1
|
Huang T, Huang W, Huang G, Wei X, Huang Y, Liu T, Liu Y, Ni W, Che C. Pincer-Type Pt(II)-NHC Antibody-Drug Conjugate for HER-2-Targeted Chemoimmunotherapy. Adv Healthc Mater 2025; 14:e2403449. [PMID: 39950551 PMCID: PMC11973945 DOI: 10.1002/adhm.202403449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/28/2025] [Indexed: 04/08/2025]
Abstract
Platinum-based chemotherapy drugs play an indispensable role in clinical cancer treatment, but exhibit considerable side effects due to their non-specific mechanism of killing cancer cells and normal cells. In this regard, the use of antibodies conjugated to anti-cancer platinum complexes will enable better differentiation of cancer cells from normal cells. Here, six pincer-platinum(II) NHC (N-heterocyclic carbene) complexes are reported, one of which has an amino group on the N-alkyl group of the NHC ligand. This platinum(II) complex is used as the payload for platinum(II)-based antibody-drug conjugate (ADC) targeting human epidermal growth factor receptor 2 (HER-2). Notably, this ADC can specifically bind to the HER-2 antigen, distinguish target cells from non-target cells, and exhibit good anti-tumor activity in vitro and in vivo.
Collapse
Affiliation(s)
- Tao Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Wan‐Qiong Huang
- Department of PathologyCancer Hospital of Shantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Gui‐Feng Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Xiao‐Long Wei
- Department of PathologyCancer Hospital of Shantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Yong‐Liang Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Tao Liu
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Yungen Liu
- Department of ChemistrySouthern University of Science and TechnologyShenzhenGuangdong518055P. R. China
| | - Wen‐Xiu Ni
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Chi‐Ming Che
- State Key Laboratory of Synthetic Chemistry and Department of ChemistryThe University of Hong KongHong KongP. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology LimitedHong Kong Science and Technology ParksUnits 1503–1511, 15/F, Building 17W, New TerritoriesHong KongP. R. China
| |
Collapse
|
2
|
Dykman L, Khlebtsov B, Khlebtsov N. Drug delivery using gold nanoparticles. Adv Drug Deliv Rev 2025; 216:115481. [PMID: 39617254 DOI: 10.1016/j.addr.2024.115481] [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: 07/12/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Modern nanotechnologies provide various possibilities for efficiently delivering drugs to biological targets. This review focuses on using functionalized gold nanoparticles (GNPs) as a drug delivery platform. Owing to their exceptional size and surface characteristics, GNPs are a perfect drug delivery vehicle for targeted and selective distribution. Several in vitro and in vivo tests have shown how simple it is to tailor these particles to administer chemical medications straight to tumors. The GNP surface can also be coated with ligands to modify drug release or improve selectivity. Moreover, the pharmacological activity can be enhanced by using the photothermal characteristics of the particles.
Collapse
Affiliation(s)
- Lev Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Boris Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Nikolai Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia; Saratov State University, 83 Ulitsa Astrakhanskaya, Saratov 410012, Russia.
| |
Collapse
|
3
|
Sun X, Zhou X, Shi X, Abed OA, An X, Lei YL, Moon JJ. Strategies for the development of metalloimmunotherapies. Nat Biomed Eng 2024; 8:1073-1091. [PMID: 38914800 PMCID: PMC11410547 DOI: 10.1038/s41551-024-01221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/30/2024] [Indexed: 06/26/2024]
Abstract
Metal ions play crucial roles in the regulation of immune pathways. In fact, metallodrugs have a long record of accomplishment as effective treatments for a wide range of diseases. Here we argue that the modulation of interactions of metal ions with molecules and cells involved in the immune system forms the basis of a new class of immunotherapies. By examining how metal ions modulate the innate and adaptive immune systems, as well as host-microbiota interactions, we discuss strategies for the development of such metalloimmunotherapies for the treatment of cancer and other immune-related diseases.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Editas Medicine, Cambridge, MA, USA.
| | - Xingwu Zhou
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoyue Shi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Omar A Abed
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Xinran An
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yu Leo Lei
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Tonon G, Rizzolio F, Visentin F, Scattolin T. Antibody Drug Conjugates for Cancer Therapy: From Metallodrugs to Nature-Inspired Payloads. Int J Mol Sci 2024; 25:8651. [PMID: 39201338 PMCID: PMC11355040 DOI: 10.3390/ijms25168651] [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: 07/24/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
This review highlights significant advancements in antibody-drug conjugates (ADCs) equipped with metal-based and nature-inspired payloads, focusing on synthetic strategies for antibody conjugation. Traditional methods such us maleimide and succinimide conjugation and classical condensation reactions are prevalent for metallodrugs and natural compounds. However, emerging non-conventional strategies such as photoconjugation are gaining traction due to their milder conditions and, in an aspect which minimizes side reactions, selective formation of ADC. The review also summarizes the therapeutic and diagnostic properties of these ADCs, highlighting their enhanced selectivity and reduced side effects in cancer treatment compared to non-conjugated payloads. ADCs combine the specificity of monoclonal antibodies with the cytotoxicity of chemotherapy drugs, offering a targeted approach to the elimination of cancer cells while sparing healthy tissues. This targeted mechanism has demonstrated impressive clinical efficacy in various malignancies. Key future advancements include improved linker technology for enhanced stability and controlled release of cytotoxic agents, incorporation of novel, more potent, cytotoxic agents, and the identification of new cancer-specific antigens through genomic and proteomic technologies. ADCs are also expected to play a crucial role in combination therapies with immune checkpoint inhibitors, CAR-T cells, and small molecule inhibitors, leading to more durable and potentially curative outcomes. Ongoing research and clinical trials are expanding their capabilities, paving the way for more effective, safer, and personalized treatments, positioning ADCs as a cornerstone of modern medicine and offering new hope to patients.
Collapse
Affiliation(s)
- Giovanni Tonon
- Department of Molecular Sciences and Nanosystems, Università Ca’ Foscari Campus Scientifico, Via Torino 155, 30174 Venezia-Mestre, Italy; (G.T.); (F.R.)
