Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Neurotoxicology ; 86: 162-165, 2021 09.
Article in English | MEDLINE | ID: mdl-34363843

ABSTRACT

The persisting need for effective clinical treatment of chemotherapy-induced neurotoxicity (CIN) motivates critical evaluation of preclinical models of CIN for their translational relevance. The present study aimed to provide the first quantitative evaluation of neural tissue exposed in vivo to a platinum-based anticancer compound, oxaliplatin (OX) during and after two commonly used dosing regimens: slow IV infusion used clinically and bolus IP injection used preclinically. Inductively-coupled plasma mass spectrometry analysis of dorsal root ganglia indicated that while differences in the temporal dynamics of platinum distribution exist, key drivers of neurotoxicity, e.g. peak concentrations and exposure, were not different across the two routes of administration. We conclude that the IP route of OX administration achieves clinically relevant pharmacokinetic exposure of neural tissues in a rodent model of CIN.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Oxaliplatin/administration & dosage , Oxaliplatin/pharmacokinetics , Administration, Intravenous , Animals , Drug Administration Routes , Infusions, Parenteral , Platinum Compounds/administration & dosage , Platinum Compounds/pharmacokinetics , Rats , Rats, Inbred F344
2.
Biochem Pharmacol ; 177: 113959, 2020 07.
Article in English | MEDLINE | ID: mdl-32272110

ABSTRACT

Breast cancer is the most prevalent type of tumor and the second leading cause of death due to cancer among women. Although screening methods, diagnosis and therapeutic options have improved in the last decade, chemoresistance remains an important challenge. There is evidence relating breast cancer resistance with signaling pathways involving hormone and growth receptors, survival, apoptosis and the activation of efflux pumps. However, the resistance mechanisms linked to drug uptake are poorly understood, despite it often being observed that the drug content is lower in resistant cancer cells and that the entry of the drug into these cells is a limiting process for the subsequent therapeutic effect.In this review, we provide an overview of drug uptake-based resistance mechanisms developed by cancer cells in the four main types of chemotherapy used in breast cancer: anthracyclines, taxanes, oxazaphosphorines and platinum-based drugs. The contribution of tumor microenvironment to reduced drug-uptake and multidrug resistance is also analyzed. As a developing field, nanomedicine-based approaches provide promising opportunities to improve drug specific targeting, cell interaction and uptake into cancer cells. The endocytic-mediated pathways attributed to the different types of nanoformulations as well as the contribution of nanotherapeutics to overcoming chemoresistance affecting drug uptake in breast cancer will be described. New approaches focusing on drug uptake mechanisms could improve breast cancer chemotherapy, obtaining better dose-response outcomes and reducing toxic side effects.


Subject(s)
Anthracyclines/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Oxazines/therapeutic use , Platinum Compounds/therapeutic use , Taxoids/therapeutic use , Anthracyclines/pharmacokinetics , Breast Neoplasms/metabolism , Female , Humans , Nanomedicine/methods , Nanomedicine/trends , Oxazines/pharmacokinetics , Platinum Compounds/pharmacokinetics , Taxoids/pharmacokinetics , Tumor Microenvironment/drug effects
3.
Xenobiotica ; 50(8): 980-987, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32072840

ABSTRACT

LLC-1903, a novel anticancer compound, was synthesized by optimizing the structure, which was derived from altering the leaving group of lobaplatin. It has an excellent in vitro anti-cancer activity, high water solubility, high stability in solution and low in vivo toxicity according to our former study.The plasma pharmacokinetics (PK) and tissue distribution of LLC-1903 and lobaplatin in rats were determined after intravenous administration of a single dose (0.06 mmol/kg body weight). Inductively coupled plasma mass spectrometry (ICP-MS) was used to measure the concentration of platinum (Pt) in plasma and tissue samples.Most PK parameters of the Pt in LLC-1903 showed a significant difference from those of lobaplatin. The plasma level of LLC-1903 is only half of that of lobaplatin (p < 0.01) which could be the direct result of faster drug clearance. The tissue distribution showed that both LLC-1903 and lobaplatin were mainly found in the liver and kidney, and less in other organs. At four time points (0.083, 0.5, 1 and 4 h) after administration, the tissue concentrations of LLC-1903 were almost always significantly higher than those of lobaplatin (p < 0.05 or p < 0.01).


