1
|
Fadaly WAA, Nemr MTM, Abd El-Hameed AM, Giovannuzzi S, Alkabbani MA, Hefina MM, Nocentini A, Mohamed MFA, Supuran CT, Eldehna WM, Zidan TH. Novel benzenesulfonamide derivatives linked to diaryl pyrazole tail as potential carbonic anhydrase II/VII inhibitors with anti-epileptic activity. Eur J Med Chem 2025; 291:117619. [PMID: 40249969 DOI: 10.1016/j.ejmech.2025.117619] [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: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Two new series of 1,2,3-triazole benzenesulfonamide derivatives 16a-f and imino-thiazolidinone benzenesulfonamide derivatives 19a-f with diaryl pyrazole tail were synthesized as carbonic anhydrase (CA) II, VII inhibitors and assessed for antiepileptic activity. All compounds were tested in vitro for their inhibition activity against the human (h) carbonic anhydrase I, II, and VII isoforms. Among these series, compounds 16b, 16d, 19b, and 19d exhibited exceptional inhibitory activity against hCA II, with Ki 10.9-47.1 nM, and hCA VII, with Ki 8.4-23.6 nM, while the two series did not show significant activity against hCA I. Furthermore, 16b, 16d, 19b, and 19d were tested against in vivo pilocarpine-induced seizure model, and they showed excellent neuroprotective activity; they delayed seizure onset, reduced seizure severity, and improved survival rates compared to the pilocarpine group, which highlighted their efficacy in regulating neuronal excitability through CA inhibition and chloride homeostasis. Also, hippocampal levels of KCC2 and mTOR were analyzed, as these markers are critical in regulating neuronal excitability and are closely linked to epilepsy. Noteworthy, Compounds 16d and 19b surpassed the standard anti-convulsant valproic acid in key parameters, underscoring their superior efficacy. In addition, they do not show any significant neurotoxic effects or alterations in liver and kidney function. Moreover, the results of in vitro cytotoxicity of compounds 16d and 19b against Vero cells indicate their safety at the doses given (IC50 = 59.7, 71.9 μM respectively) compared to acetazolamide (IC50 = 32.3 μM). Finally, molecular docking of sulfonamide derivatives with hCA II (PDB code: 2h4h) and hCA VII (PDB code: 3ml5) was performed.
Collapse
Affiliation(s)
- Wael A A Fadaly
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini street 11562, Cairo, Egypt
| | - Abeer M Abd El-Hameed
- Chemistry Department, Faculty of Science, Taibah University, P.O. BOX 30002, Al-Madinah Al-Munawarah, 14177, Saudi Arabia
| | - Simone Giovannuzzi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, Sesto Fiorentino, 50019, Firenze, Italy
| | - Mahmoud Abdelrahman Alkabbani
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo, 11829, Egypt
| | - Mohamed M Hefina
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt
| | - Alessio Nocentini
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, Sesto Fiorentino, 50019, Firenze, Italy
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, 82524, Sohag, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, New Valley University, New Valley, 72511, Egypt
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, Sesto Fiorentino, 50019, Firenze, Italy
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria, 21648, Egypt.
| | - Taha H Zidan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| |
Collapse
|
2
|
Yang X, He C, Wang F, Wen L, Huang H, Wang J, Yang Z, Zhu H. Synthesis and Construction of I-124 Labeled Small Molecular Probe for Noninvasive PET Imaging of CAIX Expression. Mol Imaging Biol 2025:10.1007/s11307-025-02004-5. [PMID: 40205275 DOI: 10.1007/s11307-025-02004-5] [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: 11/22/2024] [Revised: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE Carbonic anhydrase IX (CAIX) which is high expression in the most of hypoxic tumor than normal tissue, promoting the growth, invasion, and metastasis of the tumor. Therefore, the study aimed to evaluate the retention and diagnostic ability of [124I]I-XYIMSR- 01 in CAIX-overexpression tumor by using positron emission tomography (PET) imaging. PROCEDURES [124/125I]I-XYIMSR- 01 was labeled by 124/125I, and its CAIX-targeting properties in different cell lines were assayed by cell uptaken study. Its diagnose and retention ability in vivo were verified in different CAIX-expression models using PET imaging and biodistribution study. Pathological tissues were obtained for immunohistochemical (IHC) and Hematoxylin-Eosin (HE) staining to explore the relationship between CAIX and hypoxic, and further analyze PET/CT results. RESULTS [124I]I-XYIMSR- 01 was obtained with high specific activity, good radiochemical purity, and good stability. The uptake of of [124I]I-XYIMSR- 01 in HT- 29 cells, which have high CAIX expression, was significant higher than that in HCT116 cells with low CAIX expression (12.78 ± 0.47 vs 1.06 ± 0.10, p = 0.000, at 1 h). This indicated that the probe has good targeting capability and specificity for CAIX. In Micro-PET imaging, clear molecular images lasting for 48 h were achieved in HT29 model. Quantitative biodistribution results showed that the tumor and digestive tract background tissues had a good signal-to-noise ratio within 24 h after injection, indicating [124I]I-XYIMSR- 01 could enable delayed imaging in digestive tract tumors (tumor-to-small intestine: 8.79 ± 0.98). Tumors uptakes were also confirmed by IHC pathology. CONCLUSION The study have shown that [124I]I-XYIMSR- 01 is an ideal molecular probe for tumor hypoxia, enabling long-term dynamic monitoring and imaging of hypoxic tumors.
Collapse
Affiliation(s)
- Xianteng Yang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Chengxue He
- Department of Nuclear Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Feng Wang
- Department of Nuclear Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Li Wen
- Department of Nuclear Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Haifeng Huang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Jing Wang
- Department of Chemical Biology, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhi Yang
- Department of Nuclear Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Hua Zhu
- Department of Nuclear Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
3
|
Vaškevičius A, Trumpickaitė G, Parafjanovič E, Manakova E, Mickevičiūtė A, Gedgaudas M, Kojis T, Paketurytė-Latvė V, Smirnov A, Baranauskiene L, Gražulis S, Zubrienė A, Dudutienė V, Matulis D. Design of Rigid Compounds to Enhance Selectivity for Carbonic Anhydrase IX. Chemistry 2025; 31:e202404409. [PMID: 39905940 DOI: 10.1002/chem.202404409] [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: 11/29/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/06/2025]
Abstract
High affinity and selectivity for intended targets is an important goal of small molecule design in drug discovery, yet balancing molecular flexibility and rigidity remains a challenge. While flexible compounds can increase target affinity, they often result in non-specific interactions and reduced selectivity. In contrast, rigid compounds may recognize their target more precisely and have lower off-target effects. In this study, we incorporated a 1,1-dioxido-1,4-thiazine ring into fluorinated benzenesulfonamide derivatives with bulky meta-substituents to enhance selectivity for human carbonic anhydrase IX (CAIX), an important cancer-associated target. Due to the structural similarities of CAIX with other carbonic anhydrase isozymes, selective inhibition remains a significant challenge. A series of 3,4-substituted trifluorobenzenesulfonamides containing oxidized thiazine rings were synthesized using a novel synthetic pathway. Although the potency against CAIX was modestly reduced compared to more flexible analogs, selectivity increased significantly, with lead compounds 7 d and 7 e exhibiting over 1000-fold selectivity for CAIX over most other isozymes. X-ray crystallography revealed the structural basis for this selectivity, confirming the advantageous positioning of rigidified compounds within some CA isozyme active sites. These findings highlight the potential of molecular rigidity in the design of highly selective inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Aivaras Vaškevičius
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Gabrielė Trumpickaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Edvin Parafjanovič
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Elena Manakova
- Department of Protein - DNA Interactions, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, 10257, Vilnius, Lithuania
| | - Aurelija Mickevičiūtė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Marius Gedgaudas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Tautvydas Kojis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Vaida Paketurytė-Latvė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Alexey Smirnov
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Lina Baranauskiene
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Saulius Gražulis
- Sector of Crystallography and Chemical Informatics, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, 10257, Vilnius, Lithuania
| | - Asta Zubrienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Virginija Dudutienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius, LT-10257, Lithuania
| |
Collapse
|
4
|
Nencetti S, Cuffaro D, Ciccone L, Nocentini A, Di Stefano M, Poli G, Macchia M, Tuccinardi T, Nuti E, Supuran CT, Rossello A, Orlandini E. A series of benzensulfonamide derivatives as new potent carbonic anhydrase IX and XII inhibitors. Future Med Chem 2025; 17:271-285. [PMID: 39878534 PMCID: PMC11792798 DOI: 10.1080/17568919.2025.2453420] [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: 06/28/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
AIM Human carbonic anhydrases (hCAs) are involved in many physiological processes including respiration, pH control, ion transport, bone resorption, and gastric fluid secretion. Recently, CA IX and CA XII have been studied for their role in cancer diseases, motivating the design of inhibitors of these isoforms. MATERIAL AND METHOD Here, we used the tail approach to design a new series of monoaryl (1a-i) and bicyclic (1j-n) benzensulfonamide derivatives CA IX and CA XII inhibitors. All synthesized compounds were investigated toward a panel of hCAs, and most of them exhibited potent CA inhibitory activity for CA II, CA IX and CA XII with Ki values. In silico studies were performed to investigate the binding mode between inhibitors and CA. RESULTS AND CONCLUSION The best compound was 1i that showed a low nanomolar range of Ki value as CA inhibitor (Ki = 9.4, 5.6 and 6.3 nM hCA II, IX and XII, respectively).
Collapse
Affiliation(s)
| | | | - Lidia Ciccone
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Alessio Nocentini
- Department of Neurofarba, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Sesto Fiorentino, Italy
| | | | - Giulio Poli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Elisa Nuti
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Claudiu T. Supuran
- Department of Neurofarba, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Sesto Fiorentino, Italy
| | - Armando Rossello
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Research Center “E. Piaggio” Università di, Pisa, Italy
| | - Elisabetta Orlandini
- Research Center “E. Piaggio” Università di, Pisa, Italy
- Department of Earth Sciences, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
Maddipatla S, Bakchi B, Shinde MA, Bonardi A, Raman PK, Bhalerao HA, Singampalli A, Nanduri S, Godugu C, Sonti R, Supuran CT, Yaddanapudi VM. New 7-hydroxycoumarin acetamide derivatives as human carbonic anhydrase IX and XII inhibitors: Design, synthesis, biological evaluation and molecular docking studies. Arch Pharm (Weinheim) 2025; 358:e2400482. [PMID: 39686882 DOI: 10.1002/ardp.202400482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
Carbonic anhydrases (CAs) are crucial in regulating various physiological processes in the body. The overexpression of isoforms human carbonic anhydrases (hCA) IX and hCA XII is linked to tumour progression. The selective inhibition of CA IX and CA XII isoforms can result in the development of better cancer treatment strategies. The tail approach based on coumarin derivatives was known for selective inhibition of isoforms IX and XII. This study explores the potential of coumarin derivatives (7a-k, 8a-s and 9a-g) as selective hCA IX and hCA XII inhibitors. The synthesised derivatives exhibited potent and selective inhibition towards hCA IX and XII, with Ki values in the range of 0.58‒3.33 µM and 0.48‒2.59 µM, respectively. The oxime ether derivative 7d was found to be the most potent one against hCA IX, with a Ki value of 0.58 µM, and phenyl hydrazine derivative 8a, with a Ki value of 0.48 µM against hCA XII, was the most potent one among the synthesised molecules. The potent isoform-specific carbonic anhydrase IX and XII inhibition suggests that 7d and 8a can be taken further towards the development of potent anticancer agents.
Collapse
Affiliation(s)
- Sarvan Maddipatla
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Bulti Bakchi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Mayura Anil Shinde
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Alessandro Bonardi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Sesto Fiorentino, Firenze, Italy
| | - Preethi K Raman
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Harshada Anil Bhalerao
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Anuradha Singampalli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Sesto Fiorentino, Firenze, Italy
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| |
Collapse
|
6
|
Fadaly WAA, Mohamed FEA, Nemr MTM, Sayed AM, Khalil RG, Zidan TH. Novel benzenesulfonamide derivatives as potential selective carbonic anhydrase IX, XII inhibitors with anti-proliferative activity: Design, synthesis and in silico studies. Bioorg Chem 2024; 153:107881. [PMID: 39396453 DOI: 10.1016/j.bioorg.2024.107881] [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: 08/02/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
As inhibitors of carbonic anhydrases (CAs) IX and XII, a novel series of 1,2,3-triazole benzenesulfonamide derivatives 17a-l containing pyrazolyl-thiazole moiety was designed, synthesized, and tested for anti-proliferative activity. Compounds 17e-h demonstrated more effective inhibitory activity than acetazolamide (IC50 63 nM CA IX and IC50 92 nM CA XII), with IC50 range of 25-52 nM against CA IX and IC50 range of 31-80 nM against CA XII. To verify selectivity against CA IX and CA XII, carbonic anhydrase inhibitory activity of compounds 17e-h against the physiological CA I and CA II isoforms was carried out. The results showed that compounds 17e-h induced lower inhibitory activity against CA I and CA II with IC50 range of 0.428-0.638 μM (CA I) and 0.095-0.164 μM (CA II), in addition to higher selectivity indices (CA I/CA IX S.I. 8.9-19.92, CA I/CA XII S.I. 5.78-16.06) and (CA II/CA IX S.I. 2.83-4.35, CA II/CA XII S.I. 2.05-3.15) when compared to that of acetazolamide, IC50 of 0.199 μM (CA I), 0.133 μM (CA II) (CA I/CA IX S.I. 3.15, CA I/CA XII S.I. 2.16) and (CA II/CA IX S.I. 2.11, CA II/CA XII S.I. 1.44). Concerning anti-proliferative activity of compounds 17e-h, investigations were done on HEPG-2 cell line with IC50 ranges of 3.44-15.03 μM in comparison, 5-FU and doxorubicin showed IC50 values of 11.80 and 9.53 μM, respectively. Furthermore IC50 of MCF-7 and MDA-MB-231 were determined under both normoxic and hypoxic conditions with IC50 values ranging from 3.18-8.26 μM MCF-7 (normoxic), 1.39-6.05 μM MCF-7 (hypoxic), 7.13-26.3 μM MDA-MB-231 (normoxic), 0.76-16.3 μM MDA-MB-231 (hypoxic) using acetazolamide and SLC-0111 as selective CA inhibition references. Moreover, compounds 17e-h demonstrated greater safety against the normal cell line, MCF-10A, with IC50 of 23.06-99.50 μM in comparison to 5-FU and doxorubicin IC50 of 59.8 and 71.8 μM respectively. They also demonstrated (MCF-7 S.I. range of 3.77-31.28) in contrast to doxorubicin (S.I. 13.72) and (HepG-2 S.I. range of 3.60-6.95) in comparison to doxorubicin (S.I. 7.53). In relation to CA IX, XII inhibition, molecular docking of and ADME studies of sulfonamide derivatives 17a-l with CA IX (PDB: 5FL6) and CA XII (PDB: 1JD0) was carried out. Additionally, molecular dynamic simulation was carried out for compounds 17e and 17g which maintained good stability inside the active sites of both enzymes, with average RMSDs of 2.3 Å and 2.1 Å, respectively.