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Università Ca’ Foscari Campus Scientifico, Via Torino 155, 30174 Venezia-Mestre, Italy; (G.T.); (F.R.)
- Pathology Unit, Department of Molecular Biology and Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via Franco Gallini 2, 33081 Aviano, Italy
| | - Fabiano Visentin
- Department of Molecular Sciences and Nanosystems, Università Ca’ Foscari Campus Scientifico, Via Torino 155, 30174 Venezia-Mestre, Italy; (G.T.); (F.R.)
| | - Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
5
|
Sadique FL, Subramaiam H, Krishnappa P, Chellappan DK, Ma JH. Recent advances in breast cancer metastasis with special emphasis on metastasis to the brain. Pathol Res Pract 2024; 260:155378. [PMID: 38850880 DOI: 10.1016/j.prp.2024.155378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Understanding the underlying mechanisms of breast cancer metastasis is of vital importance for developing treatment approaches. This review emphasizes contemporary breakthrough studies with special focus on breast cancer brain metastasis. Acquired mutational changes in metastatic lesions are often distinct from the primary tumor, suggesting altered mutagenesis pathways. The concept of micrometastases and heterogeneity within the tumors unravels novel therapeutic targets at genomic and molecular levels through epigenetic and proteomic profiling. Several pre-clinical studies have identified mechanisms involving the immune system, where tumor associated macrophages are key players. Expression of cell proteins like Syndecan1, fatty acid-binding protein 7 and tropomyosin kinase receptor B have been implicated in aiding the transmigration of breast cancer cells to the brain. Changes in the proteomic landscape of the blood-brain-barrier show altered permeability characteristics, supporting entry of cancer cells. Findings from laboratory studies pave the path for the emergence of new biomarkers, especially blood-based miRNA and circulating tumor cell markers for prognostic staging. The constantly evolving therapeutics call for clinical trials backing supportive evidence of efficacies of both novel and existing approaches. The challenge lying ahead is discovering innovative techniques to replace use of human samples and optimize small-scale patient recruitment in trials.
Collapse
Affiliation(s)
- Fairooz Labiba Sadique
- Department of Biomedical Science, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Hemavathy Subramaiam
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia.
| | - Purushotham Krishnappa
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Jin Hao Ma
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
6
|
Casini A, Pöthig A. Metals in Cancer Research: Beyond Platinum Metallodrugs. ACS CENTRAL SCIENCE 2024; 10:242-250. [PMID: 38435529 PMCID: PMC10906246 DOI: 10.1021/acscentsci.3c01340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 03/05/2024]
Abstract
The discovery of the medicinal properties of platinum complexes has fueled the design and synthesis of new anticancer metallodrugs endowed with unique modes of action (MoA). Among the various families of experimental antiproliferative agents, organometallics have emerged as ideal platforms to control the compounds' reactivity and stability in a physiological environment. This is advantageous to efficiently deliver novel prodrug activation strategies, as well as to design metallodrugs acting only via noncovalent interactions with their pharmacological targets. Noteworthy, another justification for the advance of organometallic compounds for therapy stems from their ability to catalyze bioorthogonal reactions in cancer cells. When not yet ideal as drug leads, such compounds can be used as selective chemical tools that benefit from the advantages of catalytic amplification to either label the target of interest (e.g., proteins) or boost the output of biochemical signals. Examples of metallodrugs for the so-called "catalysis in cells" are considered in this Outlook together with other organometallic drug candidates. The selected case studies are discussed in the frame of more general challenges in the field of medicinal inorganic chemistry.
Collapse
Affiliation(s)
- Angela Casini
- Chair
of Medicinal and Bioinorganic Chemistry, Department of Chemistry,
School of Natural Sciences, Technical University
of Munich, Lichtenbergstraße 4, D-85748 Garching b. München, Germany
| | - Alexander Pöthig
- Catalysis
Research Center & Department of Chemistry, School of Natural Sciences, Technical University of Munich, Ernst-Otto-Fischer Str. 1, D-85748 Garching b. München, Germany
| |
Collapse
|
7
|
Xia Q, Liang T, Zhou Y, Liu J, Tang Y, Liu F. Recent Advances in Biomedical Nanotechnology Related to Natural Products. Curr Pharm Biotechnol 2024; 25:944-961. [PMID: 37605408 DOI: 10.2174/1389201024666230821090222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/24/2023] [Accepted: 07/07/2023] [Indexed: 08/23/2023]
Abstract
Natural product processing via nanotechnology has opened the door to innovative and significant applications in medical fields. On one hand, plants-derived bioactive ingredients such as phenols, pentacyclic triterpenes and flavonoids exhibit significant pharmacological activities, on another hand, most of them are hydrophobic in nature, posing challenges to their use. To overcome this issue, nanoencapsulation technology is employed to encapsulate these lipophilic compounds and enhance their bioavailability. In this regard, various nano-sized vehicles, including degradable functional polymer organic compounds, mesoporous silicon or carbon materials, offer superior stability and retention for bioactive ingredients against decomposition and loss during delivery as well as sustained release. On the other hand, some naturally occurring polymers, lipids and even microorganisms, which constitute a significant portion of Earth's biomass, show promising potential for biomedical applications as well. Through nano-processing, these natural products can be developed into nano-delivery systems with desirable characteristics for encapsulation a wide range of bioactive components and therapeutic agents, facilitating in vivo drug transport. Beyond the presentation of the most recent nanoencapsulation and nano-processing advancements with formulations mainly based on natural products, this review emphasizes the importance of their physicochemical properties at the nanoscale and their potential in disease therapy.
