EARE or EAAPE, cell cycle showed remarkable increase in sub-G1 population and a decrease of U266 cells proportion in G1 phase. In addition, EARE increased cell percentage in S phase. Moreover, analysis revealed that EAAPE or EARE induced apoptosis of U266 cells after 24 h of treatment. Interestingly, depolarization of mitochondrial membrane potential and activation of caspase 3/7 were demonstrated in treated U266 cells. Phytochemical analysis of E. arenarium extracts showed that EARE exhibited the highest content of total phenolic content. Interestingly, six phenolic compounds were identified. Myricitrin was the major compound in EARE, followed by luteolin 7-O-glucoside, resorcinol, polydatin, Trans-hydroxycinnamic acid, and hyperoside. These findings proved that an intrinsic mitochondria-mediated apoptosis pathway probably mediated the apoptotic effects of E. arenarium Guss extracts on U266 cells, and this will suggest several action plans to treat MM.
Collapse
Affiliation(s)
- Inès Limam
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Fatma Ben Aissa-Fennira
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Rim Essid
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
| | - Ahlem Chahbi
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Sarra Kefi
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
| | - Khaoula Mkadmini
- Medicinal and Aromatic Plants Laboratory, Biotechnology Center of Borj Cedria, Hammam-Lif, Tunisia
| | - Salem Elkahoui
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mohamed Abdelkarim
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
10
|
Sun R, Ying Y, Tang Z, Liu T, Shi F, Li H, Guo T, Huang S, Lai R. The Emerging Role of the SLCO1B3 Protein in Cancer Resistance. Protein Pept Lett 2020; 27:17-29. [PMID: 31556849 PMCID: PMC6978646 DOI: 10.2174/0929866526666190926154248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/08/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Currently, chemotherapy is one of the mainstays of oncologic therapies. But the efficacy of chemotherapy is often limited by drug resistance and severe side effects. Consequently, it is becoming increasingly important to investigate the underlying mechanism and overcome the problem of anticancer chemotherapy resistance. The solute carrier organic anion transporter family member 1B3 (SLCO1B3), a functional transporter normally expressed in the liver, transports a variety of endogenous and exogenous compounds, including hormones and their conjugates as well as some anticancer drugs. The extrahepatic expression of SLCO1B3 has been detected in different cancer cell lines and cancer tissues. Recently, accumulating data indicates that the abnormal expression and function of SLCO1B3 are involved in resistance to anticancer drugs, such as taxanes, camptothecin and its analogs, SN-38, and Androgen Deprivation Therapy (ADT) in breast, prostate, lung, hepatic, and colorectal cancer, respectively. Thus, more investigations have been implemented to identify the potential SLCO1B3-related mechanisms of cancer drug resistance. In this review, we focus on the emerging roles of SLCO1B3 protein in the development of cancer chemotherapy resistance and briefly discuss the mechanisms of resistance. Elucidating the function of SLCO1B3 in chemoresistance may bring out novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Ruipu Sun
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China.,Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Zhimin Tang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Ting Liu
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Fuli Shi
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Huixia Li
- Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Taichen Guo
- Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Shibo Huang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Pharmacy, Medical College, Nanchang University, Nanchang 330006, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences / Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| |
Collapse
|
11
|
Ferroni C, Varchi G. Non-Steroidal Androgen Receptor Antagonists and Prostate Cancer: A Survey on Chemical Structures Binding this Fast-Mutating Target. Curr Med Chem 2019; 26:6053-6073. [PMID: 30209993 DOI: 10.2174/0929867325666180913095239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 02/01/2023]
Abstract
The Androgen Receptor (AR) pathway plays a major role in both the pathogenesis and progression of prostate cancer. In particular, AR is chiefly involved in the development of Castration-Resistant Prostate Cancer (CRPC) as well as in the resistance to the secondgeneration AR antagonist enzalutamide, and to the selective inhibitor of cytochrome P450 17A1 (CYP17A1) abiraterone. Several small molecules acting as AR antagonists have been designed and developed so far, also as a result of the ability of cells expressing this molecular target to rapidly develop resistance and turn pure receptor antagonists into ineffective or event detrimental molecules. This review covers a survey of most promising classes of non-steroidal androgen receptor antagonists, also providing insights into their mechanism of action and efficacy in treating prostate cancer.
