Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Dalton Trans ; 48(21): 7388-7393, 2019 Jun 07.
Article in English | MEDLINE | ID: mdl-30957798

ABSTRACT

Platinum(iv) carboxylate scaffolds have garnered considerable research interest because they can be engineered to function as prodrugs of clinical platinum(ii) anticancer drugs. These platinum(iv) prodrug complexes are stable and tunable, and activated by reduction to release their cytotoxic platinum(ii) cargo. Here we propose new platinum(iv) prodrug complexes designed to release cisplatin via photoreduction upon UV irradiation. The central strategy is to utilise aryl carboxylate ligands on the axial positions of that platinum(iv) scaffold that confer significant UV absorption and would stabilise carboxyl radical formation, thus favouring homolytic Pt-O bond cleavage. We isolated and identified aryl carboxyl radicals via spin-trapping and showed that the photoreduced platinum species mirror cisplatin reactivity toward DNA bases, thereby validating the efficacy of this approach.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Organoplatinum Compounds/chemical synthesis , Prodrugs/chemical synthesis , Cisplatin/chemistry , Ligands , Oxidation-Reduction , Structure-Activity Relationship , Ultraviolet Rays
2.
ChemMedChem ; 12(4): 300-311, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28028938

ABSTRACT

Platinum(IV) bis-carboxylates are highly versatile prodrug scaffolds with different axial ligands that can be functionalized while keeping the platinum(II) pharmacophore intact. Using a sequential acylation strategy, we developed a class of PtIV prodrugs of cisplatin with contrasting lipophilic and hydrophilic ligands. We investigated their stability, reduction rates, lipophilicity, aqueous solubility, and antiproliferative efficacies, and assessed for correlations among the parameters that could be useful in drug design. We showed that compounds with high lipophilicity result in better antiproliferative effects in vitro and in vivo, with one of the three compounds tested showing better efficacy than satraplatin against an animal model of colorectal cancer, owing to its higher solubility and lower reduction rates. Our asymmetric PtIV prodrugs may pave the way for a highly predictable, fine-tuned class of orally available PtIV prodrugs for the treatment of colorectal cancer.


Subject(s)
Antineoplastic Agents/chemistry , Platinum/chemistry , Prodrugs/chemistry , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Coordination Complexes/administration & dosage , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Coordination Complexes/toxicity , Disease Models, Animal , Drug Stability , Humans , Mice , Molecular Conformation , Oxidation-Reduction , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Prodrugs/toxicity , Solubility , Structure-Activity Relationship , Tissue Distribution , X-Ray Diffraction
3.
Chimia (Aarau) ; 69(3): 100-3, 2015.
Article in English | MEDLINE | ID: mdl-26507211

ABSTRACT

Kinetically-inert Pt(IV) carboxylate complexes have emerged in recent years as candidates for the development of next-generation platinum anticancer drugs. Being native prodrugs of clinically-important Pt(II) chemotherapeutic agents, the Pt(IV) scaffold can be exploited to incorporate additional functionalities while keeping the Pt(II) pharmacophore intact. This mini-review examines recent work performed to illuminate the mechanism of Pt(IV) prodrug activation and their use as versatile platforms for targeted chemotherapy.

4.
Nucleic Acids Res ; 43(11): 5284-96, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-25916851

ABSTRACT

Platinum-based anticancer drugs act therapeutically by forming DNA adducts, but suffer from severe toxicity and resistance problems, which have not been overcome in spite of decades of research. And yet defined chromatin targets have generally not been considered in the drug development process. Here we designed novel platinum-intercalator species to target a highly deformed DNA site near the nucleosome center. Between two seemingly similar structural isomers, we find a striking difference in DNA site selectivity in vitro, which comes about from stereochemical constraints that limit the reactivity of the trans isomer to special DNA sequence elements while still allowing the cis isomer to efficiently form adducts at internal sites in the nucleosome core. This gives the potential for controlling nucleosome site targeting in vivo, which would engender sensitivity to epigenetic distinctions and in particular cell type/status-dependent differences in nucleosome positioning. Moreover, while both compounds yield very similar DNA-adduct structures and display antitumor cell activity rivalling that of cisplatin, the cis isomer, relative to the trans, has a much more rapid cytotoxic effect and distinct impact on cell function. The novel stereochemical principles for controlling DNA site selectivity we discovered could aid in the design of improved site discriminating agents.


