1
|
Lima E, Ferreira O, Boto RE, Fernandes JR, Almeida P, Silvestre SM, Santos AO, Reis LV. D-(+)-Biotinylated squaraine dyes: A journey from synthetic conception, photophysical and -chemical characterization, to the exploration of their photoantitumoral action mechanisms. Eur J Med Chem 2025; 293:117699. [PMID: 40349637 DOI: 10.1016/j.ejmech.2025.117699] [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: 03/19/2025] [Revised: 04/14/2025] [Accepted: 04/27/2025] [Indexed: 05/14/2025]
Abstract
Biotin is primarily taken up by cells through sodium-dependent multivitamin transporter, which is highly expressed in aggressive cancer cell lines, often at levels surpassing those of the folate receptor. This makes biotin an attractive ligand for tumor-targeted drug delivery. Building on this rationale, this study presents a series of six D-(+)-biotin-conjugated squaraine dyes derived from benzothiazole, indolenine, and benz[e]indole, with N-ethyl and N-hexyl chains. These compounds were thoroughly characterized in terms of their photophysical and photochemical properties, revealing strong absorption in the so-called "phototherapeutic window", notable fluorescence, especially the benzothiazole derivatives, aqueous stability, particularly the indolenine-based dyes, and moderate to high photostability. Computational studies further indicated a strong binding affinity to human serum albumin and avidin proteins. All dyes exhibited photodynamic activity, with indolenine derivatives showing remarkable tumor selectivity and benz[e]indole analogs evidencing superior photocytotoxicity. The most promising compounds preferentially accumulated in mitochondria, and both singlet oxygen and other reactive oxygen species were found to play a role in their photobiological effects. Additionally, they were non-genotoxic in the absence of irradiation, and apoptosis was the primary mechanism of cell death upon light activation. This was evidenced by preserved cytoplasmic membrane integrity, nuclear fragmentation, and caspase-3/7 activation, reinforcing the safety and potential of these compounds as phototherapeutic agents. Although cellular uptake via the sodium-dependent multivitamin transporter was not established, and diffusion is expected to be the predominant mechanism, the high predicted avidin-binding affinity of these dyes opens exciting new avenues for photodynamic therapy-combined strategies.
Collapse
Affiliation(s)
- Eurico Lima
- CQ-VR - Chemistry Centre of Vila Real, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5001-801, Vila Real, Portugal; RISE-Health, Faculty of Health Sciences, University of Beira Interior, Avenida Infante D. Henrique, 6201-506, Covilhã, Portugal.
| | - Octávio Ferreira
- RISE-Health, Faculty of Health Sciences, University of Beira Interior, Avenida Infante D. Henrique, 6201-506, Covilhã, Portugal
| | - Renato E Boto
- RISE-Health, Faculty of Health Sciences, University of Beira Interior, Avenida Infante D. Henrique, 6201-506, Covilhã, Portugal
| | - José R Fernandes
- CQ-VR - Chemistry Centre of Vila Real, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5001-801, Vila Real, Portugal
| | - Paulo Almeida
- RISE-Health, Faculty of Health Sciences, University of Beira Interior, Avenida Infante D. Henrique, 6201-506, Covilhã, Portugal
| | - Samuel M Silvestre
- RISE-Health, Faculty of Health Sciences, University of Beira Interior, Avenida Infante D. Henrique, 6201-506, Covilhã, Portugal; RISE-Health, Faculty of Sciences, University of Beira Interior, Rua Marquês d'Ávila e Bolama, 6201-001, Covilhã, Portugal.
| | - Adriana O Santos
- RISE-Health, Faculty of Health Sciences, University of Beira Interior, Avenida Infante D. Henrique, 6201-506, Covilhã, Portugal.
| | - Lucinda V Reis
- CQ-VR - Chemistry Centre of Vila Real, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5001-801, Vila Real, Portugal.
