Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.303
Filter
1.
Biophys Chem ; 310: 107256, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38728807

ABSTRACT

Understanding the mechanisms by which drugs interact with cell membranes is crucial for unraveling the underlying biochemical and biophysical processes that occur on the surface of these membranes. Our research focused on studying the interaction between an ester-type derivative of tristearoyl uridine and model cell membranes composed of lipid monolayers at the air-water interface. For that, we selected a specific lipid to simulate nontumorigenic cell membranes, namely 1,2-dihexadecanoyl-sn-glycero-3-phospho-l-serine. We noted significant changes in the surface pressure-area isotherms, with a noticeable shift towards larger areas, which was lower than expected for ideal mixtures, indicating monolayer condensation. Furthermore, the viscoelastic properties of the interfacial film demonstrated an increase in both the elastic and viscous parameters for the mixed film. We also observed structural alterations using vibrational spectroscopy, which revealed an increase in the all-trans to gauche conformers ratio. This confirmed the stiffening effect of the prodrug on the lipid monolayer. In summary, this study indicates that this lipophilic prodrug significantly impacts the lipid monolayer's thermodynamic, rheological, electrical, and molecular characteristics. This information is crucial for understanding how the drug interacts with specific sites on the cellular membrane. It also has implications for drug delivery, as the drug's passage into the cytosol may involve traversing the lipid bilayer.


Subject(s)
Cell Membrane , Prodrugs , Uridine , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Uridine/chemistry , Uridine/pharmacology , Phosphatidylserines/chemistry , Thermodynamics , Surface Properties , Viscosity , Elasticity
2.
Bioorg Med Chem ; 106: 117754, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38728869

ABSTRACT

To improve the biodistribution of the drug in the tumor, a supramolecular prodrug of SN38 was fabricated in situ between endogenous albumin and SN38 prodrug modified with semaglutide side chain. Firstly, SN38 was conjugated with semaglutide side chain and octadecanedioic acid via glycine linkers to obtain SI-Gly-SN38 and OA-Gly-SN38 prodrugs, respectively. Both SI-Gly-SN38 and OA-Gly-SN38 exhibited excellent stability in PBS for over 24 h. Due to the strong binding affinity of the semaglutide side chain with albumin, the plasma half-life of SI-Gly-SN38 was 2.7 times higher than that of OA-Gly-SN38. Furthermore, with addition of HSA, the fluorescence intensity of SI-Gly-SN38 was 4 times higher than that of OA-Gly-SN38, confirming its strong binding capability with HSA. MTT assay showed that the cytotoxicity of SI-Gly-SN38 and OA-Gly-SN38 was higher than that of Irinotecan. Even incubated with HSA, the SI-Gly-SN38 and OA-Gly-SN38 still maintained high cytotoxicity, indicating minimal influence of HSA on their cytotoxicity. In vivo pharmacokinetic studies demonstrated that the circulation half-life of SI-Gly-SN38 was twice that of OA-Gly-SN38. SI-Gly-SN38 exhibited significantly reduced accumulation in the lungs, being only 0.23 times that of OA-Gly-SN38. The release of free SN38 in the lungs from SI-Gly-SN38 was only 0.4 times that from OA-Gly-SN38 and Irinotecan. The SI-Gly-SN38 showed the highest accumulation in tumors. The tumor inhibition rate of SI-Gly-SN38 was 6.42% higher than that of OA-Gly-SN38, and 8.67% higher than that of Irinotecan, respectively. These results indicate that the supramolecular prodrug delivery system can be constructed between SI-Gly-SN38 and endogenous albumin, which improves drug biodistribution in vivo, enhances tumor accumulation, and plays a crucial role in tumor growth inhibition.


