Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 177
1.
Sci Rep ; 12(1): 18461, 2022 11 02.
Article En | MEDLINE | ID: mdl-36323739

DNA polymerase η (polη) is of significant value for designing new families of anticancer drugs. This protein takes a role in many stages of the cell cycle, including DNA replication, translesion DNA synthesis, and the repairing process of DNA. According to many studies, a high level of expression of polη in most cases has been associated with low rates of patients' survival, regardless of considering the stage of tumor cells. Thus, the design of new drugs with fewer side effects to inhibit polη in cancerous cells has attracted attention in recent years. This project aims to design and explore the alternative inhibitors for polη, which are based on carbohydrates and amino acids. In terms of physicochemical properties, they are similar to the traditional anticancer drugs such as Cytarabine (cytosine arabinose). These alternative inhibitors are supposed to disrupt the DNA replication process in cancerous cells and prevent the tumor cells from mitosis. These newly designed structures, which are based on natural products, are expected to be non-toxic and to have the same chemotherapeutic impact as the traditional agents. The combinatorial use of quantum mechanics studies and molecular dynamic simulation has enabled us to precisely predict the inhibition mechanism of the newly designed structure, which is based on carbohydrates and amino acids, and compare it with that of the traditional chemotherapeutic drugs such as Cytarabine. Our results suggest that the inhibitors containing the natural building blocks of amino acid and carbohydrate could be considered alternative drugs for Cytarabine to block polη.


Amino Acids , Antineoplastic Agents , Humans , Amino Acids/genetics , DNA Replication , Cytarabine/pharmacology , Cytarabine/chemistry , Antineoplastic Agents/pharmacology , DNA Damage , DNA/metabolism , Carbohydrates
2.
J Mater Chem B ; 9(26): 5245-5254, 2021 07 07.
Article En | MEDLINE | ID: mdl-34095945

Multifunctional drug delivery systems combining two or more therapies have a wide-range of potential for high efficacy tumor treatment. Herein, we designed a novel hollow mesoporous Prussian blue nanoparticles (HMPBs)-based platform for targeted and synergetic chemo-photothermal treatment of acute myeloid leukemia (AML). The HMPBs were first loaded with the anticancer drugs daunorubicin (DNR) and cytarabine (AraC), and were subsequently coated with polyethylenimine (PEI) through electrostatic adsorption. Then, zwitterionic sulfobetaine (ZS) and CXCR4 antagonist peptide E5 were modified onto the surface of the nanoparticles via covalent bonding to fabricate a nanoplatform (denoted as HMPBs(DNR + AraC)@PEI-ZS-E5). The nanoplatform showed excellent photothermal effects, superior photothermal stability, reduced nonspecific protein adsorption, efficient targeting capability, a constant hydrodynamic diameter and good biocompatibility. Additionally, a laser-responsive drug release pattern was observed. In vitro results indicated that the nanoplatform could achieve active targeting and remarkable chemo-photothermal synergetic therapeutic effects, showcasing its great potential in AML treatment.


Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Daunorubicin/pharmacology , Ferrocyanides/chemistry , Leukemia, Myeloid, Acute/drug therapy , Nanoparticles/chemistry , Photothermal Therapy , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytarabine/chemistry , Daunorubicin/chemistry , Drug Delivery Systems , Drug Screening Assays, Antitumor , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/pathology , Particle Size , Polyethyleneimine/chemistry , Porosity , Surface Properties
3.
Food Chem Toxicol ; 151: 112101, 2021 May.
Article En | MEDLINE | ID: mdl-33684518

Diosgenin (DG) isolated from yam roots revealed various bioactivities and applications as drug carrier. In the present study, a conjugate of DG with cytarabine (Ara-C) was used to prepare the self-assembled nanoparticles (NPs) of DG-Ara-C by a nanoprecipitation method. Dynamic light scattering (DLS) as well as transmission electron microscopy (TEM) were employed to analyze the size and the morphology of NPs, respectively. The stability and absorption of DG-Ara-C NPs were measured. Additionally, the cytotoxicity of the NPs was determined via MTT assay. The results indicated that the average particle size of DG-Ara-C NPs was around 190 nm with a narrow size distribution (PDI = 0.1). TEM showed that DG-Ara-C NPs had a spherical morphology. Compared to free DG or Ara-C, the self-assembled DG-Ara-C NPs exhibited a better anti-tumor activity against solid tumor cells as well as leukemia cells. In conclusion, DG possesses dual role in the self-assembled NPs of DG-Ara-C conjugate, being as a promising anticancer drug and drug carrier.


