1
|
Acid-sensitive and L61-crosslinked hyaluronic acid nanogels for overcoming tumor drug-resistance. Int J Biol Macromol 2021; 188:11-23. [PMID: 34364934 DOI: 10.1016/j.ijbiomac.2021.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/21/2021] [Accepted: 08/01/2021] [Indexed: 01/16/2023]
Abstract
Low intracellular drugs concentration is one of the main representations of multidrug resistance (MDR), which often results in a weak or failed chemotherapy on cancer treatment. Herein, an acid-sensitive and pluronic L61-linked hyaluronic acid nanogels (HA-L61OE/NGs) were developed for solving this problem. The nanogels could well hold more drugs under neutral condition, while triggering efficiently drugs release (61.42% within 24 h) in acidic environment. In vitro cells experiments demonstrated that the nanogels greatly increased intracellular drugs concentration by CD44-mediated endocytosis and L61-mediated anti-MDR effect, resulting in the enhanced cell-killing in MDR cells. In vivo studies verified HA-L61OE/NGs could avoid drugs leakage in blood and reduce systemic toxicity. Subsequently, the specific accumulation and penetration of nanogels at tumor regions lead to the highest tumor growth inhibition (TGI, 77.42%). Overall, HA-L61OE/NGs were effective on MDR tumor therapy and expected to be further used in clinical trials.
Collapse
|
2
|
Clinical utility of ABCB1 and ABCG2 genotyping for assessing the clinical and pathological response to FAC therapy in Mexican breast cancer patients. Cancer Chemother Pharmacol 2021; 87:843-853. [PMID: 33740100 DOI: 10.1007/s00280-021-04244-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/05/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Resistance to neoadjuvant chemotherapy with 5-fluorouracil, doxorubicin, and cyclophosphamide (FAC) in some patients with locally advanced breast cancer remains one of the main obstacles to first-line treatment. We investigated clinical and pathological responses to FAC neoadjuvant chemotherapy in Mexican women with breast cancer and their possible association with SNPs present in ABC transporters as predictors of chemoresistance. MATERIALS A total of 102 patients undergoing FAC neoadjuvant chemotherapy were included in the study. SNP analysis was performed by RT-PCR from genomic DNA. Two SNPs were analyzed: ABCB1 rs1045642 (3435 C > T) and ABCG2 rs2231142 (421 G > T). RESULTS In clinical response evaluation, significant associations were found between the ABCB1 C3435T genotype and breast cancer chemoresistant and chemosensitive patients (p < 0.05). In the early clinical response, patients with genotype C/C or C/T were more likely to be chemosensitive to neoadjuvant therapy than patients with genotype T/T (OR = 4.055; p = 0.0064). Association analysis between the ABCB1 gene polymorphism and the pathologic response to FAC chemotherapy showed that the C/C + C/T genotype was a protective factor against chemoresistance (OR = 3.714; p = 0.0104). Polymorphisms in ABCG2 indicated a lack of association with resistance to chemotherapy (p = 0.2586) evaluating the clinical or pathological response rate to FAC neoadjuvant chemotherapy. CONCLUSION The early clinical response and its association with SNPs in the ABCB1 transporter are preserved until the pathological response to neoadjuvant chemotherapy; therefore, it could be used as a predictor of chemoresistance in locally advanced breast cancer patients of the Mexican population.
Collapse
|
3
|
Omidi Y, Kianinejad N, Kwon Y, Omidian H. Drug delivery and targeting to brain tumors: considerations for crossing the blood-brain barrier. Expert Rev Clin Pharmacol 2021; 14:357-381. [PMID: 33554678 DOI: 10.1080/17512433.2021.1887729] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: The blood-brain barrier (BBB) selectively impedes the transportation of drug molecules into the brain, which makes the drug delivery and targeting of brain tumors very challenging.Areas covered: Having surveyed the recent literature, comprehensive insights are given into the impacts of the BBB on the advanced drug delivery and targeting modalities for brain tumors.Expert opinion: Brain capillary endothelial cells form the BBB in association with astrocytes, pericytes, neurons, and extracellular matrix. Coop of these forms the complex setting of neurovascular unite. The BBB maintains the brain homeostasis by restrictive controlling of the blood circulating nutrients/substances trafficking. Despite substantial progress on therapy of brain tumors, there is no impeccable strategy to safely deliver chemotherapeutics into the brain. Various strategies have been applied to deliver chemotherapeutics into the brain (e.g. BBB opening, direct delivery by infusion, injection, microdialysis, and implants, and smart nanosystems), which hold different pros and cons. Of note, smart nanoscale multifunctional nanomedicines can serve as targeting, imaging, and treatment modality for brain tumors. Given that aggressive brain tumors (e.g. gliomas) are often unresponsive to any treatments, an in-depth understanding of the molecular/cellular complexity of brain tumors might help the development of smart and effective treatment modalities.