Collapse
Affiliation(s)
- Qing Xia
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Tingting Liang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Yue Tang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| |
Collapse
|
8
|
Ahad A, K. Saeed H, del Solar V, López-Hernández JE, Michel A, Mathew J, Lewis JS, Contel M. Shifting the Antibody-Drug Conjugate Paradigm: A Trastuzumab-Gold-Based Conjugate Demonstrates High Efficacy against Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer Mouse Model. ACS Pharmacol Transl Sci 2023; 6:1972-1986. [PMID: 38093840 PMCID: PMC10714425 DOI: 10.1021/acsptsci.3c00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 01/17/2024]
Abstract
Antibody-drug conjugates (ADCs) combine the selectivity of monoclonal antibodies (mAbs) with the efficacy of chemotherapeutics to target cancers without toxicity to normal tissue. Clinically, most chemotherapeutic ADCs are based on complex organic molecules, while the conjugation of metallodrugs to mAbs has been overlooked, despite the resurgent interest in metal-based drugs as cancer chemotherapeutics. In 2019, we described the first gold ADCs containing gold-triphenylphosphane fragments as a proof of concept. The ADCs (based on the antibody trastuzumab) were selective and highly active against HER2-positive breast cancer cells. In this study, we developed site-specific ADCs (Thio-1b and Thio-2b) using the cysteine-engineered trastuzumab derivative THIOMAB antibody technology with gold(I)-containing phosphanes and a maleimide-based linker amenable to bioconjugation (1b and 2b). In addition, we developed lysine-directed ADCs with gold payloads based on phosphanes and N-heterocyclic carbenes featuring an activated ester moiety (2c and 5c) with trastuzumab (Tras-2c and Tras-5c) and another anti-HER2 antibody, pertuzumab (Per-2c and Per-5c). Both sets of ADCs demonstrated significant anticancer potency in vitro assays. Based on these results, one ADC (Tras-2c), containing the [Au(PEt3)] fragment present in FDA-approved auranofin, was selected for an in vivo antitumor efficacy study. Immunocompromised mice xenografted with the HER2-positive human cancer cell line SKBR-3 exhibited almost complete tumor reduction and low toxicity with intravenous administration of Tras-2c. With this highly selective targeting system, we demonstrated that a subnanomolar cytotoxicity profile in cells is not required for an impressive antitumor effect in a mouse xenograft model.
Collapse
Affiliation(s)
- Afruja Ahad
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Hiwa K. Saeed
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Virginia del Solar
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Javier E. López-Hernández
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| | - Alexa Michel
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Joshua Mathew
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Maria Contel
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Chemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| |
Collapse
|
9
|
Wang Y, Mesdom P, Purkait K, Saubaméa B, Burckel P, Arnoux P, Frochot C, Cariou K, Rossel T, Gasser G. Ru(ii)/Os(ii)-based carbonic anhydrase inhibitors as photodynamic therapy photosensitizers for the treatment of hypoxic tumours. Chem Sci 2023; 14:11749-11760. [PMID: 37920359 PMCID: PMC10619633 DOI: 10.1039/d3sc03932c] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023] Open
Abstract
Photodynamic therapy (PDT) is a medical technique for the treatment of cancer. It is based on the use of non-toxic molecules, called photosensitizers (PSs), that become toxic when irradiated with light and produce reactive oxygen specious (ROS) such as singlet oxygen (1O2). This light-induced toxicity is rather selective since the physician only targets a specific area of the body, leading to minimal side effects. Yet, a strategy to improve further the selectivity of this medical technique is to confine the delivery of the PS to cancer cells only instead of spreading it randomly throughout the body prior to light irradiation. To address this problem, we present here novel sulfonamide-based monopodal and dipodal ruthenium and osmium polypyridyl complexes capable of targeting carbonic anhydrases (CAs) that are a major target in cancer therapy. CAs are overexpressed in the membrane or cytoplasm of various cancer cells. We therefore anticipated that the accumulation of our complexes in or outside the cell prior to irradiation would improve the selectivity of the PDT treatment. We show that our complexes have a high affinity for CAs, accumulate in cancer cells overexpressing CA cells and importantly kill cancer cells under both normoxic and hypoxic conditions upon irradiation at 540 nm. More importantly, Os(ii) compounds still exhibit some phototoxicity under 740 nm irradiation under normoxic conditions. To our knowledge, this is the first description of ruthenium/osmium-based PDT PSs that are CA inhibitors for the selective treatment of cancers.
Collapse
Affiliation(s)
- Youchao Wang
- Chimie ParisTech, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, PSL University 75005 Paris France +33185784151 https://www.gassergroup.com
| | - Pierre Mesdom
- Chimie ParisTech, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, PSL University 75005 Paris France +33185784151 https://www.gassergroup.com
| | - Kallol Purkait
- Chimie ParisTech, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, PSL University 75005 Paris France +33185784151 https://www.gassergroup.com
| | - Bruno Saubaméa
- Cellular and Molecular Imaging Facility, US25 Inserm, UAR3612 CNRS, Faculté de Pharmacie de Paris, Université Paris Cité F-75006 Paris France
| | - Pierre Burckel
- Institut de Physique du Globe de Paris, Biogéochimie à; l'Anthropocène des Eléments et Contaminants Emergents 75005 Paris France
| | | | | | - Kevin Cariou
- Chimie ParisTech, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, PSL University 75005 Paris France +33185784151 https://www.gassergroup.com
| | - Thibaud Rossel
- Institute of Chemistry, University of Neuchâtel Avenue de Bellevaux 51 2000 Neuchâtel Switzerland
| | - Gilles Gasser
- Chimie ParisTech, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, PSL University 75005 Paris France +33185784151 https://www.gassergroup.com
| |
Collapse
|
10
|
López-Hernández JE, Nayeem N, Cerón-Carrasco JP, Ahad A, Hafeez A, León IE, Contel M. Platinum(IV)-Gold(I) Agents with Promising Anticancer Activity: Selected Studies in 2D and 3D Triple-Negative Breast Cancer Models. Chemistry 2023; 29:e202302045. [PMID: 37507346 PMCID: PMC10615877 DOI: 10.1002/chem.202302045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 07/30/2023]
Abstract
New heterometallic binuclear and trinuclear platinum(IV)-gold(I) compounds of the type [Pt(L)n Cl2 (OH){(OOC-4-C6 H4 -PPh2 )AuCl}x ] (L=NH3 , n=2; x=1, 2; L=diaminocyclohexane, DACH, n=1; x=2) are described. These compounds are cytotoxic and selective against a small panel of renal, bladder, ovarian, and breast cancer cell lines. We selected a trinuclear PtAu2 compound containing the PtIV core based on oxaliplatin, to further investigate its cell-death pathway, cell and organelle uptake and anticancer effects against the triple-negative breast cancer (TNBC) MDA-MB-231 cell line. This compound induces apoptosis and accumulates mainly in the nucleus and mitochondria. It also exerts remarkable antimigratory and antiangiogenic properties, and has a potent cytotoxic effect against TNBC 3D spheroids. Trinuclear compounds do not seem to display relevant interactions with calf thymus (CT) DNA and plasmid (pBR322) even in the presence of reducing agents, but inhibit pro-angiogenic enzyme thioredoxin reductase (TrxR) in TNBC cells.