Antimetabolites, Antineoplastic/chemistry , Cell Survival/drug effects , Cytarabine/chemistry , Diosgenin/chemistry , Nanoparticles/chemistry , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cytarabine/administration & dosage , Cytarabine/pharmacology , Diosgenin/administration & dosage , Diosgenin/pharmacology , Drug Carriers , Drug Screening Assays, Antitumor , Humans
4.
J Oncol Pharm Pract ; 27(4): 827-833, 2021 Jun.
Article En | MEDLINE | ID: mdl-32605496

BACKGROUND: Cytarabine is widely used to treat leukemia and lymphoma. Currently, Cyrabol®, powder for injection, is one of the specialties marketed in Tunisia. However, no stability data when diluted with 0.9% NaCl are available. The aim of this study is to evaluate the physical and chemical stability of cytarabine (Cyrabol®) solution after dilution in 0.9% NaCl (1 mg/mL, 5 mg/mL and 10 mg/mL) in polypropylene syringes under different storage conditions. METHODS: Cytarabine solutions (1 mg/mL, 5 mg/mL and 10 mg/mL) in 0.9% NaCl were prepared in polypropylene syringes and stored for 28 days under different conditions. Cytarabine preparations in glass containers were prepared as a control to detect any adsorption. Chemical stability was assessed by a stability-indicating high-performance liquid chromatography method. The stability-indicating capacity of the method was proved by forced degradation tests. Linearity, precision and limit of detection and quantification were performed according to the International Conference on Harmonisation recommendations. Physical stability was checked by visual inspection. RESULTS: The method was proven to be a validated stability-indicating assay. At 2-8°C, all tested solutions were chemically stable for 28 days. However, at 25°C, the main degradation product gradually increased during the study and the chemical stability of 1 mg/mL, 5 mg/mL and 10 mg/mL solutions was 14 days, 8 days and 5 days, respectively. Similar results were observed in the glass containers. CONCLUSION: The highest physical and chemical stability of cytarabine diluted in 0.9% NaCl in polypropylene syringes was observed at 2-8°C. At 25°C, better stability was found in the 1 mg/mL solution compared with those at higher concentrations (5 mg/mL and 10 mg/mL).


Antimetabolites, Antineoplastic/chemistry , Cytarabine/chemistry , Chromatography, High Pressure Liquid , Drug Compounding , Drug Stability , Drug Storage , Glass , Limit of Detection , Pharmaceutical Solutions , Polypropylenes , Reproducibility of Results , Saline Solution , Syringes , Temperature
5.
Cancer Chemother Pharmacol ; 86(5): 687-691, 2020 11.
Article En | MEDLINE | ID: mdl-32990804

PURPOSE: Cytarabine, a key chemotherapy agent for acute myeloid leukemia (AML) treatment, is deaminated into inactive uracil-arabinoside by cytidine deaminase. This deamination leads to samples stability issues with respect to clinical pharmacokinetic trials. The aim of our study was to study in vitro cytarabine stability in blood samples obtained from AML patients. METHODS: Cytarabine quantification was performed using a fully validated LC/MS/MS method. In vitro cytarabine stability was assessed at room temperature over 24 h in samples coming from 14 AML patients and 7 control patients (CTRL) with no hematological malignancy. In vitro concentrations versus time data were analyzed using a noncompartmental approach. RESULTS: Cytarabine in vitro area under the curve (AUCIVlast) was 22-fold higher in AML samples as compared to CTRL samples (AML mean (standard deviation (SD)), 51,829 (27,004) h ng/mL; CTRL mean (SD), 2356 (1250) h ng/mL, p = 0.00019). This increase was associated with a prolonged in vitro degradation half-life (t1/2IVdeg AML mean (SD), 15 (11.8) h; CTRL mean (SD), 0.36 (0.37) h, p = 0.0033). Multiple linear regression analysis showed that AML diagnosis significantly influenced t1/2IVdeg and AUCIVlas relationship. CONCLUSION: Cytarabine stability is higher in AML than in CTRL samples. The absence of correlation between t1/2IVdeg and AUCIVlast in AML samples suggests that in vitro cytarabine degradation in AML is complex. These results open perspectives including the evaluation of the clinical relevance and the involved molecular mechanisms.