Collapse
Affiliation(s)
- Wael A A Fadaly
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Fatma E A Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini Street, 11562 Cairo, Egypt.
| | - Ahmed M Sayed
- Department of Pharmacognosy, Collage of Pharmacy, Almaaqal University, 61014 Basrah, Iraq
| | - Rehab G Khalil
- Immunology Division, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Taha H Zidan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
7
|
Karakuş S, Başçıl E, Tok F, Erdoğan Ö, Çevik Ö, Başoğlu F. Synthesis, biological evaluation and molecular docking studies of novel 1,3,4-thiadiazoles as potential anticancer agents and human carbonic anhydrase inhibitors. Mol Divers 2024; 28:3801-3815. [PMID: 38123787 DOI: 10.1007/s11030-023-10778-5] [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: 09/02/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
Thiosemicarbazide and also 1,3,4-thiadiazole derivatives have been garnering substantial attention from researchers worldwide due to their expansive range of biological activities, encompassing antimicrobial, anti-inflammatory, and anticancer properties. Herein, we embarked on a comprehensive investigation in this study, introducing a novel series of thiosemicarbazides (3a-3i) and their corresponding 1,3,4-thiadiazole (4a-4i) derivatives. The compounds were meticulously designed, synthesized, and subjected to meticulous characterization using various spectroscopic methods such as FT-IR, 1H-NMR, 13C-NMR, and elemental analysis. Afterward, their potential anti-proliferative effectiveness was assessed using MTT assay against two cancer cell lines (U87 and HeLa) and normal fibroblast cells (L929). Among the compounds, 4d showed the highest cytotoxic activity against U87 and 4i against HeLa. Compound 3b exhibited selective cytotoxic activity against both cancer cells. Among the molecules with selective activity against the U87 cell line; 3a, 3b, 4d and 4e were further evaluated by caspase-3 activity levels, Bax and Bcl-2 protein expression, and total oxidant status assay. Besides, carbonic anhydrase IX activity studies were also performed in order to understand the underlying mechanism of action. The results indicated that compound 4e showed higher efficacy than standard acetazolamide (IC50 = 0.58 ± 0.02 µM) with an IC50 value of 0.03 ± 0.01 µM. Furthermore, molecular docking studies were carried out using carbonic anhydrase IX crystals to determine the compound's interactions with the enzyme's active sites. This comprehensive investigation sheds light on the intricate interplay between molecular structure and biological activity, providing valuable insights into the therapeutic potential of these compounds.
Collapse
Affiliation(s)
- Sevgi Karakuş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, 34854, Istanbul, Türkiye
| | - Elif Başçıl
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, 34854, Istanbul, Türkiye
| | - Fatih Tok
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, 34854, Istanbul, Türkiye.
| | - Ömer Erdoğan
- Department of Medical Biochemistry, Faculty of Medicine, Gaziantep Islam Science and Technology University, 27010, Gaziantep, Türkiye
| | - Özge Çevik
- Department of Biochemistry, School of Medicine, Aydın Adnan Menderes University, 09010, Aydın, Türkiye
| | - Faika Başoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, European University of Lefke, Northern Cyprus TR-10, Mersin, Türkiye.
| |
Collapse
|
8
|
Mapar M, Taghdir M, Ranjbar B. Comparative study of stability and activity of wild-type and mutant human carbonic anhydrase II enzymes using molecular dynamics and docking simulations. Biochem Biophys Res Commun 2024; 734:150720. [PMID: 39353361 DOI: 10.1016/j.bbrc.2024.150720] [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: 05/08/2024] [Revised: 08/31/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
The human carbonic anhydrase II (HCA II) enzyme is a cytosolic protein located in the membrane of red blood cells that reversible hydration of carbon dioxide (CO2). Considering the critical role of the HCA II and the effects of some mutations on the activity and stability of the enzyme in humans, several computational methods are used to study the structure and dynamics of the wild-type and the mutant enzymes with three ligands, CO2, 4-nitrophenyl acetate and acetazolamide. Our results of MD simulation of a wild-type enzyme with 4-nitrophenyl acetate show that it created essential effects on the fluctuation of this enzyme and made it more unstable and less compact than the same enzyme without ligand. In the MD of the mutant enzyme with 4-nitrophenyl acetate ligand, no significant difference is observed between with and without ligand. The affinity of the wild-type enzyme to the 4-nitrophenyl acetate is notably higher than the mutant enzyme with the same ligand. Furthermore, results showed that wild-type and mutant enzymes with CO2 are more favorable in stability and flexibility than the same enzymes without ligand. The MD results of wild-type with acetazolamide indicate instability compare without ligand, but in MD of mutant enzyme with acetazolamide show that it more stable and compact than the same enzyme without ligand. Finally, Comparing protein trajectories to assess the impact of ligands on the stability and activity of HCA II enzymes can have medical applications and can in the engineering and design of new variants of carbonic anhydrase enzyme.
Collapse
Affiliation(s)
- Maryam Mapar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Taghdir
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Bijan Ranjbar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran; Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
Jaitak A, Kumari K, Kounder S, Monga V. Carbonic anhydrases: Moiety appended derivatives, medicinal and pharmacological implications. Bioorg Med Chem 2024; 114:117933. [PMID: 39378610 DOI: 10.1016/j.bmc.2024.117933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/10/2024]
Abstract
In the realm of enzymology, Carbonic anhydrase (CA) emerges as a pivotal protagonist orchestrating the rapid conversion of carbon dioxide and water into bicarbonate ions and hydrogen ions, respectively. Carbonic anhydrase inhibitors (CAIs) are the class of drugs that target various isoforms of the enzyme, and these inhibitors play a crucial role in the treatment and management of multiple diseases such as cancer, glaucoma, high altitude sickness, rheumatoid arthritis, obesity, epilepsy, and sleep apnea. Several structural classes of CAIs developed till date possess unique architects of the pharmacophoric requirements around the central core moiety for the selective targeting of various isoforms of the CA. Recent advancements in drug design and development, along with technologies that aid in structure determination, have led to the development of several isoform-selective inhibitors of CA enzymes. However, their clinical development was hampered by the lack of desired therapeutic efficacy, isoform selectivity and safety profile. This review covers the most recent approaches used by different researchers concerned with the development of isoform-selective carbonic anhydrase inhibitors belonging to distinct structural classes like sulphonamides, carbazoles, selenols, coumarin, organotelluride, topiramate, thiophene, triazole, uracil-modified benzylic amines, and thiourea etc. In addition, their structure-activity relationships, biological evaluation, and in silico studies inlcuding the forthcoming avenues of advancements have been discussed. This review serves as a valuable resource for developing potent and efficacious CAIs with remarkable therapeutic implications; offering insights into their potency, specificity, and potential clinical applications.
Collapse
Affiliation(s)
- Aashish Jaitak
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda 151401, Punjab, India
| | - Khushi Kumari
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda 151401, Punjab, India
| | - Sanjay Kounder
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda 151401, Punjab, India
| | - Vikramdeep Monga
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda 151401, Punjab, India.
| |
Collapse
|
10
|
Dai T, Wang Q, Zhu L, Luo Q, Yang J, Meng X, Wang H, Sun Z. Combined UTMD-Nanoplatform for the Effective Delivery of Drugs to Treat Renal Cell Carcinoma. Int J Nanomedicine 2024; 19:8519-8540. [PMID: 39185349 PMCID: PMC11345023 DOI: 10.2147/ijn.s459960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction The effective accumulation of nanoparticles (NPs) in the tumour area is an important goals of current nanotechnology research, and a targeted nanoplatform is an effective solution. So we designed a multifunctional sound-sensitive targeted NP that combines a sonosensitizer to enable precisely targeted, deep-penetration sonodynamic therapy (SDT) in combination with multimodal imaging for the diagnosis and monitoring of renal cell carcinoma (RCC). Methods ZnPP@PP NPs (ZnPP@PLGA- PFP NPs) were prepared via a double emulsion method, and G250 was covalently attached to the NPs shell via the carbon diimide method. Physicochemical property tests were conducted on the ZnPP@G-PP NPs, including tests of particle size, potential distribution, encapsulation efficiency and drug loading capability. We assessed the targeting ability, the production of reactive oxygen species (ROS) and permeability of the NPs in vitro. Moreover, we evaluated the nanoparticle's multimodal imaging capabilities and therapeutic ability against RCC, both in vitro and in vivo. Results The Znpp@G-PP NPs were successfully constructed, and their general properties showed uniform particle size, negative potential and good stability. The nanoparticles were successfully loaded with ZnPP and connected with G250, showing tumor-specific targeting ability. Under LIFU irradiation, the nanoparticles produced 1O2 by SDT. For RCC, PA/US multi-modal imaging of Znpp@G-PP NPs provide diagnostic information and monitor therapies in real time in 786-O RCC xenografts, with good biocompatibility. With the UTMD, nanoparticles can be effectively targeted into the tumor cells and penetrate into the tumor interior, significantly improving the SDT effect. Experiments in vitro and in vivo showed that the combination of the nanoparticles and LIFU could suppress the tumor, and the therapeutic effect was confirmed by immunohistochemistry. Conclusion ZnPP@G-PP NPs provide a promising theranostic strategy for RCC and a platform for further research on improving the efficacy of diagnosis and treatment.
Collapse
Affiliation(s)
- Ting Dai
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Qimeihui Wang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Lingyu Zhu
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Qiang Luo
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Jiayu Yang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Xia Meng
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Hui Wang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Zhixia Sun
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
11
|
Sheta YS, Sarg MT, Abdulrahman FG, Nossier ES, Husseiny EM. Novel imidazolone derivatives as potential dual inhibitors of checkpoint kinases 1 and 2: Design, synthesis, cytotoxicity evaluation, and mechanistic insights. Bioorg Chem 2024; 149:107471. [PMID: 38823311 DOI: 10.1016/j.bioorg.2024.107471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Applying various drug design strategies including ring variation, substituents variation, and ring fusion, two series of 2-(alkylthio)-5-(arylidene/heteroarylidene)imidazolones and imidazo[1,2-a]thieno[2,3-d]pyrimidines were designed and prepared as dual potential Chk1 and Chk2 inhibitors. The newly synthesized hybrids were screened in NCI 60 cell line panel where the most active derivatives 4b, d-f, and 6a were further estimated for their five dose antiproliferative activity against the most sensitive tumor cells including breast MCF-7 and MDA-MB-468 and non-small cell lung cancer EKVX as well as normal WI-38 cell. Noticeably, increasing the carbon chain attached to thiol moiety at C-2 of imidazolone scaffold elevated the cytotoxic activity. Hence, compounds 4e and 4f, containing S-butyl fragment, exhibited the most antiproliferative activity against the tested cells where 4f showed extremely potent selectivity toward them. As well, compound 6a, containing imidazothienopyrimidine core, exerted significant cytotoxic activity and selectivity toward the examined cells. The mechanistic investigation of the most active cytotoxic analogs was achieved through the evaluation of their inhibitory activity against Chk1 and Chk2. Results revealed that 4f displayed potent dual inhibition of both Chk1 and Chk2 with IC50 equal 0.137 and 0.25 μM, respectively. It also promoted its antiproliferative and Chk suppression activity via EKVX cell cycle arrest at S phase through stimulating the apoptotic approach. The apoptosis induction was also emphasized by elevating the expression of Caspase-3 and Bax, that are accompanied by Bcl-2 diminution. The in silico molecular docking and ADMET profiles of the most active analogs have been carried out to evaluate their potential as significant anticancer drug candidates.
