Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 464
1.
AAPS PharmSciTech ; 25(5): 116, 2024 May 20.
Article En | MEDLINE | ID: mdl-38769223

Oral dispersible films have received broad interest due to fast drug absorption and no first-path metabolism, leading to high bioavailability and better patient compliance. Saxagliptin (SXG) is an antidiabetic drug that undergoes first-path metabolism, resulting in a less active metabolite, so the development of SXG oral dispersible films (SXG-ODFs) improves SXG bioavailability. The formula optimisation included a response surface experimental design and the impact of three formulation factors, the type and concentration of polymer and plasticiser concentration on in-vitro disintegration time and folding endurance. Two optimised SXG-ODFs prepared using either polyvinyl alcohol (PVA) or hydroxypropyl methylcellulose were investigated. SXG-ODFs prepared with PVA demonstrated a superior rapid disintegration time, ranging from 17 to 890 s, with the fastest disintegration time recorded at 17 s. These short durations can be attributed to the hydrophilic nature of PVA, facilitating rapid hydration and disintegration upon contact with saliva. Additionally, PVA-based films displayed remarkable folding endurance, surpassing 200 folds without rupture, indicating flexibility and stability. The high tensile strength of PVA-based films further underscores their robust mechanical properties, with tensile strength values reaching up to 4.53 MPa. SXG exhibits a UV absorption wavelength of around 212 nm, posing challenges for traditional quantitative spectrophotometric analysis, so a polyaniline nanoparticles-based solid-contact screen-printed ion-selective electrode (SP-ISE) was employed for the determination of SXG release profile effectively in comparison to HPLC. SP-ISE showed a better real-time release profile of SXG-ODFs, and the optimised formula showed lower blood glucose levels than commercial tablets.


Adamantane , Aniline Compounds , Dipeptides , Drug Liberation , Nanoparticles , Polyvinyl Alcohol , Adamantane/chemistry , Adamantane/analogs & derivatives , Dipeptides/chemistry , Dipeptides/pharmacokinetics , Dipeptides/administration & dosage , Aniline Compounds/chemistry , Nanoparticles/chemistry , Administration, Oral , Polyvinyl Alcohol/chemistry , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Humans , Hypromellose Derivatives/chemistry , Tensile Strength , Chemistry, Pharmaceutical/methods , Biological Availability , Solubility , Electrodes
2.
Toxicol Appl Pharmacol ; 486: 116934, 2024 May.
Article En | MEDLINE | ID: mdl-38663673

The development of diabetes mellitus (DM) is generally accompanied by erectile dysfunction (ED) and pulmonary arterial hypertension (PAH), which increases the use of combination drug therapy and the risk of drug-drug interactions. Saxagliptin for the treatment of DM, sildenafil for the treatment of ED and PAH, and macitentan for the treatment of PAH are all substrates of CYP3A4, which indicates their potential involvement in drug-drug interactions. Therefore, we investigated potential pharmacokinetic interactions between saxagliptin and sildenafil/macitentan. We investigated this speculation both in vitro and in vivo, and explored the underlying mechanism using in vitro hepatic metabolic models and molecular docking assays. The results showed that sildenafil substantially inhibited the metabolism of saxagliptin by occupying the catalytic site of CYP3A4 in a competitive manner, leading to the alterations in the pharmacokinetic properties of saxagliptin in terms of increased maximum plasma concentration (Cmax), area under the plasma concentration-time curve from time 0 to 24 h (AUC(0-t)), area under the plasma concentration-time curve from time 0 extrapolated to infinite time (AUC(0-∞)), decreased clearance rate (CLz/F), and prolonged terminal half-life (t1/2). In contrast, a slight inhibition was observed in saxagliptin metabolism when concomitantly used with macitentan, as no pharmacokinetic parameters were altered, except for CLz/F. Thus, dosage adjustment of saxagliptin may be required in combination with sildenafil to achieve safe therapeutic plasma concentrations and reduce the risk of potential toxicity, but it is not necessary for co-administration with macitentan.


Adamantane , Dipeptides , Drug Interactions , Pyrimidines , Sildenafil Citrate , Sulfonamides , Sildenafil Citrate/pharmacokinetics , Sildenafil Citrate/pharmacology , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Dipeptides/pharmacokinetics , Dipeptides/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Humans , Adamantane/analogs & derivatives , Adamantane/pharmacokinetics , Adamantane/pharmacology , Male , Animals , Cytochrome P-450 CYP3A/metabolism , Molecular Docking Simulation , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacology
3.
J Clin Pharmacol ; 63(7): 848-858, 2023 07.
Article En | MEDLINE | ID: mdl-36869593

