1
|
Fernandes DA. Multifunctional gold nanoparticles for cancer theranostics. 3 Biotech 2024; 14:267. [PMID: 39416669 PMCID: PMC11473483 DOI: 10.1007/s13205-024-04086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
The diagnosis and treatment of cancer can often be challenging requiring more attractive options. Some types of cancers are more aggressive than others and symptoms for many cancers are subtle, especially in the early stages. Nanotechnology provides high sensitivity, specificity and multimodal capability for cancer detection, treatment and monitoring. In particular, metal nanoparticles (NPs) such as gold nanoparticles (AuNPs) are attractive nanosystems for researchers interested in bioimaging and therapy. The size, shape and surface of AuNPs can be modified for improving targeting and accumulation in cancer cells, for example through introduction of ligands and surface charge. The interactions of AuNPs with electromagnetic radiation (e.g., visible-near-infrared, X-rays) can be used for photothermal therapy and radiation therapy, through heat generated from light absorption and emission of Auger electrons, respectively. The subsequent expansion and high X-ray attenuation from AuNPs can be used for enhancing contrast for tumor detection (e.g., using photoacoustic, computed tomography imaging). Multi-functionality can be further extended through covalent/non-covalent functionalization, for loading additional imaging/therapeutic molecules for combination therapy and multimodal imaging. In order to cover the important aspects for designing and using AuNPs for cancer theranostics, this review focuses on the synthesis, functionalization and characterization methods that are important for AuNPs, and presents their unique properties and different applications in cancer theranostics.
Collapse
|
2
|
Xiao-Qun Z, Xian-Li M, Ariffin NS. The potential of carbonic anhydrase enzymes as a novel target for anti-cancer treatment. Eur J Pharmacol 2024; 976:176677. [PMID: 38825301 DOI: 10.1016/j.ejphar.2024.176677] [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: 12/29/2023] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
Carbonic anhydrase (CA) is a zinc-dependent metal enzyme that maintains the pH and carbon dioxide (CO2) homeostasis in cells by catalyzing the reversible hydration and dehydration of CO2 and bicarbonate (HCO3-). In mammals, there are 16 isozymes of CA existed, namely CAI to CAXIV, but only 15 isozymes are found in humans except CAXV. Human CAs have highly conserved catalytic domains, all of which are distributed in different tissues and play important physiological roles. Changes in their functions may disrupt the typical distribution of CAs throughout human body and therefore CAs can be used as diagnostic biomarkers for many diseases. Furthermore, the expression of CAs is correlated to the progression of numerous tumors, therapeutic sensitivity and patient prognosis. In this review, we discuss thoroughly the structure of CAs, their functional activities in human physiology, dysregulations and diseases related to CAs, and different types of CA inhibitors that can reverse their dysregulation.
Collapse
Affiliation(s)
- Zhou Xiao-Qun
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, 42300, Bandar Puncak Alam, Selangor, Malaysia; Guilin Medical University, GuiLin, China
| | | | - Nur Syamimi Ariffin
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, 42300, Bandar Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
3
|
Yapar G, Lolak N, Bonardi A, Akocak S, Supuran CT. Exploring the potency of diazo-coumarin containing hybrid molecules: Selective inhibition of tumor-associated carbonic anhydrase isoforms IX and XII. ChemMedChem 2024; 19:e202300626. [PMID: 38193633 DOI: 10.1002/cmdc.202300626] [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: 11/11/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024]
Abstract
This study introduces a series of ten hybrid molecules DK(1-10), which combine diazo and coumarin moieties along with diverse aromatic substitutions. The primary objective was to evaluate the inhibitory capabilities of these compounds against four prominent isoforms: the cytosolic hCA I and II, as well as the tumor-associated membrane-bound hCA IX and XII. Impressively, the majority of the tested compounds exhibited significant inhibition activity against the tumor-associated isoforms hCA IX and XII, with KI values ranging from 29.2 to 293.3 nM. Notably, compound DK-8 displayed particularly robust inhibitory activity against the tumor-associated membrane-bound isoforms, hCA IX and XII, yielding KI values of 32.5 and 29.2 nM, respectively. Additionally, another derivative, DK-9, containing a primary sulfonamide, exhibited notable inhibition against hCA XII with a KI value of 36.4 nM. This investigation aimed to explore the structure-activity relationships within these compounds, shedding light on how various substitutions and structural components influence their inhibitory potential. As a result, these compounds present promising candidates for further exploration in medicinal and pharmacological research. Their ability to selectively inhibit specific isoforms, particularly those associated with hypoxic tumors, suggests their potential as foundational compounds for the development of novel therapeutic agents.
Collapse
Affiliation(s)
- Gönül Yapar
- Department of Chemistry, Faculty of Arts and Sciences, Istanbul Technical University, Istanbul, 34469, Türkiye
| | - Nebih Lolak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, 02040, Adıyaman, Türkiye
| | - Alessandro Bonardi
- Università degli Studi di Firenze, NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019, Sesto Fiorentino (Florence), Italy
| | - Suleyman Akocak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, 02040, Adıyaman, Türkiye
| | - Claudiu T Supuran
- Università degli Studi di Firenze, NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019, Sesto Fiorentino (Florence), Italy
| |
Collapse
|
4
|
Lolak N, Akocak S, Petreni A, Budak Y, Bozgeyik E, Gurdere MB, Ceylan M, Supuran CT. 1,3-Diaryl Triazenes Incorporating Disulfonamides Show Both Antiproliferative Activity and Effective Inhibition of Tumor-associated Carbonic Anhydrases IX and XII. Anticancer Agents Med Chem 2024; 24:755-763. [PMID: 38362678 DOI: 10.2174/0118715206285326240207045249] [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: 11/09/2023] [Revised: 12/31/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
AIM The aim of this study was to synthesize a library of novel di-sulfa drugs containing 1,3- diaryltriazene derivatives TS (1-13) by conjugation of diazonium salts of primary sulfonamides with sulfa drugs to investigate the cytotoxic effect of these new compounds in different cancer types and to determine their inhibitory activity against tumor-associated carbonic anhydrases IX and XII. MATERIALS AND METHODS A carbonic anhydrase inhibitory activity of the obtained compounds was evaluated against four selected human carbonic anhydrase isoforms (hCA I, hCA II, hCA IX and hCA XII) by a stoppedflow CO2 hydrase assay. In addition, in vitro, cytotoxicity studies were applied by using A549 (lung cancer), BEAS-2B (normal lung), MCF-7 (breast cancer), MDA-MB-231 (breast cancer), CRL-4010 (normal breast epithelium), HT-29 (colon cancer), and HCT -116 (colon cancer) cell lines. RESULTS As a result of the inhibition data, the 4-aminobenzenesulfonamide derivatives were more active than their 3-aminobenzenesulfonamide counterparts. More specifically, compounds TS-1 and TS-2, both of which have primary sulfonamides on both sides of the triazene linker, showed the best inhibitory activity against hCA IX with Ki values of 19.5 and 13.7 nM and also against hCA XII with Ki values of 6.6 and 8.3 nM, respectively. In addition, in vitro cytotoxic activity on the human breast cancer cell line MCF-7 showed that some derivatives of di-sulfa triazenes, such as TS-5 and TS-13, were more active than SLC-0111. CONCLUSION With the aim of developing more potent and isoform-selective CA inhibitors, these novel hybrid molecules containing sulfa drugs, triazene linkers, and the classical primary sulfonamide chemotype may be considered an interesting example of effective enzyme inhibitors and important anticancer agents.
Collapse
Affiliation(s)
- Nebih Lolak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, 02040 Adıyaman, Türkiye
| | - Suleyman Akocak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, 02040 Adıyaman, Türkiye
| | - Andrea Petreni
- Università Degli Studi di Firenze, NEUROFARBA Department Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Yakup Budak
- Department of Chemistry, Faculty of Arts and Sciences, Gaziosmanpaşa University, 60250 Tokat, Türkiye
| | - Esra Bozgeyik
- Department of Medical Services and Techniques, Vocational School of Health Services, Adıyaman University, 02040 Adıyaman, Türkiye
| | - Meliha Burcu Gurdere
- Department of Chemistry, Faculty of Arts and Sciences, Gaziosmanpaşa University, 60250 Tokat, Türkiye
| | - Mustafa Ceylan
- Department of Chemistry, Faculty of Arts and Sciences, Gaziosmanpaşa University, 60250 Tokat, Türkiye
| | - Claudiu Trandafir Supuran
- Università Degli Studi di Firenze, NEUROFARBA Department Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| |
Collapse
|
5
|
Tekeli T, Akocak S, Petreni A, Lolak N, Çete S, Supuran CT. Potent carbonic anhydrase I, II, IX and XII inhibition activity of novel primary benzenesulfonamides incorporating bis-ureido moieties. J Enzyme Inhib Med Chem 2023; 38:2185762. [PMID: 36880350 PMCID: PMC9987750 DOI: 10.1080/14756366.2023.2185762] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
A novel series of twelve aromatic bis-ureido-substituted benzenesulfonamides was synthesised by conjugation of aromatic aminobenzenesulfonamides with aromatic bis-isocyanates. The obtained bis-ureido-substituted derivatives were tested against four selected human carbonic anhydrase isoforms (hCA I, hCA II, hCA IX and hCA XII). Most of the new compounds showed an effective inhibitory profile against isoforms hCA IX and hCA XII, also having some selectivity with respect to hCA I and hCA II. The inhibition constants of these compounds against isoforms hCA IX and XII were in the range of 6.73-835 and 5.02-429 nM, respectively. Since hCA IX and hCA XII are important drug targets for anti-cancer/anti-metastatic drugs, these effective inhibitors reported here may be considered of interest for cancer related studies in which these enzymes are involved.