Antimetabolites, Antineoplastic/blood , Cytarabine/blood , Cytidine Deaminase/metabolism , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Adult , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/isolation & purification , Case-Control Studies , Chromatography, High Pressure Liquid , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Cytarabine/administration & dosage , Cytarabine/chemistry , Cytarabine/isolation & purification , Cytidine Deaminase/isolation & purification , Deamination , Drug Stability , Female , Half-Life , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Randomized Controlled Trials as Topic , Specimen Handling , Tandem Mass Spectrometry , Time Factors , Young Adult
6.
Comput Biol Chem ; 88: 107334, 2020 Oct.
Article En | MEDLINE | ID: mdl-32759050

Development in two-dimensional (2D) drug-delivery materials have quickly translated into biological and pharmacological fields. In this present work, pristine graphene (PG) and hexagonal boron nitride (h-BN) sheets are explored as a drug carrier for cytarabine (CYT) and clofarabine (CLF) anti-cancer drugs using density functional theory (DFT). The obtained geometrical, energetic and electronic properties revealed that the PG sheet is more reactive and it adsorbs CYT and CLF anti-cancer drugs better than the h-BN sheet. The adsorption energies of CYT and CLF on PG sheet is -24.293 and -23.308 kcal/mol respectively, this is due to the delocalized electrons present in the PG sheet. The flow of electron direction between anti-cancer drugs and 2D sheet are calculated by ΔN, ΔEA(B), and ΔEB(A) parameters and Natural bond orbital analysis (NBO). The electronic and optical properties are calculated to understand the chemical reactivity and stability of the complex systems. The obtained results exhibit that the PG sheet retains significant therapeutic potential as a drug delivery vehicle for a drug molecule to treat cancer therapy.


Antimetabolites, Antineoplastic/chemistry , Boron Compounds/chemistry , Clofarabine/chemistry , Cytarabine/chemistry , Density Functional Theory , Graphite/chemistry , Nanoparticles/chemistry , Drug Carriers/chemistry , Humans
7.
G Ital Med Lav Ergon ; 42(2): 109-120, 2020 06.
Article En | MEDLINE | ID: mdl-32614541

SUMMARY: Antineoplastic drugs are used to treat cancer, having their therapeutic effect by inhibiting the cell division process. Although cancer cells, due to their rapid growth, are more sensitive to the toxic effects of chemotherapeutic agents, healthy cells and tissues may also be damaged. Many studies show acute and chronic toxicity both in patients treated with chemotherapy and in exposed workers. In fact, exposure to these substances can also be linked to the formation of different types of secondary tumors. The International Agency on Research on Cancer (IARC) included some antineplastic drugs in Group 1 (carcinogenic to humans), in Group 2A (probable carcinogens for In recent years, many studies have evidenced the presence of antineoplastic drug contamination on work surfaces, materials and floors and based on these observations, international and national guidelines have been published to limit occupational exposure, with particular attention to procedures post-preparation of chemotherapy to limit as much as possible the accumulation of contaminated residues. The aim of the following study is to determine the effectiveness of the degradation of four antineoplastic drugs: 5-fluorouracil, azacitidine, cytarabine and irinotecan using a low concentration of sodium hypochlorite solution (0.115%). The analytical platform used to monitor the degradation course of the substances under examination was hydrogen nuclear magnetic spectroscopy (1H NMR). In the same experimental conditions the effectiveness of the degradation of the same antineoplastic drugs with a 99.9% ethanol solution was also evaluated. The study showed that the best degradation efficiency (> 90% ) is obtained with the hypochlorite solution after 15 minutes.


Antineoplastic Agents/chemistry , Carcinogens/chemistry , Magnetic Resonance Spectroscopy/methods , Sodium Hypochlorite/pharmacology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/classification , Azacitidine/chemistry , Carcinogens/classification , Cytarabine/chemistry , Decontamination/methods , Drug Interactions , Ethanol/pharmacology , Fluorouracil/chemistry , Humans , Hypochlorous Acid/pharmacology , Irinotecan/chemistry , Occupational Exposure/adverse effects , Occupational Exposure/prevention & control , Preliminary Data , Sodium Chloride/pharmacology , Time Factors
8.
Mater Sci Eng C Mater Biol Appl ; 112: 110888, 2020 Jul.
Article En | MEDLINE | ID: mdl-32409046