| |
Collapse
|
2
|
Gu D, Liang X, Shao Z, He H, Zhu L, Qiu S, Liu Z, Wang S, Chen H, Ci S, Wu H, Wang Y. A cascaded amplification carrier-free nanoplatform for synergistic photothermal/ferroptosis therapy via dual antioxidant pathway disruption in cervical cancer. J Mater Chem B 2025; 13:6128-6143. [PMID: 40337781 DOI: 10.1039/d5tb00627a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Cellular defense mechanisms against ferroptosis are primarily mediated by antiferroptotic regulators, particularly glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1). Notably, singlet oxygen (1O2) generated through photoactivation of organic small-molecule photosensitizers (PSs) has been demonstrated to deplete both glutathione (GSH) and nicotinamide adenine dinucleotide phosphate (NADPH). This dual depletion mechanism effectively disrupts the GSH/GPX4 redox axis and the NADPH/FSP1/ubiquinone (CoQ) antioxidant system, thereby potentiating ferroptosis. In this study, we engineered a tumor-targeting amphiphilic iridium-based photosensitizer nanoplatform (Ir-TCF3P-FA NPs) for synergistic photothermal-ferroptosis therapy. Specifically, GSH depletion and NADPH oxidation by 1O2 produced via Ir-TCF3P-FA NPs at 450 nm can suppress the expression of GPX4 and FSP1, amplifying ferroptosis. Additionally, TCF3P exhibited high photothermal conversion efficiency at 808 nm, which not only can enhance photothermal therapy (PTT) efficacy but also facilitated 1O2 generation. The Ir-TCF3P-FA NPs enable effective tumor-targeted delivery and fluorescence/photoacoustic imaging for in vivo distribution tracking. In vivo studies revealed that dual-laser irradiation of Ir-TCF3P-FA NPs provided potent therapeutic efficacy, significantly inhibiting human cervical cancer progression in murine models. This cascaded amplification carrier-free nanoplatform holds promise for clinical multimodal treatment of cervical cancer.
Collapse
Affiliation(s)
- Dihai Gu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| | - Xiao Liang
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Zitong Shao
- The First Clinical Medical College, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Haonan He
- The First Clinical Medical College, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Lin Zhu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Shali Qiu
- Department of Pathology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Zhen Liu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Senlin Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| | - Haijiao Chen
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Shusheng Ci
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Hongshuai Wu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Yihong Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| |
Collapse
|
3
|
Huang HY, Xue RY, Xiao SX, Huang LT, Liao XW, Wang JT, Duan XM, Yu RJ, Xiong YS. AIE-based ruthenium complexes as photosensitizers for specifically photo-inactivate gram-positive bacteria. J Inorg Biochem 2025; 262:112755. [PMID: 39388808 DOI: 10.1016/j.jinorgbio.2024.112755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
The emergence of multidrug-resistant bacterial have caused severe burden for public health. Particularly, Staphylococcus aureus as one of ESKAPE pathogens have induced various infectious diseases and resulted in increasing deaths. Developing new antibacterial agents is still urgent and challenging. Fortunately, in this study, based on aggregation-induced emission (AIE) ruthenium complexes were designed and synthesized, which realized the high efficiency of reactive oxygen species generation and remarkably killed S. aureus unlike conventional antibiotics action. Significantly, owing to good singlet oxygen production ability, Ru1 at only 4 μg/mL of concentration displayed good antibacterial photodynamic therapy effect upon white light irradiation and could deplete essential coenzyme NADH to disrupt intracellular redox balance. Also, the electrostatic interaction between Ru1 and bacteria enhanced the possibility of antibacterial. Under light irradiation, Ru1 could efficiently inhibit the biofilm growth and avoid the development of drug-resistant. Furthermore, Ru1 possessed excellent biocompatibility and displayed remarkable therapy effect in treating mice-wound infections in vivo. These findings indicated that AIE-based ruthenium complexes as new antibacterial agent had great potential in photodynamic therapy of bacteria and addressing the drug-resistance crisis.
Collapse
Affiliation(s)
- Hai-Yan Huang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Run-Yu Xue
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Su-Xin Xiao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Li-Ting Huang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Xiang-Wen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Jin-Tao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Xue-Min Duan
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Ru-Jian Yu
- School of life science, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Yan-Shi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China.