Collapse
Affiliation(s)
- Tuba Tekeli
- Vocational School of Technical Science, Department of Chemistry and Chemical Processing Technologies, Adıyaman University, Adıyaman, Türkiye.,Department of Chemistry, Faculty of Science, Gazi University, Ankara, Türkiye
| | - Suleyman Akocak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman, Türkiye
| | - Andrea Petreni
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, Sesto Fiorentino (Florence), Italy
| | - Nebih Lolak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman, Türkiye
| | - Servet Çete
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Türkiye
| | - Claudiu T Supuran
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, Sesto Fiorentino (Florence), Italy
| |
Collapse
|
6
|
Akocak S, Lolak N, Giovannuzzi S, Supuran CT. Potent and selective carbonic anhydrase inhibition activities of pyrazolones bearing benzenesulfonamides. Bioorg Med Chem Lett 2023; 95:129479. [PMID: 37704010 DOI: 10.1016/j.bmcl.2023.129479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
This research introduces a series of fourteen 4-aryl-hydrazonopyrazolone sulfonamide derivatives, denoted as 3(a-g) and 4(a-g), which encompass various aromatic substitutions. The aim was to assess the inhibitory potential of these compounds against four significant isoforms, including the cytosolic isoforms hCA I and II, as well as the tumor-associated membrane-bound isoforms hCA IX and XII. Most of the tested compounds exhibited substantial inhibition against the tumor-associated isoform hCA IX, with Ki values spanning from 1.1 to 158.2 nM. Notably, compounds 3e and 3g showed particularly strong inhibitory activity against the tumor-associated membrane-bound isoforms, hCA IX and XII, while maintaining a high selectivity ratio over cytosolic off-target isoforms hCA I and II. This selectivity is vital due to the potential of hCA IX and hCA XII as drug targets for hypoxic tumors. In an effort to create novel analogs that exhibit enhanced carbonic anhydrase inhibitory activity and specificity, we investigated the structure-activity relationships of these compounds and provided a concise interpretation of our findings. Consequently, these compounds merit consideration for subsequent medicinal and pharmacological research, holding potential for developing novel therapeutic agents targeting specific isoforms in hypoxic tumors.
Collapse
Affiliation(s)
- Suleyman Akocak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, 02040 Adıyaman, Turkey.
| | - Nebih Lolak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, 02040 Adıyaman, Turkey
| | - Simone Giovannuzzi
- Università degli Studi di Firenze, Dipartimento Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Via U. Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Claudiu T Supuran
- Università degli Studi di Firenze, Dipartimento Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Via U. Schiff 6, 50019 Sesto Fiorentino (Florence), Italy.
| |
Collapse
|
7
|
Tan KF, In LLA, Vijayaraj Kumar P. Surface Functionalization of Gold Nanoparticles for Targeting the Tumor Microenvironment to Improve Antitumor Efficiency. ACS APPLIED BIO MATERIALS 2023; 6:2944-2981. [PMID: 37435615 DOI: 10.1021/acsabm.3c00202] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Gold nanoparticles (AuNPs) have undergone significant research for their use in the treatment of cancer. Numerous researchers have established their potent antitumor properties, which have greatly impacted the treatment of cancer. AuNPs have been used in four primary anticancer treatment modalities, namely radiation, photothermal therapy, photodynamic therapy, and chemotherapy. However, the ability of AuNPs to destroy cancer is lacking and can even harm healthy cells without the right direction to transport them to the tumor microenvironment. Consequently, a suitable targeting technique is needed. Based on the distinct features of the human tumor microenvironment, this review discusses four different targeting strategies that target the four key features of the tumor microenvironment, including abnormal vasculature, overexpression of specific receptors, an acidic microenvironment, and a hypoxic microenvironment, to direct surface-functionalized AuNPs to the tumor microenvironment and increase antitumor efficacies. In addition, some current completed or ongoing clinical trials of AuNPs will also be discussed below to further reinforce the concept of using AuNPs in anticancer therapy.
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
8
|
Li Z, Guo L, Lin L, Wang T, Jiang Y, Song J, Feng J, Huang J, Li H, Bai Z, Liu W, Zhang J. Porous SiO 2-Based Reactor with Self-Supply of O 2 and H 2O 2 for Synergistic Photo-Thermal/Photodynamic Therapy. Int J Nanomedicine 2023; 18:3623-3639. [PMID: 37427365 PMCID: PMC10327690 DOI: 10.2147/ijn.s387505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
Purpose Although the combined photo-thermal (PTT) and photodynamic therapy (PDT) of tumors have demonstrated promise as effective cancer therapy, the hypoxic and insufficient H2O2 supply of tumors seriously limits the efficacy of PDT, and the acidic environment reduces the catalytic activity of nanomaterial in the tumor microenvironment. To develop a platform for efficiently addressing these challenges, we constructed a nanomaterial of Aptamer@dox/GOD-MnO2-SiO2@HGNs-Fc@Ce6 (AMS) for combination tumor therapy. The treatment effects of AMS were evaluated both in vitro and in vivo. Methods In this work, Ce6 and hemin were loaded on graphene (GO) through π-π conjugation, and Fc was connected to GO via amide bond. The HGNs-Fc@Ce6 was loaded into SiO2, and coated with dopamine. Then, MnO2 was modified on the SiO2. Finally, AS1411-aptamer@dox and GOD were fixed to gain AMS. We characterized the morphology, size, and zeta potential of AMS. The oxygen and reactive oxygen species (ROS) production properties of AMS were analyzed. The cytotoxicity of AMS was detected by MTT and calcein-AM/PI assays. The apoptosis of AMS to a tumor cell was estimated with a JC-1 probe, and the ROS level was detected with a 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA) probe. The anticancer efficacy in vivo was analyzed by the changes in the tumor size in different treatment groups. Results AMS was targeted to the tumor cell and released doxorubicin. It decomposed glucose to produce H2O2 in the GOD-mediated reaction. The generated sufficient H2O2 was catalyzed by MnO2 and HGNs-Fc@Ce6 to produce O2 and free radicals (•OH), respectively. The increased oxygen content improved the hypoxic environment of the tumor and effectively reduced the resistance to PDT. The generated •OH enhanced the ROS treatment. Moreover, AMS depicted a good photo-thermal effect. Conclusion The results revealed that AMS had an excellent enhanced therapy effect by combining synergistic PTT and PDT.
Collapse
Affiliation(s)
- Zhengzhao Li
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Lianshan Guo
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Liqiao Lin
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Tongting Wang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Yanqiu Jiang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jin Song
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jihua Feng
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jianfeng Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Haoyu Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Zhihao Bai
- College of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, China
| | - Wenqi Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jianfeng Zhang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| |
Collapse
|
9
|
Sufian MA, Zamanova S, Shabana AM, Kemp B, Mondal UK, Supuran CT, Ilies MA. Expression Dynamics of CA IX Epitope in Cancer Cells under Intermittent Hypoxia Correlates with Extracellular pH Drop and Cell Killing by Ureido-Sulfonamide CA IX Inhibitors. Int J Mol Sci 2023; 24:4595. [PMID: 36902027 PMCID: PMC10002582 DOI: 10.3390/ijms24054595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/25/2023] [Accepted: 02/25/2023] [Indexed: 03/02/2023] Open
Abstract
Carbonic anhydrase IX (CA IX) is a membrane-bound CA isozyme over-expressed in many hypoxic tumor cells, where it ensures pH homeostasis and has been implicated in tumor survival, metastasis and resistance to chemotherapy and radiotherapy. Given the functional importance of CA IX in tumor biochemistry, we investigated the expression dynamics of CA IX in normoxia, hypoxia and intermittent hypoxia, which are typical conditions experienced by tumor cells in aggressive carcinomas. We correlated the CA IX epitope expression dynamics with extracellular pH acidification and with viability of CA IX-expressing cancer cells upon treatment with CA IX inhibitors (CAIs) in colon HT-29, breast MDA-MB-231 and ovarian SKOV-3 tumor cell models. We observed that the CA IX epitope expressed under hypoxia by these cancer cells is retained in a significant amount upon reoxygenation, probably to preserve their proliferation ability. The extracellular pH drop correlated well with the level of CA IX expression, with the intermittent hypoxic cells showing a similar pH drop to fully hypoxic ones. All cancer cells showed higher sensitivity to CA IX inhibitors (CAIs) under hypoxia as compared to normoxia. The tumor cell sensitivity to CAIs under hypoxia and intermittent hypoxia were similar and higher than in normoxia and appeared to be correlated with the lipophilicity of the CAI.