The aim of the present study is to develop physiologically based pharmacokinetic (PBPK) models for saxagliptin and its active metabolite, 5-hydroxy saxagliptin, and to predict the effect of coadministration of rifampicin, a strong inducer of cytochrome P450 3A4 enzymes, on the pharmacokinetics of saxagliptin and 5-hydroxy saxagliptin in patients with renal impairment. The PBPK models of saxagliptin and 5-hydroxy saxagliptin were developed and validated in GastroPlus for healthy adults with or without rifampicin and adults with varying renal functions. Then, the effect of renal impairment combined with drug-drug interaction on saxagliptin and 5-hydroxy saxagliptin pharmacokinetics was investigated. The PBPK models successfully predicted the pharmacokinetics. For saxagliptin, the prediction suggests that rifampin greatly weakened the effect of renal impairment on reducing clearance, and the inductive effect of rifampin on parent drug metabolism seems to be increased with an increase in the degree of renal impairment severity. For patients with the same degree of renal impairment, rifampicin would have a slightly synergistic effect on the increase of 5-hydroxy saxagliptin exposure compared with dosed alone. There is an unsignificant decline for the saxagliptin total active moiety exposure values in patients with the same degree of renal impairment. It seems that patients with renal impairment are unlikely to require additional dose adjustments when coadministered with rifampicin, compared with saxagliptin alone. Our study provides a reasonable approach to explore unknown DDI potential in renal impairment.


Adamantane , Rifampin , Adult , Humans , Rifampin/pharmacokinetics , Dipeptides/pharmacokinetics , Drug Interactions , Cytochrome P-450 CYP3A/metabolism , Models, Biological
4.
J Clin Pharmacol ; 62(8): 1018-1029, 2022 08.
Article En | MEDLINE | ID: mdl-35247279

We characterized the impact of chronic kidney disease (CKD) on the cytochrome P450 (CYP) 3A4-mediated metabolism of saxagliptin to its metabolite, 5-hydroxysaxagliptin, using a physiologically based pharmacokinetic (PBPK) model. A PBPK model of saxagliptin and its CYP3A4 metabolite, 5-hydroxysaxagliptin, was constructed and validated for oral doses ranging from 5 to 100 mg. The observed ratios of area under the plasma concentration-time curve (AUC) and maximum plasma concentration (Cmax ) between healthy subjects and subjects with CKD were compared with those predicted using PBPK model simulations. Simulations were performed with virtual CKD populations having decreased CYP3A4 activity (ie, 64%-75% of the healthy subjects' CYP3A4 abundance) and preserved CYP3A4 activity (ie, 100% of the healthy subjects' CYP3A4 abundance). We found that simulations using decreased CYP3A4 activity generally overpredicted the ratios of saxagliptin AUC and Cmax in CKD compared with those using preserved CYP3A4 activity. Similarly, simulations using decreased CYP3A4 activity underpredicted the ratio of 5-hydroxysaxagliptin AUC in moderate and severe CKD compared with simulations using preserved CYP3A4 activity. These findings suggest that decreased CYP3A4 activity in CKD underpredicts saxagliptin clearance compared with that observed clinically. Preserving CYP3A4 activity in CKD more closely estimates saxagliptin clearance and 5-hydroxysaxagliptin exposure changes observed in vivo. Our findings suggest that there is no clinically meaningful impact of CKD on the metabolism of saxagliptin by CYP3A4. Since saxagliptin is not a highly sensitive substrate and validated probe for CYP3A4, this work represents a case study of a CYP3A4 substrate-metabolite pair and is not a generalization for all CYP3A4 substrates.


Adamantane , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A , Dipeptides , Renal Insufficiency, Chronic , Adamantane/analogs & derivatives , Adamantane/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Dipeptides/pharmacokinetics , Drug Interactions , Humans , Models, Biological
5.
Biomed Res Int ; 2021: 1555712, 2021.
Article En | MEDLINE | ID: mdl-34845436

OBJECTIVE: In the recent time, endoradionuclide therapy for metastatic castration-resistant prostate carcinoma employing 177Lu-PSMA-617 has yielded encouraging results and several clinical trials with the agent are currently ongoing. Routine preparation of 177Lu-PSMA-617 patient doses can be made simpler and convenient, if the ingredients essential for radiolabeling are made available in a ready-to-use lyophilized form. METHODS: PSMA-617 freeze-dried kit was formulated and used for the preparation of 177Lu-PSMA-617 clinical dose with high radiochemical purity using low/medium specific activity 177Lu. Detailed radiochemical studies were performed to determine the maximum activity and volume of 177LuCl3, which can be added in the kit for the formulation of 177Lu-PSMA-617. Studies were also performed to determine the shelf life of the kit to ensure its long-term usage. Studies were performed in buffer as well as human serum medium to determine the stability of the 177Lu-PSMA-617 complex after storing in respective media up to 7 days postpreparation. About ten patient doses of 177Lu-PSMA-617 were administered, and posttherapy scans were acquired. RESULTS: The formulated freeze-dried kit of PSMA-617 could be radiolabeled with an average percentage radiochemical purity > 98.53 ± 0.38. The freeze-dried kit was found suitable for tolerating up to 0.5 mL of 177LuCl3 (in 0.01 N HCl) and specific activity of 555 MBq/µg (15 mCi/µg) for the preparation of the patient dose of 177Lu-PSMA-617. The 177Lu-PSMA-617 complex prepared using the freeze-dried kit of PSMA-617 was observed to maintain % radiochemical purity (RCP) of 96.74 ± 0.87 and 94.81 ± 2.66, respectively, even after storing up to 7 days in buffer and human serum, respectively. 177Lu-PSMA-617 prepared using the in-house formulated freeze-dried kit of PSMA-617 exhibited accumulation in metastatic lesions picked up in a pretherapy PET scan. Reduction in number as well as size of lesions was observed in posttherapy scans acquired after two months of administering the first therapeutic dose of 177Lu-PSMA-617. CONCLUSIONS: The freeze-dried kit of PSMA-617 could be used for the preparation of 177Lu-PSMA-617 with high radiochemical purity (>98%) in a reproducible manner. 177Lu-PSMA-617 prepared using the developed kit was successfully evaluated in patients suffering from metastatic prostate cancer.


