1
|
Belhadj Z, Akther T, Wang Z, Xie J. Characterization of a deazaflavin analog as a potent inhibitor of multidrug resistance-associated protein 1. Biomed Pharmacother 2024; 178:117167. [PMID: 39032285 DOI: 10.1016/j.biopha.2024.117167] [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: 02/14/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
Selective inhibition of overexpressed ATP binding cassette (ABC) transporters is an attractive approach to enhancing the efficacy of chemotherapeutics in multidrug resistant cancers. Previously, we reported that the cancer sensitizing effect of deazaflavin analogs, an important chemotype for developing combination treatments with topoisomerase II (TOP2) poisons, is associated with increased intracellular drug accumulation. Here we report the characterization of ZW-1226, a deazaflavin analog, as a potent inhibitor of multidrug resistance-associated protein 1 (MRP1). Specifically, ZW-1226 inhibited MRP1 with a 16-fold higher potency than the most widely used positive control MK-571 in vesicular transport assay and displayed excellent selectivity indices exceeding 100 over other major ABC transporters, including P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), MRP2 and MRP3. Mechanistically, we revealed that its MRP1 inhibitory action requires the participation of GSH. In chemo-sensitization test, ZW-1226 fully reversed the MRP1-mediated drug resistance to TOP2 poisons etoposide (ETP) and doxorubicin (DOX) in H69AR cells and conferred CC50s comparable to those in the sensitive parental NCI-H69 cells. The sensitization was associated with boosted intracellular accumulation of ETP and DOX and elevated endogenous GSH. Moreover, ZW-1226 showed potential to occupy the leukotriene C4 binding site in molecular docking with bovine MRP1, presumably with the help of GSH. Lastly, ZW-1226 exhibited high tissue to plasma partitions in mice but did not alter ETP distribution to normal tissues, suggesting it could be a viable lead with desirable pharmacokinetic properties to warrant further investigation.
Collapse
Affiliation(s)
- Zakia Belhadj
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thamina Akther
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
2
|
Yi X, Hussain I, Zhang P, Xiao C. Nuclear-Targeting Peptides for Cancer Therapy. Chembiochem 2024:e202400596. [PMID: 39215136 DOI: 10.1002/cbic.202400596] [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: 07/15/2024] [Revised: 08/19/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Nucleus is the central regulator of cells that controls cell proliferation, metabolism, and cell cycle, and is considered the most important organelle in cells. The precision medicine that can achieve nuclear targeting has achieved good therapeutic effects in anti-tumor therapy. However, the presence of biological barriers such as cell membranes and nuclear membranes in cells limit the delivery of therapeutic agents to the nucleus. Therefore, developing effective nuclear-targeting drug delivery strategies is particularly important. Nuclear-targeting peptides are a class of functional peptides that can penetrate cell membranes and target the nucleus. They mainly recognize and bind to the nuclear transport molecules (such as Importin-α/β) and transport the therapeutic agents to the nucleus through nuclear pore complexes (NPC). This review summarizes the most recent developments of strategies for anti-tumor therapy utilizing nuclear-targeting peptides, which will ultimately contribute to the development of more effective nuclear-targeting strategies to achieve better anti-tumor outcomes.
Collapse
Affiliation(s)
- Xuan Yi
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| | - Irshad Hussain
- Department of Chemistry and Chemical Engineering, SBA School of Science & Engineering, Lahore University of Management Sciences (LUMS). DHA, Lahore, 54792, Pakistan
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| |
Collapse
|
3
|
Kruschel RD, Barbosa MG, Almeida MJ, Xavier CPR, Vasconcelos MH, McCarthy FO. Discovery of Potent Isoquinolinequinone N-Oxides to Overcome Cancer Multidrug Resistance. J Med Chem 2024; 67:13909-13924. [PMID: 39093920 PMCID: PMC11345829 DOI: 10.1021/acs.jmedchem.4c00705] [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: 03/26/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
Multidrug resistance (MDR) of human tumors has resulted in an immediate need to develop appropriate new drugs. This work outlines the development of 20 potent IQQ N-oxide derivatives in two isomeric families, both exhibiting nanomolar GI50 against human tumor cell lines. Preliminary NCI-60 tumor screening sees the C(6) isomers achieve a mean GI50 > 2 times lower than the corresponding C(7) isomers. MDR evaluation of nine selected compounds reveals that each presents lower GI50 concentrations in two MDR tumor cell lines. Four of the series display nanomolar GI50 values against MDR cells, having selectivity ratios up to 2.7 versus the sensitive (parental) cells. The most potent compound 25 inhibits the activity of drug efflux pumps in MDR cells, causes significant ROS accumulation, and potently inhibits cell proliferation, causing alterations in the cell cycle profile. Our findings are confirmed by 3D spheroid models, providing new candidates for studies against MDR cancers.
Collapse
Affiliation(s)
- Ryan D. Kruschel
- School
of Chemistry, Analytical and Biological Chemistry Research Facility, University College Cork, Cork T12 K8AF, Ireland
| | - Mélanie
A. G. Barbosa
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
- FFUP−Faculty
of Pharmacy of the University of Porto, 4050-313 Porto Portugal
| | - Maria João Almeida
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
| | - Cristina P. R. Xavier
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
| | - M. Helena Vasconcelos
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
- FFUP−Faculty
of Pharmacy of the University of Porto, 4050-313 Porto Portugal
| | - Florence O. McCarthy
- School
of Chemistry, Analytical and Biological Chemistry Research Facility, University College Cork, Cork T12 K8AF, Ireland
| |
Collapse
|
4
|
Braconi L, Riganti C, Parenti A, Cecchi M, Nocentini A, Bartolucci G, Menicatti M, Contino M, Colabufo NA, Manetti D, Romanelli MN, Supuran CT, Teodori E. Dual Inhibitors of P-gp and Carbonic Anhydrase XII (hCA XII) against Tumor Multidrug Resistance with Piperazine Scaffold. Molecules 2024; 29:3290. [PMID: 39064868 PMCID: PMC11279465 DOI: 10.3390/molecules29143290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
A new series of piperazine derivatives were synthesized and studied with the aim of obtaining dual inhibitors of P-glycoprotein (P-gp) and carbonic anhydrase XII (hCA XII) to synergistically overcome the P-gp-mediated multidrug resistance (MDR) in cancer cells expressing the two proteins, P-gp and hCA XII. Indeed, these hybrid compounds contain both P-gp and hCA XII binding groups on the two nitrogen atoms of the heterocyclic ring. All compounds showed good inhibitory activity on each protein (P-gp and hCA XII) studied individually, and many of them showed a synergistic effect in the resistant HT29/DOX and A549/DOX cell lines which overexpress both the target proteins. In particular, compound 33 displayed the best activity by enhancing the cytotoxicity and intracellular accumulation of doxorubicin in HT29/DOX and A549/DOX cells, thus resulting as promising P-gp-mediated MDR reverser with a synergistic mechanism. Furthermore, compounds 13, 27 and 32 induced collateral sensitivity (CS) in MDR cells, as they were more cytotoxic in resistant cells than in the sensitive ones; their CS mechanisms were extensively investigated.
Collapse
Affiliation(s)
- Laura Braconi
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| | - Chiara Riganti
- Oncological Pharmacology Laboratory and Molecular Biotechnology Center “Guido Tarone”, Department of Oncology, University of Turin, Piazza Nizza 44, 10126 Torino, Italy;
| | - Astrid Parenti
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Marta Cecchi
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Alessio Nocentini
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| | - Gianluca Bartolucci
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| | - Marta Menicatti
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| | - Marialessandra Contino
- Department of Pharmacy-Drug Sciences, University of Bari “A. Moro”, via Orabona 4, 70125 Bari, Italy; (M.C.); (N.A.C.)
| | - Nicola Antonio Colabufo
- Department of Pharmacy-Drug Sciences, University of Bari “A. Moro”, via Orabona 4, 70125 Bari, Italy; (M.C.); (N.A.C.)
| | - Dina Manetti
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| | - Maria Novella Romanelli
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| | - Claudiu T. Supuran
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| | - Elisabetta Teodori
- Section of Pharmaceutical and Nutraceutical Sciences, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (A.N.); (G.B.); (M.M.); (D.M.); (M.N.R.); (C.T.S.)
| |
Collapse
|
5
|
Featherston T, Paumann-Page M, Hampton MB. Melanoma redox biology and the emergence of drug resistance. Adv Cancer Res 2024; 162:145-171. [PMID: 39069368 DOI: 10.1016/bs.acr.2024.06.004] [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] [Indexed: 07/30/2024]
Abstract
Melanoma is the deadliest form of skin cancer, with the loss of approximately 60,000 lives world-wide each year. Despite the development of targeted therapeutics, including compounds that have selectivity for mutant oncoproteins expressed only in cancer cells, many patients are either unresponsive to initial therapy or their tumors acquire resistance. This results in five-year survival rates of below 25%. New strategies that either kill drug-resistant melanoma cells or prevent their emergence would be extremely valuable. Melanoma, like other cancers, has long been described as being under increased oxidative stress, resulting in an increased reliance on antioxidant defense systems. Changes in redox homeostasis are most apparent during metastasis and during the metabolic reprogramming associated with the development of treatment resistance. This review discusses oxidative stress in melanoma, with a particular focus on targeting antioxidant pathways to limit the emergence of drug resistant cells.
Collapse
Affiliation(s)
- Therese Featherston
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Martina Paumann-Page
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| | - Mark B Hampton
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
6
|
Milunovic MM, Ohui K, Besleaga I, Petrasheuskaya TV, Dömötör O, Enyedy ÉA, Darvasiova D, Rapta P, Barbieriková Z, Vegh D, Tóth S, Tóth J, Kucsma N, Szakács G, Popović-Bijelić A, Zafar A, Reynisson J, Shutalev AD, Bai R, Hamel E, Arion VB. Copper(II) Complexes with Isomeric Morpholine-Substituted 2-Formylpyridine Thiosemicarbazone Hybrids as Potential Anticancer Drugs Inhibiting Both Ribonucleotide Reductase and Tubulin Polymerization: The Morpholine Position Matters. J Med Chem 2024; 67:9069-9090. [PMID: 38771959 PMCID: PMC11181322 DOI: 10.1021/acs.jmedchem.4c00259] [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: 01/29/2024] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 05/23/2024]
Abstract
The development of copper(II) thiosemicarbazone complexes as potential anticancer agents, possessing dual functionality as inhibitors of R2 ribonucleotide reductase (RNR) and tubulin polymerization by binding at the colchicine site, presents a promising avenue for enhancing therapeutic effectiveness. Herein, we describe the syntheses and physicochemical characterization of four isomeric proligands H2L3-H2L6, with the methylmorpholine substituent at pertinent positions of the pyridine ring, along with their corresponding Cu(II) complexes 3-6. Evidently, the position of the morpholine moiety and the copper(II) complex formation have marked effects on the in vitro antiproliferative activity in human uterine sarcoma MES-SA cells and the multidrug-resistant derivative MES-SA/Dx5 cells. Activity correlated strongly with quenching of the tyrosyl radical (Y•) of mouse R2 RNR protein, inhibition of RNR activity in the cancer cells, and inhibition of tubulin polymerization. Insights into the mechanism of antiproliferative activity, supported by experimental results and molecular modeling calculations, are presented.
Collapse
Affiliation(s)
| | - Katerina Ohui
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
| | - Iuliana Besleaga
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
| | - Tatsiana V. Petrasheuskaya
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Orsolya Dömötör
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Éva A. Enyedy
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Denisa Darvasiova
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Peter Rapta
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Zuzana Barbieriková
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Daniel Vegh
- Institute
of Organic Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Bratislava SK-81237, Slovakia
| | - Szilárd Tóth
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Judit Tóth
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Nóra Kucsma
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Gergely Szakács
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
- Center
for Cancer Research, Medical University
of Vienna, Vienna A-1090, Austria
| | - Ana Popović-Bijelić
- Faculty
of Physical Chemistry, University of Belgrade, Belgrade 11158, Serbia
| | - Ayesha Zafar
- School
of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jóhannes Reynisson
- School
of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, United
Kingdom
| | - Anatoly D. Shutalev
- N.