Collapse
Affiliation(s)
- Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Nazanin Kianinejad
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Young Kwon
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Hossein Omidian
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| |
Collapse
|
4
|
Sri Laasya T, Thakur S, Poduri R, Joshi G. Current insights toward kidney injury: Decrypting the dual role and mechanism involved of herbal drugs in inducing kidney injury and its treatment. CURRENT RESEARCH IN BIOTECHNOLOGY 2020. [DOI: 10.1016/j.crbiot.2020.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
5
|
Sáfár Z, Kecskeméti G, Molnár J, Kurunczi A, Szabó Z, Janáky T, Kis E, Krajcsi P. Inhibition of ABCG2/BCRP-mediated transport-correlation analysis of various expression systems and probe substrates. Eur J Pharm Sci 2020; 156:105593. [PMID: 33059043 DOI: 10.1016/j.ejps.2020.105593] [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: 07/03/2020] [Revised: 09/23/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022]
Abstract
BCRP / ABCG2 is a key determinant of pharmacokinetics of substrate drugs. Several BCRP substrates and inhibitors are of low passive permeability, and the vesicular transport assay works well in this permeability space. Membranes were prepared from BCRP-HEK293, MCF-7/MX, and baculovirus-infected Sf9 cells with (BCRP-Sf9-HAM), and without (BCRP-Sf9) cholesterol loading. Km values for three substrates - estrone-3-sulfate, sulfasalazine, topotecan - correlated well between the four expression systems. In contrast, a 10-20-fold range in Vmax values was observed, with BCRP-HEK293 membranes possessing the largest dynamic range. IC50 values of the different test systems were similar to each other, with 94.4% of pairwise comparisons being within 3-fold. Substrate dependent inhibition showed somewhat greater variation, as 81.4% of IC50 values in the BCRP-HEK293 membranes were within 3-fold in pairwise comparisons. Overall, BCRP-HEK293 membranes demonstrated the highest activity. The IC50 values showed good concordance but substrate dependent inhibition was observed for some drugs.
Collapse
Affiliation(s)
- Zsolt Sáfár
- Solvo Biotechnology, a Charles River Company, 52 Közép fasor, Szeged H-6726, Hungary.
| | - Gábor Kecskeméti
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, Szeged H-6720, Hungary.
| | - Judit Molnár
- Solvo Biotechnology, a Charles River Company, 52 Közép fasor, Szeged H-6726, Hungary.
| | - Anita Kurunczi
- Solvo Biotechnology, a Charles River Company, 52 Közép fasor, Szeged H-6726, Hungary.
| | - Zoltán Szabó
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, Szeged H-6720, Hungary.
| | - Tamás Janáky
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, Szeged H-6720, Hungary.
| | - Emese Kis
- Solvo Biotechnology, a Charles River Company, 52 Közép fasor, Szeged H-6726, Hungary.
| | - Péter Krajcsi
- Solvo Biotechnology, a Charles River Company, 52 Közép fasor, Szeged H-6726, Hungary; Solvo Biotechnology, a Charles River Company, 4-20 Irinyi J str, Budapest H-1117, Hungary; Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter str 50/a, Budapest H-1083, Hungary; Semmelweis University, Faculty of Health Sciences, Vas str 17, Budapest H-1088, Hungary.