Dipeptides/isolation & purification , Dipeptides/therapeutic use , Heterocyclic Compounds, 1-Ring/isolation & purification , Heterocyclic Compounds, 1-Ring/therapeutic use , Lutetium/isolation & purification , Lutetium/therapeutic use , Prostate-Specific Antigen/isolation & purification , Prostate-Specific Antigen/therapeutic use , Prostatic Neoplasms/radiotherapy , Radioisotopes/isolation & purification , Radioisotopes/therapeutic use , Radiopharmaceuticals/isolation & purification , Radiopharmaceuticals/therapeutic use , Animals , Dipeptides/pharmacokinetics , Drug Compounding/methods , Drug Stability , Freeze Drying , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , In Vitro Techniques , Lutetium/pharmacokinetics , Male , Nuclear Pharmacy/methods , Pharmacy Service, Hospital , Positron Emission Tomography Computed Tomography , Prostate-Specific Antigen/pharmacokinetics , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radiochemistry/methods , Radiochemistry/standards , Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution
6.
Biomed Res Int ; 2021: 3849093, 2021.
Article En | MEDLINE | ID: mdl-34722762

The development and optimization of controlled release lipospheres (LS) from safe biocompatible behenic acid (BA) was performed for not only enhancing patient's compliance against highly prevailed chronic diabetes but also to vanquish the insufficiencies of traditional methods of drug delivery. The Box-Bhenken design (BBD) was utilized to statistically investigate the impact of formulation variables on percentage yield (Y 1), entrapment efficiency (Y 2), and SG-release (Y 3) from saxagliptin- (SG-) loaded LS, and the chosen optimized LS were subjected to a comparative in vivo pharmacokinetic analysis against commercially available SG brand. The compatibility analysis performed by DSC and FTIR established a complete lack of interaction of formulation components with SG, while p-XRD suggested a mild transformation of crystalline drug to its amorphous form during encapsulation process. The spherical, free flowing smooth surface LS having zeta potential of -32 mV and size range of 11-20 µm were conveniently formulated. The obtained data for Y 1 (30-80%), Y 2 (30-70%), and Y 3 (40-90%) showed a best fit with quadratic model. The pharmacokinetics analysis of LS showed a significantly decreased C max of SG (75.63 ± 3.85) with a sufficiently elevated T max (10.53 h) as compared to commercial brand of SG (99.66 ± 2.97 ng/mL and 3.55 ± 2.18 h). The achievement of greater bioavailability of SG was most probably attributed to higher level of half-life, mean residence time (MRT), and AUC0-24 for SG released from LS. Conclusively, the novel approach of SG-loaded LS had successfully sustained the plasma SG level for a prolonged time without increasing C max which would ultimately bring an effective management of chronic diabetes.


Adamantane/analogs & derivatives , Dipeptides/administration & dosage , Liposomes/pharmacokinetics , Adamantane/administration & dosage , Adamantane/pharmacokinetics , Adamantane/pharmacology , Administration, Oral , Adult , Biological Availability , Delayed-Action Preparations/pharmacokinetics , Dipeptides/pharmacokinetics , Dipeptides/pharmacology , Drug Compounding/methods , Drug Delivery Systems/methods , Drug Liberation/physiology , Fatty Acids/pharmacokinetics , Fatty Acids/pharmacology , Half-Life , Healthy Volunteers , Humans , Liposomes/pharmacology , Male , Models, Statistical , Solubility
7.
Mol Pharm ; 18(12): 4511-4519, 2021 12 06.
Article En | MEDLINE | ID: mdl-34714082

Lu-177-based, targeted radiotherapeutics/endoradiotherapies are an emerging clinical tool for the management of various cancers. The chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) remains the workhorse for such applications but can limit apparent molar activity or efficient charge modulation, which can impact target binding and, as a consequence, target efficacy. Previously, our lab had developed the small, rare earth selective bifunctional chelator, picaga, as an efficient bifunctional chelator for scandium and lutetium isotopes. Here, we assess the performance of these constructs for therapy in prostate-specific membrane antigen (PSMA)-expressing tumor xenografts. To assess the viability of picaga conjugates in conjunction with long in vivo circulation, a picaga conjugate functionalized with a serum albumin binding moiety, 177Lu-picaga-Alb53-PSMA, was also synthesized. A directly comparative, low, single 3.7 MBq dose treatment study with Lu-PSMA-617 was conducted. Treatment with 177Lu-picaga-Alb53-PSMA resulted in tumor regression and lengthened median survival (54 days) when compared with the vehicle (16 days), 47Sc-picaga-DUPA-, 177Lu-picaga-DUPA-, and 177Lu-PSMA-617-treated cohorts (21, 23, and 21 days, respectively).


