1
|
Alkhaldi O, Abusulieh S, Abusara OH, Sunoqrot S. Development of Mitoxantrone-Loaded Quercetin Nanoparticles for Breast Cancer Therapy with Potential for Synergism with Bioactive Natural Products. Int J Pharm 2024; 665:124674. [PMID: 39245083 DOI: 10.1016/j.ijpharm.2024.124674] [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: 04/16/2024] [Revised: 08/23/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Nanoparticle (NP)-based drug delivery systems have caused a paradigm shift in cancer treatment by enabling drug targeting, sustaining drug release, and reducing systemic toxicity of chemotherapy. Here we developed a novel NP formulation for the anticancer drug mitoxantrone (MTZ) by loading it into an emerging nanomaterial derived from the plant polyphenol quercetin (QCT). QCT was partially oxidized to produce amphiphilic oxQCT which was co-assembled with poly(ethylene glycol) (PEG) and MTZ by nanoprecipitation to form MTZ NPs. The optimal NPs exhibited an average diameter of 128 nm, a polydispersity index of 0.22, and a drug loading efficiency of 76%. While only a small fraction of the loaded drug was released at physiologic pH, a significantly higher fraction was released at acidic pH. The anticancer activity of MTZ NPs was assessed in MCF-7 and MDA-MB-231 breast cancer cell lines, alone and in combination with the bioactive natural products curcumin (CUR) and thymoquinone (TQ). In cell viability assays, MTZ NPs were slightly less potent than free MTZ, most likely due to their sustained release properties, but their cytotoxicity was greatly enhanced in the presence of TQ (in MCF-7 cells) as well as CUR (in MDA-MB-231 cells). The results were corroborated by apoptosis assays such as mitochondrial membrane potential measurement, acridine orange/ethidium bromide staining, in addition to caspase activity assays. The assays revealed that the NPs' proapoptotic effect was enhanced in the presence of CUR or TQ, depending on the cell line. Our work presents a promising nanocarrier platform for MTZ with the potential to enhance its bioactivity against breast cancer when combined with bioactive natural products.
Collapse
Affiliation(s)
- Otrujja Alkhaldi
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Samah Abusulieh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Osama H Abusara
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Suhair Sunoqrot
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan.
| |
Collapse
|
2
|
Dias-Carvalho A, Ferreira M, Reis-Mendes A, Ferreira R, de Lourdes Bastos M, Fernandes E, Sá SI, Capela JP, Carvalho F, Costa VM. Doxorubicin-induced neurotoxicity differently affects the hippocampal formation subregions in adult mice. Heliyon 2024; 10:e31608. [PMID: 38868005 PMCID: PMC11168325 DOI: 10.1016/j.heliyon.2024.e31608] [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: 06/24/2023] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Doxorubicin (DOX) is an anthracycline used to treat a wide range of tumours. Despite its effectiveness, it is associated with a long range of adverse effects, of which cognitive deficits stand out. The present study aimed to assess the neurologic adverse outcome pathways of two clinically relevant cumulative doses of DOX. Adult male CD-1 mice received biweekly intraperitoneal administrations for 3 weeks until reaching cumulative doses of 9 mg/kg (DOX9) or 18 mg/kg (DOX18). Animals were euthanized one week after the last administration, and biomarkers of oxidative stress and brain metabolism were evaluated in the whole brain. Coronal sections of fixed brains were used for specific determinations of the prefrontal cortex (PFC) and hippocampal formation (HF). In the whole brain, DOX18 tended to disrupt the antioxidant defences, affecting glutathione levels and manganese superoxide dismutase expression. Considering the regional analysis, DOX18 increased the volume of all brain areas evaluated, while GFAP-immunoreactive astrocytes decreased in the dentate gyrus (DG) and increased in the CA3 region of HF, both in a dose-dependent manner. Concerning the apoptosis pathway, whereas Bax increased in the DOX9 group, it decreased in the DOX18 group. Only in the latter group did Bcl-2 levels also decrease. While p53 only increased in the CA3 region of the DOX9 group, AIF increased in the PFC and DG of DOX18. Finally, phosphorylation of Tau decreased with the highest DOX dose in DG and CA3, while TNF-α levels increased in CA1 of DOX18. Our results indicate new pathways not yet described that could be responsible for the cognitive impairments observed in treated patients.
Collapse
Affiliation(s)
- Ana Dias-Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO–Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‐313, Porto, Portugal
| | - Mariana Ferreira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO–Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‐313, Porto, Portugal
- LAQV/REQUIMTE, Chemistry Department, University of Aveiro, Aveiro, Portugal
| | - Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO–Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‐313, Porto, Portugal
| | - Rita Ferreira
- LAQV/REQUIMTE, Chemistry Department, University of Aveiro, Aveiro, Portugal
| | - Maria de Lourdes Bastos
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO–Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‐313, Porto, Portugal
| | - Eduarda Fernandes
- LAQV/REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Susana Isabel Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Porto, Portugal
| | - João Paulo Capela
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO–Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‐313, Porto, Portugal
- FP-I3ID, Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Porto, 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, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‐313, Porto, 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, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‐313, Porto, Portugal
| |
Collapse
|
3
|
Mwema A, Muccioli GG, des Rieux A. Innovative drug delivery strategies to the CNS for the treatment of multiple sclerosis. J Control Release 2023; 364:435-457. [PMID: 37926243 DOI: 10.1016/j.jconrel.2023.10.052] [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: 07/05/2023] [Revised: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Disorders of the central nervous system (CNS), such as multiple sclerosis (MS) represent a great emotional, financial and social burden. Despite intense efforts, great unmet medical needs remain in that field. MS is an autoimmune, chronic inflammatory demyelinating disease with no curative treatment up to date. The current therapies mostly act in the periphery and seek to modulate aberrant immune responses as well as slow down the progression of the disease. Some of these therapies are associated with adverse effects related partly to their administration route and show some limitations due to their rapid clearance and inability to reach the CNS. The scientific community have recently focused their research on developing MS therapies targeting different processes within the CNS. However, delivery of therapeutics to the CNS is mainly limited by the presence of the blood-brain barrier (BBB). Therefore, there is a pressing need to develop new drug delivery strategies that ensure CNS availability to capitalize on identified therapeutic targets. Several approaches have been developed to overcome or bypass the BBB and increase delivery of therapeutics to the CNS. Among these strategies, the use of alternative routes of administration, such as the nose-to-brain (N2B) pathway, offers a promising non-invasive option in the scope of MS, as it would allow a direct transport of the drugs from the nasal cavity to the brain. Moreover, the combination of bioactive molecules within nanocarriers bring forth new opportunities for MS therapies, allowing and/or increasing their transport to the CNS. Here we will review and discuss these alternative administration routes as well as the nanocarrier approaches useful to deliver drugs for MS.
Collapse
Affiliation(s)
- Ariane Mwema
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium; Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium.
| | - Anne des Rieux
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium.
| |
Collapse
|
4
|
Brandão SR, Reis-Mendes A, Neuparth MJ, Carvalho F, Ferreira R, Costa VM. The Metabolic Fingerprint of Doxorubicin-Induced Cardiotoxicity in Male CD-1 Mice Fades Away with Time While Autophagy Increases. Pharmaceuticals (Basel) 2023; 16:1613. [PMID: 38004479 PMCID: PMC10675798 DOI: 10.3390/ph16111613] [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: 10/02/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
The cardiotoxicity of doxorubicin (DOX) may manifest at the beginning/during treatment or years after, compromising patients' quality of life. We intended to study the cardiac pathways one week (short-term, control 1 [CTRL1] and DOX1 groups) or five months (long-term, CTRL2 and DOX2 groups) after DOX administration in adult male CD-1 mice. Control groups were given saline, and DOX groups received a 9.0 mg/Kg cumulative dose. In the short-term, DOX decreased the content of AMP-activated protein kinase (AMPK) while the electron transfer flavoprotein-ubiquinone oxidoreductase (ETF-QO) increased compared to CTRL1, suggesting the upregulation of fatty acids oxidation. Moreover, mitofusin1 (Mfn1) content was decreased in DOX1, highlighting decreased mitochondrial fusion. In addition, increased B-cell lymphoma-2 associated X-protein (BAX) content in DOX1 pointed to the upregulation of apoptosis. Conversely, in the long-term, DOX decreased the citrate synthase (CS) activity and the content of Beclin1 and autophagy protein 5 (ATG5) compared to CTRL2, suggesting decreased mitochondrial density and autophagy. Our study demonstrates that molecular mechanisms elicited by DOX are modulated at different extents over time, supporting the differences on clinic cardiotoxic manifestations with time. Moreover, even five months after DOX administration, meaningful heart molecular changes occurred, reinforcing the need for the continuous cardiac monitoring of patients and determination of earlier biomarkers before clinical cardiotoxicity is set.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.B.); (A.R.-M.); (F.C.)
- UCIBIO-REQUIMTE, Laboratory of Toxicology, 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; (S.R.B.); (A.R.-M.); (F.C.)
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, 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
| | - Félix Carvalho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.B.); (A.R.-M.); (F.C.)
- UCIBIO-REQUIMTE, Laboratory of Toxicology, 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; (S.R.B.); (A.R.-M.); (F.C.)