Collapse
Affiliation(s)
- Yasmin S Sheta
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Marwa T Sarg
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Eman S Nossier
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt; The National Committee of Drugs, Academy of Scientific Research and Technology, Cairo 11516, Egypt
| | - Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt.
| |
Collapse
|
12
|
Albelwi FF, Nafie MS, Albujuq NR, Hourani W, Aljuhani A, Darwish KM, Tawfik MM, Rezki N, Aouad MR. Design and synthesis of chromene-1,2,3-triazole benzene sulfonamide hybrids as potent carbonic anhydrase-IX inhibitors against prostate cancer. RSC Med Chem 2024; 15:2440-2461. [PMID: 39026656 PMCID: PMC11253856 DOI: 10.1039/d4md00302k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
Considering the promising effects of molecular hybridization on drug discovery in recent years and the ongoing endeavors to develop bioactive scaffolds tethering the 1,2,3-triazole core, the present study sought to investigate whether the 1,2,3-triazole-linked chromene and benzene sulfonamide nucleus could exhibit activity against the human breast cancer cell line MCF-7 and prostate cancer cell line PC-3. To this end, three focused bioactive series of mono- and -bis-1,2,3-triazoles were effectively synthesized via copper-assisted cycloaddition of mono- and/or di-alkyne chromenone derivatives 2a and b and 9 with several sulfa drug azides 4a-d and 6. The resulting molecular derivatives were tested for cytotoxicity against prostate and breast cancer cells. Among the derivatives, 10a, 10c, and 10e exhibited potent cytotoxicity against PC-3 cells with IC50 values of 2.08, 7.57, and 5.52 μM compared to doxorubicin (IC50 = 2.31 μM) with potent inhibition of CA IX with IC50 values of 0.113, 0.134, and 0.214 μM. The most active compound, 10a, was tested for apoptosis-induction; it induced apoptosis by 31.9-fold cell cycle arrest at the G1-phase. Further, the molecular modeling approach highlighted the relevant binding affinity for the top-active compound 10a against CA IX as one of the most prominent PC-3 prostate cancer-associated biotargets.
Collapse
Affiliation(s)
- Fawzia F Albelwi
- Department of Chemistry, Faculty of Science, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah P.O. 27272 Sharjah United Arab Emirates
- Chemistry Department, Faculty of Science, Suez Canal University P.O. 41522 Ismailia Egypt
| | - Nader R Albujuq
- Department of Chemistry, School of Science, The University of Jordan Amman 11942 Jordan
| | - Wafa Hourani
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University Amman 19392 Jordan
| | - Ateyatallah Aljuhani
- Department of Chemistry, Faculty of Science, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| | - Khaled M Darwish
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University Ismailia 41522 Egypt
| | - Mohamed M Tawfik
- Zoology Department, Faculty of Science, Port Said University Port Said 42526 Egypt
| | - Nadjet Rezki
- Department of Chemistry, Faculty of Science, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| | - Mohamed Reda Aouad
- Department of Chemistry, Faculty of Science, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| |
Collapse
|
13
|
Ronca R, Supuran CT. Carbonic anhydrase IX: An atypical target for innovative therapies in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189120. [PMID: 38801961 DOI: 10.1016/j.bbcan.2024.189120] [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: 12/19/2023] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Carbonic anhydrases (CAs), are metallo-enzymes implicated in several pathophysiological processes where tissue pH regulation is required. CA IX is a tumor-associated CA isoform induced by hypoxia and involved in the adaptation of tumor cells to acidosis. Indeed, several tumor-driving pathways can induce CA IX expression, and this in turn has been associated to cancer cells invasion and metastatic features as well as to induction of stem-like features, drug resistance and recurrence. After its functional and structural characterization CA IX targeting approaches have been developed to inhibit its activity in neoplastic tissues, and to date this field has seen an incredible acceleration in terms of therapeutic options and biological readouts. Small molecules inhibitors, hybrid/dual targeting drugs, targeting antibodies and adoptive (CAR-T based) cell therapy have been developed at preclinical level, whereas a sulfonamide CA IX inhibitor and an antibody entered Phase Ib/II clinical trials for the treatment and imaging of different solid tumors. Here recent advances on CA IX biology and pharmacology in cancer, and its therapeutic targeting will be discussed.
Collapse
Affiliation(s)
- Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; Consorzio Interuniversitario per le Biotecnologie (CIB), Italy.
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, University of Florence, Florence 50019, Italy.
| |
Collapse
|
14
|
Knopf P, Stowbur D, Hoffmann SHL, Hermann N, Maurer A, Bucher V, Poxleitner M, Tako B, Sonanini D, Krishnamachary B, Sinharay S, Fehrenbacher B, Gonzalez-Menendez I, Reckmann F, Bomze D, Flatz L, Kramer D, Schaller M, Forchhammer S, Bhujwalla ZM, Quintanilla-Martinez L, Schulze-Osthoff K, Pagel MD, Fransen MF, Röcken M, Martins AF, Pichler BJ, Ghoreschi K, Kneilling M. Acidosis-mediated increase in IFN-γ-induced PD-L1 expression on cancer cells as an immune escape mechanism in solid tumors. Mol Cancer 2023; 22:207. [PMID: 38102680 PMCID: PMC10722725 DOI: 10.1186/s12943-023-01900-0] [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: 04/14/2023] [Accepted: 11/12/2023] [Indexed: 12/17/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy, yet the efficacy of these treatments is often limited by the heterogeneous and hypoxic tumor microenvironment (TME) of solid tumors. In the TME, programmed death-ligand 1 (PD-L1) expression on cancer cells is mainly regulated by Interferon-gamma (IFN-γ), which induces T cell exhaustion and enables tumor immune evasion. In this study, we demonstrate that acidosis, a common characteristic of solid tumors, significantly increases IFN-γ-induced PD-L1 expression on aggressive cancer cells, thus promoting immune escape. Using preclinical models, we found that acidosis enhances the genomic expression and phosphorylation of signal transducer and activator of transcription 1 (STAT1), and the translation of STAT1 mRNA by eukaryotic initiation factor 4F (elF4F), resulting in an increased PD-L1 expression. We observed this effect in murine and human anti-PD-L1-responsive tumor cell lines, but not in anti-PD-L1-nonresponsive tumor cell lines. In vivo studies fully validated our in vitro findings and revealed that neutralizing the acidic extracellular tumor pH by sodium bicarbonate treatment suppresses IFN-γ-induced PD-L1 expression and promotes immune cell infiltration in responsive tumors and thus reduces tumor growth. However, this effect was not observed in anti-PD-L1-nonresponsive tumors. In vivo experiments in tumor-bearing IFN-γ-/- mice validated the dependency on immune cell-derived IFN-γ for acidosis-mediated cancer cell PD-L1 induction and tumor immune escape. Thus, acidosis and IFN-γ-induced elevation of PD-L1 expression on cancer cells represent a previously unknown immune escape mechanism that may serve as a novel biomarker for anti-PD-L1/PD-1 treatment response. These findings have important implications for the development of new strategies to enhance the efficacy of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Philipp Knopf
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Dimitri Stowbur
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Sabrina H L Hoffmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Natalie Hermann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Valentina Bucher
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Marilena Poxleitner
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Bredi Tako
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanhita Sinharay
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | | | - Irene Gonzalez-Menendez
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Institute of Pathology and Neuropathology, Department of Pathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, Tübingen University Hospital, Tübingen, Germany
| | - Felix Reckmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - David Bomze
- Department of Dermatology, Tel-Aviv Medical Center, Tel-Aviv, Israel
| | - Lukas Flatz
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Daniela Kramer
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tübingen, Germany
| | - Martin Schaller
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | | | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Leticia Quintanilla-Martinez
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Institute of Pathology and Neuropathology, Department of Pathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, Tübingen University Hospital, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Mark D Pagel
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Martin Röcken
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - André F Martins
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 10117, Berlin, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany.
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
15
|
Mahammad Ghouse S, Bahatam K, Angeli A, Pawar G, Chinchilli KK, Yaddanapudi VM, Mohammed A, Supuran CT, Nanduri S. Synthesis and biological evaluation of new 3-substituted coumarin derivatives as selective inhibitors of human carbonic anhydrase IX and XII. J Enzyme Inhib Med Chem 2023; 38:2185760. [PMID: 36876597 PMCID: PMC10013565 DOI: 10.1080/14756366.2023.2185760] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
The Carbonic anhydrase isoforms IX and XII play a significant role in regulating the intracellular and extracellular pH in hypoxic tumours abetting the metastasis of solid tumours. Selective and potent inhibitors targeting carbonic anhydrase IX and XII reduce the activity of these isoforms in hypoxic tumours, representing an antitumor and antimetastatic mechanism. Coumarin-based derivatives are selective inhibitors of CA isoforms IX and XII. In this study, we report the design and synthesis of new 3-substituted coumarin derivatives with different functional moieties and their inhibitory activity against various carbonic anhydrase isoforms. We found that the tertiary sulphonamide derivative 6c showed selective inhibition against CA IX with IC50 of 4.1 µM. Similarly, the carbothioamides 7c, 7b and oxime ether derivative 20a exhibited good inhibition against CA IX and CA XII. Additionally, the binding mode was predicted and validated using molecular docking and dynamic simulations.
Collapse
Affiliation(s)
- Shaik Mahammad Ghouse
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Kavyaraj Bahatam
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Andrea Angeli
- Neurofarba Deptartment, Sezione di ScienzeFarmaceutiche e Nutraceutiche, Università Degli Studi di Firenze, Florence, Italy
| | - Gaurav Pawar
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Krishna Kartheek Chinchilli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Arifuddin Mohammed
- Department of Chemistry, Directorate of Distance Education, Maulana Azad National Urdu University, Hyderabad, India
| | - Claudiu T Supuran
- Neurofarba Deptartment, Sezione di ScienzeFarmaceutiche e Nutraceutiche, Università Degli Studi di Firenze, Florence, Italy
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
16
|
Ibrahim SA, Al-Mhyawi SR, Atlam FM. New imidazole-2-ones and their 2-thione analogues as anticancer agents and CAIX inhibitors: Synthesis, in silico ADME and molecular modeling studies. Bioorg Chem 2023; 141:106872. [PMID: 37776683 DOI: 10.1016/j.bioorg.2023.106872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/02/2023] [Accepted: 09/16/2023] [Indexed: 10/02/2023]
Abstract
The present study involves the synthesis of a series of new imidazole-2-ones derivatives and their 2-thione analogs using conventional heating and the environmentally friendly benign technique, the microwave technique. Structure of the compounds was well elucidated by considering the data of both elemental and spectral analyses. The obtained data and theoretical values of the synthesized molecules correlated with the proposed molecular structure. Moreover, all the synthesized compounds were evaluated in vitro for antitumor activity against HCT-116 and HeP2 human cancer cell panels and assessed as selective carbonic anhydrase IX isozyme (CA9/CAIX) inhibitors, thereby providing useful preliminary evidence for drug development. In addition, computational techniques were used to investigate the molecular and electronic characteristics of the investigated organic compounds. The 4b compound exhibited the best quantum chemistry features, as the highest occupied molecular orbital, softness, energy gap, and dipole moment, indicating the highest biological activity. This was supported by the experimental findings. Moreover, the in silico evaluation of drug candidates was also investigated. Thereafter, the anticancer activity of the most reactive candidate was studied via molecular docking to determine the types of interactions between this molecule and CAIX. According to the docking experiments, the 4b molecule generates five hydrogen bond interactions with active amino acid residues, Gln 92, Gln 67, and Thr 200.
Collapse
Affiliation(s)
- Seham A Ibrahim
- Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Saedah R Al-Mhyawi
- Chemistry Department, College of Science, University of Jeddah, Jeddah 22233, Saudi Arabia
| | - Faten M Atlam
- Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
17
|
Parn S, Lewis G, Knight M. Inhibition of carbonic anhydrase using aspirin is a novel method to block schistosomiasis infection of the parasitic trematode, Schistosoma mansoni, in the intermediate snail host, Biomphalaria glabrata. Exp Parasitol 2023; 254:108618. [PMID: 37696327 DOI: 10.1016/j.exppara.2023.108618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/18/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Schistosomiasis is a major public health concern worldwide. Although praziquantel is currently available as the only treatment option for schistosomiasis, the absence of reliable diagnostic and prognostic tools highlights the need for the identification and characterization of new drug targets. Recently, we identified the B. glabrata homolog (accession number XP_013075832.1) of human CAXIV, showing 37% amino acid sequence identity, from a BLAST search in NCBI (National Center for Biotechnology Information). Carbonic Anhydrases (CAs) are metalloenzymes that catalyze the reversible hydration/dehydration of CO2/HCO3. These enzymes are associated with many physiological processes, and their role in tumorigenesis has been widely implicated. CAs create an acidic extracellular environment that facilitates the survival, metastasis, and growth of cancer cells. In this study, we investigated the role of CA inhibition in B. glabrata snails exposed to S. mansoni miracidia. We analyzed the expression of the B. glabrata CA encoding transcript in juvenile susceptible and resistant snails, with and without exposure to S. mansoni. Our results showed that the expression of the CA mRNA encoding transcript was upregulated during early and prolonged infection in susceptible snails (BBO2), but not in the resistant BS-90 stock. Notably, sodium salicylate, a form of aspirin, inhibited the expression of CA, post-exposure, to the parasite. Increasing research between parasites and cancer has shown that schistosomes and cancer cells share similarities in their capacity to proliferate, survive, and evade host immune mechanisms. Here, we show that this model system is a potential new avenue for understanding the role of CA in the metastasis and proliferation of cancer cells. Further studies are needed to explore the potential of CA as a biomarker for infection in other schistosomiasis-causing parasites, including S. japonicum and S. haematobium.