Chelating Agents/chemistry , Dipeptides/therapeutic use , Heterocyclic Compounds, 1-Ring/therapeutic use , Lutetium/therapeutic use , Prostate-Specific Antigen/therapeutic use , Prostatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Scandium/therapeutic use , Animals , Dipeptides/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Male , Mice , Prostate-Specific Antigen/pharmacokinetics , Prostatic Neoplasms/mortality , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
8.
Pharmacol Res ; 171: 105798, 2021 09.
Article En | MEDLINE | ID: mdl-34352400

Skeletal muscle atrophy occurs in response to various pathophysiological stimuli, including disuse, aging, and neuromuscular disorders, mainly due to an imbalance of anabolic/catabolic signaling. Branched Chain Amino Acids (BCAAs: leucine, isoleucine, valine) supplements can be beneficial for counteracting muscle atrophy, in virtue of their reported anabolic properties. Here, we carried out a proof-of-concept study to assess the in vivo/ex vivo effects of a 4-week treatment with BCAAs on disuse-induced atrophy, in a murine model of hind limb unloading (HU). BCAAs were formulated in drinking water, alone, or plus two equivalents of L-Alanine (2 ALA) or the dipeptide L-Alanyl-L-Alanine (Di-ALA), to boost BCAAs bioavailability. HU mice were characterized by reduction of body mass, decrease of soleus - SOL - muscle mass and total protein, alteration of postural muscles architecture and fiber size, dysregulation of atrophy-related genes (Atrogin-1, MuRF-1, mTOR, Mstn). In parallel, we provided new robust readouts in the HU murine model, such as impaired in vivo isometric torque and ex vivo SOL muscle contractility and elasticity, as well as altered immune response. An acute pharmacokinetic study confirmed that L-ALA, also as dipeptide, enhanced plasma exposure of BCAAs. Globally, the most sensitive parameters to BCAAs action were muscle atrophy and myofiber cross-sectional area, muscle force and compliance to stress, protein synthesis via mTOR and innate immunity, with the new BCAAs + Di-ALA formulation being the most effective treatment. Our results support the working hypothesis and highlight the importance of developing innovative formulations to optimize BCAAs biodistribution.


Alanine/therapeutic use , Amino Acids, Branched-Chain/therapeutic use , Dipeptides/therapeutic use , Muscular Atrophy/drug therapy , Alanine/pharmacokinetics , Amino Acids, Branched-Chain/pharmacokinetics , Animals , Dipeptides/pharmacokinetics , Disease Models, Animal , Hindlimb Suspension , Male , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Proteome/drug effects , Transcriptome/drug effects
9.
Angew Chem Int Ed Engl ; 60(43): 23327-23334, 2021 10 18.
Article En | MEDLINE | ID: mdl-34416073

Focal adhesion kinase (FAK) is a key mediator of tumour progression and metastasis. To date, clinical trials of FAK inhibitors have reported disappointing efficacy for oncology indications. We report the design and characterisation of GSK215, a potent, selective, FAK-degrading Proteolysis Targeting Chimera (PROTAC) based on a binder for the VHL E3 ligase and the known FAK inhibitor VS-4718. X-ray crystallography revealed the molecular basis of the highly cooperative FAK-GSK215-VHL ternary complex, and GSK215 showed differentiated in-vitro pharmacology compared to VS-4718. In mice, a single dose of GSK215 induced rapid and prolonged FAK degradation, giving a long-lasting effect on FAK levels (≈96 h) and a marked PK/PD disconnect. This tool PROTAC molecule is expected to be useful for the study of FAK-degradation biology in vivo, and our results indicate that FAK degradation may be a differentiated clinical strategy versus FAK inhibition for the treatment of cancer.


Antineoplastic Agents/pharmacology , Focal Adhesion Kinase 1/antagonists & inhibitors , Proteolysis/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzamides/chemistry , Benzamides/pharmacokinetics , Benzamides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Dipeptides/chemistry , Dipeptides/pharmacokinetics , Dipeptides/pharmacology , Focal Adhesion Kinase 1/metabolism , Humans , Mice , Molecular Structure , Ubiquitin-Protein Ligases/metabolism
10.
Molecules ; 26(9)2021 Apr 30.
Article En | MEDLINE | ID: mdl-33946599

It has been herein presented that a microemulsion, known to be an effective and safe drug delivery system following intravenous administration, can be loaded with traces of [68Ga]Ga-PSMA-617 without losing its properties or causing toxicity. Following tolerated IV injections the capability of the microemulsion in altering [68Ga]Ga-PSMA-617 distribution was presented at 120 min post injection based on its ex vivo biodistribution results.