Subject(s)
Antineoplastic Agents/pharmacokinetics , Platinum Compounds/pharmacokinetics , Administration, Intravenous , Animals , Metabolic Clearance Rate , Tissue Distribution
4.
Radiol Oncol ; 53(2): 148-158, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30956230

ABSTRACT

Background Platinum-based anticancer drugs are widely used in the chemotherapy of human neoplasms. The major obstacle for the clinical use of this class of drugs is the development of resistance and toxicity. It is therefore very important to understand the chemical properties, transport and metabolic pathways and mechanism of actions of these compounds. There is a large body of evidence that therapeutic and toxic effects of platinum drugs on cells are not only a consequence of covalent adducts formation between platinum complexes and DNA but also with RNA and many proteins. These processes determine molecular mechanisms that underlie resistance to platinum drugs as well as their toxicity. Increased expression levels of various transporters and increased repair of platinum-DNA adducts are both considered as the most significant processes in the development of drug resistance. Functional genomics has an increasing role in predicting patients' responses to platinum drugs. Genetic polymorphisms affecting these processes may play an important role and constitute the basis for individualized approach to cancer therapy. Similar processes may also influence therapeutic potential of nonplatinum metal compounds with anticancer activity. Conclusions Cisplatin is the most frequently used platinum based chemotherapeutic agent that is clinically proven to combat different types of cancers and sarcomas.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/physiology , Neoplasms/drug therapy , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cisplatin/adverse effects , Cisplatin/chemistry , Cisplatin/pharmacokinetics , DNA Adducts/chemistry , DNA Repair , DNA, Neoplasm/drug effects , Drug Hypersensitivity/etiology , Drug Resistance, Neoplasm/genetics , Gold Compounds/pharmacology , Humans , Neoplasms/blood , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Palladium/pharmacology , Platinum Compounds/adverse effects , Platinum Compounds/pharmacokinetics , Platinum Compounds/pharmacology , Polymorphism, Genetic , Ruthenium/pharmacology
5.
J Pharmacol Exp Ther ; 363(2): 221-239, 2017 11.
Article in English | MEDLINE | ID: mdl-28916659

ABSTRACT

Two hybrids of Pt(IV) species were designed and prepared by addition of a chlorambucil unit to the axial positions of the Pt(IV) complexes derived from DN603 and DN604. In vitro studies of two hybrids against two pairs of cisplatin sensitive and resistant cancer cell lines indicated that compound 5 had superior antitumor activity to cisplatin and chlorambucil via suppressing DNA damage repair to reverse drug resistance. Mechanistic investigation suggested that the potent antitumor activity of compound 5 arose from its major suppression of CK2-mediated MRE11-RAD50-NBS1(MRN) complex promotion of DNA double-strand break (DSB) repair. In nude mice with A549/CDDP xenografts, compound 5 exhibited higher anticancer efficacy than cisplatin and chlorambucil by reversing drug resistance, displayed improved effectiveness, and had no toxicity effects. Overall, compound 5 is a promising drug candidate, which could promote the anticancer activity and reverse drug resistance by attenuating CK2-induced MRN-dependent DSB repair.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents/pharmacology , Chlorambucil/pharmacology , DNA, Neoplasm/drug effects , Drug Resistance, Neoplasm/drug effects , Platinum Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents, Alkylating/pharmacokinetics , Calcium/metabolism , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , Chlorambucil/pharmacokinetics , Cisplatin/pharmacology , DNA Repair/drug effects , Drug Design , Female , Humans , Male , Mice , Mice, Inbred BALB C , Platinum/metabolism , Platinum Compounds/pharmacokinetics , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
6.
Drug Resist Updat ; 27: 72-88, 2016 07.
Article in English | MEDLINE | ID: mdl-27449599