Collapse
Affiliation(s)
- Claudia Ferroni
- Institute of Organic Synthesis and Photoreactivity - ISOF, Italian National Research Council, Bologna, Italy
| | - Greta Varchi
- Institute of Organic Synthesis and Photoreactivity - ISOF, Italian National Research Council, Bologna, Italy
| |
Collapse
|
12
|
Cancer Stem Cells: Powerful Targets to Improve Current Anticancer Therapeutics. Stem Cells Int 2019; 2019:9618065. [PMID: 31781251 PMCID: PMC6874936 DOI: 10.1155/2019/9618065] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/25/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023] Open
Abstract
A frequent observation in several malignancies is the development of resistance to therapy that results in frequent tumor relapse and metastasis. Much of the tumor resistance phenotype comes from its heterogeneity that halts the ability of therapeutic agents to eliminate all cancer cells effectively. Tumor heterogeneity is, in part, controlled by cancer stem cells (CSC). CSC may be considered the reservoir of cancer cells as they exhibit properties of self-renewal and plasticity and the capability of reestablishing a heterogeneous tumor cell population. The endowed resistance mechanisms of CSC are mainly attributed to several factors including cellular quiescence, accumulation of ABC transporters, disruption of apoptosis, epigenetic reprogramming, and metabolism. There is a current need to develop new therapeutic drugs capable of targeting CSC to overcome tumor resistance. Emerging in vitro and in vivo studies strongly support the potential benefits of combination therapies capable of targeting cancer stem cell-targeting agents. Clinical trials are still underway to address the pharmacokinetics, safety, and efficacy of combination treatment. This review will address the main characteristics, therapeutic implications, and perspectives of targeting CSC to improve current anticancer therapeutics.
Collapse
|
13
|
Zhang L, Pattanayak A, Li W, Ko HK, Fowler G, Gordon R, Bergan R. A Multifunctional Therapy Approach for Cancer: Targeting Raf1- Mediated Inhibition of Cell Motility, Growth, and Interaction with the Microenvironment. Mol Cancer Ther 2019; 19:39-51. [PMID: 31582531 DOI: 10.1158/1535-7163.mct-19-0222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/17/2019] [Accepted: 09/26/2019] [Indexed: 11/16/2022]
Abstract
Prostate cancer cells move from their primary site of origin, interact with a distant microenvironment, grow, and thereby cause death. It had heretofore not been possible to selectively inhibit cancer cell motility. Our group has recently shown that inhibition of intracellular activation of Raf1 with the small-molecule therapeutic KBU2046 permits, for the first time, selective inhibition of cell motility. We hypothesized that simultaneous disruption of multiple distinct functions that drive progression of prostate cancer to induce death would result in advanced disease control. Using a murine orthotopic implantation model of human prostate cancer metastasis, we demonstrate that combined treatment with KBU2046 and docetaxel retains docetaxel's antitumor action, but provides improved inhibition of metastasis, compared with monotherapy. KBU2046 does not interfere with hormone therapy, inclusive of enzalutamide-mediated inhibition of androgen receptor (AR) function and cell growth inhibition, and inclusive of the ability of castration to inhibit LNCaP-AR cell outgrowth in mice. Cell movement is necessary for osteoclast-mediated bone degradation. KBU2046 inhibits Raf1 and its downstream activation of MEK1/2 and ERK1/2 in osteoclasts, inhibiting cytoskeleton rearrangement, resorptive cavity formation, and bone destruction in vitro, with improved effects observed when the bone microenvironment is chemically modified by pretreatment with zoledronic acid. Using a murine cardiac injection model of human prostate cancer bone destruction quantified by CT, KBU2046 plus zoledronic exhibit improved inhibitory efficacy, compared with monotherapy. The combined disruption of pathways that drive cell movement, interaction with bone, and growth constitutes a multifunctional targeting strategy that provides advanced disease control.