Collapse
Affiliation(s)
- Md. Abu Sufian
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Sabina Zamanova
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Ahmed M. Shabana
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Brianna Kemp
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Utpal K. Mondal
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - 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, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
10
|
Prospects for hypoxia-based drug delivery platforms for the elimination of advanced metastatic tumors: From 3D modeling to clinical concepts. J Control Release 2023; 353:1002-1022. [PMID: 36516901 DOI: 10.1016/j.jconrel.2022.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
Hypoxia is a unique characteristic of the solid tumor microenvironment. Hypoxia contributes to multi-drug resistance, metastasis and cancer relapse through numerous molecular pathways, but at the same time provides an opportunity for the development of novel drugs or modalities specifically targeting hypoxic tumor regions. Given the high significance of tumor hypoxia in therapeutic results, we here discuss a variety of hypoxia-adopted strategies, and their potential and utility in the treatment of deep-seated hypoxic tumor cells. We discuss the merits and demerits of these approaches, as well as their combination with other approaches such as photodynamic therapy. We also survey the currently available 3D hypoxia modeling systems, in particular organoid-based microfluidics. Finally, we discuss the potential and the current status of preclinical tumor hypoxia approaches in clinical trials for advanced cancer. We believe that multi-modal imaging and therapeutic hypoxia adopted drug delivery platforms could provide better efficacy and safety profiles, and more importantly personalized therapy. Determining the hypoxia status of tumors could offer a second chance for the clinical translation of hypoxia-based agents, such as hypoxia activated prodrugs (HAPs) from bench to bedside.
Collapse
|
11
|
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
|
12
|
Nguyen PL, Elkamhawy A, Choi YH, Lee CH, Lee K, Cho J. Suppression of Tumor Growth and Cell Migration by Indole-Based Benzenesulfonamides and Their Synergistic Effects in Combination with Doxorubicin. Int J Mol Sci 2022; 23:ijms23179903. [PMID: 36077298 PMCID: PMC9456432 DOI: 10.3390/ijms23179903] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
Pharmacological inhibition of the enzyme activity targeting carbonic anhydrases (CAs) demonstrated antiglaucoma and anticancer effects through pH control. Recently, we reported a series of indole-based benzenesulfonamides as potent CA inhibitors. The present study aimed to evaluate the antitumor effects of these compounds against various cancer cell lines, including breast cancer (MDA-MB-231, MCF-7, and SK-BR-3), lung cancer (A549), and pancreatic cancer (Panc1) cells. Overall, more potent cytotoxicity was observed on MCF-7 and SK-BR-3 cells than on lung or pancreatic cancer cells. Among the 15 compounds tested, A6 and A15 exhibited potent cytotoxic and antimigratory activities against MCF-7 and SK-BR-3 cells in the CoCl2-induced hypoxic condition. While A6 and A15 markedly reduced the viability of control siRNA-treated cells, these compounds could not significantly reduce the viability of CA IX-knockdown cells, suggesting the role of CA IX in their anticancer activities. To assess whether these compounds exerted synergism with a conventional anticancer drug doxorubicin (DOX), the cytotoxic effects of A6 or A15 combined with DOX were analyzed using Chou−Talalay and Bliss independence methods. Our data revealed that both A6 and A15 significantly enhanced the anticancer activity of DOX. Among the tested pairs, the combination of DOX with A15 showed the strongest synergism on SK-BR-3 cells. Moreover, this combination further attenuated cell migration compared to the respective drug. Collectively, our results demonstrated that A6 and A15 suppressed tumor growth and cell migration of MCF-7 and SK-BR-3 cells through inhibition of CA IX, and the combination of these compounds with DOX exhibited synergistic cytotoxic effects on these breast cancer cells. Therefore, A6 and A15 may serve as potential anticancer agents alone or in combination with DOX against breast cancer.
Collapse
Affiliation(s)
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Young Hee Choi
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
- Correspondence: (K.L.); (J.C.)
| | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
- Correspondence: (K.L.); (J.C.)
| |
Collapse
|
13
|
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
|
14
|
Güller U, Beydemir Ş, Küfrevioğlu Öİ. In vitro and In silico Interactions of Antiulcer, Glucocorticoids and Urological Drugs on Human Carbonic Anhydrase I and II isozymes. Biopharm Drug Dispos 2022; 43:47-56. [PMID: 35080786 DOI: 10.1002/bdd.2309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 11/08/2022]
Abstract
Carbonic anhydrases (CAs, EC 4.2.1.1) convert carbon dioxide to bicarbonate in metabolism and use Zn2+ ions as a cofactor for their catalytic activity. The activators or inhibitors of CA-I and CA-II, which are the most abundant CA isozymes in erythrocytes, have pharmacological applications in medicine. So, investigation of drug-protein interaction of these isozymes is significant. On this basis, the objective of this study was to clarify the primer effects of widely used drugs on the activity of human CA-I and CA-II enzymes and elucidate the inhibition mechanism through molecular docking studies. For this aim isozymes were purified from human erythrocytes by affinity chromatography technique. Then inhibition profiles of antiulcer, glucocorticoids, and urological drugs were investigated. As a result, while budesonide had the highest inhibitory potency on hydratase activity of hCA-I with the IC50 of 0.08 mM, levofloxacin showed the highest inhibition effect on hCA-II with the IC50 of 0.886 mM. The most effective inhibitor on the esterase activity of isozymes was found as fluticasone propionate with the Ki values of 0.0365±0.016 mM and 0.054±0.018 mM respectively. However, by molecular docking study, it was estimated that budesonide showed maximum inhibition potency for both isozymes with the free binding energy of -7.58 and -6.97 kcal/mol respectively. Consequently, it was observed that some of the drugs studied did not show any inhibitory effect. Drug-enzyme interactions were also estimated by molecular docking. This study could contribute to the discovery of new drug candidates and as well as target proteins. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Uğur Güller
- Department of Food Engineering, Faculty of Engineering, Iğdır University, Iğdır, Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey.,The Rectorate of Bilecik Şeyh Edebali University, Bilecik, Turkey
| | | |
Collapse
|
15
|
Mahhengam N, Kazemnezhad K, Setia Budi H, Ansari MJ, Olegovich Bokov D, Suksatan W, Thangavelu L, Siahmansouri H. Targeted therapy of tumor microenvironment by gold nanoparticles as a new therapeutic approach. J Drug Target 2022; 30:494-510. [DOI: 10.1080/1061186x.2022.2032095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Negah Mahhengam
- Faculty of General Medicine, Belarusian State Medical University, Minsk, Belarus.
| | - Kimia Kazemnezhad
- Faculty of General Medicine, Belarusian State Medical University, Minsk, Belarus.
| | - Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University,Al-kharj, Saudi Arabia.
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, 119991, Russian Federation.
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand.
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India.
| | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Güller P, Dağalan Z, Güller U, Çalışır U, Nişancı B. Enzymes inhibition profiles and antibacterial activities of benzylidenemalononitrile derivatives. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
Pérez-Herrero E, Fernández-Medarde A. The reversed intra- and extracellular pH in tumors as a unified strategy to chemotherapeutic delivery using targeted nanocarriers. Acta Pharm Sin B 2021; 11:2243-2264. [PMID: 34522586 PMCID: PMC8424227 DOI: 10.1016/j.apsb.2021.01.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023] Open
Abstract
Solid tumors are complex entities, comprising a wide variety of malignancies with very different molecular alterations. Despite this, they share a set of characteristics known as "hallmarks of cancer" that can be used as common therapeutic targets. Thus, every tumor needs to change its metabolism in order to obtain the energy levels required for its high proliferative rates, and these adaptations lead to alterations in extra- and intracellular pH. These changes in pH are common to all solid tumors, and can be used either as therapeutic targets, blocking the cell proton transporters and reversing the pH changes, or as means to specifically deliver anticancer drugs. In this review we will describe how proton transport inhibitors in association with nanocarriers have been designed to block the pH changes that are needed for cancer cells to survive after their metabolic adaptations. We will also describe studies aiming to decrease intracellular pH in cancer using nanoparticles as molecular cages for protons which will be released upon UV or IR light exposure. Finally, we will comment on several studies that have used the extracellular pH in cancer for an enhanced cell internalization and tumor penetration of nanocarriers and a controlled drug delivery, describing how nanocarriers are being used to increase drug stability and specificity.
Collapse
Affiliation(s)
- Edgar Pérez-Herrero
- Departamento de Ingeniería Química y Tecnología Farmacéutica, Universidad de La Laguna, La Laguna 38206, Tenerife, Spain
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna 38206, Tenerife, Spain
- Instituto Universitario de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna 38200, Tenerife, Spain
| | - Alberto Fernández-Medarde
- Instituto de Biología Molecular y Celular Del Cáncer, Centro de Investigación Del Cáncer (USAL-CSIC), Salamanca 37007, Spain
| |
Collapse
|
18
|
Chen Y, Alba M, Tieu T, Tong Z, Minhas RS, Rudd D, Voelcker NH, Cifuentes-Rius A, Elnathan R. Engineering Micro–Nanomaterials for Biomedical Translation. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - Maria Alba
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO) Clayton VIC 3168 Australia
| | - Ziqiu Tong
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Rajpreet Singh Minhas
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - David Rudd
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
- Department of Materials Science and Engineering Monash University 22 Alliance Lane Clayton VIC 3168 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO) Clayton VIC 3168 Australia
- INM-Leibniz Institute for New Materials Campus D2 2 Saarbrücken 66123 Germany
| | - Anna Cifuentes-Rius
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
- Department of Materials Science and Engineering Monash University 22 Alliance Lane Clayton VIC 3168 Australia
| |
Collapse
|
19
|
Antibacterial properties and carbonic anhydrase inhibition profiles of azido sulfonyl carbamate derivatives. Future Med Chem 2021; 13:1285-1299. [PMID: 34075799 DOI: 10.4155/fmc-2020-0387] [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] [Indexed: 12/11/2022] Open
Abstract
Aim: The aim of this study was to identify inhibition of carbonic anhydrase I and II (CA I and II) isozymes by azido sulfonyl carbamates through both in vitro and in silico approaches and also to determine the drug-likeness properties and antibacterial activities of azido sulfonyl carbamates. Methods & Results: In vitro inhibition and molecular docking studies of azido sulfonyl carbamate derivatives (1-4) on isozymes were performed. Except for derivative 4, all derivatives inhibited human CA I and II. Almost all compounds had antibacterial effects. The docking results showed that compound 3 had the best results, with binding energy of -8.20 kcal/mol for human CA I and -8.24 kcal/mol for human CA II. Conclusion: Molecule 4 inhibited only CA I. Its usage as a potential chemotherapeutic agent specific to the CA I isozyme may be considered.