Collapse
Affiliation(s)
- Javier E López-Hernández
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - Nazia Nayeem
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - José P Cerón-Carrasco
- Centro Universitario de la Defensa, Universidad Politécnica de Cartagena, C/Coronel López Peña s/n, Base Aérea de San Javier, Santiago de la Ribera, 30720, Murcia, Spain
| | - Afruja Ahad
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
- Radiology, Molecular Pharmacology Program, and, Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, NY 11065, USA
| | - Aiman Hafeez
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Ignacio E León
- Centro de Química Inorgánica, CEQUINOR (CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N°1465, La Plata, 1900, Argentina
| | - Maria Contel
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| |
Collapse
|
11
|
Yang R, Chen L, Wang Y, Zhang L, Zheng X, Yang Y, Zhu Y. Tumor microenvironment responsive metal nanoparticles in cancer immunotherapy. Front Immunol 2023; 14:1237361. [PMID: 37575228 PMCID: PMC10413122 DOI: 10.3389/fimmu.2023.1237361] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Malignant tumors have a unique tumor microenvironment (TME), which includes mild acidity, hypoxia, overexpressed reactive oxygen species (ROS), and high glutathione (GSH) levels, among others. Recently, TME regulation approaches have attracted widespread attention in cancer immunotherapy. Nanoparticles as drug delivery systems have ability to modulate the hydrophilicity of drugs to affect drug uptake and efflux in tumor. Especially, the metal nanoparticles have been extensive applied for tumor immunotherapy due to their unique physical properties and elaborate design. However, the potential deficiencies of metal nanoparticles due to their low biodegradability, toxicity and treatment side effects restrict their clinical application. In this review, we briefly introduce the feature characteristics of the TME and the recent advances in tumor microenvironment responsive metal nanoparticles for tumor immunotherapy. In addition, nanoparticles could be combined with other treatments, such as chemotherapy, radiotherapy and photodynamic therapy also is presented. Finally, the challenges and outlook for improving the antitumor immunotherapy efficiency, side effect and potential risks of metal nanoparticles has been discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Yong Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuxuan Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
12
|
Marciano Y, del Solar V, Nayeem N, Dave D, Son J, Contel M, Ulijn RV. Encapsulation of Gold-Based Anticancer Agents in Protease-Degradable Peptide Nanofilaments Enhances Their Potency. J Am Chem Soc 2023; 145:234-246. [PMID: 36542079 PMCID: PMC10720394 DOI: 10.1021/jacs.2c09820] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We investigated the use of amphiphilic, protease-cleavable peptides as encapsulation moieties for hydrophobic metallodrugs, in order to enhance their bioavailability and consequent activity. Two hydrophobic, gold-containing anticancer agents varying in aromatic ligand distribution (Au(I)-N-heterocyclic carbene compounds 1 and 2) were investigated. These were encapsulated into amphiphilic decapeptides that form soluble filamentous structures with hydrophobic cores, varying supramolecular packing arrangements and surface charge. Peptide sequence strongly dictates the supramolecular packing within the aromatic core, which in turn dictates drug loading. Anionic peptide filaments can effectively load 1, and to a lesser extent 2, while their cationic counterparts could not, collectively demonstrating that loading efficiency is dictated by both aromatic and electrostatic (mis)matching between drug and peptide. Peptide nanofilaments were nontoxic to cancerous and noncancerous cells. By contrast, those loaded with 1 and 2 displayed enhanced cytotoxicity in comparison to 1 and 2 alone, when exposed to Caki-1 and MDA-MB-231 cancerous cell lines, while no cytotoxicity was observed in noncancerous lung fibroblasts, IMR-90. We propose that the enhanced in vitro activity results from the enhanced proteolytic activity in the vicinity of the cancer cells, thereby breaking the filaments into drug-bound peptide fragments that are taken up by these cells, resulting in enhanced cytotoxicity toward cancer cells.