Collapse
Affiliation(s)
- Limin Zhang
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Abhinandan Pattanayak
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Wenqi Li
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Hyun-Kyung Ko
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Graham Fowler
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Ryan Gordon
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Raymond Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
14
|
Kadayat TM, Park S, Shrestha A, Jo H, Hwang SY, Katila P, Shrestha R, Nepal MR, Noh K, Kim SK, Koh WS, Kim KS, Jeon YH, Jeong TC, Kwon Y, Lee ES. Discovery and Biological Evaluations of Halogenated 2,4-Diphenyl Indeno[1,2- b]pyridinol Derivatives as Potent Topoisomerase IIα-Targeted Chemotherapeutic Agents for Breast Cancer. J Med Chem 2019; 62:8194-8234. [PMID: 31398033 DOI: 10.1021/acs.jmedchem.9b00970] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the aim of developing new effective topoisomerase IIα-targeted anticancer agents, we synthesized a series of hydroxy- and halogenated 2,4-diphenyl indeno[1,2-b]pyridinols using a microwave-assisted single step synthetic method and investigated structure-activity relationships. The majority of compounds with chlorophenyl group at 2-position and phenol group at the 4-position of indeno[1,2-b]pyridinols exhibited potent antiproliferative activity and topoisomerase IIα-selective inhibition. Of the 172 compounds tested, 89 showed highly potent and selective topoisomerase IIα inhibition and antiproliferative activity in the nanomolar range against human T47D breast (2.6 nM) cancer cell lines. In addition, mechanistic studies revealed compound 89 is a nonintercalative topoisomerase II poison, and in vitro studies showed it had promising cytotoxic effects in diverse breast cancer cell lines and was particularly effective at inducing apoptosis in T47D cells. Furthermore, in vivo administration of compound 89 had significant antitumor effects in orthotopic mouse model of breast cancer.
Collapse
Affiliation(s)
- Tara Man Kadayat
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
- New Drug Development Center , Daegu-Gyeongbuk Medical Innovation Foundation , Daegu 41061 , Republic of Korea
| | - Seojeong Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 120-750 , Republic of Korea
| | - Aarajana Shrestha
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| | - Hyunji Jo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 120-750 , Republic of Korea
| | - Soo-Yeon Hwang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 120-750 , Republic of Korea
| | - Pramila Katila
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| | - Ritina Shrestha
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| | - Mahesh Raj Nepal
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| | - Keumhan Noh
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| | - Sang Kyoon Kim
- Laboratory Animal Center , Daegu-Gyeongbuk Medical Innovation Foundation , Daegu 41061 , Republic of Korea
| | - Woo-Suk Koh
- Laboratory Animal Center , Daegu-Gyeongbuk Medical Innovation Foundation , Daegu 41061 , Republic of Korea
| | - Kil Soo Kim
- Laboratory Animal Center , Daegu-Gyeongbuk Medical Innovation Foundation , Daegu 41061 , Republic of Korea
- College of Veterinary Medicine , Kyungpook National University , Daegu 41566 , Republic of Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center , Daegu-Gyeongbuk Medical Innovation Foundation , Daegu 41061 , Republic of Korea
| | - Tae Cheon Jeong
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 120-750 , Republic of Korea
| | - Eung-Seok Lee
- College of Pharmacy , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| |
Collapse
|
15
|
Mbaveng AT, Fotso GW, Ngnintedo D, Kuete V, Ngadjui BT, Keumedjio F, Andrae-Marobela K, Efferth T. Cytotoxicity of epunctanone and four other phytochemicals isolated from the medicinal plants Garcinia epunctata and Ptycholobium contortum towards multi-factorial drug resistant cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 48:112-119. [PMID: 30195869 DOI: 10.1016/j.phymed.2017.12.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/30/2017] [Accepted: 12/17/2017] [Indexed: 06/08/2023]