Collapse
|
20
|
Zhou H, Qin F, Chen C. Designing Hypoxia-Responsive Nanotheranostic Agents for Tumor Imaging and Therapy. Adv Healthc Mater 2021; 10:e2001277. [PMID: 32985141 DOI: 10.1002/adhm.202001277] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/06/2020] [Indexed: 12/15/2022]
Abstract
Hypoxia, a common feature of most solid tumors, plays an important role in tumor proliferation, metastasis, and invasion, leading to drug, radiation, and photodynamic therapy resistance, and resulting in a sharp reduction in the disease-free survival rate of tumor patients. The lack of sufficient blood supply to the interior regions of tumors hinders the delivery of traditional drugs and contrast agents, interfering with their accumulation in the hypoxic region, and preventing efficient theranostics. Thus, there is a need for the fabrication of novel tumor theranostic agents that overcome these obstacles. Reports, in recent years, of hypoxia-responsive nanomaterials may provide with such means. In this review, a comprehensive description of the physicochemical and biological characteristics of hypoxic tumor tissues is provided, the principles of designing the hypoxia-responsive tumor theranostic agents are discussed, and the recent research into hypoxia-triggered nanomaterials is examined. Additionally, other hypoxia-associated responsive strategies, the current limitations, and future prospects for hypoxia-responsive nanotheranostic agents in tumor treatment are discussed.
Collapse
Affiliation(s)
- Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| | - Fenglan Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| |
Collapse
|
21
|
Wu M, Zhong C, Zhang Q, Wang L, Wang L, Liu Y, Zhang X, Zhao X. pH-responsive delivery vehicle based on RGD-modified polydopamine-paclitaxel-loaded poly (3-hydroxybutyrate-co-3-hydroxyvalerate) nanoparticles for targeted therapy in hepatocellular carcinoma. J Nanobiotechnology 2021; 19:39. [PMID: 33549107 PMCID: PMC7866683 DOI: 10.1186/s12951-021-00783-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
A limitation of current anticancer nanocarriers is the contradiction between multiple functions and favorable biocompatibility. Thus, we aimed to develop a compatible drug delivery system loaded with paclitaxel (PTX) for hepatocellular carcinoma (HCC) therapy. A basic backbone, PTX-loaded poly (3-hydroxybutyrate-co-3-hydroxyvalerate) PHBV nanoparticle (PHBV-PTX-NPs), was prepared by emulsion solvent evaporation. As a gatekeeper, the pH-sensitive coating was formed by self-polymerization of dopamine (PDA). The HCC-targeted arginine-glycine-aspartic acid (RGD)-peptide and PDA-coated nanoparticles (NPs) were combined through the Michael addition. Subsequently, the physicochemical properties of RGD-PDA-PHBV-PTX-NPs were characterized by dynamic light scattering-autosizer, transmission electron microscope, fourier transform infrared spectroscopy, differential scanning calorimetry, thermogravimetry and X-ray spectroscopy. As expected, the RGD-PDA-PHBV-PTX-NPs showed robust anticancer efficacy in a xenograft mouse model. More importantly, they exhibited lower toxicity than PTX to normal hepatocytes and mouse in vitro and in vivo, respectively. Taken together, these results indicate that the RGD-PDA-PHBV-PTX-NPs are potentially beneficial for easing conflict between multifunction and biocompatible characters of nanocarriers. ![]()
Collapse
Affiliation(s)
- Mingfang Wu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 26 hexing road, Harbin, 150040, Heilongjiang, China.,School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, 310023, Zhejiang, China
| | - Chen Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Qian Zhang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 26 hexing road, Harbin, 150040, Heilongjiang, China.,Key Laboratory of Forest Plant Ecology, Northeast Forestry University, Ministry of Education, Harbin, 150040, Heilongjiang, China
| | - Lu Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 26 hexing road, Harbin, 150040, Heilongjiang, China.,Key Laboratory of Forest Plant Ecology, Northeast Forestry University, Ministry of Education, Harbin, 150040, Heilongjiang, China
| | - Lingling Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 26 hexing road, Harbin, 150040, Heilongjiang, China.,Key Laboratory of Forest Plant Ecology, Northeast Forestry University, Ministry of Education, Harbin, 150040, Heilongjiang, China
| | - Yanjie Liu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 26 hexing road, Harbin, 150040, Heilongjiang, China.,Key Laboratory of Forest Plant Ecology, Northeast Forestry University, Ministry of Education, Harbin, 150040, Heilongjiang, China
| | - Xiaoxue Zhang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 26 hexing road, Harbin, 150040, Heilongjiang, China.,Key Laboratory of Forest Plant Ecology, Northeast Forestry University, Ministry of Education, Harbin, 150040, Heilongjiang, China
| | - Xiuhua Zhao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 26 hexing road, Harbin, 150040, Heilongjiang, China. .,Key Laboratory of Forest Plant Ecology, Northeast Forestry University, Ministry of Education, Harbin, 150040, Heilongjiang, China.
| |
Collapse
|
22
|
Shabana AM, Kambhampati SP, Hsia RC, Kannan RM, Kokkoli E. Thermosensitive and biodegradable hydrogel encapsulating targeted nanoparticles for the sustained co-delivery of gemcitabine and paclitaxel to pancreatic cancer cells. Int J Pharm 2021; 593:120139. [PMID: 33278494 DOI: 10.1016/j.ijpharm.2020.120139] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 01/12/2023]
Abstract
Pancreatic cancer represents a life threatening disease with rising mortality. Although the synergistic combination of gemcitabine and albumin-bound paclitaxel has proven to enhance the median survival rates as compared to gemcitabine alone, their systemic and repeated co-administration has been associated with serious toxic side effects and poor patient compliance. For this purpose, we designed a thermosensitive and biodegradable hydrogel encapsulating targeted nanoparticles for the local and sustained delivery of gemcitabine (GEM) and paclitaxel (PTX) to pancreatic cancer. GEM and PTX were loaded into PR_b-functionalized liposomes targeting integrin α5β1, which was shown to be overexpressed in pancreatic cancer. PR_b is a fibronectin-mimetic peptide that binds to α5β1 with high affinity and specificity. The PR_b liposomes were encapsulated into a poly(δ-valerolactone-co-D,L-lactide)-b-poly(ethylene glycol)-b-poly(δ-valerolactone-co-D,L-lactide) (PVLA-PEG-PVLA) hydrogel and demonstrated sustained release of both drugs compared to PR_b-functionalized liposomes free in solution or free drugs in the hydrogel. Moreover, the hydrogel-nanoparticle system was proven to be very efficient towards killing monolayers of human pancreatic cancer cells (PANC-1), and showed a significant reduction in the growth pattern of PANC-1 tumor spheroids as compared to hydrogels encapsulating non-targeted liposomes with GEM/PTX or free drugs, after a one week treatment period. Our hybrid hydrogel-nanoparticle system is a promising platform for the local and sustained delivery of GEM/PTX to pancreatic cancer, with the goal of maximizing the therapeutic efficacy of this synergistic drug cocktail while potentially minimizing toxic side effects and eliminating the need for repeated co-administration.
Collapse
Affiliation(s)
- Ahmed M Shabana
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Siva P Kambhampati
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Ru-Ching Hsia
- Department of Neural and Pain Sciences, Electron Microscopy Core Imaging Facility, University of Maryland Baltimore Dental School, Baltimore, MD 21201, United States
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Efrosini Kokkoli
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States.
| |
Collapse
|
23
|
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
|
24
|
Daunys S, Janonienė A, Januškevičienė I, Paškevičiūtė M, Petrikaitė V. 3D Tumor Spheroid Models for In Vitro Therapeutic Screening of Nanoparticles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:243-270. [PMID: 33543463 DOI: 10.1007/978-3-030-58174-9_11] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The anticancer activity of compounds and nanoparticles is most often determined in the cell monolayer. However, three-dimensional (3D) systems, such as tumor spheroids, are more representing the natural tumor microenvironment. They have been shown to have higher invasiveness and resistance to cytotoxic agents and radiotherapy compared to cells growing in 2D monolayer. Furthermore, to improve the prediction of clinical efficacy of drugs, in the past decades, even more sophisticated systems, such as multicellular 3D cultures, closely representing natural tumor microenvironment have been developed. Those cultures are formed from either cell lines or patient-derived tumor cells. Such models are very attractive and could improve the selection of tested materials for clinical trials avoiding unnecessary expensive tests in vivo. The microenvironment in tumor spheroids is different, and those differences or the interaction between several cell populations may contribute to different tumor response to the treatment. Also, different types of nanoparticles may have different behavior in 3D models, depending on their nature, physicochemical properties, the presence of targeting ligands on the surface, etc. Therefore, it is very important to understand in which cases which type of tumor spheroid is more suitable for testing specific types of nanoparticles, which conditions should be used, and which analytical method should be applied.
Collapse
Affiliation(s)
- Simonas Daunys
- Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agnė Janonienė
- Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Indrė Januškevičienė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Miglė Paškevičiūtė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vilma Petrikaitė
- Life Sciences Center, Vilnius University, Vilnius, Lithuania.
