Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
1.
Dalton Trans ; 53(18): 7682-7693, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38573236

ABSTRACT

Dysregulation of Fibroblast Growth Factor Receptors (FGFRs) signaling has been associated with breast cancer, yet employing FGFR-targeted delivery systems to improve the efficacy of cytotoxic agents is still sparsely exploited. Herein, we report four new bi-functional ruthenium-peptide conjugates (RuPCs) with FGFR-targeting and pH-dependent releasing abilities, envisioning the selective delivery of cytotoxic Ru complexes to FGFR(+)-breast cancer cells, and controlled activation at the acidic tumoral microenvironment. The antiproliferative potential of the RuPCs and free Ru complexes was evaluated in four breast cancer cell lines with different FGFR expression levels (SKBR-3, MDA-MB-134-VI, MCF-7, and MDA-MB-231) and in human dermal fibroblasts (HDF), at pH 6.8 and pH 7.4 aimed at mimicking the tumor microenvironment and normal tissues/bloodstream pHs, respectively. The RuPCs showed higher cytotoxicity in cells with higher level of FGFR expression at acidic pH. Additionally, RuPCs showed up to 6-fold higher activity in the FGFR(+) breast cancer lines compared to the normal cell line. The release profile of Ru complexes from RuPCs corroborates the antiproliferative effects observed. Remarkably, the cytotoxicity and releasing ability of RuPCs were shown to be strongly dependent on the conjugation of the peptide position in the Ru complex. Complementary molecular dynamic simulations and computational calculations were performed to help interpret these findings at the molecular level. In summary, we identified a lead bi-functional RuPC that holds strong potential as a FGFR-targeted chemotherapeutic agent.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Peptides , Receptors, Fibroblast Growth Factor , Ruthenium , Female , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Drug Screening Assays, Antitumor , Hydrogen-Ion Concentration , Peptides/chemistry , Peptides/pharmacology , Peptides/therapeutic use , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Ruthenium/chemistry , Ruthenium/pharmacology , Ruthenium/therapeutic use
2.
J Inorg Biochem ; 253: 112486, 2024 04.
Article in English | MEDLINE | ID: mdl-38266323

ABSTRACT

The modular synthesis of the heteroscorpionate core is explored as a tool for the rapid development of ruthenium-based therapeutic agents. Starting with a series of structurally diverse alcohol-NN ligands, a family of heteroscorpionate-based ruthenium derivatives was synthesized, characterized, and evaluated as an alternative to platinum therapy for breast cancer therapy. In vitro, the antitumoral activity of the novel derivatives was assessed in a series of breast cancer cell lines using UNICAM-1 and cisplatin as metallodrug control. Through this approach, a bimetallic heteroscorpionate-based metallodrug (RUSCO-2) was identified as the lead compound of the series with an IC50 value range as low as 3-5 µM. Notably, RUSCO-2 was found to be highly cytotoxic in TNBC cell lines, suggesting a mode of action independent of the receptor status of the cells. As a proof of concept and taking advantage of the luminescent properties of one of the complexes obtained, uptake was monitored in human breast cancer MCF7 cell lines by fluorescence lifetime imaging microscopy (FLIM) to reveal that the compound is evenly distributed in the cytoplasm and that the incorporation of the heteroscorpionate ligand protects it from aqueous processes, conversion in another entity, or the loss of the chloride group. Finally, ROS studies were conducted, lipophilicity was estimated, the chloride/water exchange was studied, and stability studies in simulated biological media were carried out to propose structure-activity relationships.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Coordination Complexes , Ruthenium , Humans , Female , Breast Neoplasms/drug therapy , Ruthenium/pharmacology , Ruthenium/therapeutic use , Ligands , Chlorides , MCF-7 Cells , Cell Line, Tumor
3.
Adv Healthc Mater ; 13(2): e2302526, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37823717