Collapse
Affiliation(s)
- Simone Parn
- Division of Science & Mathematics, University of the District of Columbia, 4200 Connecticut Ave, NW Washington, D.C., 20008, USA
| | - Gabriela Lewis
- Division of Science & Mathematics, University of the District of Columbia, 4200 Connecticut Ave, NW Washington, D.C., 20008, USA
| | - Matty Knight
- Division of Science & Mathematics, University of the District of Columbia, 4200 Connecticut Ave, NW Washington, D.C., 20008, USA; Department of Microbiology, Immunology & Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, The George Washington University Ross Hall, 2300 I Street, NW Washington DC, 20037, USA.
| |
Collapse
|
18
|
Ghouse SM, Sinha K, Bonardi A, Pawar G, Malasala S, Danaboina S, Mohammed A, Yaddanapudi VM, Supuran CT, Nanduri S. 6-Aminocoumarin oxime-ether/sulfonamides as selective hCA IX and XII inhibitors: Synthesis, evaluation, and molecular dynamics studies. Arch Pharm (Weinheim) 2023; 356:e2300316. [PMID: 37495909 DOI: 10.1002/ardp.202300316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023]
Abstract
Carbonic anhydrase isoforms IX and XII are overexpressed in hypoxic tumor cells regulating various physiological processes such as cell proliferation, invasion, and metastasis, resulting in the onset and spread of cancer. Selective inhibition of these enzymes is a promising strategy for anticancer therapy. Coumarin derivatives were identified as selective and potent inhibitors of these isoforms. This study reports 6-aminocoumarin sulfonamide and oxime ether derivatives linked through a chloroacetyl moiety tethered to piperazine and piperidone, respectively, showing selective inhibition against human carbonic anhydrase (hCA) IX and XII with Ki ranging from 0.51 to 1.18 µM and 0.89-4.43 µM. While the sulfonamide derivative 8a exhibited submicromolar inhibition against hCA IX and XII with Ki 0.89 and 0.51 µM, the oxime ether derivatives showed lower activity than the sulfonamides, with the compound 5n inhibiting hCA IX and hCA XII with a Ki of 1.055 and 0.70 µM, respectively. The above results demonstrate the potential of these derivatives as selective, potent inhibitors of carbonic anhydrase IX and XII and provide a foundation for further optimization and development as effective anticancer agents. Further, the binding mode of the synthesized derivatives in the active site were examined using molecular docking and dynamic simulation studies.
Collapse
Affiliation(s)
- Shaik Mahammad Ghouse
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Kareena Sinha
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Alessandro Bonardi
- Neurofarba Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università Degli Studi di Firenze, Florence, Italy
| | - Gaurav Pawar
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Satyaveni Malasala
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Srikanth Danaboina
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Arifuddin Mohammed
- Department of Chemistry, Directorate of Distance Education, Maulana Azad National Urdu University, Hyderabad, India
| | - Venkata M Yaddanapudi
- Department of Chemistry, Directorate of Distance Education, Maulana Azad National Urdu University, Hyderabad, India
| | - Claudiu T Supuran
- Neurofarba Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università Degli Studi di Firenze, Florence, Italy
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
19
|
Zhu W, Li X, Zheng G, Bai C, Ji Z, Zhang H, Xing H, Zhang Y, Huo L. Preclinical and pilot clinical evaluation of a small-molecule carbonic anhydrase IX targeting PET tracer in clear cell renal cell carcinoma. Eur J Nucl Med Mol Imaging 2023; 50:3116-3125. [PMID: 37246998 DOI: 10.1007/s00259-023-06248-7] [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: 02/17/2023] [Accepted: 04/24/2023] [Indexed: 05/30/2023]
Abstract
PURPOSE Clear cell renal cell carcinoma (ccRCC) highly expresses carbonic anhydrase IX (CAIX). The purpose of this study was to evaluate 68Ga-NY104, a small-molecule CAIX-targeting PET agent, in tumor models of ccRCC and patients diagnosed with confirmed, or suspicious, ccRCC. METHODS The in vivo and ex vivo biodistribution of 68Ga-NY104 was investigated in CAIX-positive OS-RC-2 xenograft-bearing models. The binding of the tracer was further validated using autoradiography for human ccRCC samples. In addition, three patients with confirmed or suspicious ccRCC were studied. RESULTS NY104 can be labeled with high radiochemical yield and purity. It quickly cleared through kidney with α-half-life of 0.15 h. Discernible uptake is noted in the heart, lung, liver, stomach, and kidney. The OS-RC-2 xenograft demonstrated intense uptake 5 min after injection and gradually increased until 3 h after injection with ID%/g of 29.29 ± 6.82. Significant binding was detected using autoradiography on sections of human ccRCC tumor. In the three patients studied, 68Ga-NY104 was well-tolerated and no adverse events were reported. Substantial accumulation was observed in both primary and metastatic lesions in patient 1 and 2 with SUVmax of 42.3. Uptake in the stomach, pancreas, intestine, and choroid plexus was noted. The lesion in third patient was correctly diagnosed as non-metastatic for negative 68Ga-NY104 uptake. CONCLUSION 68Ga-NY104 can efficiently and specifically bind to CAIX. Given the pilot nature of our study, future clinical studies are warranted to evaluate 68Ga-NY104 for detection of CAIX-positive lesions in patients with ccRCC. TRIAL REGISTRATION The clinical evaluation part of this study was retrospectively registered at ClinicalTrial.gov (NCT05728515) as NYPILOT on 6 Feb, 2023.
Collapse
Affiliation(s)
- Wenjia Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Xiaoyuan Li
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guoyang Zheng
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Haiqiong Zhang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Haiqun Xing
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yushi Zhang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Li Huo
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
20
|
Lionetto MG. Carbonic Anhydrase and Biomarker Research: New Insights. Int J Mol Sci 2023; 24:ijms24119687. [PMID: 37298637 DOI: 10.3390/ijms24119687] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Carbonic anhydrase (CA) is a widespread metalloenzyme with eight genetically distinct families catalyzing the reversible hydration of CO2 to HCO3- and H+ [...].
Collapse
Affiliation(s)
- Maria Giulia Lionetto
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), Salento University, 73100 Lecce, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
21
|
Bhuimali M, Munshi S, Hapa K, Kadu PK, Kale PP. Evaluation of liposomes for targeted drug delivery in lung cancer treatment. INT J POLYM MATER PO 2023. [DOI: 10.1080/00914037.2022.2163639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Mitali Bhuimali
- SVKM’S Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Sunya Munshi
- SVKM’S Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Kunali Hapa
- SVKM’S Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Pramod K. Kadu
- Department of Pharmaceutics, SVKM’S Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Pravin P. Kale
- Department of Pharmacology, SVKM’S Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
22
|
Wu R, Wang K, Gai Y, Li M, Wang J, Wang C, Zhang Y, Xiao Z, Jiang D, Gao Z, Xia X. Nanomedicine for renal cell carcinoma: imaging, treatment and beyond. J Nanobiotechnology 2023; 21:3. [PMID: 36597108 PMCID: PMC9809106 DOI: 10.1186/s12951-022-01761-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
The kidney is a vital organ responsible for maintaining homeostasis in the human body. However, renal cell carcinoma (RCC) is a common malignancy of the urinary system and represents a serious threat to human health. Although the overall survival of RCC has improved substantially with the development of cancer diagnosis and management, there are various reasons for treatment failure. Firstly, without any readily available biomarkers, timely diagnosis has been greatly hampered. Secondly, the imaging appearance also varies greatly, and its early detection often remains difficult. Thirdly, chemotherapy has been validated as unavailable for treating renal cancer in the clinic due to its intrinsic drug resistance. Concomitant with the progress of nanotechnological methods in pharmaceuticals, the management of kidney cancer has undergone a transformation in the recent decade. Nanotechnology has shown many advantages over widely used traditional methods, leading to broad biomedical applications ranging from drug delivery, prevention, diagnosis to treatment. This review focuses on nanotechnologies in RCC management and further discusses their biomedical translation with the aim of identifying the most promising nanomedicines for clinical needs. As our understanding of nanotechnologies continues to grow, more opportunities to improve the management of renal cancer are expected to emerge.
Collapse
Affiliation(s)
- Ruolin Wu
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Keshan Wang
- grid.33199.310000 0004 0368 7223Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yongkang Gai
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Mengting Li
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Jingjing Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Chenyang Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Yajing Zhang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zhiwei Xiao
- grid.413247.70000 0004 1808 0969Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dawei Jiang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zairong Gao
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Xiaotian Xia
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| |
Collapse
|
23
|
Párraga-Ros E, Latorre-Reviriego R, Aparicio-González M, Boronat-Belda T, López-Albors O. The immunolocalization of HIF-2α, GLUT1 and CAIX in porcine oviduct during the estrous cycle. Anat Rec (Hoboken) 2023; 306:176-186. [PMID: 35684983 PMCID: PMC10084220 DOI: 10.1002/ar.25014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 01/29/2023]
Abstract
Oxygen (O2 ) rates in the oviduct are essential to human and animal reproduction. These rates are regulated by the activity of hypoxia markers such as the hypoxia-inducible factors (HIFs), the glucose transporters (GLUT), and the carbonic anhydrase (CA). In the porcine model, scarce studies have been reported regarding these markers and their effects in reproduction are unknown. The objective was to characterize the immunolocalization of HIF-2α, GLUT1, and CAIX in porcine oviducts throughout the estrous cycle. Oviducts (ampulla and isthmus) of adult sows (n = 45) were collected for histological and immunohistochemical analysis with HIF-2α, GLUT1, and CAIX markers. The percentage of immunopositive area was quantified, and the differences among phases of the estrous cycle were analyzed (folicular, early luteal, and late luteal). The three markers showed epithelial presence mainly. Significantly lower expression of HIF-2α was found in the luteal phases, especially in the isthmus. GLUT1 expression did not change throughout the estrous cycle, but differences were found between the ampulla and isthmus. CAIX expression showed the highest, with a significant downward trend throughout estrous cycle. The ubiquitous expression of hypoxia markers shows the porcine oviduct physiology in relation to O2 . The differential expression of HIF-2α, GLUT1, and CAIX in different subcompartments of the oviduct throughout the estrous cycle contributes to improve the knowledge of the cell physiology of the oviduct, which can be useful in fertilization studies.
Collapse
Affiliation(s)
- Ester Párraga-Ros
- Anatomy and Comparative Pathology Department, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Rafael Latorre-Reviriego
- Anatomy and Comparative Pathology Department, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Mónica Aparicio-González
- Anatomy and Comparative Pathology Department, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Talía Boronat-Belda
- Unit of Cell Physiology and Nutrition, Miguel Hernández University, Alicante, Spain
| | - Octavio López-Albors
- Anatomy and Comparative Pathology Department, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| |
Collapse
|
24
|
Tawfik HO, Belal A, Abourehab MAS, Angeli A, Bonardi A, Supuran CT, El-Hamamsy MH. Dependence on linkers' flexibility designed for benzenesulfonamides targeting discovery of novel hCA IX inhibitors as potent anticancer agents. J Enzyme Inhib Med Chem 2022; 37:2765-2785. [PMID: 36210545 PMCID: PMC9559471 DOI: 10.1080/14756366.2022.2130285] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Herein we reported the design and synthesis of two series comprising twenty-two benzenesulfonamides that integrate the s-triazine moiety. Target compounds successfully suppressed the hCA IX, with IC50 ranging from 28.6 to 871 nM. Compounds 5d, 11b, 5b, and 7b were the most active analogues, which inhibited hCA IX isoform in the low nanomolar range (KI = 28.6, 31.9, 33.4, and 36.6 nM, respectively). Furthermore, they were assessed for their cytotoxic activity against a panel of 60 cancer cell lines following US-NCI protocol. According to five-dose assay, 13c showed significant anticancer activity than 5c with GI50-MID values of 25.08 and 189.01 µM, respectively. Additionally, 13c's effects on wound healing, cell cycle disruption, and apoptosis induction in NCI-H460 cancer cells were examined. Further, docking studies combined with molecular dynamic simulation showed a stable complex with high binding affinity of 5d to hCA IX, exploiting a favourable H-bond and lipophilic interactions.HIGHLIGHTSCarbonic anhydrase (CA) inhibitors comprising rigid and flexible linkers were developed.Compound 5d is the most potent CA IX inhibitor in the study (IC50: 28.6 nM).Compounds 5c and 13c displayed the greatest antiproliferative activity towards 60 cell lines.Compound 13c exposed constructive outcomes on normal cell lines, metastasis, and wound healing.Molecular docking and molecular dynamics (MDs) simulation was utilised to study binding mode.
