Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 306
Filter
1.
Adv Sci (Weinh) ; 9(6): e2102303, 2022 02.
Article in English | MEDLINE | ID: mdl-35023320

ABSTRACT

Diabetes is directly related to the risk of breast cancer (BC) occurrence and worsened BC prognosis. Currently, there are no specific treatments for diabetes-associated BC. This paper aims to understand the fundamental mechanisms of diabetes-induced BC progression and to develop personalized treatments. It reports a metabolic reprogramming strategy (MRS) that pharmaceutical induction of glucose import and glycolysis with metformin and NF-κB inhibitor (NF-κBi) while blocking the export of excessive lactate via inhibiting monocarboxylate transporter 4 (MCT4) leads to a metabolic crisis within the cancer cells. It demonstrates that the MRS shifts the metabolism of BC cells toward higher production of lactate, blocks lactate secretion, prompts intracellular acidification and induces significant cytotoxicity. Moreover, a novel MCT4 inhibitor CB-2 has been identified by structure-based virtual screening. A triple combination of metformin, CB-2, and trabectedin, a drug that impedes NF-κB signaling, strongly inhibits BC cells. Compared to normal glucose condition, MRS elicits more potent cancer cell-killing effects under high glucose condition. Animal model studies show that diabetic conditions promote the proliferation and progression of BC xenografts in nude mice and that MRS treatment significantly inhibits HG-induced BC progression. Therefore, inhibition of MCT4 combined with metformin/NF-κBi is a promising cancer therapy, especially for diabetes-associated BC.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Diabetes Mellitus, Experimental/metabolism , Metformin/therapeutic use , Monocarboxylic Acid Transporters/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Trabectedin/therapeutic use , Animals , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Breast Neoplasms/complications , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Female , Glucose/metabolism , Glycolysis/drug effects , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Lactic Acid/metabolism , Metformin/metabolism , Mice , Prognosis , Trabectedin/metabolism
2.
Biochem Biophys Res Commun ; 577: 89-94, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34509083

ABSTRACT

The protozoan Plasmodium falciparum is the main aetiological agent of tropical malaria. Characteristic of the phylum is the presence of a plastid-like organelle which hosts several homologs of plant proteins, including a ferredoxin (PfFd) and its NADPH-dependent reductase (PfFNR). The PfFNR/PfFd redox system is essential for the parasite, while mammals share no homologous proteins, making the enzyme an attractive target for novel and much needed antimalarial drugs. Based on previous findings, three chemically reactive residues important for PfFNR activity were identified: namely, the active-site Cys99, responsible for hydride transfer; Cys284, whose oxidation leads to an inactive dimeric form of the protein; and His286, which is involved in NADPH binding. These amino acid residues were probed by several residue-specific reagents and the two cysteines were shown to be promising targets for covalent inhibition. The quantitative and qualitative description of the reactivity of few compounds, including a repurposed drug, set the bases for the development of more potent and specific antimalarial leads.


Subject(s)
Enzyme Inhibitors/pharmacology , Ferredoxin-NADP Reductase/antagonists & inhibitors , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Biocatalysis/drug effects , Carmustine/chemistry , Carmustine/metabolism , Carmustine/pharmacology , Catalytic Domain , Cysteine/chemistry , Cysteine/metabolism , Diamide/chemistry , Diamide/metabolism , Diamide/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Ferredoxin-NADP Reductase/chemistry , Ferredoxin-NADP Reductase/metabolism , Kinetics , Malaria, Falciparum/parasitology , Molecular Structure , NADP/metabolism , Organomercury Compounds/chemistry , Organomercury Compounds/metabolism , Organomercury Compounds/pharmacology , Plasmodium falciparum/enzymology , Plasmodium falciparum/physiology , Protein Binding , Protein Domains , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Substrate Specificity
3.
Int J Biol Macromol ; 187: 742-754, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34310997