ABSTRACT

Central post-stroke pain (CPSP) is a chronic neuropathic pain caused by cerebrovascular lesion or disfunction after stroke. Convincing evidence suggest that excessive reactive oxygen species (ROS), generated matrix metalloproteinase (MMPs) and neuroinflammation are largely involved in the development of pain. In this study, an effective strategy is reported for treating pain hypersensitivity using an endoplasmic reticulum (ER)-targeted metal-organic framework (MOF)-confined ruthenium (Ru) nanozyme. The Ru MOF is coated with a p-dodecylbenzene sulfonamide (p-DBSN) modified liposome with endoplasmic reticulum-targeted function. The experimental results reveals that ROS, Emmprin, MMP-2, and MMP-9 are upregulated in the brain of CPSP mice, along with the elevated expression of inflammation markers such as TNF-α and IL-6. Compared to vehicle, one-time intravenous administration of ER-Ru MOF significantly reduces mechanical hypersensitivity after CPSP for three days. Overall, ER-Ru MOF system can inhibit oxidative stress in the brain tissues of CPSP model, reduce MMPs expression, and suppress neuroinflammation response-induced injury, resulting in satisfactory prevention and effective treatment of CPSP during a hemorrhagic stroke. The ER-Ru MOF is expected to be useful for the treatment of neurological diseases associated with the vicious activation of ROS, based on the generality of the approach used in this study.


Subject(s)
Metal-Organic Frameworks , Neuralgia , Ruthenium , Mice , Animals , Reactive Oxygen Species/metabolism , Ruthenium/pharmacology , Ruthenium/therapeutic use , Neuroinflammatory Diseases , Oxidative Stress , Endoplasmic Reticulum/metabolism
4.
Nanomedicine ; 53: 102703, 2023 09.
Article in English | MEDLINE | ID: mdl-37591367

ABSTRACT

Carbosilane metallodendrimers, based on the arene Ru(II) complex (CRD13) and integrated to imino-pyridine surface groups have been investigated as an anticancer agent in a mouse model with triple-negative breast cancer. The dendrimer entered into the cells efficiently, and exhibited selective toxicity for 4T1 cells. In vivo investigations proved that a local injection of CRD13 caused a reduction of tumour mass and was non-toxic. ICP analyses indicated that Ru(II) accumulated in all tested tissues with a greater content detected in the tumour.


Subject(s)
Antineoplastic Agents , Ruthenium , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Ruthenium/pharmacology , Ruthenium/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor
5.
Photodiagnosis Photodyn Ther ; 42: 103615, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37201769

ABSTRACT

Recent studies involving photosensitizing agents containing ruthenium (Ru) are interpreted as an indication that this approach might be useful for treatment of bladder cancer. The absorbance spectrum of such agents tends to be limited to wavelengths < 600 nm. While this can spare underlying tissues from photodamage, this will limit applications to instances where only a thin layer of malignant cells is present. Among the more potentially interesting results is a protocol that uses only Ru nanoparticles. Other issues in Ru-based photodynamic therapy are discussed including the limited absorbance spectrum, questions relating to methodology and a general lack of details concerning localization and death pathways.


Subject(s)
Photochemotherapy , Ruthenium , Photosensitizing Agents/therapeutic use , Photochemotherapy/methods , Ruthenium/therapeutic use
6.
J Med Chem ; 66(10): 6922-6937, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37185020

ABSTRACT

Synergistic drug combinations can extend the use of poly(ADP-ribose) polymerase inhibitors (PARPi) such as Olaparib to BRCA-proficient tumors and overcome acquired or de novo drug resistance. To identify new synergistic combinations for PARPi, we screened a "micro-library" comprising a mix of commercially available drugs and DNA-binding ruthenium(II) polypyridyl complexes (RPCs) for Olaparib synergy in BRCA-proficient triple-negative breast cancer cells. This identified three hits: the natural product Curcumin and two ruthenium(II)-rhenium(I) polypyridyl metallomacrocycles. All combinations identified were effective in BRCA-proficient breast cancer cells, including an Olaparib-resistant cell line, and spheroid models. Mechanistic studies indicated that synergy was achieved via DNA-damage enhancement and resultant apoptosis. Combinations showed low cytotoxicity toward non-malignant breast epithelial cells and low acute and developmental toxicity in zebrafish embryos. This work identifies RPC metallomacrocycles as a novel class of agents for cancer combination therapy and provides a proof of concept for the inclusion of metallocompounds within drug synergy screens.