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania.
- Institute of Physiology and Pharmacology, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| |
Collapse
|
25
|
Oguz M, Kalay E, Akocak S, Nocentini A, Lolak N, Boga M, Yilmaz M, Supuran CT. Synthesis of calix[4]azacrown substituted sulphonamides with antioxidant, acetylcholinesterase, butyrylcholinesterase, tyrosinase and carbonic anhydrase inhibitory action. J Enzyme Inhib Med Chem 2020; 35:1215-1223. [PMID: 32401067 PMCID: PMC7269057 DOI: 10.1080/14756366.2020.1765166] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
A series of novel calix[4]azacrown substituted sulphonamide Schiff bases was synthesised by the reaction of calix[4]azacrown aldehydes with different substituted primary and secondary sulphonamides. The obtained novel compounds were investigated as inhibitors of six human (h) isoforms of carbonic anhydrases (CA, EC 4.2.1.1). Their antioxidant profile was assayed by various bioanalytical methods. The calix[4]azacrown substituted sulphonamide Schiff bases were also investigated as inhibitors of acetylcholinesterase (AChE), butyrylcholinesterase (BChE) and tyrosinase enzymes, associated with several diseases such as Alzheimer, Parkinson, and pigmentation disorders. The new sulphonamides showed low to moderate inhibition against hCAs, AChE, BChE, and tyrosinase enzymes. However, some of them possessed relevant antioxidant activity, comparable with standard antioxidants used in the study.
Collapse
Affiliation(s)
- Mehmet Oguz
- Department of Chemistry, University of Selcuk, Konya, Turkey
- Department of Advanced Material and Nanotechnology, Selcuk University, Konya, Turkey
| | - Erbay Kalay
- Kars Vocational School, Kafkas University, Kars, Turkey
| | - Suleyman Akocak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adiyaman University, Adiyaman, Turkey
| | - Alessio Nocentini
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, Florence, Italy
| | - Nebih Lolak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adiyaman University, Adiyaman, Turkey
| | - Mehmet Boga
- Department of Analytical Chemistry, Faculty of Pharmacy, Dicle University, Diyarbakir, Turkey
| | - Mustafa Yilmaz
- Department of Chemistry, University of Selcuk, Konya, Turkey
| | - Claudiu T. Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
26
|
Li J, Tan T, Zhao L, Liu M, You Y, Zeng Y, Chen D, Xie T, Zhang L, Fu C, Zeng Z. Recent Advancements in Liposome-Targeting Strategies for the Treatment of Gliomas: A Systematic Review. ACS APPLIED BIO MATERIALS 2020; 3:5500-5528. [PMID: 35021787 DOI: 10.1021/acsabm.0c00705] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignant tumors represent some of the most intractable diseases that endanger human health. A glioma is a tumor of the central nervous system that is characterized by severe invasiveness, blurred boundaries between the tumor and surrounding normal tissue, difficult surgical removal, and high recurrence. Moreover, the blood-brain barrier (BBB) and multidrug resistance (MDR) are important factors that contribute to the lack of efficacy of chemotherapy in treating gliomas. A liposome is a biofilm-like drug delivery system with a unique phospholipid bilayer that exhibits high affinities with human tissues/organs (e.g., BBB). After more than five decades of development, classical and engineered liposomes consist of four distinct generations, each with different characteristics: (i) traditional liposomes, (ii) stealth liposomes, (iii) targeting liposomes, and (iv) biomimetic liposomes, which offer a promising approach to promote drugs across the BBB and to reverse MDR. Here, we review the history, preparatory methods, and physicochemical properties of liposomes. Furthermore, we discuss the mechanisms by which liposomes have assisted in the diagnosis and treatment of gliomas, including drug transport across the BBB, inhibition of efflux transporters, reversal of MDR, and induction of immune responses. Finally, we highlight ongoing and future clinical trials and applications toward further developing and testing the efficacies of liposomes in treating gliomas.
Collapse
Affiliation(s)
- Jie Li
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Tiantian Tan
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Liping Zhao
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Mengmeng Liu
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Yu You
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yiying Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Dajing Chen
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Tian Xie
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Lele Zhang
- School of Medicine, Chengdu University, Chengdu 610106, Sichuan, China
| | - Chaomei Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Zhaowu Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| |
Collapse
|
27
|
Akocak S, Güzel-Akdemir Ö, Kishore Kumar Sanku R, Russom SS, Iorga BI, Supuran CT, Ilies MA. Pyridinium derivatives of 3-aminobenzenesulfonamide are nanomolar-potent inhibitors of tumor-expressed carbonic anhydrase isozymes CA IX and CA XII. Bioorg Chem 2020; 103:104204. [PMID: 32891000 DOI: 10.1016/j.bioorg.2020.104204] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/26/2022]
Abstract
Building on the conclusions of previous inhibition studies with pyridinium-benzenesulfonamides from our team and on the X-ray crystal structure of the lead compound identified, a series of 24 pyridinium derivatives of 3-aminobenzenesulfonamide was synthesized and investigated for carbonic anhydrase inhibition. The new pyridinium-sulfonamides were evaluated as inhibitors of four human carbonic anhydrase (CA, EC 4.2.1.1) isoforms, namely CA I, CA II (cytosolic), CA IX and XII (transmembrane, tumor-associated forms). Excellent inhibitory activity in the nanomolar range was observed against CA IX with most of these sulfonamides, and against CA XII (nanomolar/sub-nanomolar) with some of the new compounds. These sulfonamides were generally potent inhibitors of CA II and CA I too. Docking studies revealed a preference of these compounds to bind the P1 hydrophobic site of CAs, supporting the observed inhibition profile. The salt-like nature of these positively charged sulfonamides can further focus the inhibitory ability on membrane-bound CA IX and CA XII and could efficiently decrease the viability of three human carcinomas under hypoxic conditions where these isozymes are over-expressed, thus recommending the new compounds as potential diagnostic tools or therapeutic agents.
Collapse
Affiliation(s)
- Suleyman Akocak
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N Broad Street, Philadelphia PA-19140, United States
| | - Özlen Güzel-Akdemir
- NEUROFARBA Department, Università degli Studi di Firenze, Polo Scientifico, Via della Lastruccia 3, 50019 Sesto Fiorentino (Florence), Italy; Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34116 Beyazıt, Istanbul, Turkey
| | - Rajesh Kishore Kumar Sanku
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N Broad Street, Philadelphia PA-19140, United States
| | - Samson S Russom
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N Broad Street, Philadelphia PA-19140, United States
| | - Bogdan I Iorga
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles (ICSN), 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Claudiu T Supuran
- NEUROFARBA Department, Università degli Studi di Firenze, Polo Scientifico, Via della Lastruccia 3, 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 PA-19140, United States.
| |
Collapse
|
28
|
Mó I, Sabino IJ, Melo-Diogo DD, Lima-Sousa R, Alves CG, Correia IJ. The importance of spheroids in analyzing nanomedicine efficacy. Nanomedicine (Lond) 2020; 15:1513-1525. [PMID: 32552537 DOI: 10.2217/nnm-2020-0054] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of nanomedicines for cancer treatment holds a great potential due to their improved efficacy and safety. During the nanomedicine preclinical in vitro evaluation stage, these are mainly tested on cell culture monolayers. However, these 2D models are an unrealistic representation of the in vivo tumors, leading to an inaccurate screening of the candidate formulations. To address this problem, spheroids are emerging as an additional tool to validate the efficacy of new therapeutics due to the ability of these 3D in vitro cancer models to mimic the key features displayed by in vivo solid tumors. In this review, the application of spheroids for the evaluation of nanomedicines' physicochemical properties and therapeutic efficacy is discussed.
Collapse
Affiliation(s)
- Inês Mó
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Ivo J Sabino
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Duarte de Melo-Diogo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Rita Lima-Sousa
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Cátia G Alves
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Ilídio J Correia
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal.,CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, 3030-790, Coimbra, Portugal
| |
Collapse
|
29
|
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
|
30
|
Lepeltier E, Rijo P, Rizzolio F, Popovtzer R, Petrikaite V, Assaraf YG, Passirani C. Nanomedicine to target multidrug resistant tumors. Drug Resist Updat 2020; 52:100704. [PMID: 32512316 DOI: 10.1016/j.drup.2020.100704] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/21/2019] [Accepted: 05/13/2020] [Indexed: 12/12/2022]
Abstract
Nanomedicine employs nanotechnologies to develop innovative applications, and more specifically nano-objects in the field of human health, through exploitation of the physical, chemical and biological properties of materials at the nanoscale. The use of nanovehicles capable of transporting and releasing the active therapeutic payload into target cells, particularly in the case of cancer or inflammatory diseases, can also enhance diagnosis. Therefore, nanomedicines improve the benefit/risk ratio of drugs by increasing their bioavailability, selectivity, and efficacy in the target tissue, while reducing the necessary doses and hence diminishing untoward toxicity to healthy tissues. Overcoming multidrug resistance (MDR) to antitumor agents is a central goal of cancer research and therapeutics, making it possible to treat these diseases more accurately and effectively. The adaptability of nanomedicines e.g. modulation of their components, surface functionalization, encapsulation of various active therapeutics as well as the possibility of combining several treatments using a single nanoparticle platform, are characteristics which are perfectly poised to address classical chemoresistance, a major obstacle towards curative cancer therapy. In this review, we discuss an assortment of nanomedicines along with those that should be developed in order to surmount cancer MDR; these include exosomes, natural compounds, lipid nanocapsules, prodrug self-assemblies, and gold nanoparticles.