D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119991, Russian Federation
| | - Ruoli Bai
- Molecular
Pharmacology Branch, Developmental Therapeutics Program, Division
of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick
National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Ernest Hamel
- Molecular
Pharmacology Branch, Developmental Therapeutics Program, Division
of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick
National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Vladimir B. Arion
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
- Inorganic
Polymers Department, “Petru Poni”
Institute of Macromolecular Chemistry, Aleea Gr. Ghica Voda 41 A, Iasi 700487, Romania
| |
Collapse
|
7
|
Kovács H, Jakusch T, May NV, Tóth S, Szakács G, Enyedy ÉA. Complex formation of ML324, the histone demethylase inhibitor, with essential metal ions: Relationship between solution chemistry and anticancer activity. J Inorg Biochem 2024; 255:112540. [PMID: 38552361 DOI: 10.1016/j.jinorgbio.2024.112540] [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: 12/20/2023] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/16/2024]
Abstract
N-(3-(dimethylamino)propyl-4-(8-hydroxyquinolin-6-yl)benzamide (ML324, HL) is a potent inhibitor of the iron-containing histone demethylase KDM4, a recognized potential target of cancer therapeutics. Herein, we report the proton dissociation and complex formation processes of ML324 with essential metal ions such as Fe(II), Fe(III), Cu(II) and Zn(II) using UV-visible, fluorescence, electron paramagnetic resonance and 1H NMR spectroscopic methods. The electrochemical behaviour of the copper and iron complexes was characterized by cyclic voltammetry and spectroelectrochemistry. The solid phase structure of ML324 analysed by X-ray crystallography is also provided. Based on the solution equilibrium data, ML324 is present in solution in H2L+ form with a protonated dimethylammonium moiety at pH 7.4, and this (N,O) donor bearing ligand forms mono and bis complexes with all the studied metal ions and the tris-ligand species is also observed with Fe(III). At pH 7.4 the metal binding ability of ML324 follows the order: Fe(II) < Zn(II) < Cu(II) < Fe(III). Complexation with iron resulted in a negative redox potential (E'1/2 = -145 mV vs. NHE), further suggesting that the ligand has a preference for Fe(III) over Fe(II). ML324 was tested for its anticancer activity in chemosensitive and resistant human cancer cells overexpressing the efflux pump P-glycoprotein. ML324 exerted similar activity in all tested cells (IC50 = 1.9-3.6 μM). Co-incubation and complexation of the compound with Cu(II) and Zn(II) had no impact on the cytotoxicity of ML324, whereas Fe(III) decreased the toxicity in a concentration-dependent manner, and this effect was more pronounced in the multidrug resistant cells.
Collapse
Affiliation(s)
- Hilda Kovács
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary; Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary
| | - Tamás Jakusch
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary
| | - Nóra V May
- Centre for Structural Science, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Szilárd Tóth
- Drug Resistance Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar Tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Gergely Szakács
- Drug Resistance Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar Tudósok krt. 2, H-1117 Budapest, Hungary; Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary; Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary.
| |
Collapse
|
8
|
Podolski-Renić A, Čipak Gašparović A, Valente A, López Ó, Bormio Nunes JH, Kowol CR, Heffeter P, Filipović NR. Schiff bases and their metal complexes to target and overcome (multidrug) resistance in cancer. Eur J Med Chem 2024; 270:116363. [PMID: 38593587 DOI: 10.1016/j.ejmech.2024.116363] [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: 02/27/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Overcoming multidrug resistance (MDR) is one of the major challenges in cancer therapy. In this respect, Schiff base-related compounds (bearing a R1R2CNR3 bond) gained high interest during the past decades. Schiff bases are considered privileged ligands for various reasons, including the easiness of their preparation and the possibility to form complexes with almost all transition metal ions. Schiff bases and their metal complexes exhibit many types of biological activities and are used for the treatment and diagnosis of various diseases. Until now, 13 Schiff bases have been investigated in clinical trials for cancer treatment and hypoxia imaging. This review represents the first collection of Schiff bases and their complexes which demonstrated MDR-reversal activity. The areas of drug resistance covered in this article involve: 1) Modulation of ABC transporter function, 2) Targeting lysosomal ABCB1 overexpression, 3) Circumvention of ABC transporter-mediated drug efflux by alternative routes of drug uptake, 4) Selective activity against MDR cancer models (collateral sensitivity), 5) Targeting GSH-detoxifying systems, 6) Overcoming apoptosis resistance by inducing necrosis and paraptosis, 7) Reactivation of mutated p53, 8) Restoration of sensitivity to DNA-damaging anticancer therapy, and 9) Overcoming drug resistance through modulation of the immune system. Through this approach, we would like to draw attention to Schiff bases and their metal complexes representing highly interesting anticancer drug candidates with the ability to overcome MDR.
Collapse
Affiliation(s)
- Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | | | - Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Óscar López
- Departamento de Química Organica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Julia H Bormio Nunes
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Nenad R Filipović
- Department of Chemistry and Biochemistry, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
9
|
Yang GZ, Wang L, Gao K, Zhu X, Lou LG, Yue JM. Design and Synthesis of Cyclolipopeptide Mimics of Dysoxylactam A and Evaluation of the Reversing Potencies against P-Glycoprotein-Mediated Multidrug Resistance. J Med Chem 2024. [PMID: 38502936 DOI: 10.1021/acs.jmedchem.3c01920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Inspired by the structure of dysoxylactam A (DLA) that has been demonstrated to reverse P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) effectively, 61 structurally simplified cyclolipopeptides were thus designed and synthesized via an effective method, and their reversing P-gp-mediated MDR potentials were evaluated, which provided a series of more potent analogues and allowed us to explore their structure-activity relationship (SAR). Among them, a well-simplified compound, 56, with only two chiral centers that all derived from amino acids dramatically reversed drug resistance in KBV200 cells at 10 μM in combination with vinorelbine (VNR), paclitaxel (PTX), and adriamycin (ADR), respectively, which is more promising than DLA. The mechanism study showed that 56 reversed the MDR of tumor cells by inhibiting the transport function of P-gp rather than reducing its expression. Notably, compound 56 effectively restored the sensitivity of MDR tumors to VNR in vivo at a dosage without obvious toxicity.
Collapse
Affiliation(s)
- Guan-Zhou Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Lei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Kun Gao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xi Zhu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Li-Guang Lou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Jian-Min Yue
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
- Research Units of Discovery of New Drug Lead Molecules, Chinese Academy of Medical Sciences, Shanghai 201203, People's Republic of China
| |
Collapse
|
10
|
Maltas J, Tadele DS, Durmaz A, McFarland CD, Hinczewski M, Scott JG. Frequency-dependent ecological interactions increase the prevalence, and shape the distribution, of pre-existing drug resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.16.533001. [PMID: 36993678 PMCID: PMC10055114 DOI: 10.1101/2023.03.16.533001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The evolution of resistance remains one of the primary challenges for modern medicine from infectious diseases to cancers. Many of these resistance-conferring mutations often carry a substantial fitness cost in the absence of treatment. As a result, we would expect these mutants to undergo purifying selection and be rapidly driven to extinction. Nevertheless, pre-existing resistance is frequently observed from drug-resistant malaria to targeted cancer therapies in non-small cell lung cancer (NSCLC) and melanoma. Solutions to this apparent paradox have taken several forms from spatial rescue to simple mutation supply arguments. Recently, in an evolved resistant NSCLC cell line, we found that frequency-dependent ecological interactions between ancestor and resistant mutant ameliorate the cost of resistance in the absence of treatment. Here, we hypothesize that frequency-dependent ecological interactions in general play a major role in the prevalence of pre-existing resistance. We combine numerical simulations with robust analytical approximations to provide a rigorous mathematical framework for studying the effects of frequency-dependent ecological interactions on the evolutionary dynamics of pre-existing resistance. First, we find that ecological interactions significantly expand the parameter regime under which we expect to observe pre-existing resistance. Next, even when positive ecological interactions between mutants and ancestors are rare, these resistant clones provide the primary mode of evolved resistance because even weak positive interaction leads to significantly longer extinction times. We then find that even in the case where mutation supply alone is sufficient to predict pre-existing resistance, frequency-dependent ecological forces still contribute a strong evolutionary pressure that selects for increasingly positive ecological effects (negative frequency-dependent selection). Finally, we genetically engineer several of the most common clinically observed resistance mechanisms to targeted therapies in NSCLC, a treatment notorious for pre-existing resistance. We find that each engineered mutant displays a positive ecological interaction with their ancestor. As a whole, these results suggest that frequency-dependent ecological effects can play a crucial role in shaping the evolutionary dynamics of pre-existing resistance.
Collapse
Affiliation(s)
- Jeff Maltas
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
- Case Western Reserve University, School of Medicine, Cleveland, OH
| | - Dagim Shiferaw Tadele
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
- Oslo University Hospital, Ullevål, Department of Medical Genetics, Oslo, Norway
| | - Arda Durmaz
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
| | - Christopher D. McFarland
- Case Western Reserve University, School of Medicine, Cleveland, OH
- Case Comprehensive Cancer Center, Cleveland, OH
| | | | - Jacob G. Scott
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
- Case Western Reserve University, School of Medicine, Cleveland, OH
- Case Western Reserve University, Department of Physics, Cleveland, OH
- Case Comprehensive Cancer Center, Cleveland, OH
| |
Collapse
|
11
|
Chen Q, Fang C, Xia F, Wang Q, Li F, Ling D. Metal nanoparticles for cancer therapy: Precision targeting of DNA damage. Acta Pharm Sin B 2024; 14:1132-1149. [PMID: 38486992 PMCID: PMC10934341 DOI: 10.1016/j.apsb.2023.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/30/2023] [Accepted: 08/15/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer, a complex and heterogeneous disease, arises from genomic instability. Currently, DNA damage-based cancer treatments, including radiotherapy and chemotherapy, are employed in clinical practice. However, the efficacy and safety of these therapies are constrained by various factors, limiting their ability to meet current clinical demands. Metal nanoparticles present promising avenues for enhancing each critical aspect of DNA damage-based cancer therapy. Their customizable physicochemical properties enable the development of targeted and personalized treatment platforms. In this review, we delve into the design principles and optimization strategies of metal nanoparticles. We shed light on the limitations of DNA damage-based therapy while highlighting the diverse strategies made possible by metal nanoparticles. These encompass targeted drug delivery, inhibition of DNA repair mechanisms, induction of cell death, and the cascading immune response. Moreover, we explore the pivotal role of physicochemical factors such as nanoparticle size, stimuli-responsiveness, and surface modification in shaping metal nanoparticle platforms. Finally, we present insights into the challenges and future directions of metal nanoparticles in advancing DNA damage-based cancer therapy, paving the way for novel treatment paradigms.
Collapse
Affiliation(s)
- Qian Chen
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunyan Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fan Xia
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiyue Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
| | - Daishun Ling
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| |
Collapse
|
12
|
Lei ZN, Albadari N, Teng QX, Rahman H, Wang JQ, Wu Z, Ma D, Ambudkar SV, Wurpel JND, Pan Y, Li W, Chen ZS. ABCB1-dependent collateral sensitivity of multidrug-resistant colorectal cancer cells to the survivin inhibitor MX106-4C. Drug Resist Updat 2024; 73:101065. [PMID: 38367548 DOI: 10.1016/j.drup.2024.101065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/28/2024] [Accepted: 02/02/2024] [Indexed: 02/19/2024]
Abstract
AIMS To investigate the collateral sensitivity (CS) of ABCB1-positive multidrug resistant (MDR) colorectal cancer cells to the survivin inhibitor MX106-4C and the mechanism. METHODS Biochemical assays (MTT, ATPase, drug accumulation/efflux, Western blot, RT-qPCR, immunofluorescence, flow cytometry) and bioinformatic analyses (mRNA-sequencing, reversed-phase protein array) were performed to investigate the hypersensitivity of ABCB1 overexpressing colorectal cancer cells to MX106-4C and the mechanisms. Synergism assay, long-term selection, and 3D tumor spheroid test were used to evaluate the anti-cancer efficacy of MX106-4C. RESULTS MX106-4C selectively killed ABCB1-positive colorectal cancer cells, which could be reversed by an ABCB1 inhibitor, knockout of ABCB1, or loss-of-function ABCB1 mutation, indicating an ABCB1 expression and function-dependent mechanism. MX106-4C's selective toxicity was associated with cell cycle arrest and apoptosis through ABCB1-dependent survivin inhibition and activation on caspases-3/7 as well as modulation on p21-CDK4/6-pRb pathway. MX106-4C had good selectivity against ABCB1-positive colorectal cancer cells and retained this in multicellular tumor spheroids. In addition, MX106-4C could exert a synergistic anti-cancer effect with doxorubicin or re-sensitize ABCB1-positive cancer cells to doxorubicin by reducing ABCB1 expression in the cell population via long-term exposure. CONCLUSIONS MX106-4C selectively kills ABCB1-positive MDR colorectal cancer cells via a novel ABCB1-dependent survivin inhibition mechanism, providing a clue for designing CS compound as an alternative strategy to overcome ABCB1-mediated colorectal cancer MDR.