Subject(s)
Ovarian Neoplasms , Ruthenium , Humans , Animals , Female , Ruthenium/pharmacology , Ruthenium/therapeutic use , Zebrafish , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Ovarian Neoplasms/drug therapy , Phthalazines/pharmacology , Phthalazines/therapeutic use , DNA , Cell Line, Tumor
7.
Int J Mol Sci ; 24(7)2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37047448

ABSTRACT

Based on compelling preclinical evidence concerning the progress of our novel ruthenium-based metallotherapeutics, we are focusing research efforts on challenging indications for the treatment of invasive neoplasms such as the triple-negative breast cancer (TNBC). This malignancy mainly afflicts younger women, who are black, or who have a BRCA1 mutation. Because of faster growing and spreading, TNBC differs from other invasive breast cancers having fewer treatment options and worse prognosis, where existing therapies are mostly ineffective, resulting in a large unmet biomedical need. In this context, we benefited from an experimental model of TNBC both in vitro and in vivo to explore the effects of a biocompatible cationic liposomal nanoformulation, named HoThyRu/DOTAP, able to effectively deliver the antiproliferative ruthenium(III) complex AziRu, thus resulting in a prospective candidate drug. As part of the multitargeting mechanisms featuring metal-based therapeutics other than platinum-containing agents, we herein validate the potential of HoThyRu/DOTAP liposomes to act as a multimodal anticancer agent through inhibition of TNBC cell growth and proliferation, as well as migration and invasion. The here-obtained preclinical findings suggest a potential targeting of the complex pathways network controlling invasive and migratory cancer phenotypes. Overall, in the field of alternative chemotherapy to platinum-based drugs, these outcomes suggest prospective brand-new settings for the nanostructured AziRu complex to get promising goals for the treatment of metastatic TNBC.


Subject(s)
Antineoplastic Agents , Ruthenium , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/pathology , Ruthenium/pharmacology , Ruthenium/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Fatty Acids, Monounsaturated , Liposomes/therapeutic use , Cell Line, Tumor
8.
Int J Mol Sci ; 23(21)2022 Nov 06.
Article in English | MEDLINE | ID: mdl-36362381

ABSTRACT

Ruthenium(II) arene complexes exhibit promising chemotherapeutic properties. In this study, the effect of the counter anion in Ru(II) complexes was evaluated by analyzing the biological effect of two Ru(II) p-cymene derivatives with the 1,10-phenanthroline-5,6-dione ligand of general-formula [(η6-arene)Ru(L)Cl][X] X = CF3SO3 (JHOR10) and PF6 (JHOR11). The biological activity of JHOR10 and JHOR11 was examined in the ovarian carcinoma cell line A2780, colorectal carcinoma cell line HCT116, doxorubicin-resistant HCT116 (HCT116-Dox) and in normal human dermal fibroblasts. Both complexes JHOR10 and JHOR11 displayed an antiproliferative effect on A2780 and HCT116 cell lines, and low cytotoxicity in fibroblasts. Interestingly, JHOR11 also showed antiproliferative activity in the HCT116-Dox cancer cell line, while JHOR10 was inactive. Studies in A2780 cells showed that JHOR11 induced the production of reactive oxygen species (ROS) that trigger autophagy and cellular senescence, but no apoptosis induction. Further analysis showed that JHOR11 presented no tumorigenicity, with no effect in the cellular mobility, as evaluated by thye wound scratch assay, and no anti- or pro-angiogenic effect, as evaluated by the ex-ovo chorioallantoic membrane (CAM) assay. Importantly, JHOR11 presented no toxicity in chicken and zebrafish embryos and reduced in vivo the proliferation of HCT116 injected into zebrafish embryos. These results show that these are suitable complexes for clinical applications with improved tumor cell cytotoxicity and low toxicity, and that counter-anion alteration might be a viable clinical strategy for improving chemotherapy outcomes in multidrug-resistant (MDR) tumors.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Ovarian Neoplasms , Ruthenium , Animals , Humans , Female , Ruthenium/pharmacology , Ruthenium/therapeutic use , Zebrafish , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Ovarian Neoplasms/drug therapy , Coordination Complexes/pharmacology , Cell Proliferation
9.
Ophthalmic Genet ; 43(6): 756-761, 2022 12.
Article in English | MEDLINE | ID: mdl-36317817