| |
Collapse
|
6
|
Norouzi S, Yazdian Robati R, Ghandadi M, Abnous K, Behravan J, Mosaffa F. Comparative proteomics study of proteins involved in induction of higher rates of cell death in mitoxantrone-resistant breast cancer cells MCF-7/MX exposed to TNF-α. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:663-672. [PMID: 32742605 PMCID: PMC7374993 DOI: 10.22038/ijbms.2020.40029.9486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Objective(s): Resistance to medications is one of the main complications in chemotherapy of cancer. It has been shown that some multidrug resistant cancer cells indicate more sensitivity against cytotoxic effects of TNF-α compared to their parental cells. Our previous findings indicated vulnerability of the mitoxantrone-resistant breast cancer cells MCF-7/MX to cell death induced by TNF-α compared to the parent cells MCF-7. In this study, we performed a comparative proteomics analysis for identification of proteins involved in induction of higher susceptibility of MCF-7/MX cells to cytotoxic effect of TNF-α. Materials and Methods: Intensity of protein spots in 2D gel electrophoresis profiles of MCF-7 and MCF-7/MX cells were compared with Image Master Platinum 6.0 software. Selected differential protein-spots were identified with MALDI-TOF/TOF mass spectrometry and database searching. Pathway analyses of identified proteins were performed using PANTHER, KEGG PATHWAY, Gene MANIA and STRING databases. Western blot was performed for confirmation of the proteomics results. Results: Our results indicated that 48 hr exposure to TNF-α induced 87% death in MCF-7/MX cells compared to 19% death in MCF-7 cells. Forty landmarks per 2D gel electrophoresis were matched by Image Master Software. Six proteins were identified with mass spectrometry. Western blot showed that 14-3-3γ and p53 proteins were expressed higher in MCF-7/MX cells treated with TNF-α compared to MCF-7 cells treated with TNF-α. Conclusion: Our results showed that 14-3-3 γ, prohibitin, peroxiredoxin 2 and P53 proteins which were expressed differentially in MCF-7/MX cells treated with TNF-α may involve in the induction of higher rates of cell death in these cells compared to TNF-α-treated MCF-7 cells.
Collapse
Affiliation(s)
- Saeed Norouzi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rezvan Yazdian Robati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Morteza Ghandadi
- Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Behravan
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Reis MA, Matos AM, Duarte N, Ahmed OB, Ferreira RJ, Lage H, Ferreira MJU. Epoxylathyrane Derivatives as MDR-Selective Compounds for Disabling Multidrug Resistance in Cancer. Front Pharmacol 2020; 11:599. [PMID: 32457612 PMCID: PMC7226783 DOI: 10.3389/fphar.2020.00599] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
Background Multidrug resistance (MDR) has been regarded as one of the major hurdles for the successful outcome of cancer chemotherapy. The collateral sensitivity (CS) effect is one the most auspicious anti-MDR strategies. Epoxylathyrane derivatives 1–16 were obtained by derivatization of the macrocyclic diterpene epoxyboetirane A (17), a lathyrane-type macrocyclic diterpene isolated from Euphorbia boetica. Some of these compounds were found to strongly modulate P-glycoprotein (P-gp/ABCB1) efflux. Purpose The main goal was to develop lathyrane-type macrocyclic diterpenes with improved MDR-modifying activity, by targeting more than one anti-MDR mechanism. Study design/methods In this study, the potential CS effect of compounds 1–16 was evaluated against gastric (EPG85-257), pancreatic (EPP85-181), and colon (HT-29) human cancer cells and their drug-resistant counterparts, respectively selected against mitoxantrone (EPG85-257RNOV; EPP85-181RNOV; HT-RNOV) or daunorubicin (EPG85-257RDB; EPP85-181RDB; HT-RDB). The most promising compounds (8, 15, and 16) were investigated as apoptosis inducers, using the assays annexin V/PI and active caspase-3. Results The compounds were more effective against the resistant gastric cell lines, being the CS effect more significant in EPG85-257RDB cells. Taking together the IC50 values and the CS effect, compounds 8, 15, and 16 exhibited the best results. Epoxyboetirane P (8), with the strongest MDR-selective antiproliferative activity against gastric carcinoma EPG85-257RDB cells (IC50 of 0.72 µM), being 10-fold more active against this resistant subline than in sensitive gastric carcinoma cells. The CS effect elicited by compounds 15 and 16 appeared to be by inducing apoptosis via caspase-3 activation. Structure-activity relationships of the compounds were additionally obtained through regression models to clarify the structural determinants associated to the CS effect. Conclusions This study reinforces the importance of lathyrane-type diterpenes as lead molecules for the research of MDR-modifying agents.