Collapse
Affiliation(s)
- Haytham O. Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt,CONTACT H. O. Tawfik Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amany Belal
- Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt,Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mohammed A. S. Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Andrea Angeli
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Alessandro Bonardi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy,C. T. Supuran Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Mervat H. El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
25
|
Amin MU, Ali S, Ali MY, Fuhrmann DC, Tariq I, Seitz BS, Preis E, Brüßler J, Brüne B, Bakowsky U. Co-delivery of carbonic anhydrase IX inhibitor and doxorubicin as a promising approach to address hypoxia-induced chemoresistance. Drug Deliv 2022; 29:2072-2085. [PMID: 35848469 PMCID: PMC9297722 DOI: 10.1080/10717544.2022.2092234] [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] [Indexed: 11/03/2022] Open
Abstract
Hypoxia, an oxygen-deprived condition of the tumor, is one of the major reasons for resistance to chemotherapy. Carbonic anhydrases are generally involved in pH homeostasis in normal conditions, but in solid tumors having a strong relation with hypoxia, the carbonic anhydrase IX (CA-IX) enzyme is overexpressed and results in an extracellular acidic environment. For most weakly basic anticancer drugs, including doxorubicin (Dox), the ionization in an acidic environment limits their cellular uptake, and consequently, the tumor exposure to the drug at sub-therapeutic concentration comes out as chemoresistance. Herein, a combined drug delivery system of liposomes and mesoporous silica nanoparticles (MSNPs) was developed for the co-delivery of the CA-IX enzyme inhibitor and Dox in hypoxic condition. The unique structure of MSNPs with higher surface area was utilized for higher drug loading and sustained release of Dox. Additionally, the biocompatible nature of liposomal coating as a second loading site for the CA-IX enzyme inhibitor has provided gatekeeping effects at pore opening to avoid premature drug release. Lipid coated MSNPs as a co-delivery system for Dox and the CA-IX inhibitor have synergistic cytotoxic effects against MDA-MB 231 breast cancer cells in hypoxic conditions. These findings assure the potential of this drug delivery system to overcome hypoxia-related chemoresistance.
Collapse
Affiliation(s)
- Muhammad Umair Amin
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Sajid Ali
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany.,Department of Chemistry, Angström Laboratory, Uppsala University, Uppsala, Sweden
| | - Muhammad Yasir Ali
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany.,Faculty of Pharmaceutical Sciences, GC University Faisalabad, Faisalabad, Pakistan
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Imran Tariq
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany.,Punjab University College of Pharmacy, University of Punjab, Lahore, Pakistan
| | - Benjamin S Seitz
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Jana Brüßler
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| |
Collapse
|
26
|
Geçkil AA, Kıran TR, Berber NK, Otlu Ö, Erdem M, İn E. Carbonic Anhydrase IX as a Marker of Disease Severity in Obstructive Sleep Apnea. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58111643. [PMID: 36422182 PMCID: PMC9695925 DOI: 10.3390/medicina58111643] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022]
Abstract
Background and Objectives: Carbonic anhydrase (CA) enzymes are a family of metalloenzymes that contain a zinc ion in their active sites. CA enzymes have been implied in important situations such as CO2 transport, pH regulation, and oncogenesis. CA-IX is a transmembrane glycoprotein and stimulates the expression of hypoxia-inducible factor-1 (HIF-1) CA-IX. This study aimed to determine serum CA-IX levels in OSA patients in whom intermittent hypoxia is important and to investigate the relationship between serum CA-IX levels and disease severity. Materials and Methods: The study included 88 people who applied to Malatya Turgut Özal University Training and Research Hospital Sleep Disorders Center without a history of respiratory disease, malignancy, and smoking. Patients were divided into three groups: control (AHI < 5, n = 31), mild−moderate OSA (AHI = 5−30, n = 27) and severe OSA (AHI > 30, n = 30). The analysis of the data included in the research was carried out with the SPSS (IBM Statistics 25, NY, USA). The Shapiro−Wilk Test was used to check whether the data included in the study had a normal distribution. Comparisons were made with ANOVA in multivariate groups and the t-test in bivariate groups. ANCOVA was applied to determine the effect of the CA-IX parameter for OSA by controlling the effect of independent variables. The differentiation in CA-IX and OSA groups was analyzed regardless of BMI, age, gender, and laboratory variables. ROC analysis was applied to determine the parameter cut-off point. Sensitivity, specificity, and cut-off were calculated, and the area under the curve (AUC) value was calculated. Results: Serum CA-IX levels were 126.3 ± 24.5 pg/mL in the control group, 184.6 ± 59.1 pg/mL in the mild−moderate OSA group, and 332.0 ± 39.7 pg/mL in the severe OSA group. Serum CA-IX levels were found to be higher in the severe OSA group compared to the mild−moderate OSA group and control group and higher in the mild−moderate OSA group compared to the control group (p < 0.001, p < 0.001, p < 0.001, respectively). In addition, a negative correlation between CA-IX and minimum SaO2 and mean SaO2 (r = −0.371, p = 0.004; r = −0.319, p = 0.017, respectively). A positive correlation between CA-IX and desaturation index (CT90) was found (r = 0.369, p = 0.005). A positive correlation was found between CA-IX and CRP (r = 0.340, p = 0.010). When evaluated by ROC curve analysis, the area under the curve (AUC) value was determined as 0.940 (95% CI 0.322−0.557; p < 0.001). When the cut-off value for CA-IX was taken as 254.5 pg/mL, it was found to have 96.7% sensitivity and 94.8% specificity in demonstrating severe OSA. Conclusions: Our study found that serum CA-IX value was higher in OSA patients than in control patients, and this elevation was associated with hypoxemia and inflammation. CA-IX value can be a fast, precise, and useful biomarker to predict OSA.
Collapse
Affiliation(s)
- Ayşegül Altıntop Geçkil
- Department of Chest Diseases, Malatya Turgut Özal University, Malatya 44210, Turkey
- Correspondence: ; Tel.: +90-042-2502-8001
| | - Tuğba Raika Kıran
- Department of Biochemistry, Malatya Turgut Özal University, Malatya 44210, Turkey
| | - Nurcan Kırıcı Berber
- Department of Chest Diseases, Malatya Turgut Özal University, Malatya 44210, Turkey
| | - Önder Otlu
- Department of Biochemistry, Malatya Turgut Özal University, Malatya 44210, Turkey
| | - Mehmet Erdem
- Department of Biochemistry, Malatya Turgut Özal University, Malatya 44210, Turkey
| | - Erdal İn
- Department of Chest Diseases, Malatya Turgut Özal University, Malatya 44210, Turkey
| |
Collapse
|
27
|
Functionalization of Nanoparticulate Drug Delivery Systems and Its Influence in Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14051113. [PMID: 35631699 PMCID: PMC9145684 DOI: 10.3390/pharmaceutics14051113] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 12/13/2022] Open
Abstract
Research into the application of nanocarriers in the delivery of cancer-fighting drugs has been a promising research area for decades. On the other hand, their cytotoxic effects on cells, low uptake efficiency, and therapeutic resistance have limited their therapeutic use. However, the urgency of pressing healthcare needs has resulted in the functionalization of nanoparticles' (NPs) physicochemical properties to improve clinical outcomes of new, old, and repurposed drugs. This article reviews recent research on methods for targeting functionalized nanoparticles to the tumor microenvironment (TME). Additionally, the use of relevant engineering techniques for surface functionalization of nanocarriers (liposomes, dendrimers, and mesoporous silica) and their critical roles in overcoming the current limitations in cancer therapy-targeting ligands used for targeted delivery, stimuli strategies, and multifunctional nanoparticles-were all reviewed. The limitations and future perspectives of functionalized nanoparticles were also finally discussed. Using relevant keywords, published scientific literature from all credible sources was retrieved. A quick search of the literature yielded almost 400 publications. The subject matter of this review was addressed adequately using an inclusion/exclusion criterion. The content of this review provides a reasonable basis for further studies to fully exploit the potential of these nanoparticles in cancer therapy.
Collapse
|
28
|
Kciuk M, Gielecińska A, Mujwar S, Mojzych M, Marciniak B, Drozda R, Kontek R. Targeting carbonic anhydrase IX and XII isoforms with small molecule inhibitors and monoclonal antibodies. J Enzyme Inhib Med Chem 2022; 37:1278-1298. [PMID: 35506234 PMCID: PMC9090362 DOI: 10.1080/14756366.2022.2052868] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carbonic anhydrases IX and CAXII (CAIX/CAXII) are transmembrane zinc metalloproteins that catalyze a very basic but crucial physiological reaction: the conversion of carbon dioxide into bicarbonate with a release of the proton. CA, especially CAIX and CAXII isoforms gained the attention of many researchers interested in anticancer drug design due to pivotal functions of enzymes in the cancer cell metastasis and response to hypoxia, and their expression restricted to malignant cells. This offers an opportunity to develop new targeted therapies with fewer side effects. Continuous efforts led to the discovery of a series of diverse compounds with the most abundant sulphonamide derivatives. Here we review current knowledge considering small molecule and antibody-based targeting of CAIX/CAXII in cancer.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland.,Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Somdutt Mujwar
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| |
Collapse
|
29
|
de Campos NSP, Souza BS, da Silva GCP, Porto VA, Chalbatani GM, Lagreca G, Janji B, Suarez ER. Carbonic Anhydrase IX: A Renewed Target for Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14061392. [PMID: 35326544 PMCID: PMC8946730 DOI: 10.3390/cancers14061392] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Carbonic anhydrase IX (CAIX) has been explored for a long time as a therapeutic target in the fight against clear cell renal cell carcinoma and several hypoxic tumors, usually offering modest results followed by adverse effects. However, recent studies using different antibodies and adoptive cell therapies against CAIX have generated exciting prospects for the immunotherapy of these tumors. This complete review will approach the past and future of anti-CAIX immunotherapies. Abstract The carbonic anhydrase isoform IX (CAIX) enzyme is constitutively overexpressed in the vast majority of clear cell renal cell carcinoma (ccRCC) and can also be induced in hypoxic microenvironments, a major hallmark of most solid tumors. CAIX expression is restricted to a few sites in healthy tissues, positioning this molecule as a strategic target for cancer immunotherapy. In this review, we summarized preclinical and clinical data of immunotherapeutic strategies based on monoclonal antibodies (mAbs), fusion proteins, chimeric antigen receptor (CAR) T, and NK cells targeting CAIX against different types of solid malignant tumors, alone or in combination with radionuclides, cytokines, cytotoxic agents, tyrosine kinase inhibitors, or immune checkpoint blockade. Most clinical studies targeting CAIX for immunotherapy were performed using G250 mAb-based antibodies or CAR T cells, developed primarily for bioimaging purposes, with a limited clinical response for ccRCC. Other anti-CAIX mAbs, CAR T, and NK cells developed with therapeutic intent presented herein offered outstanding preclinical results, justifying further exploration in the clinical setting.
Collapse
Affiliation(s)
- Najla Santos Pacheco de Campos
- Center for Natural and Human Sciences, Federal University of ABC, Santo André 09210-580, SP, Brazil; (N.S.P.C.); (B.S.S.); (G.C.P.S.); (V.A.P.); (G.L.)
| | - Bruna Santos Souza
- Center for Natural and Human Sciences, Federal University of ABC, Santo André 09210-580, SP, Brazil; (N.S.P.C.); (B.S.S.); (G.C.P.S.); (V.A.P.); (G.L.)
| | - Giselle Correia Próspero da Silva
- Center for Natural and Human Sciences, Federal University of ABC, Santo André 09210-580, SP, Brazil; (N.S.P.C.); (B.S.S.); (G.C.P.S.); (V.A.P.); (G.L.)
| | - Victoria Alves Porto
- Center for Natural and Human Sciences, Federal University of ABC, Santo André 09210-580, SP, Brazil; (N.S.P.C.); (B.S.S.); (G.C.P.S.); (V.A.P.); (G.L.)
| | - Ghanbar Mahmoodi Chalbatani
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health, 1445 Luxembourg, Luxembourg;
| | - Gabriela Lagreca
- Center for Natural and Human Sciences, Federal University of ABC, Santo André 09210-580, SP, Brazil; (N.S.P.C.); (B.S.S.); (G.C.P.S.); (V.A.P.); (G.L.)
| | - Bassam Janji
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health, 1445 Luxembourg, Luxembourg;
- Correspondence: (B.J.); (E.R.S.)
| | - Eloah Rabello Suarez
- Center for Natural and Human Sciences, Federal University of ABC, Santo André 09210-580, SP, Brazil; (N.S.P.C.); (B.S.S.); (G.C.P.S.); (V.A.P.); (G.L.)
- Correspondence: (B.J.); (E.R.S.)
| |
Collapse
|
30
|
Tawfik HO, Petreni A, Supuran CT, El-Hamamsy MH. Discovery of new carbonic anhydrase IX inhibitors as anticancer agents by toning the hydrophobic and hydrophilic rims of the active site to encounter the dual-tail approach. Eur J Med Chem 2022; 232:114190. [PMID: 35182815 DOI: 10.1016/j.ejmech.2022.114190] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/19/2022] [Accepted: 02/06/2022] [Indexed: 12/13/2022]
Abstract
The hydrophobic and the hydrophilic rims in the active site of human carbonic anhydrase IX (hCA IX) which as well contains a zinc ion as part of the catalytic core, were simultaneously matched to design and synthesize potent and selective inhibitors using a dual-tail approach. Seventeen new compounds, 5a-q, were designed to have the benzenesulfonamide moiety as a zinc binding group. In addition, N-substituted hydrazone and N-phenyl fragments were chosen as the hydrophilic and hydrophobic parts, respectively to achieve favorable interactions with the corresponding halves of the active site. All synthesized compounds successfully suppressed the CA IX, with IC50 values in nanomolar range from 13.3 to 259 nM. Compounds, 5h, 5c, 5m, 5e, and 5k were the top-five compounds efficiently inhibited the tumor-related CA IX isoform in the low nanomolar range (KI = 13.3, 22.6, 25.8, 26.9 and 27.2 nM, respectively). The target compounds 5a-q developed remarkable selectivity toward the tumor-associated isoforms (hCA IX and XII) over the off-target isoforms (hCA I and II). Furthermore, they were assessed for their anti-proliferative activity, according to US-NCI protocol, against a panel of fifty-nine cancer cell lines. Compounds 5d, 5k and 5o were passed the criteria for activity and scheduled automatically for evaluation at five concentrations with 10-fold dilutions. Compound 5k exhibited significant in vitro anticancer activity with GI50-MID; 8.68 μM compared to compounds 5d and 5o with GI50-MID; 25.76 μM and 34.97 μM respectively. The most selective compounds 5h and 5k were further screened for their in vitro cytotoxic activity against SK-MEL-5, HCC-2998 and RXF 393 cancer cell lines under hypoxic conditions. Furthermore, 5k was screened for cell cycle disturbance, apoptosis induction and intracellular reactive oxygen species (ROS) production in SK-MEL-5 cancer cells. Finally, molecular docking studies were performed to gain insights for the plausible binding interactions and affinities for selected compounds within hCA IX active site.