Collapse
Affiliation(s)
- Yaron Marciano
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Virginia del Solar
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
| | - Nazia Nayeem
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program inBiology, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Dhwanit Dave
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Chemistry, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| | - Jiye Son
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
| | - María Contel
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Ph.D. Program in Biochemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Ph.D. Program inBiology, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Rein V. Ulijn
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Ph.D. Program in Biochemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Chemistry, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| |
Collapse
|
13
|
Gascón E, Otal I, Maisanaba S, Llana-Ruiz-Cabello M, Valero E, Repetto G, Jones PG, Oriol L, Jiménez J. Gold(I) metallocyclophosphazenes with antibacterial potency and antitumor efficacy. Synergistic antibacterial action of a heterometallic gold and silver-cyclophosphazene. Dalton Trans 2022; 51:13657-13674. [PMID: 36040292 DOI: 10.1039/d2dt01963a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
One of the most important uses of phosphazenes today involves its biomedical applications. They can also be employed as scaffolds for the design and construction of a variety of ligands in order to coordinate them to metallic drugs. The coordination chemistry of the (amino)cyclotriphosphazene ligand, [N3P3(NHCy)6], towards gold(I) complexes has been studied. Neutral complexes, [N3P3(NHCy)6{AuX}n] (X = Cl or C6F5; n = 1 or 2) (1-4), cationic complexes, [N3P3(NHCy)6{Au(PR3)}n](NO3)n (PR3 = PPh3, PPh2Me, TPA; n = 1, 2 or 3) (6-12) [TPA = 1,3,5-triaza-7-phosphaadamantane] and a heterometallic compound [N3P3(NHCy)6{Au(PPh3)}2{Ag(PPh3)}](NO3)3 (13) have been obtained and characterized by various methods including single-crystal X-ray diffraction for 7, which confirms the coordination of gold atoms to the nitrogens of the phosphazene ring. Compounds 1, 4, 6-13 were screened for in vitro cytotoxic activity against two tumor human cell lines, MCF7 (breast adenocarcinoma) and HepG2 (hepatocellular carcinoma), and for antimicrobial activity against five bacterial species including Gram-positive, Gram-negative, and Mycobacteria. Both the median inhibitory concentration (IC50) and minimum inhibitory concentration (MIC) values are among the lowest found for any gold or silver derivatives against the cell lines and particularly against the Gram-positive (S. aureus) strain and the mycobacteria used in this work. Structure-activity relationships are discussed in order to determine the influence of ancillary ligands and the number and type of metal atoms (silver or gold). Compounds 4 and 8 showed not only maximal potency on human cells but also some tumour selectivity. Remarkably, compound 13, with both gold and silver atoms, showed outstanding activity against both Gram-positive and Gram-negative strains (nanomolar range), thus having a cooperative effect between gold and silver, with MIC values which are similar or lower than those of gentamicine, ciprofloxacin and rifampicine. The broad spectrum antimicrobial efficacy of all these metallophosphazenes and particularly of heterometallic compound 13 could be very useful to obtain materials for surfaces with antimicrobial properties that are increasingly in demand.
Collapse
Affiliation(s)
- Elena Gascón
- Departamento de Química Inorgánica, Facultad de Ciencias, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain.
| | - Isabel Otal
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Universidad de Zaragoza, Zaragoza, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Sara Maisanaba
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - María Llana-Ruiz-Cabello
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - Eva Valero
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área Nutrición y Bromatología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - Guillermo Repetto
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - Peter G Jones
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106, Braunschweig, Germany
| | - Luis Oriol
- Departamento de Química Orgánica, Instituto de Nanociencia y Materiales de Aragón-Facultad de Ciencias, Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Josefina Jiménez
- Departamento de Química Inorgánica, Facultad de Ciencias, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain.
| |
Collapse
|
14
|
In Situ Prodrug Activation by an Affibody‐Ruthenium Catalyst Hybrid for HER2‐Targeted Chemotherapy. Angew Chem Int Ed Engl 2022; 61:e202202855. [DOI: 10.1002/anie.202202855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Indexed: 11/07/2022]
|
15
|
In Situ Prodrug Activation by an Affibody–Ruthenium Catalyst Hybrid for HER2‐Targeted Chemotherapy. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202202855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
16
|
Scattolin T, Valente G, Luzietti L, Piva M, Demitri N, Lampronti I, Gambari R, Visentin F. Synthesis and anticancer activity of Pt(0)‐olefin complexes bearing 1,3,5‐triaza‐7‐phosphaadamantane and
N
‐heterocyclic carbene ligands. Appl Organomet Chem 2021. [DOI: 10.1002/aoc.6438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Thomas Scattolin
- Dipartimento di Scienze Molecolari e Nanosistemi Università Ca' Foscari Venezia‐Mestre Italy
| | - Giorgia Valente
- Dipartimento di Scienze Molecolari e Nanosistemi Università Ca' Foscari Venezia‐Mestre Italy
| | - Lara Luzietti
- Dipartimento di Scienze della Vita e Biotecnologie Università degli Studi di Ferrara Ferrara Italy
| | - Michele Piva
- Dipartimento di Scienze Molecolari e Nanosistemi Università Ca' Foscari Venezia‐Mestre Italy
| | - Nicola Demitri
- S.S. 14 Km 163.5 in Area Science Park Elettra–Sincrotrone Trieste Trieste Italy
| | - Ilaria Lampronti
- Dipartimento di Scienze della Vita e Biotecnologie Università degli Studi di Ferrara Ferrara Italy
| | - Roberto Gambari
- Dipartimento di Scienze della Vita e Biotecnologie Università degli Studi di Ferrara Ferrara Italy
| | - Fabiano Visentin
- Dipartimento di Scienze Molecolari e Nanosistemi Università Ca' Foscari Venezia‐Mestre Italy
| |
Collapse
|
17
|