Collapse
Affiliation(s)
- Elise Lepeltier
- Micro et Nanomédecines Translationnelles, MINT, UNIV Angers, UMR INSERM 1066, UMR CNRS 6021, Angers, France
| | - Patricia Rijo
- Research Center for Biosciences & Health Technologies (CBIOS), Lisboa, Portugal; iMed.ULisboa - Research Institute for Medicines, Lisboa, Portugal
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, 30123 Venezia, Italy; Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology & Advanced Materials Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Vilma Petrikaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių Av. 13, LT-50161 Kaunas, Lithuania; Institute of Physiology and Pharmacology, Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus 9, LT-44307 Kaunas, Lithuania
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Catherine Passirani
- Micro et Nanomédecines Translationnelles, MINT, UNIV Angers, UMR INSERM 1066, UMR CNRS 6021, Angers, France.
| |
Collapse
|
31
|
An P, Fan F, Gu D, Gao Z, Hossain AMS, Sun B. Photothermal-reinforced and glutathione-triggered in Situ cascaded nanocatalytic therapy. J Control Release 2020; 321:734-743. [PMID: 32145265 DOI: 10.1016/j.jconrel.2020.03.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/01/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
Tumor microenvironment (TME)-responsive nanoformulations that catalyze a cascade of intracellular redox reactions showed promise for tumor treatment with high specificity and efficiency. In this study, we report Cu2+-doped zeolitic imidazolate frameworks-coated polydopamine nanoparticles (PDA@Cu/ZIF-8 NPs) for glutathione-triggered and photothermal-reinforced sequential catalytic therapy against breast cancer. In the TME, the PDA@Cu/ZIF-8 NPs could initially react with antioxidant glutathione (GSH), inducing GSH depletion and Cu+ generation. Whereafter, the generated Cu+ would catalyze local H2O2 to produce highly toxic hydroxyl radicals (·OH) through an efficient Fenton-like reaction even in weakly acidity. Importantly, the PDA could exert excellent photothermal conversion effect to simultaneously accelerate GSH consumption and improve the Fenton-like reaction for further expanding the intracellular oxidative stress, which innovatively achieves a synergistic photothermal-chemodynamic therapy for highly efficient anticancer treatment.
Collapse
Affiliation(s)
- Peijing An
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, PR China
| | - Fengying Fan
- Shanghai Institute of Materia Medica, Chinese Academy of Science, 501 Haike Road, Shanghai 201210, PR China
| | - Dihai Gu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, PR China
| | - Zhiguo Gao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, PR China
| | | | - Baiwang Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, PR China.
| |
Collapse
|
32
|
Caballero AB, Cardo L, Claire S, Craig JS, Hodges NJ, Vladyka A, Albrecht T, Rochford LA, Pikramenou Z, Hannon MJ. Assisted delivery of anti-tumour platinum drugs using DNA-coiling gold nanoparticles bearing lumophores and intercalators: towards a new generation of multimodal nanocarriers with enhanced action. Chem Sci 2019; 10:9244-9256. [PMID: 32055309 PMCID: PMC7003971 DOI: 10.1039/c9sc02640a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
New gold and lipoic based nanocarriers for the delivery of platinum(ii) and platinum(iv) drugs are developed, which allow enhanced loading of the drug on the surface of the nanocarriers and release in a pH-dependent fashion, with superior release at lower pHs which are associated with many tumours. The conjugate nanoparticles and their conjugates enter cells rapidly (within 3 hours). They tend to cluster in vesicles and are also observed by light and electron microscopies in the cytoplasm, endoplasmic reticulum and nucleus. We further incorporate aminoanthraquinone units that are both fluorophores and DNA intercalators. This results in nanocarriers that after drug release will remain surface decorated with DNA-binders challenging the conventional design of the nanocarrier as an inert component. The outcome is nanocarriers that themselves have distinctive, remarkable and unusual DNA binding properties being able to bind and wrap DNA (despite their anionic charge) and provide enhanced cytotoxic activity beyond that conferred by the platinum agents they release. DNA coiling is usually associated with polycations which can disrupt cell membranes; anionic nanoparticles that can cause novel and dramatic effects on DNA may have fascinating potential for new approaches to in-cell nucleic acid recognition. Our findings have implications for the understanding and interpretation of the biological activities of nanoparticles used to deliver other DNA-binding drugs including clinical drug doxorubicin and its formulations.
Collapse
Affiliation(s)
- Ana B Caballero
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| | - Lucia Cardo
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| | - Sunil Claire
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| | - James S Craig
- Physical Sciences for Health Centre , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK
| | - Nikolas J Hodges
- School of Biosciences , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK
| | - Anton Vladyka
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| | - Tim Albrecht
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| | - Luke A Rochford
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| | - Zoe Pikramenou
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| | - Michael J Hannon
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , UK . ;
| |
Collapse
|
33
|
Multivalent Carbonic Anhydrases Inhibitors. Int J Mol Sci 2019; 20:ijms20215352. [PMID: 31661796 PMCID: PMC6862271 DOI: 10.3390/ijms20215352] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Biomolecular recognition using a multivalent strategy has been successfully applied, this last decade on several biological targets, especially carbohydrate-processing enzymes, proteases, and phosphorylases. This strategy is based on the fact that multivalent interactions of several inhibitory binding units grafted on a presentation platform may enhance the binding affinity and selectivity. The zinc metalloenzymes carbonic anhydrases (CAs, EC 4.2.1.1) are considered as drug targets for several pathologies, and different inhibitors found clinical applications as diuretics, antiglaucoma agents, anticonvulsants, and anticancer agents/diagnostic tools. Their main drawback is related to the lack of isoform selectivity leading to serious side effects for all pathologies in which they are employed. Thus, the multivalent approach may open new opportunities in the drug design of innovative isoform-selective carbonic anhydrase inhibitors with biomedical applications.
Collapse
|
34
|
Deng L, Feng Z, Deng H, Jiang Y, Song K, Shi Y, Liu S, Zhang J, Bai S, Qin Z, Dong A. Rational Design of Nanoparticles to Overcome Poor Tumor Penetration and Hypoxia-Induced Chemotherapy Resistance: Combination of Optimizing Size and Self-Inducing High Level of Reactive Oxygen Species. ACS APPLIED MATERIALS & INTERFACES 2019; 11:31743-31754. [PMID: 31389686 DOI: 10.1021/acsami.9b12129] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One tough question induced by the hypoxia in cancer tissue is resistance to anticancer drugs basing on the reactive oxygen species (ROS) mechanism. Furthermore, the hypoxic regions locate in the center of tumor where tumor cells are easily residual and survival due to the poor drug-delivery efficiency even with nanocarriers. In this paper, these problems were well addressed through the rational combination of the enhanced penetration, self-inducing high level of intracellular ROS, and synchronously pH-sensitive drug release, realized by a simple structural and accessible copolymer, poly(poly(ethylene glycol) methyl ether methacrylate-co-(2-methylpropenoic acid-glycerol-cinnamaldehyde)) (PgEMC). For one thing, PgEMC could self-assemble into stable nanoparticles with PEG shell and optimizing diameters of 60 nm to simultaneously facilitate long blood circulation and deep tumor penetration. Second, cinnamylaldehyde moieties could detach from PgEMC NPs in intracellular acidic environment and trigger high level of ROS to allay the doxorubicin (DOX) resistance induced by hypoxia in solid malignancies. Furthermore, the DOX payload in PgEMC NPs could be synchronously released with the intracellular disassembly of PgEMC NPs due to the detaching of cinnamylaldehyde moieties. In 4T1 cells treated with PgEMC/DOX NPs, remarkable elevation of ROS level and enhanced DOX sensitivity in hypoxia environment were observed in in vitro studies. The results of tumor spheroid penetration indicated that 60 nm sized DOX-loaded PgEMC NPs (PgEMC60/DOX) could distribute into deep site of tumor at a high intensity. In vivo studies using a 4T1 breast tumor model, PgEMC60/DOX NPs, showed significant inhibition over 95.4% of the tumor growth. These results reveal that integrating optimizing size, self-inducing ROS, and pH-sensitive drug release into one small-sized nanoparticle can efficiently overcome the poor tumor penetration and hypoxia-induced chemotherapy resistance.