Collapse
Affiliation(s)
- Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, PR China
| | - Najah Albadari
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Hadiar Rahman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zhongzhi Wu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dejian Ma
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | - John N D Wurpel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Yihang Pan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, PR China.
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
13
|
Dhanyamraju PK. Drug resistance mechanisms in cancers: Execution of pro-survival strategies. J Biomed Res 2024; 38:95-121. [PMID: 38413011 PMCID: PMC11001593 DOI: 10.7555/jbr.37.20230248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 02/29/2024] Open
Abstract
One of the quintessential challenges in cancer treatment is drug resistance. Several mechanisms of drug resistance have been described to date, and new modes of drug resistance continue to be discovered. The phenomenon of cancer drug resistance is now widespread, with approximately 90% of cancer-related deaths associated with drug resistance. Despite significant advances in the drug discovery process, the emergence of innate and acquired mechanisms of drug resistance has impeded the progress in cancer therapy. Therefore, understanding the mechanisms of drug resistance and the various pathways involved is integral to treatment modalities. In the present review, I discuss the different mechanisms of drug resistance in cancer cells, including DNA damage repair, epithelial to mesenchymal transition, inhibition of cell death, alteration of drug targets, inactivation of drugs, deregulation of cellular energetics, immune evasion, tumor-promoting inflammation, genome instability, and other contributing epigenetic factors. Furthermore, I highlight available treatment options and conclude with future directions.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
14
|
Liu Z, Cai J, Jiang G, Wang M, Wu C, Su K, Hu W, Huang Y, Yu C, Huang X, Cao G, Wang H. Novel Platinum(IV) complexes intervene oxaliplatin resistance in colon cancer via inducing ferroptosis and apoptosis. Eur J Med Chem 2024; 263:115968. [PMID: 37995563 DOI: 10.1016/j.ejmech.2023.115968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Platinum-based chemotherapeutics are widely used for cancer treatment but are frequently limited because of dosage-dependent side effects and drug resistance. To attenuate these drawbacks, a series of novel platinum(IV) prodrugs (15a-18c) were synthesized and evaluated for anti-cancer activity. Among them, 17a demonstrated superior anti-proliferative activity compared with oxaliplatin (OXA) in the cisplatin-resistant lung cancer cell line A549/CDDP and OXA-resistant colon cancer cell line HCT-116/OXA but showed a lower cytotoxic effect toward human normal cell lines HUVEC and L02. Mechanistic investigations suggested that 17a efficiently enhanced intracellular platinum accumulation, induced DNA damage, disturbed the homeostasis of intracellular reactive oxygen molecules and mitochondrial membrane potential, and thereby activated the mitochondrion-dependent apoptosis pathway. Moreover, 17a significantly induced ferroptosis in HCT-116/OXA via triggering the accumulation of lipid peroxides, disrupting iron homeostasis, and inhibiting solute carrier family 7 member 11 and glutathione peroxidase 4 axial pathway transduction by inhibiting the expression of the phosphorylated signal transducer and activator of transcription 3 and nuclear factor erythroid 2-related factor 2. Moreover, 17a exerted remarkable in vivo antitumor efficacy in the HCT-116/OXA xenograft models but showed attenuated toxicity. These results indicated that these novel platinum(IV) complexes provided an alternative strategy to develop novel platinum-based antineoplastic agents for cancer treatment.
Collapse
Affiliation(s)
- Zhikun Liu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Jinyuan Cai
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Guiyang Jiang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Meng Wang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China
| | - Chuang Wu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Kangning Su
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Weiwei Hu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Yaxian Huang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Chunhao Yu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Xiaochao Huang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| | - Guoxiu Cao
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China.
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| |
Collapse
|
15
|
Dutra JDP, Scheiffer G, Kronenberger T, Gomes LJC, Zanzarini I, dos Santos KK, Tonduru AK, Poso A, Rego FGDM, Picheth G, Valdameri G, Moure VR. Structural and molecular characterization of lopinavir and ivermectin as breast cancer resistance protein (BCRP/ABCG2) inhibitors. EXCLI JOURNAL 2023; 22:1155-1172. [PMID: 38204967 PMCID: PMC10776880 DOI: 10.17179/excli2023-6427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/02/2023] [Indexed: 01/12/2024]
Abstract
A current clinical challenge in cancer is multidrug resistance (MDR) mediated by ABC transporters. Breast cancer resistance protein (BCRP) or ABCG2 transporter is one of the most important ABC transporters implicated in MDR and the use of inhibitors is a promising approach to overcome the resistance in cancer. This study aimed to characterize the molecular mechanism of ABCG2 inhibitors identified by a repurposing drug strategy using antiviral, anti-inflammatory and antiparasitic agents. Lopinavir and ivermectin can be considered as pan-inhibitors of ABC transporters, since both compounds inhibited ABCG2, P-glycoprotein and MRP1. They inhibited ABCG2 activity showing IC50 values of 25.5 and 23.4 µM, respectively. These drugs were highly cytotoxic and not transported by ABCG2. Additionally, these drugs increased the 5D3 antibody binding and did not affect the mRNA and protein expression levels. Cell-based analysis of the type of inhibition suggested a non-competitive inhibition, which was further corroborated by in silico approaches of molecular docking and molecular dynamics simulations. These results showed an overlap of the lopinavir and ivermectin binding sites on ABCG2, mainly interacting with E446 residue. However, the substrate mitoxantrone occupies a different site, binding to the F436 region, closer to the L554/L555 plug. In conclusion, these results revealed the mechanistic basis of lopinavir and ivermectin interaction with ABCG2. See also the Graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Julia de Paula Dutra
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, Brazil
| | - Gustavo Scheiffer
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, Brazil
| | - Thales Kronenberger
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- (a) Department of Internal Medicine VIII, University Hospital Tuebingen, Otfried-Müller-Strasse 14, Tuebingen DE 72076, Germany, (b) Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-Universität, Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany, (c) Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany, (d) Tuebingen Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tuebingen, Germany
| | - Lucas Julian Cruz Gomes
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, Brazil
| | - Isadora Zanzarini
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, Brazil
| | - Kelly Karoline dos Santos
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, Brazil
| | - Arun K. Tonduru
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Antti Poso
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- (a) Department of Internal Medicine VIII, University Hospital Tuebingen, Otfried-Müller-Strasse 14, Tuebingen DE 72076, Germany, (b) Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-Universität, Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany, (c) Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany, (d) Tuebingen Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tuebingen, Germany
| | | | - Geraldo Picheth
- Graduate Program in Pharmaceutical Sciences, Federal University of Parana, Curitiba, PR, Brazil
| | - Glaucio Valdameri
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, Brazil
| | - Vivian Rotuno Moure
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, Brazil
| |
Collapse
|
16
|
Zhu J, Lei S, Lu J, Hao Y, Qian Q, Devanathan AS, Feng Z, Xie XQ, Wipf P, Ma X. Metabolism-guided development of Ko143 analogs as ABCG2 inhibitors. Eur J Med Chem 2023; 259:115666. [PMID: 37482017 PMCID: PMC10529637 DOI: 10.1016/j.ejmech.2023.115666] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
ATP-binding cassette subfamily G member 2 (ABCG2), an efflux transporter, is involved in multiple pathological processes. Ko143 is a potent ABCG2 inhibitor; however, it is quickly metabolized through carboxylesterase 1-mediated hydrolysis of its t-butyl ester moiety. The current work aimed to develop more metabolically stable ABCG2 inhibitors. Novel Ko143 analogs were designed and synthesized by replacing the unstable t-butyl ester moiety in Ko143 with an amide group. The synthesized Ko143 analogs were evaluated for their ABCG2 inhibitory activity, binding mode with ABCG2, cytotoxicity, and metabolic stability. We found that the amide modification of Ko143 led to metabolically stable ABCG2 inhibitors. Among these Ko143 analogs, K2 and K34 are promising candidates with favorable oral pharmacokinetic profiles in mice. In summary, we synthesized novel Ko143 analogs with improved metabolic stability, which can potentially be used as lead compounds for the future development of ABCG2 inhibitors.
Collapse
Affiliation(s)
- Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Saifei Lei
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Lu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yixuan Hao
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qi Qian
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aaron S Devanathan
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter Wipf
- Department of Chemistry and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
17
|
Teixeira R, Salaroglio IC, Oliveira NFB, Sequeira JGN, Fontrodona X, Romero I, Machuqueiro M, Tomaz AI, Garcia MH, Riganti C, Valente A. Fighting Multidrug Resistance with Ruthenium-Cyclopentadienyl Compounds: Unveiling the Mechanism of P-gp Inhibition. J Med Chem 2023; 66:14080-14094. [PMID: 37616241 PMCID: PMC10614197 DOI: 10.1021/acs.jmedchem.3c01120] [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: 06/21/2023] [Indexed: 08/26/2023]
Abstract
The search for more effective and selective drugs to overcome cancer multidrug resistance is urgent. As such, a new series of ruthenium-cyclopentadienyl ("RuCp") compounds with the general formula [Ru(η5-C5H4R)(4,4'-R'-2,2'-bipy)(PPh3)] were prepared and fully characterized. All compounds were evaluated toward non-small cell lung cancer cells with different degrees of cisplatin sensitivity (A549, NCI-H2228, Calu-3, and NCI-H1975), showing better cytotoxicity than the first-line chemotherapeutic drug cisplatin. Compounds 2 and 3 (R' = -OCH3; R = CHO (2) or CH2OH (3)) further inhibited the activity of P-gp and MRP1 efflux pumps by impairing their catalytic activity. Molecular docking calculations identified the R-site P-gp pocket as the preferred one, which was further validated using site-directed mutagenesis experiments in P-gp. Altogether, our results unveil the first direct evidence of the interaction between P-gp and "RuCp" compounds in the modulation of P-gp activity and establish them as valuable candidates to circumvent cancer MDR.
Collapse
Affiliation(s)
- Ricardo
G. Teixeira
- Centro
de Química Estrutural, Institute of Molecular Sciences and
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | | | - Nuno F. B. Oliveira
- BioISI:
Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - João G. N. Sequeira
- BioISI:
Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Xavier Fontrodona
- Departament
de Química and Serveis Tècnics de Recerca, Universitat de Girona, C/M. Aurèlia Campmany, 69, E-17003 Girona, Spain
| | - Isabel Romero
- Departament
de Química and Serveis Tècnics de Recerca, Universitat de Girona, C/M. Aurèlia Campmany, 69, E-17003 Girona, Spain
| | - Miguel Machuqueiro
- BioISI:
Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Ana Isabel Tomaz
- Centro
de Química Estrutural, Institute of Molecular Sciences and
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - M. Helena Garcia
- Centro
de Química Estrutural, Institute of Molecular Sciences and
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Chiara Riganti
- Department
of Oncology, University of Torino, 10126 Torino, Italy
- Molecular
Biotechnology Center “Guido Tarone”, University of Torino, 10126 Torino, Italy
| | - Andreia Valente
- Centro
de Química Estrutural, Institute of Molecular Sciences and
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
18
|
Liu Z, Feng Z, Chen M, Zhan J, Wu R, Shi Y, Xue Y, Liu R, Zhu JJ, Zhang J. An orthogonally activatable CRISPR-Cas13d nanoprodrug to reverse chemoresistance for enhanced chemo-photodynamic therapy. Chem Sci 2023; 14:4102-4113. [PMID: 37063792 PMCID: PMC10094006 DOI: 10.1039/d3sc00020f] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Orthogonal therapy that combines CRISPR-based gene editing and prodrug-based chemotherapy is a promising approach to combat multidrug-resistant cancer. However, its potency to precisely regulate different therapeutic modalities in vivo is limited due to the lack of an integrated platform with high spatiotemporal resolution. Taking advantage of CRISPR technology, a Pt(iv)-based prodrug and orthogonal emissive upconversion nanoparticles (UCNPs), we herein rationally designed the first logic-gated CRISPR-Cas13d-based nanoprodrug for orthogonal photomodulation of gene editing and prodrug release for enhanced cancer therapy. The nanoprodrug (URL) was constructed by encapsulating a green light-activatable Pt(iv) prodrug and UV light-activatable Cas13d gene editing tool into UCNPs. We demonstrated that URL maintained excellent orthogonal emission behaviors under 808 and 980 nm excitations, allowing wavelength-selective photoactivation of Cas13d and the prodrug for downregulation of the resistance-related gene and induction of chemo-photodynamic therapy, respectively. Moreover, the photomodulation superiority of URL for overcoming drug resistance was highlighted by integrating it with a Boolean logic gate for programmable modulation of multiple cell behaviors. Importantly, in vivo studies demonstrated that URL can promote Pt(iv) prodrug activation and ROS generation and massively induce on-target drug accumulation by Cas13d-mediated drug resistance attenuation, delivering an ultimate chemo-photodynamic therapeutic performance in efficiently eradicating primary tumors and preventing further liver metastasis. Collectively, our results suggest that URL expands the Cas13d-based genome editing toolbox into prodrug nanomedicine and accelerates the discovery of new orthogonal therapeutic approaches.