ABSTRACT

A surface modulated biodegradable transdermal strategy has been exploited for improving the biopharmaceutical properties of Temozolomide augmented in Poly Lactic-co-glycolic acid (PLGA) chitosan double walled nanogel (PCNGL). The PCNGL was synthesized by dual approach methodology showing consistent nanosize particle range of 210 nm and PDI 0.325 ± 0.43 with cationic zeta potential values +29.34 ± 0.79 mV. The PCNGL showed qualitative endothermic & exothermic temperature dependent degradation peaks by thermogravimetry analysis. Blood hemolysis and coagulation assay showed 3.37 ± 0.19 as hemolytic ratio, validating biologically safe margin for transdermal delivery. The in vitro drug release showed 85% transdermal release at slightly acidic pH mimicking skin microenvironment. The ex vivo studies displayed noteworthy penetration potential validated by concentration depth assay and confocal laser scanning microscopy, exhibiting 80% Temozolomide uptake in porcine epidermal tissue. The current research demonstrated the biodegradable controlled delivery of chemotherapeutic Temozolomide leading to biologically safe transdermal therapy.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Drug Carriers , Nanogels , Poloxamer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Temozolomide/chemistry , Administration, Cutaneous , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/metabolism , Delayed-Action Preparations , Drug Compounding , Drug Liberation , Epidermis/metabolism , Hydrogen-Ion Concentration , Nanotechnology , Skin Absorption , Surface Properties , Sus scrofa , Temozolomide/administration & dosage , Temozolomide/metabolism
4.
Chem Commun (Camb) ; 56(69): 9986-9989, 2020 Sep 07.
Article in English | MEDLINE | ID: mdl-32720950

ABSTRACT

We report a two-photon responsive drug delivery system (DDS), namely, p-hydroxyphenacyl-naphthalene-chlorambucil (pHP-Naph-Cbl), having a two-photon absorption (TPA) cross-section of ≥20 GM in the phototherapeutic window (700 nm). Our DDS exhibited both AIE and ESIPT phenomena, which were utilized for the real-time monitoring of anti-cancer drug release.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Chlorambucil/chemistry , Drug Carriers/chemistry , Naphthalenes/chemistry , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Cell Survival/drug effects , Chlorambucil/metabolism , Chlorambucil/pharmacology , Drug Liberation , Humans , Light , MCF-7 Cells , Microscopy, Confocal , Photons
5.
Arch Toxicol ; 93(11): 3291-3303, 2019 11.
Article in English | MEDLINE | ID: mdl-31598736

ABSTRACT

The clinical use of cyclophosphamide and ifosfamide is limited by a resultant bladder toxicity which has been attributed to the metabolite acrolein. Another metabolite chloroacetaldehyde (CAA) associated with nephrotoxicity, has not been investigated for toxicity in the bladder and this study investigates the effects of acrolein and CAA on human urothelial cells in vitro. Human urothelial cells (RT4 and T24) were treated with acrolein or CAA and changes in cell viability, reactive oxygen species, caspase-3 activity and release of urothelial mediators ATP, acetylcholine, PGE2 were measured. The protective effects of N-acetyl cysteine (NAC) were also assessed. Both metabolites were toxic to human urothelial cells, however, CAA significantly decreased cell viability at a ten-fold lower concentration (10 µM) than acrolein (100 µM). This was associated with increased ROS production and caspase-3 activity. NAC protected cells from these changes. In RT4 cells 100 µM acrolein caused a significant increase in basal and stretch-induced ATP, Ach and PGE2 release. In T24 cells chloroacetaldehyde (10 µM) increased basal and stimulated ATP and PGE2 levels. Again, NAC protected against changes in urothelial mediator release following acrolein or CAA. This study is the first to report that CAA in addition to acrolein contributes to the urotoxicity of cyclophosphamide and ifosfamide. Both metabolites altered urothelial mediator levels which could contribute to the sensory and functional bladder changes experienced by patients after treatment with cyclophosphamide or ifosfamide. Alterations in urothelial cell viability and mediator release may be causally linked to oxidative stress, with NAC providing protection against these changes.