Collapse
Affiliation(s)
- Mariana Alves Reis
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Ana M Matos
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Noélia Duarte
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Omar Bauomy Ahmed
- Institute of Pathology, University Hospital Charité, Berlin, Germany
| | - Ricardo J Ferreira
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal.,Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Hermann Lage
- Institute of Pathology, University Hospital Charité, Berlin, Germany
| | - Maria-José U Ferreira
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
8
|
Zhang W, Liu H, Liu C. Biopharmaceutics classification and intestinal absorption of chikusetsusaponin IVa. Biopharm Drug Dispos 2019; 40:276-281. [PMID: 31294470 DOI: 10.1002/bdd.2200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/03/2019] [Accepted: 07/05/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Wenzhou Zhang
- Department of PharmacyLiaocheng People's Hospital Liaocheng China
| | - Hui Liu
- Pharmacy DepartmentLiaocheng Third People's Hospital Liaocheng China
| | - Chongfeng Liu
- Department of PharmacyLiaocheng People's Hospital Liaocheng China
| |
Collapse
|
9
|
Ravegnini G, Sammarini G, Moran S, Calice G, Indio V, Urbini M, Astolfi A, Zanotti F, Pantaleo MA, Hrelia P, Angelini S. Mechanisms of resistance to a PI3K inhibitor in gastrointestinal stromal tumors: an omic approach to identify novel druggable targets. Cancer Manag Res 2019; 11:6229-6244. [PMID: 31308757 PMCID: PMC6615718 DOI: 10.2147/cmar.s189661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 04/16/2019] [Indexed: 12/22/2022] Open
Abstract
Background: Gastrointestinal stromal tumors (GISTs) represent a worldwide paradigm of target therapy. The introduction of tyrosine kinase inhibitors has deeply changed the prognosis of GIST patients, however, the majority of them acquire secondary mutations and progress. Unfortunately, besides tyrosine-kinase inhibitors, no other therapeutic options are available. Therefore, it is mandatory to identify novel molecules and/or strategies to overcome the inevitable resistance. In this context, after promising preclinical data on the novel PI3K inhibitor BYL719, the NCT01735968 trial in GIST patients who had previously failed treatment with imatinib and sunitinib started. BYL719 has attracted our attention, and we comprehensively characterized genomic and transcriptomic changes taking place during resistance. Methods: For this purpose, we generated two in vitro GIST models of acquired resistance to BYL719 and performed an omic-based analysis by integrating RNA-sequencing, miRNA, and methylation profiles in sensitive and resistant cells. Results: We identified novel epigenomic mechanisms of pharmacological resistance in GISTs suggesting the existence of pathways involved in drug resistance and alternatively acquired mutations. Therefore, epigenomics should be taken into account as an alternative adaptive mechanism. Conclusion: Despite the fact that currently we do not have patients in treatment with BYL719 to verify this hypothesis, the most intriguing result is the involvement of H19 and PSTA1 in GIST resistance, which might represent druggable targets.