ABSTRACT

It is now widely accepted that organic anion-transporting polypeptides (OATPs), especially members of the OATP1A/1B family, can have a major impact on the disposition and elimination of a variety of endogenous molecules and drugs. Owing to their prominent expression in the sinusoidal plasma membrane of hepatocytes, OATP1B1 and OATP1B3 play key roles in the hepatic uptake and plasma clearance of a multitude of structurally diverse anti-cancer and other drugs. Here, we present a thorough assessment of the currently available OATP1A and OATP1B knockout and transgenic mouse models as key tools to study OATP functions in vivo. We discuss recent studies using these models demonstrating the importance of OATPs, primarily in the plasma and hepatic clearance of anticancer drugs such as taxanes, irinotecan/SN-38, methotrexate, doxorubicin, and platinum compounds. We further discuss recent work on OATP-mediated drug-drug interactions in these mouse models, as well as on the role of OATP1A/1B proteins in the phenomenon of hepatocyte hopping, an efficient and flexible way of liver detoxification for both endogenous and exogenous substrates. Interestingly, glucuronide conjugates of both the heme breakdown product bilirubin and the protein tyrosine kinase-targeted anticancer drug sorafenib are strongly affected by this process. The clinical relevance of variation in OATP1A/1B activity in patients has been previously revealed by the effects of polymorphic variants and drug-drug interactions on drug toxicity. The development of in vivo tools to study OATP1A/1B functions has greatly advanced our mechanistic understanding of their functional role in drug pharmacokinetics, and their implications for therapeutic efficacy and toxic side effects of anticancer and other drug treatments.


Subject(s)
Antineoplastic Agents/toxicity , Inactivation, Metabolic/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Neoplasms/metabolism , Organic Cation Transport Proteins/genetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Camptothecin/toxicity , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/toxicity , Drug Interactions , Gene Expression , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Irinotecan , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver-Specific Organic Anion Transporter 1/metabolism , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Methotrexate/toxicity , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Niacinamide/analogs & derivatives , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Niacinamide/toxicity , Organic Cation Transport Proteins/deficiency , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Phenylurea Compounds/toxicity , Platinum Compounds/pharmacokinetics , Platinum Compounds/pharmacology , Platinum Compounds/toxicity , Sorafenib , Taxoids/pharmacokinetics , Taxoids/pharmacology , Taxoids/toxicity
7.
Pharmacogenomics ; 17(12): 1365-78, 2016 08.
Article in English | MEDLINE | ID: mdl-27462924

ABSTRACT

Lung cancer is one of the leading causes of cancer-related death in the world. Platinum-based chemotherapy is the first-line treatment for non-small-cell lung cancer (NSCLC), however, the therapeutic efficiency varies remarkably among individuals. A large number of pharmacogenomics studies aimed to identify genetic variations which can be used to predict platinum response. Those studies are leading NSCLC treatment to the new era of precision medicine. In the current review, we provided a comprehensive update on the main recent findings of genetic variations which can be used to predict platinum sensitivity in the NSCLC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Pharmacogenetics , Platinum Compounds/therapeutic use , Precision Medicine , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Genome-Wide Association Study , Humans , Lung Neoplasms/metabolism , Platinum Compounds/pharmacokinetics
8.
Drug Dev Ind Pharm ; 42(11): 1792-9, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27042965

ABSTRACT

LLC-0601(S,S) and LLC-0601(R,R) are two novel synthesized isomerism platinum compounds both with encouraging anticancer activity. However, the previous study showed that toxicity of LLC-0601(R,R) was much higher than that of LLC-0601(S,S) with higher body weight loss and mortality rate of tested rats. This paper is focused on the comparison of the two compounds with their pharmacokinetic (PK) profiles in rats and tissue distribution in mice after intravenous administration. The atomic absorption spectrometry (AAS) method was successfully developed and applied for the determination of platinum in plasma and tissues. The results showed that main PK parameters such as half-life, AUC and MRT of the two compounds had no significant difference after intravenous administration to rats (p  > 0.05). The tissue distribution after intravenous administration to mice showed that the concentration of LLC-0601(R,R) in heart at 0.083 h was higher than that of LLC-0601(S,S) (p < 0.05) and it was the same case for AUC5min-4 h (p < 0.05). Different distribution of the two compounds in heart was possibly the main reason of different toxicity and more in-depth research on the metabolites and other mechanism are needed to investigate the toxicity.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Organoplatinum Compounds/pharmacokinetics , Platinum Compounds/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Isomerism , Mice , Organoplatinum Compounds/chemistry , Platinum Compounds/chemistry , Rats , Tissue Distribution
9.
Prog. obstet. ginecol. (Ed. impr.) ; 58(6): 269-274, jun.-jul. 2015. tab, graf
Article in English | IBECS | ID: ibc-139293