Graphene quantum dots (GQDs) were prepared by the pyrolysis of citric acid (CA), which were used for the loading of hydrophilic cytarabine (Cyt), an anti-cancer drug, and then wrapped with chitosan (CS) gels for the encapsulation of the loaded Cyt. The fluorescent stability of GQDs was significantly enhanced in the presence of CS, which might be attributed to the inhibited agglomeration of GQDs by the CS gels. In addition, the burst release of Cyt from the developed carrier was also effectively relieved by the CS coating. Since the incorporation of Cyt into GQDs was achieved by amidation reaction, the delivery of Cyt from the carrier was pH-sensitive due to the hydrolysis of the amido linkage between GQDs and Cyt in acidic medium.


Antineoplastic Agents/chemistry , Chitosan/chemistry , Cytarabine/chemistry , Drug Carriers/chemistry , Graphite/chemistry , Quantum Dots/chemistry , Antineoplastic Agents/metabolism , Cytarabine/metabolism , Drug Liberation , Fluorescent Dyes/chemistry , Gels/chemistry , Hydrogen-Ion Concentration , Kinetics
9.
Int J Med Sci ; 17(6): 824-833, 2020.
Article En | MEDLINE | ID: mdl-32218704

Due to the high toxicity of currently used chemotherapeutics, novel methods of cancer treatment are needed. Gold nanoparticles (AuNPs) seem to be an interesting alternative due to penetration through biological membranes and systemic barriers. AuNPs as carriers of chemotherapeutics allow for reduced concentrations whilst maintaining the expected effect, and thus reducing the costs of therapy and adverse effects. We synthesized AuNPs stabilized with reduced glutathione (GSH) and conjugated with doxorubicin (DOX), gemcitabine (GEM) or cytarabine (CTA). This is the first study in which cytarabine-AuNPs were synthesized and characterized. Transmission electron microscopy (TEM), thermogravimetric analysis (TGA), nuclear magnetic resonance spectroscopy (NMR) and high-performance liquid chromatography (HPLC) were used to chemically characterize obtained nanoparticles. Antitumor activity and safety of application were assessed by MTT assay in in vitro model (human osteosarcoma cells -143B, human osteoblast- hFOB1.19, breast cancer cells - MCF7, breast epithelial cells - MCF10A, pancreatic cancer cells - PANC-1, and pancreatic cells - hTERT-HPNE cells). We have shown that cellular response varies according to the type and concentration of AuNPs. At some concentrations, we were able to show selective cytotoxicity of our AuNPs conjugates only to cancer cell lines. Synthesized nanoparticles were more cytotoxic to tumor cell lines than chemotherapeutics alone.


Glutathione/pharmacology , Gold/chemistry , Metal Nanoparticles/chemistry , Neoplasms/drug therapy , Chromatography, High Pressure Liquid , Cytarabine/chemistry , Cytarabine/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Glutathione/chemistry , Gold/adverse effects , Humans , MCF-7 Cells , Metal Nanoparticles/adverse effects , Microscopy, Electron, Transmission , Neoplasms/genetics , Neoplasms/pathology , Osteoblasts/drug effects , Telomerase/chemistry , Gemcitabine
10.
Cancer Chemother Pharmacol ; 85(4): 661-672, 2020 04.
Article En | MEDLINE | ID: mdl-32072218

PURPOSE: We postulate that the deoxyguanosine analogue CNDAG [9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)guanine] likely causes a single-strand break after incorporation into DNA, similar to the action of its cytosine congener CNDAC, and that subsequent DNA replication across the unrepaired nick would generate a double-strand break. This study aimed at identifying cellular responses and repair mechanisms for CNDAG prodrugs, 2-amino-9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)-6-methoxy purine (6-OMe) and 9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)-2,6-diaminopurine (6-NH2). Each compound is a substrate for adenosine deaminase, the action of which generates CNDAG. METHODS: Growth inhibition assay, clonogenic survival assay, immunoblotting, and cytogenetic analyses (chromosomal aberrations and sister chromatid exchanges) were used to investigate the impact of CNDAG on cell lines. RESULTS: The 6-NH2 derivative was selectively potent in T cell malignant cell lines. Both prodrugs caused increased phosphorylation of ATM and its downstream substrates Chk1, Chk2, SMC1, NBS1, and H2AX, indicating activation of ATM-dependent DNA damage response pathways. In contrast, there was no increase in phosphorylation of DNA-PKcs, which participates in repair of double-strand breaks by non-homologous end-joining. Deficiency in ATM, RAD51D, XRCC3, BRCA2, and XPF, but not DNA-PK or p53, conferred significant clonogenic sensitivity to CNDAG or the prodrugs. Moreover, hamster cells lacking XPF acquired remarkably more chromosomal aberrations after incubation for two cell cycle times with CNDAG 6-NH2, compared to the wild type. Furthermore, CNDAG 6-NH2 induced greater levels of sister chromatid exchanges in wild-type cells exposed for two cycles than those for one cycle, consistent with increased double-strand breaks after a second S phase. CONCLUSION: CNDAG-induced double-strand breaks are repaired mainly through homologous recombination.