Collapse
Affiliation(s)
- Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giulia Sammarini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sebastian Moran
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institue (Idibell), l'Hospitalet de Llobregat, Barcelona, Spain
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Valentina Indio
- Giorgio Prodi Cancer Research Center, University of Bologna, Bologna, Italy
| | - Milena Urbini
- Giorgio Prodi Cancer Research Center, University of Bologna, Bologna, Italy
| | - Annalisa Astolfi
- Giorgio Prodi Cancer Research Center, University of Bologna, Bologna, Italy
| | - Federica Zanotti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Maria A Pantaleo
- Giorgio Prodi Cancer Research Center, University of Bologna, Bologna, Italy.,Department of Specialized, Experimental, and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
10
|
Abstract
Treatment of certain central nervous system disorders, including different types of cerebral malignancies, is limited by traditional oral or systemic administrations of therapeutic drugs due to possible serious side effects and/or lack of the brain penetration and, therefore, the efficacy of the drugs is diminished. During the last decade, several new technologies were developed to overcome barrier properties of cerebral capillaries. This review gives a short overview of the structural elements and anatomical features of the blood–brain barrier. The various in vitro (static and dynamic), in vivo (microdialysis), and in situ (brain perfusion) blood–brain barrier models are also presented. The drug formulations and administration options to deliver molecules effectively to the central nervous system (CNS) are presented. Nanocarriers, nanoparticles (lipid, polymeric, magnetic, gold, and carbon based nanoparticles, dendrimers, etc.), viral and peptid vectors and shuttles, sonoporation and microbubbles are briefly shown. The modulation of receptors and efflux transporters in the cell membrane can also be an effective approach to enhance brain exposure to therapeutic compounds. Intranasal administration is a noninvasive delivery route to bypass the blood–brain barrier, while direct brain administration is an invasive mode to target the brain region with therapeutic drug concentrations locally. Nowadays, both technological and mechanistic tools are available to assist in overcoming the blood–brain barrier. With these techniques more effective and even safer drugs can be developed for the treatment of devastating brain disorders.
Collapse
|
11
|
Wang Y, Sun H, Xiao Z, Zhang G, Zhang D, Bao X, Li F, Wu S, Gao Y, Wei N. DNA damage and apoptosis induced by a potent orally podophyllotoxin derivative in breast cancer. Cell Commun Signal 2018; 16:52. [PMID: 30176902 PMCID: PMC6122736 DOI: 10.1186/s12964-018-0263-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Targeting TopoisomeraseII (TopoII) and generate enzyme mediated DNA damage is an effective strategy for treatment of breast cancer. TopoII is known as a validated target for drug discovery and cancer chemotherapy. METHODS XWL-1-48, a new orally podophyllotoxin derivative, was designed and synthesized. The effect of XWL-1-48 on TopoII binding and activity was determined by molecular docking software and kDNA-decatenation assay, respectively. In vitro and in vivo breast cancer models were used to document the antitumor activity of XWL-1-48. Cellular apoptosis, cell cycle and ROS were analyzed by flow cytometry. Alteration of XWL-1-48-mediated downstream pathways was determined by western blot analysis. RESULTS The cytotoxicity of XWL-1-48 is more potent than that of its congener GL331. Molecular docking demonstrated that XWL-1-48 could bind to TopoII through forming two strong hydrogen bonds and potential pi-pi interactions. Noticeably, XWL-1-48 exerts potent antitumor activity in in vitro and in vivo breast cancer model. Treatment with XWL-1-48 caused ROS generation and triggered DNA damage through induction of γ-H2AX and activation of ATM/p53/p21 pathway. Further studies showed that XWL-1-48 led to S-phase arrest and mitochondrial apoptosis. Meanwhile, XWL-1-48 significantly blocked PI3K/Akt/Mdm2 pathway and enhanced Mdm2 degradation. CONCLUSION XWL-1-48 may be a promising orally topoII inhibitor, its mechanisms are associated with suppression of TopoII, induction of DNA damage and apoptosis, blockage of PI3K/AKT/Mdm2 pathway.
Collapse
Affiliation(s)
- Yajie Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hua Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China.