Collapse
Affiliation(s)
- Liandong Deng
- College of Pharmacy , Xinxiang Medical University , Xinxiang 453003 , China
- Department of Polymer Science and Technology, Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
| | - Zujian Feng
- College of Pharmacy , Xinxiang Medical University , Xinxiang 453003 , China
- Department of Polymer Science and Technology, Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
| | - Hongzhang Deng
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
- Department of Polymer Science and Technology, Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300072 , China
| | - Yujia Jiang
- Department of Polymer Science and Technology, Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
| | - Kun Song
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Yongli Shi
- College of Pharmacy , Xinxiang Medical University , Xinxiang 453003 , China
| | - Shuangqing Liu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Jianhua Zhang
- Department of Polymer Science and Technology, Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
| | - Suping Bai
- College of Pharmacy , Xinxiang Medical University , Xinxiang 453003 , China
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Anjie Dong
- College of Pharmacy , Xinxiang Medical University , Xinxiang 453003 , China
- Department of Polymer Science and Technology, Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300072 , China
| |
Collapse
|
35
|
Roma-Rodrigues C, Pombo I, Raposo L, Pedrosa P, Fernandes AR, Baptista PV. Nanotheranostics Targeting the Tumor Microenvironment. Front Bioeng Biotechnol 2019; 7:197. [PMID: 31475143 PMCID: PMC6703081 DOI: 10.3389/fbioe.2019.00197] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer is considered the most aggressive malignancy to humans, and definitely the major cause of death worldwide. Despite the different and heterogenous presentation of the disease, there are pivotal cell elements involved in proliferation, differentiation, and immortalization, and ultimately the capability to evade treatment strategies. This is of utmost relevance when we are just beginning to grasp the complexity of the tumor environment and the molecular "evolution" within. The tumor micro-environment (TME) is thought to provide for differentiation niches for clonal development that results in tremendous cancer heterogeneity. To date, conventional cancer therapeutic strategies against cancer are failing to tackle the intricate interplay of actors within the TME. Nanomedicine has been proposing innovative strategies to tackle this TME and the cancer cells that simultaneously provide for biodistribution and/or assessment of action. These nanotheranostics systems are usually multi-functional nanosystems capable to carry and deliver active cargo to the site of interest and provide diagnostics capability, enabling early detection, and destruction of cancer cells in a more selective way. Some of the most promising multifunctional nanosystems are based on gold nanoparticles, whose physic-chemical properties have prompt for the development of multifunctional, responsive nanomedicines suitable for combinatory therapy and theranostics. Herein, we shall focus on the recent developments relying on the properties of gold nanoparticles as the basis for nanotheranostics systems against the heterogeneity within the TME.
Collapse
Affiliation(s)
| | | | | | | | | | - Pedro V. Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Costa da Caparica, Portugal
| |
Collapse
|
36
|
Abstract
INTRODUCTION The physiologic importance of fast CO2/HCO3- interconversion in various tissues requires the presence of carbonic anhydrase (CA, EC 4.2.1.1). Fourteen CA isozymes are present in humans, all of them being used as biomarkers. AREAS COVERED A great number of patents and articles were focused on the use of CA isozymes as biomarkers for various diseases and syndromes in the recent years, in an ascending trend over the last decade. The review highlights the most important studies related with each isozyme and covers the most recent patent literature. EXPERT OPINION The CAs biomarker research area expanded significantly in recent years, shifting from the predominant use of CA IX and CA XII in cancer diagnostic, staging, and prognosis towards a wider use of CA isozymes as disease biomarkers. CA isozymes are currently used either alone, in tandem with other CA isozymes and/or in combination with other proteins for the detection, staging, and prognosis of a huge repertoire of human dysfunctions and diseases, ranging from mild transformation of the normal tissues to extreme shifts in tissue organization and function. The techniques used for their detection/quantitation and the state-of-the-art in each clinical application are presented through relevant clinical examples and corresponding statistical data.
Collapse
Affiliation(s)
- Sabina Zamanova
- a Department of Pharmaceutical Sciences and Moulder Center of Drug Discovery Research , Temple University School of Pharmacy , Philadelphia , PA , USA
| | - Ahmed M Shabana
- a Department of Pharmaceutical Sciences and Moulder Center of Drug Discovery Research , Temple University School of Pharmacy , Philadelphia , PA , USA
| | - Utpal K Mondal
- a Department of Pharmaceutical Sciences and Moulder Center of Drug Discovery Research , Temple University School of Pharmacy , Philadelphia , PA , USA
| | - Marc A Ilies
- a Department of Pharmaceutical Sciences and Moulder Center of Drug Discovery Research , Temple University School of Pharmacy , Philadelphia , PA , USA.,b Temple Fox Chase Cancer Center , Philadelphia , PA , USA
| |
Collapse
|
37
|
Bugárová N, Špitálsky Z, Mičušík M, Bodík M, Šiffalovič P, Koneracká M, Závišová V, Kubovčíková M, Kajanová I, Zaťovičová M, Pastoreková S, Šlouf M, Majková E, Omastová M. A Multifunctional Graphene Oxide Platform for Targeting Cancer. Cancers (Basel) 2019; 11:cancers11060753. [PMID: 31146494 PMCID: PMC6627436 DOI: 10.3390/cancers11060753] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 12/21/2022] Open
Abstract
Diagnosis of oncological diseases remains at the forefront of current medical research. Carbonic Anhydrase IX (CA IX) is a cell surface hypoxia-inducible enzyme functionally involved in adaptation to acidosis that is expressed in aggressive tumors; hence, it can be used as a tumor biomarker. Herein, we propose a nanoscale graphene oxide (GO) platform functionalized with magnetic nanoparticles and a monoclonal antibody specific to the CA IX marker. The GO platforms were prepared by a modified Hummers and Offeman method from exfoliated graphite after several centrifugation and ultrasonication cycles. The magnetic nanoparticles were prepared by a chemical precipitation method and subsequently modified. Basic characterization of GO, such as the degree of oxidation, nanoparticle size and exfoliation, were determined by physical and chemical analysis, including X-ray photoelectron spectroscopy (XPS), transmission electron microscopy (TEM), energy dispersive X-ray analysis (EDX), and atomic force microscopy (AFM). In addition, the size and properties of the poly-L-lysine-modified magnetic nanoparticles were characterized. The antibody specific to CA IX was linked via an amidic bond to the poly-L-lysine modified magnetic nanoparticles, which were conjugated to GO platform again via an amidic bond. The prepared GO-based platform with magnetic nanoparticles combined with a biosensing antibody element was used for a hypoxic cancer cell targeting study based on immunofluorescence.
Collapse
Affiliation(s)
- Nikola Bugárová
- Polymer Institute, SAS, Dúbravská cesta 9, 845 41 Bratislava, Slovakia.
| | - Zdenko Špitálsky
- Polymer Institute, SAS, Dúbravská cesta 9, 845 41 Bratislava, Slovakia.
| | - Matej Mičušík
- Polymer Institute, SAS, Dúbravská cesta 9, 845 41 Bratislava, Slovakia.
| | - Michal Bodík
- Institute of Physics, SAS, Dúbravská cesta 9, 845 11 Bratislava, Slovakia.
| | - Peter Šiffalovič
- Institute of Physics, SAS, Dúbravská cesta 9, 845 11 Bratislava, Slovakia.
| | - Martina Koneracká
- Institute of Experimental Physics, SAS, Watsonova 47, 040 01 Košice, Slovakia.
| | - Vlasta Závišová
- Institute of Experimental Physics, SAS, Watsonova 47, 040 01 Košice, Slovakia.
| | - Martina Kubovčíková
- Institute of Experimental Physics, SAS, Watsonova 47, 040 01 Košice, Slovakia.
| | - Ivana Kajanová
- Institute of Virology, Biomedical Research Center, SAS, Dúbravská cesta 9, 845 11 Bratislava, Slovakia.
| | - Miriam Zaťovičová
- Institute of Virology, Biomedical Research Center, SAS, Dúbravská cesta 9, 845 11 Bratislava, Slovakia.
| | - Silvia Pastoreková
- Institute of Virology, Biomedical Research Center, SAS, Dúbravská cesta 9, 845 11 Bratislava, Slovakia.
| | - Miroslav Šlouf
- Institute of Macromolecular Chemistry AS CR, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic.
| | - Eva Majková
- Institute of Physics, SAS, Dúbravská cesta 9, 845 11 Bratislava, Slovakia.
| | - Mária Omastová
- Polymer Institute, SAS, Dúbravská cesta 9, 845 41 Bratislava, Slovakia.
| |
Collapse
|
38
|
Lolak N, Akocak S, Bua S, Sanku RKK, Supuran CT. Discovery of new ureido benzenesulfonamides incorporating 1,3,5-triazine moieties as carbonic anhydrase I, II, IX and XII inhibitors. Bioorg Med Chem 2019; 27:1588-1594. [PMID: 30846402 DOI: 10.1016/j.bmc.2019.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 12/22/2022]
Abstract
A series of twenty novel ureido benzenesulfonamides incorporating 1,3,5-triazine moieties substituted on one side with aromatic amines and on the other side with dimethylamine, morpholine and piperidine is reported. The compounds were synthesized from the 4-(3-(4,6-dichloro-1,3,5-triazin-2-yl)ureido)benzensulfonamide (1) by using stepwise nucleophilic substitution of the chlorine atoms of cyanuric chloride. The intermediates 2(a-e) and final compounds 3(a-o) were tested for their efficiency as carbonic anhydrase (CA) inhibitors against four selected physiologically relevant human carbonic anhydrase (CA, EC 4.2.1.1) isoforms, namely, the cytosolic ones hCA I and II, and the transmembrane, tumor associated ones hCA IX, and XII. The compounds 2a, 2e and 3m showed the highest activity for hCA IX with Kis in the range of 11.8-14.6 nM. Most of the compounds showed high hCA IX selectivity over the abundant off-target isoforms hCA I and II. Since hCA IX is a validated drug target for anticancer/antimetastatic agents, these isoform-selective and potent inhibitors may be considered of interest for further medicinal/pharmacologic studies.