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
5
|
Kim H, Jung I, Lee CH, An J, Ko M. Development of Novel Epigenetic Anti-Cancer Therapy Targeting TET Proteins. Int J Mol Sci 2023; 24:16375. [PMID: 38003566 PMCID: PMC10671484 DOI: 10.3390/ijms242216375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Epigenetic dysregulation, particularly alterations in DNA methylation and hydroxymethylation, plays a pivotal role in cancer initiation and progression. Ten-eleven translocation (TET) proteins catalyze the successive oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) and further oxidized methylcytosines in DNA, thereby serving as central modulators of DNA methylation-demethylation dynamics. TET loss of function is causally related to neoplastic transformation across various cell types while its genetic or pharmacological activation exhibits anti-cancer effects, making TET proteins promising targets for epigenetic cancer therapy. Here, we developed a robust cell-based screening system to identify novel TET activators and evaluated their potential as anti-cancer agents. Using a carefully curated library of 4533 compounds provided by the National Cancer Institute, Bethesda, MD, USA, we identified mitoxantrone as a potent TET agonist. Through rigorous validation employing various assays, including immunohistochemistry and dot blot studies, we demonstrated that mitoxantrone significantly elevated 5hmC levels. Notably, this elevation manifested only in wild-type (WT) but not TET-deficient mouse embryonic fibroblasts, primary bone marrow-derived macrophages, and leukemia cell lines. Furthermore, mitoxantrone-induced cell death in leukemia cell lines occurred in a TET-dependent manner, indicating the critical role of TET proteins in mediating its anti-cancer effects. Our findings highlight mitoxantrone's potential to induce tumor cell death via a novel mechanism involving the restoration of TET activity, paving the way for targeted epigenetic therapies in cancer treatment.
Collapse
Affiliation(s)
- Hyejin Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea; (H.K.); (I.J.)
| | - Inkyung Jung
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea; (H.K.); (I.J.)
| | - Chan Hyeong Lee
- Department of Life Sciences, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Jungeun An
- Department of Life Sciences, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Myunggon Ko
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea; (H.K.); (I.J.)
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| |
Collapse
|
6
|
van Gelder M, van der Zanden SY, Vriends MBL, Wagensveld RA, van der Marel GA, Codée JDC, Overkleeft HS, Wander DPA, Neefjes JJC. Re-Exploring the Anthracycline Chemical Space for Better Anti-Cancer Compounds. J Med Chem 2023; 66:11390-11398. [PMID: 37561481 PMCID: PMC10461226 DOI: 10.1021/acs.jmedchem.3c00853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Indexed: 08/11/2023]
Abstract
The anthracycline anti-cancer drugs are intensely used in the clinic to treat a wide variety of cancers. They generate DNA double strand breaks, but recently the induction of chromatin damage was introduced as another major determinant of anti-cancer activity. The combination of these two events results in their reported side effects. While our knowledge on the structure-activity relationship of anthracyclines has improved, many structural variations remain poorly explored. Therefore, we here report on the preparation of a diverse set of anthracyclines with variations within the sugar moiety, amine alkylation pattern, saccharide chain and aglycone. We assessed the cytotoxicity in vitro in relevant human cancer cell lines, and the capacity to induce DNA- and chromatin damage. This coherent set of data allowed us to deduce a few guidelines on anthracycline design, as well as discover novel, highly potent anthracyclines that may be better tolerated by patients.
Collapse
Affiliation(s)
- Merle
A. van Gelder
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Sabina Y. van der Zanden
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Merijn B. L. Vriends
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Roos A. Wagensveld
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | | | - Jeroen D. C. Codée
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Herman S. Overkleeft
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Dennis P. A. Wander
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Jacques J. C. Neefjes
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
7
|
Dias-Carvalho A, Margarida-Araújo A, Reis-Mendes A, Sequeira CO, Pereira SA, Guedes de Pinho P, Carvalho F, Sá SI, Fernandes E, Costa VM. A Clinically Relevant Dosage of Mitoxantrone Disrupts the Glutathione and Lipid Metabolic Pathways of the CD-1 Mice Brain: A Metabolomics Study. Int J Mol Sci 2023; 24:13126. [PMID: 37685929 PMCID: PMC10488007 DOI: 10.3390/ijms241713126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/26/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023] Open
Abstract
Long-term cognitive dysfunction, or "chemobrain", has been observed in cancer patients treated with chemotherapy. Mitoxantrone (MTX) is a topoisomerase II inhibitor that binds and intercalates with DNA, being used in the treatment of several cancers and multiple sclerosis. Although MTX can induce chemobrain, its neurotoxic mechanisms are poorly studied. This work aimed to identify the adverse outcome pathways (AOPs) activated in the brain upon the use of a clinically relevant cumulative dose of MTX. Three-month-old male CD-1 mice were given a biweekly intraperitoneal administration of MTX over the course of three weeks until reaching a total cumulative dose of 6 mg/kg. Controls were given sterile saline in the same schedule. Two weeks after the last administration, the mice were euthanized and their brains removed. The left brain hemisphere was used for targeted profiling of the metabolism of glutathione and the right hemisphere for an untargeted metabolomics approach. The obtained results revealed that MTX treatment reduced the availability of cysteine (Cys), cysteinylglycine (CysGly), and reduced glutathione (GSH) suggesting that MTX disrupts glutathione metabolism. The untargeted approach revealed metabolic circuits of phosphatidylethanolamine, catecholamines, unsaturated fatty acids biosynthesis, and glycerolipids as relevant players in AOPs of MTX in our in vivo model. As far as we know, our study was the first to perform such a broad profiling study on pathways that could put patients given MTX at risk of cognitive deficits.
Collapse
Affiliation(s)
- Ana Dias-Carvalho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana Margarida-Araújo
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana Reis-Mendes
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Catarina Oliveira Sequeira
- iNOVA4Health, LS4Future, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Sofia Azeredo Pereira
- iNOVA4Health, LS4Future, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, 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, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Susana Isabel Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Eduarda Fernandes
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, 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, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
8
|
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
|
9
|
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: 8] [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
|
10
|
Gaikwad M, Konkimalla VB, Salunke-Gawali S. Metal complexes as topoisomerase inhibitors. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.121089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
11
|
Dias-Carvalho A, Ferreira M, Reis-Mendes A, Ferreira R, Bastos ML, Fernandes E, Sá SI, Capela JP, Carvalho F, Costa VM. Chemobrain: mitoxantrone-induced oxidative stress, apoptotic and autophagic neuronal death in adult CD-1 mice. Arch Toxicol 2022; 96:1767-1782. [PMID: 35306571 DOI: 10.1007/s00204-022-03261-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/17/2022] [Indexed: 12/17/2022]
|
12
|
Reis-Mendes A, Padrão AI, Duarte JA, Gonçalves-Monteiro S, Duarte-Araújo M, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. Role of Inflammation and Redox Status on Doxorubicin-Induced Cardiotoxicity in Infant and Adult CD-1 Male Mice. Biomolecules 2021; 11:1725. [PMID: 34827723 PMCID: PMC8615472 DOI: 10.3390/biom11111725] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/21/2022] Open
Abstract
Doxorubicin (DOX) is a topoisomerase II inhibitor commonly used in the treatment of several types of cancer. Despite its efficacy, DOX can potentially cause fatal adverse effects, like cardiotoxicity. This work aimed to assess the role of inflammation in DOX-treated infant and adult mice and its possible link to underlying cardiotoxicity. Two groups of CD-1 male mice of different ages (infants or adults) were subjected to biweekly DOX administrations, to reach a cumulative dose of 18.0 mg/kg, which corresponds approximately in humans to 100.6 mg/m2 for infants and 108.9 mg/m2 for adults a clinically relevant dose in humans. The classic plasmatic markers of cardiotoxicity increased, and that damage was confirmed by histopathological findings in both groups, although it was higher in adults. Moreover, in DOX-treated adults, an increase of cardiac fibrosis was observed, which was accompanied by an increase in specific inflammatory parameters, namely, macrophage M1 and nuclear factor kappa B (NF-κB) p65 subunit, with a trend toward increased levels of the tumor necrosis factor receptor 2 (TNFR2). On the other hand, the levels of myeloperoxidase (MPO) and interleukin (IL)-6 significantly decreased in DOX-treated adult animals. In infants, a significant increase in cardiac protein carbonylation and in the levels of nuclear factor erythroid-2 related factor 2 (Nrf2) was observed. In both groups, no differences were found in the levels of tumor necrosis factor (TNF-α), IL-1β, p38 mitogen-activated protein kinase (p38 MAPK) or NF-κB p52 subunit. In conclusion, using a clinically relevant dose of DOX, our study demonstrated that cardiac effects are associated not only with the intensity of the inflammatory response but also with redox response. Adult mice seemed to be more prone to DOX-induced cardiotoxicity by mechanisms related to inflammation, while infant mice seem to be protected from the damage caused by DOX, possibly by activating such antioxidant defenses as Nrf2.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.R.); (F.C.); (M.L.B.)
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana Isabel Padrão
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal; (A.I.P.); (J.A.D.)
| | - José Alberto Duarte
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal; (A.I.P.); (J.A.D.)