Cytarabine/analogs & derivatives , DNA Damage/drug effects , DNA Repair/drug effects , Homologous Recombination/drug effects , Leukemia, T-Cell/drug therapy , Cell Survival , Cytarabine/chemistry , Cytarabine/pharmacology , Humans , Leukemia, T-Cell/pathology , Phosphorylation , Tumor Cells, Cultured
11.
Carbohydr Polym ; 234: 115901, 2020 Apr 15.
Article En | MEDLINE | ID: mdl-32070522

A therapeutic nanocarrier capable of cell targeting has the potential to reduce off-target effects of otherwise effective drugs. Nanoparticle surface modification can be tailored for specific cells, however multistep surface modification can prove slow and difficult for a variety of cell types. Here, we designed drug carrying polysaccharide based nanoparticles with a layered structure for clickable surface modification. The center of nanoparticle was composed of cationic macromer (e.g., poly-l-lysine) and anionic polysaccharide (e.g., heparin). Furthermore, a 'clickable' polysaccharide was installed on the surface of the nanoparticles to permit a wide range of bioconjugation via norbornene-tetrazine click chemistry. The utilities of these layered nanoparticles were demonstrated via enhanced protein sequestration, selective cell targeting (via PEGylation or altering polysaccharide coating), as well as loading and release of chemotherapeutic. The drug-loaded nanocarriers proved cytotoxic to J774A.1 monocytes and MOLM-14 leukemia cells.


Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Nanoparticles/chemistry , Polysaccharides/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Click Chemistry , Cytarabine/chemistry , Drug Carriers/chemistry , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Structure , Particle Size , Surface Properties
12.
Drug Dev Res ; 81(3): 274-282, 2020 05.
Article En | MEDLINE | ID: mdl-31643118

In view of the potent anticancer activity of the d-arabino-configured cytosine nucleoside (ara-C), apioarabinofuranosyl pyrimidine nucleosides were designed and synthesized from d-ribose as starting material. The synthetic strategy signifies that tosylation followed by in situ cyclization, one-pot controlled oxidative cleavage and acetylation by Pb(OAc)4 , stereoselective nucleobase condensation, inversion of hydroxyl group and uracil group converted to cytosine as the key steps. Synthesized apioarabinofuranosyl pyrimidine nucleosides were tested using breast, colon, and ovarian cancer cell lines. However, only compound 19a, 19b, and 22b have a moderate growth-suppressive effect against the luminal A breast cancer cell line MCF7.


Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Pyrimidines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cytarabine/analogs & derivatives , Cytarabine/chemistry , Female , Humans , MCF-7 Cells , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
13.
Anticancer Drugs ; 31(1): 67-72, 2020 01.
Article En | MEDLINE | ID: mdl-31633499

Cytarabine, the 4-amino-1-(ß-D-arabinofuranosyl)-2(1H)-pyrimidinone, (ARA-C) is an antimetabolite cytidine analogue used worldwide as key drug in the management of leukaemia. As specified in the manufacturers' instructions, once the components-sterile water and cytarabine powder-are unpackaged and mixed, the solution begins to degrade after 6 hours at room temperature and 12 hours at 4°C. To evaluate how to avoid wasting the drug in short-term, low-dose treatment regimens, the reconstituted samples, stored at 25°C and 4°C, were analyzed every day of the test week by reversed-phase HPLC and high-field NMR spectroscopy. All the samples remained unchanged for the entire week, which corresponds to the time required to administer the entire commercial drug package during low-dose therapeutic regimens. The drug solution was stored in a glass container at 4°C in an ordinary freezer and drawn with sterile plastic syringes; during this period, no bacterial or fungal contamination was observed. Our findings show that an cytarabine solution prepared and stored in the original vials retains its efficacy and safety and can, therefore, be divided into small doses to be administered over more days, thus avoiding unnecessary expensive and harmful waste of the drug preparation. Moreover, patients who require daily administration of the drug could undergo the infusion at home without need to go to hospital. The stability of the aliquots would help decrease hospitalization costs.