Subject(s)
Acetaldehyde/analogs & derivatives , Acrolein/toxicity , Antineoplastic Agents, Alkylating/toxicity , Cyclophosphamide/toxicity , Epithelial Cells/drug effects , Urinary Bladder/drug effects , Urothelium/drug effects , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Acrolein/metabolism , Antineoplastic Agents, Alkylating/metabolism , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cyclophosphamide/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urothelium/metabolism , Urothelium/pathology
7.
Drug Metab Dispos ; 47(5): 547-555, 2019 05.
Article in English | MEDLINE | ID: mdl-30858239

ABSTRACT

Duocarmycins [including cyclopropyl pyrroloindole (CPI) or cyclopropyl benzoindole (CBI)] are a class of DNA minor-groove alkylators and seco-CPI/CBIs are synthetic pro-forms that can spirocyclize to CPI/CBI. Bis-CPI/CBIs are potential drug candidates because of their enhanced cytotoxicity from DNA crosslinking, but it is difficult to analyze them for structure-activity correlation because of their DNA reactivity. To study their DNA alkylation, neutral thermal hydrolysis has been frequently applied to process depurination. However, unwanted side reactions under this condition have been reported, which could lead to poor correlation of DNA alkylation data with efficacy results, especially for bis-CPI/CBIs. In this study, an acidic depurination method was developed and applied for analysis of DNA alkylation and shown to be an easier and milder method than the traditional neutral thermal hydrolysis. DNA alkylation and stability of three bis-seco-CBIs were characterized in comparison with two mono-seco-CPIs. The results suggested that: 1) The acidic depurination method was capable of capturing a more representative population, sometimes a different population, of DNA adducts as they existed on DNA compared with the heat depurination method. 2) Di-adenine adducts were captured as expected for the CBI dimers, although the major type of adduct was still mono-adenine adducts. 3) The rate of DNA alkylation, DNA adduct profile, and relative amounts of di-adduct versus mono-adduct were significantly affected by the size, and possibly lipophilicity, of the nonalkylating part of the molecules. 4) Spirocyclization and amide hydrolysis represented two major pathways of degradation. Overall, by applying acidic depurination analyses, this study has illustrated DNA adduct characteristics of novel bis-seco-CBIs with dominating mono-alkylation and provides an alternative method for evaluating DNA minor-groove alkylators. These findings provide an effective analytical tool to evaluate DNA alkylators and to study the DNA alkylation that is a disposition mechanism of these compounds.


Subject(s)
Alkylation/physiology , Antineoplastic Agents, Alkylating/metabolism , DNA/metabolism , Duocarmycins/metabolism , Adenine/metabolism , Alkylating Agents/metabolism , DNA Adducts/metabolism
8.
Drug Deliv ; 26(1): 34-44, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30744436

ABSTRACT

The addition of temozolomide (TMZ) to radiotherapy (RT) improves survival of patients with glioblastoma (GBM). However, TMZ + RT causes excess toxicity in patients. In this study, we prepared angiopep-2 (A2) modified lipid-poly (hypoxic radiosensitized polyprodrug) nanoparticles for TMZ delivery (A2-P(MIs)25/TMZ) to achieve synergistic effects against glioma. This A2-P(MIs)25/TMZ display highly promising advantages: (1) a hydrophobic P-(MIs)25 core where poorly water-soluble TMZ can be encapsulated; (2) nitro groups of the hydrophobic P-(MIs)25 core that are converted into hydrophilic amino groups (P(NH2s)25) under low oxygen conditions to mimic the oxygen-increased sensitization to RT; (3) a lipid monolayer at the interface of the core and the shell to modify the A2 (a specific ligand for low-density lipoprotein receptor-related protein-1 (LRP-1), which are expressed in the blood-brain barrier (BBB) and human glioma cells), thereby enhancing the drug encapsulation efficiency in glioma. These nanoparticles appear as a promising and robust nanoplatforms for TMZ and hypoxic cell radiosensitization delivery.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Glioma/therapy , Nanoparticles/administration & dosage , Peptides/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Temozolomide/administration & dosage , Animals , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/metabolism , Cell Line, Tumor , Combined Modality Therapy/methods , Drug Delivery Systems/methods , Glioma/metabolism , Humans , Male , Mice , Mice, Inbred ICR , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemical synthesis , Polylactic Acid-Polyglycolic Acid Copolymer/metabolism , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/metabolism , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/metabolism , Radiotherapy/methods , Temozolomide/chemical synthesis , Temozolomide/metabolism , Xenograft Model Antitumor Assays/methods
9.
Eur J Pharm Biopharm ; 136: 156-163, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30682492

ABSTRACT

Temozolomide (TMZ) is the first-line treatment for Glioblastoma Multiforme (GBM). After administration, TMZ is rapidly converted into its active metabolite (MTIC). However, its pharmacological activity is reduced due MTIC low bioavailability in the brain. Since drugs' permeability through biological barriers and tumor cell membranes affects its bioavailability, the ability of MTIC to interact with the biological membranes presents a major contribution on its pharmacological properties and activity. Biomembrane models mimic the physiological conditions, allowing to predict the drug's behavior at biological membranes and its effects on drug biodistribution profiles. In this work, lipid bilayer models using liposomes were applied for the drug-membrane interaction studies. The zwitterionic phospholipid, 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), and cholesterol were chosen for the composition of the model, since they represent the major components of the membranes of GBM cells and brain capillary endothelial cell. Thus, the molecular interactions between MTIC and these models were studied by the evaluation of the partition of the drug into the phospholipid's membrane, its location within the bilayer and its effect on the fluidity of the membrane. The attained results suggest that the composition of membranes affects drugs partition, showing that drug biodistribution depends not only on its physicochemical features, but also depends on the characteristics of the membrane such as the packing of the lipid molecules. Also, MTIC exhibited low affinity to biological membranes, explaining its low bioavailability on the target cells.


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Cholesterol/metabolism , Dimyristoylphosphatidylcholine/metabolism , Glioblastoma/metabolism , Membranes, Artificial , Temozolomide/metabolism , Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/metabolism , Drug Interactions/physiology , Glioblastoma/drug therapy , Humans , Temozolomide/administration & dosage
10.
Chem Commun (Camb) ; 55(7): 953-956, 2019 Jan 17.
Article in English | MEDLINE | ID: mdl-30604783

ABSTRACT

Supramolecular drug delivery systems are becoming an increasingly important part in controlled drug release. In this work, we report a novel enzyme-responsive supramolecular assembly directly constructed using biocompatible sulfato-ß-cyclodextrin (SCD) and an anti-cancer prodrug, i.e. choline modified anti-cancer drug chlorambucil (QA-Cbl). The supramolecular assembly acts as an effective drug delivery system via the controlled drug loading and enzyme-responsive drug release, because the butyrylcholinesterase (BChE) can cleave the ester bond of QA-Cbl prodrug, resulting in the release of anti-cancer drug chlorambucil (Cbl). Compared to other sophisticated drug delivery systems, the present system provides a feasible and functional approach for achievement of controlled drug release.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Butyrylcholinesterase/metabolism , Chlorambucil/administration & dosage , Cyclodextrins/chemistry , Drug Liberation , Prodrugs/chemistry , Antineoplastic Agents, Alkylating/metabolism , Chlorambucil/metabolism , Delayed-Action Preparations , Humans
11.
Biomater Sci ; 7(3): 1161-1178, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30652182

ABSTRACT

Melanoma is a highly aggressive skin cancer. A paclitaxel formulation of solid lipid nanoparticles modified with Tyr-3-octreotide (PSM) is employed to treat melanoma that highly expresses somatostatin receptors (SSTRs). PSM exerts more apoptotic and anti-invasive effects in B16F10 mice melanoma cells as compared to dacarbazine (DTIC), an approved chemotherapeutic drug for treating aggressive melanoma. Besides, PSM induces one of the biomarkers of immunogenic cell death in vitro and in vivo as confirmed by calreticulin exposure on the B16F10 cell surface. We observed a significant number of CD8 positive T cells in the tumor bed of the PSM treated group. As a result, PSM effectively reduces tumor volume in vivo as compared to DTIC. PSM also induces a favorable systemic immune response as determined in the spleen and sera of the treated animals. Importantly, PSM can reduce the number of nodule formations in the experimental lung metastasis model. Our experimentations indicate that the metronomic PSM exhibits remarkable anti-melanoma activities without any observable toxicity. This immune modulation behavior of PSM can be exploited for the therapy of melanoma and probably for other malignancies.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Nanoparticles/chemistry , Paclitaxel/chemistry , Peptides/chemistry , Animals , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Calreticulin/chemistry , Calreticulin/pharmacology , Cell Line, Tumor , Cytokines/metabolism , Dacarbazine/chemistry , Dacarbazine/metabolism , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Disease Models, Animal , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Paclitaxel/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Survival Rate , Tissue Distribution
12.
J Oncol Pharm Pract ; 25(1): 226-228, 2019 Jan.
Article in English | MEDLINE | ID: mdl-28857712

ABSTRACT

Busulfan is an alkylating agent used in pre-transplant conditioning for patients undergoing hematopoietic stem cell transplantation. Several factors contribute to variations in busulfan drug disposition including bioavailability, age, liver function, genetic polymorphisms, and concurrent administration of other drugs. Busulfan is metabolized by hepatic oxidation via the cytochrome P450 3A4 system as well as through conjugation with glutathione. Interactions with drugs such as phenytoin, itraconazole, and metronidazole have been reported to alter busulfan clearance and result in sub- or supra-therapeutic concentrations. We report a case of a clinically significant drug interaction between intravenous busulfan and the bifunctional T-cell engager, blinatumomab, observed through busulfan therapeutic drug monitoring. We found that busulfan clearance was reduced resulting in a higher area under the concentration-time curve when it was administered 48 h after blinatumomab. Repeat busulfan pharmacokinetic testing two weeks later demonstrated increased clearance of the drug and a 31% higher dose recommendation. Similar to other protein therapeutics, cytokine elevations during blinatumomab treatment can lead to cytochrome 3A4 suppression. We hypothesize that the increased busulfan levels observed could be related to a cytokine-mediated CYP3A4 suppression. This represents a unique pharmacologic consideration in hematopoietic stem cell transplantation which would impact several drugs that undergo CYP3A4 metabolism, including calcineurin inhibitors, cyclophosphamide, sirolimus, and triazole antifungals. Additionally, this mechanism of CYP3A4 suppression may be relevant in treatments and disease states where cytokine levels are elevated such as haploidentical stem cell transplantation, graft-versus-host disease, and use of chimeric antigen receptor T-cell therapy.


Subject(s)
Antibodies, Bispecific/metabolism , Antineoplastic Agents/metabolism , Busulfan/metabolism , Drug Monitoring/methods , Adult , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents, Alkylating/metabolism , Busulfan/therapeutic use , Drug Interactions/physiology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/methods , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Male , Transplantation Conditioning/methods
13.
J Endocrinol ; 240(2): 243-256, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30530902

ABSTRACT

Cancer therapy can cause off-target effects including ovarian damage, which may result in primary ovarian insufficiency in girls and premenopausal women. Loss of ovarian follicles within the ovarian reserve leads to ovarian endocrine dysfunction and impaired fertility. Cyclophosphamide (CPA), a commonly used chemotherapeutic and immunosuppressant agent, is a gonadotoxic agent that destroys ovarian cells by crosslinking DNA. To protect the ovary against CPA damage, we sought to precisely map the mechanism by which the ovarian reserve is depleted by CPA. We found that CPA specifically depletes primordial follicles without affecting primary and secondary follicles in three independent murine strains (CD-1, C57BL/6J and BALB/cJ) in vivo. We directly tested the effect of the active metabolite of CPA, 1 µM 4-hydroxyperoxycyclophophamide (4-HC), in vitro and confirmed the loss of primordial oocytes but no change in the number of primary and secondary follicles. We demonstrated that phospho-AKT (p-AKT) and cleaved PARP (cPARP) are present in primordial oocytes 3 days after CPA injection, consistent with the role of these markers as part of the apoptotic cascade. Interestingly, p-AKT positive primordial oocytes co-expressed cPARP. Treatment of animals with specific inhibitors of apoptotic pathway components, ETP46464 and CHK2, blocked 4-HC‒induced DNA damage in vitro. These data suggest that CPA targets primordial germ cells in the ovarian reserve by stimulating apoptosis pathways. Adjuvant therapies to protect primordial germ cells from the off-target effects of CPA may reduce the risk of POI.


Subject(s)
Apoptosis/drug effects , Cyclophosphamide/toxicity , Ovarian Reserve/drug effects , Oxazines/pharmacology , Quinolines/pharmacology , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/toxicity , Cyclophosphamide/administration & dosage , Cyclophosphamide/metabolism , Female , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Ovarian Reserve/physiology , Poly(ADP-ribose) Polymerases/metabolism , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/prevention & control , Protective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism
14.
Biomed Pharmacother ; 109: 386-390, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30399573

ABSTRACT

We showed previously that Dihydrotanshinone (DHT) augments temozolomide (TMZ) efficacy by inducing reactive oxygen species production in an in vitro model. Here, the underlying basis of the synergistic effect and the ability of DHT to potentially pass the blood brain barrier (BBB) is investigated using an in vitro model. Trypan blue exclusion assays were used to determine effects of DHT/TMZ combinatorial treatment on GBM cell viability. ELISA was utilized to determine effects on NFkB levels after singular and combinatorial treatment. An in vitro model of the BBB was constructed to predict the potential of DHT to penetrate the BBB in vivo. DHT and TMZ synergistically reduce cancer cell viability, NFkB activity, and markedly halt cell cycle progression. This regimen was also shown to exert minimal effects on astrocytes. Finally, DHT was shown to have the potential of passing through the BBB to a similar extent as TMZ and that paclitaxel's oncolytic effects are completely ablated in the presence of our in vitro BBB. Our data confirms the synergistic interaction between DHT and TMZ and also highlights the potential of combination treatment to sequester NFkB activity and inhibit cell cycle progression. The encouraging data with the BBB model show that the DHT/TMZ combination may be clinically useful and warrants future in vivo testing.


Subject(s)
Abietanes/metabolism , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Glioblastoma/metabolism , Temozolomide/metabolism , Tumor Suppressor Proteins/metabolism , Abietanes/administration & dosage , Abietanes/chemistry , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/metabolism , Blood-Brain Barrier/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Coculture Techniques , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drug Synergism , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Predictive Value of Tests , Temozolomide/administration & dosage , Tumor Suppressor Proteins/genetics
15.
Drug Metab Dispos ; 47(3): 257-268, 2019 03.
Article in English | MEDLINE | ID: mdl-30567881

ABSTRACT

Cyclophosphamide (CP), a prodrug that is enzymatically converted to the cytotoxic 4-hydroxycyclophosphamide (4OHCP) by hepatic enzymes, is commonly used in both human and veterinary medicine to treat cancers and modulate the immune system. We investigated the metabolism of CP in humans, dogs, cats, and mice using liver microsomes; apparent K M, V max, and intrinsic clearance (V max/K M) parameters were estimated. The interspecies and intraspecies variations in kinetics were vast. Dog microsomes were, on average, 55-fold more efficient than human microsomes, 2.8-fold more efficient than cat microsomes, and 1.2-fold more efficient than mouse microsomes at catalyzing CP bioactivation. These differences translated to cell-based systems. Breast cancer cells exposed to 4OHCP via CP bioactivation by microsomes resulted in a stratification of cytotoxicity that was dependent on the species of microsomes measured by IC50: dog (31.65 µM), mouse (44.95 µM), cat (272.6 µM), and human (1857 µM). The contributions of cytochrome P450s, specifically, CYP2B, CYP2C, and CYP3A, to CP bioactivation were examined: CYP3A inhibition resulted in no change in 4OHCP formation; CYP2B inhibition slightly reduced 4OHCP in humans, cats, and mice; and CYP2C inhibition drastically reduced 4OHCP formation in each species. Semiphysiologic modeling of CP metabolism using scaled metabolic parameters resulted in simulated data that closely matched published pharmacokinetic profiles, determined by noncompartmental analysis. The results highlight differential CP metabolism delineated by species and demonstrate the importance of metabolism on CP clearance.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Cyclophosphamide/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Models, Biological , Prodrugs/pharmacokinetics , Animals , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Cat Diseases/drug therapy , Cat Diseases/immunology , Cats , Cell Line, Tumor , Cyclophosphamide/metabolism , Cyclophosphamide/therapeutic use , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/pharmacology , Dog Diseases/drug therapy , Dog Diseases/immunology , Dogs , Female , Humans , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/therapeutic use , Male , Mice , Microsomes, Liver , Neoplasms/drug therapy , Oxidation-Reduction/drug effects , Prodrugs/metabolism , Prodrugs/therapeutic use
16.
Int J Nanomedicine ; 13: 7971-7985, 2018.
Article in English | MEDLINE | ID: mdl-30538471

ABSTRACT

INTRODUCTION: Macrocrystalline oxides of alkaline earth metals (Mg and Ca) or light metals (Al and Ti) can respond to standard warfare agents such as sulfur mustard, soman, or agent VX. In this paper, we compared the decontamination ability of sodium hydroxide (NaOH) and sodium hypochlorite (NaClO) for nitrogen mustards (cyclophosphamide [CP] and ifosfamide [IFOS]) with a new procedure using a destructive sorbent based on nanocrystalline and nanodispersive titanium dioxide (TiO2) as a new efficient and cheap material for complete decontamination of surfaces. METHODS: Titanium (IV) dioxide nanoparticles were prepared by the homogeneous hydrolysis of titanium(IV) oxysulfate (TiOSO4) with urea. The as-prepared TiO2 nanoparticles were used for the fast and safe decontamination of cytostatics from the nitrogen mustard family (CP and IFOS) in water. The adsorption-degradation process of cytostatics in the presence of TiO2 was compared with decontamination agents (0.01 M solution of sodium hydroxide and 5% solution of sodium hypochlorite). The mechanism of the decontamination process and the degradation efficiency were determined by high-performance liquid chromatography with mass spectrometry. RESULTS: It was demonstrated that a 0.01 M solution of sodium hydroxide (NaOH) decomposes CP to 3-((amino(bis(2-chloroethyl)amino)phosphoryl)oxy)propanoic acid and sodium hypochlorite formed two reaction products, namely, IFOS and 4-hydroxy-cyclophosphamide. IFOS is cytotoxic, and 4-hydroxy-cyclophosphamide is a known metabolite of CP after its partial metabolism by CYP/CYP450. IFOS degrades in the pres¬ence of NaOH to toxic IFOS mustard. Titanium(IV) dioxide nanoparticles adsorbed on its surface CP after 5 minutes and on IFOS after 10 minutes. The adsorption-degradation process of CP in water and in the presence of TiO2 led to 4-hydroxy-cyclophosphamide and IFOS, respectively, which decayed to oxidation product 4-hydroxy-ifosfamide. CONCLUSION: Nanodispersive TiO2 is an effective degradation agent for decontamination of surfaces from cytostatics in medical facilities.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Cyclophosphamide/chemistry , Cytostatic Agents/chemistry , Decontamination/methods , Ifosfamide/chemistry , Nanoparticles/chemistry , Titanium/chemistry , Antineoplastic Agents, Alkylating/metabolism , Cyclophosphamide/metabolism , Cytostatic Agents/metabolism , Humans , Ifosfamide/metabolism
17.
J Med Chem ; 61(20): 9132-9145, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30247905

ABSTRACT

We describe several new aromatic nitrogen mustards with various aromatic substituents and boronic esters that can be activated with H2O2 to efficiently cross-link DNA. In vitro studies demonstrated the anticancer potential of these compounds at lower concentrations than those of other clinically used chemotherapeutics, such as melphalan and chlorambucil. In particular, compound 10, bearing an amino acid ester chain, is selectively cytotoxic toward breast cancer and leukemia cells that have inherently high levels of reactive oxygen species. Importantly, 10 was 10-14-fold more efficacious than melphalan and chlorambucil for triple-negative breast-cancer (TNBC) cells. Similarly, 10 is more toxic toward primary chronic-lymphocytic-leukemia cells than either chlorambucil or the lead compound, 9. The introduction of an amino acid side chain improved the solubility and permeability of 10. Furthermore, 10 inhibited the growth of TNBC tumors in xenografted mice without obvious signs of general toxicity, making this compound an ideal drug candidate for clinical development.


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Drug Design , Hydrogen Peroxide/metabolism , Nitrogen Mustard Compounds/metabolism , Nitrogen Mustard Compounds/pharmacology , Antineoplastic Agents, Alkylating/chemistry , Boronic Acids/chemistry , Cell Line, Tumor , Esters/chemistry , Humans , Nitrogen Mustard Compounds/chemistry
18.
Int J Pharm ; 545(1-2): 84-92, 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29715532

ABSTRACT

Glioblastoma multiforme is the most lethal type of brain tumor and the established therapy only extends patients survival to approximately one year. Its first-line treatment is based on of chemotherapy with the alkylating agent temozolomide (TMZ). As many other chemotherapeutic drugs, TMZ presents several limitations as high toxicity and low bioavailability. The delivery of TMZ using poly(lactic-co-glycolic acid) nanoparticles is proposed in this work. Stable nanoparticles functionalized with a OX26 type monoclonal antibody for transferrin receptor were developed, targeting the glioblastoma tumor cells, since these cells are known for overexpressing this receptor. The release profile of TMZ from the nanoparticles was studied mimicking physiological conditions, and targeted cellular internalization was also investigated. Two glioblastoma cell lines - U215 and U87 - were used to evaluate the in vitro cytotoxicity of the drug, showing that the prepared nanocarriers enhance the anticancer activity of TMZ. The functionalization with the monoclonal antibody for transferrin receptor proved to be advantageous in enhancing the cellular internalization in glioblastoma cells.