| |
Collapse
|
4
|
Monisha S, Myithili T, Ajithkumar S, Sudharsan K, Keerthana T, Sarikalakshmi B, Pandi M, Kalimuthu P. Aggregation assisted enhancement of singlet oxygen generation by 4-ethynylphenyl substituted porphyrin photosensitizer for photodynamic therapy. Photochem Photobiol 2024. [PMID: 39706810 DOI: 10.1111/php.14049] [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: 08/17/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024]
Abstract
Modulating the photophysical properties of photosensitizers is an effective approach to enhance singlet oxygen generation for photodynamic therapy. Porphyrins are the most widely used photosensitizers due to their biocompatible nature. Aggregation-induced emission (AIE) characteristics of photosensitizers are one of the advantageous features that will enhance fluorescence, intersystem crossing, and efficient triplet state generation. Herein, we demonstrate two glycosylated porphyrin photosensitizers, ZnGEPOH (with two ethynyl groups) and ZnGPOH (without two ethynyl groups), which exhibit AIE. Detailed studies revealed that ZnGEPOH exhibited a two-fold increase in singlet oxygen production than ZnGPOH due to AIE. The photo-cytotoxicity of ZnGPOH and ZnGEPOH were evaluated using cancer cell lines A549 and AGS. ZnGEPOH shows superior photo-cytotoxicity with cell viability of 21% and 19% for A549 and AGS, respectively, at 250 μg/mL concentration in 48 h. Moreover, ZnGEPOH exhibits minimal photo-cytotoxicity towards the control cell line HEK 293.
Collapse
Affiliation(s)
- Sekar Monisha
- Department of Chemistry, The Gandhigram Rural Institute-Deemed to be University, Dindigul, Tamilnadu, India
| | - Thangavel Myithili
- School of Biotechnology, Madurai Kamaraj University, Madurai, Tamilnadu, India
| | - Samuthirakani Ajithkumar
- Department of Chemistry, The Gandhigram Rural Institute-Deemed to be University, Dindigul, Tamilnadu, India
| | - Kumaresan Sudharsan
- Department of Chemistry, The Gandhigram Rural Institute-Deemed to be University, Dindigul, Tamilnadu, India
| | - Thangaraj Keerthana
- Department of Chemistry, The Gandhigram Rural Institute-Deemed to be University, Dindigul, Tamilnadu, India
| | - Baskaran Sarikalakshmi
- Department of Chemistry, The Gandhigram Rural Institute-Deemed to be University, Dindigul, Tamilnadu, India
| | - Mohan Pandi
- School of Biotechnology, Madurai Kamaraj University, Madurai, Tamilnadu, India
| | - Palanisamy Kalimuthu
- Department of Chemistry, The Gandhigram Rural Institute-Deemed to be University, Dindigul, Tamilnadu, India
| |
Collapse
|
5
|
Zheng H, Wang K, Ji D, Liu X, Wang C, Jiang Y, Jia Z, Xiong B, Ling Y, Miao J. Novel tris-bipyridine based Ru(II) complexes as type-I/-II photosensitizers for antitumor photodynamic therapy through ferroptosis and immunogenic cell death. Eur J Med Chem 2024; 279:116909. [PMID: 39357314 DOI: 10.1016/j.ejmech.2024.116909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Ru(II) complexes have attracted attention as photosensitizers for their promising photodynamic properties. Herein, novel tris-bipyridine based Ru(II) complexes (6a-e) were synthesized by introducing saturated heterocycles to improve photodynamic properties and lipid-water partition coefficients. Among them, 6d demonstrated significant phototoxicity towards three cancer cells, with IC50 values of 5.66-7.17 μM, exceeding values in dark (IC50s > 100 μM). Under hypoxic conditions, 6d maintained excellent photodynamic activity in A549 cells, with PI values exceeding 24, highlighting its potential for highly effective type-I/-II photodynamic therapy by inducing ROS generation, oxidative stress, and mitochondrial damage. Additionally, it induced ferroptosis and immunogenic cell death of A549 cells by regulating the expression of relevant markers. Finally, 6d remarkably inhibited the growth of A549 transplanted tumor growth by 95.4 %. This Ru(II) complex shows great potential for cancer treatment with its potent photodynamic activity and diverse mechanisms of tumor cell death.