Cytarabine/chemistry , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/economics , Chromatography, High Pressure Liquid , Cost Savings , Cytarabine/administration & dosage , Cytarabine/economics , Drug Costs , Drug Stability , Drug Storage , Humans , Leukemia, Myeloid, Acute/drug therapy , Medication Adherence , Nuclear Magnetic Resonance, Biomolecular/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Solutions/chemistry
14.
Biomolecules ; 9(12)2019 11 23.
Article En | MEDLINE | ID: mdl-31771220

Cancer is the second leading cause of death worldwide. The main modality to fight against cancer is surgery, radiotherapy, and chemotherapy, and more recently targeted therapy, gene therapy and immunotherapy, which play important roles in treating cancer patients. In the last decades, chemotherapy has been well developed. Nonetheless, administration of the drug is not always successful, as limited drug dosage can reach the tumor cells.. In this context, the possibility to use an encapsulated anti-cancer drug may potentially solve the problem. Liposomal cytarabine is a formulation with pronounced effectiveness in lymphomatous meningitis and reduced cardiotoxicity if compared to liposomal anthracyclines. Thus, the future liposomal cytarabine use could be extended to other diseases given its reduction in cytotoxic side effects compared to the free formulation. This review summarizes the chemistry and biology of liposomal cytarabine, with exploration of its clinical implications.


Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cytarabine/administration & dosage , Cytarabine/chemistry , Liposomes/chemistry , Neoplasms/drug therapy , Animals , Drug Compounding , Humans
15.
J Biol Chem ; 294(50): 19048-19054, 2019 12 13.
Article En | MEDLINE | ID: mdl-31685662

Cytarabine (AraC) is the mainstay for the treatment of acute myeloid leukemia. Although complete remission is observed in a large proportion of patients, relapse occurs in almost all the cases. The chemotherapeutic action of AraC derives from its ability to inhibit DNA synthesis by the replicative polymerases (Pols); the replicative Pols can insert AraCTP at the 3' terminus of the nascent DNA strand, but they are blocked at extending synthesis from AraC. By extending synthesis from the 3'-terminal AraC and by replicating through AraC that becomes incorporated into DNA, translesion synthesis (TLS) DNA Pols could reduce the effectiveness of AraC in chemotherapy. Here we identify the TLS Pols required for replicating through the AraC templating residue and determine their error-proneness. We provide evidence that TLS makes a consequential contribution to the replication of AraC-damaged DNA; that TLS through AraC is conducted by three different pathways dependent upon Polη, Polι, and Polν, respectively; and that TLS by all these Pols incurs considerable mutagenesis. The prominent role of TLS in promoting proficient and mutagenic replication through AraC suggests that TLS inhibition in acute myeloid leukemia patients would increase the effectiveness of AraC chemotherapy; and by reducing mutation formation, TLS inhibition may dampen the emergence of drug-resistant tumors and thereby the high incidence of relapse in AraC-treated patients.


Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , DNA Replication/drug effects , DNA-Directed DNA Polymerase/biosynthesis , Leukemia, Myeloid, Acute/drug therapy , Antimetabolites, Antineoplastic/chemistry , Cytarabine/chemistry , DNA Replication/genetics , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/drug effects , Humans , Leukemia, Myeloid, Acute/metabolism , Nucleic Acid Conformation
16.
Sci Rep ; 9(1): 16400, 2019 11 08.
Article En | MEDLINE | ID: mdl-31704958

Cytarabine (AraC) is the mainstay chemotherapy for acute myeloid leukemia (AML). Whereas initial treatment with AraC is usually successful, most AML patients tend to relapse, and AraC treatment-induced mutagenesis may contribute to the development of chemo-resistant leukemic clones. We show here that whereas the high-fidelity replicative polymerase Polδ is blocked in the replication of AraC, the lower-fidelity translesion DNA synthesis (TLS) polymerase Polη is proficient, inserting both correct and incorrect nucleotides opposite a template AraC base. Furthermore, we present high-resolution crystal structures of human Polη with a template AraC residue positioned opposite correct (G) and incorrect (A) incoming deoxynucleotides. We show that Polη can accommodate local perturbation caused by the AraC via specific hydrogen bonding and maintain a reaction-ready active site alignment for insertion of both correct and incorrect incoming nucleotides. Taken together, the structures provide a novel basis for the ability of Polη to promote AraC induced mutagenesis in relapsed AML patients.


Cytarabine/pharmacology , DNA Polymerase II/chemistry , DNA Polymerase II/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytarabine/analogs & derivatives , Cytarabine/chemistry , DNA Replication/drug effects , Humans , Models, Molecular , Molecular Structure , Protein Conformation
17.
Curr Med Chem ; 26(28): 5278-5292, 2019.
Article En | MEDLINE | ID: mdl-31099318

Acute Myeloid Leukaemia (AML) is the neoplastic transformation of Hematopoietic Stem Cells (HSC) and relapsed disease is a major challenge in the treatment. Despite technological advances in the field of medicine and our heightened knowledge regarding the pathogenesis of AML, the initial therapy of "7+3" Cytarabine and Daunorubicin has remained mainly unchanged since 1973. AML is a disease of the elderly, and increased morbidity in this patient group does not allow the full use of the treatment and drug-resistant relapse is common. Nanocarriers are drug-delivery systems that can be used to transport drugs to the bone marrow and target Leukemic Stem Cells (LSC), conferring less side-effects compared to the free-drug alternative. Nanocarriers also can be used to favour the transport of drugs that otherwise would not have been used clinically due to toxicity and poor efficacy. Liposomes are a type of nanocarrier that can be used as a dedicated drug delivery system, which can also have active ligands on the surface in order to interact with antigens on the target cells or tissues. In addition to using small molecules, it is possible to attach antibodies to the liposome surface, generating so-called immunoliposomes. By using immunoliposomes as a drug-delivery system, it is possible to minimize the toxic side effects caused by the chemotherapeutic drug on healthy organs, and at the same time direct the drugs towards the remaining AML blasts and stem cells. This article aims to explore the possibilities of using immunoliposomes as a drug carrier in AML therapy. Emphasis will be on possible target molecules on the AML cells, leukaemic stem cells, as well as bone marrow constituents relevant to AML therapy. Further, some conditions and precautions that must be met for immunoliposomes to be used in AML therapy will be discussed.


Antineoplastic Agents/therapeutic use , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Drug Carriers/chemistry , Leukemia, Myeloid, Acute/drug therapy , Animals , Antineoplastic Agents/chemistry , Cytarabine/chemistry , Daunorubicin/chemistry , Drug Screening Assays, Antitumor , Humans , Leukemia, Myeloid, Acute/pathology , Liposomes/chemistry , Nanoparticles/chemistry
18.
Acta Biomater ; 94: 306-319, 2019 08.
Article En | MEDLINE | ID: mdl-30836199

Brain access remains a major challenge in drug testing. The nearly 'impermeable' blood-brain-barrier (BBB) prevents most drugs from gaining access to brain cells via systematic intravenous (IV) injection. In this study, silk fibroin films were used as drug carrier as well as cell culture substrate to simulate the in vivo interface between drug reservoir and brain cells for testing drug delivery in the brain. In in vitro studies, film-released arabinofuranosyl cytidine (AraC), a mitotic inhibitor, selectively killed glial cells in film-supported mixed neural cell cultures; with widened dosage windows for drug efficacy and tolerance compared to drugs in solution. In the brain, the presence of silk films was well tolerated with no signs of acute neuroinflammation, cell death, or altered brain function. Topical application of silk films on the cortical surface delivered Evans blue, a BBB-impenetrable fluorescent marker, through the intact dura matter into the parenchyma of the ipsilateral hemisphere as deep as the hippocampal region, but not the contralateral hemisphere. In a mouse traumatic brain injury (TBI) model, necrosis markers by film delivery accessed more cells in the lesion core than by con-current IV delivery; whereas the total coverage including the peri-lesional area appeared to be comparable between the two routes. The complementary distribution patterns of co-delivered markers provided direct evidence of the partial confinement of either route's access to brain cells by a restrictive zone near the lesion border. Finally, film-delivered necrostatin-1 reduced overall cell necrosis by approximately 40% in the TBI model. These findings from representative small molecules of delivery route-dependent drug access are broadly applicable for evaluating drug actions both in vitro and in vivo. Combined with its demonstrated role of supporting neuron-electrode interfaces, the film system can be further developed for testing a range of neuromodulation approaches (i.e., drug delivery, electrical stimulation, cell graft) in the brain. STATEMENT OF SIGNIFICANCE: This study demonstrated that silk fibroin films can be used to evaluate drug actions both in vitro and in vivo, partially overcoming the significant delivery barriers of the brain. This system can be adapted for efficient drug access to specific brain regions and/or cell types. The film system can be further developed for testing a range of interventions with drugs, electrical signals or cell graft for analysis of treatment outcomes including cell responses and brain function.


Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic , Hippocampus/metabolism , Membranes, Artificial , Neuroglia/metabolism , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cytarabine/chemistry , Cytarabine/pharmacokinetics , Cytarabine/pharmacology , Drug Implants/chemistry , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Rats
19.
ACS Chem Biol ; 14(3): 348-355, 2019 03 15.
Article En | MEDLINE | ID: mdl-30763062

Cancer therapies are plagued by resistance. Previously, we discovered a novel form of cancer drug resistance where the Glioma-associated protein 1 (GLI1) elevates UGT1A glucuronidation enzymes, thereby glucuronidating cytarabine and ribavirin, leading to resistance in leukemia patients. Here, we demonstrate that GLI1 imparts resistance to ∼40 compounds, including FDA-approved drugs with disparate chemotypes ( e.g., methotrexate and venetoclax). GLI1 indirectly elevates UGT1As via the chaperone calreticulin, which is required for resistance. Further, we demonstrate that resistant cells are more sensitive to ATP inhibitors, suggesting an Achilles' heel, which could be exploited in the future. In all, we identify GLI1-inducible glucuronidation as a broad-spectrum multidrug resistance pathway.


Antimetabolites, Antineoplastic/chemistry , Cytarabine/chemistry , Glucuronosyltransferase/metabolism , Small Molecule Libraries/chemistry , Zinc Finger Protein GLI1/antagonists & inhibitors , Adenosine Triphosphate/antagonists & inhibitors , Antimetabolites, Antineoplastic/metabolism , Calreticulin/metabolism , Cytarabine/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Small Molecule Libraries/metabolism
20.
Mol Cancer Ther ; 18(3): 567-578, 2019 03.
Article En | MEDLINE | ID: mdl-30679386

More than 40 years ago, the present standard induction therapy for acute myeloid leukemia (AML) was developed. This consists of the metabolic inhibitor cytarabine (AraC) and the cytostatic topoisomerase 2 inhibitor daunorubucin (DNR). In light of the high chance for relapse, as well as the large heterogeneity, novel therapies are needed to improve patient outcome. We have tested the anti-AML activity of 15 novel compounds based on the scaffolds pyrrolo[2,3-a]carbazole-3-carbaldehyde, pyrazolo[3,4-c]carbazole, pyrazolo[4,3-a]phenanthridine, or pyrrolo[2,3-g]indazole. The compounds were inhibitors of Pim kinases, but could also have inhibitory activity against other protein kinases. Ser/Thr kinases like the Pim kinases have been identified as potential drug targets for AML therapy. The compound VS-II-173 induced AML cell death with EC50 below 5 µmol/L, and was 10 times less potent against nonmalignant cells. It perturbed Pim-kinase-mediated AML cell signaling, such as attenuation of Stat5 or MDM2 phosphorylation, and synergized with DNR to induce AML cell death. VS-II-173 induced cell death also in patients with AML blasts, including blast carrying high-risk FLT3-ITD mutations. Mutation of nucleophosmin-1 was associated with good response to VS-II-173. In conclusion new scaffolds for potential AML drugs have been explored. The selective activity toward patient AML blasts and AML cell lines of the pyrazolo-analogue VS-II-173 make it a promising drug candidate to be further tested in preclinical animal models for AML.


Carbazoles/chemistry , Indazoles/chemistry , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytarabine/chemistry , Cytarabine/pharmacology , Humans , Indazoles/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics , Signal Transduction/drug effects , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology
...