ABSTRACT

BACKGROUND: Two major treatment modalities for retinoblastoma, intraarterial chemotherapy (IAC) and intravitreal chemotherapy (IVitC), have superseded external beam radiotherapy for eye salvage. In this new setting our objectives were to evaluate the indications for plaque radiotherapy, complications, and recurrence rates. METHODS: Retrospective detailed review of patient's charts was performed for all subjects treated with plaque radiotherapy for retinoblastoma between January 2015 and December 2020. RESULTS: A total of 12 eyes of 12 patients were included. Mean age at plaque insertion was 45 months (median 29, range 17-150). The treatment dose was 40 Gy to the tumor apex. The indication for plaque radiotherapy was salvage therapy in 11 eyes (92%) and primary treatment in one eye (8%). At last follow-up from plaque insertion (mean 36 months, range 3-67), four (33%) patients had visual acuity better than 0.5 LogMAR and four (33%) had visual acuity worse than 1.0 LogMAR. Radiation-related complications were: one (8%) vitreous haemorrhage, two (16%) non-proliferative radiation retinopathy and one (8%) cataract. Recurrence was detected in four (33%) patients at a mean of 7.8 months (median 5, range 1-20) post-plaque. Globe salvage rate was 75%, as three eyes required enucleation, one to treat recurrence of the tumor treated with plaque and two to treat recurrence of other tumors. CONCLUSIONS: In the current era of retinoblastoma management, a role for plaque radiotherapy remains for salvage or primary treatment in eyes with localised active tumor, providing tumor control in 66%. Close observation is recommended to both detect recurrence and radiation-related complications.


Subject(s)
Cataract , Retinal Neoplasms , Retinoblastoma , Ruthenium , Humans , Infant , Child, Preschool , Retinoblastoma/drug therapy , Retinoblastoma/radiotherapy , Retinoblastoma/pathology , Retinal Neoplasms/drug therapy , Retinal Neoplasms/radiotherapy , Retinal Neoplasms/pathology , Ruthenium/therapeutic use , Retrospective Studies , Treatment Outcome
10.
Molecules ; 27(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36235023

ABSTRACT

Cancer treatments which include conventional chemotherapy have not proven very successful in curing human malignancies. The failures of these treatment modalities include inherent resistance, systemic toxicity and severe side effects. Out of 50% patients administrated to chemotherapy, only 5% survive. For these reasons, the identification of new drug designs and therapeutic strategies that could target cancer cells while leaving normal cells unaffected still continues to be a challenge. Despite advances that have led to the development of new therapies, treatment options are still limited for many types of cancers. This review provides an overview of platinum, copper and ruthenium metal based anticancer drugs in clinical trials and in vitro/in vivo studies. Presumably, copper and ruthenium complexes have greater potential than Pt(II) complexes, showing reduced toxicity, a new mechanism of action, a different spectrum of activity and the possibility of non-cross-resistance. We focus the discussion towards past, present and future aspects.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Ruthenium , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Copper/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Platinum/therapeutic use , Ruthenium/therapeutic use
11.
Biomaterials ; 289: 121757, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36058028

ABSTRACT

The alternations in the hypoxic and immune microenvironment are closely related to the therapeutic effect and prognosis of oral squamous cell carcinoma (OSCC). Herein, a new nanocomposite, TiO2@Ru@siRNA is constructed from a ruthenium-based photosensitizer (Ru) modified-TiO2 nanoparticles (NPs) loaded with siRNA of hypoxia-inducible factor-1α (HIF-1α). Under visible light irradiation, TiO2@Ru@siRNA can elicit both Type I and Type II photodynamic effects, which causes lysosomal damage, HIF-1α gene silencing, and OSCC cell elimination efficiently. As a consequence of hypoxia relief and pyroptosis induction, TiO2@Ru@siRNA reshapes the immune microenvironment by downregulation of key immunosuppressive factors, upregulation of immune cytokines, and activation of CD4+ and CD8+ T lymphocytes. Furthermore, patient-derived xenograft (PDX) and rat oral experimental carcinogenesis models prove that TiO2@Ru@siRNA-mediated photodynamic therapy significantly inhibits the tumor growth and progression, and markedly enhances cancer immunity. In all, this study presents an effective hypoxia-adaptive photo-immunotherapeutic nanosystem with great potential for OSCC prevention and treatment.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Nanoparticles , Ruthenium , Animals , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cytokines , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit , Immunotherapy , Metal Nanoparticles , Mouth Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , RNA, Small Interfering/genetics , Rats , Ruthenium/therapeutic use , Squamous Cell Carcinoma of Head and Neck , Titanium , Tumor Microenvironment
12.
J Med Chem ; 65(19): 13041-13051, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36134739