Collapse
Affiliation(s)
- Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Andrea Petreni
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Mervat H El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
31
|
Zhang Z, Yang H, Zhong Y, Wang Y, Wang J, Cheng M, Liu Y. Synthesis, Molecular Docking Analysis, and Biological Evaluations of Saccharide-Modified Sulfonamides as Carbonic Anhydrase IX Inhibitors. Int J Mol Sci 2021; 22:ijms222413610. [PMID: 34948406 PMCID: PMC8704611 DOI: 10.3390/ijms222413610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022] Open
Abstract
Based on the strategy of the “tail approach”, 15 novel saccharide-modified sulfonamides were designed and synthesised. The novel compounds were evaluated as inhibitors of three human carbonic anhydrase (CA) isoforms, namely cytoplasmic CA II, transmembrane CA IX, and XII. Most of these compounds showed good activity against CAs and high topological polar surface area (TPSA) values, which had a positive effect on the selective inhibition of transmembrane isoforms CA IX and XII. In the in vitro activity studies, compounds 16a, 16b, and 16e reduced the viability of HT-29 and MDA-MB-231 cells with a high expression of CA IX under hypoxia. The inhibitory activity of compound 16e on the human osteosarcoma cell line MG-63 with a high expression of CA IX and XII was better than that of AZM. Moreover, high concentrations of compounds 16a and 16b reversed the acidification of the tumour microenvironment. In addition, compound 16a had a certain inhibitory effect on the migration of MDA-MB-231 cells. All the above results indicate that the saccharide-modified sulfonamide has further research value for the development of CA IX inhibitors.
Collapse
|
32
|
Lenferink AEG, McDonald PC, Cantin C, Grothé S, Gosselin M, Baardsnes J, Banville M, Lachance P, Robert A, Cepero-Donates Y, Radinovic S, Salois P, Parat M, Oamari H, Dulude A, Patel M, Lafrance M, Acel A, Bousquet-Gagnon N, L'Abbé D, Pelletier A, Malenfant F, Jaramillo M, O'Connor-Mccourt M, Wu C, Durocher Y, Duchesne M, Gadoury C, Marcil A, Fortin Y, Paul-Roc B, Acchione M, Chafe SC, Nemirovsky O, Lau J, Bénard F, Dedhar S. Isolation and characterization of monoclonal antibodies against human carbonic anhydrase-IX. MAbs 2021; 13:1999194. [PMID: 34806527 PMCID: PMC8632296 DOI: 10.1080/19420862.2021.1999194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The architectural complexity and heterogeneity of the tumor microenvironment (TME) remains a substantial obstacle in the successful treatment of cancer. Hypoxia, caused by insufficient oxygen supply, and acidosis, resulting from the expulsion of acidic metabolites, are prominent features of the TME. To mitigate the consequences of the hostile TME, cancer cells metabolically rewire themselves and express a series of specific transporters and enzymes instrumental to this adaptation. One of these proteins is carbonic anhydrase (CA)IX, a zinc-containing extracellular membrane bound enzyme that has been shown to play a critical role in the maintenance of a neutral intracellular pH (pHi), allowing tumor cells to survive and thrive in these harsh conditions. Although CAIX has been considered a promising cancer target, only two antibody-based therapeutics have been clinically tested so far. To fill this gap, we generated a series of novel monoclonal antibodies (mAbs) that specifically recognize the extracellular domain (ECD) of human CAIX. Here we describe the biophysical and functional properties of a set of antibodies against the CAIX ECD domain and their applicability as: 1) suitable for development as an antibody-drug-conjugate, 2) an inhibitor of CAIX enzyme activity, or 3) an imaging/detection antibody. The results presented here demonstrate the potential of these specific hCAIX mAbs for further development as novel cancer therapeutic and/or diagnostic tools.
Collapse
Affiliation(s)
- Anne E G Lenferink
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Paul C McDonald
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Christiane Cantin
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Suzanne Grothé
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Mylene Gosselin
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Myriam Banville
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Paul Lachance
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Alma Robert
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Yuneivy Cepero-Donates
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Stevo Radinovic
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Patrick Salois
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Marie Parat
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Hafida Oamari
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Annie Dulude
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Mehul Patel
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Martin Lafrance
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Andrea Acel
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Nathalie Bousquet-Gagnon
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Denis L'Abbé
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Alex Pelletier
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Félix Malenfant
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Maria Jaramillo
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Maureen O'Connor-Mccourt
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Cunle Wu
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Mélanie Duchesne
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Christine Gadoury
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Anne Marcil
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Yves Fortin
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Beatrice Paul-Roc
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Maurizio Acchione
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Shawn C Chafe
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Oksana Nemirovsky
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Joseph Lau
- Department of Molecular Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Francois Bénard
- Department of Molecular Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| |
Collapse
|
33
|
Wang Y, Yin L, Cui Y, Wang L, Wu J, Wang J, Zhao H, Liu C, Cui Y, Zhang Y, Li X, Zhu Z, Yang L. Prognostic Significance of Membranous Carbonic Anhydrase IX Expression in Patients with Nonmetastatic Clear Cell Renal Cell Carcinoma of Different Tumor Stages. Cancer Biother Radiopharm 2021; 37:494-502. [PMID: 34714117 DOI: 10.1089/cbr.2020.3948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background: There are paradoxical results regarding whether carbonic anhydrase IX (CAIX) is a prognostic biomarker for patients with clear cell renal cell carcinoma (ccRCC). The objective of this study was to evaluate prognostic significance of CAIX in nonmetastatic ccRCC patients of different stages. Materials and Methods: This is a retrospective study on 1263 patients with nonmetastatic ccRCC from January 2005 to June 2018. Patients were stratified into eight subgroups (pT1a, pT1b, pT2a, pT2b, pT3a, pT3b, pT3c, and pT4) according to the 2016 TNM classification system. Immunohistochemical staining of membranous CAIX was quantified. Cancer-specific survival (CSS) rates in patients with high (>85%) and low (<85%) CAIX expressions were compared by Kaplan-Meier curves with log-rank test. Results: There were 220 tumors (17.42%) with low CAIX expression and 1043 tumors (82.58%) with high CAIX expression. The cumulative CSS rates were statistically significant between all patients with low and high CAIX expression (p-value <0.001). In pT2a, pT2b, and pT3a subgroups, the patients with low CAIX expression exhibited markedly decreased cumulative CSS rates compared to patients with high CAIX expression (p-value <0.05). Univariable and multivariable Cox regression analysis showed that CAIX expression was an independent predictor of prognosis in patients with pT2a, pT2b, and pT3a ccRCC (p-value <0.05), rather than in all nonmetastatic patients. Conclusion: CAIX expression is of independent prognostic value for ccRCC patients in pT2a, pT2b, and pT3a stages. CAIX expression combined with tumor stage would further improve risk stratification of nonmetastatic ccRCC patients and provide directions for therapies.
Collapse
Affiliation(s)
- Yongqiang Wang
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Likui Yin
- Clinical Laboratory, Dongying People's Hospital, Dongying, P.R. China
| | - Yupeng Cui
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Lin Wang
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Jitao Wu
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Jipeng Wang
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Hongwei Zhao
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Chu Liu
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Yuanshan Cui
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| | - Yongfu Zhang
- Department of Urology, Affiliated Yantai Hospital of Binzhou Medical University, Yantai, P.R. China
| | - Xiaofei Li
- Department of Urology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Zhe Zhu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, School of Medicine, La Jolla, California, USA
| | - Liqing Yang
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P.R. China
| |
Collapse
|
34
|
Janoniene A, Mazutis L, Matulis D, Petrikaite V. Inhibition of Carbonic Anhydrase IX Suppresses Breast Cancer Cell Motility at the Single-Cell Level. Int J Mol Sci 2021; 22:11571. [PMID: 34769000 PMCID: PMC8584155 DOI: 10.3390/ijms222111571] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 12/12/2022] Open
Abstract
Protein Carbonic Anhydrase IX (CA IX), which is expressed in various hypoxic solid tumors in order to maintain proper pH, is also related to cancer cell adhesion, invasion, and metastasis processes. Here, we investigated whether CA IX inhibition by a highly CA IX selective agent benzenesulfonamide VD11-4-2 triggers changes in individual cell motility. We seeded breast cancer cells on an extracellular matrix-coated glass-bottomed dish and in a microfluidic device with a gradient flow of epidermal growth factor (EGF), tracked individual cell movement, calculated their migration speeds, and/or followed movement direction. Our results showed that the inhibitor VD11-4-2 decreased the speed of CA IX positive breast cancer cells by 20-26% while not affecting non-cancerous cell migration. The inhibitor suppressed the cell migration velocity increment and hindered cells from reaching their maximum speed. VD11-4-2 also reduced CA IX, expressing cell movement towards the growth factor as a chemoattractant. Such a single cell-based migration assay enabled the comprehensive investigation of the cell motility and revealed that VD11-4-2 shows the ability to suppress breast cancer cell migration at a lower concentration than previously tested CA IX inhibitors.
Collapse
Affiliation(s)
| | | | | | - Vilma Petrikaite
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania; (A.J.); (L.M.); (D.M.)
| |
Collapse
|
35
|
Saleem S, Khan R, Haider G, Hasan S, Fatima F, Zehra S, Azhar A. Association of genetic polymorphism rs2071676 in carbonic anhydrase gene (CA9) with the risk of squamous cell carcinoma of lungs and esophagus. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00812-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Guan SS, Wu CT, Liao TZ, Lin KL, Peng CL, Shih YH, Weng MF, Chen CT, Yeh CH, Wang YC, Liu SH. A novel 111indium-labeled dual carbonic anhydrase 9-targeted probe as a potential SPECT imaging radiotracer for detection of hypoxic colorectal cancer cells. Eur J Pharm Biopharm 2021; 168:38-52. [PMID: 34450241 DOI: 10.1016/j.ejpb.2021.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/21/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
Tumor hypoxia is a common feature in colorectal cancer (CRC), and is associated with resistance to radiotherapy and chemotherapy. Thus, a specifically targeted probe for the detection of hypoxic CRC cells is urgently needed. Carbonic anhydrase 9 (CA9) is considered to be a specific marker for hypoxic CRC diagnosis. Here, a nuclear imaging Indium-111 (111In)-labeled dual CA9-targeted probe was synthesized and evaluated for CA9 detection in in vitro, in vivo, and in human samples. The CA9-targeted peptide (CA9tp) and CA9 inhibitor acetazolamide (AAZ) were combined to form a dual CA9-targeted probe (AAZ-CA9tp) using an automatic microwave peptide synthesizer, which then was conjugated with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) for radioisotope (111In) labeling (111In-DOTA-AAZ-CA9tp). The assays for cell binding, stability, and toxicity were conducted in hypoxic CRC HCT15 cells. The analyses for imaging and biodistribution were performed in an HCT15 xenograft mouse model. The binding and distribution of 111In-DOTA-AAZ-CA9tp were detected in human CRC samples using microautoradiography. AAZ-CA9tp possessed good CA9-targeting ability in hypoxic HCT15 cells. The dual CA9-targeted radiotracer showed high serum stability, high surface binding, and high affinity in vitro. After exposure of 111In-DOTA-AAZ-CA9tp to the HCT15-bearing xenograft mice, the levels of 111In-DOTA-AAZ-CA9tp were markedly and specifically increased in the hypoxic tumor tissues compared to control mice. 111In-DOTA-AAZ-CA9tp also targeted the areas of CA9 overexpression in human colorectal tumor tissue sections. The results of this study suggest that the novel 111In-DOTA-AAZ-CA9tp nuclear imaging agent may be a useful tool for the detection of hypoxic CRC cells in clinical practice.