Collapse
Affiliation(s)
- Zheng Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Zhiyuan Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Mohan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Jiayin Zhan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Rong Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Yang Shi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Yunsheng Xue
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University Xuzhou 221004 China
| | - Ran Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Jingjing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| |
Collapse
|
19
|
Damiani D, Tiribelli M. ABCG2 in Acute Myeloid Leukemia: Old and New Perspectives. Int J Mol Sci 2023; 24:ijms24087147. [PMID: 37108308 PMCID: PMC10138346 DOI: 10.3390/ijms24087147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Despite recent advances, prognosis of acute myeloid leukemia (AML) remains unsatisfactory due to poor response to therapy or relapse. Among causes of resistance, over-expression of multidrug resistance (MDR) proteins represents a pivotal mechanism. ABCG2 is an efflux transporter responsible for inducing MDR in leukemic cells; through its ability to extrude many antineoplastic drugs, it leads to AML resistance and/or relapse, even if conflicting data have been reported to date. Moreover, ABCG2 may be co-expressed with other MDR-related proteins and is finely regulated by epigenetic mechanisms. Here, we review the main issues regarding ABCG2 activity and regulation in the AML clinical scenario, focusing on its expression and the role of polymorphisms, as well as on the potential ways to inhibit its function to counteract drug resistance to, eventually, improve outcomes in AML patients.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, P.le Santa Maria della Misericordia, 5, 33100 Udine, Italy
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, P.le Santa Maria della Misericordia, 5, 33100 Udine, Italy
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| |
Collapse
|
20
|
Lin-Rahardja K, Weaver DT, Scarborough JA, Scott JG. Evolution-Informed Strategies for Combating Drug Resistance in Cancer. Int J Mol Sci 2023; 24:6738. [PMID: 37047714 PMCID: PMC10095117 DOI: 10.3390/ijms24076738] [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: 02/27/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
The ever-changing nature of cancer poses the most difficult challenge oncologists face today. Cancer's remarkable adaptability has inspired many to work toward understanding the evolutionary dynamics that underlie this disease in hopes of learning new ways to fight it. Eco-evolutionary dynamics of a tumor are not accounted for in most standard treatment regimens, but exploiting them would help us combat treatment-resistant effectively. Here, we outline several notable efforts to exploit these dynamics and circumvent drug resistance in cancer.
Collapse
Affiliation(s)
- Kristi Lin-Rahardja
- Systems Biology & Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Davis T. Weaver
- Systems Biology & Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jessica A. Scarborough
- Systems Biology & Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jacob G. Scott
- Systems Biology & Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Translational Hematology & Oncology, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Semi-Synthetic Dihydrotestosterone Derivatives Modulate Inherent Multidrug Resistance and Sensitize Colon Cancer Cells to Chemotherapy. Pharmaceutics 2023; 15:pharmaceutics15020584. [PMID: 36839907 PMCID: PMC9966060 DOI: 10.3390/pharmaceutics15020584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/21/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Multidrug resistance (MDR) is a serious hurdle to successful cancer therapy. Here, we examined the efficiency of novel semi-synthetic dihydrotestosterone derivatives, more specifically androstano-arylpyrimidines in inhibiting the efflux activity of ATP-binding cassette (ABC) transporters and sensitizing inherently MDR colon cancer cells to various chemotherapy drugs. Using the Rhodamine123 accumulation assay, we evaluated the efflux activity of cancer cells following treatments with androstano-arylpyrimidines. We found that acetylated compounds were capable of attenuating the membrane efflux of inherently MDR cells; however, deacetylated counterparts were ineffective. To delineate the possible molecular mechanisms underlying these unique activities of androstano-arylpyrimidines, the degree of apoptosis induction was assessed by AnnexinV-based assays, both upon the individual as well as by steroid and chemotherapy agent combination treatments. Five dihydrotestosterone derivatives applied in combination with Doxorubicin or Epirubicin triggered massive apoptosis in MDR cells, and these combinations were more efficient than chemotherapy drugs together with Verapamil. Furthermore, our results revealed that androstano-arylpyrimidines induced significant endoplasmic reticulum stress (ER stress) but did not notably modulate ABC transporter expression. Therefore, ER stress triggered by acetylated androstano-arylpyrimidines is probably involved in the mechanism of efflux pump inhibition and drug sensitization which can be targeted in future drug developments to defeat inherently multidrug-resistant cancer.
Collapse
|
22
|
Jha NG, Dkhar DS, Singh SK, Malode SJ, Shetti NP, Chandra P. Engineered Biosensors for Diagnosing Multidrug Resistance in Microbial and Malignant Cells. BIOSENSORS 2023; 13:235. [PMID: 36832001 PMCID: PMC9954051 DOI: 10.3390/bios13020235] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/17/2023] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
To curtail pathogens or tumors, antimicrobial or antineoplastic drugs have been developed. These drugs target microbial/cancer growth and survival, thereby improving the host's health. In attempts to evade the detrimental effects of such drugs, these cells have evolved several mechanisms over time. Some variants of the cells have developed resistances against multiple drugs or antimicrobial agents. Such microorganisms or cancer cells are said to exhibit multidrug resistance (MDR). The drug resistance status of a cell can be determined by analyzing several genotypic and phenotypic changes, which are brought about by significant physiological and biochemical alterations. Owing to their resilient nature, treatment and management of MDR cases in clinics is arduous and requires a meticulous approach. Currently, techniques such as plating and culturing, biopsy, gene sequencing, and magnetic resonance imaging are prevalent in clinical practices for determining drug resistance status. However, the major drawbacks of using these methods lie in their time-consuming nature and the problem of translating them into point-of-care or mass-detection tools. To overcome the shortcomings of conventional techniques, biosensors with a low detection limit have been engineered to provide quick and reliable results conveniently. These devices are highly versatile in terms of analyte range and quantities that can be detected to report drug resistance in a given sample. A brief introduction to MDR, along with a detailed insight into recent biosensor design trends and use for identifying multidrug-resistant microorganisms and tumors, is presented in this review.
Collapse
Affiliation(s)
- Niharika G. Jha
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, Uttar Pradesh, India
| | - Daphika S. Dkhar
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, Uttar Pradesh, India
| | - Sumit K. Singh
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, Uttar Pradesh, India
| | - Shweta J. Malode
- Center for Energy and Environment, School of Advanced Sciences, KLE Technological University, Hubballi 580031, Karnataka, India
| | - Nagaraj P. Shetti
- Center for Energy and Environment, School of Advanced Sciences, KLE Technological University, Hubballi 580031, Karnataka, India
- University Center for Research & Development (UCRD), Chandigarh University, Mohali 140413, Panjab, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
23
|
Chen Z, Wen T, Wang X, Yang L, Wang Z, Qin Y, Hu Y, Zhang T, Wang D, Liu A, Zhang L, Lei M, Zhu Y. Co-delivery of immunochemotherapeutic by classified targeting based on chitosan and cyclodextrin derivatives. Int J Biol Macromol 2023; 226:1396-1410. [PMID: 36442558 DOI: 10.1016/j.ijbiomac.2022.11.253] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Herein, a cyclodextrin derivative (R6RGD-CMβCD) with tumor target and a carboxymethyl chitosan derivative (M2pep-CMCS) with tumor-associated macrophages 2 (TAM2) target were successfully synthesized, respectively. DOX-loaded nanoparticles (R6RGD-CMβCD@DOX NPs, RCNPDOX) and R848-loaded nanoparticles (M2pep-CMCS@R848 NPs, MCNPR848) were prepared. Furthermore, the RCNPDOX and MCNPR848 exhibited good DOX and R848 absorption. Meanwhile, the synergetic cell toxicity of RCNPDOX and MCNPR848 was found. Additionally, RCNPDOX + MCNPR848 nanoparticles greatly promoted the expression levels of cleaved Caspase3, which indicated that the nanoparticles could induce cell apoptosis. At the same time, the immunohistochemical images exhibited that RCNPDOX + MCNPR848 group could effectively transform the phenotype of tumor-associated macrophages. Importantly, in vivo experiments revealed that RCNPDOX + MCNPR848 NPs exerted excellent anticancer effects in tumor-bearing mice. To summarize, RCNPDOX + MCNPR848 NPs are effective anticancer treatment combining chemotherapy and immunotherapy, M2pep-CMCS and R6RGD-CMβCD are good delivery materials.
Collapse
Affiliation(s)
- Zhimeng Chen
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Tiantian Wen
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Xueyuan Wang
- College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Lin Yang
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Zhongjie Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yanru Qin
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yixue Hu
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Tianyu Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Dongna Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Amin Liu
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Liefeng Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Meng Lei
- College of Science, Nanjing Forestry University, Nanjing 210037, China.
| | - Yongqiang Zhu
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China; College of Life Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
24
|
Dömötör O, Teixeira RG, Spengler G, Avecilla F, Marques F, Lenis-Rojas OA, Matos CP, de Almeida RFM, Enyedy ÉA, Tomaz AI. Ruthenium(II) polypyridyl complexes with benzothiophene and benzimidazole derivatives: Synthesis, antitumor activity, solution studies and biospeciation. J Inorg Biochem 2023; 238:112058. [PMID: 36375357 DOI: 10.1016/j.jinorgbio.2022.112058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/08/2022]
Abstract
With the aim to incorporate pharmacophore motifs into the Ru(II)-polypyridyl framework, compounds [Ru(II)(1,10-phenantroline)2(2-(2-pyridyl)benzo[b]thiophene)](CF3SO3)2 (1) and [Ru(II)(1,10-phenantroline)2(2-(2-pyridyl)benzimidazole)](CF3SO3)2 (2) were prepared, characterized and tested for their antitumor potential. The solid-state structure of the compounds was confirmed by single-crystal X-ray diffraction analysis. The solution behavior of both complexes was investigated, namely their solubility, stability, and lipophilicity in physiological mimetic conditions, as well as an eventual uptake by passive diffusion. In vitro anticancer activity of the complexes on ovarian and different colon cancer cells and apoptosis induction by the complexes were studied. A slow transformation process was observed for complex 1 in aqueous solution when exposed to sunlight, while complex 2 undergoes deprotonation (pKa = 7.59). The lipophilicity of this latter complex depends strongly on the pH and ionic strength. In contrast, 1 is rather hydrophilic under various conditions. Complex 1 was highly cytotoxic on Colo-205 human colon (IC50 = 7.87 μM) and A2780 ovarian (IC50 = 2.2 μM) adenocarcinoma cell lines, while 2 displayed moderate anticancer activity (30.9 μM and 18.0 μM, respectively). The complexes induced late apoptosis and necrosis. Only a weak binding of the complexes to human serum albumin, the main transport protein in blood serum, was found. However, a more significant binding to calf thymus DNA was observed in UV-visible titrations and fluorometric dye displacement studies. Detailed analysis of fluorescence lifetime data collected for the latter systems reveals not only the partial intercalation of the complexes, but goes beyond the usual simplified interpretations.
Collapse
Affiliation(s)
- Orsolya Dömötör
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary; MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary.
| | - Ricardo G Teixeira
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1049-016 Lisboa, Portugal
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary; Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Fernando Avecilla
- Universidade da Coruña, Grupo NanoToxGen, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Química, Facultade de Ciencias, Campus de A Coruña, 15071A Coruña, Spain
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares and Departamento de Ciências e Engenharia Nucleares, Instituto Superior Técnico, Universidade de Lisboa, EN 10 (km 139,7), 2695-066 Bobadela, Loures, Portugal
| | - Oscar A Lenis-Rojas
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1049-016 Lisboa, Portugal
| | - Cristina P Matos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1049-016 Lisboa, Portugal
| | - Rodrigo F M de Almeida
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1049-016 Lisboa, Portugal
| | - Éva A Enyedy
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary; MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Ana Isabel Tomaz
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1049-016 Lisboa, Portugal.
| |
Collapse
|
25
|
Moore PC, Henderson KW, Classon M. The epigenome and the many facets of cancer drug tolerance. Adv Cancer Res 2023; 158:1-39. [PMID: 36990531 DOI: 10.1016/bs.acr.2022.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The use of chemotherapeutic agents and the development of new cancer therapies over the past few decades has consequently led to the emergence of myriad therapeutic resistance mechanisms. Once thought to be explicitly driven by genetics, the coupling of reversible sensitivity and absence of pre-existing mutations in some tumors opened the way for discovery of drug-tolerant persisters (DTPs): slow-cycling subpopulations of tumor cells that exhibit reversible sensitivity to therapy. These cells confer multi-drug tolerance, to targeted and chemotherapies alike, until the residual disease can establish a stable, drug-resistant state. The DTP state can exploit a multitude of distinct, yet interlaced, mechanisms to survive otherwise lethal drug exposures. Here, we categorize these multi-faceted defense mechanisms into unique Hallmarks of Cancer Drug Tolerance. At the highest level, these are comprised of heterogeneity, signaling plasticity, differentiation, proliferation/metabolism, stress management, genomic integrity, crosstalk with the tumor microenvironment, immune escape, and epigenetic regulatory mechanisms. Of these, epigenetics was both one of the first proposed means of non-genetic resistance and one of the first discovered. As we describe in this review, epigenetic regulatory factors are involved in most facets of DTP biology, positioning this hallmark as an overarching mediator of drug tolerance and a potential avenue to novel therapies.