Subject(s)
Antineoplastic Agents/chemistry , Intercalating Agents/chemistry , Naphthalimides/chemistry , Nucleosomes/chemistry , Organoplatinum Compounds/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , DNA Adducts/analysis , Humans , Intercalating Agents/toxicity , Naphthalimides/toxicity , Organoplatinum Compounds/toxicity , Stereoisomerism
5.
Nanoscale ; 7(12): 5383-94, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25727105

ABSTRACT

The use of platinum-based chemotherapeutic drugs in cancer therapy still suffers from severe disadvantages, such as lack of appropriate selectivity for tumor tissues and insurgence of multi-drug resistance. Moreover, drug efficacy can be attenuated by several mechanisms such as premature drug inactivation, reduced drug uptake inside cells and increased drug efflux once internalized. The use of functionalized carbon nanotubes (CNTs) as chemotherapeutic drug delivery systems is a promising strategy to overcome such limitations due to their ability to enhance cellular internalization of poorly permeable drugs and thus increase the drug bioavailability at the diseased site, compared to the free drug. Furthermore, the possibility to encapsulate agents in the nanotubes' inner cavity can protect the drug from early inactivation and their external functionalizable surface is useful for selective targeting. In this study, a hydrophobic platinum(IV) complex was encapsulated within the inner space of two different diameter functionalized multi-walled CNTs (Pt(IV)@CNTs). The behavior of the complexes, compared to the free drug, was investigated on both HeLa human cancer cells and RAW 264.7 murine macrophages. Both CNT samples efficiently induced cell death in HeLa cancer cells 72 hours after the end of exposure to CNTs. Although the larger diameter CNTs were more cytotoxic on HeLa cells compared to both the free drug and the smaller diameter nanotubes, the latter allowed a prolonged release of the encapsulated drug, thus increasing its anticancer efficacy. In contrast, both Pt(IV)@CNT constructs were poorly cytotoxic on macrophages and induced negligible cell activation and no pro-inflammatory cytokine production. Both CNT samples were efficiently internalized by the two types of cells, as demonstrated by transmission electron microscopy observations and flow cytometry analysis. Finally, the platinum levels found in the cells after Pt(IV)@CNT exposure demonstrate that they can promote drug accumulation inside cells in comparison with treatment with the free complex. To conclude, our study shows that CNTs are promising nanocarriers to improve the accumulation of a chemotherapeutic drug and its slow release inside tumor cells, by tuning the CNT diameter, without inducing a high inflammatory response.


Subject(s)
Cell Survival/drug effects , Cisplatin/chemistry , Delayed-Action Preparations/chemistry , Nanocapsules/chemistry , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/ultrastructure , Absorption, Physicochemical , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cisplatin/administration & dosage , Delayed-Action Preparations/administration & dosage , Diffusion , HeLa Cells , Humans , Nanocapsules/ultrastructure , Particle Size , Prodrugs/administration & dosage , Prodrugs/chemistry
6.
Nanomedicine ; 10(7): 1465-75, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24486857