Subject(s)
Antibodies, Monoclonal/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Drug Carriers , Glioblastoma/drug therapy , Lactic Acid/chemistry , Nanoparticles , Polyglycolic Acid/chemistry , Receptors, Transferrin/metabolism , Antibodies, Monoclonal/chemistry , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/chemistry , Dacarbazine/metabolism , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Compounding , Drug Liberation , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Kinetics , Nanotechnology , Polylactic Acid-Polyglycolic Acid Copolymer , Receptors, Transferrin/immunology , Technology, Pharmaceutical/methods , Temozolomide
19.
PLoS One ; 13(4): e0195366, 2018.
Article in English | MEDLINE | ID: mdl-29621334

ABSTRACT

The waste of commonly used medicines is known to contaminate freshwater ecosystems. Pharmaceuticals can be toxic, mutagenic, or modifying to freshwater organisms even at low concentrations if consider their permanent presence in the environment. Chemotherapeutics used to treat cancer, and in particular alkylating agents, contribute significantly to this form of pollution, the latter introducing cytotoxic and/or mutagenic lesions to the DNA and RNA of organisms which can be disruptive to their cells. The aim of the present study was to investigate the influence of the alkylating anticancer agent cyclophosphamide (CP) on Daphnia magna clones. We evaluated the life history parameters and protein profiles of this crustacean following exposure to environmentally relevant CP concentration of 10 ng L-1. Even at this low concentration, the alkylating agent caused modification of the life history parameters and proteome profile of the Daphnia. These changes were clone-specific and involved growth rate, age at first reproduction, neonate number, and proteins related to cell cycle and redox state regulation. The disturbance caused by pharmaceuticals contaminating freshwater ecosystem is probably weaker and unlikely to be cytotoxic in character due to the high dilution of these substances in the water. However, our results indicate that prolonged exposure of organisms to these toxins may lead to modifications on the organismal and molecular levels with unpredictable significance for the entire ecosystem.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Cell Cycle/drug effects , Cyclophosphamide/toxicity , Daphnia/growth & development , Water Pollutants, Chemical/toxicity , Animals , Antineoplastic Agents, Alkylating/metabolism , Cyclophosphamide/metabolism , Daphnia/metabolism , Oxidation-Reduction/drug effects , Water Pollutants, Chemical/metabolism , Water Pollution, Chemical/analysis
20.
Mol Cancer Ther ; 17(3): 650-660, 2018 03.
Article in English | MEDLINE | ID: mdl-29440292

ABSTRACT

Tumor-selective delivery of cytotoxic agents in the form of antibody-drug conjugates (ADCs) is now a clinically validated approach for cancer treatment. In an attempt to improve the clinical success rate of ADCs, emphasis has been recently placed on the use of DNA-cross-linking pyrrolobenzodiazepine compounds as the payload. Despite promising early clinical results with this class of ADCs, doses achievable have been low due to systemic toxicity. Here, we describe the development of a new class of potent DNA-interacting agents wherein changing the mechanism of action from a cross-linker to a DNA alkylator improves the tolerability of the ADC. ADCs containing the DNA alkylator displayed similar in vitro potency, but improved bystander killing and in vivo efficacy, compared with those of the cross-linker. Thus, the improved in vivo tolerability and antitumor activity achieved in rodent models with ADCs of the novel DNA alkylator could provide an efficacious, yet safer option for cancer treatment. Mol Cancer Ther; 17(3); 650-60. ©2018 AACR.


Subject(s)
Immunoconjugates/pharmacology , Intercalating Agents/pharmacology , Neoplasms/drug therapy , Therapeutic Index, Drug , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cross-Linking Reagents/chemistry , DNA/genetics , DNA/metabolism , Drug Design , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Mice , Neoplasms/pathology , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...