Collapse
Affiliation(s)
- Hongwei Zheng
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Kai Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Dongliang Ji
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Xiao Liu
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Chen Wang
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Yangyang Jiang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Zihan Jia
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Biao Xiong
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China.
| | - Yong Ling
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China.
| | - Jiefei Miao
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China.
| |
Collapse
|
6
|
Lee LC, Lo KK. Leveraging the Photofunctions of Transition Metal Complexes for the Design of Innovative Phototherapeutics. SMALL METHODS 2024; 8:e2400563. [PMID: 39319499 PMCID: PMC11579581 DOI: 10.1002/smtd.202400563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/03/2024] [Indexed: 09/26/2024]
Abstract
Despite the advent of various medical interventions for cancer treatment, the disease continues to pose a formidable global health challenge, necessitating the development of new therapeutic approaches for more effective treatment outcomes. Photodynamic therapy (PDT), which utilizes light to activate a photosensitizer to produce cytotoxic reactive oxygen species (ROS) for eradicating cancer cells, has emerged as a promising approach for cancer treatment due to its high spatiotemporal precision and minimal invasiveness. However, the widespread clinical use of PDT faces several challenges, including the inefficient production of ROS in the hypoxic tumor microenvironment, the limited penetration depth of light in biological tissues, and the inadequate accumulation of photosensitizers at the tumor site. Over the past decade, there has been increasing interest in the utilization of photofunctional transition metal complexes as photosensitizers for PDT applications due to their intriguing photophysical and photochemical properties. This review provides an overview of the current design strategies used in the development of transition metal complexes as innovative phototherapeutics, aiming to address the limitations associated with PDT and achieve more effective treatment outcomes. The current challenges and future perspectives on the clinical translation of transition metal complexes are also discussed.
Collapse
Affiliation(s)
- Lawrence Cho‐Cheung Lee
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| | - Kenneth Kam‐Wing Lo
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
- State Key Laboratory of Terahertz and Millimeter WavesCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| |
Collapse
|
7
|
Moreth D, Stevens-Cullinane L, Rees TW, Müller VVL, Pasquier A, Song OR, Warchal S, Howell M, Hess J, Schatzschneider U. Antibacterial activity of Au(I), Pt(II), and Ir(III) biotin conjugates prepared by the iClick reaction: influence of the metal coordination sphere on the biological activity. J Biol Inorg Chem 2024; 29:573-582. [PMID: 39198276 PMCID: PMC7616682 DOI: 10.1007/s00775-024-02073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/10/2024] [Indexed: 09/01/2024]
Abstract
A series of biotin-functionalized transition metal complexes was prepared by iClick reaction from the corresponding azido complexes with a novel alkyne-functionalized biotin derivative ([Au(triazolatoR,R')(PPh3)], [Pt(dpb)(triazolatoR,R')], [Pt(triazolatoR,R')(terpy)]PF6, and [Ir(ppy)(triazolatoR,R')(terpy)]PF6 with dpb = 1,3-di(2-pyridyl)benzene, ppy = 2-phenylpyridine, and terpy = 2,2':6',2''-terpyridine and R = C6H5, R' = biotin). The complexes were compared to reference compounds lacking the biotin moiety. The binding affinity toward avidin and streptavidin was evaluated with the HABA assay as well as isothermal titration calorimetry (ITC). All compounds exhibit the same binding stoichiometry of complex-to-avidin of 4:1, but the ITC results show that the octahedral Ir(III) compound exhibits a higher binding affinity than the square-planar Pt(II) complex. The antibacterial activity of the compounds was evaluated on a series of Gram-negative and Gram-positive bacterial strains. In particular, the neutral Au(I) and Pt(II) complexes showed significant antibacterial activity against Staphylococcus aureus and Enterococcus faecium at very low micromolar concentrations. The cytotoxicity against a range of eukaryotic cell lines was studied and revealed that the octahedral Ir(III) complex was non-toxic, while the square-planar Pt(II) and linear Au(I) complexes displayed non-selective micromolar activity.
Collapse
Affiliation(s)
- Dominik Moreth
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074, Würzburg, Germany
| | - Lars Stevens-Cullinane
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London, SE1 1DB, UK
| | - Thomas W Rees
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Victoria V L Müller
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074, Würzburg, Germany
| | - Adrien Pasquier
- High Throughput Screening Science and Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Ok-Ryul Song
- High Throughput Screening Science and Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Scott Warchal
- High Throughput Screening Science and Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Michael Howell
- High Throughput Screening Science and Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Jeannine Hess
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London, SE1 1DB, UK
| | - Ulrich Schatzschneider
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074, Würzburg, Germany.
| |
Collapse
|