Abstract
INTRODUCTION Resistance of cancer cells is a serious impediment to chemotherapy and several phytochemicals are active against multi-drug resistant (MDR) phenotypes. The cytotoxicity of five naturally occurring compounds: betulin (1), mundulea lactone (2), seputhecarpan A (3), seputheisoflavone (4) and epunctanone (5) was evaluated on a panel of 9 cancer cell lines including various sensitive and drug-resistant cell lines. The modes of action of compound 5 were further investigated. METHODS The resazurin reduction assay was used to evaluate cytotoxicity of samples and ferroptotic cell death induced by compound 5; caspase-Glo assay was used to detect the activation of caspases in CCRF-CEM leukemia cells treated with compound 5. Flow cytometry was used for cell cycle analysis in CCRF-CEM cells treated with compound 5, as well as detection of apoptotic cells by annexin V/PI staining, analysis of mitochondrial membrane potential (MMP) and measurement of reactive oxygen species (ROS). RESULTS Compounds 1-5 displayed cytotoxic effects in the 9 studied cancer cell lines with IC50 values below 70 µM. The IC50 values varied from 8.20 µM (in HCT116 (p53-/-) colon cancer cells) to 35.10 µM (against HepG2 hepatocarcinoma cells) for 1, from 8.84 µM (in CEM/ADR5000 leukemia cells) to 48.99 µM (in MDA-MB-231 breast adenocarcinoma cells) for 2, from 12.17 µM (in CEM/ADR5000 cells) to 65.08 µM (in MDA-MB-231 cells) for 3, from 23.80 µM (in U87MG.ΔEGFR glioblastoma cells) to 68.66 µM (in HCT116 (p53-/-) cells) for 4, from 4.84 µM (in HCT116 (p53-/-) cells) to 13.12 µM (in HepG2 cells) for 5 and from 0.02 µM (against CCRF-CEM cells) to 122.96 µM (in CEM/ADR5000 cells) for doxorubicin. Compound 5 induced apoptosis in CCRF-CEM cells through alteration of MMP and increase in ROS production. In addition to apoptosis, ferroptosis was also identified as another mode of cell death induced by epunctanone. CONCLUSIONS Compounds 1-5 are valuable cytotoxic compounds that could be used to combat MDR cancer cells. Benzophenoe 5 is the most active molecule and deserve more investigations to develop new anticancer drugs.
Collapse
Affiliation(s)
- Armelle T Mbaveng
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz, Germany; Department of Biochemistry, Faculty of Science, University of Dschang, Cameroon
| | - Ghislain W Fotso
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Dominique Ngnintedo
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Victor Kuete
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz, Germany; Department of Biochemistry, Faculty of Science, University of Dschang, Cameroon
| | - Bonaventure T Ngadjui
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon; Department of Pharmacognosy and Pharmaceutical Sciences, Faculty of Medicine and Biomedical Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Felix Keumedjio
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Kerstin Andrae-Marobela
- Department of Biological Sciences, Faculty of Science, University of Botswana, Block 235, Private Bag, 0022 Gaborone, Botswana
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
16
|
Menezes JCJMDS, Edraki N, Kamat SP, Khoshneviszadeh M, Kayani Z, Mirzaei HH, Miri R, Erfani N, Nejati M, Cavaleiro JAS, Silva T, Saso L, Borges F, Firuzi O. Long Chain Alkyl Esters of Hydroxycinnamic Acids as Promising Anticancer Agents: Selective Induction of Apoptosis in Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7228-7239. [PMID: 28718636 DOI: 10.1021/acs.jafc.7b01388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cancer is the major cause of morbidity and mortality worldwide. Hydroxycinnamic acids (HCAs) are naturally occurring compounds and their alkyl esters may possess enhanced biological activities. We evaluated C4, C14, C16, and C18 alkyl esters of p-coumaric, ferulic, sinapic, and caffeic acids (19 compounds) for their cytotoxic activity against four human cancer cells and also examined their effect on cell cycle alteration and apoptosis induction. The tetradecyl (1c) and hexadecyl (1d) esters of p-coumaric acid and tetradecyl ester of caffeic acid (4c), but not the parental HCAs, were selectively effective against MOLT-4 (human lymphoblastic leukemia) cells with IC50 values of 0.123 ± 0.012, 0.301 ± 0.069 and 1.0 ± 0.1 μM, respectively. Compounds 1c, 1d, and 4c significantly increased apoptotic cells in sub-G1 phase and activated the caspase-3 enzyme in MOLT-4 cells. Compound 1c was 15.4 and 23.6 times more potent than doxorubicin and cisplatin, respectively, against the drug resistant MES-SA-DX5 uterine sarcoma cells. These p-coumarate esters were several times less effective against NIH/3T3 fibroblast cells. Docking studies showed that 1c may cause cytotoxicity by interaction with carbonic anhydrase IX. In conclusion, long chain alkyl esters of p-coumaric acid are promising scaffolds for selective apoptosis induction in cancer cells.
Collapse
Affiliation(s)
- José C J M D S Menezes
- Department of Chemistry & QOPNA, University of Aveiro , 3810-193 Aveiro, Portugal
- Department of Chemistry, Goa University , Taleigao 403 206 Goa India
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | | | - Mahsima Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - Zahra Kayani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - Hossein Hadavand Mirzaei
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
- Department of Molecular Physiology, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO) , Karaj, Iran
| | - Ramin Miri
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - Nasrollah Erfani
- Institute for Cancer Research (ICR), School of Medicine, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Maryam Nejati
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - José A S Cavaleiro
- Department of Chemistry & QOPNA, University of Aveiro , 3810-193 Aveiro, Portugal
| | - Tiago Silva
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , 4169-007 Porto, Portugal
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer″, Sapienza University of Rome , Italy
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , 4169-007 Porto, Portugal
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| |
Collapse
|
17
|
Hall S, Anoopkumar-Dukie S, Grant GD, Desbrow B, Lai R, Arora D, Hong Y. Modulation of chemotherapy-induced cytotoxicity in SH-SY5Y neuroblastoma cells by caffeine and chlorogenic acid. Toxicol Mech Methods 2017; 27:363-369. [DOI: 10.1080/15376516.2017.1296049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Susan Hall
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy, Griffith University, Gold Coast, QLD, Australia
- Quality Use of Medicines Network, Gold Coast, QLD, Australia
| | - Shailendra Anoopkumar-Dukie
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy, Griffith University, Gold Coast, QLD, Australia
- Quality Use of Medicines Network, Gold Coast, QLD, Australia
| | - Gary D. Grant
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy, Griffith University, Gold Coast, QLD, Australia
- Quality Use of Medicines Network, Gold Coast, QLD, Australia
| | - Ben Desbrow
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Allied Health Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Richard Lai
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy, Griffith University, Gold Coast, QLD, Australia
| | - Devinder Arora
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy, Griffith University, Gold Coast, QLD, Australia
- Quality Use of Medicines Network, Gold Coast, QLD, Australia
| | - Yinna Hong
- Guangdong Pharmaceutical University, Guangzhou City, Guangdong, People’s Republic of China
| |
Collapse
|
18
|
The effects of hyperthermic intraperitoneal chemoperfusion on colonic anastomosis: an experimental study in a rat model. TUMORI JOURNAL 2017; 103:307-313. [PMID: 28291907 DOI: 10.5301/tj.5000610] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2017] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Cytoreductive surgery (CRS) with subsequent hyperthermic intraperitoneal chemotherapy (HIPEC) is a promising modality to treat and prevent peritoneal metastases. However, this treatment is associated with signficant morbidity and mortality. Whether or not CRS with HIPEC interferes with anastomotic healing has also been debated. This study was designed to investigate the effects of mitomycin C, cisplatin, oxaliplatin, and doxorubicin used in HIPEC treatment on colonic anastomosis healing in a rat model. METHODS Sixty Wistar albino rats were employed in the study. Sigmoid resection and end-to-end colorectal anastomosis was performed in all rats. Group 1 rats underwent the surgical procedure alone, while group 2 rats were given hyperthermic intraperitoneal lavage with heated saline following surgery. Groups 3, 4, 5, and 6 had surgery with concomitant HIPEC treatment with mitomycin C, cisplatin, oxaliplatin, and doxorubicin, respectively. Anastomotic bursting pressures and hydroxyproline levels were evaluated. RESULTS Regarding the hydroxyproline levels, groups 1 and 2 showed significantly higher values than other groups (p<0.001). However, there was no significant difference between the HIPEC treatment groups (groups 3, 4, 5, and 6) (p>0.05). When groups were compared regarding bursting pressure values, no significant differences were observed (p = 0.81). CONCLUSIONS This study demonstrated that the HIPEC procedure with mitomycin C, cisplatin, oxaliplatin and doxorubicin had negative effects on hydroxyproline levels, but had no detrimental effect on anastomotic bursting pressure in a rat model.
Collapse
|
19
|
Laev SS, Salakhutdinov NF, Lavrik OI. Tyrosyl-DNA phosphodiesterase inhibitors: Progress and potential. Bioorg Med Chem 2016; 24:5017-5027. [PMID: 27687971 DOI: 10.1016/j.bmc.2016.09.045] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/30/2016] [Accepted: 09/18/2016] [Indexed: 10/21/2022]
Abstract
DNA topoisomerases are essential during transcription and replication. The therapeutic mechanism of action of topoisomerase inhibitors is enzyme poisoning rather than catalytic inhibition. Tyrosyl-DNA phosphodiesterases 1 or 2 were found as DNA repair enzymes hydrolyzing the covalent bond between the tyrosyl residue of topoisomerases I or II and the 3'- or 5'-phosphate groups in DNA, respectively. Tyrosyl-DNA phosphodiesterase 1 is a key enzyme in DNA repair machinery and a promising target for antitumor and neurodegenerative therapy. Inhibitors of tyrosyl-DNA phosphodiesterase 1 could act synergistically with topoisomerase I inhibitors and thereby potentiate the effects of topoisomerase I poisons. Tyrosyl-DNA phosphodiesterase 2 is an enzyme that specifically repairs DNA damages induced by topoisomerase II poisons and causes resistance to these drugs. Selective inhibition of tyrosyl-DNA phosphodiesterase 2 may be a novel approach to overcome intrinsic or acquired resistance to topoisomerase II-targeted drug therapy. Thus, agents that inhibit tyrosyl-DNA phosphodiesterases 1 and 2 have many applications in biochemical and physiological research and they have the potential to become anticancer and antiviral drugs. The structures, mechanism of action and therapeutic rationale of tyrosyl-DNA phosphodiesterase inhibitors and their development for combinations with topoisomerase inhibitors and DNA damaging agents are discussed.
Collapse
Affiliation(s)
- Sergey S Laev
- Vorozhtsov Institute of Organic Chemistry, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 9, Novosibirsk 630090, Russian Federation.
| | - Nariman F Salakhutdinov
- Vorozhtsov Institute of Organic Chemistry, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 9, Novosibirsk 630090, Russian Federation; Novosibirsk State University, Pirogova Str. 2, Novosibirsk 630090, Russian Federation
| | - Olga I Lavrik
- Novosibirsk State University, Pirogova Str. 2, Novosibirsk 630090, Russian Federation; Novosibirsk Institute of Chemical Biology and Fundamental Medicine, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 8, Novosibirsk 630090, Russian Federation
| |
Collapse
|