| | - Zhiyan Xiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Gang Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Xiuqi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Fangfang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Shaoyu Wu
- Guangdong Province Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, China
| | - Yuanchao Gao
- Beijing Tsinghua Changgeng Hospital, Beijing, China
| | - Ning Wei
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Groba SR, Guttmann S, Niemietz C, Bernick F, Sauer V, Hachmöller O, Karst U, Zischka H, Zibert A, Schmidt HH. Downregulation of hepatic multi-drug resistance protein 1 (MDR1) after copper exposure. Metallomics 2018; 9:1279-1287. [PMID: 28805879 DOI: 10.1039/c7mt00189d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Copper homeostasis is strictly regulated in mammalian cells. We investigated the adaptation of hepatocytes after long-term copper exposure. Copper-resistant hepatoma HepG2 cell lines lacking ATP7B were generated. Growth, copper accumulation, gene expression, and transport were determined. Hepatocyte-like cells derived from a Wilson disease (WD) patient and the liver of a WD animal model were also studied. The rapidly gained copper resistance was found to be stable, as subculturing of cells in the absence of added copper (weaning) did not restore copper sensitivity. Intracellular copper levels and the expression of MT1 and HSP70 were increased, whereas the expression of CTR1 was reduced. However, the values normalized after weaning. In contrast, downregulation of multi-drug resistance protein 1 (MDR1), encoding P-glycoprotein (P-gp), was shown to be permanent. Calcein assays confirmed the downregulation of MDR1 in the resistant cell lines. MDR1 knockdown by siRNA resulted in increased copper resistance and decreased intracellular copper. Treatment of the resistant cells with verapamil, a known inducer of MDR1, was followed by increased copper-induced toxicity. Downregulation of MDR1 was also observed in hepatocyte-like cells derived from a WD patient after copper exposure. In addition, MDR1 was downregulated in Long-Evans Cinnamon rats when the liver copper was elevated. The results indicate that downregulation of MDR1 is an adaptation of hepatic cells after sustained copper exposure when ATP7B is non-functional. Our data add to the versatile functions of MDR1 in the hepatocyte and may have an impact on the treatment of copper-related diseases, prominently WD.
Collapse
Affiliation(s)
- Sara Reinartz Groba
- Klinik für Transplantationsmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, 48149 Münster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Maia RC, Vasconcelos FC, Souza PS, Rumjanek VM. Towards Comprehension of the ABCB1/P-Glycoprotein Role in Chronic Myeloid Leukemia. Molecules 2018; 23:molecules23010119. [PMID: 29316665 PMCID: PMC6017716 DOI: 10.3390/molecules23010119] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/25/2017] [Accepted: 01/05/2018] [Indexed: 12/22/2022] Open
Abstract
Abstract: The introduction of imatinib (IM), a BCR-ABL1 tyrosine kinase inhibitor (TKI), has represented a significant advance in the first-line treatment of chronic myeloid leukemia (CML). However, approximately 30% of patients need to discontinue IM due to resistance or intolerance to this drug. Both resistance and intolerance have also been observed in treatment with the second-generation TKIs-dasatinib, nilotinib, and bosutinib-and the third-generation TKI-ponatinib. The mechanisms of resistance to TKIs may be BCR-ABL1-dependent and/or BCR-ABL1-independent. Although the role of efflux pump P-glycoprotein (Pgp), codified by the ABCB1 gene, is unquestionable in drug resistance of many neoplasms, a longstanding question exists about whether Pgp has a firm implication in TKI resistance in the clinical scenario. The goal of this review is to offer an overview of ABCB1/Pgp expression/activity/polymorphisms in CML. Understanding how interactions, associations, or cooperation between Pgp and other molecules-such as inhibitor apoptosis proteins, microRNAs, or microvesicles-impact IM resistance risk may be critical in evaluating the response to TKIs in CML patients. In addition, new non-TKI compounds may be necessary in order to overcome the resistance mediated by Pgp in CML.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/physiology
- Animals
- Drug Resistance, Neoplasm
- Genetic Predisposition to Disease
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Polymorphism, Single Nucleotide
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
Collapse
Affiliation(s)
- Raquel C Maia
- Laboratório de Hemato-Oncologia Celular e Molecular and Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Praça da Cruz Vermelha, 23, 6° andar, CEP 20230-130 Rio de Janeiro, Brazil.
| | - Flavia C Vasconcelos
- Laboratório de Hemato-Oncologia Celular e Molecular and Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Praça da Cruz Vermelha, 23, 6° andar, CEP 20230-130 Rio de Janeiro, Brazil.
| | - Paloma S Souza
- Laboratório de Hemato-Oncologia Celular e Molecular and Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Praça da Cruz Vermelha, 23, 6° andar, CEP 20230-130 Rio de Janeiro, Brazil.