Collapse
Affiliation(s)
- Siao-Syun Guan
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Cheng-Tien Wu
- Department of Nutrition, China Medical University, Taichung 40402, Taiwan; Master Program of Food and Drug Safety, China Medical University, Taichung 40402, Taiwan
| | - Tse-Zung Liao
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Kun-Liang Lin
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Cheng-Liang Peng
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Ying-Hsia Shih
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Mao-Feng Weng
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Chun-Tang Chen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Chung-Hsin Yeh
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Ying-Chieh Wang
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, No.1, Jen-Ai Road Section 1, Taipei 10051, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
37
|
Ullah A, Leong SW, Wang J, Wu Q, Ghauri MA, Sarwar A, Su Q, Zhang Y. Cephalomannine inhibits hypoxia-induced cellular function via the suppression of APEX1/HIF-1α interaction in lung cancer. Cell Death Dis 2021; 12:490. [PMID: 33990544 PMCID: PMC8121842 DOI: 10.1038/s41419-021-03771-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022]
Abstract
Lung cancer (LC) is one of the leading causes of cancer-related death. As one of the key features of tumor microenvironment, hypoxia conditions are associated with poor prognosis in LC patients. Upregulation of hypoxic-induced factor-1α (HIF-1α) leads to the activation of various factors that contribute to the increased drug resistance, proliferation, and migration of tumor cells. Apurinic/apyrimidinic endonuclease-1 (APEX1) is a multi-functional protein that regulates several transcription factors, including HIF-1α, that contribute to tumor growth, oxidative stress responses, and DNA damage. In this study, we explored the mechanisms underlying cell responses to hypoxia and modulation of APEX1, which regulate HIF-1α and downstream pathways. We found that hypoxia-induced APEX1/HIF-1α pathways regulate several key cellular functions, including reactive oxygen species (ROS) production, carbonic anhydrase 9 (CA9)-mediated intracellular pH, migration, and angiogenesis. Cephalomannine (CPM), a natural compound, exerted inhibitory effects in hypoxic LC cells via the inhibition of APEX1/HIF-1α interaction in vitro and in vivo. CPM can significantly inhibit cell viability, ROS production, intracellular pH, and migration in hypoxic LC cells as well as angiogenesis of HUVECs under hypoxia through the inhibition of APEX1/HIF-1α interaction. Taken together, CPM could be considered as a promising compound for LC treatment.
Collapse
Affiliation(s)
- Asmat Ullah
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Sze Wei Leong
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - Jingjing Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Qing Wu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Mohsin Ahmad Ghauri
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Ammar Sarwar
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Qi Su
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China.
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China.
| |
Collapse
|
38
|
Kugler M, Holub J, Brynda J, Pospíšilová K, Anwar SE, Bavol D, Havránek M, Král V, Fábry M, Grüner B, Řezáčová P. The structural basis for the selectivity of sulfonamido dicarbaboranes toward cancer-associated carbonic anhydrase IX. J Enzyme Inhib Med Chem 2021; 35:1800-1810. [PMID: 32962427 PMCID: PMC7534198 DOI: 10.1080/14756366.2020.1816996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human carbonic anhydrase IX (CA IX), a protein specifically expressed on the surface of solid tumour cells, represents a validated target both for anticancer therapy and diagnostics. We recently identified sulfonamide dicarbaboranes as promising inhibitors of CA IX with favourable activities both in vitro and in vivo. To explain their selectivity and potency, we performed detailed X-ray structural analysis of their interactions within the active sites of CA IX and CA II. Series of compounds bearing various aliphatic linkers between the dicarbaborane cluster and sulfonamide group were examined. Preferential binding towards the hydrophobic part of the active site cavity was observed. Selectivity towards CA IX lies in the shape complementarity of the dicarbaborane cluster with a specific CA IX hydrophobic patch containing V131 residue. The bulky side chain of F131 residue in CA II alters the shape of the catalytic cavity, disrupting favourable interactions of the spherical dicarbaborane cluster.
Collapse
Affiliation(s)
- Michael Kugler
- Deparment of Structural Biology, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.,Deparment of Structural Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Josef Holub
- Department of Syntheses, Institute of Inorganic Chemistry of the Czech Academy of Sciences, Řež, Czech Republic
| | - Jiří Brynda
- Deparment of Structural Biology, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.,Deparment of Structural Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Klára Pospíšilová
- Deparment of Structural Biology, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Suzan El Anwar
- Department of Syntheses, Institute of Inorganic Chemistry of the Czech Academy of Sciences, Řež, Czech Republic
| | - Dmytro Bavol
- Department of Syntheses, Institute of Inorganic Chemistry of the Czech Academy of Sciences, Řež, Czech Republic
| | | | - Vlastimil Král
- Deparment of Structural Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Milan Fábry
- Deparment of Structural Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Bohumír Grüner
- Department of Syntheses, Institute of Inorganic Chemistry of the Czech Academy of Sciences, Řež, Czech Republic
| | - Pavlína Řezáčová
- Deparment of Structural Biology, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.,Deparment of Structural Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
39
|
An ultra-high-affinity small organic ligand of fibroblast activation protein for tumor-targeting applications. Proc Natl Acad Sci U S A 2021; 118:2101852118. [PMID: 33850024 DOI: 10.1073/pnas.2101852118] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We describe the development of OncoFAP, an ultra-high-affinity ligand of fibroblast activation protein (FAP) for targeting applications with pan-tumoral potential. OncoFAP binds to human FAP with affinity in the subnanomolar concentration range and cross-reacts with the murine isoform of the protein. We generated various fluorescent and radiolabeled derivatives of OncoFAP in order to study biodistribution properties and tumor-targeting performance in preclinical models. Fluorescent derivatives selectively localized in FAP-positive tumors implanted in nude mice with a rapid and homogeneous penetration within the neoplastic tissue. Quantitative in vivo biodistribution studies with a lutetium-177-labeled derivative of OncoFAP revealed a preferential localization in tumors at doses of up to 1,000 nmol/kg. More than 30% of the injected dose had already accumulated in 1 g of tumor 10 min after intravenous injection and persisted for at least 3 h with excellent tumor-to-organ ratios. OncoFAP also served as a modular component for the generation of nonradioactive therapeutic products. A fluorescein conjugate mediated a potent and FAP-dependent tumor cell killing activity in combination with chimeric antigen receptor (CAR) T cells specific to fluorescein. Similarly, a conjugate of OncoFAP with the monomethyl auristatin E-based Vedotin payload was well tolerated and cured tumor-bearing mice in combination with a clinical-stage antibody-interleukin-2 fusion. Collectively, these data support the development of OncoFAP-based products for tumor-targeting applications in patients with cancer.
Collapse
|
40
|
Millul J, Krudewig C, Zana A, Dakhel Plaza S, Puca E, Villa A, Neri D, Cazzamalli S. Immunotherapy with Immunocytokines and PD-1 Blockade Enhances the Anticancer Activity of Small Molecule-Drug Conjugates Targeting Carbonic Anhydrase IX. Mol Cancer Ther 2021; 20:512-522. [PMID: 33443104 PMCID: PMC7617078 DOI: 10.1158/1535-7163.mct-20-0361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/12/2020] [Accepted: 12/16/2020] [Indexed: 11/16/2022]
Abstract
Small molecule-drug conjugates (SMDCs) represent an alternative to conventional antitumor chemotherapeutic agents, with the potential to improve the therapeutic window of cytotoxic payloads through active delivery at the site of the disease. In this article, we describe novel combination therapies consisting of anti-carbonic anhydrase IX SMDCs combined with different immunomodulatory products. The therapeutic effect of the SMDCs was potentiated by combination with PD-1 blockade and with tumor-homing antibody-cytokine fusions in mouse models of renal cell carcinoma and colorectal cancer. The combination with L19-IL12, a fusion protein specific to the alternatively spliced EDB domain of fibronectin containing the murine IL12 moiety, was also active against large established tumors. Analysis of the microscopic structures of healthy organs performed 3 months after tumor eradication confirmed absence of pathologic abnormalities in the healthy kidney, liver, lung, stomach, and intestine. Our findings may be of clinical significance as they provide motivation for the development of combinations based on SMDCs and immunotherapy for the treatment of renal cell carcinoma and hypoxic tumors.
Collapse
Affiliation(s)
| | - Christiane Krudewig
- Laboratory for Animal Model Pathology, Universität Zürich, Zurich, Switzerland
| | | | | | | | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
41
|
PEG Linker Length Strongly Affects Tumor Cell Killing by PEGylated Carbonic Anhydrase Inhibitors in Hypoxic Carcinomas Expressing Carbonic Anhydrase IX. Int J Mol Sci 2021; 22:ijms22031120. [PMID: 33498779 PMCID: PMC7866101 DOI: 10.3390/ijms22031120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
Hypoxic tumors overexpress membrane-bound isozymes of carbonic anhydrase (CA) CA IX and CA XII, which play key roles in tumor pH homeostasis under hypoxia. Selective inhibition of these CA isozymes has the potential to generate pH imbalances that can lead to tumor cell death. Since these isozymes are dimeric, we designed a series of bifunctional PEGylated CA inhibitors (CAIs) through the attachment of our preoptimized CAI warhead 1,3,4-thiadiazole-2-sulfonamide to polyethylene glycol (PEG) backbones with lengths ranging from 1 KDa to 20 KDa via a succinyl linker. A detailed structure−thermal properties and structure–biological activity relationship study was conducted via differential scanning calorimetry (DSC) and via viability testing in 2D and 3D (tumor spheroids) cancer cell models, either CA IX positive (HT-29 colon cancer, MDA-MB 231 breast cancer, and SKOV-3 ovarian cancer) or CA IX negative (NCI-H23 lung cancer). We identified PEGylated CAIs DTP1K 28, DTP2K 23, and DTP3.4K 29, bearing short and medium PEG backbones, as the most efficient conjugates under both normoxic and hypoxic conditions, and in the tumor spheroid models. PEGylated CAIs did not affect the cell viability of CA IX-negative NCI-H23 tumor spheroids, thus confirming a CA IX-mediated cell killing for these potential anticancer agents.
Collapse
|
42
|
Nemr MTM, AboulMagd AM, Hassan HM, Hamed AA, Hamed MIA, Elsaadi MT. Design, synthesis and mechanistic study of new benzenesulfonamide derivatives as anticancer and antimicrobial agents via carbonic anhydrase IX inhibition. RSC Adv 2021; 11:26241-26257. [PMID: 35479426 PMCID: PMC9037358 DOI: 10.1039/d1ra05277b] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 07/26/2021] [Indexed: 02/04/2023] Open
Abstract
Changes in gene expression cause uncontrolled cell proliferation and consequently tumor hypoxia. The tumor cells shift their metabolism to anaerobic glycolysis with a significant modification in pH. Therefore, an over expression of carbonic anhydrase IX (CA IX) genes was detected in many solid tumors. Accordingly, selective inhibition of CA IX can be a useful target for discovering novel antiproliferative agents. The present study described the synthesis of new aryl thiazolone–benzenesulfonamides 4a–j as well as their carbonic anhydrase IX inhibitory effect. All the designed derivatives were evaluated for their anti-proliferative activity against triple-negative breast cancer cell line (as MDA-MB-231) and another breast cancer cell line (MCF-7) in addition to normal breast cell line MCF-10A. Compounds 4b–c, 4e, 4g–h showed significant inhibitory effect against both cancer cell lines at concentration ranges from 1.52–6.31 μM, with a high selectivity against breast cancer cell lines ranges from 5.5 to 17.5 times. Moreover, three sulfonamides derivatives 4e, 4g and 4h showed excellent enzyme inhibition against CA IX with IC50 10.93–25.06 nM and against CA II with IC50 1.55–3.92 μM that revealed their remarkable selectivity for CA IX over CA II. Additionally, 4e was able to induce apoptosis in MDA-MB-231 with a significant increase in the annexin V-FITC percent by 22 fold as compared with control. Cellular uptake on MDA-MB-231 cell lines were carried out using HPLC method on the three active compounds (4e, 4g and 4h). On the other hand inhibition of one or more CAs present in bacteria was reported to interfere with bacterial growth. So, the new benzenesulfonamides were evaluated against their antibacterial and anti-biofilm activities. Analogues 4e, 4g and 4h exhibited significant inhibition at 50 μg mL−1 concentration with 80.69%, 69.74% and 68.30% against S. aureus compared to the positive control CIP which was 99.2%, while compounds 4g and 4h showed potential anti-biofilm inhibition 79.46% and 77.52% against K. pneumonia. Furthermore, the designed compounds were docked into CA IX (human) protein (PDB ID: 5FL6) and molecular modeling studies revealed favorable binding interactions for the active inhibitors. Finally, the predictive ADMET studies showed that, compounds 4e, 4g and 4h possessed promising pharmacokinetic properties. New benzenesulfonamide derivatives as anticancer and antimicrobial agents via CA IX inhibition.![]()
Collapse
Affiliation(s)
- Mohamed T. M. Nemr
- Pharmaceutical Organic Chemistry Department
- Faculty of Pharmacy
- Cairo University
- Cairo
- Egypt
| | - Asmaa M. AboulMagd
- Pharmaceutical Chemistry Department
- Faculty of Pharmacy
- Nahda University
- Beni-Suef 62513
- Egypt
| | - Hossam M. Hassan
- Pharmacognosy Department
- Faculty of Pharmacy
- Beni-Suef University
- Beni-Suef 62513
- Egypt
| | - Ahmed A. Hamed
- Microbial Chemistry Department
- National Research Center
- Giza
- Egypt
| | - Mohamed I. A. Hamed
- Organic and Medicinal Chemistry Department
- Faculty of Pharmacy
- Fayoum University
- Fayoum 63514
- Egypt
| | - Mohamed T. Elsaadi
- Pharmaceutical Chemistry Department
- Faculty of Pharmacy
- Beni-Suef University
- Beni-Suef 62513
- Egypt
| |
Collapse
|
43
|
Havránková E, Peña-Méndez EM, Csöllei J, Havel J. Prediction of biological activity of compounds containing a 1,3,5-triazinyl sulfonamide scaffold by artificial neural networks using simple molecular descriptors. Bioorg Chem 2020; 107:104565. [PMID: 33418318 DOI: 10.1016/j.bioorg.2020.104565] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022]
Abstract
Simple molecular descriptors of extensive series of 1,3,5-triazinyl sulfonamide derivatives, based on the structure of sulfonamides and their physicochemical properties, were designed and calculated. These descriptors were successfully applied as inputs for artificial neural network (ANN) modelling of the relationship between the structure and biological activity. The optimized ANN architecture was applied to the prediction of the inhibition activity of 1,3,5-triazinyl sulfonamides against human carbonic anhydrase (hCA) II, tumour-associated hCA IX, and their selectivity (hCA II/hCA IX).