Collapse
|
26
|
Pivarcsik T, Pósa V, Kovács H, May NV, Spengler G, Pósa SP, Tóth S, Nezafat Yazdi Z, Özvegy-Laczka C, Ugrai I, Szatmári I, Szakács G, Enyedy ÉA. Metal Complexes of a 5-Nitro-8-Hydroxyquinoline-Proline Hybrid with Enhanced Water Solubility Targeting Multidrug Resistant Cancer Cells. Int J Mol Sci 2022; 24:ijms24010593. [PMID: 36614037 PMCID: PMC9820345 DOI: 10.3390/ijms24010593] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Multidrug resistance (MDR) in cancer is one of the major obstacles of chemotherapy. We have recently identified a series of 8-hydroxyquinoline Mannich base derivatives with MDR-selective toxicity, however with limited solubility. In this work, a novel 5-nitro-8-hydroxyquinoline-proline hybrid and its Rh(η5-C5Me5) and Ru(η6-p-cymene) complexes with excellent aqueous solubility were developed, characterized, and tested against sensitive and MDR cells. Complex formation of the ligand with essential metal ions was also investigated using UV-visible, circular dichroism, 1H NMR (Zn(II)), and electron paramagnetic resonance (Cu(II)) spectroscopic methods. Formation of mono and bis complexes was found in all cases with versatile coordination modes, while tris complexes were also formed with Fe(II) and Fe(III) ions, revealing the metal binding affinity of the ligand at pH 7.4: Cu(II) > Zn(II) > Fe(II) > Fe(III). The ligand and its Rh(III) complex displayed enhanced cytotoxicity against the resistant MES-SA/Dx5 and Colo320 human cancer cell lines compared to their chemosensitive counterparts. Both organometallic complexes possess high stability in solution, however the Ru(II) complex has lower chloride ion affinity and slower ligand exchange processes, along with the readiness to lose the arene ring that is likely connected to its inactivity.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Vivien Pósa
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Hilda Kovács
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Nóra V. May
- Centre for Structural Science, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Szonja P. Pósa
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Szilárd Tóth
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Zeinab Nezafat Yazdi
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Imre Ugrai
- Institute of Pharmaceutical Chemistry and Stereochemistry Research Group, Eötvös Loránd Research Network, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - István Szatmári
- Institute of Pharmaceutical Chemistry and Stereochemistry Research Group, Eötvös Loránd Research Network, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Gergely Szakács
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Éva A. Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
27
|
Todosenko N, Yurova K, Khaziakhmatova O, Malashchenko V, Khlusov I, Litvinova L. Heparin and Heparin-Based Drug Delivery Systems: Pleiotropic Molecular Effects at Multiple Drug Resistance of Osteosarcoma and Immune Cells. Pharmaceutics 2022; 14:pharmaceutics14102181. [PMID: 36297616 PMCID: PMC9612132 DOI: 10.3390/pharmaceutics14102181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/23/2022] Open
Abstract
One of the main problems of modern health care is the growing number of oncological diseases both in the elderly and young population. Inadequately effective chemotherapy, which remains the main method of cancer control, is largely associated with the emergence of multidrug resistance in tumor cells. The search for new solutions to overcome the resistance of malignant cells to pharmacological agents is being actively pursued. Another serious problem is immunosuppression caused both by the tumor cells themselves and by antitumor drugs. Of great interest in this context is heparin, a biomolecule belonging to the class of glycosaminoglycans and possessing a broad spectrum of biological activity, including immunomodulatory and antitumor properties. In the context of the rapid development of the new field of “osteoimmunology,” which focuses on the collaboration of bone and immune cells, heparin and delivery systems based on it may be of intriguing importance for the oncotherapy of malignant bone tumors. Osteosarcoma is a rare but highly aggressive, chemoresistant malignant tumor that affects young adults and is characterized by constant recurrence and metastasis. This review describes the direct and immune-mediated regulatory effects of heparin and drug delivery systems based on it on the molecular mechanisms of (multiple) drug resistance in (onco) pathological conditions of bone tissue, especially osteosarcoma.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Vladimir Malashchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Igor Khlusov
- Department of Morphology and General Pathology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Correspondence:
| |
Collapse
|
28
|
Kalave S, Hegde N, Juvale K. Applications of Nanotechnology-based Approaches to Overcome Multi-drug Resistance in Cancer. Curr Pharm Des 2022; 28:3140-3157. [PMID: 35366765 DOI: 10.2174/1381612828666220401142300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/27/2022] [Indexed: 01/28/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Chemotherapy and radiation therapy are the major treatments used for the management of cancer. Multidrug resistance (MDR) is a major hindrance faced in the treatment of cancer and is also responsible for cancer relapse. To date, several studies have been carried out on strategies to overcome or reverse MDR in cancer. Unfortunately, the MDR reversing agents have been proven to have minimal clinical benefits, and eventually, no improvement has been made in therapeutic efficacy to date. Thus, several investigational studies have also focused on overcoming drug resistance rather than reversing the MDR. In this review, we focus primarily on nanoformulations regarded as a novel approach to overcome or bypass the MDR in cancer. The nanoformulation systems serve as an attractive strategy as these nanosized materials selectively get accumulated in tumor tissues, thereby improving the clinical outcomes of patients suffering from MDR cancer. In the current work, we present an overview of recent trends in the application of various nano-formulations, belonging to different mechanistic classes and functionalization like carbon nanotubes, carbon nanohorns, carbon nanospheres, liposomes, dendrimers, etc., to overcome MDR in cancer. A detailed overview of these techniques will help researchers in exploring the applicability of nanotechnologybased approaches to treat MDR.
Collapse
Affiliation(s)
- Sana Kalave
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle [W], Mumbai, India
| | - Namita Hegde
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle [W], Mumbai, India
| | - Kapil Juvale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle [W], Mumbai, India
| |
Collapse
|
29
|
Żesławska E, Tejchman W, Kincses A, Spengler G, Nitek W, Żuchowski G, Szymańska E. 5-Arylidenerhodanines as P-gp Modulators: An Interesting Effect of the Carboxyl Group on ABCB1 Function in Multidrug-Resistant Cancer Cells. Int J Mol Sci 2022; 23:ijms231810812. [PMID: 36142724 PMCID: PMC9503420 DOI: 10.3390/ijms231810812] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Multidrug resistance (MDR) is considered one of the major mechanisms responsible for the failure of numerous anticancer and antiviral chemotherapies. Various strategies to overcome the MDR phenomenon have been developed, and one of the most attractive research directions is focused on the inhibition of MDR transporters, membrane proteins that extrude cytotoxic drugs from living cells. Here, we report the results of our studies on a series newly synthesized of 5-arylidenerhodanines and their ability to inhibit the ABCB1 efflux pump in mouse T-lymphoma cancer cells. In the series, compounds possessing a triphenylamine moiety and the carboxyl group in their structure were of particular interest. These amphiphilic compounds showed over 17-fold stronger efflux pump inhibitory effects than verapamil. The cytotoxic and antiproliferative effects of target rhodanines on T-lymphoma cells were also investigated. A putative binding mode for 11, one of the most potent P-gp inhibitors tested here, was predicted by molecular docking studies and discussed with regard to the binding mode of verapamil.
Collapse
Affiliation(s)
- Ewa Żesławska
- Institute of Biology, Pedagogical University of Krakow, Podchorążych 2, 30-084 Kraków, Poland
- Correspondence: (E.Ż.); (E.S.)
| | - Waldemar Tejchman
- Institute of Biology, Pedagogical University of Krakow, Podchorążych 2, 30-084 Kraków, Poland
| | - Annamária Kincses
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Gabriella Spengler
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Wojciech Nitek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Grzegorz Żuchowski
- Chair of Organic Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Ewa Szymańska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
- Correspondence: (E.Ż.); (E.S.)
| |
Collapse
|
30
|
Pósa SP, Dargó G, Nagy S, Kisszékelyi P, Garádi Z, Hámori L, Szakács G, Kupai J, Tóth S. Cytotoxicity of cinchona alkaloid organocatalysts against MES-SA and MES-SA/Dx5 multidrug-resistant uterine sarcoma cell lines. Bioorg Med Chem 2022; 67:116855. [PMID: 35640378 DOI: 10.1016/j.bmc.2022.116855] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/10/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022]
Abstract
Since the first application of natural quinine as an anti-malarial drug, cinchona alkaloids and their derivatives have been exhaustively studied for their biological activity. In our work, we tested 13 cinchona alkaloid organocatalysts, synthesised from quinine. These derivatives were screened against MES-SA and Dx5 uterine sarcoma cell lines for in vitro anticancer activity and to investigate their potential to overcome P-glycoprotein (P-gp) mediated multidrug resistance (MDR). Decorating quinine with hydrogen-bond donor units, such as thiourea and (thio)squaramide, resulted in decreased half-maximal growth inhibition values on both cell lines (1.3-21 µM) compared to quinine and other cinchona alcohols (47-111 µM). Further cytotoxicity studies conducted in the presence of the P-gp inhibitor tariquidar indicated that several analogues, especially cinchona amines and squaramides, but not thiosquaramide, were expelled from MDR cells by P-gp. Similarly to the established P-gp inhibitor quinine, 6 cinchona analogues were shown to inhibit calcein-AM efflux. Interestingly, quinine and didehydroquinine exhibited a marginally increased toxicity against the multidrug resistant Dx5 cells. Collateral sensitivity of the MDR cell line was more pronounced when the cinchona thiosquaramide was complexed with Cu(II) acetate. Based on the results, cinchona derivatives are good anticancer candidates for further drug development.
Collapse
Affiliation(s)
- Szonja Polett Pósa
- Department of Organic Chemistry & Technology, Budapest University of Technology & Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary; Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gyula Dargó
- Department of Organic Chemistry & Technology, Budapest University of Technology & Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary
| | - Sándor Nagy
- Department of Organic Chemistry & Technology, Budapest University of Technology & Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary
| | - Péter Kisszékelyi
- Department of Organic Chemistry & Technology, Budapest University of Technology & Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary
| | - Zsófia Garádi
- Department of Pharmacognosy Semmelweis University, Üllői út. 26, H-1085 Budapest, Hungary
| | - Lilla Hámori
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Institute of Cancer Research, Medical University Vienna, Vienna, Austria
| | - József Kupai
- Department of Organic Chemistry & Technology, Budapest University of Technology & Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary.
| | - Szilárd Tóth
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary.
| |
Collapse
|
31
|
Farrokhian N, Maltas J, Dinh M, Durmaz A, Ellsworth P, Hitomi M, McClure E, Marusyk A, Kaznatcheev A, Scott JG. Measuring competitive exclusion in non-small cell lung cancer. SCIENCE ADVANCES 2022; 8:eabm7212. [PMID: 35776787 PMCID: PMC10883359 DOI: 10.1126/sciadv.abm7212] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In this study, we experimentally measure the frequency-dependent interactions between a gefitinib-resistant non-small cell lung cancer population and its sensitive ancestor via the evolutionary game assay. We show that cost of resistance is insufficient to accurately predict competitive exclusion and that frequency-dependent growth rate measurements are required. Using frequency-dependent growth rate data, we then show that gefitinib treatment results in competitive exclusion of the ancestor, while the absence of treatment results in a likely, but not guaranteed, exclusion of the resistant strain. Then, using simulations, we demonstrate that incorporating ecological growth effects can influence the predicted extinction time. In addition, we show that higher drug concentrations may not lead to the optimal reduction in tumor burden. Together, these results highlight the potential importance of frequency-dependent growth rate data for understanding competing populations, both in the laboratory and as we translate adaptive therapy regimens to the clinic.