Matlou GG, Abrahamse H. Hybrid Inorganic-Organic Core-Shell Nanodrug Systems in Targeted Photodynamic Therapy of Cancer. Pharmaceutics 2021; 13:1773. [PMID: 34834188 PMCID: PMC8625656 DOI: 10.3390/pharmaceutics13111773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Hybrid inorganic-organic core-shell nanoparticles (CSNPs) are an emerging paradigm of nanodrug carriers in the targeted photodynamic therapy (TPDT) of cancer. Typically, metallic cores and organic polymer shells are used due to their submicron sizes and high surface to volume ratio of the metallic nanoparticles (NPs), combined with enhances solubility, stability, and absorption sites of the organic polymer shell. As such, the high loading capacity of therapeutic agents such as cancer specific ligands and photosensitizer (PS) agents is achieved with desired colloidal stability, drug circulation, and subcellular localization of the PS agents at the cancer site. This review highlights the synthesis methods, characterization techniques, and applications of hybrid inorganic-organic CSNPs as loading platforms of therapeutic agents for use in TPDT. In addition, cell death pathways and the mechanisms of action that hybrid inorganic-organic core-shell nanodrug systems follow in TPDT are also reviewed. Nanodrug systems with cancer specific properties are able to localize within the solid tumor through the enhanced permeability effect (EPR) and bind with affinity to receptors on the cancer cell surfaces, thus improving the efficacy of short-lived cytotoxic singlet oxygen. This ability by nanodrug systems together with their mechanism of action during cell death forms the core basis of this review and will be discussed with an overview of successful strategies that have been reported in the literature.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein 2028, South Africa;
| |
Collapse
|
18
|
Fotopoulou E, Titilas I, Ronconi L. Metallodrugs as Anticancer Chemotherapeutics and Diagnostic Agents: A Critical Patent Review (2010-2020). Recent Pat Anticancer Drug Discov 2021; 17:42-54. [PMID: 34493191 DOI: 10.2174/1574892816666210907101146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/07/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The development of metallodrugs with potential applications in cancer treatment and diagnosis has been a hot topic since the approval and subsequent marketing of the anticancer drug cisplatin in 1978. Since then, thousands of metal-based derivatives have been reported and evaluated for their chemotherapeutic or tumor imaging properties, but only a very limited number gained clinical status. Nonetheless, research in the field has been increasing exponentially over the years, especially in a view to exploiting novel drug designing approaches and strategies aimed at improving pharmacological outcomes and, at the same time, reducing side-effects. OBJECTIVE This review article reports on the patents filed during the last decade and strictly focusing on the development of metal-based anticancer and diagnostic agents. The goal is to identify the latest trends and designing strategies in the field, which would represent a valuable starting point to researchers interested in the development of metallodrugs. METHODS The most relevant patents filed in the 2010-2020 timeframe have been retrieved from various databases using dedicated search engines (such as SciFinder, Google Patents, PatentPak, Espacenet, Global Dossier, PatentScope), sorted by type of metallodrug and screened to include those reporting a substantial amount of biological data. RESULTS The majority of patents here reviewed are concerned with metallodrugs (mostly platinum-based) showing interesting pharmacological properties but no specific tumor-targeting features. Nonetheless, some promising trends in the development of novel drug delivery strategies and/or metallodrugs with potential applications in targeted chemotherapy are envisaged. CONCLUSION In this review, the latest trends in the development of metallodrugs from recent patents are summarized and critically discussed. Such trends would be of interest not only to the scientific community but also to lay audiences aiming to broaden their knowledge of the field and industrial stakeholders potentially interested in the exploitation and commercialization of this class of pharmaceuticals.
Collapse
Affiliation(s)
- Eirini Fotopoulou
- School of Chemistry, College of Science and Engineering, National University of Ireland Galway, University Road, Galway H91 TK33. Ireland
| | - Ioannis Titilas
- School of Chemistry, College of Science and Engineering, National University of Ireland Galway, University Road, Galway H91 TK33. Ireland
| | - Luca Ronconi
- School of Chemistry, College of Science and Engineering, National University of Ireland Galway, University Road, Galway H91 TK33. Ireland
| |
Collapse
|
19
|
Mo J, Mai Le NP, Priefer R. Evaluating the mechanisms of action and subcellular localization of ruthenium(II)-based photosensitizers. Eur J Med Chem 2021; 225:113770. [PMID: 34403979 DOI: 10.1016/j.ejmech.2021.113770] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/23/2021] [Accepted: 08/09/2021] [Indexed: 01/16/2023]
Abstract
The identification of ruthenium(II) polypyridyl complexes as photosensitizers in photodynamic therapy (PDT) for the treatment of cancer is progressing rapidly. Due to their favorable photophysical and photochemical properties, Ru(II)-based photosensitizers have absorption in the visible spectrum, can be irradiated via one- and two-photon excitation within the PDT window, and yield potent oxygen-dependent and/or oxygen-independent photobiological activities. Herein, we present a current overview of the mechanisms of action and subcellular localization of Ru(II)-based photosensitizers in the treatment of cancer. These photosensitizers are highlighted from a medicinal chemistry and chemical biology perspective. However, although this field is burgeoning, challenges and limitations remain in the photosensitization strategies and clinical translation.
Collapse
Affiliation(s)
- Jiancheng Mo
- Massachusetts College of Pharmacy and Health Sciences University, Boston, MA, USA
| | - Ngoc Phuong Mai Le
- Massachusetts College of Pharmacy and Health Sciences University, Boston, MA, USA
| | - Ronny Priefer
- Massachusetts College of Pharmacy and Health Sciences University, Boston, MA, USA.
| |
Collapse
|
20
|
Pohanka M. Current Biomedical and Diagnostic Applications of Gold Micro and Nanoparticles. Mini Rev Med Chem 2021; 21:1085-1095. [PMID: 32744971 DOI: 10.2174/1389557520666200730155616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 11/22/2022]
Abstract
Production of particles and their adaptation in the pharmacology became an object of interest, and they are the currently introduced therapies based on the use of micro and nanoparticles. The use of gold particles is not an exception. This review has focused on the application of gold micro and nanoparticles in pharmacology and biomedicine. The particles can be used for diagnosis respective theranostic of cancer, rheumatoid arthritis and as antimicrobial means. Besides these applications, specifications of gold, gold particles, and colloidal gold manufacturing and their comparison with the solid gold, are described as well. This review is based on a survey of actual scientific literature.
