Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Bioconjug Chem ; 33(7): 1340-1349, 2022 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-35767675

RESUMO

It is particularly challenging to develop a truly effective pharmacotherapy for cocaine use disorder (CUD) treatment. Accelerating cocaine metabolism via hydrolysis at cocaine benzoyl ester using an efficient cocaine hydrolase (CocH) is known as a promising pharmacotherapeutic approach to CUD treatment. Preclinical and clinical studies on our first CocH (CocH1), in its human serum albumin-fused form known as TV-1380, have demonstrated the promise of a general concept of CocH-based pharmacotherapy for CUD treatment. However, the biological half-life of TV-1380 (t1/2 = 8 h in rats, associated with t1/2 = 43-77 h in humans) is not long enough for practical treatment of cocaine dependence, which requires enzyme injection for no more than once weekly. Through protein fusion of a human butyrylcholinesterase mutant (denoted as CocH5) with a mutant (denoted as Fc(M6)) of Fc from human IgG1, we have designed, prepared, and tested a new fusion protein (denoted as CocH5-Fc(M6)) for its pharmacokinetic profile and in vivo catalytic activity against (-)-cocaine. CocH5-Fc(M6) represents the currently most efficient long-acting cocaine hydrolase with both the highest catalytic activity against (-)-cocaine and the longest elimination half-life (t1/2 = 229 ± 5 h) in rats. As a result, even at a single modest dose of 3 mg/kg, CocH5-Fc(M6) can significantly and effectively accelerate the metabolism of cocaine in rats for at least 60 days. In addition, ∼70 nM CocH5-Fc(M6) in plasma was able to completely block the toxicity and physiological effects induced by intraperitoneal injection of a lethal dose of cocaine (60 mg/kg).


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Animais , Butirilcolinesterase/genética , Butirilcolinesterase/farmacocinética , Hidrolases de Éster Carboxílico/genética , Cocaína/metabolismo , Cocaína/uso terapêutico , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Humanos , Ratos , Proteínas Recombinantes
2.
Addict Biol ; 27(4): e13179, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35754103

RESUMO

Dysregulation of dopamine transporters (DAT) within the dopaminergic system is an important biomarker of cocaine exposure. Depending on cocaine amount in-taken, one-time exposure in rats could lead to most (>95% of total) of DAT translocating to plasma membrane of the dopaminergic neurons compared to normal DAT distribution (~5.7% on the plasma membrane). Without further cocaine exposure, the time course of striatal DAT distribution, in terms of intracellular and plasma membrane fractions of DAT, represents a recovery process of the dopaminergic system. In this study, we demonstrated that after an acute cocaine exposure of 20 mg/kg (i.p.), the initial recovery process from days 1 to 15 in rats was relatively faster (from >95% on day 1 to ~35.4% on day 15). However, complete recovery of the striatal DAT distribution may take about 60 days. In another situation, with repeated cocaine exposures for once every other day for a total of 17 doses of 20 mg/kg cocaine (i.p.) from days 0 to 32, the complete recovery of striatal DAT distribution may take an even longer time (about 90 days), which represents a consequence of chronic cocaine use. Further, we demonstrated that a highly efficient Fc-fused cocaine hydrolase, CocH5-Fc(M6), effectively blocked cocaine-induced hyperactivity and DAT trafficking with repeated cocaine exposures by maintaining a plasma CocH5-Fc(M6) concentration ≥58.7 ± 2.9 nM in rats. The cocaine hydrolase protected dopaminergic system and helped the cocaine-altered DAT distribution to recover by preventing the dopaminergic system from further damage by cocaine.


Assuntos
Cocaína , Animais , Hidrolases de Éster Carboxílico , Cocaína/farmacologia , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Ratos , Proteínas Recombinantes
3.
Addict Biol ; 27(1): e13089, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34363291