Collapse
Affiliation(s)
| | - Jeff Maltas
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Mina Dinh
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | | | | | - Masahiro Hitomi
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Erin McClure
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Andriy Marusyk
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Artem Kaznatcheev
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob G Scott
- CWRU School of Medicine, Cleveland, OH, USA
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
32
|
Hanssen KM, Wheatley MS, Yu DMT, Conseil G, Norris MD, Haber M, Cole SPC, Fletcher JI. GSH facilitates the binding and inhibitory activity of novel multidrug resistance protein 1 (MRP1) modulators. FEBS J 2022; 289:3854-3875. [PMID: 35080351 DOI: 10.1111/febs.16374] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/29/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
MRP1 (ABCC1) is a membrane transporter that confers multidrug resistance in cancer cells by exporting chemotherapeutic agents, often in a reduced glutathione (GSH)-dependent manner. This transport activity can be altered by compounds (modulators) that block drug transport while simultaneously stimulating GSH efflux by MRP1. In MRP1-expressing cells, modulator-stimulated GSH efflux can be sufficient to deplete GSH and increase sensitivity to chemotherapy, enhancing cancer cell death. Further development of clinically useful MRP1 modulators requires a better mechanistic understanding of modulator binding and its relationship to GSH binding and transport. Here, we explore the mechanism of two MRP1 small molecule modulators, 5681014 and 7914321, in relation to a bipartite substrate-binding cavity of MRP1. Binding of these modulators to MRP1 was dependent on the presence of GSH but not its reducing capacity. Accordingly, the modulators poorly inhibited organic anion transport by K332L-mutant MRP1, where GSH binding and transport is limited. However, the inhibitory activity of the modulators was also diminished by mutations that limit E2 17βG but spare GSH-conjugate binding and transport (W553A, M1093A, W1246A), suggesting overlap between the E2 17βG and modulator binding sites. Immunoblots of limited trypsin digests of MRP1 suggest that binding of GSH, but not the modulators, induces a conformation change in MRP1. Together, these findings support the model, in which GSH binding induces a conformation change that facilitates binding of MRP1 modulators, possibly in a proposed hydrophobic binding pocket of MRP1. This study may facilitate the structure-guided design of more potent and selective MRP1 modulators.
Collapse
Affiliation(s)
- Kimberley M Hanssen
- Lowy Cancer Research Centre, Children's Cancer Institute Australia, UNSW Sydney, Kensington, Australia.,School of Women's and Children's Health, UNSW Sydney, Kensington, Australia
| | - Madeleine S Wheatley
- Lowy Cancer Research Centre, Children's Cancer Institute Australia, UNSW Sydney, Kensington, Australia
| | - Denise M T Yu
- Lowy Cancer Research Centre, Children's Cancer Institute Australia, UNSW Sydney, Kensington, Australia.,School of Women's and Children's Health, UNSW Sydney, Kensington, Australia
| | - Gwenaëlle Conseil
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen's University Cancer Research Institute, Kingston, Canada
| | - Murray D Norris
- Lowy Cancer Research Centre, Children's Cancer Institute Australia, UNSW Sydney, Kensington, Australia.,University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, Australia
| | - Michelle Haber
- Lowy Cancer Research Centre, Children's Cancer Institute Australia, UNSW Sydney, Kensington, Australia.,School of Women's and Children's Health, UNSW Sydney, Kensington, Australia
| | - Susan P C Cole
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen's University Cancer Research Institute, Kingston, Canada
| | - Jamie I Fletcher
- Lowy Cancer Research Centre, Children's Cancer Institute Australia, UNSW Sydney, Kensington, Australia.,School of Women's and Children's Health, UNSW Sydney, Kensington, Australia
| |
Collapse
|
33
|
Chi S, Zuo M, Zhu M, Wang Z, Liu Z. Loading Drugs in Natural Phospholipid Bilayers of Cell Membrane Shells to Construct Biomimetic Nanocomposites for Enhanced Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:28671-28682. [PMID: 35703029 DOI: 10.1021/acsami.2c08587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drug-based oncotherapy is seriously challenged by insufficient drug accumulation at tumor sites, mainly resulting from low drug loading efficiency and poor tumor-targeting ability of drug carriers. We herein proposed a "one-stone, two-bird" strategy to circumvent both obstacles, utilizing the source cancer cell membrane (CM) as a dual-function carrier to simultaneously achieve sufficient drug loading and homologous tumor targeting. Combining the use of TPGS (d-α-tocopherol polyethylene glycol 1000 succinate) to inhibit the drug efflux process of drug-resistant tumor, we constructed core-shell-structured nanocomposites CMGNPs consisting of ICG (indocyanine green)/DOX (doxorubicin)-loaded, TPGS/OA (oleic acid)-stabilized upconversion nanoparticles as the core and ICG-loaded MCF7/ADR CMs as the shell, for combined chemo/phototherapy of MCF7/ADR tumor. The employment of phospholipid bilayers of CMs as natural pockets for extra drug loading while preserving the homologous targeting ability greatly enhanced drug concentration at tumor sites, endowing CMGNPs with excellent therapeutic efficacy. Our effort provides a versatile approach for facilitating drug delivery in diverse therapeutic systems.
Collapse
Affiliation(s)
- Siyu Chi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Miaomiao Zuo
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Mengting Zhu
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Zijun Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Zhihong Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
34
|
Zhang J, Cunningham J, Brown J, Gatenby R. Evolution-based mathematical models significantly prolong response to abiraterone in metastatic castrate-resistant prostate cancer and identify strategies to further improve outcomes. eLife 2022; 11:e76284. [PMID: 35762577 PMCID: PMC9239688 DOI: 10.7554/elife.76284] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/01/2022] [Indexed: 11/15/2022] Open
Abstract
Background Abiraterone acetate is an effective treatment for metastatic castrate-resistant prostate cancer (mCRPC), but evolution of resistance inevitably leads to progression. We present a pilot study in which abiraterone dosing is guided by evolution-informed mathematical models to delay onset of resistance. Methods In the study cohort, abiraterone was stopped when PSA was <50% of pretreatment value and resumed when PSA returned to baseline. Results are compared to a contemporaneous cohort who had >50% PSA decline after initial abiraterone administration and met trial eligibility requirements but chose standard of care (SOC) dosing. Results 17 subjects were enrolled in the adaptive therapy group and 16 in the SOC group. All SOC subjects have progressed, but four patients in the study cohort remain stably cycling (range 53-70 months). The study cohort had significantly improved median time to progression (TTP; 33.5 months; p<0.001) and median overall survival (OS; 58.5 months; hazard ratio, 0.41, 95% confidence interval (CI), 0.20-0.83, p<0.001) compared to 14.3 and 31.3 months in the SOC cohort. On average, study subjects received no abiraterone during 46% of time on trial. Longitudinal trial data demonstrated the competition coefficient ratio (αRS/αSR) of sensitive and resistant populations, a critical factor in intratumoral evolution, was two- to threefold higher than pre-trial estimates. Computer simulations of intratumoral evolutionary dynamics in the four long-term survivors found that, due to the larger value for αRS/αSR, cycled therapy significantly decreased the resistant population. Simulations in subjects who progressed predicted further increases in OS could be achieved with prompt abiraterone withdrawal after achieving 50% PSA reduction. Conclusions Incorporation of evolution-based mathematical models into abiraterone monotherapy for mCRPC significantly increases TTP and OS. Computer simulations with updated parameters from longitudinal trial data can estimate intratumoral evolutionary dynamics in each subject and identify strategies to improve outcomes. Funding Moffitt internal grants and NIH/NCI U54CA143970-05 (Physical Science Oncology Network).
Collapse
Affiliation(s)
- Jingsong Zhang
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research InstituteTampaUnited States
| | - Jessica Cunningham
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center and Research InstituteTampaUnited States
| | - Joel Brown
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center and Research InstituteTampaUnited States
- Department of Biological Sciences, University of Illinois at ChicagoChicagoUnited States
| | - Robert Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center and Research InstituteTampaUnited States
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research InstituteTampaUnited States
| |
Collapse
|
35
|
Yang G, Wang L, Fan Y, Lai Z, Yu X, Lou L, Gao K, Yue J. Concise Total Synthesis of Dysoxylactam A and a Simplified Analog. CHINESE J CHEM 2022. [DOI: 10.1002/cjoc.202200123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Guan‐Zhou Yang
- State Key Laboratory of Applied Organic Chemistry College of Chemistry and Chemical Engineering, Lanzhou Uni‐versity 222 Tianshui South Road Lanzhou 730000 China
| | - Lei Wang
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| | - Yao‐Yue Fan
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| | - Zeng‐Wei Lai
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| | - Xue‐Ni Yu
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Li‐Guang Lou
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| | - Kun Gao
- State Key Laboratory of Applied Organic Chemistry College of Chemistry and Chemical Engineering, Lanzhou Uni‐versity 222 Tianshui South Road Lanzhou 730000 China
| | - Jian‐Min Yue
- State Key Laboratory of Applied Organic Chemistry College of Chemistry and Chemical Engineering, Lanzhou Uni‐versity 222 Tianshui South Road Lanzhou 730000 China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road Shanghai 201203 China
| |
Collapse
|
36
|
Pape VFS, Palkó R, Tóth S, Szabó MJ, Sessler J, Dormán G, Enyedy ÉA, Soós T, Szatmári I, Szakács G. Structure-Activity Relationships of 8-Hydroxyquinoline-Derived Mannich Bases with Tertiary Amines Targeting Multidrug-Resistant Cancer. J Med Chem 2022; 65:7729-7745. [PMID: 35613553 PMCID: PMC9189845 DOI: 10.1021/acs.jmedchem.2c00076] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
![]()
A recently proposed
strategy to overcome multidrug resistance (MDR)
in cancer is to target the collateral sensitivity of otherwise resistant
cells. We designed a library of 120 compounds to explore the chemical
space around previously identified 8-hydroxyquinoline-derived Mannich
bases with robust MDR-selective toxicity. We included compounds to
study the effect of halogen and alkoxymethyl substitutions in R5 in
combination with different Mannich bases in R7, a shift of the Mannich
base from R7 to R5, as well as the introduction of an aromatic moiety.
Cytotoxicity tests performed on a panel of parental and MDR cells
highlight a strong influence of experimentally determined pKa values of the donor atom moieties, indicating
that protonation and metal chelation are important factors modulating
the MDR-selective anticancer activity of the studied compounds. Our
results identify structural requirements increasing MDR-selective
anticancer activity, providing guidelines for the development of more
effective anticancer chelators targeting MDR cancer.
Collapse
Affiliation(s)
- Veronika F S Pape
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary.,Department of Physiology, Semmelweis University, Faculty of Medicine, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
| | - Roberta Palkó
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| | - Szilárd Tóth
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| | | | - Judit Sessler
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| | - György Dormán
- TargetEx Ltd., Madách Imre u 31/2., H-2120 Dunakeszi, Hungary
| | - Éva A Enyedy
- Department of Inorganic and Analytical Chemistry, MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Tibor Soós
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| | - István Szatmári
- Institute of Pharmaceutical Chemistry and Stereochemistry Research Group of Hungarian Academy of Sciences, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary.,Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| |
Collapse
|
37
|
Targeting breast cancer resistance protein (BCRP/ABCG2): Functional inhibitors and expression modulators. Eur J Med Chem 2022; 237:114346. [DOI: 10.1016/j.ejmech.2022.114346] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/15/2022] [Accepted: 04/01/2022] [Indexed: 12/16/2022]
|
38
|
Díaz-Alejo JF, González Gómez I, Earl J. Ultrasounds in cancer therapy: A summary of their use and unexplored potential. Oncol Rev 2022; 16:531. [PMID: 35340884 PMCID: PMC8941342 DOI: 10.4081/oncol.2022.531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 09/17/2021] [Indexed: 11/26/2022] Open
Abstract
Ultrasounds (US) are a non-ionizing mechanical wave, with less adverse effects than conventional pharmacological or surgical treatments. Different biological effects are induced in tissues and cells by ultrasound actuation depending on acoustic parameters, such as the wave intensity, frequency and treatment dose. This non-ionizing radiation has considerable applications in biomedicine including surgery, medical imaging, physical therapy and cancer therapy. Depending on the wave intensity, US are applied as high-intensity ultrasounds (HIUS) and low-intensity pulsed ultrasounds (LIPUS), with different effects on cells and tissues. HIUS produce thermal and mechanical effects, resulting in a large localized temperature increase, leading to tissue ablation and even tumor necrosis. This can be achieved by focusing low intensity waves emitted from different electrically shifted transducers, known as high-intensity focused ultrasounds (HIFU). LIPUS have been used extensively as a therapeutic, surgical and diagnostic tool, with diverse biological effects observed in tissues and cultured cells. US represent a non-invasive treatment strategy that can be applied to selected areas of the body, with limited adverse effects. In fact, tumor ablation using HIFU has been used as a curative treatment in patients with an early-stage pancreatic tumor and is an effective palliative treatment in patients with advanced stage disease. However, the biological effects, dose standardization, benefit-risk ratio and safety are not fully understood. Thus, it is an emerging field that requires further research in order to reach its full potential.
