Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 573
Filter
1.
Bioorg Med Chem Lett ; 105: 129730, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38583784

ABSTRACT

Chlorambucil is an alkylating drug that finds application towards chemotherapy of different types of cancers. In order to explore the possibility of utilization of this drug as an imaging agent for early diagnosis of solid tumors, attempt was made to synthesize a 99mTc complex of chlorambucil and evaluate its potential in tumor bearing small animal model. HYNIC-chlorambucil was synthesized by conjugation of HYNIC with chlorambucil via an ethylenediamine linker. All the intermediates and final product were purified and characterized by standard spectroscopic techniques viz. FT-IR, 1H/13C-NMR as well as by mass spectrometry. HYNIC-chlorambucil conjugate was radiolabeled with [99mTc]Tc and found to be formed with > 95 % radiochemical purity via RP-HPLC studies. The partition coefficient (Log10Po/w) of the synthesized complex was found to be -0.78 ± 0.25 which indicated the moderate hydrophilic nature for the complex. Biological behaviour of [99mTc]Tc-HYNIC-chlorambucil, studied in fibrosarcoma bearing Swiss mice, revealed a tumor uptake of about 4.16 ± 1.52 %IA/g at 30 min post-administration, which declined to 1.91 ± 0.13 % IA/g and 1.42 ± 0.14 %IA/g at 1 h and 2 h post-administration, respectively. A comparison of different [99mTc]Tc-chlorambucil derivatives (reported in the contemporary literature) formulated using different methodologies revealed that tumor uptake and pharmacokinetics exhibited by these agents strongly depend on the lipophilicity/hydrophilicity of such agents, which in turn is dependent on the bifunctional chelators used for formulating the radiolabeled chlorambucils.


Subject(s)
Chlorambucil , Organotechnetium Compounds , Animals , Humans , Mice , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Chlorambucil/chemistry , Chlorambucil/chemical synthesis , Chlorambucil/pharmacology , Molecular Structure , Nicotinic Acids/chemistry , Nicotinic Acids/chemical synthesis , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/chemical synthesis , Organotechnetium Compounds/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Technetium/chemistry , Tissue Distribution
2.
Science ; 377(6605): 502-511, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35901163

ABSTRACT

Approximately half of glioblastoma and more than two-thirds of grade II and III glioma tumors lack the DNA repair protein O6-methylguanine methyl transferase (MGMT). MGMT-deficient tumors respond initially to the DNA methylation agent temozolomide (TMZ) but frequently acquire resistance through loss of the mismatch repair (MMR) pathway. We report the development of agents that overcome this resistance mechanism by inducing MMR-independent cell killing selectively in MGMT-silenced tumors. These agents deposit a dynamic DNA lesion that can be reversed by MGMT but slowly evolves into an interstrand cross-link in MGMT-deficient settings, resulting in MMR-independent cell death with low toxicity in vitro and in vivo. This discovery may lead to new treatments for gliomas and may represent a new paradigm for designing chemotherapeutics that exploit specific DNA repair defects.


Subject(s)
Antineoplastic Agents, Alkylating , Brain Neoplasms , DNA Modification Methylases , DNA Repair Enzymes , Drug Design , Drug Resistance, Neoplasm , Glioblastoma , Tumor Suppressor Proteins , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , DNA Methylation/genetics , DNA Modification Methylases/genetics , DNA Repair/genetics , DNA Repair Enzymes/genetics , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Tumor Suppressor Proteins/genetics
3.
Bioorg Med Chem ; 53: 116533, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34863065