RESUMO

Cocaine blocks dopamine uptake via dopamine transporter (DAT) on plasma membrane of neuron cells and, as a result, produces the high and induces DAT trafficking to plasma membrane which contributes to the drug seeking or craving. In this study, we first examined the dose dependence of cocaine-induced DAT trafficking and hyperactivity in rats, demonstrating that cocaine at an intraperitoneal dose of 10 mg/kg or higher led to redistribution of most DAT to the plasma membrane while inducing significant hyperactivity in rats. However, administration of 5-mg/kg cocaine (ip) did not significantly induce DAT trafficking or hyperactivity in rats. So the threshold (intraperitoneal) dose of cocaine that can significantly induce DAT trafficking or hyperactivity should be between 5 and 10 mg/kg. These data suggest that when a cocaine dose is high enough to induce significant hyperactivity, it can also significantly induce DAT trafficking to the plasma membrane. Further, the threshold brain cocaine concentration required to induce significant hyperactivity and DAT trafficking was estimated to be ~2.0 ± 0.8 µg/g. Particularly, for treatment of cocaine abuse, previous studies demonstrated that an exogenous cocaine-metabolizing enzyme, for example, CocH3-Fc(M3), can effectively block cocaine-induced hyperactivity. However, it was unknown whether an enzyme could also effectively block cocaine-induced DAT trafficking to the plasma membrane. This study demonstrates, for the first time, that the enzyme is also capable of effectively blocking cocaine from reaching the brain even with a lethal dose of 60-mg/kg cocaine (ip) and, thus, powerfully preventing cocaine-induced physiological effects such as the hyperactivity and DAT trafficking.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Membrana Celular/efeitos dos fármacos , Cocaína/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Hipercinese/patologia , Proteínas Recombinantes/metabolismo , Animais , Transtornos Relacionados ao Uso de Cocaína , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Sprague-Dawley
4.
Addict Biol ; 27(6): e13236, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36301205

RESUMO

As well known, cocaine induces stimulant effects and dopamine transporter (DAT) trafficking to the plasma membrane of dopaminergic neurons. In the present study, we examined cocaine-induced hyperactivity along with cocaine-induced DAT trafficking and the recovery rate of the dopaminergic system in female rats in comparison with male rats, demonstrating interesting gender differences. Female rats are initially more sensitive to cocaine than male rats in terms of both the DAT trafficking and hyperactivity induced by cocaine. Particularly, intraperitoneal (i.p.) administration of 5 mg/kg cocaine induced significant hyperactivity and DAT trafficking in female rats but not in male rats. After repeated cocaine exposures (i.e., i.p. administration of 20 mg/kg cocaine every other day from Day 0 to Day 32), cocaine-induced hyperactivity in female rats gradually became a clear pattern of two phases, with the first phase of the hyperactivity lasting for only a few minutes and the second phase lasting for over an hour beginning at ~30 min, which is clearly different from that of male rats. It has also been demonstrated that the striatal DAT distribution of female rats may recover faster than that of male rats after multiple cocaine exposures. Nevertheless, despite the remarkable gender differences, our recently developed long-acting cocaine hydrolase, known as CocH5-Fc(M6), can similarly and effectively block cocaine-induced DAT trafficking and hyperactivity in both male and female rats.


Assuntos
Cocaína , Proteínas da Membrana Plasmática de Transporte de Dopamina , Masculino , Feminino , Ratos , Animais , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Cocaína/farmacologia , Cocaína/metabolismo , Fatores Sexuais , Inibidores da Captação de Dopamina/farmacologia , Inibidores da Captação de Dopamina/metabolismo , Membrana Celular/metabolismo
5.
Org Biomol Chem ; 18(10): 1968-1977, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32101217

RESUMO

A majority of cocaine users also consume alcohol. The concurrent use of cocaine and alcohol produces the pharmacologically active metabolites cocaethylene and norcocaethylene, in addition to norcocaine. Both cocaethylene and norcocaethylene are more toxic than cocaine itself. Hence, a truly valuable cocaine-metabolizing enzyme for cocaine abuse/overdose treatment should be effective for the hydrolysis of not only cocaine, but also its metabolites norcocaine, cocaethylene, and norcocaethylene. However, there has been no report on enzymes capable of hydrolyzing norcocaethylene (the most toxic metabolite of cocaine). The catalytic efficiency parameters (kcat and KM) of human butyrylcholinesterase (BChE) and two mutants (known as cocaine hydrolases E14-3 and E12-7) against norcocaethylene have been characterized in the present study for the first time, and they are compared with those against cocaine. According to the obtained kinetic data, wild-type human BChE showed a similar catalytic efficiency against norcocaethylene (kcat = 9.5 min-1, KM = 11.7 µM, and kcat/KM = 8.12 × 105 M-1 min-1) to that against (-)-cocaine (kcat = 4.1 min-1, KM = 4.5 µM, and kcat/KM = 9.1 × 105 M-1 min-1). E14-3 and E12-7 showed an improved catalytic activity against norcocaethylene compared to wild-type BChE. E12-7 showed a 39-fold improved catalytic efficiency against norcocaethylene (kcat = 210 min-1, KM = 6.6 µM, and kcat/KM = 3.18 × 107 M-1 min-1). It has been demonstrated that E12-7 as an exogenous enzyme can efficiently metabolize norcocaethylene in rats.