Collapse
Affiliation(s)
- Jesús Frutos Díaz-Alejo
- Molecular Epidemiology and Predictive Tumor Markers Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid
- Faculty of Medicine and Health Sciences, University of Alcalá de Henares (UAH), Madrid
| | | | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid
- Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| |
Collapse
|
39
|
Abd-Ellatef GEF, Gazzano E, El-Desoky AH, Hamed AR, Kopecka J, Belisario DC, Costamagna C, S Marie MA, Fahmy SR, Abdel-Hamid AHZ, Riganti C. Glabratephrin reverses doxorubicin resistance in triple negative breast cancer by inhibiting P-glycoprotein. Pharmacol Res 2022; 175:105975. [PMID: 34785319 DOI: 10.1016/j.phrs.2021.105975] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/27/2021] [Accepted: 11/06/2021] [Indexed: 11/28/2022]
Abstract
Triple-negative breast cancer is one of the most aggressive breast cancer. The first therapeutic option is chemotherapy, often based on anthracycline as doxorubicin. However, chemotherapy efficacy is limited in by the presence of P-glycoprotein (Pgp), a membrane transporter protein that effluxes doxorubicin, reducing its cellular accumulation and toxicity. Inhibiting Pgp activity with effective and non-toxic products is still an open challenge. In this work, we demonstrated that the natural product Glabratephrin (Glab), a prenylated flavonoid from Tephrosia purpurea with a unique chemical structure, increased doxorubicin accumulation and cytotoxicity in triple negative breast cancer cells with high levels of Pgp, characterized by both acquired or intrinsic resistance to doxorubicin. Glab also reduced the growth of Pgp-expressing tumors, without adding significant extra-toxicities to doxorubicin treatment. Interestingly, Glab did not change the expression of Pgp, but it reduced the affinity for Pgp and the efflux of doxorubicin, as suggested by the increased Km and the reduced Vmax. In silico molecular docking predicted that Glab binds two residues (phenylalanine 322, glutamine 721) localized in the transmembrane domains of Pgp, facing the extracellular environment. Moreover, site-directed mutagenesis identified glycine 185 as a critical residue mediating the reduced catalytic efficacy of Pgp elicited by Glab. We propose Glab as an effective and safe compound able to reverse doxorubicin resistance mediated by Pgp in triple negative breast cancers, opening the way to a new combinatorial approach that may improve chemotherapy efficacy in the most refractory and aggressive breast cancer.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cell Line, Tumor
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Female
- Flavonoids/pharmacology
- Flavonoids/therapeutic use
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Mice, Inbred BALB C
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Mice
Collapse
Affiliation(s)
- Gamal Eldein Fathy Abd-Ellatef
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy; Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El Bohouth St., 12622 Dokki, Giza, Egypt
| | - Elena Gazzano
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | - Ahmed H El-Desoky
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El Bohouth St., 12622 Dokki, Giza, Egypt
| | - Ahmed R Hamed
- Chemistry of Medicinal Plants Department & Biology Unit of Central Laboratory, Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El Bohouth St., 12622 Dokki, Giza, Egypt
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | | | - Costanzo Costamagna
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | | | - Sohair R Fahmy
- Zoology Department, Faculty of Science, Cairo University, Gamaa Street, Giza, Egypt
| | - Abdel-Hamid Z Abdel-Hamid
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El Bohouth St., 12622 Dokki, Giza, Egypt
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy.
| |
Collapse
|
40
|
Ramalhete C, Gonçalves BMF, Barbosa F, Duarte N, Ferreira MJU. Momordica balsamina: phytochemistry and pharmacological potential of a gifted species. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2022; 21:617-646. [PMID: 35153639 PMCID: PMC8821832 DOI: 10.1007/s11101-022-09802-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 01/09/2022] [Indexed: 05/07/2023]
Abstract
Momordica balsamina L. (Cucurbitaceae), frequently named balsam apple, southern balsam pear or African pumpkin, is a vegetable with high nutritional value, being mostly used as food in sub-Saharan Africa. It has also been largely used in traditional medicine to treat several diseases, such as malaria fevers and diabetes. As a member of the Cucurbitaceae family, the main constituents are cucurbitane-type triterpenoids, with different oxidation patterns, named cucurbitacins. This review aims at summarizing our contribution to the phytochemical study of M. balsamina and the evaluation of the isolated cucurbitacins and derivatives as multidrug resistance reversers in cancer cells and bacteria. In this way, the selective antiproliferative activity against multidrug resistant cancer cells of cucurbitacins obtained from M. balsamina, their ability as P-glycoprotein inhibitors in cancer cells overexpressing this ABC transporter, as well as efflux pump inhibitors in resistant bacteria strains are reviewed. Moreover, the in vitro antimalarial activity of cucurbitacins and acyl derivatives against the blood and liver-stages of Plasmodium strains, and the in vivo activity of selected compounds is also reviewed. Besides our work, edible and medicinal uses, and other studies mainly reporting the biological activities of M. balsamina extracts, such as antidiabetic, antibacterial, anti-inflammatory, and antioxidant properties are also addressed.
Collapse
Affiliation(s)
- Cátia Ramalhete
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- ATLÂNTICA – Instituto Universitário, Fábrica da Pólvora de Barcarena, Barcarena, Oeiras, 2730-036 Portugal
| | - Bruno M. F. Gonçalves
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Filipa Barbosa
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Noélia Duarte
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria-José U. Ferreira
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
41
|
Mechanistic insight into photoactivation of small inorganic molecules from the biomedical applications perspectives. BIOMEDICAL APPLICATIONS OF INORGANIC PHOTOCHEMISTRY 2022. [DOI: 10.1016/bs.adioch.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
42
|
Hanssen KM, Haber M, Fletcher JI. Targeting multidrug resistance-associated protein 1 (MRP1)-expressing cancers: Beyond pharmacological inhibition. Drug Resist Updat 2021; 59:100795. [PMID: 34983733 DOI: 10.1016/j.drup.2021.100795] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 12/30/2022]
Abstract
Resistance to chemotherapy remains one of the most significant obstacles to successful cancer treatment. While inhibiting drug efflux mediated by ATP-binding cassette (ABC) transporters is a seemingly attractive and logical approach to combat multidrug resistance (MDR), small molecule inhibition of ABC transporters has so far failed to confer clinical benefit, despite considerable efforts by medicinal chemists, biologists, and clinicians. The long-sought treatment to eradicate cancers displaying ABC transporter overexpression may therefore lie within alternative targeting strategies. When aberrantly expressed, the ABC transporter multidrug resistance-associated protein 1 (MRP1, ABCC1) confers MDR, but can also shift cellular redox balance, leaving the cell vulnerable to select agents. Here, we explore the physiological roles of MRP1, the rational for targeting this transporter in cancer, the development of small molecule MRP1 inhibitors, and the most recent developments in alternative therapeutic approaches for targeting cancers with MRP1 overexpression. We discuss approaches that extend beyond simple MRP1 inhibition by exploiting the collateral sensitivity to glutathione depletion and ferroptosis, the rationale for targeting the shared transcriptional regulators of both MRP1 and glutathione biosynthesis, advances in gene silencing, and new molecules that modulate transporter activity to the detriment of the cancer cell. These strategies illustrate promising new approaches to address multidrug resistant disease that extend beyond the simple reversal of MDR and offer exciting routes for further research.
Collapse
Affiliation(s)
- Kimberley M Hanssen
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
43
|
Zhou Q, Mohammed F, Wang Y, Wang J, Lu N, Li J, Ge Z. Hypoxia-responsive block copolymer polyprodrugs for complementary photodynamic-chemotherapy. J Control Release 2021; 339:130-142. [PMID: 34560158 DOI: 10.1016/j.jconrel.2021.09.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 09/18/2021] [Indexed: 01/12/2023]
Abstract
The inherent hypoxic microenvironment of solid tumors has an important influence on tumor growth, distant metastasis, and invasiveness. The heterogeneous distribution of hypoxic regions inside tumors limits the therapeutic efficacy of O2-assisted therapeutic strategy (e.g. photodynamic therapy (PDT)). On the other hand, the hypoxia-activable prodrugs cannot work effectively in the regions with enough O2 concentration. To address the issues, we prepare a block copolymer polyprodrug consisting of polyethylene glycol (PEG) and copolymerized segments of nitroimidazole-linked camptothecin (CPT) methacrylate and 5,10,15,20-tetraphenylporphyrin (TPP)-containing methacrylate monomers for complementary photodynamic-chemotherapy. The polyprodrug can self-assemble into polymeric micelles in aqueous solution with suitable size and high stability. After intravenous injection, the polyprodrug micelles show tumor accumulation. Followed by light irradiation (650 nm) at tumor sites, TPP moieties induce singlet oxygen (1O2) production in the oxygen-rich area to exert PDT and cause transformation of the oxygen-rich areas into hypoxia. Simultaneously, in the hypoxic areas, the hypoxia-responsive polyprodrugs can be activated to release free CPT due to the cleavage of nitroimidazole linkages. The polyprodrug micelles with the segments for PDT and hypoxia-activable CPT efficiently suppress the growth of HeLa tumors. The well-defined polyprodrug amphiphiles offer an effective strategy to overcome the disadvantages of single treatment of PDT or hypoxia-responsive prodrugs for complementary photodynamic-chemotherapy of cancers.
Collapse
Affiliation(s)
- Qinghao Zhou
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Fathelrahman Mohammed
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yuheng Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Jingbo Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Nannan Lu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Junjie Li
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
44
|
Wölfl B, te Rietmole H, Salvioli M, Kaznatcheev A, Thuijsman F, Brown JS, Burgering B, Staňková K. The Contribution of Evolutionary Game Theory to Understanding and Treating Cancer. DYNAMIC GAMES AND APPLICATIONS 2021; 12:313-342. [PMID: 35601872 PMCID: PMC9117378 DOI: 10.1007/s13235-021-00397-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 05/05/2023]
Abstract
Evolutionary game theory mathematically conceptualizes and analyzes biological interactions where one's fitness not only depends on one's own traits, but also on the traits of others. Typically, the individuals are not overtly rational and do not select, but rather inherit their traits. Cancer can be framed as such an evolutionary game, as it is composed of cells of heterogeneous types undergoing frequency-dependent selection. In this article, we first summarize existing works where evolutionary game theory has been employed in modeling cancer and improving its treatment. Some of these game-theoretic models suggest how one could anticipate and steer cancer's eco-evolutionary dynamics into states more desirable for the patient via evolutionary therapies. Such therapies offer great promise for increasing patient survival and decreasing drug toxicity, as demonstrated by some recent studies and clinical trials. We discuss clinical relevance of the existing game-theoretic models of cancer and its treatment, and opportunities for future applications. Moreover, we discuss the developments in cancer biology that are needed to better utilize the full potential of game-theoretic models. Ultimately, we demonstrate that viewing tumors with evolutionary game theory has medically useful implications that can inform and create a lockstep between empirical findings and mathematical modeling. We suggest that cancer progression is an evolutionary competition between different cell types and therefore needs to be viewed as an evolutionary game.