Dipeptides/pharmacokinetics , Edetic Acid/analogs & derivatives , Emulsions , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Oligopeptides/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals , Administration, Intravenous , Animals , Biomarkers , Chemical Phenomena , Dipeptides/administration & dosage , Dipeptides/adverse effects , Edetic Acid/administration & dosage , Edetic Acid/adverse effects , Edetic Acid/pharmacokinetics , Emulsions/chemistry , Gallium Isotopes , Gallium Radioisotopes , Heterocyclic Compounds, 1-Ring/administration & dosage , Heterocyclic Compounds, 1-Ring/adverse effects , Male , Mice , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Positron Emission Tomography Computed Tomography , Prostate-Specific Antigen , Tissue Distribution , Toxicity Tests, Acute , Zinc Isotopes
11.
J Med Chem ; 64(5): 2534-2575, 2021 03 11.
Article En | MEDLINE | ID: mdl-33596065

The biological and medicinal impacts of proteolysis-targeting chimeras (PROTACs) and related chimeric molecules that effect intracellular degradation of target proteins via ubiquitin ligase-mediated ubiquitination continue to grow. However, these chimeric entities are relatively large compounds that often possess molecular characteristics, which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. We therefore explored the conjugation of such molecules to monoclonal antibodies using technologies originally developed for cytotoxic payloads so as to provide alternate delivery options for these novel agents. In this report, we describe the first phase of our systematic development of antibody-drug conjugates (ADCs) derived from bromodomain-containing protein 4 (BRD4)-targeting chimeric degrader entities. We demonstrate the antigen-dependent delivery of the degrader payloads to PC3-S1 prostate cancer cells along with related impacts on MYC transcription and intracellular BRD4 levels. These experiments culminate with the identification of one degrader conjugate, which exhibits antigen-dependent antiproliferation effects in LNCaP prostate cancer cells.


Cell Cycle Proteins/antagonists & inhibitors , Dipeptides/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Immunoconjugates/pharmacology , Proteolysis/drug effects , Transcription Factors/antagonists & inhibitors , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Dipeptides/chemical synthesis , Dipeptides/pharmacokinetics , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Oxidoreductases/immunology , PC-3 Cells , Transcription Factors/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
12.
J Med Chem ; 64(5): 2576-2607, 2021 03 11.
Article En | MEDLINE | ID: mdl-33596073

Heterobifunctional compounds that direct the ubiquitination of intracellular proteins in a targeted manner via co-opted ubiquitin ligases have enormous potential to transform the field of medicinal chemistry. These chimeric molecules, often termed proteolysis-targeting chimeras (PROTACs) in the chemical literature, enable the controlled degradation of specific proteins via their direction to the cellular proteasome. In this report, we describe the second phase of our research focused on exploring antibody-drug conjugates (ADCs), which incorporate BRD4-targeting chimeric degrader entities. We employ a new BRD4-binding fragment in the construction of the chimeric ADC payloads that is significantly more potent than the corresponding entity utilized in our initial studies. The resulting BRD4-degrader antibody conjugates exhibit potent and antigen-dependent BRD4 degradation and antiproliferation activities in cell-based experiments. Multiple ADCs bearing chimeric BRD4-degrader payloads also exhibit strong, antigen-dependent antitumor efficacy in mouse xenograft assessments that employ several different tumor models.


Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Proteolysis/drug effects , Transcription Factors/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Dipeptides/chemical synthesis , Dipeptides/pharmacokinetics , Dipeptides/therapeutic use , Female , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Mice, SCID , Oxidoreductases/immunology , Transcription Factors/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Xenograft Model Antitumor Assays
13.
J Cardiovasc Pharmacol ; 76(2): 189-196, 2020 08.
Article En | MEDLINE | ID: mdl-32769849

OBJECTIVE: The natural molecule α-lipoic acid has been shown to be partially cytoprotective through antioxidant and antiapoptotic mechanisms. To obtain an initial assessment of the safety and potential efficacy of a synthetic derivative, CMX-2043, in preventing ischemic complications of percutaneous coronary intervention (PCI) we conducted the Subjects Undergoing PCI and Perioperative Reperfusion Treatment (SUPPORT-1) trial, the first patient experience with this agent. METHODS AND RESULTS: SUPPORT-1 was a phase 2a, 6-center, international, placebo-controlled, randomized, double-blind trial. A total of 142 patients were randomized to receive a single intravenous bolus dose of drug or placebo administered 15-60 minutes before PCI. Cardiac biomarker assessments included serial measurements of creatine kinase myocardial band (CK-MB) at 6, 12, 18, and 24 hours after PCI and a single measurement of troponin T (TnT) at 24 hours. Peak concentrations of CK-MB and TnT were significantly reduced in the 2.4 mg/kg group compared with placebo (P = 0.05 and 0.03, respectively). No subject administered 2.4 mg/kg of CMX-2043 had an increase of CK-MB to ≥3X upper limit of normal versus 16% for placebo (P = 0.02); 16% of the 2.4-mg/kg dose group developed an elevation of TnT to ≥3X upper limit of normal versus 39% in the placebo group (P = 0.05). No drug-related serious adverse events were observed in any group. CONCLUSION: These data suggest that CMX-2043 may reduce PCI periprocedural myonecrosis and support further clinical evaluation of this novel agent for its potential cytoprotective effects.