ABSTRACT

The curative effect of sorafenib in hepatocellular carcinoma (HCC) is limited and sorafenib resistance remains a major obstacle for HCC. To overcome this obstacle, a new photoactive sorafenib-Ru(II) complex Ru-Sora has been designed. Upon irradiation (λ = 465 nm), Ru-Sora rapidly releases sorafenib and generates reactive oxygen species, which can oxidize intracellular substances such as GSH. Cellular experiments show that irradiated Ru-Sora is highly cytotoxic toward Hep-G2 cells, including sorafenib-resistant Hep-G2-SR cells. Compared to sorafenib, Ru-Sora has a significant photoactivated chemotherapeutic effect against Hep-G2-SR cancer cells and 3D Hep-G2 multicellular tumor spheroids. Furthermore, Ru-Sora inducing apoptosis and ferroptosis is proved by GSH depletion, GPX4 downregulation, and lipid peroxide accumulation. Metabolomics results suggest that Ru-Sora exerts photocytotoxicity by disrupting the purine metabolism, which is expected to inhibit tumor development. This study provides a promising strategy for enhancing chemotherapy and combating drug-resistant HCC disease.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , Prodrugs , Ruthenium , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Hep G2 Cells , Humans , Lipid Peroxides/pharmacology , Liver Neoplasms/pathology , Prodrugs/pharmacology , Prodrugs/therapeutic use , Purines/pharmacology , Reactive Oxygen Species/metabolism , Ruthenium/pharmacology , Ruthenium/therapeutic use , Sorafenib/pharmacology
13.
Dalton Trans ; 51(25): 9673-9680, 2022 Jun 27.
Article in English | MEDLINE | ID: mdl-35670555

ABSTRACT

Ruthenium-based assemblies containing tetrapyridylporphyrins (TPyP) in their structure have been evaluated as photosensitizers (PS) to treat rheumatoid arthritis (RA) by photodynamic therapy (PDT). TPyP is useless by itself as a PS due to its low solubility in biological media, however, incorporated in metallacages it can be internalized in cells. The study shows a cellular antiproliferative activity in fibroblast-like synoviocyte (FLS) in the lower nanomolar range in the presence of light, and no dark toxicity at 1 µM concentration, thus having an excellent photoactivity index. The presence of diamagnetic (Zn2+) and paramagnetic (Co2+) metals in the center of TPyP impairs the effectiveness of PDT, showing no (Co) or reduced (Zn) photoactivity. A total of five metallacages with different structural characteristics have been evaluated, and our results suggest that the incorporation of PS in metalla-assemblies is not only an elegant method to increase solubility in biological media for TPyP but also appears to be an efficient hybrid system to treat RA by PDT.


Subject(s)
Arthritis, Rheumatoid , Photochemotherapy , Ruthenium , Arthritis, Rheumatoid/drug therapy , Fibroblasts , Humans , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Ruthenium/pharmacology , Ruthenium/therapeutic use
14.
Recent Pat Anticancer Drug Discov ; 18(2): 174-186, 2022.
Article in English | MEDLINE | ID: mdl-35770412

ABSTRACT

BACKGROUND: Doxorubicin is a significant drug for the treatment of breast cancer, but its cardiotoxicity is an obvious obstacle. Previously, we confirmed that ruthenium complex (Δ-Ru1) and doxorubicin (Δ-Ru1/Dox) combination had a synergistic effect in MCF-7 cells, but its biological effect in vivo is unknown. PURPOSES: To find a way to overcome the toxicity of doxorubicin and build MCF-7 xenograft tumor mouse model to test whether this potential combination has better efficacy and less toxicity. METHODS: The tumor model of nude mice was established to verify the synergistic antitumor effect of the drug combination in vivo. H&E staining was used to detect the toxicity of major organs in mice. Sirius red staining and transmission electron microscopy were used to detect cardiotoxicity. Prussian blue was used to measure iron accumulation in heart tissue. TUNEL staining was used to detect the antitumor effect in vivo. Immunohistochemical staining was used to detect the expression of iron death-related pathway proteins. High-throughput sequencing techniques were used to determine the molecular mechanism of ferroptosis. RESULTS: Histopathological analysis of tumor tissues indicated that the Δ-Ru1/Dox combination significantly promoted tumor cell apoptosis. Doxorubicin damaged cardiac tissue by inducing fibrosis and iron accumulation, but it was reversed by the Δ-Ru1/Dox combination treatment. Further exploration found that doxorubicin could regulate iron accumulation in the ferroptosis pathway and the expression of lipid peroxidation-related proteins, including upregulation of Tf, DMT1, and HO-1, and downregulation of Nrf2, SLC7A11, and GPX4. CONCLUSION: Δ-Ru1/Dox combination synergistically inhibits tumor growth, and it can significantly reduce and alleviate the toxic side effects of doxorubicin, especially cardiac injury.