Collapse
Affiliation(s)
- Benjamin Wölfl
- Department of Mathematics, University of Vienna, Vienna, Austria
- Vienna Graduate School of Population Genetics, Vienna, Austria
| | - Hedy te Rietmole
- Department of Molecular Cancer Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Monica Salvioli
- Department of Mathematics, University of Trento, Trento, Italy
- Department of Data Science and Knowledge Engineering, Maastricht University, Maastricht, The Netherlands
| | - Artem Kaznatcheev
- Department of Biology, University of Pennsylvania, Philadelphia, USA
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Frank Thuijsman
- Department of Data Science and Knowledge Engineering, Maastricht University, Maastricht, The Netherlands
| | - Joel S. Brown
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL USA
| | - Boudewijn Burgering
- Department of Molecular Cancer Research, University Medical Center Utrecht, Utrecht, The Netherlands
- The Oncode Institute, Utrecht, The Netherlands
| | - Kateřina Staňková
- Department of Data Science and Knowledge Engineering, Maastricht University, Maastricht, The Netherlands
- Department of Engineering Systems and Services, Faculty of Technology, Policy and Management, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
45
|
Valente A, Podolski-Renić A, Poetsch I, Filipović N, López Ó, Turel I, Heffeter P. Metal- and metalloid-based compounds to target and reverse cancer multidrug resistance. Drug Resist Updat 2021; 58:100778. [PMID: 34403910 DOI: 10.1016/j.drup.2021.100778] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022]
Abstract
Drug resistance remains the major cause of cancer treatment failure especially at the late stage of the disease. However, based on their versatile chemistry, metal and metalloid compounds offer the possibility to design fine-tuned drugs to circumvent and even specifically target drug-resistant cancer cells. Based on the paramount importance of platinum drugs in the clinics, two main areas of drug resistance reversal strategies exist: overcoming resistance to platinum drugs as well as multidrug resistance based on ABC efflux pumps. The current review provides an overview of both aspects of drug design and discusses the open questions in the field. The areas of drug resistance covered in this article involve: 1) Altered expression of proteins involved in metal uptake, efflux or intracellular distribution, 2) Enhanced drug efflux via ABC transporters, 3) Altered metabolism in drug-resistant cancer cells, 4) Altered thiol or redox homeostasis, 5) Altered DNA damage recognition and enhanced DNA damage repair, 6) Impaired induction of apoptosis and 7) Altered interaction with the immune system. This review represents the first collection of metal (including platinum, ruthenium, iridium, gold, and copper) and metalloid drugs (e.g. arsenic and selenium) which demonstrated drug resistance reversal activity. A special focus is on compounds characterized by collateral sensitivity of ABC transporter-overexpressing cancer cells. Through this approach, we wish to draw the attention to open research questions in the field. Future investigations are warranted to obtain more insights into the mechanisms of action of the most potent compounds which target specific modalities of drug resistance.
Collapse
Affiliation(s)
- Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Isabella Poetsch
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nenad Filipović
- Department of Chemistry and Biochemistry, Faculty of Agriculture, University of Belgrade, Belgrade, Serbia
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
46
|
Amplified antitumor efficacy by a targeted drug retention and chemosensitization strategy-based "combo" nanoagent together with PD-L1 blockade in reversing multidrug resistance. J Nanobiotechnology 2021; 19:200. [PMID: 34225744 PMCID: PMC8256488 DOI: 10.1186/s12951-021-00947-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/28/2021] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND Recent studies have demonstrated that multidrug resistance (MDR) is a critical factor in the low efficacy of cancer chemotherapy. The main mechanism of MDR arises from the overexpression of P-glycoprotein (P-gp), which actively enhances drug efflux and limits the effectiveness of chemotherapeutic agents. RESULTS In this study, we fabricated a "combo" nanoagent equipping with triple synergistic strategies for enhancing antitumor efficacy against MDR cells. Tumor homing-penetrating peptide endows the nanosystem with targeting and penetrating capabilities in the first stage of tumor internalization. The abundant amine groups of polyethylenimine (PEI)-modified nanoparticles then trigger a proton sponge effect to promote endo/lysosomal escape, which enhances the intracellular accumulation and retention of anticancer drugs. Furthermore, copper tetrakis(4-carboxyphenyl)porphyrin (CuTCPP) encapsulated in the nanosystem, effectively scavenges endogenous glutathione (GSH) to reduce the detoxification mediated by GSH and sensitize the cancer cells to drugs, while simultaneously serving as a photoacoustic imaging (PAI) contrast agent for image visualization. Moreover, we also verify that these versatile nanoparticles in combination with PD-1/PD-L1 blockade therapy can not only activate immunological responses but also inhibit P-gp expression to obliterate primary and metastatic tumors. CONCLUSION This work shows a significant enhancement in therapeutic efficacy against MDR cells and syngeneic tumors by using multiple MDR reversing strategies compared to an equivalent dose of free paclitaxel.
Collapse
|
47
|
Kang W, Ji Y, Cheng Y. Van der Waals force-driven indomethacin-ss-paclitaxel nanodrugs for reversing multidrug resistance and enhancing NSCLC therapy. Int J Pharm 2021; 603:120691. [PMID: 33965541 DOI: 10.1016/j.ijpharm.2021.120691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/12/2021] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
The high expression of multidrug resistance-associated protein 1 (MRP1) in cancer cells caused serious multidrug resistance (MDR), which limited the effectiveness of paclitaxel (PTX) in non-small cell lung cancer (NSCLC) chemotherapy. Indomethacin (IND), a kind of non-steroidal anti-inflammatory drugs (NSAIDs), which has been confirmed to be a potential MRP1 inhibitor. Taking into account the advantages of old drugs without extra controversial biosafety issue, in this manuscript, the disulfide bond (-S-S-) was employed for connecting IND and PTX to construct conjugate IND-S-S-PTX, which was further self-assembled and formed nanodrug (IND-S-S-PTX NPs). The particle size of IND-S-S-PTX NPs was ~160 nm with a narrow PDI value of 0.099, which distributed well in water and also exhibited a stable characteristic. Moreover, due to the existence of disulfide bond, the NPs were sensitive to the high level of glutathione (GSH) in tumor microenvironment. Molecular dynamics (MD) simulation presented the process of self-assembly in detail. Density functional theory (DFT) calculations revealed that the main driving force in self-assembly process was originated from the van der waals force. In addition, this carrier-free nano drug delivery systems (nDDs) could reverse the MDR by downregulating the expression of MRP1 protein in A549/taxol.
Collapse
Affiliation(s)
- Wenbo Kang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| | - Yu Cheng
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| |
Collapse
|
48
|
Teng YN, Lin KI, Lin YC, Thang TD, Lan YH, Hung CC. A novel flavonoid from Fissistigma cupreonitens, 5‑hydroxy‑7,8‑dimethoxyflavanone, competitively inhibited the efflux function of human P-glycoprotein and reversed cancer multi-drug resistance. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153528. [PMID: 33735724 DOI: 10.1016/j.phymed.2021.153528] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/03/2021] [Accepted: 02/22/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND P-glycoprotein (P-gp) over-expression plays a vital role in not only systemic drug bioavailability but also cancer multi-drug resistance (MDR). Develop functional inhibitors of P-gp can conquer both problems. PURPOSE AND STUDY DESIGN The aim of the present study was to research the P-gp modulating effects and MDR reversing ability of a novel flavonoid from Fissistigma cupreonitens, the underlying inhibitory mechanisms were further elucidated as well. METHODS Calcein-AM, rhodamine 123, and doxorubicin were fluorescent substrates for the evaluation of P-gp inhibitory function and detailed drug binding modes. Docking simulation was performed to reveal the in silico molecular bonding. ATPase assay and MDR1 shift assay were adopted to reveal the ATP consumption and conformational change of P-gp. The MDR reversing effects were demonstrated through cytotoxicity, cell cycle, and apoptosis analyses. RESULTS 5‑hydroxy‑7,8‑dimethoxyflavanone inhibited the efflux of rhodamine 123 and doxorubicin in a competitive manner, and increased the intracellular fluorescence of calcein at a concentration as low as 2.5 μg/ml. 5‑hydroxy‑7,8‑dimethoxyflavanone slightly changed P-gp's conformation and only stimulated ATPase at very high concentration (100 μg/ml). The docking results showed that 5‑hydroxy‑7,8‑dimethoxyflavanone and verapamil exhibited similar binding affinity to P-gp. The MDR reversing effects were prominent in the vincristine group, the reversal folds were 23.01 and 13.03 when combined with 10 μg/ml 5‑hydroxy‑7,8‑dimethoxyflavanone in the P-gp over-expressing cell line (ABCB1/Flp-In™-293) and MDR cancer cell line (KB/VIN), respectively. CONCLUSION The present study demonstrated that 5‑hydroxy‑7,8‑dimethoxyflavanone was a novel effective flavonoid in the P-gp efflux inhibition and in vitro cancer MDR reversion.
Collapse
Affiliation(s)
- Yu-Ning Teng
- School of Medicine, College of Medicine, I-Shou University, 8 Yida Road, Kaohsiung 82445, Taiwan, R.O.C..
| | - Kun-I Lin
- Department of Obstetrics and Gynecology, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.; Department of Cosmetic Science, Providence University, Taichung, Taiwan, R.O.C..
| | - Yu-Chao Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, R.O.C.; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan, R.O.C.
| | - Tran-Dinh Thang
- Institute of Biotechnology and Food Technology, Industrial University of Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| | - Yu-Hsuan Lan
- Department of Pharmacy, College of Pharmacy, China Medical University, 100, Sec. 1, Jingmao Rd., Beitun Dist., Taichung City 406040, Taiwan, R.O.C..
| | - Chin-Chuan Hung
- Department of Pharmacy, College of Pharmacy, China Medical University, 100, Sec. 1, Jingmao Rd., Beitun Dist., Taichung City 406040, Taiwan, R.O.C.; Department of Pharmacy, China Medical University Hospital, 2 Yude Road, Taichung 40447, Taiwan, R.O.C.; Department of Healthcare Administration, Asia University, 500, Lioufeng Rd., Wufeng, Taichung, 41354, Taiwan.
| |
Collapse
|
49
|
Stefan E, Obexer R, Hofmann S, Vu Huu K, Huang Y, Morgner N, Suga H, Tampé R. De novo macrocyclic peptides dissect energy coupling of a heterodimeric ABC transporter by multimode allosteric inhibition. eLife 2021; 10:67732. [PMID: 33929325 PMCID: PMC8116058 DOI: 10.7554/elife.67732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/29/2021] [Indexed: 12/16/2022] Open
Abstract
ATP-binding cassette (ABC) transporters constitute the largest family of primary active transporters involved in a multitude of physiological processes and human diseases. Despite considerable efforts, it remains unclear how ABC transporters harness the chemical energy of ATP to drive substrate transport across cell membranes. Here, by random nonstandard peptide integrated discovery (RaPID), we leveraged combinatorial macrocyclic peptides that target a heterodimeric ABC transport complex and explore fundamental principles of the substrate translocation cycle. High-affinity peptidic macrocycles bind conformationally selective and display potent multimode inhibitory effects. The macrocycles block the transporter either before or after unidirectional substrate export along a single conformational switch induced by ATP binding. Our study reveals mechanistic principles of ATP binding, conformational switching, and energy transduction for substrate transport of ABC export systems. We highlight the potential of de novo macrocycles as effective inhibitors for membrane proteins implicated in multidrug resistance, providing avenues for the next generation of pharmaceuticals.
Collapse
Affiliation(s)
- Erich Stefan
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt, Germany
| | - Richard Obexer
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Susanne Hofmann
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt, Germany
| | - Khanh Vu Huu
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| | - Yichao Huang
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Nina Morgner
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
50
|
Mehendale-Munj S, Sawant S. Breast Cancer Resistance Protein: A Potential Therapeutic Target for Cancer. Curr Drug Targets 2021; 22:420-428. [PMID: 33243119 DOI: 10.2174/1389450121999201125200132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/05/2020] [Accepted: 10/14/2020] [Indexed: 11/22/2022]
Abstract
Breast Cancer Resistance Protein (BCRP) is an efflux transporter responsible for causing multidrug resistance (MDR). It is known to expel many potent antineoplastic drugs, owing to its efflux function. Efflux of chemotherapeutics because of BCRP develops resistance to many drugs, leading to failure in cancer treatment. BCRP plays an important role in physiology by protecting the organism from xenobiotics and other toxins. It is a half-transporter affiliated to the ATP- binding cassette (ABC) superfamily of transporters, encoded by the gene ABCG2 and functions in response to adenosine triphosphate (ATP). Regulation of BCRP expression is critically controlled at molecular levels, which help in maintaining the balance of xenobiotics and nutrients inside the body. Expression of BCRP can be found in brain, liver, lung cancers and acute myeloid leukemia (AML). Moreover, it is also expressed at high levels in stem cells and many cell lines. This frequent expression of BCRP has an impact on the treatment procedures and, if not scrutinized, may lead to the failure of many cancer therapies.
Collapse
Affiliation(s)
- Sonali Mehendale-Munj
- Department of Pharmaceutical Chemistry, Vivekanand Education Society's College of Pharmacy, Hashu Advani Memorial Complex, Behind Collector's Colony, Chembur (E), Mumbai 400074, Affiliated to University of Mumbai, Maharashtra, India
| | - Shivangi Sawant
- Department of Pharmaceutical Chemistry, Vivekanand Education Society's College of Pharmacy, Hashu Advani Memorial Complex, Behind Collector's Colony, Chembur (E), Mumbai 400074, Affiliated to University of Mumbai, Maharashtra, India
| |
Collapse
|