Angioplasty, Balloon, Coronary , Cardiovascular Agents/therapeutic use , Coronary Artery Disease/therapy , Dipeptides/therapeutic use , Myocytes, Cardiac/drug effects , Thioctic Acid/analogs & derivatives , Aged , Angioplasty, Balloon, Coronary/adverse effects , Biomarkers/blood , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Coronary Artery Disease/diagnostic imaging , Creatine Kinase, MB Form/blood , Dipeptides/adverse effects , Dipeptides/pharmacokinetics , Double-Blind Method , Female , Humans , India , Male , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Prospective Studies , Thioctic Acid/adverse effects , Thioctic Acid/pharmacokinetics , Thioctic Acid/therapeutic use , Time Factors , Treatment Outcome , Troponin T/blood , United States
14.
J Med Chem ; 63(17): 9977-9989, 2020 09 10.
Article En | MEDLINE | ID: mdl-32787082

The aberrant expression of protein arginine methyltransferase 5 (PRMT5) has been associated with multiple cancers. Using the proteolysis targeting chimera technology, we discovered a first-in-class PRMT5 degrader 15 (MS4322). Here, we report the design, synthesis, and characterization of compound 15 and two structurally similar controls 17 (MS4370) and 21 (MS4369), with impaired binding to the von Hippel-Lindau E3 ligase and PRMT5, respectively. Compound 15, but not 17 and 21, effectively reduced the PRMT5 protein level in MCF-7 cells. Our mechanism studies indicate that compound 15 degraded PRMT5 in an E3 ligase- and proteasome-dependent manner. Compound 15 also effectively reduced the PRMT5 protein level and inhibited growth in multiple cancer cell lines. Moreover, compound 15 was highly selective for PRMT5 in a global proteomic study and exhibited good plasma exposure in mice. Collectively, compound 15 and its two controls 17 and 21 are valuable chemical tools for exploring the PRMT5 functions in health and disease.


Dipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Animals , Cell Line, Tumor , Dipeptides/chemical synthesis , Dipeptides/pharmacokinetics , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , Ligands , Male , Mice , Molecular Structure , Protein-Arginine N-Methyltransferases/metabolism , Proteolysis/drug effects , Structure-Activity Relationship , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
15.
Drug Metab Pharmacokinet ; 35(4): 374-382, 2020 Aug.
Article En | MEDLINE | ID: mdl-32651148

To develop a novel intestinal drug absorption system using intestinal epithelial cells derived from human induced pluripotent stem (iPS) cells, the cells must possess sufficient pharmacokinetic functions. However, the CYP3A4/5 activities of human iPS cell-derived small intestinal epithelial cells prepared using conventional differentiation methods is low. Further, studies of the CYP3A4/5 activities of human iPS-derived and primary small intestinal cells are not available. To fill this gap in our knowledge, here we used forskolin to develop a new differentiation protocol that activates adenosine monophosphate signaling. mRNA expressions of human iPS cell-derived small intestinal epithelial cells, such as small intestine markers, drug-metabolizing enzymes, and drug transporters, were comparable to or greater than those of the adult small intestine. The activities of CYP3A4/5 in the differentiated cells were equal to those of human primary small intestinal cells. The differentiated cells had P-glycoprotein and PEPT1 activities equivalent to those of Caco-2 cells. Differentiated cells were superior to Caco-2 cells for predicting the membrane permeability of drugs that were absorbed through a paracellular pathway and via drug transporters. In summary, here we produced human iPS cell-derived small intestinal epithelial cells with CYP3A4/5 activities equivalent to those of human primary small intestinal cells.


Epithelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Intestine, Small/metabolism , Alkanesulfonic Acids/pharmacokinetics , Caco-2 Cells , Cells, Cultured , Cyclosporins/pharmacokinetics , Digoxin/pharmacokinetics , Dipeptides/pharmacokinetics , Humans , Ibuprofen/pharmacokinetics , Intestine, Small/cytology , Morpholines/pharmacokinetics
16.
AAPS PharmSciTech ; 21(5): 188, 2020 Jul 10.
Article En | MEDLINE | ID: mdl-32651896