ABSTRACT

Glioblastoma (GBM) is the most aggressive and treatment-refractory malignant adult brain cancer. After standard of care therapy, the overall median survival for GBM is only ∼6 months with a 5-year survival <10%. Although some patients initially respond to the DNA alkylating agent temozolomide (TMZ), unfortunately most patients become resistant to therapy and brain tumors eventually recur. We previously found that knockout of BRG1 or treatment with PFI-3, a small molecule inhibitor of the BRG1 bromodomain, enhances sensitivity of GBM cells to temozolomide in vitro and in vivo GBM animal models. Those results demonstrated that the BRG1 catalytic subunit of the SWI/SNF chromatin remodeling complex appears to play a critical role in regulating TMZ-sensitivity. In the present study we designed and synthesized Structurally Related Analogs of PFI-3 (SRAPs) and tested their bioactivity in vitro. Among of the SRAPs, 9f and 11d show better efficacy than PFI-3 in sensitizing GBM cells to the antiproliferative and cell death inducing effects of temozolomide in vitro, as well as enhancing the inhibitor effect of temozolomide on the growth of subcutaneous GBM tumors.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Azabicyclo Compounds/pharmacology , DNA Helicases/antagonists & inhibitors , Glioblastoma/drug therapy , Nuclear Proteins/antagonists & inhibitors , Pyridines/pharmacology , Temozolomide/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents, Alkylating/chemistry , Azabicyclo Compounds/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , DNA Helicases/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Congenic , Mice, Inbred NOD , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/metabolism , Pyridines/chemistry , Structure-Activity Relationship , Temozolomide/chemistry , Transcription Factors/metabolism
4.
ACS Appl Mater Interfaces ; 13(47): 55851-55861, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34788006

ABSTRACT

Temozolomide (TMZ) is a prodrug of 5-(3-methyltriazene-1-yl)imidazole-4-carboxamide (MTIC, short-lived) and used as a first-line therapy drug for glioblastoma multiforme (GBM). However, little progress has been made in regulating the kinetics of TMZ to MTIC degradation to improve the therapeutic effect, particularly in the case of TMZ-resistant GBM. In this work, we introduced a strategy to cage MTIC by N-acylation of the triazene moiety to boost the MTIC stability, designed a diblock copolymer-based MTIC prodrug installed with a disulfide linkage, and achieved self-assembled polymer micelles without the concern of MTIC leakage under physiological conditions. Polymer micelles could be induced to disassemble by stimuli factors such as glutathione (GSH) and visible light irradiation through thiol/sulfide exchange and homolytic sulfide scission mechanisms, which contributed to MTIC release in GSH-dependent and GSH-independent pathways. The in vitro results demonstrated that microenvironment-responsive polymeric micelles benefited the suppression of both TMZ-sensitive and TMZ-resistant GBM cells. The chemistry of polymer-MTIC prodrug provided a new option for TMZ-based glioma treatment.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Glioblastoma/drug therapy , Glutathione/chemistry , Light , Polymers/chemistry , Prodrugs/pharmacology , Temozolomide/pharmacology , Antineoplastic Agents, Alkylating/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Delivery Systems , Drug Liberation , Drug Screening Assays, Antitumor , Glioblastoma/pathology , Humans , Molecular Structure , Particle Size , Prodrugs/chemistry , Surface Properties , Temozolomide/chemistry
5.
J Mol Model ; 27(12): 347, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34748097

ABSTRACT

The remarkable properties of pristine B3O3 nanosheet as a nanocarrier for adsorption and desorption of TEPA anticancer drug for designing potential drug delivery platform were investigated using periodic DFT calculations. We studied the adsorption energy of all stable complexes formed between the drug molecule and B3O3 in gas and aqueous phases along with electronic structure analysis of complexes. Different adsorption configurations were studied for drug/B3O3 complexes, including the interaction of the C atom of the triangular ring, O atom in the TEPA drug with the B atom in B3O3, and indirect drug interaction the middle of the R1 ring cavity of the B3O3 nanosheet. The take-up of TEPA prompts a substantial change of 68.13% in the band gap (Eg) of the B3O3 nanosheet in the most stable complex. The present study results affirmed the application of B3O3 nanosheet as a potential vehicle for TEPA drugs in the treatment of cancerous tissues.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Boron Compounds/chemistry , Drug Delivery Systems/methods , Triethylenephosphoramide/administration & dosage , Triethylenephosphoramide/chemistry , Adsorption , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacokinetics , Density Functional Theory , Drug Liberation , Gases/chemistry , Nanostructures/chemistry , Solvents/chemistry , Triethylenephosphoramide/pharmacokinetics , Water/chemistry
6.
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
7.
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
8.
Chem Res Toxicol ; 34(7): 1790-1799, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34133118

ABSTRACT

Nitrogen mustards are a widely used class of antitumor agents that exert their cytotoxic effects through the formation of DNA interstrand cross-links (ICLs). Despite being among the first antitumor agents used, the biological responses to NM ICLs remain only partially understood. We have previously reported the generation of NM ICL mimics by incorporation of ICL precursors into DNA using solid-phase synthesis at defined positions, followed by a double reductive amination reaction. However, the structure of these mimics deviated from the native NM ICLs. Using further development of our approach, we report a new class of NM ICL mimics that only differ from their native counterpart by substitution of dG with 7-deaza-dG at the ICL. Importantly, this approach allows for the synthesis of diverse NM ICLs, illustrated here with a mimic of the adduct formed by chlorambucil. We used the newly generated ICLs in reactions with replicative and translesion synthesis DNA polymerase to demonstrate their stability and utility for functional studies. These new NM ICLs will allow for the further characterization of the biological responses to this important class of antitumor agents.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , DNA/chemistry , Intercalating Agents/chemistry , Mechlorethamine/analogs & derivatives , Antineoplastic Agents, Alkylating/chemical synthesis , DNA/chemical synthesis , DNA-Directed DNA Polymerase/chemistry , Humans , Intercalating Agents/chemical synthesis , Mechlorethamine/chemical synthesis
9.
Eur J Pharm Biopharm ; 161: 29-36, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33567313

ABSTRACT

Glioblastoma multiforme is the most aggressive and lethal form of brain tumour due to the high degree of cancer cells infiltration into surrounding brain tissue. No form of monotherapy can guarantee satisfactory patient outcomes and is only of palliative importance. To find a potential option of glioblastoma treatment the bioresorbable, layer nonwoven mats for controlled temozolomide and nimorazole release were obtained by classical and coaxial electrospinning. Optimization of fibre structure that enables delayed and controlled drug release was performed. The studied bioresorbable polymers were poly(L-lactide-co-ε-caprolactone) and poly(L-lactide-co-glycolide-co-trimethylene carbonate). The physicochemical properties of polymers were determined as well as drug release profiles of nonwoven mats. A combination of coaxial electrospinning and electrospray technique provided three-phased release profiles of temozolomide and nimorazole: the slow release of very low drug doses followed by accelerated release and saturation phase. Results form the basis for further investigation since both studied polymers possess a great potential as nimorazole and temozolomide delivery systems in the form of layered nonwoven implants.


Subject(s)
Absorbable Implants , Drug Carriers/chemistry , Nimorazole/administration & dosage , Temozolomide/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/chemistry , Brain Neoplasms/drug therapy , Chemistry, Pharmaceutical , Delayed-Action Preparations , Dioxanes/chemistry , Drug Liberation , Glioblastoma/drug therapy , Nimorazole/chemistry , Polyesters/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polymers/chemistry , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/chemistry , Temozolomide/chemistry
10.
ACS Appl Mater Interfaces ; 13(5): 6099-6108, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33507729

ABSTRACT

The blood-brain barrier (BBB) is a physical barrier that selectively prevents certain substances from entering the brain through the blood. The BBB protects the brain from germs and causes difficulty in intracranial treatment. The chemotherapy drug temozolomide (TMZ), embedded in nanobubbles (NBs) and combined with persistent luminescent nanoparticles (PLNs), has been used to treat glioblastoma (GBM) effectively through image tracking. Through ultrasound induction, NBs produce cavitation that temporarily opens the BBB. Additionally, the PLNs release near-infrared emission and afterglow, which can penetrate deep tissues and improve the signal-to-noise ratio of bioimages. In this work, the nanosystem crossed the BBB for drug delivery and image tracking over time, allowing the enhancement of the drug's therapeutic effect on GBM. We hope that this nanosystem can be applied to the treatment of different brain diseases by embedding different drugs in NBs.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/therapy , Glioblastoma/therapy , Nanocomposites/chemistry , Temozolomide/pharmacology , Ultrasonic Therapy , Animals , Antineoplastic Agents, Alkylating/chemistry , Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Glioblastoma/pathology , Humans , Infrared Rays , Molecular Structure , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Particle Size , Surface Properties , Temozolomide/chemistry , Ultrasonic Waves
11.
Chembiochem ; 22(9): 1538-1545, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33453075

ABSTRACT

Pyrrole-imidazole (PI) polyamides, which target specific DNA sequences, have been studied as a class of DNA minor-groove-binding molecules. To investigate the potential of compounds for cancer treatment, PI polyamides were conjugated with DNA-alkylating agents, such as seco-CBI and chlorambucil. DNA-alkylating PI polyamides have attracted attention because of their sequence-specific alkylating activities, which contribute to reducing the severe side effects of current DNA-damaging drugs. Many of these types of conjugates have been developed as new candidates for anticancer drugs. Herein, we review recent progress into research on DNA-alkylating PI polyamides and their sequence-specific action on targets associated with cancer development.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Imidazoles/chemistry , Nylons/chemistry , Pyrroles/chemistry , Animals , Antineoplastic Agents, Alkylating/therapeutic use , DNA/chemistry , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Telomere/chemistry
12.
ACS Appl Bio Mater ; 4(3): 2628-2638, 2021 03 15.
Article in English | MEDLINE | ID: mdl-35014379

ABSTRACT

Enhanced drug localization at the tumor sites with minimal toxicity was demonstrated using dendrimer-conjugated temozolomide for treating experimental lymphoma, developed as a solid tumor. Herein, we have constructed a polyamidoamine (PAMAM) dendrimer conjugated with temozolomide to enhance the stability of the active drug metabolites, derived from the prodrug temozolomide. Our results suggest that the active drug (5-(3-methyltriazen-1-yl)imidazole-4-carboxamide) (MTIC) (derived from temozolomide) showed stable and sustained release from the dendrimer-temozolomide conjugate, suggesting the suitability of the construct for therapy. Besides growth inhibition and direct killing, the dendrimer-temozolomide construct induced extensive apoptosis not only in parental Dalton lymphoma tumor cells but also in the doxorubicin-resistant form of the tumor cells. Dendrimer-temozolomide conjugation significantly reduced the solid tumor growth and increased the lifespan with better prognosis, including improved histopathology of the treated mice, while untreated littermates developed extensive metastasis and succumbed to death.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Biocompatible Materials/pharmacology , Dendrimers/pharmacology , Drug Development , Lymphoma/drug therapy , Temozolomide/pharmacology , Animals , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/chemistry , Apoptosis/drug effects , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dendrimers/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Female , Humans , Lymphoma/pathology , Materials Testing , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Particle Size , Temozolomide/chemistry
13.
J Oncol Pharm Pract ; 27(1): 78-87, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32228132

ABSTRACT

BACKGROUND: Temozolomide oral suspension is not commercially available. OBJECTIVE: To evaluate the stability of three temozolomide 10 mg/mL suspensions prepared in Oral Mix SF® in three container types stored at 4°C and 23°C. METHODS: Using commercial capsules, three separate batches of three different temozolomide 10 mg/mL formulations (Oral Mix SF® with PK-30; PK-30 and citric acid; and neither PK-30 nor citric acid) were made and stored in three container types (amber glass bottles, amber polyethylene terephthalate bottles, and polypropylene oral syringes). The aliquots in each container type were stored protected from light, half at 25°C and half at 4°C. On study days 0, 5, 8, 14, 21, 28, 35, 42, and 56, physical properties of samples from each container type at each temperature were assessed, and the temozolomide concentration was determined using a stability-indicating method. The beyond-use-date (time to achieve 90% of initial concentration calculated using the lower limit of the 95% confidence interval of the observed degradation rate) was calculated. RESULTS: Samples stored at 25°C turned from white to orange within seven days. Temozolomide crystals were observed in all samples. Concentration changes due to study day and temperature (p < 0.001) were observed but not due to container (p = 0.991) or formulation (p = 0.987). The beyond-use-date of all formulations in all container types was 56 days at 4°C and 6 days at 23°C. CONCLUSIONS: We recommend that these temozolomide 10 mg/mL formulations be stored at 4°C and be assigned a beyond-use-date of 30 days.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Temozolomide/chemistry , Crystallization , Drug Compounding , Drug Packaging , Drug Stability , Drug Storage , Glass , Plastics , Syringes , Temperature
14.
Bioorg Med Chem Lett ; 31: 127697, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33220402

ABSTRACT

Tumor hypoxia has been widely explored over the years as a diagnostic and therapeutic marker. Herein, we have reported the design and synthesis of a series of dinitrobenzamide mustards (DNBM) based on the PR-104A hypoxia-selective prodrug. Specifically, we explored the impact of various leaving groups and the introduction of a carboxylic acid group on the biological performance of the DNBM constructs. Once in hand, the Log D values, cytotoxicity in PC-3 and DU-145 human prostate cancer cells lines and the hypoxia selectivities of the DNBM analogs were examined. Overall, the DNBM constructs were found to be tolerant to modifications with none of the explored modifications substantially degrading the cytotoxic potential of the constructs.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cell Hypoxia/drug effects , Nitrogen Mustard Compounds/pharmacology , Prostatic Neoplasms/drug therapy , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Molecular Structure , Nitrogen Mustard Compounds/chemical synthesis , Nitrogen Mustard Compounds/chemistry , Prostatic Neoplasms/pathology , Structure-Activity Relationship
15.
Drug Discov Today ; 26(2): 577-584, 2021 02.
Article in English | MEDLINE | ID: mdl-33232841

ABSTRACT

The duocarmycins belong to a class of agent that has fascinated scientists for over four decades. Their exquisite potency, unique mechanism of action, and efficacy in multidrug-resistant tumour models makes them attractive to medicinal chemists and drug hunters. However, despite great advances in fine-tuning biological activity through structure-activity relationship studies (SARS), no duocarmycin-based therapeutic has reached clinical approval. In this review, we provide an overview of the most promising strategies currently used and include both tumour-targeted prodrug approaches and antibody-directed technologies.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Duocarmycins/pharmacology , Neoplasms/drug therapy , Animals , Antibodies/immunology , Antineoplastic Agents, Alkylating/chemistry , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Duocarmycins/administration & dosage , Duocarmycins/chemistry , Humans , Prodrugs , Structure-Activity Relationship
16.
Biochem Biophys Res Commun ; 533(3): 325-331, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32958252