Assuntos
Butirilcolinesterase/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Cocaína/análogos & derivados , Proteínas Recombinantes/metabolismo , Animais , Biocatálise , Butirilcolinesterase/química , Butirilcolinesterase/genética , Células CHO , Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/genética , Cocaína/química , Cocaína/metabolismo , Cocaína/farmacocinética , Cricetulus , Ensaios Enzimáticos , Humanos , Hidrólise , Cinética , Masculino , Simulação de Acoplamento Molecular , Mutação , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
6.
Bioorg Med Chem Lett ; 28(5): 858-862, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29456107

RESUMO

Human mPGES-1 has emerged as a promising target in exploring a next generation of anti-inflammatory drugs, as selective mPGES-1 inhibitors are expected to discriminatively suppress the production of induced PGE2 without blocking the normal biosynthesis of other prostanoids including homeostatic PGE2. Therefore, this therapeutic approach is believed to reduce the adverse effects associated with the application of traditional non-steroidal anti-inflammatory drugs (tNSAIDs) and selective COX-2 inhibitors (coxibs). Identified from structure-based virtue screening, the compound with (Z)-5-benzylidene-2-iminothiazolidin-4-one scaffold was used as lead in rational design of novel inhibitors. Besides, we further designed, synthesized, and evaluated 5-((1,3-diphenyl-1H-pyrazol-4-yl)methylene)pyrimidine-2,4,6(1H,3H,5H)-triones and structurally related derivatives for their in vitro inhibitory activities. According to in vitro activity assays, a number of these compounds were capable of inhibiting human mPGES-1, with the desirable selectivity for mPGES-1 over COX isozymes.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Prostaglandina-E Sintases/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinas/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Estrutura Molecular , Prostaglandina-E Sintases/metabolismo , Pirazóis/síntese química , Pirazóis/química , Pirimidinas/síntese química , Pirimidinas/química , Relação Estrutura-Atividade
7.
Biochem J ; 466(2): 243-51, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25486543

RESUMO

Mouse butyrylcholinesterase (mBChE) and an mBChE-based cocaine hydrolase (mCocH, i.e. the A¹99S/S²²7A/S²87G/A³²8W/Y³³²G mutant) have been characterized for their catalytic activities against cocaine, i.e. naturally occurring (-)-cocaine, in comparison with the corresponding human BChE (hBChE) and an hBChE-based cocaine hydrolase (hCocH, i.e. the A¹99S/F²²7A/S²87G/A³²8W/Y³³²G mutant). It has been demonstrated that mCocH and hCocH have improved the catalytic efficiency of mBChE and hBChE against (-)-cocaine by ~8- and ~2000-fold respectively, although the catalytic efficiencies of mCocH and hCocH against other substrates, including acetylcholine (ACh) and butyrylthiocholine (BTC), are close to those of the corresponding wild-type enzymes mBChE and hBChE. According to the kinetic data, the catalytic efficiency (k(cat)/K(M)) of mBChE against (-)-cocaine is comparable with that of hBChE, but the catalytic efficiency of mCocH against (-)-cocaine is remarkably lower than that of hCocH by ~250-fold. The remarkable difference in the catalytic activity between mCocH and hCocH is consistent with the difference between the enzyme-(-)-cocaine binding modes obtained from molecular modelling. Further, both mBChE and hBChE demonstrated substrate activation for all of the examined substrates [(-)-cocaine, ACh and BTC] at high concentrations, whereas both mCocH and hCocH showed substrate inhibition for all three substrates at high concentrations. The amino-acid mutations have remarkably converted substrate activation of the enzymes into substrate inhibition, implying that the rate-determining step of the reaction in mCocH and hCocH might be different from that in mBChE and hBChE.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Cocaína/metabolismo , Modelos Moleculares , Acetilcolina/química , Acetilcolina/metabolismo , Substituição de Aminoácidos , Animais , Sítios de Ligação , Biocatálise , Butirilcolinesterase/química , Butirilcolinesterase/genética , Butirilcolinesterase/metabolismo , Butiriltiocolina/química , Butiriltiocolina/metabolismo , Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/genética , Cocaína/química , Ativação Enzimática , Humanos , Cinética , Camundongos , Simulação de Dinâmica Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformação Proteica , Engenharia de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
8.
Biochem J ; 460(3): 447-57, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24870023