| | - Vivian M Rumjanek
- Laboratório de Imunologia Tumoral, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Cidade Universitária, CEP 21941-902 Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Prokocimer M, Molchadsky A, Rotter V. Dysfunctional diversity of p53 proteins in adult acute myeloid leukemia: projections on diagnostic workup and therapy. Blood 2017; 130:699-712. [PMID: 28607134 PMCID: PMC5659817 DOI: 10.1182/blood-2017-02-763086] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022] Open
Abstract
The heterogeneous nature of acute myeloid leukemia (AML) and its poor prognosis necessitate therapeutic improvement. Current advances in AML research yield important insights regarding AML genetic, epigenetic, evolutional, and clinical diversity, all in which dysfunctional p53 plays a key role. As p53 is central to hematopoietic stem cell functions, its aberrations affect AML evolution, biology, and therapy response and usually predict poor prognosis. While in human solid tumors TP53 is mutated in more than half of cases, TP53 mutations occur in less than one tenth of de novo AML cases. Nevertheless, wild-type (wt) p53 dysfunction due to nonmutational p53 abnormalities appears to be rather frequent in various AML entities, bearing, presumably, a greater impact than is currently appreciated. Hereby, we advocate assessment of adult AML with respect to coexisting p53 alterations. Accordingly, we focus not only on the effects of mutant p53 oncogenic gain of function but also on the mechanisms underlying nonmutational wtp53 inactivation, which might be of therapeutic relevance. Patient-specific TP53 genotyping with functional evaluation of p53 protein may contribute significantly to the precise assessment of p53 status in AML, thus leading to the tailoring of a rationalized and precision p53-based therapy. The resolution of the mechanisms underlying p53 dysfunction will better address the p53-targeted therapies that are currently considered for AML. Additionally, a suggested novel algorithm for p53-based diagnostic workup in AML is presented, aiming at facilitating the p53-based therapeutic choices.
Collapse
MESH Headings
- Adult
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- DNA Damage/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Genomic Instability/drug effects
- Hematopoiesis/drug effects
- Humans
- Karyopherins/genetics
- Karyopherins/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Targeted Therapy/methods
- Mutation/drug effects
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nucleophosmin
- Protein Interaction Maps/drug effects
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Translocation, Genetic
- Tumor Suppressor Protein p53/analysis
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Exportin 1 Protein
Collapse
Affiliation(s)
- Miron Prokocimer
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Alina Molchadsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
15
|
Miyata KI, Nakagawa Y, Kimura Y, Ueda K, Akamatsu M. Structure–activity relationships of dibenzoylhydrazines for the inhibition of P-glycoprotein-mediated quinidine transport. Bioorg Med Chem 2016; 24:3184-91. [DOI: 10.1016/j.bmc.2016.05.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/18/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022]
|
16
|
Yu X, Yang G, Shi Y, Su C, Liu M, Feng B, Zhao L. Intracellular targeted co-delivery of shMDR1 and gefitinib with chitosan nanoparticles for overcoming multidrug resistance. Int J Nanomedicine 2015; 10:7045-56. [PMID: 26648717 PMCID: PMC4648604 DOI: 10.2147/ijn.s92436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nowadays, multidrug resistance and side effects of drugs limit the effectiveness of chemotherapies in clinics. P-glycoprotein (P-gp) (MDR1), as a member of the ATP-binding cassette family, acts on transporting drugs into cell plasma across the membrane of cancer cells and leads to the occurrence of multidrug resistance, thus resulting in the failure of chemotherapy in cancer. The main aims of this research were to design a nanodelivery system for accomplishing the effective co-delivery of gene and antitumor drug and overcoming multidrug resistance effect. In this study, shMDR1 and gefitinib-encapsulating chitosan nanoparticles with sustained release, small particle size, and high encapsulation efficiency were prepared. The serum stability, protection from nuclease, and transfection efficiency of gene in vitro were investigated. The effects of co-delivery of shMDR1 and gefitinib in nanoparticles on reversing multidrug resistance were also evaluated by investigating the cytotoxicity, cellular uptake mechanism, and cell apoptosis on established gefitinib-resistant cells. The results demonstrated that chitosan nanoparticles entrapping gefitinib and shMDR1 had the potential to overcome the multidrug resistance and improve cancer treatment efficacy, especially toward resistant cells.
Collapse
Affiliation(s)
- Xiwei Yu
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Guang Yang
- Department of Oncology, BenQ Medical Center, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yijie Shi
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Chang Su
- School of Veterinary Medicine, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Ming Liu
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Bo Feng
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Liang Zhao
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| |
Collapse
|