ABSTRACT

Background: Claudin-4, a component of the tight junction, plays an important role in tumorigenesis and metastasis of ovarian cancer, but its role in platinum resistance has not been elucidated. Aim of the work: To determine the presence of claudin-4 in ovarian cancer tissues in relation to platinum compounds resistance. Patients and methods: Patients with advanced ovarian malignancy (FIGO stages III and IV) that have undergone primary surgery for maximal cytoreduction, followed by first line chemotherapy with platinum compounds and paclitaxel, were followed up for 6 months to determine chemotherapeutic response. Claudin-4 expression in ovarian cancer tissue resected from the patients surgically was evaluated immunohistochemically. Results: Claudin-4 is associated with more aggressive behavior of ovarian tumors and the advanced stage of the tumors. High expression of claudin-4 was found in high grade tumors, of the papillary serous subtype. High expression is linked to chemotherapeutic resistance, whereas low expression is associated with good response to first line chemotherapy. Conclusion: High claudin-4 expression can predict poor chemotherapeutic response in advanced ovarian cancer (AU)


Antecedentes: La claudina-4, componente de la zona de oclusión, desempeña un papel importante en la oncogenia y metástasis del cáncer ovárico, pero su papel en la resistencia al platino no se ha aclarado todavía. Objetivo: Establecer la presencia de claudina-4 en los tejidos de cáncer ovárico con relación a la resistencia a compuestos de platino. Pacientes y métodos: Pacientes con neoplasia maligna de ovario (clasificación FIGO fase III y IV) sometidos a una intervención citorreductora primaria seguida de quimioterapia de primera línea con compuestos de platino y paclitaxel; se realizó un seguimiento durante 6 meses para valorar la reacción a la quimioterapia. Se llevó a cabo un análisis inmunohistoquímico de la expresión de la claudina-4 en los tejidos de cáncer ovárico. Resultados: La claudina-4 se asocia con un comportamiento más agresivo de los tumores ováricos y con una fase más avanzada de los mismos. Se encontró una alta manifestación de claudina-4 en tumores de grado alto, de subtipo papiloma seroso. Se relaciona la expresión alta con la resistencia quimioterápica, mientras que una expresión baja se asocia a una buena reacción a la quimioterapia de primera línea. Conclusión: Una expresión alta de claudina-4 puede predecir una mala respuesta quimioterápica en el cáncer de ovario avanzado (AU)


Subject(s)
Humans , Claudin-4/analysis , Ovarian Neoplasms/pathology , Platinum Compounds/pharmacokinetics , Paclitaxel/pharmacokinetics , Biomarkers, Tumor/analysis , Drug Resistance , Treatment Outcome
10.
J Trace Elem Med Biol ; 31: 178-82, 2015.
Article in English | MEDLINE | ID: mdl-24703712

ABSTRACT

Copper (Cu) is an essential metal for growth and development that has the potential to be toxic if levels accumulate beyond the ability of cells to homeostatically balance uptake with detoxification. One system for Cu acquisition is the integral membrane Cu(+) transporter, Ctr1, which has been quite well characterized in terms of its function and physiology. The mammalian Ctr2 protein has been a conundrum for the copper field, as it is structurally closely related to the high affinity Cu transporter Ctr1, sharing important motifs for Cu transport activity. However, in contrast to mammalian Ctr1, Ctr2 fails to suppress the Cu-dependent growth phenotype of yeast cells defective in Cu(+) import, nor does it appreciably stimulate Cu acquisition when over-expressed in mammalian cells, underscoring important functional dissimilarities between the two proteins. Several roles for the mammalian Ctr2 have been suggested both in vitro and in vivo. Here, we summarize and discuss current insights into the Ctr2 protein and its interaction with Ctr1, its functions in mammalian Cu homeostasis and platinum-based chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cation Transport Proteins/metabolism , Copper/metabolism , Models, Biological , Organoplatinum Compounds/pharmacokinetics , Platinum Compounds/pharmacokinetics , Amino Acid Motifs , Animals , Antineoplastic Agents/therapeutic use , Biological Transport , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Copper Transporter 1 , Gene Expression Regulation , Homeostasis , Humans , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Organoplatinum Compounds/therapeutic use , Platinum Compounds/therapeutic use , Protein Conformation , Protein Processing, Post-Translational , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , SLC31 Proteins
11.
J Inorg Biochem ; 140: 219-27, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25171667