RESUMO

It is known that the majority of cocaine users also consume alcohol. Alcohol can react with cocaine to produce a significantly more cytotoxic compound, cocaethylene. Hence a truly valuable cocaine-metabolizing enzyme as treatment for cocaine abuse/overdose should be efficient for not only cocaine itself, but also cocaethylene. The catalytic parameters (kcat and KM) of human BChE (butyrylcholinesterase) and two mutants (known as cocaine hydrolases E14-3 and E12-7) for cocaethylene are characterized in the present study, for the first time, in comparison with those for cocaine. On the basis of the obtained kinetic data, wild-type human BChE has a lower catalytic activity for cocaethylene (kcat=3.3 min(-1), KM=7.5 µM and kcat/KM=4.40 × 10(5) M(-1)·min(-1)) compared with its catalytic activity for (-)-cocaine. E14-3 and E12-7 have a considerably improved catalytic activity against cocaethylene compared with the wild-type BChE. E12-7 is identified as the most efficient enzyme for hydrolysing cocaethylene in addition to its high activity for (-)-cocaine. E12-7 has an 861-fold improved catalytic efficiency for cocaethylene (kcat=3600 min(-1), KM=9.5 µM and kcat/KM=3.79 × 10(8) M(-1)·min(-1)). It has been demonstrated that E12-7 as an exogenous enzyme can indeed rapidly metabolize cocaethylene in rats. Further kinetic modelling has suggested that E12-7 with an identical concentration as that of the endogenous BChE in human plasma can effectively eliminate (-)-cocaine, cocaethylene and norcocaine in simplified kinetic models of cocaine abuse and overdose associated with the concurrent use of cocaine and alcohol.


Assuntos
Butirilcolinesterase/genética , Butirilcolinesterase/metabolismo , Cocaína/análogos & derivados , Animais , Células CHO , Catálise , Cocaína/metabolismo , Cocaína/farmacocinética , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Cricetulus , Etanol/farmacologia , Células HEK293 , Humanos , Hidrólise , Cinética , Masculino , Simulação de Acoplamento Molecular , Ratos
9.
Clin Pharmacol Ther ; 114(3): 664-672, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37422675

RESUMO

Recently, multiple chimeric antigen receptor T-cell (CAR-T)-based therapies have been approved for treating hematological malignancies, targeting CD19 and B-cell maturation antigen. Unlike protein or antibody therapies, CAR-T therapies are "living cell" therapies whose pharmacokinetics are characterized by expansion, distribution, contraction, and persistence. Therefore, this unique modality requires a different approach for quantitation compared with conventional ligand binding assays implemented for most biologics. Cellular (flow cytometry) or molecular assays (polymerase chain reaction (PCR)) can be deployed with each having unique advantages and disadvantages. In this article, we describe the molecular assays utilized: quantitative PCR (qPCR), which was the initial platform used to estimate transgene copy numbers and more recently droplet digital PCR (ddPCR) which quantitates the absolute copy numbers of CAR transgene. The comparability of the two methods in patient samples and of each method across different matrices (isolated CD3+ T-cells or whole blood) was also performed. The results show a good correlation between qPCR and ddPCR for the amplification of same gene in clinical samples from a CAR-T therapy trial. In addition, our studies show that the qPCR-based amplification of transgene levels was well-correlated, independent of DNA sources (either CD3+ T-cells or whole blood). Our results also highlight that ddPCR can be a better platform for monitoring samples at the early phase of CAR-T dosing prior to expansion and during long-term monitoring as they can detect samples with very low copy numbers with high sensitivity, in addition to easier implementation and sample logistics.


Assuntos
Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Cinética , Reação em Cadeia da Polimerase/métodos , Linfócitos T/metabolismo , Imunoterapia Adotiva/métodos
10.
Toxicol Rep ; 9: 1586-1594, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518391

RESUMO

As most cocaine users drink alcohol, it is interesting to understand how a non-lethal dose of alcohol affects the metabolism and toxicity of cocaine. In this study, we examined the correlation between dose-dependent toxicity and the metabolism/pharmacokinetic (PK) profile of cocaine with or without alcohol (ethanol, 1 g/kg) co-administration in rats. The cocaine toxicity in rats with or without alcohol co-administration is characterized by not only the commonly used LD50, but also the average times for the appearance of convulsion and death as well as total toxicity level (TTL) in the blood. All these data have consistently demonstrated that co-administration of alcohol increased cocaine toxicity, and that the alcohol-enhanced toxicity of cocaine is mainly attributed to the observed two additional metabolites (cocaethylene and norcocaethylene - products of chemical reactions of cocaine with alcohol catalyzed by metabolic enzymes carboxylesterase-1 and liver microsomal cytochrome P450 3A4) that are more toxic than cocaine itself. So, evaluation of the substance TTL should account for the blood levels of not only cocaine itself, but also its all toxic metabolites. In addition, for rats died of a lethal dose of cocaine (60 or 100 mg/kg) combined with 1 g/kg alcohol, we also determined the TTL at the time of death, demonstrating that death would occur once the TTL reached a threshold (~16 µM).