ABSTRACT

Carbon nanotubes (CNTs) are promising drug delivery systems due to their external functionalizable surface and their hollowed cavity that can encapsulate several bioactive molecules. In this study, the chemotherapeutic drug cisplatin or an inert platinum(IV) complex were entrapped inside functionalized-multi-walled-CNTs and intravenously injected into mice to investigate the influence of CNTs on the biodistribution of Pt-based molecules. The platinum levels in vital organs suggested that functionalized-CNTs did not affect cisplatin distribution, while they significantly enhanced the accumulation of Pt(IV) sample in some tissues (e.g. in the lungs, suggesting their potential application in lung cancer therapy) and reduced both kidney and liver accumulation (thus decreasing eventual nephrotoxicity, a typical side effect of cisplatin). Concurrently, CNTs did not induce any intrinsic abnormal immune response or inflammation, as confirmed by normal cytokine levels and histological evaluations. Therefore, functionalized nanotubes represent an efficient nano-carrier to improve accumulation of Pt species in targeted tissues/organs. From the clinical editor: In this preclinical study functionalized carbon nanotubes are reported to be safe and efficient for targeted delivery of platinum-containing compounds in rodents. Approaches like this may improve the treatment of specific cancers, since platinum based chemotherapies are commonly used, yet limited by toxicity and relatively poor target tissue concentration.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Nanotubes, Carbon , Platinum Compounds/pharmacokinetics , Animals , Drug Carriers , Female , Mice , Mice, Inbred BALB C , Tissue Distribution
7.
Curr Med Chem ; 21(2): 251-60, 2014.
Article in English | MEDLINE | ID: mdl-24059229

ABSTRACT

Drug resistance is a major hurdle to the success of chemotherapy. The permeability glycoprotein (P-gp) is an important factor dictating drug access to the cells, as it controls the efflux of chemotherapeutic agents against the concentration gradient. Pmd1, a P-gp-like protein, was recently isolated as a doxorubicin resistance gene in fission yeast. Although the null mutant of pmd1 (Δpmd1) exhibited sensitivity to doxorubicin, it showed an unexpectedly high resistance to the drug at relatively high concentrations. The data presented here suggest that this is due to the presence of cooperative processes that can complement and counteract drug cytotoxicity in the absence of Pmd1. One such factor, Rav1, is an essential factor in controlling the assembly of the pH-regulating transporter vacuolar-ATPase (V-ATPase) in fission yeast. The simultaneous disruption of Pmd1 and Rav1 resulted in a prominent accumulation of doxorubicin in the cytoplasm of cells, accompanied by a decline in cell viability. With concurrent treatment of pharmacological inhibitors in human cervical cancer cells, P-gp and V-ATPase were further shown to act synergistically to sensitize cells to doxorubicin also in the human cells. Furthermore, a novel Cornichon-like protein SPAC2C4.05 (herein named as Cor1) was demonstrated for the first time to be involved in the interaction with P-gp and V-ATPase to counteract doxorubicin-dependent cytotoxicity. Therefore this study identified a molecular cooperation between multiple membrane transporter proteins that confers chemoresistance to cells against the chemical insult of doxorubicin. Interestingly, this network exhibited differential effects to doxorubicin as compared with its close epimeric analog epirubicin, suggestive of the intricacy of the drug response regulated by this synergistic interaction. A model is discussed on how the versatility of this network can differentiate closely related chemical drug structures yet allow for the robustness to counteract a vast range of drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphatases/metabolism , Anthracyclines/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance/drug effects , Schizosaccharomyces/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adenosine Triphosphatases/antagonists & inhibitors , Anthracyclines/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
8.
Biomaterials ; 35(2): 748-59, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24140044