ABSTRACT

The biological properties of a series of cisplatin-based Pt(IV) prodrug candidates, namely trans,cis,cis-[Pt(carboxylato)2Cl2(NH3)2], where carboxylato=CH3(CH2)nCOO(-) [(1), n=0; (2), n=2; (3), n=4; (4), n=6] having a large interval of lipophilicity are discussed. The stability of the complexes was tested in different pH conditions (i.e. from 1.0 to 9.0) to simulate the hypothetical conditions for an oral route of administration, showing a high stability (>90%). The transformation into their active Pt(II) metabolites was demonstrated in the presence of ascorbic acid, with a pseudo-first order kinetics, the half-time of which smoothly decreases as the chain length of carboxylic acid increases. Their antiproliferative activity has been evaluated in vitro on a large panel of human cancer cell lines. As expected, the potency increases with the chain length: 3 and 4 resulted by far more active than cisplatin on all cell lines of about one or two orders of magnitude, respectively. Both complexes retained their activity also on cisplatin-resistant cell line, and exhibited a progressive increase of the selectivity compared with non-tumor cells. These results were confirmed with more prolonged treatment (up to 14days) studied on multicellular tumor spheroids (MCTSs). In this case the Pt(IV) complexes exert a protracted antiproliferative action, even if the drug is removed from the culture medium. Finally, in a time-course experiment of the total platinum evaluation in mice blood (after a single oral administration of the title complexes), 2 gave the best results, representing a good compromise between lipophilicity and water solubility, that increase and decrease respectively on passing from 1 to 4.


Subject(s)
Antineoplastic Agents/pharmacology , Platinum Compounds/pharmacology , Prodrugs/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Humans , Oxidation-Reduction , Platinum Compounds/chemistry , Platinum Compounds/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics
12.
J Pharmacol Exp Ther ; 349(3): 458-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24696041

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) has been implicated as a hub for multiple oncogenic pathways. The constitutive activation of STAT3 is present in several cancers, including gliomas (GBMs), and is associated with poor therapeutic responses. Phosphorylation of STAT3 triggers its dimerization and nuclear transport, where it promotes the transcription of genes that stimulate tumor growth. In light of this role, inhibitors of the STAT3 pathway are attractive therapeutic targets for cancer. To this end, we evaluated the STAT3-inhibitory activities of three compounds (CPA-7 [trichloronitritodiammineplatinum(IV)], WP1066 [(S,E)-3-(6-bromopyridin-2-yl)-2-cyano-N-(1-phenylethyl)acrylamide, C17H14BrN3O], and ML116 [4-benzyl-1-{thieno[2,3-d]pyrimidin-4-yl}piperidine, C18H19N3S]) in cultured rodent and human glioma cells, including GBM cancer stem cells. Our results demonstrate a potent induction of growth arrest in GBM cells after drug treatment with a concomitant induction of cell death. Although these compounds were effective at inhibiting STAT3 phosphorylation, they also displayed variable dose-dependent inhibition of STAT1, STAT5, and nuclear factor κ light-chain enhancer of activated B cells. The therapeutic efficacy of these compounds was further evaluated in peripheral and intracranial mouse tumor models. Whereas CPA-7 elicited regression of peripheral tumors, both melanoma and GBM, its efficacy was not evident when the tumors were implanted within the brain. Our data suggest poor permeability of this compound to tumors located within the central nervous system. WP1066 and ML116 exhibited poor in vivo efficacy. In summary, CPA-7 constitutes a powerful anticancer agent in models of peripheral solid cancers. Our data strongly support further development of CPA-7-derived compounds with increased permeability to enhance their efficacy in primary and metastatic brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/secondary , STAT3 Transcription Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorine Compounds/pharmacokinetics , Chlorine Compounds/pharmacology , Chlorine Compounds/therapeutic use , Drug Screening Assays, Antitumor , Glioma/metabolism , Glioma/pathology , HEK293 Cells , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/pharmacology , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Piperidines/pharmacokinetics , Piperidines/pharmacology , Piperidines/therapeutic use , Platinum Compounds/pharmacokinetics , Platinum Compounds/pharmacology , Platinum Compounds/therapeutic use , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/therapeutic use , STAT3 Transcription Factor/genetics , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/therapeutic use , Tissue Distribution , Tyrphostins/pharmacokinetics , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
13.
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
14.
Adv Drug Deliv Rev ; 65(13-14): 1667-85, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24113520