Subject(s)
Breast Neoplasms , Ruthenium , Mice , Humans , Animals , Female , Ruthenium/pharmacology , Ruthenium/therapeutic use , Cardiotoxicity , Mice, Nude , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Breast Neoplasms/pathology , Iron/pharmacology , Iron/therapeutic use , Apoptosis
15.
Curr Med Chem ; 29(31): 5159-5178, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35366762

ABSTRACT

Human parasitic infections cause a combined global mortality rate of over one million people per annum and represent some of the most challenging diseases for medical intervention. Current chemotherapeutic strategies often require prolonged treatment, coupled with subsequent drug-induced cytotoxic morbidity to the host, while resistance generation is also a major concern. Metals have been used extensively throughout the history of medicine, with more recent applications as anticancer and antimicrobial agents. Ruthenium metallotherapeutic antiparasitic agents are highly effective at targeting a range of key parasites, including the causative agents of malaria, trypanosomiasis, leishmaniasis, amoebiasis, toxoplasmosis and other orphan diseases, while demonstrating lower cytotoxicity profiles than current treatment strategies. Generally, such compounds also demonstrate activity against multiple cellular target sites within parasites, including inhibition of enzyme function, cell membrane perturbation, and alterations to metabolic pathways, therefore reducing the opportunity for resistance generation. This review provides a comprehensive and subjective analysis of the rapidly developing area of ruthenium metal- based antiparasitic chemotherapeutics, in the context of rational drug design and potential clinical approaches to combatting human parasitic infections.


Subject(s)
Anti-Infective Agents , Leishmaniasis , Parasitic Diseases , Ruthenium , Trypanosomiasis , Anti-Infective Agents/therapeutic use , Antiparasitic Agents/pharmacology , Antiparasitic Agents/therapeutic use , Humans , Leishmaniasis/drug therapy , Parasitic Diseases/drug therapy , Ruthenium/pharmacology , Ruthenium/therapeutic use , Trypanosomiasis/drug therapy
16.
Chem Asian J ; 17(13): e202200270, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35419865

ABSTRACT

Metal complexes have shown great potential in cancer immunotherapy. This review briefly introduces the basic concepts and strategies of cancer immunotherapy and summarizes the recent discoveries on the immune effects of traditional platinum-based anticancer compounds. In addition, we also outline the latest research progresses on metal complexes for cancer immunotherapy focusing on platinum, ruthenium, iridium, rhenium and copper complexes. Finally, the research perspectives and unsolved problems on the applications of metallo-anticancer agents in cancer immunotherapy are purposed.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Ruthenium , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Humans , Immunotherapy , Iridium , Neoplasms/drug therapy , Ruthenium/pharmacology , Ruthenium/therapeutic use
17.
J Inorg Biochem ; 226: 111652, 2022 01.
Article in English | MEDLINE | ID: mdl-34741931

ABSTRACT

In recent years, Ru polypyridyl complexes have been intensively studied for their anticancer activity. The vast majority of research focuses on assessing their cytotoxic activity, as well as targeting cancer cells with them. Since the formation of metastases poses a greater risk than primary tumors, scientists recently began evaluating these compounds as potential metastasis inhibitors. This review highlights the latest achievements in this field with particular attention to the identification of the target proteins responsible for such activity. Cell migration, invasion, and adhesion are key components of metastasis, therefore understanding how they are affected by Ru polypyridyl complexes is of great importance. KEYWORDS: Ruthenium polypyridyl complexes Antimetastatic Migration Invasion Adhesion Metalloproteinases.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Pyridines , Ruthenium , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Movement/drug effects , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use , Humans , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Pyridines/chemistry , Pyridines/therapeutic use , Ruthenium/chemistry , Ruthenium/therapeutic use
18.
Int J Mol Sci ; 24(1)2022 Dec 30.
Article in English | MEDLINE | ID: mdl-36614133