The current study aims at formulating and optimizing lipospheres (LS) by the Box-Behnken design (BBD) from safe biodegradable carnauba wax (CW) to co-administer saxagliptin (SG) and enalapril (EP) for co-existing chronic hypertensive diabetes in order to overcome inadequacies of conventional modes of drug administration. Optimized liposphere formulation (OLF) was selected by a numerical optimization procedure and a comparative in vivo pharmacokinetic study of OLF and commercial brands was also performed. Discrete, free-flowing, spherical, smooth-surface LS having a size range of 5-10 µm and zeta potential of - 20 to - 30 mV were successfully formulated. Compatibility studies by FTIR and DSC proved the lack of interaction of components while XRD suggested the transformation of crystalline drugs to amorphous form. Outcomes of dependent optimizing variables like percentage yield (30-90%), EP-release (32-92%), and SG-release (28-95%) followed a polynomial quadratic model. Pharmacokinetics studies indicated a significantly lower Cmax of EP (125.22 ± 6.32) and SG (75.63 ± 3.85) and higher mean Tmax values (9.4 h for EP and 10.73 h for SG) from OLF in comparison with reference brands of EP (257.54 ± 8.23 ng/mL) and SG (393.66 ± 2.97 ng/mL). Additionally, a potential rise in half-life and MRT of SG and EP was achieved reaching approximately 2- to 3-fold higher than noted for reference brands. Importantly, the enhanced Tmax and AUC0-24 specified the achievement of enhanced bioavailability of both drugs from LS. Consequently, such an innovative approach could not only control drug release in both in vitro and in vivo analyses but also maintain plasma drug concentration for a longer time without maximizing Cmax leading towards effective management of chronic illnesses.


Adamantane/analogs & derivatives , Dipeptides/pharmacokinetics , Enalapril/pharmacokinetics , Adamantane/administration & dosage , Adamantane/pharmacokinetics , Administration, Oral , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacokinetics , Biological Availability , Delayed-Action Preparations/chemistry , Dipeptides/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Drug Liberation , Enalapril/administration & dosage , Half-Life , Humans , Particle Size , Waxes/chemistry
17.
Int J Pharm ; 586: 119575, 2020 Aug 30.
Article En | MEDLINE | ID: mdl-32622809

Stable-isotope-labeled probes suitable for magnetic resonance imaging (MRI) would have various potential medical applications, such as tumor imaging. Here, with the aim of developing MRI probes targeting peptide transporters, we synthesized a series of [19F]dipeptides by introducing one or two fluorine atoms or a trifluoromethyl group into the benzene ring of l-phenylalanyl-ψ[CS-N]-l-alanine (Phe-ψ-Ala), which is resistant to cleavage by peptidases. The mono- and difluoro dipeptides were efficiently transported by PEPT1 and PEPT2. Moreover, (3,5)-difluoro Phe-ψ-Ala was metabolically stable in human hepatocyte culture, and had a low distribution volume in mice. An acute toxicity study in mice revealed no apparent effect on body weight or behavior. The biodistribution and biodynamics of this compound could be clearly visualized by 19F-MRI in vivo, although specific signal enhancement was observed only in the bladder, but not in the tumor of tumor-xenografted mice. Although there was no specific signal enhancement of the tested compound at the tumor, the present study provides some challenging points regarding 19F-MRI probes for future investigation.


Dipeptides/pharmacokinetics , Neoplasms/diagnostic imaging , Peptide Transporter 1/metabolism , Symporters/metabolism , Animals , Biological Transport , Dipeptides/chemistry , Dipeptides/toxicity , Fluorine-19 Magnetic Resonance Imaging , HEK293 Cells , HeLa Cells , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Tissue Distribution , Toxicity Tests, Acute
18.
Clin Drug Investig ; 40(6): 555-566, 2020 Jun.
Article En | MEDLINE | ID: mdl-32277364

BACKGROUND AND OBJECTIVE: Bentysrepinine (Y101), a derivative of repensine (a compound isolated from Dichondra repens Forst), is a novel phenylalanine dipeptide currently under development for the treatment of hepatitis B virus (HBV). The objectives of these studies were to assess the safety, tolerability and pharmacokinetics of bentysrepinine in healthy Chinese subjects. METHODS: Two randomised, double-blind, placebo-controlled trials evaluated a single oral dose (50-900 mg, study 01) and multiple doses (300 mg and 600 mg, study 02), and a randomised, open, crossover food-effect study (600 mg, study 03) of bentysrepinine was established. Safety and tolerability were assessed by adverse event (AE) reporting, clinical laboratory tests, physical examinations, vital sign monitoring and electrocardiogram (ECG). Plasma, urine and faecal samples were analysed using validated liquid chromatography tandem mass spectrometry (LC-MS/MS) methods to investigate the pharmacokinetics of bentysrepinine. RESULTS: Ninety-four subjects were enrolled, and bentysrepinine was well tolerated. Mild and reversible AEs occurred for single and multiple oral doses between 50 and 900 mg. The most common adverse effects were increased alanine aminotransferase (ALT) and aspartate transaminase (AST). Other clinically significant AEs included nausea and elevated urine leukocytes, urine red blood cells, transaminase, creatine kinase, total cholesterol, triglycerides, and low-density cholesterol. There were no clinically significant changes in the ECG, vital signs or laboratory assessments during the studies. The maximum tolerated dose (MTD) was not reached in the dose escalation study. Bentysrepinine was rapidly absorbed and metabolised with a mean time to reach maximum concentration (Tmax) between 1-2 h and a mean terminal elimination half-life (t1/2) of approximately 1-3 h. In the single ascending dose study, the exposure including the area under the concentration-time curve (AUC) and the maximum plasma concentration (Cmax) of bentysrepinine generally increased in a dose-dependent but not dose-proportional manner in the 50-900 mg dose range. The urinary excretion and faecal excretion of unchanged bentysrepinine were 2.98% and 4.58% of the total dose, respectively. In the multiple-dose study, no accumulation was found after repeated administration at the 300 mg and 600 mg dose levels. The food-effect study using a 600 mg single dose showed that food intake has an obvious effect on the absorption of bentysrepinine from tablets. No experimental differences were found based on sex. CONCLUSION: Bentysrepinine exhibited acceptable safety and tolerability in healthy subjects in the dose range of 50-900 mg in both single- and multiple-dose studies. The drug did not exhibit linear pharmacokinetic characteristics. No accumulation was observed after the administration of multiple 300 and 600 mg doses. Bentysrepinine is extensively metabolised in the body. Food may increase its bioavailability. TRIALS REGISTRATION: CFDA registration numbers CTR20160096, CTR20160094, and CTR20140543 (www.chinadrugtrials.org.cn).