ABSTRACT

Platinum based anticancer drugs have revolutionized cancer chemotherapy, and continue to be in widespread clinical use especially for management of tumors of the ovary, testes, and the head and neck. However, several dose limiting toxicities associated with platinum drug use, partial anti-tumor response in most patients, development of drug resistance, tumor relapse, and many other challenges have severely limited the patient quality of life. These limitations have motivated an extensive research effort towards development of new strategies for improving platinum therapy. Nanocarrier-based delivery of platinum compounds is one such area of intense research effort beginning to provide encouraging preclinical and clinical results and may allow the development of the next generation of platinum chemotherapy. This review highlights current understanding on the pharmacology and limitations of platinum compounds in clinical use, and provides a comprehensive analysis of various platinum-polymer complexes, micelles, dendrimers, liposomes and other nanoparticles currently under investigation for delivery of platinum drugs.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Drug Resistance, Neoplasm , Nanoparticles/chemistry , Organoplatinum Compounds/administration & dosage , Platinum Compounds/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Dose-Response Relationship, Drug , Drug Discovery , Humans , Molecular Structure , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/therapeutic use , Platinum Compounds/chemistry , Platinum Compounds/pharmacokinetics , Platinum Compounds/therapeutic use
15.
Expert Opin Drug Metab Toxicol ; 9(10): 1381-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23829480

ABSTRACT

INTRODUCTION: Picoplatin was developed as platinum coordination complex to overcome development of resistance, through conjugation to thioles, by the introduction of a methyl-pyridine moiety into the cisplatin parent structure. Pharmacokinetic parameters of the drug, after intravenous and oral application, were studied in solid tumors and clinical Phase I - III trials performed, in particular in NSCLC and small cell lung cancer (SCLC). Results showed low clinical activity of picoplatin. AREAS COVERED: This article presents an overview of the pharmacokinetic assessments of picoplatin in lung cancer. Specifically, the authors address the relationship between disposition and clinical activity of the drug. EXPERT OPINION: Picoplatin failed to overcome resistance to platinum compounds in lung cancer to achieve significant improved survival of most patients. Even highest doses of the drug reaching 150 m/m² given intravenously every 3 weeks were not sufficient to achieve better response than existing chemotherapeutics and the oral bioavailability of a dose of 200 - 400 mg corresponded only to 80 mg/m² iv. Picoplatin therefore seem to be quite ineffective. Picoplatin is expected to overcome tumor resistance in cases which overexpress thiol-conjugating pathways; however, this was not proved in clinical trials. To conclude, this blocked platinum complex is not able to reverse cisplatin resistance to a significant extent in vivo and its mechanisms and kinetics and of DNA damage failed to produce significant clinical results compared to second-line standard therapy for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Organoplatinum Compounds/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Humans , Organoplatinum Compounds/administration & dosage , Platinum Compounds/pharmacokinetics , Randomized Controlled Trials as Topic
16.
Drug Metab Pharmacokinet ; 28(1): 19-27, 2013.
Article in English | MEDLINE | ID: mdl-22986709