ABSTRACT

Among the arsenal of nano-materials, carbon nanotubes (CNTs) are becoming more prominent due to favorable attributes including their unique shape, which promotes cellular-uptake, and large aspect-ratio that facilitates functionalization of bioactive molecules on their surface. In this study, multi-walled carbon nanotubes (MWCNTs) were functionalized with either mitochondrial-targeting fluorescent rhodamine-110 (MWCNT-Rho) or non-targeting fluorescein (MWCNT-Fluo). Despite structural similarities, MWCNT-Rho associated well with mitochondria (ca. 80% co-localization) in contrast to MWCNT-Fluo, which was poorly localized (ca. 21% co-localization). Additionally, MWCNT-Rho entrapping platinum(IV) pro-drug of cisplatin (PtBz) displayed enhanced potency (IC50 = 0.34 ± 0.07 µM) compared to a construct based on MWCNT-Fluo (IC50 ≥ 2.64 µM). Concurrently, preliminary in vitro toxicity evaluation revealed that empty MWCNT-Rho neither decreased cell viability significantly nor interfered with mitochondrial membrane-potential, while seemingly being partially expelled from cells. Due to its targeting capability and apparent lack of cytotoxicity, MWCNT-Rho complex was used to co-encapsulate PtBz and a chemo-potentiator, 3-bromopyruvate (BP), and the resulting MWCNT-Rho(PtBz+BP) construct demonstrated superior efficacy over PtBz free drug in several cancer cell lines tested. Importantly, a 2-fold decrease in mitochondrial potential was observed, implying that mitochondrial targeting of compounds indeed incurred additional intended damage to mitochondria.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Mitochondria/drug effects , Nanotubes, Carbon/chemistry , Platinum/chemistry , Prodrugs/pharmacology , Cell Survival/drug effects , Drug Delivery Systems , Drug Synergism , Flow Cytometry , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial , Microscopy, Confocal , Microscopy, Electron, Transmission , Mitochondria/metabolism , Pyruvates/pharmacology , Rhodamines/pharmacology
9.
J Med Chem ; 55(17): 7571-82, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22876932

ABSTRACT

Platinum(II) anticancer drug cisplatin is one of the most important chemotherapeutic agents in clinical use but is limited by its high toxicity and severe side effects. Platinum(IV) anticancer prodrugs can overcome these limitations by resisting premature aquation and binding to essential plasma proteins. Structure-activity relationship studies revealed a link between the efficacy of platinum(IV) complexes with the nature of their axial ligands, which can be modified to enhance the properties of the prodrug. The existing paradigm of employing platinum(IV) complexes with symmetrical axial carboxylate ligands does not fully exploit their vast potential. A new approach was conceived to control properties of platinum(IV) prodrugs using contrasting axial ligands via sequential acylation. We report a novel class of asymmetric platinum(IV) carboxylates based on the cisplatin template containing both hydrophilic and lipophilic ligands on the same scaffold designed to improve their aqueous properties and enhance their efficacy against cancer cells in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Platinum/pharmacology , Acylation , Antineoplastic Agents/chemistry , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Models, Molecular , Platinum/chemistry , Solubility , Spectrometry, Mass, Electrospray Ionization
10.
Curr Top Med Chem ; 11(21): 2602-12, 2011.
Article in English | MEDLINE | ID: mdl-22039869

ABSTRACT

Over the past four decades, the search for improved platinum drugs based on the classical platinum (II)-diam(m)ine pharmacophore has yielded only a handful of successful candidates. New methodologies centred on platinum (IV) complexes, with better stability and expanded coordination spheres, offer the possibility of overcoming limitations inherent to platinum (II) drugs. In this review, novel strategies of targeting and killing cancer cells using platinum (IV) constructs are discussed. These approaches exploit the unique electrochemical characteristics and structural attributes of platinum (IV) complexes as a means of developing anticancer prodrugs that can target and selectively destroy cancer cells. Anticancer platinum (IV) prodrugs represent promising new strategies as targeted chemotherapeutic agents in the ongoing battle against cancer.


Subject(s)
Antineoplastic Agents/chemistry , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Organoplatinum Compounds/chemistry , Platinum/chemistry , Prodrugs/chemistry , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Humans , Nanotubes, Carbon/chemistry , Neoplasms/pathology , Organoplatinum Compounds/therapeutic use , Phototherapy , Prodrugs/therapeutic use
11.
Dalton Trans ; 40(44): 11690-2, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-21987084

ABSTRACT

Bridge cleavage reactions of the dimeric monocarbene complex [PdBr(2)((i)Pr(2)-bimy)](2) can be effectively used to end-cap pyridine containing pseudorotaxanes affording stable [2]rotaxanes.

SELECTION OF CITATIONS
SEARCH DETAIL
...