Collapse
Affiliation(s)
- Miroslav Pohanka
- Faculty of Military Health Sciences, University of Defense, Trebesska 1575, Hradec Kralove CZ-50001, Czech Republic
| |
Collapse
|
21
|
|
22
|
Lengacher R, Braband H, Csucker J, Alberto R. Convenient Cyclopentadiene Modifications for Building Versatile (Radio‐)Metal Cyclopentadienyl Frameworks. Eur J Inorg Chem 2021. [DOI: 10.1002/ejic.202100163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Raphael Lengacher
- Department of Chemistry University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Henrik Braband
- Department of Chemistry University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Joshua Csucker
- Department of Chemistry University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Roger Alberto
- Department of Chemistry University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| |
Collapse
|
23
|
Merkul E, Muns JA, Sijbrandi NJ, Houthoff H, Nijmeijer B, Rheenen G, Reedijk J, Dongen GAMS. An Efficient Conjugation Approach for Coupling Drugs to Native Antibodies via the Pt
II
Linker
Lx
for Improved Manufacturability of Antibody–Drug Conjugates. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202011593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Eugen Merkul
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Joey A. Muns
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Niels J. Sijbrandi
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Hendrik‐Jan Houthoff
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Bart Nijmeijer
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Gerro Rheenen
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Jan Reedijk
- Leiden Institute of Chemistry Leiden University PO Box 9502 2300 RA Leiden The Netherlands
| | - Guus A. M. S. Dongen
- Department of Radiology and Nuclear Medicine Amsterdam UMC, location VU medical center Amsterdam The Netherlands
| |
Collapse
|
24
|
Merkul E, Muns JA, Sijbrandi NJ, Houthoff H, Nijmeijer B, van Rheenen G, Reedijk J, van Dongen GAMS. An Efficient Conjugation Approach for Coupling Drugs to Native Antibodies via the Pt II Linker Lx for Improved Manufacturability of Antibody-Drug Conjugates. Angew Chem Int Ed Engl 2021; 60:3008-3015. [PMID: 33185916 PMCID: PMC7986738 DOI: 10.1002/anie.202011593] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/29/2020] [Indexed: 12/20/2022]
Abstract
The PtII linker [ethylenediamineplatinum(II)]2+ , coined Lx, has emerged as a novel non-conventional approach to antibody-drug conjugates (ADCs) and has shown its potential in preclinical in vitro and in vivo benchmark studies. A crucial improvement of the Lx conjugation reaction from initially <15 % to ca. 75-90 % conjugation efficiency is described, resulting from a systematic screening of all relevant reaction parameters. NaI, a strikingly simple inorganic salt additive, greatly improves the conjugation efficiency as well as the conjugation selectivity simply by exchanging the leaving chloride ligand on Cl-Lx-drug complexes (which are direct precursors for Lx-ADCs) for iodide, thus generating I-Lx-drug complexes as more reactive species. Using this iodide effect, we developed a general and highly practical conjugation procedure that is scalable: our lead Lx-ADC was produced on a 5 g scale with an outstanding conjugation efficiency of 89 %.
Collapse
Affiliation(s)
- Eugen Merkul
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Joey A. Muns
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Niels J. Sijbrandi
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | | | - Bart Nijmeijer
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Gerro van Rheenen
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Jan Reedijk
- Leiden Institute of ChemistryLeiden UniversityPO Box 95022300RALeidenThe Netherlands
| | - Guus A. M. S. van Dongen
- Department of Radiology and Nuclear MedicineAmsterdam UMC, location VU medical centerAmsterdamThe Netherlands
| |
Collapse
|
25
|
Encapsidation of Different Plasmonic Gold Nanoparticles by the CCMV CP. Molecules 2020; 25:molecules25112628. [PMID: 32516956 PMCID: PMC7321416 DOI: 10.3390/molecules25112628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/26/2022] Open
Abstract
Different types of gold nanoparticles have been synthesized that show great potential in medical applications such as medical imaging, bio-analytical sensing and photothermal cancer therapy. However, their stability, polydispersity and biocompatibility are major issues of concern. For example, the synthesis of gold nanorods, obtained through the elongated micelle process, produce them with a high positive surface charge that is cytotoxic, while gold nanoshells are unstable and break down in a few weeks due to the Ostwald ripening process. In this work, we report the self-assembly of the capsid protein (CP) of cowpea chlorotic mottle virus (CCMV) around spherical gold nanoparticles, gold nanorods and gold nanoshells to form virus-like particles (VLPs). All gold nanoparticles were synthesized or treated to give them a negative surface charge, so they can interact with the positive N-terminus of the CP leading to the formation of the VLPs. To induce the protein self-assembly around the negative gold nanoparticles, we use different pH and ionic strength conditions determined from a CP phase diagram. The encapsidation with the viral CP will provide the nanoparticles better biocompatibility, stability, monodispersity and a new biological substrate on which can be introduced ligands toward specific cells, broadening the possibilities for medical applications.
Collapse
|
26
|
Kuchur OA, Tsymbal SA, Shestovskaya MV, Serov NS, Dukhinova MS, Shtil AA. Metal-derived nanoparticles in tumor theranostics: Potential and limitations. J Inorg Biochem 2020; 209:111117. [PMID: 32473483 DOI: 10.1016/j.jinorgbio.2020.111117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/19/2022]
Abstract
Initially, metal derived nanoparticles have been used exclusively as contrasting agents in magnetic resonance imaging. Today, green routes of chemical synthesis together with numerous modifications of the core and surface gave rise to a plethora of biomedical applications of metal derived nanoparticles including tumor imaging, diagnostics, and therapy. These materials are an emerging class of tools for tumor theranostics. Nevertheless, the spectrum of clinically approved metal nanoparticles remains narrow, as the safety, specificity and efficiency still have to be improved. In this review we summarize the major directions for development and biomedical applications of metal based nanoparticles and analyze their effects on tumor cells and microenvironment. We discuss the advantages and possible limitations of metal nanoparticle-based tumor theranostics, as well as the potential strategies to improve the in vivo performance of these unique materials.
Collapse
Affiliation(s)
- O A Kuchur
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - S A Tsymbal
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - M V Shestovskaya
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - N S Serov
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - M S Dukhinova
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia.