ABSTRACT

Several solute carriers and ATP-binding cassette transporters have been implicated in the influx or efflux of platinum-based chemotherapeutic agents such as cisplatin, carboplatin, and oxaliplatin. Given that many of these proteins are highly polymorphic, the genetic status of these proteins could be an important contributor to the extensive interindividual pharmacokinetic variability associated with the clinical use of these agents. In this review article, we provide an updated overview of the various transporters that have shown promise in animal models or patient populations in facilitating the movement of platinum-based agents across cell membranes, and how their function is associated with drug disposition or pharmacodynamic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Membrane Transport Proteins/genetics , Platinum Compounds/pharmacology , Platinum Compounds/pharmacokinetics , Animals , Cation Transport Proteins/genetics , Copper Transporter 1 , Humans , Multidrug Resistance-Associated Proteins/genetics , Organic Cation Transport Proteins/genetics , Polymorphism, Single Nucleotide
17.
Proc Natl Acad Sci U S A ; 109(30): 11987-92, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22773807

ABSTRACT

Monofunctional platinum(II) complexes of general formula cis-[Pt(NH(3))(2)(N-heterocycle)Cl]Cl bind DNA at a single site, inducing little distortion in the double helix. Despite this behavior, these compounds display significant antitumor properties, with a different spectrum of activity than that of classic bifunctional cross-linking agents like cisplatin. To discover the most potent monofunctional platinum(II) compounds, the N-heterocycle was systematically varied to generate a small library of new compounds, with guidance from the X-ray structure of RNA polymerase II (Pol II) stalled at a monofunctional pyriplatin-DNA adduct. In pyriplatin, the N-heterocycle is pyridine. The most effective complex evaluated was phenanthriplatin, cis-[Pt(NH(3))(2)(phenanthridine)Cl]NO(3), which exhibits significantly greater activity than the Food and Drug Administration-approved drugs cisplatin and oxaliplatin. Studies of phenanthriplatin in the National Cancer Institute 60-cell tumor panel screen revealed a spectrum of activity distinct from that of these clinically validated anticancer agents. The cellular uptake of phenanthriplatin is substantially greater than that of cisplatin and pyriplatin because of the hydrophobicity of the phenanthridine ligand. Phenanthriplatin binds more effectively to 5'-deoxyguanosine monophosphate than to N-acetyl methionine, whereas pyriplatin reacts equally well with both reagents. This chemistry supports DNA as a viable cellular target for phenanthriplatin and suggests that it may avoid cytoplasmic platinum scavengers with sulfur-donor ligands that convey drug resistance. With the use of globally platinated Gaussia luciferase vectors, we determined that phenanthriplatin inhibits transcription in live mammalian cells as effectively as cisplatin, despite its inability to form DNA cross-links.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Models, Molecular , Organoplatinum Compounds/pharmacology , Phenanthridines/pharmacology , Platinum Compounds/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Crystallography, X-Ray , Deoxyguanine Nucleotides/metabolism , Drug Discovery/methods , Genetic Vectors , Inhibitory Concentration 50 , Luciferases , Molecular Structure , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/metabolism , Organoplatinum Compounds/pharmacokinetics , Phenanthridines/chemistry , Phenanthridines/metabolism , Phenanthridines/pharmacokinetics , Platinum Compounds/chemistry , Platinum Compounds/metabolism , Platinum Compounds/pharmacokinetics
18.
Curr Cancer Drug Targets ; 12(8): 962-86, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22794121

ABSTRACT

Membrane transporters govern the movement of drugs and their metabolites across biological membranes, thereby determining their pharmacokinetics, efficacy and adverse drug reactions. Platinum-based anticancer drugs are a mainstay of chemotherapy for many human malignancies. However, their clinical utility is limited by tumor resistance and normal tissue toxicities, which are determined at least in part by the level of tissue accumulation of platinum. Recently, several members of the ATP-binding cassette (ABC), solute carrier (SLC) and ATPase membrane protein superfamilies have been found to contribute to the net accumulation of platinum drugs in malignant and normal tissues. Herein, a review has been carried out to critically evaluate current preclinical and clinical evidence implicating membrane transporters as determinants of the pharmacology of cisplatin, oxaliplatin, carboplatin and related investigational compounds. The evidence includes studies of recombinant cell systems with genetically modified expression of individual membrane transporters, platinum-resistant or -sensitive human cancer cells and in vivo xenografted tumors, animal models of platinum-induced nephro-, oto- or neurotoxicity, and clinical studies of associations between the membrane transporter tumor expression and patient outcomes from platinum-based chemotherapy. Understanding the role of membrane transporters as determinants of the pharmacology of platinum drugs will be a basis for targeting these drug transporters in individualized and optimized platinum-based cancer therapy, and new drug development.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Membrane Transport Proteins/metabolism , Platinum Compounds/pharmacology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cisplatin/pharmacology , Cisplatin/toxicity , Copper-Transporting ATPases , Humans , Membrane Transport Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Platinum/pharmacology , Platinum/therapeutic use , Platinum/toxicity , Platinum Compounds/pharmacokinetics , SLC31 Proteins , Treatment Outcome
19.
Expert Opin Drug Metab Toxicol ; 7(7): 919-28, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21599552