11.
CPT Pharmacometrics Syst Pharmacol ; 10(4): 362-376, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33565700

RESUMO

Despite tremendous success of chimeric antigen receptor (CAR) T cell therapy in clinical oncology, the dose-exposure-response relationship of CAR-T cells in patients is poorly understood. Moreover, the key drug-specific and system-specific determinants leading to favorable clinical outcomes are also unknown. Here we have developed a multiscale mechanistic pharmacokinetic (PK)-pharmacodynamic (PD) model for anti-B-cell maturation antigen (BCMA) CAR-T cell therapy (bb2121) to characterize (i) in vitro target cell killing in multiple BCMA expressing tumor cell lines at varying effector to target cell ratios, (ii) preclinical in vivo tumor growth inhibition and blood CAR-T cell expansion in xenograft mice, and (iii) clinical PK and PD biomarkers in patients with multiple myeloma. Our translational PK-PD relationship was able to effectively describe the commonly observed multiphasic CAR-T cell PK profile in the clinic, consisting of the rapid distribution, expansion, contraction, and persistent phases, and accounted for the categorical individual responses in multiple myeloma to effectively calculate progression-free survival rates. Preclinical and clinical data analysis revealed comparable parameter estimates pertaining to CAR-T cell functionality and suggested that patient baseline tumor burden could be more sensitive than dose levels toward overall extent of exposure after CAR-T cell infusion. Virtual patient simulations also suggested a very steep dose-exposure-response relationship with CAR-T cell therapy and indicated the presence of a "threshold" dose, beyond which a flat dose-response curve could be observed. Our simulations were concordant with multiple clinical observations discussed in this article. Moving forward, this framework could be leveraged a priori to explore multiple infusions and support the preclinical/clinical development of future CAR-T cell therapies.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Antígeno de Maturação de Linfócitos B/antagonistas & inibidores , Mieloma Múltiplo/terapia , Receptores de Antígenos Quiméricos/uso terapêutico , Animais , Antineoplásicos Imunológicos/administração & dosagem , Biomarcadores Farmacológicos/análise , Linhagem Celular Tumoral/efeitos dos fármacos , Simulação por Computador , Relação Dose-Resposta a Droga , Humanos , Imunoterapia Adotiva/métodos , Infusões Intravenosas , Camundongos , Camundongos Endogâmicos NOD , Modelos Teóricos , Farmacocinética , Intervalo Livre de Progressão , Receptores de Antígenos Quiméricos/administração & dosagem , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
12.
Clin Pharmacol Ther ; 109(3): 716-727, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33002189

RESUMO

Chimeric antigen receptor (CAR)-T cell therapy has achieved considerable success in treating B-cell hematologic malignancies. However, the challenges of extending CAR-T therapy to other tumor types, particularly solid tumors, remain appreciable. There are substantial variabilities in CAR-T cellular kinetics across CAR-designs, CAR-T products, dosing regimens, patient responses, disease types, tumor burdens, and lymphodepletion conditions. As a "living drug," CAR-T cellular kinetics typically exhibit four distinct phases: distribution, expansion, contraction, and persistence. The cellular kinetics of CAR-T may correlate with patient responses, but which factors determine CAR-T cellular kinetics remain poorly defined. Herein, we developed a cellular kinetic model to retrospectively characterize CAR-T kinetics in 217 patients from 7 trials and compared CAR-T kinetics across response status, patient populations, and tumor types. Based on our analysis results, CAR-T cells exhibited a significantly higher cell proliferation rate and capacity but a lower contraction rate in patients who responded to treatment. CAR-T cells proliferate to a higher degree in hematologic malignancies than in solid tumors. Within the assessed dose ranges (107 -109 cells), CAR-T doses were weakly correlated with CAR-T cellular kinetics and patient response status. In conclusion, the developed CAR-T cellular kinetic model adequately characterized the multiphasic CAR-T cellular kinetics and supported systematic evaluations of the potential influencing factors, which can have significant implications for the development of more effective CAR-T therapies.