ABSTRACT

Cancer is one of the leading cause of lethality worldwide, CRC being the third most common cancer reported worldwide, with 1.85 million cases and 850,000 deaths annually. As in all other cancers, kinases are one of the major enzymes that play an essential role in the incidence and progression of CRC. Thus, using multi-kinase inhibitors is one of the therapeutic strategies used to counter advanced-stage CRC. Regorafenib is an FDA-approved drug in the third-line therapy of refractory metastatic colorectal cancer. Acquired resistance to cancers and higher toxicity of these drugs are disadvantages to the patients. To counter this, combination therapy is used as a strategy where a minimal dose of drugs can be used to get a higher efficacy and reduce drug resistance development. Ruthenium-based compounds are observed to be a potential alternative to platinum-based drugs due to their significant safety and effectiveness. Formerly, our lab reported Ru-1, a ruthenium-based compound, for its anticancer activity against multiple cancer cells, such as HepG2, HCT116, and MCF7. This study evaluates Ru-1's activity against regorafenib-resistant HCT116 cells and as a combination therapeutic with regorafenib. Meanwhile, the mechanism of the effect of Ru-1 alone and with regorafenib as a combination is still unknown. In this study, we tested a drug combination (Ru-1 and regorafenib) against a panel of HT29, HCT116, and regorafenib-resistant HCT116 cells. The combination showed a synergistic inhibitory activity. Several mechanisms underlying these numerous synergistic activities, such as anti-proliferative efficacy, indicated that the combination exhibited potent cytotoxicity and enhanced apoptosis induction. Disruption of mitochondrial membrane potential increased intracellular ROS levels and decreased migratory cell properties were observed. The combination exhibited its activity by regulating PI3K/Akt and p38 MAP kinase signalling. This indicates that the combination of REG/Ru-1 targets cancer cells by modulating the PI3K/Akt and ERK signalling.


Subject(s)
Colorectal Neoplasms , Ruthenium , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Ruthenium/pharmacology , Ruthenium/therapeutic use , Apoptosis , Colorectal Neoplasms/pathology , Phenylurea Compounds/therapeutic use , Cell Line, Tumor
19.
ACS Appl Mater Interfaces ; 13(38): 45269-45278, 2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34520159

ABSTRACT

Rationally constructing single-atom enzymes (SAEs) with superior activity, robust stability, and good biocompatibility is crucial for tumor therapy but still remains a substantial challenge. In this work, we adopt biocompatible carbon dots as the carrier material to load Ru single atoms, achieving Ru SAEs with superior multiple enzyme-like activity and stability. Ru SAEs behave as oxidase, peroxidase, and glutathione oxidase mimics to synchronously catalyze the generation of reactive oxygen species (ROS) and the depletion of glutathione, thus amplifying the ROS damage and finally causing the death of cancer cells. Notably, Ru SAEs exhibit excellent peroxidase-like activity with a specific activity of 7.5 U/mg, which surpasses most of the reported SAEs and is 20 times higher than that of Ru/C. Theoretical results reveal that the electrons of the Ru 4d orbital in Ru SAEs are transferred to O atoms in H2O2 and then efficiently activate H2O2 to produce •OH. Our work may provide some inspiration for the design of SAEs for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Quantum Dots/therapeutic use , Ruthenium/therapeutic use , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carbon/chemistry , Catalysis , Cell Line, Tumor , Glutathione/metabolism , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Kinetics , Mice , Oxidation-Reduction , Oxidative Stress/drug effects , Quantum Dots/chemistry , Ruthenium/chemistry
20.
Molecules ; 26(15)2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34361543

ABSTRACT

Lung cancer is one of the most common malignancies with the highest mortality rate and the second-highest incidence rate after breast cancer, posing a serious threat to human health. The accidental discovery of the antitumor properties of cisplatin in the early 1960s aroused a growing interest in metal-based compounds for cancer treatment. However, the clinical application of cisplatin is limited by serious side effects and drug resistance. Therefore, other transition metal complexes have been developed for the treatment of different malignant cancers. Among them, Ru(II/III)-based complexes have emerged as promising anticancer drug candidates due to their potential anticancer properties and selective cytotoxic activity. In this review, we summarized the latest developments of Ru(II/III) complexes against lung cancer, focusing mainly on the mechanisms of their biological activities, including induction of apoptosis, necroptosis, autophagy, cell cycle arrest, inhibition of cell proliferation, and invasion and metastasis of lung cancer cells.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Cytotoxins , Lung Neoplasms/drug therapy , Ruthenium , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use , Cytotoxins/chemistry , Cytotoxins/therapeutic use , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Ruthenium/chemistry , Ruthenium/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...