Collapse
Affiliation(s)
- Eva Havránková
- Masaryk University, Faculty of Pharmacy, Department of Chemical Drugs, Palackého 1-3, CZ-612 42 Brno, Czech Republic
| | - E M Peña-Méndez
- Universidad de La Laguna (ULL), Facultad de Ciencias, Departamento de Química, Unidad Departamental de Química Analítica, 38201 La Laguna, Spain
| | - Jozef Csöllei
- Masaryk University, Faculty of Pharmacy, Department of Chemical Drugs, Palackého 1-3, CZ-612 42 Brno, Czech Republic
| | - Josef Havel
- Masaryk University, Faculty of Science, Department of Chemistry, University Campus, Kamenice 753/5, CZ-625 00 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Pekařská 53, 656 91 Brno, Czech Republic
| |
Collapse
|
44
|
Nekvinda J, Kugler M, Holub J, El Anwar S, Brynda J, Pospíšilová K, Růžičková Z, Řezáčová P, Grüner B. Direct Introduction of an Alkylsulfonamido Group on C‐sites of Isomeric Dicarba‐
closo
‐dodecaboranes: The Influence of Stereochemistry on Inhibitory Activity against the Cancer‐Associated Carbonic Anhydrase IX Isoenzyme. Chemistry 2020; 26:16541-16553. [DOI: 10.1002/chem.202002809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/28/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Jan Nekvinda
- Department of Synthesis Institute of Inorganic Chemistry of, the Czech Academy of Sciences 25068 Řež Czech Republic
| | - Michael Kugler
- Institute of Molecular Genetics Czech Academy of Sciences Vídeňská 1083 14220 Prague 4 Czech Republic
- Institute of Organic Chemistry and Biochemistry Czech Academy of, Sciences Flemingovo nám. 2 16610 Prague Czech Republic
| | - Josef Holub
- Department of Synthesis Institute of Inorganic Chemistry of, the Czech Academy of Sciences 25068 Řež Czech Republic
| | - Suzan El Anwar
- Department of Synthesis Institute of Inorganic Chemistry of, the Czech Academy of Sciences 25068 Řež Czech Republic
| | - Jiří Brynda
- Institute of Molecular Genetics Czech Academy of Sciences Vídeňská 1083 14220 Prague 4 Czech Republic
- Institute of Organic Chemistry and Biochemistry Czech Academy of, Sciences Flemingovo nám. 2 16610 Prague Czech Republic
| | - Klára Pospíšilová
- Institute of Organic Chemistry and Biochemistry Czech Academy of, Sciences Flemingovo nám. 2 16610 Prague Czech Republic
| | - Zdeňka Růžičková
- Department of General and Inorganic Chemistry Faculty of Chemical Technology University of Pardubice Studentská 573 53210 Pardubice Czech Republic
| | - Pavlína Řezáčová
- Institute of Molecular Genetics Czech Academy of Sciences Vídeňská 1083 14220 Prague 4 Czech Republic
- Institute of Organic Chemistry and Biochemistry Czech Academy of, Sciences Flemingovo nám. 2 16610 Prague Czech Republic
| | - Bohumír Grüner
- Department of Synthesis Institute of Inorganic Chemistry of, the Czech Academy of Sciences 25068 Řež Czech Republic
| |
Collapse
|
45
|
Andring JT, Fouch M, Akocak S, Angeli A, Supuran CT, Ilies MA, McKenna R. Structural Basis of Nanomolar Inhibition of Tumor-Associated Carbonic Anhydrase IX: X-Ray Crystallographic and Inhibition Study of Lipophilic Inhibitors with Acetazolamide Backbone. J Med Chem 2020; 63:13064-13075. [PMID: 33085484 DOI: 10.1021/acs.jmedchem.0c01390] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This study provides a structure-activity relationship study of a series of lipophilic carbonic anhydrase (CA) inhibitors with an acetazolamide backbone. The inhibitors were tested against the tumor-expressed CA isozyme IX (CA IX), and the cytosolic CA I, CA II, and membrane-bound CA IV. The study identified several low nanomolar potent inhibitors against CA IX, with lipophilicities spanning two log units. Very potent pan-inhibitors with nanomolar potency against CA IX and sub-nanomolar potency against CA II and CA IV, and with potency against CA I one order of magnitude better than the parent acetazolamide 1 were also identified in this study, together with compounds that displayed selectivity against membrane-bound CA IV. A comprehensive X-ray crystallographic study (12 crystal structures), involving both CA II and a soluble CA IX mimetic (CA IX-mimic), revealed the structural basis of this particular inhibition profile and laid the foundation for further developments toward more potent and selective inhibitors for the tumor-expressed CA IX.
Collapse
Affiliation(s)
- Jacob T Andring
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Mallorie Fouch
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Suleyman Akocak
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Andrea Angeli
- NEUROFARBA Department, Pharmaceutical Sciences Section, Universita degli Studi di Firenze, Polo Scientifico, Via Ugo Schiff no. 6, 50019 Sesto Fiorentino, Florence, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Pharmaceutical Sciences Section, Universita degli Studi di Firenze, Polo Scientifico, Via Ugo Schiff no. 6, 50019 Sesto Fiorentino, Florence, Italy
| | - Marc A Ilies
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
46
|
The Anticancer Activity for the Bumetanide-Based Analogs via Targeting the Tumor-Associated Membrane-Bound Human Carbonic Anhydrase-IX Enzyme. Pharmaceuticals (Basel) 2020; 13:ph13090252. [PMID: 32961906 PMCID: PMC7558282 DOI: 10.3390/ph13090252] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/05/2020] [Accepted: 09/16/2020] [Indexed: 12/20/2022] Open
Abstract
The membrane-bound human carbonic anhydrase (hCA) IX is widely recognized as a marker of tumor hypoxia and a prognostic factor within several human cancers. Being undetected in most normal tissues, hCA-IX implies the pharmacotherapeutic advent of reduced off-target adverse effects. We assessed the potential anticancer activity of bumetanide-based analogues to inhibit the hCA-IX enzymatic activity and cell proliferation of two solid cancer cell lines, namely kidney carcinoma (A-498) and bladder squamous cell carcinoma (SCaBER). Bumetanide analogues efficiently inhibit the target hCA-IX in low nanomolar activity (IC50 = 4.4–23.7 nM) and have an excellent selectivity profile (SI = 14.5–804) relative to the ubiquitous hCA-II isoform. Additionally, molecular docking studies provided insights into the compounds’ structure–activity relationship and preferential binding of small-sized as well as selective bulky ligands towards the hCA-IX pocket. In particular, 2,4-dihydro-1,2,4-triazole-3-thione derivative 9c displayed pronounced hCA-IX inhibitory activity and impressive antiproliferative activity on oncogenic A-498 kidney carcinoma cells and is being considered as a promising anticancer candidate. Future studies will aim to optimize this compound to fine-tune its anticancer activity as well as explore its potential through in-vivo preclinical studies.
Collapse
|
47
|
Xu J, Zhu S, Xu L, Liu X, Ding W, Wang Q, Chen Y, Deng H. CA9 Silencing Promotes Mitochondrial Biogenesis, Increases Putrescine Toxicity and Decreases Cell Motility to Suppress ccRCC Progression. Int J Mol Sci 2020; 21:E5939. [PMID: 32824856 PMCID: PMC7460829 DOI: 10.3390/ijms21165939] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 02/04/2023] Open
Abstract
Carbonic anhydrase IX (CA9), a pH-regulating transmembrane protein, is highly expressed in solid tumors, and particularly in clear cell renal cell carcinoma (ccRCC). The catalytic mechanisms of CA9 are well defined, but its roles in mediating cell migration/invasion and survival in ccRCC remain to be determined. Here, we confirmed that the mRNA expression of CA9 in ccRCC was significantly higher than that in para-carcinoma tissues from analysis of the datasets in The Cancer Genome Atlas. CA9 knockdown upregulated oxidative phosphorylation-associated proteins and increased mitochondrial biogenesis, resulting in the reversal of the Warburg phenotype and the inhibition of cell growth. Our study revealed that CA9 knockdown upregulated mitochondrial arginase 2 (ARG2), leading to the accumulation of putrescine, which suppressed ccRCC proliferation. Surfaceomics analysis revealed that CA9 knockdown downregulated proteins associated with extracellular matrix (ECM)-receptor interaction and cell adhesion, resulting in decreased cell migration. CA9 silencing also downregulated amino acid transporters, leading to reduced cellular amino acids. Collectively, our data show that CA9 knockdown suppresses proliferation via metabolic reprogramming and reduced cell migration, reaffirming that CA9 is a potential therapeutic target for ccRCC treatment.
Collapse
Affiliation(s)
- Jiatong Xu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Songbiao Zhu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Lina Xu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Xiaohui Liu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Wenxi Ding
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Qingtao Wang
- Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China;
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| |
Collapse
|
48
|
Mishra CB, Tiwari M, Supuran CT. Progress in the development of human carbonic anhydrase inhibitors and their pharmacological applications: Where are we today? Med Res Rev 2020; 40:2485-2565. [PMID: 32691504 DOI: 10.1002/med.21713] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/14/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022]
Abstract
Carbonic anhydrases (CAs, EC 4.2.1.1) are widely distributed metalloenzymes in both prokaryotes and eukaryotes. They efficiently catalyze the reversible hydration of carbon dioxide to bicarbonate and H+ ions and play a crucial role in regulating many physiological processes. CAs are well-studied drug target for various disorders such as glaucoma, epilepsy, sleep apnea, and high altitude sickness. In the past decades, a large category of diverse families of CA inhibitors (CAIs) have been developed and many of them showed effective inhibition toward specific isoforms, and effectiveness in pathological conditions in preclinical and clinical settings. The discovery of isoform-selective CAIs in the last decade led to diminished side effects associated with off-target isoforms inhibition. The many new classes of such compounds will be discussed in the review, together with strategies for their development. Pharmacological advances of the newly emerged CAIs in diseases not usually associated with CA inhibition (neuropathic pain, arthritis, cerebral ischemia, and cancer) will also be discussed.
Collapse
Affiliation(s)
- Chandra B Mishra
- Department of Bioorganic Chemistry, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India.,Department of Pharmaceutical Chemistry, College of Pharmacy, Sookmyung Women's University, Seoul, South Korea
| | - Manisha Tiwari
- Department of Bioorganic Chemistry, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Claudiu T Supuran
- Dipartimento Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
49
|
Janoniene A, Petrikaite V. In Search of Advanced Tumor Diagnostics and Treatment: Achievements and Perspectives of Carbonic Anhydrase IX Targeted Delivery. Mol Pharm 2020; 17:1800-1815. [PMID: 32374612 DOI: 10.1021/acs.molpharmaceut.0c00180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The research of how cells sense and adapt the oxygen deficiency has been recognized as worth winning a Nobel Prize in 2019. Understanding hypoxia-driven molecular machinery paved a path for novel strategies in fighting hypoxia-related diseases including cancer. The oxygen depletion inside the tumor provokes HIF-1 dependent gene and protein expression which helps the tumor to survive. For this reason, tumor related molecules are in the spotlight for scientists developing anticancer agents. One such target is carbonic anhydrase IX (CA IX)-a protein located on the outer cell membrane of most hypoxic tumor cells. This offers the opportunity to exploit it as a target for delivery of cytotoxic drugs, dyes, or radioisotopes to cancer cells. Therefore, researchers investigate CA IX specific small molecules and antibodies as tumor-targeting moieties in nanosystems and conjugates which are expected to overcome the limitations of some existing diagnostic and treatment strategies. This review covers the vast majority of CA IX-targeted systems (nanoparticle and conjugate based) for both therapeutic and imaging purposes published up to now. Furthermore, it shows their stage of development and gives an assessment of their clinical translation possibilities.
Collapse
Affiliation(s)
- Agne Janoniene
- Vilnius University Life Science Center, Institute of Biotechnology, LT-10257 Vilnius, Lithuania
| | - Vilma Petrikaite
- Vilnius University Life Science Center, Institute of Biotechnology, LT-10257 Vilnius, Lithuania.,Lithuanian University of Health Sciences, Institute of Cardiology, LT-50162 Kaunas, Lithuania
| |
Collapse
|
50
|
Tumor microenvironment, immune response and post-radiotherapy tumor clearance. Clin Transl Oncol 2020; 22:2196-2205. [PMID: 32445035 DOI: 10.1007/s12094-020-02378-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
Radiotherapy is the treatment of choice for many cancer patients. Residual tumor leads to local recurrence after a period of an equilibrium created between proliferating, quiescent and dying cancer cells. The tumor microenvironment is a main obstacle for the efficacy of radiotherapy, as impaired blood flow leads to hypoxia, acidity and reduced accessibility of radiosensitizers. Eradication of remnant disease is an intractable clinical quest. After more than a century of research, anti-tumor immunity has gained a dominant position in oncology research and therapy. Immune cells play a significant role in the eradication of tumors during and after the completion of radiotherapy. The tumor equilibrium reached in the irradiated tumor may shift towards cancer cell eradication if the immune response is appropriately modulated. In the modern immunotherapy era, clinical trials are urged to standardize immunotherapy schemes that could be safely applied to improve clearance of the post-radiotherapy remnant disease.
Collapse
|