Antiviral Agents/adverse effects , Benzamides/administration & dosage , Dipeptides/administration & dosage , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Area Under Curve , Asian People , Benzamides/adverse effects , Benzamides/pharmacokinetics , Chromatography, Liquid , Cross-Over Studies , Dipeptides/adverse effects , Dipeptides/pharmacokinetics , Double-Blind Method , Female , Healthy Volunteers , Hepatitis B virus , Humans , Male , Placebos , Tandem Mass Spectrometry
19.
J Nucl Med ; 61(11): 1602-1606, 2020 11.
Article En | MEDLINE | ID: mdl-32169910

Neuroendocrine differentiation is associated with treatment failure and poor outcome in metastatic castration-resistant prostate cancer. We investigated the effect of circulating neuroendocrine biomarkers on the efficacy of prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT). Methods: Neuroendocrine biomarker profiles (progastrin-releasing peptide, neuron-specific enolase, and chromogranin-A) were analyzed in 50 patients commencing 177Lu-PSMA-617 RLT. The primary endpoint was a prostate-specific antigen response in relation to baseline neuroendocrine marker profiles. An additional endpoint was progression-free survival. Tumor uptake on posttherapeutic scans, a known predictive marker for response, was used as a control variable. Results: Neuroendocrine biomarker profiles were abnormal in most patients. Neuroendocrine biomarker levels did not predict treatment failure or early progression (P ≥ 0.13). By contrast, intense PSMA-ligand uptake in metastases predicted both treatment response (P = 0.0030) and reduced risk of early progression (P = 0.0111). Conclusion: Neuroendocrine marker profiles do not predict an adverse outcome from RLT. By contrast, high ligand uptake was confirmed to be crucial for achieving a tumor response.


Antigens, Surface/metabolism , Chromogranin A/blood , Dipeptides/therapeutic use , Glutamate Carboxypeptidase II/metabolism , Heterocyclic Compounds, 1-Ring/therapeutic use , Peptide Fragments/blood , Phosphopyruvate Hydratase/blood , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Biomarkers , Cell Differentiation , Dipeptides/adverse effects , Dipeptides/pharmacokinetics , Heterocyclic Compounds, 1-Ring/adverse effects , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Lutetium , Male , Neuroendocrine Cells/chemistry , Neuroendocrine Cells/cytology , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Recombinant Proteins/blood
20.
Xenobiotica ; 50(4): 468-478, 2020 Apr.
Article En | MEDLINE | ID: mdl-31329010

The objective of this study was to clarify the species differences of pharmacokinetics of Y101 (N-[N-benzoyl-O-(2-dimethylaminoethyl)-l-tyrosyl]-l-phenylalaninol hydrochloride), a derivative of herbal ingredient with anti-HBV hepatitis activity, in rats, dogs, monkeys and humans.The metabolic stability and metabolite identification studies using liver microsomes in vitro, plasma protein binding using a rapid equilibrium dialysis in vitro, pharmacokinetic studies in vivo were carried out to evaluate the interspecies differences. The toxicokinetic study in monkeys was also investigated.The metabolic profiles were similar in monkeys and humans, which were significant different from rats and dogs in vitro. In vitro plasma protein binding showed no major differences between species with medium to high protein binding rates. After single oral dose to rats, dogs, and monkeys, the absolute oral bioavailability of Y101 was 44.9%, 43.1%, and 19.2%, respectively. There was no accumulation for Y101 toxicokinetics in monkeys after oral administration for 90 d.The metabolic profiles indicated monkey was the very animal model for preclinical safety evaluation of Y101. Our results have demonstrated the favorable pharmacokinetics profile of Y101, which supports the clinical trials in humans.


Antiviral Agents/metabolism , Benzamides/metabolism , Dipeptides/metabolism , Animals , Antiviral Agents/pharmacokinetics , Benzamides/pharmacokinetics , Dipeptides/pharmacokinetics , Dogs , Hepatitis B , Humans , Microsomes, Liver/metabolism , Rats , Species Specificity
...