Assuntos
Proliferação de Células , Imunoterapia Adotiva , Ativação Linfocitária , Modelos Imunológicos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Morte Celular , Ensaios Clínicos como Assunto , Simulação por Computador , Humanos , Memória Imunológica , Cinética , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Estudos Retrospectivos , Linfócitos T/metabolismo
13.
PhytoKeys ; 138: 163-177, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31988611

RESUMO

A new genus of Arundinarieae, Khoonmengia, is established to accommodate a unique new bamboo species, K. honbaensis, from central-southern Vietnam. The morphological features, habitats and distribution of Khoonmengia and related genera, i.e. Ampelocalamus and Hsuehochloa, are compared. The characters of its scrambling habit, internodes with brownish green dots, conspicuous nodes swollen at one side, elliptic buds wholly sunken into culm, extravaginal branching pattern, mid-culm branch complement with one central dominant branch elongating to reiterate the culm accompanied by several lateral slender branches, swollen culm sheath base with a distinctive zone of transverse wrinkles, synflorescence composed of only one spikelet, single or several to many synflorescences arranged into a raceme or panicle terminal on leafy branches, purple anthers and nut-like caryopsis with hardened pericarp and loosely adherent lemma and palea distinguish K. honbaensis from morphologically similar taxa. In order to investigate the phylogenetic position of this unknown bamboo, molecular phylogenetic analyses based on the nuclear gene GBSSI were also conducted, and the results proved that K. honbaensis is definitely a member of Arundinarieae with an isolated position, which also indicated that this species could not be assigned to any of the already described genera and supported the establishment of the new genus.

14.
MAbs ; 12(1): 1688616, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31852337

RESUMO

The development of mechanism-based, multiscale pharmacokinetic-pharmacodynamic (PK-PD) models for chimeric antigen receptor (CAR)-T cells is needed to enable investigation of in vitro and in vivo correlation of CAR-T cell responses and to facilitate preclinical-to-clinical translation. Toward this goal, we first developed a cell-level in vitro PD model that quantitatively characterized CAR-T cell-induced target cell depletion, CAR-T cell expansion and cytokine release. The model accounted for key drug-specific (CAR-affinity, CAR-densities) and system-specific (antigen densities, E:T ratios) variables and was able to characterize comprehensive in vitro datasets from multiple affinity variants of anti-EGFR and anti-HER2 CAR-T cells. Next, a physiologically based PK (PBPK) model was developed to simultaneously characterize the biodistribution of untransduced T-cells, anti-EGFR CAR-T and anti-CD19 CAR-T cells in xenograft -mouse models. The proposed model accounted for the engagement of CAR-T cells with tumor cells at the site of action. Finally, an integrated PBPK-PD relationship was established to simultaneously characterize expansion of CAR-T cells and tumor growth inhibition (TGI) in xenograft mouse model, using datasets from anti-BCMA, anti-HER2, anti-CD19 and anti-EGFR CAR-T cells. Model simulations provided potential mechanistic insights toward the commonly observed multiphasic PK profile (i.e., rapid distribution, expansion, contraction and persistence) of CAR-T cells in the clinic. Model simulations suggested that CAR-T cells may have a steep dose-exposure relationship, and the apparent Cmax upon CAR-T cell expansion in blood may be more sensitive to patient tumor-burden than CAR-T dose levels. Global sensitivity analysis described the effect of other drug-specific parameters toward CAR-T cell expansion and TGI. The proposed modeling framework will be further examined with the clinical PK and PD data, and the learnings can be used to inform design and development of future CAR-T therapies.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Animais , Movimento Celular , Proliferação de Células , Simulação por Computador , Receptores ErbB/imunologia , Xenoenxertos , Humanos , Camundongos , Modelos Teóricos , Neoplasias/terapia , Ligação Proteica , Receptor ErbB-2/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia
15.
Drug Alcohol Depend ; 204: 107462, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31499241