ABSTRACT

INTRODUCTION: Pemetrexed is a multi-targeted antifolate cytotoxic agent that has demonstrated activity in a number of very common cancer types including NSCLC in both first- and second-line settings and in the treatment of malignant mesothelioma. AREAS COVERED: This article focuses on all of the currently published pharmacokinetic data of pemetrexed reviewing a number of different scenarios and patient populations. All the articles reviewed in this manuscript are from peer-reviewed English-spoken literature without any limitations to the time of publication. EXPERT OPINION: Pemetrexed's clearance correlates with renal function and it may be safely used with vitamin supplementation in patients with creatinine clearance ≥ 45 ml/min. The pharmacokinetics of pemetrexed is also largely unchanged in third-space fluids and can be feasibly and safely administered in combination with several other cytotoxic or targeted agents. It is the author's opinion that pemetrexed is already a valuable cytotoxic agent which has proved useful in several malignancies. However, future trials might expand on the combined use of pemetrexed with other targeted agents that could be beneficial to other selected patients harboring relevant mutations or other biological features.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Folic Acid Antagonists/pharmacokinetics , Glutamates/pharmacokinetics , Guanine/analogs & derivatives , Mesothelioma/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Brain/drug effects , Disease Models, Animal , Drug Evaluation , Drug Interactions , Folic Acid Antagonists/administration & dosage , Glutamates/administration & dosage , Guanine/administration & dosage , Guanine/pharmacokinetics , Humans , Kidney Neoplasms/drug therapy , Liver/drug effects , Pemetrexed , Platinum Compounds/administration & dosage , Platinum Compounds/pharmacokinetics , Tissue Distribution , Vitamins/administration & dosage , Vitamins/pharmacokinetics
20.
Mol Cancer Ther ; 9(4): 1058-69, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20371711

ABSTRACT

Picoplatin, a third-generation platinum agent, is efficacious against lung cancers that are otherwise resistant or become refractory during platinum treatment. This effort was aimed at the determination of the influence of organic cation transporters 1, 2, and 3 (OCT1, OCT2, and OCT3) and their genetic variants on cellular uptake of picoplatin and on the individual components of the ensuing cytotoxicity such as DNA adduct formation. The effect of OCT1 on picoplatin pharmacokinetics and antitumor efficacy was determined using OCT knockout mice and HEK293 xenografts stably expressing OCT1. The uptake and DNA adduct formation of picoplatin were found to be significantly enhanced by the expression of the OCTs. Expression of OCT1 and OCT2, but not OCT3, significantly enhanced picoplatin cytotoxicity, which was reduced in the presence of an OCT inhibitor. Common reduced functional variants of OCT1 and OCT2 led to reduction in uptake and DNA adduct formation of picoplatin in comparison with the reference OCT1 and OCT2. Pharmacokinetic parameters of picoplatin in Oct1(-/-) and Oct1(+/+) mice were not significantly different, suggesting that the transporters do not influence the disposition of the drug. In contrast, the volume of OCT1-expressing xenografts in mice was significantly reduced by picoplatin treatment, suggesting that OCT1 may enhance the antitumor efficacy of picoplatin. These studies provide a basis for follow-up clinical studies that would seek to examine the relationship between the anticancer efficacy of picoplatin and expression levels of OCTs and their genetic variants in tumors. Mol Cancer Ther; 9(4); 1058-69. (c)2010 AACR.


Subject(s)
Organic Cation Transport Proteins/metabolism , Platinum Compounds/pharmacology , Platinum Compounds/pharmacokinetics , Animals , Cell Death/drug effects , Cell Line, Tumor , Cimetidine/pharmacology , DNA Adducts/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Tissue Distribution/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...