- TOXRUN–Toxicology Research Unit, University Institute of Health Sciences, Advanced Polytechnic and University Cooperative (CESPU), CRL, 4585-116 Gandra, Portugal
| | - Salomé Gonçalves-Monteiro
- Outcomes Research Laboratory, MOREHealth, Outcomes Research Laboratory, Portuguese Institute of Oncology at Porto Francisco Gentil (IPO Porto), 4200-072 Porto, Portugal;
| | - Margarida Duarte-Araújo
- Department of Immuno-Physiology and Pharmacology, ICBAS—Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal;
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.R.); (F.C.); (M.L.B.)
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.R.); (F.C.); (M.L.B.)
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- CIIMAR–Interdisciplinary Centre of Marine and Environmental Research, 4450-208 Porto, Portugal
| | - Maria Lourdes Bastos
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.R.); (F.C.); (M.L.B.)
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.R.); (F.C.); (M.L.B.)
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
13
|
Dias-Carvalho A, Ferreira M, Ferreira R, Bastos MDL, Sá SI, Capela JP, Carvalho F, Costa VM. Four decades of chemotherapy-induced cognitive dysfunction: comprehensive review of clinical, animal and in vitro studies, and insights of key initiating events. Arch Toxicol 2021; 96:11-78. [PMID: 34725718 DOI: 10.1007/s00204-021-03171-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/23/2021] [Indexed: 01/22/2023]
Abstract
Cognitive dysfunction has been one of the most reported and studied adverse effects of cancer treatment, but, for many years, it was overlooked by the medical community. Nevertheless, the medical and scientific communities have now recognized that the cognitive deficits caused by chemotherapy have a strong impact on the morbidity of cancer treated patients. In fact, chemotherapy-induced cognitive dysfunction or 'chemobrain' (also named also chemofog) is at present a well-recognized effect of chemotherapy that could affect up to 78% of treated patients. Nonetheless, its underlying neurotoxic mechanism is still not fully elucidated. Therefore, this work aimed to provide a comprehensive review using PubMed as a database to assess the studies published on the field and, therefore, highlight the clinical manifestations of chemobrain and the putative neurotoxicity mechanisms.In the last two decades, a great number of papers was published on the topic, mainly with clinical observations. Chemotherapy-treated patients showed that the cognitive domains most often impaired were verbal memory, psychomotor function, visual memory, visuospatial and verbal learning, memory function and attention. Chemotherapy alters the brain's metabolism, white and grey matter and functional connectivity of brain areas. Several mechanisms have been proposed to cause chemobrain but increase of proinflammatory cytokines with oxidative stress seem more relevant, not excluding the action on neurotransmission and cellular death or impaired hippocampal neurogenesis. The interplay between these mechanisms and susceptible factors makes the clinical management of chemobrain even more difficult. New studies, mainly referring to the underlying mechanisms of chemobrain and protective measures, are important in the future, as it is expected that chemobrain will have more clinical impact in the coming years, since the number of cancer survivors is steadily increasing.
Collapse
Affiliation(s)
- Ana Dias-Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal. .,UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - Mariana Ferreira
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rita Ferreira
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Maria de Lourdes Bastos
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Susana Isabel Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Porto, Portugal
| | - João Paulo Capela
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal. .,UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
14
|
Inhibition of the DSB repair protein RAD51 potentiates the cytotoxic efficacy of doxorubicin via promoting apoptosis-related death pathways. Cancer Lett 2021; 520:361-373. [PMID: 34389435 DOI: 10.1016/j.canlet.2021.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 01/04/2023]
Abstract
The anthracycline derivative doxorubicin (Doxo) induces DNA double-strand breaks (DSBs) by inhibition of DNA topoisomerase type II. Defective mismatch repair (MMR) contributes to Doxo resistance and has been reported for colon and mammary carcinomas. Here, we investigated the outcome of pharmacological inhibition of various DNA repair-related mechanisms on Doxo-induced cytotoxicity employing MMR-deficient HCT-116 colon carcinoma cells. Out of different inhibitors tested (i.e. HDACi, PARPi, MRE11i, RAD52i, RAD51i), we identified the RAD51-inhibitor B02 as the most powerful compound to synergistically increase Doxo-induced cytotoxicity. B02-mediated synergism rests on pleiotropic mechanisms, including pronounced G2/M arrest, damage to mitochondria and caspase-driven apoptosis. Of note, B02 also promotes the cytotoxicity of oxaliplatin and 5-fluoruracil (5-FU) in HCT-116 cells and, furthermore, also increases Doxo-induced cytotoxicity in MMR-proficient colon and mammary carcinoma cells. Summarizing, pharmacological inhibition of RAD51 is suggested to synergistically increase the cytotoxic efficacy of various types of conventional anticancer drugs in different tumor entities. Hence, pre-clinical in vivo studies are preferable to determine the therapeutic window of B02 in a clinically oriented therapeutic regimen.
Collapse
|
15
|
Kaur M, Mehta V, Arora S, Munshi A, Singh S, Kumar R. Design, Synthesis and Biological Evaluation of New 5‐(2‐Nitrophenyl)‐1‐aryl‐1
H
‐pyrazoles as Topoisomerase Inhibitors. ChemistrySelect 2021. [DOI: 10.1002/slct.202101459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Manpreet Kaur
- Laboratory for Drug Design and Synthesis Department of Pharmaceutical Sciences and Natural Products School of Pharmaceutical Sciences Central University of Punjab Ghudda Bathinda 151401 India
| | - Vikrant Mehta
- Department of Human Genetics and Molecular Medicine Central University of Punjab Ghudda Bathinda 151401 India
| | - Sahil Arora
- Laboratory for Drug Design and Synthesis Department of Pharmaceutical Sciences and Natural Products School of Pharmaceutical Sciences Central University of Punjab Ghudda Bathinda 151401 India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine Central University of Punjab Ghudda Bathinda 151401 India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine Central University of Punjab Ghudda Bathinda 151401 India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis Department of Pharmaceutical Sciences and Natural Products School of Pharmaceutical Sciences Central University of Punjab Ghudda Bathinda 151401 India
| |
Collapse
|
16
|
Reis-Mendes A, Dores-Sousa JL, Padrão AI, Duarte-Araújo M, Duarte JA, Seabra V, Gonçalves-Monteiro S, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. Inflammation as a Possible Trigger for Mitoxantrone-Induced Cardiotoxicity: An In Vivo Study in Adult and Infant Mice. Pharmaceuticals (Basel) 2021; 14:ph14060510. [PMID: 34073506 PMCID: PMC8229902 DOI: 10.3390/ph14060510] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023] Open
Abstract
Mitoxantrone (MTX) is a pharmaceutical drug used in the treatment of several cancers and refractory multiple sclerosis (MS). Despite its therapeutic value, adverse effects may be severe, namely the frequently reported cardiotoxicity, whose mechanisms need further research. This work aimed to assess if inflammation or oxidative stress-related pathways participate in the cardiotoxicity of MTX, using the mouse as an animal model, at two different age periods (infant or adult mice) using two therapeutic relevant cumulative doses. Histopathology findings showed that MTX caused higher cardiac toxicity in adults. In MTX-treated adults, at the highest dose, noradrenaline cardiac levels decreased, whereas at the lowest cumulative dose, protein carbonylation increased and the expression of nuclear factor kappa B (NF-κB) p65 subunit and of M1 macrophage marker increased. Moreover, MTX-treated adult mice had enhanced expression of NF-κB p52 and tumour necrosis factor (TNF-α), while decreasing interleukin-6 (IL-6). Moreover, while catalase expression significantly increased in both adult and infant mice treated with the lowest MTX cumulative dose, the expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and glutathione peroxidase only significantly increased in infant animals. Nevertheless, the ratio of GAPDH to ATP synthase subunit beta decreased in adult animals. In conclusion, clinically relevant doses of MTX caused dissimilar responses in adult and infant mice, being that inflammation may be an important trigger to MTX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.L.D.-S.); (F.R.); (F.C.); (M.L.B.)
- Correspondence: (A.R.-M.); (V.M.C.); Tel.: +351-220-428-599 (A.R.-M.); +351-220-428-599 (V.M.C.)
| | - José Luís Dores-Sousa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.L.D.-S.); (F.R.); (F.C.); (M.L.B.)
| | - Ana Isabel Padrão
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal; (A.I.P.); (J.A.D.)
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, University of Porto, 4050-313 Porto, Portugal;
- Department of Immune-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - José Alberto Duarte
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal; (A.I.P.); (J.A.D.)
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, University Institute of Health Sciences (IUCS), CESPU, 4585-116 Paredes, Portugal;
| | - Vítor Seabra
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, University Institute of Health Sciences (IUCS), CESPU, 4585-116 Paredes, Portugal;
| | - Salomé Gonçalves-Monteiro
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- MOREHealth, Outcomes Research Lab, Portuguese Institute of Oncology at Porto Francisco Gentil (IPO Porto), 4200-072 Porto, Portugal
| | - Fernando Remião
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.L.D.-S.); (F.R.); (F.C.); (M.L.B.)
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.L.D.-S.); (F.R.); (F.C.); (M.L.B.)
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- CIIMAR—Interdisciplinary Centre of Marine and Environmental Research, 4450-208 Porto, Portugal
| | - Maria Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.L.D.-S.); (F.R.); (F.C.); (M.L.B.)
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.L.D.-S.); (F.R.); (F.C.); (M.L.B.)
- Correspondence: (A.R.-M.); (V.M.C.); Tel.: +351-220-428-599 (A.R.-M.); +351-220-428-599 (V.M.C.)
| |
Collapse
|
17
|
Inhibition of histone deacetylases, topoisomerases and epidermal growth factor receptor by metal-based anticancer agents: Design & synthetic strategies and their medicinal attributes. Bioorg Chem 2020; 105:104396. [PMID: 33130345 DOI: 10.1016/j.bioorg.2020.104396] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 12/22/2022]
Abstract
Metal-based inhibitors of histone deacetylases (HDAC), DNA topoisomerases (Topos) and Epidermal Growth Factor Receptor (EGFR) have demonstrated their cytotoxic potential against various cancer types such as breast, lung, uterus, colon, etc. Additionally, these have proven their role in resolving the resistance issues, enhancing the affinity, lipophilicity, stability, and biocompatibility and therefore, emerged as potential candidates for molecularly targeted therapeutics. This review focusses on nature and role of metals and organic ligands in tuning the anticancer activity in multiple modes of inhibition considering HDACs, Topos or EGFR as one of the primary targets. The conceptual design and synthetic approaches of platinum and non-platinum metal complexes comprising of chiefly ruthenium, rhodium, palladium, copper, iron, nickel, cobalt, zinc metals coordinated with organic scaffolds, along with their biological activity profiles, structure-activity relationships (SARs), docking studies, possible modes of action, and their scope and limitations are discussed in detail.
Collapse
|
18
|
Costa VM, Capela JP, Sousa JR, Eleutério RP, Rodrigues PRS, Dores-Sousa JL, Carvalho RA, Lourdes Bastos M, Duarte JA, Remião F, Almeida MG, Varner KJ, Carvalho F. Mitoxantrone impairs proteasome activity and prompts early energetic and proteomic changes in HL-1 cardiomyocytes at clinically relevant concentrations. Arch Toxicol 2020; 94:4067-4084. [DOI: 10.1007/s00204-020-02874-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/12/2020] [Indexed: 11/24/2022]
|
19
|
Wan Z, Sun R, Moharil P, Chen J, Liu Y, Song X, Ao Q. Research advances in nanomedicine, immunotherapy, and combination therapy for leukemia. J Leukoc Biol 2020; 109:425-436. [PMID: 33259068 DOI: 10.1002/jlb.5mr0620-063rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/29/2020] [Accepted: 07/12/2020] [Indexed: 12/17/2022] Open
Abstract
In the past decade, clinical and laboratory studies have led to important new insights into the biology of leukemia and its treatment. This review describes the progress of leukemia research in the United States in recent years. Whereas the traditional method of treatment is chemotherapy, it is nonselective and could induce systemic toxicities. Thus, in parallel with research on new chemotherapies, great emphasis has been placed on developing immunotherapies. Here, we will review the current immunotherapies available in research and development that overcome current challenges, specifically looking in the field of chimeric antigen receptor T-cell (CAR-T) therapies, checkpoint inhibitors, and antibody-drug conjugates. With about 100 clinical trials for CAR-T therapies and 30 in checkpoint inhibitors for leukemia treatment, scientists are trying to make these technologies cheaper, faster, and more feasible. Further describing the delivery of these therapeutics, we look at the current progress, clinical, and preclinical status of nano-based medicines such as liposomes, polymeric micelles, and metal nanoparticles. Taking advantage of their physicochemical and biologic properties, nanoparticles have been shown to increase the efficacy of commonly administered chemotherapies with reduced adverse effects.
Collapse
Affiliation(s)
- Zhuoya Wan
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Runzi Sun
- Department of Immunology, School of Medicine, University of Pittsburgh, Pennsylvania, USA
| | - Pearl Moharil
- Department of Cell Biology, Harvard Medical School, Harvard University, Massachusetts, USA.,Department of Pharmaceutical Science, School of Pharmacy, University of Pittsburgh, Pennsylvania, USA
| | - Jing Chen
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.,Department of Pharmaceutical Science, School of Pharmacy, University of Pittsburgh, Pennsylvania, USA
| | - Yuzhe Liu
- Department of Materials Engineering, Purdue University, Indiana, USA
| | - Xu Song
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Qiang Ao
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Guloglu S, Kirmaci FN, Çetinkol ÖP, Forough M, Gulkaya A. Azacyanines as Novel Topoisomerase II Alpha Inhibitors. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190628161945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Introduction:
Topoisomerase II alpha (Topo IIα) has become one of the extensively exploited
targets in chemotherapy due to its role in regulating the topological constraints of DNA during
replication and transcription. Small molecules targeting Topo IIα’s activity such as etoposide
(VP-16) and doxorubicin are extensively used in the treatment of many different types of cancer.
Objective:
Here, the effects of three small molecules, named as azacyanines, on Topo IIα have been
assessed.
Methods:
In-vitro Topoisomerase IIα drug screening kit and agarose gel imaging were used for the
assessment of Topo IIα’s activity.
Results:
Our results revealed that all the azacyanines investigated decreased the catalytic activity of
Topo IIα dramatically. More importantly, the decrease in the catalytic activity of Topo IIα in the
presence of azacyanines was higher than the presence of VP-16, which is a commercially available
chemotherapy drug. Upon further investigation, it has been observed that Azamethyl’s catalytic inhibition
of Topo IIα was concentration dependent and the catalytic activity of Topo IIα was almost
completely abolished in the presence of 100.0 μM of Azamethyl.
Conclusion:
These findings reveal the potential of azacyanines as effective Topo IIα inhibitors and
chemotherapeutic agents.
Collapse
Affiliation(s)
- Sercan Guloglu
- Biochemistry Program, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Fahriye Nur Kirmaci
- Biochemistry Program, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Özgül Persil Çetinkol
- Biochemistry Program, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Mehrdad Forough
- Department of Chemistry, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Aybuke Gulkaya
- Department of Chemistry, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| |
Collapse
|
21
|
A Precise Nanostructure of Folate-Overhung Mitoxantrone DNA Tetrahedron for Targeted Capture Leukemia. NANOMATERIALS 2020; 10:nano10050951. [PMID: 32429472 PMCID: PMC7279534 DOI: 10.3390/nano10050951] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/28/2020] [Accepted: 05/14/2020] [Indexed: 12/31/2022]
Abstract
Regular chemotherapy cannot eliminate leukemic cells, due to the sparse distribution of cancer cells in leukemia patients. Here, we report a precise nanostructure of folate-overhung mitoxantrone DNA tetrahedron that enables the treatment of leukemic cells by targeted action. Folate is used as a targeting molecule and synthesized with DNA strand in forming the folate-overhang DNA complement, and the complement is then separately base-paired onto six sides of the fabricated DNA tetrahedron. Mitoxantrone is used as an anticancer agent and intercalated into the double strands of the folate-overhung DNA tetrahedron for drug loading. The evaluation studies are performed on leukemia BALL-1 and K562 cells. The results demonstrate that the folate-overhung mitoxantrone DNA tetrahedra (approximately 25 nm) are able to target leukemic cells, transport across the nuclei membrane, induce the apoptosis, and enhance the overall efficacy of treating leukemic cells in vitro and in leukemia-bearing mice. This study provides a potential drug-containing DNA nanostructure, to clean the sparsely distributed leukemic cells in patients.
Collapse
|
22
|
Filipczak N, Jaromin A, Piwoni A, Mahmud M, Sarisozen C, Torchilin V, Gubernator J. A Triple Co-Delivery Liposomal Carrier That Enhances Apoptosis via an Intrinsic Pathway in Melanoma Cells. Cancers (Basel) 2019; 11:cancers11121982. [PMID: 31835393 PMCID: PMC6966600 DOI: 10.3390/cancers11121982] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 11/29/2022] Open
Abstract
The effectiveness of existing anti-cancer therapies is based mainly on the stimulation of apoptosis of cancer cells. Most of the existing therapies are somewhat toxic to normal cells. Therefore, the quest for nontoxic, cancer-specific therapies remains. We have demonstrated the ability of liposomes containing anacardic acid, mitoxantrone and ammonium ascorbate to induce the mitochondrial pathway of apoptosis via reactive oxygen species (ROS) production by the killing of cancer cells in monolayer culture and shown its specificity towards melanoma cells. Liposomes were prepared by a lipid hydration, freeze-and-thaw (FAT) procedure and extrusion through polycarbonate filters, a remote loading method was used for dug encapsulation. Following characterization, hemolytic activity, cytotoxicity and apoptosis inducing effects of loaded nanoparticles were investigated. To identify the anticancer activity mechanism of these liposomes, ROS level and caspase 9 activity were measured by fluorescence and by chemiluminescence respectively. We have demonstrated that the developed liposomal formulations produced a high ROS level, enhanced apoptosis and cell death in melanoma cells, but not in normal cells. The proposed mechanism of the cytotoxic action of these liposomes involved specific generation of free radicals by the iron ions mechanism.
Collapse
Affiliation(s)
- Nina Filipczak
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.J.); (A.P.); (M.M.); (J.G.)
- Correspondence: or ; Tel.: +48-713-756-318
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.J.); (A.P.); (M.M.); (J.G.)
| | - Adriana Piwoni
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.J.); (A.P.); (M.M.); (J.G.)
| | - Mohamed Mahmud
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.J.); (A.P.); (M.M.); (J.G.)
- Department of Food Science and Technology, Faculty of Agriculture, University of Misurata, Misurata 2478, Libya
| | - Can Sarisozen
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; (C.S.); (V.T.)
| | - Vladimir Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; (C.S.); (V.T.)
- Department of Oncology, Radiotherapy and Plastic Surgery I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.J.); (A.P.); (M.M.); (J.G.)
| |
Collapse
|
23
|
Johnson AD, Zammit R, Vella J, Valentino M, Buhagiar JA, Magri DC. Aminonaphthalimide hybrids of mitoxantrone and amonafide as anticancer and fluorescent cellular imaging agents. Bioorg Chem 2019; 93:103287. [PMID: 31561011 DOI: 10.1016/j.bioorg.2019.103287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/28/2019] [Accepted: 09/15/2019] [Indexed: 01/10/2023]
Abstract
Novel water-soluble 4-aminonaphthalimides were synthesised and their cellular fluorescent imaging, cytotoxicity and ability to induced apoptosis evaluated. The lead compound 1 was designed from the cross-fertilisation of the basic hydrophilic amino pharmacophore of mitoxantrone, and an aminonaphthalimide scaffold of the drug candidate, amonafide. The compounds are also fluorescent pH probes based on photoinduced electron transfer (PET) and internal charge transfer (ICT). The compounds are sensitive to solvent polarity with large Stoke shifts (>90 nm) and provide emissive-coloured solutions (blue to yellow). Excited state pKas of 9.0-9.3 and fluorescence quantum yields of 0.47-0.58 were determined in water. The cytotoxicity and cellular fluorescent imaging properties of the compounds were tested on human cancer cell lines K562 and MCF-7 by the MTT assay, phase contrast and fluorescence microscopy. Compounds 1 and 3 with flexible aminoalkyl chains exhibited GI50 comparable to amonafide, while 2 and 4 with a rigid piperazine moiety and butyl chain are less cytotoxic. Fluorescence microscopy with 1 allowed for the visualization of the intracellular microenvironment exemplifying the potential utility of such hybrid molecules as anticancer and fluorescent cellular imaging agents.
Collapse
Affiliation(s)
- Alex D Johnson
- Department of Chemistry, Faculty of Science, University of Malta, Msida, MSD 2080, Malta
| | - Rodrianne Zammit
- Department of Biology, Faculty of Science, University of Malta, Msida, MSD 2080, Malta
| | - Jasmine Vella
- Department of Physiology and Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD 2080, Malta
| | - Mario Valentino
- Department of Physiology and Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD 2080, Malta
| | - Joseph A Buhagiar
- Department of Biology, Faculty of Science, University of Malta, Msida, MSD 2080, Malta
| | - David C Magri
- Department of Chemistry, Faculty of Science, University of Malta, Msida, MSD 2080, Malta.
| |
Collapse
|
24
|
Chen KTJ, Gilabert-Oriol R, Bally MB, Leung AWY. Recent Treatment Advances and the Role of Nanotechnology, Combination Products, and Immunotherapy in Changing the Therapeutic Landscape of Acute Myeloid Leukemia. Pharm Res 2019; 36:125. [PMID: 31236772 PMCID: PMC6591181 DOI: 10.1007/s11095-019-2654-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 06/01/2019] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia that is becoming more prevalent particularly in the older (65 years of age or older) population. For decades, "7 + 3" remission induction therapy with cytarabine and an anthracycline, followed by consolidation therapy, has been the standard of care treatment for AML. This stagnancy in AML treatment has resulted in less than ideal treatment outcomes for AML patients, especially for elderly patients and those with unfavourable profiles. Over the past two years, six new therapeutic agents have received regulatory approval, suggesting that a number of obstacles to treating AML have been addressed and the treatment landscape for AML is finally changing. This review outlines the challenges and obstacles in treating AML and highlights the advances in AML treatment made in recent years, including Vyxeos®, midostaurin, gemtuzumab ozogamicin, and venetoclax, with particular emphasis on combination treatment strategies. We also discuss the potential utility of new combination products such as one that we call "EnFlaM", which comprises an encapsulated nanoformulation of flavopiridol and mitoxantrone. Finally, we provide a review on the immunotherapeutic landscape of AML, discussing yet another angle through which novel treatments can be designed to further improve treatment outcomes for AML patients.
Collapse
Affiliation(s)
- Kent T J Chen
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Interdisciplinary Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Roger Gilabert-Oriol
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Marcel B Bally
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- Cuprous Pharmaceuticals Inc., Vancouver, British Columbia, Canada.
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Ada W Y Leung
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Cuprous Pharmaceuticals Inc., Vancouver, British Columbia, Canada
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
Cheng Y, Fan Y, Liu X, Liu Y, Liu J, Wang D, Yu Y, Qin S, Liu W, Huang C, Zhang H, Liang J, Shi J, Sheng L, Yu H. Randomized controlled trial of lobaplatin plus etoposide vs. cisplatin plus etoposide as first-line therapy in patients with extensive-stage small cell lung cancer. Oncol Lett 2019; 17:4701-4709. [PMID: 30988825 DOI: 10.3892/ol.2019.10125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 02/04/2019] [Indexed: 12/11/2022] Open
Abstract
The majority of previous studies of lobaplatin in small cell lung cancer (SCLC) are small phase I-II studies. The present study aimed to verify the non-inferiority (in terms of efficacy) of lobaplatin plus etoposide (EL) vs. cisplatin plus etoposide (EP) in patients with previously untreated extensive-stage SCLC (ES-SCLC). This phase III non-inferiority randomized clinical trial enrolled patients at 17 sites between September 2010 and May 2013. Patients were randomized to EL (30 mg/m2 lobaplatin on day 1 and 100 mg/m2 etoposide on days 1-3, for 21-day cycles) or EP (80 mg/m2 cisplatin on day 1 and 100 mg/m2 etoposide on days 1-3, for 21-day cycles). The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), objective response rate, disease control rate (DCR), toxicity and quality of life (QoL). A total of 234 patients were randomized to the EL (n=122) and EP (n=112) treatment groups. The median PFS, median OS and DCR were 5.1 vs. 5.3 months (P=0.786), 10.6 vs. 9.7 months (P=0.701) and 85.5 vs. 86.7% (P=0.848) in the EL vs. EP groups, respectively. Patients in the EL group had significantly lower frequencies of nephrotoxicity (2.5 vs. 11.7%; P=0.008), nausea (22.3 vs. 40.5%; P=0.003) and vomiting (14.1 vs. 35.1%; P<0.001) than those in the EP group. Overall, EL was not inferior to EP in terms of PFS and OS. The tolerance and QoL of the EL regimen were better than those of the EP regimen. EL is thus an alternative choice for the first-line treatment of ES-SCLC.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Oncology, Jilin Cancer Hospital, Changchun, Jilin 130021, P.R. China
| | - Yun Fan
- Department of Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiaoqing Liu
- Department of Chest Tumor, 307th Hospital of The Academy of Military Medical Sciences, Beijing 100071, P.R. China
| | - Yunpeng Liu
- Department of Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Dong Wang
- Department of Oncology, The Daping Hospital of Third Military Medical University, Chongqing 400042, P.R. China
| | - Yan Yu
- Department of Oncology, The Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shukui Qin
- Department of Oncology, People's Liberation Army Cancer Center of Nanjing 81 Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Wei Liu
- Department of Oncology, Hebei Cancer Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Cheng Huang
- Department of Oncology, Fujian Province Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Helong Zhang
- Department of Oncology, The Tangdu Hospital of Fourth Military Medical University, Xi'an, Shanxi 710038, P.R. China
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Peking University, Beijing 102206, P.R. China
| | - Jianhua Shi
- Department of Oncology, Linyi Cancer Hospital, Linyi, Shandong 251500, P.R. China
| | - Lijun Sheng
- Department of Oncology, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| | - Hao Yu
- Department of Biostatistics, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
26
|
Jin B, Liu Y, Xie J, Luo B, Long C. Ethnobotanical survey of plant species for herbal tea in a Yao autonomous county (Jianghua, China): results of a 2-year study of traditional medicinal markets on the Dragon Boat Festival. JOURNAL OF ETHNOBIOLOGY AND ETHNOMEDICINE 2018; 14:58. [PMID: 30185187 PMCID: PMC6125875 DOI: 10.1186/s13002-018-0257-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 08/22/2018] [Indexed: 05/18/2023]
Abstract
BACKGROUND Herbal tea is widely consumed in Jianghua, a Yao autonomous county in Hunan Province, China, to prevent and treat diseases. The materials in herbal tea at the traditional medicinal markets at the Dragon Boat Festival remain unknown. The aims of the paper were (1) to specifically investigate the materials of herbal tea used by Yao nationalities in Hunan Province, (2) to record the most common and the culturally important medicinal plant species in the markets, and (3) to compare the medical plant tradition both used for herbal tea between the Jianghua and Lingnan regions. METHODS During 2016-2017, 215 vendors were interviewed at traditional medicinal markets at the Dragon Boat Festival in Jianghua to record plants used for herbal tea and to document traditional knowledge of their medicinal function, habitat, and conservation status. Bunches of medicinal plants were purchased to identify the species and to prepare voucher specimens. Cognitive salience (CS) based on free-lists and use value (UV) were calculated to analyze the cultural importance of medical plants; other quantitative methods (coefficient of similarity and chi-square analysis) were applied for comparison of herbal tea tradition between the Jianghua and Lingnan regions. RESULTS A total of 169 species belonging to 66 families and 142 genera were recorded in herbal tea to treat health conditions in the study area. There were 30 health conditions that were recorded, with heat-clearing and detoxifying being the most common medicinal function, followed by treating rheumatism and promoting blood circulation. Of the 169 species, 97 were herbs. The whole plant was the most commonly used plant part in the preparation of herbal tea. According to the national evaluation criteria, three of these species are listed on "China's red list" and registered as vulnerable (VU). By comparing the coefficient of similarity of herbal tea plants and the number of mentions for part(s) used in Jianghua and Lingnan, the medicinal plant tradition is different in two areas. CONCLUSIONS Herbal tea in Jianghua reflects the cultural diversity of the Yao people and the plant diversity of the region. Future research on the safety, efficacy, and the adulterants of herbal tea are needed for sustainable utilization.
Collapse
Affiliation(s)
- Bing Jin
- College of Sciences, Nanjing Agricultural University, Nanjing, 210095 China
| | - Yujing Liu
- Ecological Institute, Jiangsu University, Zhenjiang, 212013 China
| | - Jiaxi Xie
- Ecological Institute, Jiangsu University, Zhenjiang, 212013 China
| | - Binsheng Luo
- College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081 China
| | - Chunlin Long
- Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081 China
| |
Collapse
|
27
|
Damiani RM, Moura DJ, Viau CM, Brito V, Morás AM, Henriques JAP, Saffi J. Influence of PARP-1 inhibition in the cardiotoxicity of the topoisomerase 2 inhibitors doxorubicin and mitoxantrone. Toxicol In Vitro 2018; 52:203-213. [PMID: 29913208 DOI: 10.1016/j.tiv.2018.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/17/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023]
Abstract
Doxorubicin (DOX) and Mitoxantrone (MTX) are very effective drugs for a range of tumors despite being highly cardiotoxic. DNA topoisomerase 2 beta (Top2ß) was revealed as key mediator of DOX-induced cardiotoxicity, although ROS generation is also an important mechanism. Oxidative stress is also an important issue in MTX-induced cardiotoxicity that is manifested by mitochondrial dysfunction. Studies have demonstrated the relationship between PARP-1 overactivation and cell viability in DOX-treated cardiomyocytes. In reference of MTX, data regarding PARP-1 overactivation as the mechanism responsible for cardiotoxicity is difficult to find. The aim of this study was to evaluate the influence of PARP-1 inhibitor DPQ on DOX- and MTX-mediated cardiotoxicity. Cells were exposed for 24 h to DOX or MTX in the presence or absence of DPQ. Viability, apoptosis, and genotoxicity assays were carried out. Immunofluorescence of phosphorylated histone H2AX was analyzed in H9c2 cells and cardiomyocytes from neonatal rats. Results demonstrated that DPQ co-treatment increases DOX-induced apoptosis in H9c2 cells. DPQ also prevents DOX and MTX-ROS generation in part by increasing SOD and CAT activities. Furthermore, DPQ co-treatment increased the generation of DNA strand breaks by DOX and MTX whilst also inducing phosphorylation of H2AX, MRE11, and ATM in H9c2 cells. Our results demonstrated that as well as increasing DNA damage and inducing apoptotic cell death, DPQ enhances DOX- and MTX-mediated cytotoxicity in H9c2.
Collapse
Affiliation(s)
- Roberto Marques Damiani
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil; Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil; Centro Universitário Ritter dos Reis (UniRitter), Orfanotrófio st, 555, Porto Alegre, RS, Brazil.
| | - Dinara Jaqueline Moura
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - Cassiana Macagnan Viau
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - Verônica Brito
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - Ana Moira Morás
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - João Antonio Pêgas Henriques
- Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil; Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil
| | - Jenifer Saffi
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil; Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil
| |
Collapse
|
28
|
Enache M, Toader AM. Insights Into Molecular Interaction of Antitumor Drug Mitoxantrone With Anionic Surfactant Sodium Dodecyl Sulfate at Different Temperatures. J SURFACTANTS DETERG 2018. [DOI: 10.1002/jsde.12014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Mirela Enache
- Institute of Physical Chemistry “Ilie Murgulescu,” Romanian Academy, Splaiul Independentei 202; Bucharest 060021 Romania
| | - Ana Maria Toader
- Institute of Physical Chemistry “Ilie Murgulescu,” Romanian Academy, Splaiul Independentei 202; Bucharest 060021 Romania
| |
Collapse
|
29
|
Mature Ovarian Teratoma After Treatment for Unilateral Retinoblastoma With Nonmutated RB1. J Pediatr Hematol Oncol 2017; 39:485-486. [PMID: 28719514 DOI: 10.1097/mph.0000000000000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Spectroscopic Investigation of the Interaction of the Anticancer Drug Mitoxantrone with Sodium Taurodeoxycholate (NaTDC) and Sodium Taurocholate (NaTC) Bile Salts. Molecules 2017; 22:molecules22071079. [PMID: 28657593 PMCID: PMC6152313 DOI: 10.3390/molecules22071079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/14/2017] [Accepted: 06/21/2017] [Indexed: 12/25/2022] Open
Abstract
The focus of the present work was to investigate the interaction of the anticancer drug mitoxantrone with two bile salts, sodium taurodeoxycholate (NaTDC) and sodium taurocholate (NaTC). Ultraviolet-visible (UV-Vis) absorption and electron paramagnetic resonance (EPR) spectroscopy were used to quantify the interaction and to obtain information on the location of mitoxantrone in bile salt micelles. The presence of submicellar concentrations of both bile salts induces mitoxantrone aggregation and the extent of drug aggregation in NaTDC is higher than in NaTC. For micellar bile salts concentrations, mitoxantrone monomers are entrapped in the micellar core. Binding constants, micelle/water partition coefficients and the corresponding thermodynamic parameters for binding and partitioning processes were estimated using the changes in monomer absorbance in the presence of bile salts. Binding interaction of mitoxantrone is stronger for NaTDC than NaTC micelles, whereas partitioning efficiency is higher for NaTC micelles for all investigated temperatures. Thermodynamic parameters indicate that both binding and partitioning processes are spontaneous and entropy controlled. The spectral behavior and thermodynamic parameters indicate distinct types of mitoxantrone interaction with NaTDC and NaTC micelles supported by the differences in nature and structure of bile salts micelles.
Collapse
|
31
|
Advances in the Chemistry of Natural and Semisynthetic Topoisomerase I/II Inhibitors. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2017. [DOI: 10.1016/b978-0-444-63929-5.00002-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
32
|
Reis-Mendes A, Gomes AS, Carvalho RA, Carvalho F, Remião F, Pinto M, Bastos ML, Sousa E, Costa VM. Naphthoquinoxaline metabolite of mitoxantrone is less cardiotoxic than the parent compound and it can be a more cardiosafe drug in anticancer therapy. Arch Toxicol 2016; 91:1871-1890. [PMID: 27629428 DOI: 10.1007/s00204-016-1839-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022]
Abstract
Mitoxantrone (MTX) is an antineoplastic agent used to treat several types of cancers and on multiple sclerosis, which shows a high incidence of cardiotoxicity. Still, the underlying mechanisms of MTX cardiotoxicity are poorly understood and the potential toxicity of its metabolites scarcely investigated. Therefore, this work aimed to synthesize the MTX-naphthoquinoxaline metabolite (NAPHT) and to compare its cytotoxicity to the parent compound in 7-day differentiated H9c2 cells using pharmacological relevant concentrations (0.01-5 µM). MTX was more toxic in equivalent concentrations in all cytotoxicity tests performed [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide reduction, neutral red uptake, and lactate dehydrogenase release assays] and times tested (24 and 48 h). Both MTX and NAPHT significantly decreased mitochondrial membrane potential in 7-day differentiated H9c2 cells after a 12-h incubation. However, energetic pathways were affected in a different manner after MTX or NAPHT incubation. ATP increased and lactate levels decreased after a 24-h incubation with MTX, whereas for the same incubation time and concentrations, NAPHT did not cause any significant effect. The increased activity of ATP synthase seems responsible for MTX-induced increases in ATP levels, as oligomycin (an inhibitor of ATP synthase) abrogated this effect on 5 µM MTX-incubated cells. 3-Methyladenine (an autophagy inhibitor) was the only molecule to give a partial protection against the cytotoxicity produced by MTX or NAPHT. To the best of our knowledge, this was the first broad study on NAPHT cardiotoxicity, and it revealed that the parent drug, MTX, caused a higher disruption in the energetic pathways in a cardiac model in vitro, whereas autophagy is involved in the toxicity of both compounds. In conclusion, NAPHT is claimed to largely contribute to MTX-anticancer properties; therefore, this metabolite should be regarded as a good option for a safer anticancer therapy since it is less cardiotoxic than MTX.
Collapse
Affiliation(s)
- A Reis-Mendes
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - A S Gomes
- UCIBIO-REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.,Lab. Química Orgânica e Farmacêutica, Dep. Química, Faculdade de Farmácia, U. Porto, Porto, Portugal
| | - R A Carvalho
- Centre for Functional Ecology, Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - F Carvalho
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - F Remião
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - M Pinto
- Lab. Química Orgânica e Farmacêutica, Dep. Química, Faculdade de Farmácia, U. Porto, Porto, Portugal.,CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - M L Bastos
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - E Sousa
- Lab. Química Orgânica e Farmacêutica, Dep. Química, Faculdade de Farmácia, U. Porto, Porto, Portugal.,CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - V M Costa
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
33
|
Ahmed SH, Moussa Sherif DE, Fouad Y, Kelany M, Abdel-Rahman O. Principles of a risk evaluation and mitigation strategy (REMS) for breast cancer patients receiving potentially cardiotoxic adjuvant treatments. Expert Opin Drug Saf 2016; 15:911-23. [DOI: 10.1517/14740338.2016.1170115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
34
|
Structural basis of DNA topoisomerase II-α (Top2-α) inhibition: a computational analysis of interactions between Top2-α and its inhibitors. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1567-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
35
|
Inhibition of DNA Topoisomerase Type IIα (TOP2A) by Mitoxantrone and Its Halogenated Derivatives: A Combined Density Functional and Molecular Docking Study. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6817502. [PMID: 27088089 PMCID: PMC4754470 DOI: 10.1155/2016/6817502] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023]
Abstract
In this study, mitoxantrone and its halogenated derivatives have been designed by density functional theory (DFT) to explore their structural and thermodynamical properties. The performance of these drugs was also evaluated to inhibit DNA topoisomerase type IIα (TOP2A) by molecular docking calculation. Noncovalent interactions play significant role in improving the performance of halogenated drugs. The combined quantum and molecular mechanics calculations revealed that CF3 containing drug shows better preference in inhibiting the TOP2A compared to other modified drugs.
Collapse
|
36
|
Synthesis and biological activity of ferrocenyl indeno[1,2-c]isoquinolines as topoisomerase II inhibitors. Bioorg Med Chem 2016; 24:651-60. [DOI: 10.1016/j.bmc.2015.12.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/15/2015] [Accepted: 12/17/2015] [Indexed: 12/18/2022]
|
37
|
Nabors LB, Surboeck B, Grisold W. Complications from pharmacotherapy. HANDBOOK OF CLINICAL NEUROLOGY 2016; 134:235-250. [PMID: 26948358 DOI: 10.1016/b978-0-12-802997-8.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The pharmacotherapy management of cancers of the nervous system has significant overlap with systemic solid cancers that may utilize similar drugs or agents. There is however a unique aspect related to central nervous system (CNS) cancers where therapies directed against a malignant process may have enhanced toxicities or toxicities unique to the CNS. In addition, many agents used to treat CNS malignancies have unique CNS toxicities that may require a specific intervention. This chapter attempts to review conventional and biologic therapies utilized for CNS malignancies and characterize expected and, if known, unique toxicities.
Collapse
Affiliation(s)
- L Burt Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Birgit Surboeck
- Department of Neurology, Kaiser-Franz-Josef Hospital, Vienna, Austria
| | - Wolfgang Grisold
- Department of Neurology, Kaiser-Franz-Josef Hospital, Vienna, Austria; Medical University of Vienna, Vienna, Austria
| |
Collapse
|
38
|
Efficacy of doxorubicin-transferrin conjugate in apoptosis induction in human leukemia cells through reactive oxygen species generation. Cell Oncol (Dordr) 2015; 39:107-18. [PMID: 26611752 PMCID: PMC4820500 DOI: 10.1007/s13402-015-0256-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2015] [Indexed: 12/18/2022] Open
Abstract
Background Doxorubicin (DOX) is a small molecular cytotoxic agent that can be transferred efficiently to cancer cells by nanocarriers. This anthracycline antibiotic serves as an effective anti-neoplastic drug against both hematological and solid malignancies. Here, we set out to assess the capacity of a novel doxorubicin - transferrin conjugate (DOX-TRF) to provoke apoptosis in human normal and leukemia cells through free radicals produced via a redox cycle of doxorubicin (DOX) when released from its conjugate. Methods After DOX-TRF exposure, we determined the time-course of apoptotic and necrotic events, the generation of reactive oxygen species (ROS), changes in mitochondrial membrane potential, as well as alterations in cytochrome c levels and intracellular calcium concentrations in human leukemia-derived cell lines (CCRF-CEM, K562 and its doxorubicin-resistant derivative K562/DOX) and normal peripheral blood-derived mononuclear cells (PBMC). Results We found that DOX-TRF can induce apoptosis in all leukemia-derived cell lines tested, which was associated with morphological changes and decreases in mitochondrial membrane potential. In comparison to free DOX treated cells, we observed a time-dependency between a higher level of ROS generation and a higher drop in mitochondrial membrane potential, particularly in the doxorubicin-resistant cell line. In addition, we found that the apoptotic cell death induced by DOX-TRF was directly associated with a release of cytochrome c from the mitochondria and an increase in intracellular calcium level in all human leukemia-derived cell lines tested. Conclusions Our data indicate that DOX-TRF is considerably more cytotoxic to human leukemia cells than free DOX. In addition, we show that DOX-TRF can effectively produce free radicals, which are directly involved in apoptosis induction.
Collapse
|
39
|
Evison BJ, Sleebs BE, Watson KG, Phillips DR, Cutts SM. Mitoxantrone, More than Just Another Topoisomerase II Poison. Med Res Rev 2015; 36:248-99. [PMID: 26286294 DOI: 10.1002/med.21364] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 02/06/2023]
Abstract
Mitoxantrone is a synthetic anthracenedione originally developed to improve the therapeutic profile of the anthracyclines and is commonly applied in the treatment of breast and prostate cancers, lymphomas, and leukemias. A comprehensive overview of the drug's molecular, biochemical, and cellular pharmacology is presented here, beginning with the cardiotoxic nature of its predecessor doxorubicin and how these properties shaped the pharmacology of mitoxantrone itself. Although mitoxantrone is firmly established as a DNA topoisomerase II poison within mammalian cells, it is now clear that the drug interacts with a much broader range of biological macromolecules both covalently and noncovalently. Here, we consider each of these interactions in the context of their wider biological relevance to cancer therapy and highlight how they may be exploited to further enhance the therapeutic value of mitoxantrone. In doing so, it is now clear that mitoxantrone is more than just another topoisomerase II poison.
Collapse
Affiliation(s)
- Benny J Evison
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Keith G Watson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Don R Phillips
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Suzanne M Cutts
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| |
Collapse
|
40
|
Shaul P, Steinbuch KB, Blacher E, Stein R, Fridman M. Exploring the Effects of Glycosylation and Etherification of the Side Chains of the Anticancer Drug Mitoxantrone. ChemMedChem 2015; 10:1528-38. [DOI: 10.1002/cmdc.201500274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 02/02/2023]
|
41
|
The age factor for mitoxantrone’s cardiotoxicity: Multiple doses render the adult mouse heart more susceptible to injury. Toxicology 2015; 329:106-19. [PMID: 25582955 DOI: 10.1016/j.tox.2015.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/04/2015] [Accepted: 01/08/2015] [Indexed: 11/30/2022]
|
42
|
Li J, Feng J, Luo C, Herman HYS, Jiang RW. Absolute configuration of podophyllotoxone and its inhibitory activity against human prostate cancer cells. Chin J Nat Med 2015; 13:59-64. [PMID: 25660289 DOI: 10.1016/s1875-5364(15)60007-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Indexed: 11/24/2022]
Abstract
Podophyllotoxone (1) was isolated from the roots of Dysosma versipellis. The structure was determined by spectroscopic analysis in combination with single-crystal X-ray analysis. The absolute configuration of compound 1 was assigned based on the Flack parameter. It showed significant inhibitory activities against human prostate cancer cells PC3 and DU145 with IC50 values being 14.7 and 20.6 μmol·L(-1), respectively. It also arrested the cells at G2/M phase. Tubulin polymerization assay showed that it inhibited the tubulin polymerization in a dose-dependent manner, and molecular docking analysis revealed a different binding mode with tubulin as compared with those known tubulin inhibitors.
Collapse
Affiliation(s)
- Juan Li
- Guangxi Key Laboratory of Functional Phytochemicals Research and Utilization, Guangxi Institute of Botany, Guangxi Zhuang Autonomous Region and Chinese Academy of Sciences, Guilin 541006, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Juan Feng
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Cheng Luo
- State Key Laboratory of New Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Ho-Yung Sung Herman
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Ren-Wang Jiang
- Guangxi Key Laboratory of Functional Phytochemicals Research and Utilization, Guangxi Institute of Botany, Guangxi Zhuang Autonomous Region and Chinese Academy of Sciences, Guilin 541006, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.
| |
Collapse
|
43
|
Zhu SJ, Ying HZ, Wu Y, Qiu N, Liu T, Yang B, Dong XW, Hu YZ. Design, synthesis and biological evaluation of novel podophyllotoxin derivatives bearing 4β-disulfide/trisulfide bond as cytotoxic agents. RSC Adv 2015. [DOI: 10.1039/c5ra12837d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A novel series of podophyllotoxin derivatives bearing 4β-disulfide/trisulfide were designed, synthesized and biologically evaluated for their cytotoxic activities against KB cells and KB/VCR cells.
Collapse
Affiliation(s)
- Shi-Jun Zhu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Hua-Zhou Ying
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Yan Wu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Ni Qiu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Tao Liu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Bo Yang
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Xiao-Wu Dong
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Yong-Zhou Hu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| |
Collapse
|
44
|
Vitamin C effect on mitoxantrone-induced cytotoxicity in human breast cancer cell lines. PLoS One 2014; 9:e115287. [PMID: 25531443 PMCID: PMC4274052 DOI: 10.1371/journal.pone.0115287] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/20/2014] [Indexed: 12/02/2022] Open
Abstract
In recent years the use of natural dietary antioxidants to minimize the cytotoxicity and the damage induced in normal tissues by antitumor agents is gaining consideration. In literature, it is reported that vitamin C exhibits some degree of antineoplastic activity whereas Mitoxantrone (MTZ) is a synthetic anti-cancer drug with significant clinical effectiveness in the treatment of human malignancies but with severe side effects. Therefore, we have investigated the effect of vitamin C alone or combined with MTZ on MDA-MB231 and MCF7 human breast cancer cell lines to analyze their dose-effect on the tumor cellular growth, cellular death, cell cycle and cell signaling. Our results have evidenced that there is a dose-dependence on the inhibition of the breast carcinoma cell lines, MCF7 and MDA-MB231, treated with vitamin C and MTZ. Moreover, their combination induces: i) a cytotoxic effect by apoptotic death, ii) a mild G2/M elongation and iii) H2AX and mild PI3K activation. Hence, the formulation of vitamin C with MTZ induces a higher cytotoxicity level on tumor cells compared to a disjointed treatment. We have also found that the vitamin C enhances the MTZ effect allowing the utilization of lower chemotherapic concentrations in comparison to the single treatments.
Collapse
|
45
|
Mitochondrial cumulative damage induced by mitoxantrone: late onset cardiac energetic impairment. Cardiovasc Toxicol 2014; 14:30-40. [PMID: 24096626 DOI: 10.1007/s12012-013-9230-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitoxantrone (MTX) is a chemotherapeutic agent, which presents late irreversible cardiotoxicity. This work aims to highlight the mechanisms involved in the MTX-induced cardiotoxicity, namely the effects toward mitochondria using in vivo and in vitro studies. Male Wistar rats were treated with 3 cycles of 2.5 mg/kg MTX at day 0, 10, and 20. One treated group was euthanized on day 22 (MTX22) to evaluate the early MTX cardiac toxic effects, while the other was euthanized on day 48 (MTX48), to allow the evaluation of MTX late cardiac effects. Cardiac mitochondria isolated from 4 adult untreated rats were also used to evaluate in vitro the MTX (10 nM, 100 nM, and 1 μM) direct effects upon mitochondria functionality. Two rats of MTX48 died on day 35, and MTX treatment caused a reduction in relative body weight gain in both treated groups with no significant changes in water and food intake. Decreased levels of plasma total creatine kinase and CK-MB were detected in the MTX22 group, and increased plasma levels of lactate were seen in MTX48. Increased cardiac relative mass and microscopic changes were evident in both treated groups. Considering mitochondrial effects, for the first time, it was evidenced that MTX induced an increase in the complex IV and complex V activities in MTX22 group, while a decrease in the complex V activity was accompanied by the reduction in ATP content in the MTX48 rats. No alterations in mitochondria transmembrane potential were found in isolated mitochondria from MTX48 rats or in isolated mitochondria directly incubated with MTX. This study highlights the relevance of the cumulative MTX-induced in vivo mitochondriopathy to the MTX cardiotoxicity.
Collapse
|
46
|
Therapeutic concentrations of mitoxantrone elicit energetic imbalance in H9c2 cells as an earlier event. Cardiovasc Toxicol 2014; 13:413-25. [PMID: 24046265 DOI: 10.1007/s12012-013-9224-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mitoxantrone (MTX) is a chemotherapeutic agent that emerged as an alternative to anthracycline therapy. However, MTX also causes late cardiotoxicity, being oxidative stress and mitochondrial-impaired function proposed as possible mechanisms. This work aimed to investigate the relevance of these mechanisms to the MTX toxicity in H9c2 cells, using therapeutic concentrations. The observed cytotoxicity of MTX was time and concentration dependent in both lactate dehydrogenase leakage assay and MTT reduction assay. Two therapeutic concentrations (100 nM and 1 μM) and three time points were selected (24, 48, and 96 h) for further studies. Both MTX concentrations caused a significant increase in caspase-3 activity, which was not prevented by inhibiting MTX CYP450-metabolism. Significant decreases were observed in the total and reduced glutathione levels only in MTX 100 nM at 96 h; however, neither alterations in oxidized glutathione nor increases in the malondialdehyde levels were observed at any time or concentrations tested. On the other hand, changes in the intracellular ATP levels, mitochondrial membrane potential, and intracellular calcium levels were observed in both concentrations and all time tested. Noteworthy, decreased levels of ATP-synthase expression and activity and increases in the reactive species generation were observed at 96 h in both working concentrations. However, the radical scavenger N-acetylcysteine or the mitochondrial function enhancer L-carnitine did not prevent MTX cytotoxicity. Thus, this work evidenced the early MTX-induced energetic crisis as a possible key factor in the cell injury.
Collapse
|
47
|
Phase I clinical trial of pegylated liposomal mitoxantrone plm60-s: pharmacokinetics, toxicity and preliminary efficacy. Cancer Chemother Pharmacol 2014; 74:637-46. [PMID: 25034977 DOI: 10.1007/s00280-014-2523-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 07/06/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Plm60-s is a pegylated liposomal mitoxantrone formulation, in which mitoxantrone was loaded into small unilamellar vesicles (~60 nm) made from solid lipid membrane. This two-arm, dose-escalating phase I study was designed to determine safety and pharmacokinetics of plm60-s, and to compare with those of conventional mitoxantrone injection (c-MI). METHODS Patients received an intravenous infusion of plm60-s at 6, 10, 12, 14, 16 and 18 mg/m(2) every 4 weeks. Three or 6 patients were in each group of dose level. If more than one third patients of a group experienced dose-limiting toxicity, dose climbing will stop. The control group of 3 patients received c-MI at 10 mg/m(2) every 28 days. Samples for pharmacokinetic studies were collected. The analysis of the safety and tolerability was done according to the record and laboratory examination, etc. RESULTS Twenty patients were enrolled. One grade 3 leukocytopenia occurred in plm60-s groups. Plm60-s was safer than c-MI at the same dose of 10 mg/m(2). Two complete responses and one partial response occurred in plm60-s group. In plasma, plm60-s exhibited sustained release of the content, resulting in the reduced peak concentrations and enhanced AUC of released MIT. Total mitoxantrone was linearly cleared, and mitoxantrone was predominantly in the liposomal encapsulation form. Repeated administration of plm60-s did not affect the clearance kinetics. CONCLUSIONS At a dose of up to 18 mg/m(2), plm60-s could be well tolerated and potential efficacy could be observed. The pharmacokinetic profile of plm60-s was remarkably altered. Further investigations are in progression.
Collapse
|
48
|
Winkler GC, Barle EL, Galati G, Kluwe WM. Functional differentiation of cytotoxic cancer drugs and targeted cancer therapeutics. Regul Toxicol Pharmacol 2014; 70:46-53. [PMID: 24956585 DOI: 10.1016/j.yrtph.2014.06.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/05/2014] [Accepted: 06/15/2014] [Indexed: 11/16/2022]
Abstract
There is no nationally or internationally binding definition of the term "cytotoxic drug" although this term is used in a variety of regulations for pharmaceutical development and manufacturing of drugs as well as in regulations for protecting medical personnel from occupational exposure in pharmacy, hospital, and other healthcare settings. The term "cytotoxic drug" is frequently used as a synonym for any and all oncology or antineoplastic drugs. Pharmaceutical companies generate and receive requests for assessments of the potential hazards of drugs regularly - including cytotoxicity. This publication is intended to provide functional definitions that help to differentiate between generically-cytotoxic cancer drugs of significant risk to normal human tissues, and targeted cancer therapeutics that pose much lesser risks. Together with specific assessments, it provides comprehensible guidance on how to assess the relevant properties of cancer drugs, and how targeted therapeutics discriminate between cancer and normal cells. The position of several regulatory agencies in the long-term is clearly to regulate all drugs regardless of classification, according to scientific risk based data. Despite ongoing discussions on how to replace the term "cytotoxic drugs" in current regulations, it is expected that its use will continue for the near future.
Collapse
Affiliation(s)
- Gian C Winkler
- Novartis Pharma AG NIBR, Postfach, CH-4002 Basel, Switzerland.
| | | | - Giuseppe Galati
- Patheon Inc., 2100 Syntex Court, Mississauga, Ontario L5N 7K9, Canada.
| | - William M Kluwe
- Novartis Pharmaceuticals Corporation, NIBR, One Health Plaza, East Hanover, NJ 07936-1080, USA.
| |
Collapse
|
49
|
Li C, Zhao X, Deng C, Wang C, Wei N, Cui J. Pegylated liposomal mitoxantrone is more therapeutically active than mitoxantrone in L1210 ascitic tumor and exhibits dose-dependent activity saturation effect. Int J Pharm 2014; 460:165-72. [DOI: 10.1016/j.ijpharm.2013.10.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 10/03/2013] [Indexed: 11/30/2022]
|
50
|
Wartlick F, Bopp A, Henninger C, Fritz G. DNA damage response (DDR) induced by topoisomerase II poisons requires nuclear function of the small GTPase Rac. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3093-3103. [DOI: 10.1016/j.bbamcr.2013.08.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/14/2013] [Accepted: 08/23/2013] [Indexed: 01/12/2023]
|