RESUMO

BACKGROUND: Majority of cocaine users also consume alcohol, and concurrent use of cocaine and alcohol produces cocaethylene, norcocaine, norcocaethylene, and other non-toxic metabolites. It is essential to know their relative toxicity for development of a truly effective therapeutics for cocaine toxicity treatment. METHODS: Drug (norcocaethylene or norcocaine)-induced acute toxicity was characterized by the occurrence (and the timing) of prostration, seizure, and death after intraperitoneal administration of the drug (n = 15) using the same strain (Swiss Webster) of male mice reported in previous study by Hearn et al. to determine LD50 of cocaine and cocaethylene. In addition, drug (cocaine, cocaethylene, norcocaine, or norcocaethylene)-induced hyperactivity was determined by locomotor activity testing (n = 8). RESULTS: According to the animal data, norcocaethylene (LD50=∼39.4 mg/kg) and norcocaine (LD50=∼49.7 mg/kg) are the most toxic metabolites, but they do not induce significant hyperactivity. In addition, the relative toxicity of drugs correlates with the time to the occurrence of prostration/seizure/death after the drug administration. CONCLUSIONS: The relative toxicity of these toxic drugs can be ranked in this order: norcocaethylene > norcocaine > cocaethylene > cocaine. The data suggest that norcocaethylene, norcocaine, and cocaethylene are all significant contributors to acute toxicity of cocaine in concurrent use of cocaine and alcohol. Hence, future therapeutic development for cocaine toxicity treatment must account for detoxification of these more toxic metabolites. In addition, the relative toxicity of different drugs correlates with the average time to the occurrence of death, seizure, or prostration after the drug administration with a same dose close to their LD50 values.


Assuntos
Cocaína/análogos & derivados , Locomoção/efeitos dos fármacos , Animais , Cocaína/metabolismo , Cocaína/toxicidade , Relação Dose-Resposta a Droga , Dose Letal Mediana , Locomoção/fisiologia , Masculino , Camundongos
16.
Protein Eng Des Sel ; 32(3): 159-166, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31711233

RESUMO

Prostate apoptosis response-4 (Par-4) is a tumor suppressor which protects against neoplastic transformation. Remarkably, Par-4 is capable of inducing apoptosis selectively in cancer cells without affecting the normal cells. In this study, we found that recombinant Par-4 protein had limited serum persistence in mice that may diminish its anti-tumor activity in vivo. To improve the in vivo performance of the short-lived Par-4 protein, we aimed to develop a novel, long-lasting form of Par-4 with extended sequence, denoted as Par-4Ex, without affecting the desirable molecular function of the natural Par-4. We demonstrate that the Par-4Ex protein entity, produced by using the Escherichia coli expression system suitable for large-scale production, fully retains the desirable pro-apoptotic activity of Par-4 protein, but with ~7-fold improved biological half-life. Further in vivo tests confirmed that, due to the prolonged biological half-life, the Par-4Ex protein is indeed more potent in suppressing metastatic tumor growth in mice.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/farmacologia , Engenharia de Proteínas , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/farmacocinética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Distribuição Tecidual
17.
Chem Biol Interact ; 306: 89-95, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-30986387

RESUMO

Human butyrylcholinesterase (BChE) is known as a safe and effective protein for detoxification of organophosphorus (OP) nerve agents. Its rationally designed mutants with considerably improved catalytic activity against cocaine, known as cocaine hydrolases (CocHs), are recognized as the most promising drug candidates for the treatment of cocaine abuse. However, it is a grand challenge to efficiently produce active recombinant BChE and CocHs with a sufficiently long biological half-life. In the present study, starting from a promising CocH, known as CocH3 (i.e. A199S/F227A/S287G/A328W/Y332G mutant of human BChE), which has a ~2000-fold improved catalytic activity against cocaine compared to wild-type BChE, we designed an N-terminal fusion protein, Fc(M3)-(PAPAP)2-CocH3, which was constructed by fusing Fc of human IgG1 to the N-terminal of CocH3 and further optimized by inserting a linker between the two protein domains. Without lowering the enzyme activity, Fc(M3)-(PAPAP)2-CocH3 expressed in Chinese hamster ovary (CHO) cells has not only a long biological half-life of 105 ±â€¯7 h in rats, but also a high yield of protein expression. Particularly, Fc(M3)-(PAPAP)2-CocH3 has a ~21-fold increased protein expression yield in CHO cells compared to CocH3 under the same experimental conditions. Given the observations that Fc(M3)-(PAPAP)2-CocH3 has not only a high catalytic activity against cocaine and a long biological half-life, but also a high yield of protein expression, this new protein entity reported in this study would be a more promising candidate for therapeutic treatment of cocaine overdose and addiction.