Subject(s)
Irinotecan , Prodrugs , Irinotecan/chemistry , Irinotecan/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Animals , Humans , Mice , Tissue Distribution , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Molecular Structure , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects , Cell Line, Tumor , Mice, Inbred BALB C , Mice, Nude , Albumins/chemistry , Male , Structure-Activity Relationship , Serum Albumin, Human/chemistry , Glucagon-Like Peptides
3.
ACS Appl Mater Interfaces ; 16(20): 25665-25675, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38735053

ABSTRACT

Tumor-associated macrophages (TAMs) usually adopt a tumor-promoting M2-like phenotype, which largely impedes the immune response and therapeutic efficacy of solid tumors. Repolarizing TAMs from M2 to the antitumor M1 phenotype is crucial for reshaping the tumor immunosuppressive microenvironment (TIME). Herein, we developed self-assembled nanoparticles from the polymeric prodrug of resiquimod (R848) to reprogram the TIME for robust cancer immunotherapy. The polymeric prodrug was constructed by conjugating the R848 derivative to terminal amino groups of the linear dendritic polymer composed of linear poly(ethylene glycol) and lysine dendrimer. The amphiphilic prodrug self-assembled into nanoparticles (PLRS) of around 35 nm with a spherical morphology. PLRS nanoparticles could be internalized by antigen-presenting cells (APCs) in vitro and thus efficiently repolarized macrophages from M2 to M1 and facilitated the maturation of APCs. In addition, PLRS significantly inhibited tumor growth in the 4T1 orthotopic breast cancer model with much lower systemic side effects. Mechanistic studies suggested that PLRS significantly stimulated the TIME by repolarizing TAMs into the M1 phenotype and increased the infiltration of cytotoxic T cells into the tumor. This study provides an effective polymeric prodrug-based strategy to improve the therapeutic efficacy of R848 in cancer immunotherapy.


Subject(s)
Imidazoles , Immunotherapy , Nanoparticles , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use , Animals , Mice , Imidazoles/chemistry , Imidazoles/pharmacology , Nanoparticles/chemistry , Female , Mice, Inbred BALB C , Cell Line, Tumor , Humans , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , RAW 264.7 Cells , Polyethylene Glycols/chemistry , Tumor Microenvironment/drug effects , Dendrimers/chemistry , Dendrimers/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism
4.
J Med Chem ; 67(10): 8296-8308, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38739678

ABSTRACT

Platinum-drug-based chemotherapy in clinics has achieved great success in clinical malignancy therapy. However, unpredictable off-target toxicity and the resulting severe side effects in the treatment are still unsolved problems. Although metabolic glycan labeling-mediated tumor-targeted therapy has been widely reported, less selective metabolic labeling in vivo limited its wide application. Herein, a novel probe of B-Ac3ManNAz that is regulated by reactive oxygen species in tumor cells is introduced to enhance the recognition and cytotoxicity of DBCO-modified oxaliplatin(IV) via bioorthogonal chemistry. B-Ac3ManNAz was synthesized from Ac4ManNAz by incorporation with 4-(hydroxymethyl) benzeneboronic acid pinacol ester (HBAPE) at the anomeric position, which is confirmed to be regulated by ROS and could robustly label glycans on the cell surface. Moreover, N3-treated tumor cells could enhance the tumor accumulation of DBCO-modified oxaliplatin(IV) via click chemistry meanwhile reduce the off-target distribution in normal tissue. Our strategy provides an effective metabolic precursor for tumor-specific labeling and targeted cancer therapies.


Subject(s)
Antineoplastic Agents , Oxaliplatin , Polysaccharides , Prodrugs , Reactive Oxygen Species , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Oxaliplatin/pharmacology , Oxaliplatin/chemistry , Humans , Reactive Oxygen Species/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice , Cell Line, Tumor , Mice, Inbred BALB C , Mice, Nude
5.
J Med Chem ; 67(9): 7470-7486, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38690769

ABSTRACT

We assessed factors that determine the tissue-specific bioactivation of ProTide prodrugs by comparing the disposition and activation of remdesivir (RDV), its methylpropyl and isopropyl ester analogues (MeRDV and IsoRDV, respectively), the oral prodrug GS-621763, and the parent nucleotide GS-441524 (Nuc). RDV and MeRDV yielded more active metabolite remdesivir-triphosphate (RDV-TP) than IsoRDV, GS-621763, and Nuc in human lung cell models due to superior cell permeability and higher susceptivity to cathepsin A. Intravenous administration to mice showed that RDV and MeRDV delivered significantly more RDV-TP to the lung than other compounds. Nevertheless, all four ester prodrugs exhibited very low oral bioavailability (<2%), with Nuc being the predominant metabolite in blood. In conclusion, ProTides prodrugs, such as RDV and MeRDV, are more efficient in delivering active metabolites to the lung than Nuc, driven by high cell permeability and susceptivity to cathepsin A. Optimizing ProTides' ester structures is an effective strategy for enhancing prodrug activation in the lung.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents , Cathepsin A , Lung , Prodrugs , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Animals , Mice , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Humans , Cathepsin A/metabolism , Lung/metabolism , Cell Membrane Permeability/drug effects , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacokinetics , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/pharmacokinetics , Alanine/metabolism , Alanine/pharmacology , Permeability , ProTides
6.
ACS Appl Bio Mater ; 7(5): 3431-3440, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38697834

ABSTRACT

Light-induced release of cisplatin from Pt(IV) prodrugs represents a promising approach for precise control over the antiproliferative activity of Pt-based chemotherapeutic drugs. This method has the potential to overcome crucial drawbacks of conventional cisplatin therapy, such as high general toxicity toward healthy organs and tissues. Herein, we report two Pt(IV) prodrugs with BODIPY-based photoactive ligands Pt-1 and Pt-2, which were designed using carbamate and triazole linkers, respectively. Both prodrugs demonstrated the ability to release cisplatin under blue light irradiation without the requirement of an external reducing agent. Dicarboxylated Pt-2 prodrug turned out to be more stable in the dark and more sensitive to light than its monocarbamate Pt-1 counterpart; these observations were explained using DFT calculations. The investigation of the photoreduction mechanism of Pt-1 and Pt-2 prodrugs using DFT modeling and ΔG0 PET estimation suggests that the photoinduced electron transfer from the singlet excited state of the BODIPY axial ligand to the Pt(IV) center is the key step in the light-induced release of cisplatin from the complexes. Cytotoxicity studies demonstrated that both prodrugs were nontoxic in the dark and toxic to MCF-7 cells under low-dose irradiation with blue light, and the observed effect was solely due to the cisplatin release from the Pt(IV) prodrugs. Our research presents an elegant synthetic approach to light-activated Pt(IV) prodrugs and presents findings that may contribute to the future rational design of photoactivatable Pt(IV) prodrugs.


Subject(s)
Antineoplastic Agents , Drug Screening Assays, Antitumor , Light , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Molecular Structure , Materials Testing , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemical synthesis , Cell Survival/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cisplatin/chemistry , Particle Size , Boron Compounds/chemistry , Boron Compounds/pharmacology , Boron Compounds/chemical synthesis , Photochemical Processes , Density Functional Theory
7.
Carbohydr Polym ; 338: 122196, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38763723

ABSTRACT

Triple negative breast cancer (TNBC) represents the most aggressive and heterogenous disease, and combination therapy holds promising potential. Here, an enzyme-responsive polymeric prodrug with self-assembly properties was synthesized for targeted co-delivery of paclitaxel (PTX) and ursolic acid (UA). Hyaluronic acid (HA) was conjugated with UA, yielding an amphiphilic prodrug with 13.85 mol% UA and a CMC of 32.3 µg/mL. The HA-UA conjugate exhibited ∼14 % and 47 % hydrolysis at pH 7.4 and in tumor cell lysate. HA-UA/PTX NPs exhibited a spherical structure with 173 nm particle size, and 0.15 PDI. The nanoparticles showed high drug loading (11.58 %) and entrapment efficiency (76.87 %) of PTX. Release experiments revealed accelerated drug release (∼78 %) in the presence of hyaluronidase enzyme. Cellular uptake in MDA-MB-231 cells showed enhanced uptake of HA-UA/PTX NPs through CD44 receptor-mediated endocytosis. In vitro, HA-UA/PTX NPs exhibited higher cytotoxicity, apoptosis, and mitochondrial depolarization compared to PTX alone. In vivo, HA-UA/PTX NPs demonstrated improved pharmacokinetic properties, with 2.18, 2.40, and 2.35-fold higher AUC, t1/2, and MRT compared to free PTX. Notably, HA-UA/PTX NPs exhibited superior antitumor efficacy with a 90 % tumor inhibition rate in 4T1 tumor model and low systemic toxicity, showcasing their significant potential as carriers for TNBC combination therapy.


Subject(s)
Hyaluronic Acid , Nanoparticles , Paclitaxel , Triple Negative Breast Neoplasms , Triterpenes , Ursolic Acid , Triterpenes/chemistry , Triterpenes/pharmacology , Hyaluronic Acid/chemistry , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Humans , Nanoparticles/chemistry , Animals , Female , Paclitaxel/pharmacology , Paclitaxel/chemistry , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Cell Line, Tumor , Drug Liberation , Apoptosis/drug effects , Mice , Drug Carriers/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Mice, Inbred BALB C , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemistry
8.
J Labelled Comp Radiopharm ; 67(5): 186-196, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38661253

ABSTRACT

Malaria continues to be a serious and debilitating disease. The emergence and spread of high-level resistance to multiple antimalarial drugs by Plasmodium falciparum has brought about an urgent need for new treatments that will be active against multidrug resistant malaria infections. One such treatment, ELQ-331 (MMV-167), an alkoxy carbonate prodrug of 4(1H)-quinolone ELQ-300, is currently in preclinical development with the Medicines for Malaria Venture. Clinical development of ELQ-331 or similar compounds will require the availability of isotopically labeled analogs. Unfortunately, a suitable method for the deuteration of these important compounds was not found in the literature. Here, we describe a facile and scalable method for the deuteration of 4(1H)-quinolone ELQ-300, its alkoxycarbonate prodrug ELQ-331, and their respective N-oxides using deuterated acetic acid.


Subject(s)
Chemistry Techniques, Synthetic , Deuterium , Quinolones , Quinolones/chemical synthesis , Quinolones/chemistry , Deuterium/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antimalarials/pharmacology
9.
J Inorg Biochem ; 256: 112567, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38669911

ABSTRACT

AS101 (Ammonium trichloro (dioxoethylene-O,O') tellurate) is an important hypervalent Te-based prodrug. Recently, we started a systematic investigation on AS101 with the aim to correlate its promising biological effects as a potent immunomodulator drug with multiple medicinal applications and its specific chemical properties. To date, a substantial agreement on the rapid conversion of the initial AS101 species into the corresponding TeOCl3- anion does exist, and this latter species is reputed as the pharmacologically active one. However, we realized that TeOCl3- could quickly undergo further steps of conversion in an aqueous medium, eventually producing the TeO2 species. Using a mixed experimental and theoretical investigation approach, we characterized the conversion process leading to TeO2 occurring both in pure water and in reference buffers at physiological-like pH. Our findings may offer a valuable "chemical tool" for a better description, interpretation -and optimization- of the mechanism of action of AS101 and Te-based compounds. This might be a starting point for improved AS101-based medicinal application.


Subject(s)
Prodrugs , Prodrugs/pharmacology , Prodrugs/chemistry
10.
J Control Release ; 369: 622-629, 2024 May.
Article in English | MEDLINE | ID: mdl-38604383

ABSTRACT

Enhancing the delivery and release efficiency of hydroxyl agents, constrained by high pKa values and issues of release rate or unstable linkage, is a critical challenge. To address this, a self-immolative linker, composed of a modifiable p-hydroxybenzyl ether and a fast cyclization adapter (N-(ortho-hydroxyphenyl)-N-methylcarbamate) was strategically designed, for the synthesis of prodrugs. The innovative linker not only provides a side chain modification but also facilitates the rapid release of the active payloads, thereby enabling precise drug delivery. Particularly, five prodrug model compounds (J1, J2, J3, J5 and J6) were synthesized to evaluate the release rates by using ß-glucuronic acid as trigger and five hydroxyl compounds as model payloads. Significantly, all prodrug model compounds could efficiently release the hydroxyl payloads under the action of ß-glucuronidase, validating the robustness of the linker. And then, to assess the drug delivery and release efficiency using endogenous albumin as a transport vehicle, J1148, a SN38 prodrug modified with maleimide side chain was synthesized. Results demonstrated that J1148 covalently bound to plasma albumin through in situ Michael addition, effectively targeting the tumor microenvironment. Activated by ß-glucuronidase, J1148 underwent a classical 1, 6-elimination, followed by rapid cyclization of the adapter, thereby releasing SN38. Impressively, J1148 showed excellent therapeutic efficacy against human colonic cancer xenograft model, leading to a significant reduction or even disappearance of tumors (3/6 of mice cured). These findings underscore the potential of the designed linker in the delivery system of hydroxyl agents, positioning it at the forefront of advancements in drug delivery technology.


Subject(s)
Drug Delivery Systems , Irinotecan , Prodrugs , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Animals , Humans , Irinotecan/administration & dosage , Irinotecan/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/chemistry , Drug Liberation , Mice, Nude , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Female , Mice , Albumins/administration & dosage , Albumins/chemistry , Glucuronidase/metabolism , Mice, Inbred BALB C
11.
Int J Biol Macromol ; 268(Pt 2): 131641, 2024 May.
Article in English | MEDLINE | ID: mdl-38641277

ABSTRACT

Redox nanoparticles have been extensively developed for chemotherapy. However, the intracellular oxidative stress induced by constant aberrant glutathione (GSH), reactive oxygen species (ROS) and gamma-glutamyl transpeptidase (GGT) homeostasis remains the primary cause of evading tumor apoptosis. Herein, an oxidative stress-amplification strategy was designed using a pH-GSH-H2O2-GGT sensitive nano-prodrug for precise synergistic chemotherapy. The disulfide bond- conjugated doxorubicin prodrug (DOX-ss) was constructed as a GSH-scavenger. Then, phenylboronic acid (PBA), DOX-ss and poly (γ-glutamic acid) (γ-PGA) were successively conjugated using chitosan oligosaccharide (COS) to obtain the nano-prodrug PBA-COS-ss-DOX/γ-PGA. The PBA-COS-ss-DOX/γ-PGA prodrug could tightly attach to the polymer chain segment by atom transfer radical polymerization. Simultaneously, the drug interacted relatively weakly with the polymer by encapsulating ionic crosslinkers in DOX@PBA-COS/γ-PGA. The disulfide bond of the DOX-ss prodrug as a GSH-scavenger could be activated using overexpressed GSH to release DOX. Particularly, PBA-COS-ss-DOX/γ-PGA could prevent premature drug leakage and facilitate DOX delivery by GGT-targeting and intracellular H2O2-cleavable linker in human hepatocellular carcinoma (HepG2) cells. Concurrently, the nano-prodrug induced strong oxidative stress and tumor cell apoptosis. Collectively, the pH-GSH-H2O2-GGT responsive nano-prodrug shows potential for synergistic tumor therapy.


Subject(s)
Chitosan , Doxorubicin , Nanoparticles , Oligosaccharides , Oxidative Stress , Prodrugs , Chitosan/chemistry , Oxidative Stress/drug effects , Prodrugs/chemistry , Prodrugs/pharmacology , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Nanoparticles/chemistry , Glutathione/metabolism , Glutathione/chemistry , Hep G2 Cells , Reactive Oxygen Species/metabolism , Polyglutamic Acid/chemistry , Polyglutamic Acid/analogs & derivatives , Hydrogen Peroxide/chemistry , Drug Liberation , Drug Carriers/chemistry , Apoptosis/drug effects , gamma-Glutamyltransferase/metabolism , Boronic Acids/chemistry , Hydrogen-Ion Concentration
12.
J Chem Inf Model ; 64(9): 3733-3743, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38683970

ABSTRACT

Some of the well-known drawbacks of clinically approved PtII complexes can be overcome using six-coordinate PtIV complexes as inert prodrugs, which release the corresponding four-coordinate active PtII species upon reduction by cellular reducing agents. Therefore, the key factor of PtIV prodrug mechanism of action is their tendency to be reduced which, when the involved mechanism is of outer-sphere type, is measured by the value of the reduction potential. Machine learning (ML) models can be used to effectively capture intricate relationships within PtIV complex data, leading to highly accurate predictions of reduction potentials and other properties, and offering significant insights into their electrochemical behavior and potential applications. In this study, a machine learning-based approach for predicting the reduction potentials of PtIV complexes based on relevant molecular descriptors is presented. Leveraging a data set of experimentally determined reduction potentials and a diverse range of molecular descriptors, the proposed model demonstrates remarkable predictive accuracy (MSE = 0.016 V2, RMSE = 0.13 V, R2 = 0.92). Ab initio calculations and a set of different machine learning algorithms and feature engineering techniques have been employed to systematically explore the relationship between molecular structure and similarity and reduction potential. Specifically, it has been investigated whether the reduction potential of these compounds can be described by combining ML models across different combinations of constitutional, topological, and electronic molecular descriptors. Our results not only provide insights into the crucial factors influencing reduction potentials but also offer a rapid and effective tool for the rational design of PtIV complexes with tailored electrochemical properties for pharmaceutical applications. This approach has the potential to significantly expedite the development and screening of novel PtIV prodrug candidates. The analysis of principal components and key features extracted from the model highlights the significance of structural descriptors of the 2D Atom Pairs type and the lowest unoccupied molecular orbital energy. Specifically, with just 20 appropriately selected descriptors, a notable separation of complexes based on their reduction potential value is achieved.


Subject(s)
Machine Learning , Oxidation-Reduction , Coordination Complexes/chemistry , Prodrugs/chemistry , Models, Molecular
13.
Eur J Med Chem ; 271: 116412, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38643669

ABSTRACT

New acyclic pyrimidine nucleoside phosphonate prodrugs with a 4-(2,4-diaminopyrimidin-6-yl)oxy-but-2-enyl]phosphonic acid skeleton (O-DAPy nucleobase) were prepared through a convergent synthesis by olefin cross-metathesis as the key step. Several acyclic nucleoside 4-(2,4-diaminopyrimidin-6-yl)oxy-but-2-enyl]phosphonic acid prodrug exhibited in vitro antiviral activity in submicromolar or nanomolar range against varicella zoster virus (VZV), human cytomegalovirus (HCMV), human herpes virus type 1 (HSV-1) and type 2 (HSV-2), and vaccinia virus (VV), with good selective index (SI). Among them, the analogue 9c (LAVR-289) proved markedly inhibitory against VZV wild-type (TK+) (EC50 0.0035 µM, SI 740) and for thymidine kinase VZV deficient strains (EC50 0.018 µM, SI 145), with a low morphological toxicity in cell culture at 100 µM and acceptable cytostatic activity resulting in excellent selectivity. Compound 9c exhibited antiviral activity against HCMV (EC50 0.021 µM) and VV (EC50 0.050 µM), as well as against HSV-1 (TK-) (EC50 0.0085 µM). Finally, LAVR-289 (9c) deserves further (pre)clinical investigations as a potent candidate broad-spectrum anti-herpesvirus drug.


Subject(s)
Antiviral Agents , DNA Viruses , Microbial Sensitivity Tests , Prodrugs , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Prodrugs/chemistry , Humans , DNA Viruses/drug effects , Structure-Activity Relationship , Herpesvirus 1, Human/drug effects , Molecular Structure , Herpesvirus 3, Human/drug effects , Organophosphonates/pharmacology , Organophosphonates/chemistry , Organophosphonates/chemical synthesis , Cytomegalovirus/drug effects , Dose-Response Relationship, Drug , Vaccinia virus/drug effects , Herpesvirus 2, Human/drug effects
14.
J Med Chem ; 67(9): 7033-7047, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38634331

ABSTRACT

A brand-new enhanced starvation is put forward to trigger sensitized chemotherapy: blocking tumor-relation blood vessel formation and accelerating nutrient degradation and efflux. Following this concept, two cisplatin-like gemfibrozil-derived Pt(IV) prodrugs, GP and GPG, are synthesized. GP and GPG had nanomolar IC50 against A2780 cells and higher selectivity against normal cells than cisplatin. Bioactivity results confirmed that GP and GPG highly accumulated in cells and induced DNA damage, G2-phase arrest, and p53 expression. Besides, they could increase ROS and MDA levels and reduce mitochondrial membrane potential and Bcl-2 expression to promote cell apoptosis. In vivo, GP showed superior antitumor activity in A2780 tumor-bearing mice with no observable tissue damage. Mechanistic studies suggested that highly selective chemotherapy could be due to the new enhanced starvation effect: blocking vasculature formation via inhibiting the CYP2C8/EETs pathway and VEGFR2, NF-κB, and COX-2 expression and cholesterol efflux and degradation acceleration via increasing ABCA1 and PPARα.


Subject(s)
Antineoplastic Agents , Gemfibrozil , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Gemfibrozil/pharmacology , Mice, Inbred BALB C , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/chemical synthesis
15.
ACS Infect Dis ; 10(5): 1679-1695, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38581700

ABSTRACT

Linezolid is a drug with proven human antitubercular activity whose use is limited to highly drug-resistant patients because of its toxicity. This toxicity is related to its mechanism of action─linezolid inhibits protein synthesis in both bacteria and eukaryotic mitochondria. A highly selective and potent series of oxazolidinones, bearing a 5-aminomethyl moiety (in place of the typical 5-acetamidomethyl moiety of linezolid), was identified. Linezolid-resistant mutants were cross-resistant to these molecules but not vice versa. Resistance to the 5-aminomethyl molecules mapped to an N-acetyl transferase (Rv0133) and these mutants remained fully linezolid susceptible. Purified Rv0133 was shown to catalyze the transformation of the 5-aminomethyl oxazolidinones to their corresponding N-acetylated metabolites, and this transformation was also observed in live cells of Mycobacterium tuberculosis. Mammalian mitochondria, which lack an appropriate N-acetyltransferase to activate these prodrugs, were not susceptible to inhibition with the 5-aminomethyl analogues. Several compounds that were more potent than linezolid were taken into C3HeB/FeJ mice and were shown to be highly efficacious, and one of these (9) was additionally taken into marmosets and found to be highly active. Penetration of these 5-aminomethyl oxazolidinone prodrugs into caseum was excellent. Unfortunately, these compounds were rapidly converted into the corresponding 5-alcohols by mammalian metabolism which retained antimycobacterial activity but resulted in substantial mitotoxicity.


Subject(s)
Antitubercular Agents , Mycobacterium tuberculosis , Oxazolidinones , Prodrugs , Prodrugs/pharmacology , Prodrugs/chemistry , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Mycobacterium tuberculosis/drug effects , Oxazolidinones/pharmacology , Oxazolidinones/chemistry , Animals , Microbial Sensitivity Tests , Mice , Humans , Linezolid/pharmacology , Linezolid/chemistry , Drug Resistance, Bacterial , Mitochondria/drug effects , Mitochondria/metabolism
16.
Colloids Surf B Biointerfaces ; 238: 113906, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615388

ABSTRACT

Combination chemotherapy has been recognized as a more powerful strategy for tumor treatment rather than the single chemotherapy. However, the interactive mechanism of the two hydrophobic chemotherapeutic drugs has not been explored by now. Aiming for a better synergistic effect, such interactive mechanism was investigated in the present work, by designing CPT@DOX-DPUTEA-PEG nanomedicine with encapsulated camptothecin (CPT) and conjugated doxorubicin (DOX). The synergistic controlled drug release effect was found for the two drugs loaded on the different sites of the dendritic polyurethane core. Synergism was achieved on the HepG2 cells with a combination index (CI) of 0.58 in the in vitro cellular experiments. The results demonstrated the promising application of the unimolecular micelles-based nanomedicine with independently loading of two hydrophobic chemotherapeutic drugs.


Subject(s)
Camptothecin , Doxorubicin , Drug Liberation , Micelles , Prodrugs , Doxorubicin/pharmacology , Doxorubicin/chemistry , Camptothecin/pharmacology , Camptothecin/chemistry , Humans , Hydrogen-Ion Concentration , Hep G2 Cells , Prodrugs/chemistry , Prodrugs/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Polymers/chemistry , Cell Survival/drug effects , Dendrimers/chemistry , Drug Delivery Systems , Drug Synergism , Polyethylene Glycols/chemistry
17.
Bioorg Med Chem Lett ; 104: 129729, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38583786

ABSTRACT

Aptamers have shown significant potential in treating diverse diseases. However, challenges such as stability and drug delivery limited their clinical application. In this paper, the development of AS1411 prodrug-type aptamers for tumor treatment was introduced. A Short oligonucleotide was introduced at the end of the AS1411 sequence with a disulfide bond as responsive switch. The results indicated that the aptamer prodrugs not only enhanced the stability of the aptamer against nuclease activity but also facilitated binding to serum albumin. Furthermore, in the reducing microenvironment of tumor cells, disulfide bonds triggered drug release, resulting in superior therapeutic effects in vitro and in vivo compared to original drugs. This paper proposes a novel approach for optimizing the structure of nucleic acid drugs, that promises to protect other oligonucleotides or secondary structures, thus opening up new possibilities for nucleic acid drug design.


Subject(s)
Antineoplastic Agents , Aptamers, Nucleotide , Prodrugs , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Cell Line, Tumor , Disulfides/chemistry , Drug Delivery Systems , Nucleic Acids/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Stability
18.
J Med Chem ; 67(9): 7301-7311, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38635879

ABSTRACT

Although the selective and effective clearance of senescent cancer cells can improve cancer treatment, their development is confronted by many challenges. As part of efforts designed to overcome these problems, prodrugs, whose design is based on senescence-associated ß-galactosidase (SA-ß-gal), have been developed to selectively eliminate senescent cells. However, chemotherapies relying on targeted molecular inhibitors as senolytic drugs can induce drug resistance. In the current investigation, we devised a new strategy for selective degradation of target proteins in senescent cancer cells that utilizes a prodrug composed of the SA-ß-gal substrate galactose (galacto) and the proteolysis-targeting chimeras (PROTACs) as senolytic agents. Prodrugs Gal-ARV-771 and Gal-MS99 were found to display senolytic indexes higher than those of ARV-771 and MS99. Significantly, results of in vivo studies utilizing a human lung A549 xenograft mouse model demonstrated that concomitant treatment with etoposide and Gal-ARV-771 leads to a significant inhibition of tumor growth without eliciting significant toxicity.


Subject(s)
Cellular Senescence , Galactose , Prodrugs , Proteolysis , Humans , Animals , Cellular Senescence/drug effects , Galactose/chemistry , Galactose/pharmacology , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/therapeutic use , Mice , Proteolysis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Xenograft Model Antitumor Assays , beta-Galactosidase/metabolism , Mice, Nude , Cell Line, Tumor , Cell Proliferation/drug effects , A549 Cells , Etoposide/pharmacology , Senotherapeutics/pharmacology , Senotherapeutics/chemistry , Proteolysis Targeting Chimera
19.
Nat Rev Drug Discov ; 23(5): 365-380, 2024 May.
Article in English | MEDLINE | ID: mdl-38565913

ABSTRACT

Prodrugs are derivatives with superior properties compared with the parent active pharmaceutical ingredient (API), which undergo biotransformation after administration to generate the API in situ. Although sharing this general characteristic, prodrugs encompass a wide range of different chemical structures, therapeutic indications and properties. Here we provide the first holistic analysis of the current landscape of approved prodrugs using cheminformatics and data science approaches to reveal trends in prodrug development. We highlight rationales that underlie prodrug design, their indications, mechanisms of API release, the chemistry of promoieties added to APIs to form prodrugs and the market impact of prodrugs. On the basis of this analysis, we discuss strengths and limitations of current prodrug approaches and suggest areas for future development.


Subject(s)
Prodrugs , Prodrugs/pharmacology , Prodrugs/chemistry , Humans , Animals , Drug Design , Drug Development/methods
20.
J Med Chem ; 67(8): 6410-6424, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38592014

ABSTRACT

We report two novel prodrug Pt(IV) complexes with bis-organosilane ligands in axial positions: cis-dichloro(diamine)-trans-[3-(triethoxysilyl)propylcarbamate]platinum(IV) (Pt(IV)-biSi-1) and cis-dichloro(diisopropylamine)-trans-[3-(triethoxysilyl) propyl carbamate]platinum(IV) (Pt(IV)-biSi-2). Pt(IV)-biSi-2 demonstrated enhanced in vitro cytotoxicity against colon cancer cells (HCT 116 and HT-29) compared with cisplatin and Pt(IV)-biSi-1. Notably, Pt(IV)-biSi-2 exhibited higher cytotoxicity toward cancer cells and lower toxicity on nontumorigenic intestinal cells (HIEC6). In preclinical mouse models of colorectal cancer, Pt(IV)-biSi-2 outperformed cisplatin in reducing tumor growth at lower concentrations, with reduced side effects. Mechanistically, Pt(IV)-biSi-2 induced permanent DNA damage independent of p53 levels. DNA damage such as double-strand breaks marked by histone gH2Ax was permanent after treatment with Pt(IV)-biSi-2, in contrast to cisplatin's transient effects. Pt(IV)-biSi-2's faster reduction to Pt(II) species upon exposure to biological reductants supports its superior biological response. These findings unveil a novel strategy for designing Pt(IV) anticancer prodrugs with enhanced activity and specificity, offering therapeutic opportunities beyond conventional Pt drugs.


Subject(s)
Antineoplastic Agents , Organoplatinum Compounds , Prodrugs , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/chemical synthesis , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Animals , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/chemical synthesis , Ligands , Mice , Cell Line, Tumor , Silanes/chemistry , Silanes/pharmacology , Structure-Activity Relationship , Drug Screening Assays, Antitumor , HT29 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...