| | - A A Shtil
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia; Institute of Gene Biology, Russian Academy of Science, 119334 Moscow, Russia
| |
Collapse
|
27
|
Meier-Menches SM, Casini A. Design Strategies and Medicinal Applications of Metal-Peptidic Bioconjugates. Bioconjug Chem 2020; 31:1279-1288. [DOI: 10.1021/acs.bioconjchem.0c00152] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Samuel M. Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, 1090 Vienna, Austria
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Lichtenbergstrasse 4, 85748 Garching, Germany
| |
Collapse
|
28
|
Karges J, Jakubaszek M, Mari C, Zarschler K, Goud B, Stephan H, Gasser G. Synthesis and Characterization of an Epidermal Growth Factor Receptor-Selective Ru II Polypyridyl-Nanobody Conjugate as a Photosensitizer for Photodynamic Therapy. Chembiochem 2020; 21:531-542. [PMID: 31339225 PMCID: PMC7065149 DOI: 10.1002/cbic.201900419] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Indexed: 02/06/2023]
Abstract
There is a current surge of interest in the development of novel photosensitizers (PSs) for photodynamic therapy (PDT), as those currently approved are not completely ideal. Among the tested compounds, we have previously investigated the use of RuII polypyridyl complexes with a [Ru(bipy)2 (dppz)]2+ and [Ru(phen)2 (dppz)]2+ scaffold (bipy=2,2'-bipyridine; dppz=dipyrido[3,2-a:2',3'-c]phenazine; phen=1,10-phenanthroline). These complexes selectively target DNA. However, because DNA is ubiquitous, it would be of great interest to increase the selectivity of our PDT PSs by linking them to a targeting vector in view of targeted PDT. Herein, we present the synthesis, characterization, and in-depth photophysical evaluation of a nanobody-containing RuII polypyridyl conjugate selective for the epidermal growth factor receptor (EGFR) in view of targeted PDT. Using ICP-MS and confocal microscopy, we could demonstrate that our conjugate has high selectivity for the EGFR receptor, which is a crucial oncological target because it is overexpressed and/or deregulated in a variety of solid tumors. However, in contrast to expectations, this conjugate was found to not produce reactive oxygen species (ROS) in cancer cells and is therefore not phototoxic.
Collapse
Affiliation(s)
- Johannes Karges
- Chimie ParisTechPSL UniversityCNRSInstitute of Chemistry for Life and Health SciencesLaboratory for Inorganic Chemical Biology75005ParisFrance
| | - Marta Jakubaszek
- Chimie ParisTechPSL UniversityCNRSInstitute of Chemistry for Life and Health SciencesLaboratory for Inorganic Chemical Biology75005ParisFrance
- Institut CuriePSL UniversityCNRS UMR 14426 rue d'Ulm75005ParisFrance
| | - Cristina Mari
- Department of ChemistryUniversity of ZürichWinterthurerstrasse 1908057ZürichSwitzerland
| | - Kristof Zarschler
- Helmholtz-Zentrum Dresden–RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Bruno Goud
- Institut CuriePSL UniversityCNRS UMR 14426 rue d'Ulm75005ParisFrance
| | - Holger Stephan
- Helmholtz-Zentrum Dresden–RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Gilles Gasser
- Chimie ParisTechPSL UniversityCNRSInstitute of Chemistry for Life and Health SciencesLaboratory for Inorganic Chemical Biology75005ParisFrance
| |
Collapse
|
29
|
Gascón E, Maisanaba S, Otal I, Valero E, Repetto G, Jones PG, Jiménez J. (Amino)cyclophosphazenes as Multisite Ligands for the Synthesis of Antitumoral and Antibacterial Silver(I) Complexes. Inorg Chem 2020; 59:2464-2483. [PMID: 31984738 DOI: 10.1021/acs.inorgchem.9b03334] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The reactivity of the multisite (amino)cyclotriphosphazene ligands, [N3P3(NHCy)6] and [N3P3(NHCy)3(NMe2)3], has been explored in order to obtain silver(I) metallophosphazene complexes. Two series of cationic silver(I) metallophosphazenes were obtained and characterized: [N3P3(NHCy)6{AgL}n](TfO)n [n = 2, L = PPh3 (2), PPh2Me (4); n = 3, L = PPh3 (3), PPh2Me (5), TPA (TPA = 1,3,5-triaza-7-phosphaadamantane, 6)] and nongem-trans-[N3P3(NHCy)3(NMe2)3{AgL}n](TfO)n [n = 2, L = PPh3 (7), PPh2Me (9); n = 3, L = PPh3 (8), PPh2Me (10)]. 5, 7, and 9 have also been characterized by single-crystal X-ray diffraction, thereby allowing key bonding information to be obtained. Compounds 2-6, 9, and 10 were screened for in vitro cytotoxic activity against two tumor human cell lines, MCF7 (breast adenocarcinoma) and HepG2 (hepatocellular carcinoma), and for antimicrobial activity against five bacterial species including Gram-positive, Gram-negative, and Mycobacteria strains. Both the IC50 and MIC values revealed excellent biological activity for these metal complexes, compared with their precursors and cisplatin and also AgNO3 and silver sulfadiazine, respectively. Both IC50 and MIC values are among the lowest values found for any silver derivatives against the cell lines and bacterial strains used in this work. The structure-activity relationships were clear. The most cytotoxic and antimicrobial derivatives were those with the triphenylphosphane and [N3P3(NHCy)6] ligands. A significant improvement in the activity was also observed upon a rise in the number of silver atoms linked to the phosphazene ring.
Collapse
Affiliation(s)
- Elena Gascón
- Departamento de Química Inorgánica, Facultad de Ciencias, Instituto de Síntesis Química y Catálisis Homogénea , Universidad de Zaragoza-CSIC , Pedro Cerbuna 12 , 50009 Zaragoza , Spain
| | - Sara Maisanaba
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología , Universidad Pablo de Olavide , Ctra. Utrera, Km 1 , 41013 Sevilla , Spain
| | - Isabel Otal
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Medicina Preventiva y Salud Pública , Universidad de Zaragoza , Zaragoza 50009 , Spain.,Instituto de Salud Carlos III , CIBER de Enfermedades Respiratorias , E-28029 Madrid , Spain
| | - Eva Valero
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área Nutrición y Bromatología , Universidad Pablo de Olavide , Ctra. Utrera, Km 1 , 41013 Sevilla , Spain
| | - Guillermo Repetto
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología , Universidad Pablo de Olavide , Ctra. Utrera, Km 1 , 41013 Sevilla , Spain
| | - Peter G Jones
- Institut für Anorganische und Analytische Chemie , Technische Universität Braunschweig , Hagenring 30 , D-38106 Braunschweig , Germany
| | - Josefina Jiménez
- Departamento de Química Inorgánica, Facultad de Ciencias, Instituto de Síntesis Química y Catálisis Homogénea , Universidad de Zaragoza-CSIC , Pedro Cerbuna 12 , 50009 Zaragoza , Spain
| |
Collapse
|