ABSTRACT

Busulfan is an alkylating agent used in chemotherapy conditioning regimens prior to hematopoietic stem cell transplantation (HSCT). However, its administration is associated with a great risk of adverse toxicities, which have been historically attributed to busulfan's mechanism of non-specific DNA alkylation. A phase II generated metabolite of busulfan, EdAG (γ-glutamyldehydroalanylglycine), is a dehydroalanine analog of glutathione (GSH) with an electrophilic moiety, suggesting it may bind to proteins and disrupt biological function. However, EdAG's reactions with common cellular thiols such as glutathione (GSH) and l-cysteine are understudied, along with possible inhibition of glutathionylation-dependent enzymes (with active site cysteine residues). We established a physiologically-relevant in vitro model to readily measure thiol loss over time. Using this model, we compared the apparent rates of thiol depletion in the presence of EdAG or arecoline, a toxic constituent of the areca (betel) nut and known GSH depletor. Simulated kinetic modeling revealed that the mean (±SE) alpha (α) second order rate constants describing GSH and l-cysteine depletion in the presence of EdAG were 0.00522 (0.00845) µM-1∙min-1 and 0.0207 (0.00721) µM-1∙min-1, respectively; in the presence of arecoline, the apparent rates of depletion were 0.0619 (0.009) µM-1∙min-1 and 0.2834 (0.0637) µM-1∙min-1 for GSH and l-cysteine, respectively. Under these experimental conditions, we conclude that EdAG was a weaker electrophile than arecoline. Arecoline and EdAG both depleted apparent l-cysteine concentrations to a much greater extent than GSH, approximately 4.58-fold and 3.97-fold change greater, respectively. EdAG modestly inhibited (∼20%) the human thioredoxin-1 (hTrx-1) catalyzed reduction of insulin with a mean IC50 of 93 µM [95% CI: 78.6-110 µM). In summary, EdAG's ability to spontaneously react with endogenous thiols and inhibit hTrx-1 are potentially biochemically relevant in humans. These findings continue to support the growing concept that EdAG, an underrecognized phase II metabolite of busulfan, plays a role in untoward cellular toxicities during busulfan pharmacotherapy.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Arecoline/chemistry , Busulfan/chemistry , Glutathione/analogs & derivatives , Glutathione/chemistry , Thioredoxins/chemistry , Arecoline/antagonists & inhibitors , Biotransformation , Cysteine/antagonists & inhibitors , Cysteine/chemistry , Glutathione/antagonists & inhibitors , Humans , Kinetics , Solutions , Thioredoxins/antagonists & inhibitors , Water/chemistry
17.
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
18.
Biomed Pharmacother ; 129: 110443, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32593130

ABSTRACT

The present study aims at designing a biodegradable and biocompatible nanocarrier using gelatin and reduced graphene oxide nanosheets functionalized with folic acid, for release of chlorambucil drug in controlled manner and achieving high loading efficiency. From scanning electron microscopic studies small pore like structure with rough and thick morphology on the plane of graphene oxide is clearly visible indicating high loading of drug. Further, Drug loading and encapsulation efficiency, in vitro release studies of the drug from the nanocarrier at different concentrations of reduced graphene oxide, different pH were studied. The mean particle size, entrapment efficiency (%) of optimized folic acid functionalized gelatin-graphene oxide formulation was observed to be 300 nm and 56% respectively. From the release studies it is clear that, after 24 h the release rate of the drug was found to be higher at acidic conditions compared to neutral conditions. It was found that 62.1% and 82% of the total bound drug was released from the nanocarrier at pH 5.4 and pH 1.2 respectively. Besides, under neutral conditions (pH 7.4), 43.7% of the total bound drug was released from the nanocarrier in the first 24 h. The % cell viability of free drug, drug loaded nanocomposites against human cervical adenocarcinoma cell line was found to be 11.7% and 28% respectively at the dose of 500 µg mL-1 after 24 h. IC50 values also manifest the significantly lower cytotoxicity of drug loaded nanocarrier (IC50 = 125.9 µg/mL) as compared to free-drug (IC50 = 86 µg/mL). For FAGGO, CLB and CLB-FAGGO the values of mean ± std. deviation were found to be 71.80 ± 6.66; 48.71 ± 23.15; 55.48 ± 19.65 respectively. The unique properties exhibited by biodegradable polymer like gelatin and carbon based materials such as graphene offers an excellent applications in biomedical field.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Alkylating/pharmacology , Chlorambucil/pharmacology , Drug Carriers , Graphite/chemistry , Nanoparticles , Uterine Cervical Neoplasms/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents, Alkylating/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Chlorambucil/chemistry , Delayed-Action Preparations , Dose-Response Relationship, Drug , Drug Compounding , Drug Liberation , Female , Folic Acid/chemistry , Gelatin/chemistry , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Kinetics , Uterine Cervical Neoplasms/pathology
19.
Sci Rep ; 10(1): 4479, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32161295

ABSTRACT

Chemical modification of known, effective drugs is one method to improve chemotherapy. Thus, the object of this study was to generate melphalan derivatives with improved cytotoxic activity in human cancer cells (RPMI8226, HL60 and THP1). Several melphalan derivatives were synthesised, modified in their two important functional groups. Nine analogues were tested, including melphalan compounds modified: only at the amino group, by replacing the amine with an amidine group containing a morpholine ring (MOR-MEL) or with an amidino group and dipropyl chain (DIPR-MEL); only at the carboxyl group to form methyl and ethyl esters of melphalan (EM-MEL, EE-MEL); and in a similar manner at both functional groups (EM-MOR-MEL, EE-MOR-MEL, EM-DIPR-MEL, EE-DIPR-MEL). Melphalan derivatives were evaluated for cytotoxicity (resazurin viability assay), genotoxicity (comet assay) and the ability to induce apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labelling, TUNEL, phosphatidylserine externalisation, chromatin condensation, activity of caspases 3/7, 8 and 9 and intracellular concentration of calcium ions) in comparison with the parent drug. Almost all derivatives, with the exception of MOR-MEL and DIPR-MEL, were found to be more toxic than melphalan in all cell lines evaluated. Treatment of cultures with the derivatives generated a significant higher level of DNA breaks compared to those treated with melphalan, especially after longer incubation times. In addition, all the melphalan derivatives demonstrated a high apoptosis-inducing ability in acute monocytic and promyelocytic leukemia cells. This study showed that the mechanism of action of the tested compounds differed depending on the cell line, and allowed the selection of the most active compounds for further, more detailed investigations.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Melphalan/chemistry , Melphalan/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Calcium/metabolism , Caspases/metabolism , Cell Survival , DNA Damage/drug effects , Drug Development , Drug Screening Assays, Antitumor , Hematologic Neoplasms/drug therapy , Humans , Melphalan/analogs & derivatives , Melphalan/therapeutic use , Molecular Structure , Phosphatidylserines
20.
Metallomics ; 12(5): 721-731, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32196031

ABSTRACT

A novel cyclometalated gold(iii) complex supported by chlorambucil coupled with phenylpyridine (CHL-N^C) and a hybrid of vitamin B1 with dithiocarbamate (B1-DTC) with the formula [(CHL-N^C)AuIII(B1-DTC)](Cl2), 1, was synthesized and fully characterized using different techniques, including multinuclear NMR, mass spectrometry, and elemental analysis. This complex is water-soluble and stable in a biological environment. This new complex offers a new scaffold to explore the biological properties of gold(iii) complexes as an anticancer drug. The antiproliferative activities of complex 1 and free ligands against breast and colon cancer cells showed auspicious results with IC50 values in the micromolar range for complex 1 and more active than cisplatin and free ligands with selectivity over non-tumorigenic cells human lung fibroblasts, MRC-5. The DNA binding and inhibition of thioredoxin reductase of complex 1 were studied and compared with molecular docking results. Moreover, the Au cellular uptake and apoptosis of this new complex were investigated.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Chlorambucil/pharmacology , Coordination Complexes/pharmacology , Gold/chemistry , Neoplasms/drug therapy , Thiamine/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Chlorambucil/chemistry , Cisplatin/pharmacology , Coordination Complexes/chemistry , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Ligands , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , Neoplasms/pathology , Thiamine/chemistry , Vitamin B Complex/chemistry , Vitamin B Complex/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...