Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
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.
Inorg Chem ; 63(13): 5783-5804, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38502532

ABSTRACT

In recent years, organometallic complexes have attracted much attention as anticancer therapeutics aiming at overcoming the limitations of platinum drugs that are currently marketed. Still, the development of half-sandwich organometallic cobalt complexes remains scarcely explored. Four new cobalt(III)-cyclopentadienyl complexes containing N,N-heteroaromatic bidentate, and phosphane ligands were synthesized and fully characterized by elemental analysis, spectroscopic techniques, and DFT methods. The cytotoxicity of all complexes was determined in vitro by the MTS assay in colorectal (HCT116), ovarian (A2780), and breast (MDA-MB-231 and MCF-7) human cancer cell lines and in a healthy human cell line (fibroblasts). The complexes showed high cytotoxicity in cancer cell lines, mostly due to ROS production, apoptosis, autophagy induction, and disruption of the mitochondrial membrane. Also, these complexes were shown to be nontoxic in vivo in an ex ovo chick embryo yolk sac membrane (YSM) assay.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Ovarian Neoplasms , Animals , Chick Embryo , Humans , Female , Cell Line, Tumor , Antineoplastic Agents/chemistry , Platinum/pharmacology , Cobalt/pharmacology , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Apoptosis
3.
Dalton Trans ; 51(7): 2593-2609, 2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35106525

ABSTRACT

Chemotherapeutic metallodrugs such as cisplatin and its derivatives are among the most widely applied anticancer treatments worldwide. Despite their clinical success, patients suffer from severe adverse effects while subjected to treatment due to platinum's low selectivity for tumour over healthy tissues. Additionally, intrinsic or acquired resistance to metallodrugs, as well as their inability to reach cancer metastases, often results in therapeutic failure. The evident need for highly efficient and specific treatments has driven the scientific community to research novel ways to surpass the stated limitations. Within this scenario, a rising number of smart drug delivery systems have been lately reported to target primary cancers or metastases, where the metallodrugs are released in a controlled and selective way triggered by specific tumour-related stimuli, thus suggesting a viable and attractive therapeutic approach. Herein, we discuss the main efforts undertaken in the past few years towards the smart delivery of metal-based drugs and drug candidates to tumour sites, particularly focusing on the pH- and/or redox-responsive targeted delivery of platinum and ruthenium anticancer complexes.


Subject(s)
Ruthenium
4.
Molecules ; 26(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34885928

ABSTRACT

Considering our interest in the use of peptides as potential target-specific drugs or as delivery vectors of metallodrugs for various biomedical applications, it is crucial to explore improved synthetic methodologies to accomplish the highest peptide crude purity in the shortest time possible. Therefore, we compared "classical" fluorenylmethoxycarbonyl (Fmoc)-solid phase peptide synthesis (SPPS) with ultrasound(US)-assisted SPPS based on the preparation of three peptides, namely the fibroblast growth factor receptor 3(FGFR3)-specific peptide Pep1 (VSPPLTLGQLLS-NH2) and the novel peptides Pep2 (RQMATADEA-NH2) and Pep3 (AAVALLPAVLLALLAPRQMATADEA-NH2), which are being developed aimed at interfering with the intracellular protein-protein interaction(PPI) RANK-TRAF6. Our results demonstrated that US-assisted SPPS led to a 14-fold (Pep1) and 4-fold time reduction (Pep2) in peptide assembly compared to the "classical" method. Interestingly, US-assisted SPPS yielded Pep1 in higher purity (82%) than the "classical" SPPS (73%). The significant time reduction combined with high crude peptide purity attained prompted use to apply US-assisted SPPS to the large peptide Pep3, which displays a high number of hydrophobic amino acids and homooligo-sequences. Remarkably, the synthesis of this 25-mer peptide was attained during a "working day" (347 min) in moderate purity (approx. 49%). In conclusion, we have reinforced the importance of using US-SPPS towards facilitating the production of peptides in shorter time with increased efficacy in moderate to high crude purity. This is of special importance for long peptides such as the case of Pep3.


Subject(s)
Peptides/chemical synthesis , Solid-Phase Synthesis Techniques/methods , Humans , Peptides/chemistry , Receptor Activator of Nuclear Factor-kappa B/chemistry , Receptors, Fibroblast Growth Factor/chemistry , Sonication/methods , TNF Receptor-Associated Factor 6/chemistry
5.
Dalton Trans ; 49(18): 5974-5987, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32314752

ABSTRACT

In this work we explored the possibility of improving the selectivity of a cytotoxic Ru complex [RuCp(PPh3)(2,2'-bipy)][CF3SO3] (where Cp = η5-cyclopentadienyl) TM34 towards FGFR(+) breast cancer cells. Molecular dynamics (MD) simulations of TM34 in a phosphatidylcholine membrane model pinpointed the cyclopentadienyl group as a favorable derivatization position for the peptide conjugation approach. Three new Ru(ii) complexes presenting a functionalized η5-cyclopentadienyl were synthesized, namely [Ru(η5-C5H4COOH)(2,2'-bipy)(PPh3)][CF3SO3] (TM281) and its precursors, [Ru(η5-C5H4COOCH2CH3)(η2-2,2'-bipy)(PPh3)][CF3SO3] (3) and [Ru(η5-C5H4COOCH2CH3)(PPh3)2Cl] (2). Complex TM281 was prepared by the hydrolysis of the ethyl ester group appended to the η5-cyclopentadienyl ligand of complex 3 with K2CO3 in water/acetonitrile, followed by mild protonation using an ion exchange resin. The newly synthesized complexes were fully characterized by NMR, FTIR and UV-vis spectroscopic techniques. Also, electrochemical studies were carried out by means of cyclic voltammetry in order to evaluate the stability of the compounds. Single crystal X-ray diffraction studies were carried out for compounds 3 and TM281 which crystallized in the monoclinic system, space group P21/n. The unprecedented synthesis and characterization of three half-sandwich ruthenium(ii)-cyclopentadienyl peptide conjugates and their preliminary biological evaluation against human FGFR(+) and FGFR(-) breast cancer cells are also reported.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coordination Complexes/pharmacology , Cyclopentanes/pharmacology , Peptides/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Ruthenium/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Cyclopentanes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Electrochemical Techniques , Female , Humans , Molecular Dynamics Simulation , Molecular Structure , Peptides/chemistry , Receptors, Fibroblast Growth Factor/metabolism , Ruthenium/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
6.
Molecules ; 24(1)2018 Dec 23.
Article in English | MEDLINE | ID: mdl-30583594

ABSTRACT

Precision medicine relies on individually tailored therapeutic intervention taking into account individual variability. It is strongly dependent on the availability of target-specific drugs and/or imaging agents that recognize molecular targets and patient-specific disease mechanisms. The most sensitive molecular imaging modalities, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET), rely on the interaction between an imaging radioprobe and a target. Moreover, the use of target-specific molecular tools for both diagnostics and therapy, theranostic agents, represent an established methodology in nuclear medicine that is assuming an increasingly important role in precision medicine. The design of innovative imaging and/or theranostic agents is key for further accomplishments in the field. G-protein-coupled receptors (GPCRs), apart from being highly relevant drug targets, have also been largely exploited as molecular targets for non-invasive imaging and/or systemic radiotherapy of various diseases. Herein, we will discuss recent efforts towards the development of innovative imaging and/or theranostic agents targeting selected emergent GPCRs, namely the Frizzled receptor (FZD), Ghrelin receptor (GHSR-1a), G protein-coupled estrogen receptor (GPER), and Sphingosine-1-phosphate receptor (S1PR). The pharmacological and clinical relevance will be highlighted, giving particular attention to the studies on the synthesis and characterization of targeted molecular imaging agents, biological evaluation, and potential clinical applications in oncology and non-oncology diseases. Whenever relevant, supporting computational studies will be also discussed.


Subject(s)
Ligands , Molecular Imaging , Precision Medicine , Receptors, G-Protein-Coupled/chemistry , Theranostic Nanomedicine , Animals , Drug Discovery , Humans , Molecular Imaging/methods , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Structure-Activity Relationship , Theranostic Nanomedicine/methods
SELECTION OF CITATIONS
SEARCH DETAIL