Collapse
Affiliation(s)
- Nabih Lolak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adiyaman University, 02040 Adiyaman, Turkey
| | - Suleyman Akocak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adiyaman University, 02040 Adiyaman, Turkey.
| | - Silvia Bua
- Università degli Studi di Firenze, NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Rajesh K K Sanku
- University of Pennsylvania, Perelman School of Medicine, Department of Systems Pharmacology and Translational Therapeutics, 19104 Philadelphia, United States
| | - Claudiu T Supuran
- Università degli Studi di Firenze, NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy.
| |
Collapse
|
39
|
Sarkar A, Carvalho E, D'souza AA, Banerjee R. Liposome-encapsulated fish oil protein-tagged gold nanoparticles for intra-articular therapy in osteoarthritis. Nanomedicine (Lond) 2019; 14:871-887. [PMID: 30895865 DOI: 10.2217/nnm-2018-0221] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
AIM To provide multilayered combination therapies encompassing nanoparticles and organic peptides and to assess their efficacy in the treatment of arthritis. MATERIALS & METHODS Fish oil protein (FP) was isolated from fish oil glands and tagged with spherical gold nanoparticles (GNPs). Tagged GNPs were encapsulated in DPPC liposomes (FP-GNP-DPPC) and characterized. RESULTS & CONCLUSION FP increased the hydrophilicity of GNP, while encapsulation of FP-GNP within liposomes increased the hydrophobicity. In vitro release studies of FP-GNP-DPPC exhibited sustained release of FP in simulated synovial fluid. FP-GNP-DPPC injected into intra-articular joints of rats displayed anti-osteoarthritic effects in osteoarthritic rat model. This is the first study to report the anti-osteoarthritic activity of FP and DPPC encapsulated FP-GNP liposomes.
Collapse
Affiliation(s)
- Amrita Sarkar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India.,Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Edmund Carvalho
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India.,Department of Microbiology, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Anisha A D'souza
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India
| | - Rinti Banerjee
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India
| |
Collapse
|
40
|
Jahangirian H, Kalantari K, Izadiyan Z, Rafiee-Moghaddam R, Shameli K, Webster TJ. A review of small molecules and drug delivery applications using gold and iron nanoparticles. Int J Nanomedicine 2019; 14:1633-1657. [PMID: 30880970 PMCID: PMC6417854 DOI: 10.2147/ijn.s184723] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Conventional cancer treatment techniques show several limitations including low or no specificity and consequently a low efficacy in discriminating between cancer cells and healthy cells. Recent nanotechnology developments have introduced smart and novel therapeutic nanomaterials that take advantage of various targeting approaches. The use of nanotechnology in medicine and, more specifically, drug delivery is set to spread even more rapidly than it has over the past two decades. Currently, many nanoparticles (NPs) are under investigation for drug delivery including those for cancer therapy. Targeted nanomaterials bind selectively to cancer cells and greatly affect them with only a minor effect on healthy cells. Gold nanoparticles (Au-NPs), specifically, have been identified as significant candidates for new cancer therapeutic modalities because of their biocompatibility, easy functionalization and fabrication, optical tunable characteristics, and chemophysical stability. In the last decade, there has been significant research on Au-NPs and their biomedical applications. Functionalized Au-NPs represent highly attractive and promising candidates for drug delivery, owing to their unique dimensions, tunable surface functionalities, and controllable drug release. Further, iron oxide NPs due to their "superparamagnetic" properties have been studied and have demonstrated successful employment in numerous applications. In targeted drug delivery systems, drug-loaded iron oxide NPs can accumulate at the tumor site with the aid of an external magnetic field. This can lead to incremental effectiveness in drug release to the tumor site and vanquish cancer cells without harming healthy cells. In order for the application of iron oxide NPs in the human body to be realized, they should be biodegradable and biocompatible to minimize toxicity. This review illustrates recent advances in the field drug and small molecule delivery such as fluorouracil, folic acid, doxorubicin, paclitaxel, and daunorubicin, specifically when using gold and iron oxide NPs as carriers of anticancer therapeutic agents.
Collapse
Affiliation(s)
- Hossein Jahangirian
- Department of Chemical Engineering, 313 Snell Engineering Center, Northeastern University, Boston, MA, USA,
| | - Katayoon Kalantari
- Centre of Advanced Materials (CAM), Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Zahra Izadiyan
- Department of Environment and Green Technology, Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Kuala Lumpur, Malaysia
| | - Roshanak Rafiee-Moghaddam
- Department of Chemical Engineering, 313 Snell Engineering Center, Northeastern University, Boston, MA, USA,
| | - Kamyar Shameli
- Department of Environment and Green Technology, Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Kuala Lumpur, Malaysia
| | - Thomas J Webster
- Department of Chemical Engineering, 313 Snell Engineering Center, Northeastern University, Boston, MA, USA,
| |
Collapse
|
41
|
Lolak N, Akocak S, Bua S, Supuran CT. Design, synthesis and biological evaluation of novel ureido benzenesulfonamides incorporating 1,3,5-triazine moieties as potent carbonic anhydrase IX inhibitors. Bioorg Chem 2019; 82:117-122. [DOI: 10.1016/j.bioorg.2018.10.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 10/28/2022]
|
42
|
Song N, Li Y, Chen L, Hu X, Xie Z. BODIPY derivatives as light-induced free radical generators for hypoxic cancer treatment. J Mater Chem B 2019. [DOI: 10.1039/c9tb00694j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
NBDP can self-assemble into nanoparticles (NBDP NPs), which could be internalized by cells via endocytosis. After being irradiated with an 808 nm laser, the BODIPY unit could convert photons into heat and further initiates the production of alkyl radicals. The generated tumor hyperthermia and alkyl radicals synergistically kill cancer cells.
Collapse
Affiliation(s)
- Nan Song
- Department of Chemistry
- Northeast Normal University
- Changchun 130024
- P. R. China
| | - Yuanyuan Li
- The First Hospital of Jilin University
- Changchun
- P. R. China
| | - Li Chen
- Department of Chemistry
- Northeast Normal University
- Changchun 130024
- P. R. China
| | - Xiuli Hu
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| |
Collapse
|
43
|
Supuran CT. Carbonic anhydrase inhibitors as emerging agents for the treatment and imaging of hypoxic tumors. Expert Opin Investig Drugs 2018; 27:963-970. [PMID: 30426805 DOI: 10.1080/13543784.2018.1548608] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Hypoxic tumors overexpress two carbonic anhydrases (CA, EC 4.2.1.1), CA IX and XII, involved in complex processes connected to tumorigenesis (pH regulation, metabolism, invasion, and dissemination of the tumor). The biochemical rationale behind these processes is orchestrated by the transcription factor hypoxia inducible factor 1 (HIF-1). AREAS COVERED CA IX and XII have been validated as antitumor/antimetastatic drug targets and may be used for imaging hypoxic tumors. Many CA inhibitors (CAIs) belonging to the sulfonamide, coumarin and sulfocoumarin classes selectively inhibit these two isoforms. CA IX/XII inhibitors inhibit the growth of primary tumors and the formation of metastases and deplete the cancer stem cell population, alone or in combination with other agents. These are three beneficial antitumor mechanisms that make them unique among anticancer drugs available. EXPERT OPINION Indisulam entered clinical trials as an antitumor sulfonamide; it progressed to Phase II trials but was terminated in 2016. However, SLC-0111, a sulfonamide CA IX/XII inhibitor 1, recently completed a successful Phase I clinical trial for the treatment of advanced, metastatic solid tumors. This compound is now in Phase Ib/II clinical trials and is being assessed as a monotherapy or in combination with other agents such as gemcitabine. CA IX/XII inhibitors are synergistic with other anticancer agents (cisplatin, proton pump inhibitors, doxorubicin, temozolamide) and are a versatile, emerging class of antitumor drugs.
Collapse
Affiliation(s)
- Claudiu T Supuran
- a NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Sesto Fiorentino (Firenze) , Italy
| |
Collapse
|
44
|
Akocak S, Lolak N, Bua S, Supuran CT. Discovery of novel 1,3-diaryltriazene sulfonamides as carbonic anhydrase I, II, VII, and IX inhibitors. J Enzyme Inhib Med Chem 2018; 33:1575-1580. [PMID: 30296852 PMCID: PMC6179046 DOI: 10.1080/14756366.2018.1515933] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A series of new 1,3-diaryltriazene sulfonamides was synthesised by reaction of diazonium salt of metanilamide (3-aminobenzene sulfonamide) with substituted aromatic amines. The obtained new compounds were assayed as inhibitors of four physiologically and pharmacologically relevant human (h) isoforms of carbonic anhydrases (CA, EC 4.2.1.1), specifically, hCA I, hCA II, and hCA VII (cytosolic isoforms), as well as the tumour-associated membrane-bound isoform hCA IX. All isoforms investigated here were inhibited by the newly synthesised 1,3-diaryltriazene sulfonamide derivatives from the micromolar to the nanomolar range. The cytosolic isoforms were inhibited with Kis in the range of 92.3–8371.1 nM (hCA I), 4.3–9194.0 nM (hCA II), and 15.6–9477.8 nM (hCA VII), respectively. For the membrane-bound tumour-associated isoform hCA IX, the KI-s ranged between 50.8 and 9268.5 nM. The structure–activity relationship (SAR) with these newly synthesised metanilamide derivatives are discussed in detail.
Collapse
Affiliation(s)
- Suleyman Akocak
- a Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Adiyaman University , Adiyaman , Turkey
| | - Nabih Lolak
- a Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Adiyaman University , Adiyaman , Turkey
| | - Silvia Bua
- b NEUROFARBA Dept., Sezione di Scienze Farmaceutiche , Università degli Studi di Firenze , Sesto Fiorentino (Florence) , Italy
| | - Claudiu T Supuran
- b NEUROFARBA Dept., Sezione di Scienze Farmaceutiche , Università degli Studi di Firenze , Sesto Fiorentino (Florence) , Italy
| |
Collapse
|