Assuntos
Hidrolases de Éster Carboxílico/biossíntese , Hidrolases de Éster Carboxílico/química , Fragmentos Fc das Imunoglobulinas/química , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Animais , Células CHO , Hidrolases de Éster Carboxílico/genética , Cricetulus , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética
18.
Sci Rep ; 8(1): 16762, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30425345

RESUMO

Heroin is a growing national crisis in America. There is an increasing frequency of heroin overdoses. All of the currently used therapeutic approaches to treatment of heroin abuse and other opioid drugs of abuse focus on antagonizing a brain receptor (particularly µ-opiate receptors). However, it has been known that the therapeutic use of certain µ-opiate receptor antagonist may actually increase heroin overdose. Once overdosed, heroin addicts may continue to get overdosed again and again until fatal. Here we report our design and validation of a novel therapeutic strategy targeting heroin activation based on our analysis of the chemical transformation and functional change of heroin in the body. An effective blocker of heroin activation, such as ethopropazine tested in this study, may be used as a standalone therapy or in combination with a currently available, traditional medications targeting µ-opiate receptors (e.g. naltrexone or its extended-release formulation Vivitrol). The combination therapy would be ideal for heroin abuse treatment as the effects of two therapeutic agents targeting two independent mechanisms are cooperative.


Assuntos
Overdose de Drogas/tratamento farmacológico , Heroína/toxicidade , Ativação Metabólica/efeitos dos fármacos , Animais , Biocatálise , Interações Medicamentosas , Overdose de Drogas/metabolismo , Heroína/metabolismo , Humanos , Masculino , Camundongos , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Fenotiazinas/farmacologia , Fenotiazinas/uso terapêutico , Receptores Opioides/metabolismo
19.
AAPS J ; 20(3): 53, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29556863

RESUMO

Cocaine abuse is a worldwide public health and social problem without a US Food and Drug Administration (FDA)-approved medication. Accelerating cocaine metabolism that produces biologically inactive metabolites by administration of an efficient cocaine hydrolase (CocH) has been recognized as a promising strategy for cocaine abuse treatment. However, the therapeutic effects of CocH are limited by its short biological half-life (e.g., 8 h or shorter in rats). In this study, we designed and prepared a set of Fc-fusion proteins constructed by fusing Fc(M3) with CocH3 at the N-terminus of CocH3. A linker between the two protein domains was optimized to improve both the biological half-life and catalytic activity against cocaine. It has been concluded that Fc(M3)-G6S-CocH3 not only has fully retained the catalytic efficiency of CocH3 against cocaine but also has the longest biological half-life (e.g., ∼ 136 h in rats) among all of the long-acting CocHs identified so far. A single dose (0.2 mg/kg, IV) of Fc(M3)-G6S-CocH3 was able to significantly attenuate 15 mg/kg cocaine-induced hyperactivity for at least 11 days (268 h) after the Fc(M3)-G6S-CocH3 administration.


Assuntos
Hidrolases de Éster Carboxílico/química , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Fragmentos Fc das Imunoglobulinas/química , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Recombinantes/química , Animais , Células CHO , Catálise , Cricetulus , Meia-Vida , Masculino , Engenharia de Proteínas , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/farmacocinética
20.
Sci Rep ; 8(1): 5205, 2018 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-29581541

RESUMO

Human mPGES-1 is recognized as a promising target for next generation of anti-inflammatory drugs without the side effects of currently available anti-inflammatory drugs, and various inhibitors have been reported in the literature. However, none of the reported potent inhibitors of human mPGES-1 has shown to be also a potent inhibitor of mouse or rat mPGES-1, which prevents using the well-established mouse/rat models of inflammation-related diseases for preclinical studies. Hence, despite of extensive efforts to design and discover various human mPGES-1 inhibitors, the promise of mPGES-1 as a target for the next generation of anti-inflammatory drugs has never been demonstrated in any wild-type mouse/rat model using an mPGES-1 inhibitor. Here we report discovery of a novel type of selective mPGES-1 inhibitors potent for both human and mouse mPGES-1 enzymes through structure-based rational design. Based on in vivo studies using wild-type mice, the lead compound is indeed non-toxic, orally bioavailable, and more potent in decreasing the PGE2 (an inflammatory marker) levels compared to the currently available drug celecoxib. This is the first demonstration in wild-type mice that mPGES-1 is truly a promising target for the next generation of anti-inflammatory drugs.


Assuntos
Anti-Inflamatórios/química , Inibidores Enzimáticos/química , Inflamação/tratamento farmacológico , Prostaglandina-E Sintases/química , Animais , Anti-Inflamatórios/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Humanos , Inflamação/genética , Inflamação/patologia , Camundongos , Prostaglandina-E Sintases/antagonistas & inibidores , Ratos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA