Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Chem Soc Rev ; 51(10): 3886-3897, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35506708

RESUMEN

Degrader-antibody conjugates (DACs) are novel entities that combine a proteolysis targeting chimera (PROTAC) payload with a monoclonal antibody via some type of chemical linker. This review provides a current summary of the DAC field. Many general aspects associated with the creation and biological performance of traditional cytotoxic antibody-drug conjugates (ADCs) are initially presented. These characteristics are subsequently compared and contrasted with related parameters that impact DAC generation and biological activity. Several examples of DACs assembled from both the scientific and the patent literature are utilized to highlight differing strategies for DAC creation, and specific challenges associated with DAC construction are documented. Collectively, the assembled examples demonstrate that biologically-active DACs can be successfully prepared using a variety of PROTAC payloads which employ diverse E3 ligases to degrade multiple protein targets.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Anticuerpos Monoclonales , Proteolisis
2.
Bioorg Med Chem Lett ; 59: 128576, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35065235

RESUMEN

Structure-based design was utilized to optimize 6,6-diaryl substituted dihydropyrone and hydroxylactam to obtain inhibitors of lactate dehydrogenase (LDH) with low nanomolar biochemical and single-digit micromolar cellular potencies. Surprisingly the replacement of a phenyl with a pyridyl moiety in the chemical structure revealed a new binding mode for the inhibitors with subtle conformational change of the LDHA active site. This led to the identification of a potent, cell-active hydroxylactam inhibitor exhibiting an in vivo pharmacokinetic profile suitable for mouse tumor xenograft study.


Asunto(s)
Inhibidores Enzimáticos/farmacología , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Lactamas/farmacología , Animales , Línea Celular , Perros , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Humanos , L-Lactato Deshidrogenasa/metabolismo , Lactamas/química , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Relación Estructura-Actividad
3.
Bioorg Med Chem Lett ; 30(4): 126907, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31902710

RESUMEN

Chimeric molecules which effect intracellular degradation of target proteins via E3 ligase-mediated ubiquitination (e.g., PROTACs) are currently of high interest in medicinal chemistry. However, these entities are relatively large compounds that often possess molecular characteristics which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. Accordingly, we explored whether conjugation of chimeric degraders to monoclonal antibodies using technologies originally developed for cytotoxic payloads might provide alternate delivery options for these novel agents. In this report we describe the construction of several degrader-antibody conjugates comprised of two distinct ERα-targeting degrader entities and three independent ADC linker modalities. We subsequently demonstrate the antigen-dependent delivery to MCF7-neo/HER2 cells of the degrader payloads that are incorporated into these conjugates. We also provide evidence for efficient intracellular degrader release from one of the employed linkers. In addition, preliminary data are described which suggest that reasonably favorable in vivo stability properties are associated with the linkers utilized to construct the degrader conjugates.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Portadores de Fármacos/química , Receptor alfa de Estrógeno/inmunología , Anticuerpos Monoclonales/química , Antineoplásicos/química , Antineoplásicos/inmunología , Antineoplásicos/farmacología , Diseño de Fármacos , Receptor alfa de Estrógeno/metabolismo , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoconjugados/farmacología , Células MCF-7 , Proteolisis/efectos de los fármacos , Receptor ErbB-2/metabolismo
4.
Bioconjug Chem ; 30(5): 1356-1370, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30966735

RESUMEN

This work discloses the first examples of antibody-drug conjugates (ADCs) that are constructed from linker-drugs bearing dimeric seco-CBI payloads (duocarmycin analogs). Several homogeneous, CD22-targeting THIOMAB antibody-drug conjugates (TDCs) containing the dimeric seco-CBI entities are shown to be highly efficacious in the WSU-DLCL2 and BJAB mouse xenograft models. Surprisingly, the seco-CBI-containing conjugates are also observed to undergo significant biotransformation in vivo in mice, rats, and monkeys and thereby form 1:1 adducts with the Alpha-1-Microglobulin (A1M) plasma protein from these species. Variation of both the payload mAb attachment site and length of the linker-drug is shown to alter the rates of adduct formation. Subsequent experiments demonstrated that adduct formation attenuates the in vitro antiproliferation activity of the affected seco-CBI-dimer TDCs, but does not significantly impact the in vivo efficacy of the conjugates. In vitro assays employing phosphatase-treated whole blood suggest that A1M adduct formation is likely to occur if the seco-CBI-dimer TDCs are administered to humans. Importantly, protein adduct formation leads to the underestimation of total antibody (Tab) concentrations using an ELISA assay but does not affect Tab values determined via an orthogonal LC-MS/MS method. Several recommendations regarding bioanalysis of future in vivo studies involving related seco-CBI-containing ADCs are provided based on these collective findings.


Asunto(s)
alfa-Globulinas/química , Antineoplásicos/farmacología , Inmunoconjugados/farmacología , Animales , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dimerización , Haplorrinos , Humanos , Inmunoconjugados/química , Ratones , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Drug Metab Dispos ; 47(8): 884-889, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31072822

RESUMEN

Carfilzomib (CFZ) is a proteasome inhibitor used for oncology indications including treating multiple myeloma. CFZ is a potent cytotoxic agent with an IC50 value in the nanomolar range in various cancer cell lines and was considered as a potential payload for antibody drug conjugates (ADCs); however, the conjugated CFZ to anti-CD22 or anti-HER2 antibody totally abolishes the in vitro potency. This was a surprise since with other payloads such as monomethyl auristatin E (MMAE), where potent antiproliferation efficacy was retained as MMAE alone or as a payload in an ADC. Further investigations were conducted using CFZ alone, CFZ with a linker, and CFZ-ADC with tissue matrices including lysosomal enzymes. With CFZ linked to the ADC, cathepsin B (a lysosomal enzyme) was efficient in liberating CFZ from the ADC by cleavage of the valine-citrulline linker. At the same time, the liberated CFZ in the lysosome was inactivated due to further metabolism by lysosomal enzymes. The products from epoxide and amide hydrolysis were identified from these incubations. These results suggested that the CFZ-ADC upon uptake and internalization specifically delivers CFZ payload to the lysosomes, where CFZ was inactivated. On the other hand, CFZ by itself is not as vulnerable and could reach its target. Therefore, lysosomal stability is an important criterion in the selection of a payload for making the next generation of potent ADC therapeutics.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/farmacocinética , Inmunoconjugados/farmacocinética , Lisosomas/enzimología , Oligopéptidos/farmacocinética , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Catepsina B/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Estudios de Factibilidad , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Receptor ErbB-2/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores
6.
Drug Metab Dispos ; 47(10): 1146-1155, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31358513

RESUMEN

Antibody-drug conjugates (ADCs) contain a disease-receptor antibody and a payload drug connected via a linker. The payload delivery depends on both tumor properties and ADC characteristics. In this study, we used different linkers, attachment sites, and doses to modulate payload delivery of several ADCs bearing maytansinoids (e.g., DM1), auristatins (e.g., MMAE), and DNA alkylating agents [e.g., pyrrolo[2,1-c][1,4]benzodiazepine-dimer (PBD)] as payloads in HER2- or CD22-expressing xenograft models. The tumor growth inhibition and ADC stability and exposure data were collected and analyzed from these dosed animals. The trend analysis suggests that intratumoral payload exposures that directly related the combination of conjugate linker and dose correlate with the corresponding efficacies of three payload types in two antigen-expressing xenograft models. These preliminary correlations also suggest that a minimal threshold concentration of intratumoral payload is required to support sustained efficacy. In addition, an ADC can deliver an excessive level of payload to tumors that does not enhance efficacy ("Plateau" effect). In contrast to tumor payload concentrations, the assessments of systemic exposures of total antibody (Tab) as well as the linker, dose, site of attachment, plasma stability, and drug-to-antibody ratio changes of these ADCs did not consistently rationalize the observed ADC efficacies. The requirement of a threshold payload concentration for efficacy is further supported by dose fractionation studies with DM1-, MMAE-, and PBD-containing ADCs, which demonstrated that single-dose regimens showed better efficacies than fractionated dosing. Overall, this study demonstrates that 1) the linker and dose together determine the tissue payload concentration that correlates with the antitumor efficacy of ADCs and 2) an ADC can deliver an unnecessary level of payload to tumors in xenograft models.


Asunto(s)
Antineoplásicos Inmunológicos/farmacocinética , Inmunoconjugados/farmacocinética , Receptor ErbB-2/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Ado-Trastuzumab Emtansina/administración & dosificación , Ado-Trastuzumab Emtansina/farmacocinética , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/química , Benzodiazepinas/química , Brentuximab Vedotina/administración & dosificación , Brentuximab Vedotina/farmacocinética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunoconjugados/administración & dosificación , Ratones , Ratones Transgénicos , Pirroles/química , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Drug Metab Dispos ; 47(10): 1156-1163, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31085544

RESUMEN

In cells, catalytic disulfide cleavage is an essential mechanism in protein folding and synthesis. However, detailed enzymatic catalytic mechanism relating cleavage of disulfide bonds in xenobiotics is not well understood. This study reports an enzymatic mechanism of cleavage of disulfide bonds in xenobiotic small molecules and antibody conjugate (ADC) linkers. The chemically stable disulfide bonds in substituted disulfide-containing pyrrolobenzodiazepine (PBD, pyrrolo[2,1-c][1,4]benzodiazepine) monomer prodrugs in presence of glutathione or cysteine were found to be unstable in incubations in whole blood of humans and rats. It was shown the enzymes involved were thioredoxin (TRX) and glutaredoxin (GRX). For a diverse set of drug-linker conjugates, we determined that TRX in the presence of TRX-reductase and NADPH generated the cleaved products that are consistent with catalytic disulfide cleavage and linker immolation. GRX was less rigorously studied; in the set of compounds studied, its role in the catalytic cleavage was also confirmed. Collectively, these in vitro experiments demonstrate that TRX as well as GRX can catalyze the cleavage of disulfide bonds in both small molecules and linkers of ADCs.


Asunto(s)
Glutarredoxinas/metabolismo , Inmunoconjugados/farmacocinética , Tiorredoxinas/metabolismo , Animales , Benzodiazepinas/química , Benzodiazepinas/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Femenino , Humanos , Inmunoconjugados/química , Masculino , Pirroles/química , Pirroles/metabolismo , Ratas , Proteínas Recombinantes/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo
8.
Xenobiotica ; 49(9): 1063-1077, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30257601

RESUMEN

GNE-617 (N-(4-((3,5-difluorophenyl)sulfonyl)benzyl)imidazo[1,2-a]pyridine-6-carboxamide) is a potent, selective nicotinamide phosphoribosyltransferase (NAMPT) inhibitor being explored as a potential treatment for human cancers. Plasma clearance was low in monkeys and dogs (9.14 mL min-1 kg-1 and 4.62 mL min-1 kg-1, respectively) and moderate in mice and rats (36.4 mL min-1 kg-1 and 19.3 mL min-1 kg-1, respectively). Oral bioavailability in mice, rats, monkeys and dogs was 29.7, 33.9, 29.4 and 65.2%, respectively. Allometric scaling predicted a low clearance of 3.3 mL min-1 kg-1 and a volume of distribution of 1.3 L kg-1 in human. Efficacy (57% tumor growth inhibition) in Colo-205 CRC tumor xenograft mice was observed at an oral dose of 15 mg/kg BID (AUC = 10.4 µM h). Plasma protein binding was moderately high. GNE-617 was stable to moderately stable in vitro. Main human metabolites identified in human hepatocytes were formed primarily by CYP3A4/5. Transporter studies suggested that GNE-617 is likely a substrate for MDR1 but not for BCRP. Simcyp® simulations suggested a low (CYP2C9 and CYP2C8) or moderate (CYP3A4/5) potential for drug-drug interactions. The potential for autoinhibition was low. Overall, GNE-617 exhibited acceptable preclinical properties and projected human PK and dose estimates.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Heterocíclicos con 2 Anillos/farmacología , Sulfonas/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Neoplasias Colorrectales/tratamiento farmacológico , Sistema Enzimático del Citocromo P-450/metabolismo , Citocinas/antagonistas & inhibidores , Perros , Interacciones Farmacológicas , Estabilidad de Medicamentos , Femenino , Compuestos Heterocíclicos con 2 Anillos/administración & dosificación , Compuestos Heterocíclicos con 2 Anillos/farmacocinética , Humanos , Macaca fascicularis , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Ratas Sprague-Dawley , Sulfonas/administración & dosificación , Sulfonas/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Bioconjug Chem ; 29(2): 267-274, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29369629

RESUMEN

The valine-citrulline (Val-Cit) dipeptide and p-aminobenzyl (PAB) spacer have been commonly used as a cleavable self-immolating linker in ADC design including in the clinically approved ADC, brentuximab vedotin (Adcetris). When the same linker was used to connect to the phenol of the cyclopropabenzindolone (CBI) (P1), the resulting ADC1 showed loss of potency in CD22 target-expressing cancer cell lines (e.g., BJAB, WSU-DLCL2). In comparison, the conjugate (ADC2) of a cyclopropapyrroloindolone (CPI) (P2) was potent despite the two corresponding free drugs having similar picomolar cell-killing activity. Although the corresponding spirocyclization products of P1 and P2, responsible for DNA alkylation, are a prominent component in buffer, the linker immolation was slow when the PAB was connected as an ether (PABE) to the phenol in P1 compared to that in P2. Additional immolation studies with two other PABE-linked substituted phenol compounds showed that electron-withdrawing groups accelerated the immolation to release an acidic phenol-containing payload (to delocalize the negative charge on the anticipated anionic phenol oxygen during immolation). In contrast, efficient immolation of LD4 did not result in an active ADC4 because the payload (P4) had a low potency to kill cells. In addition, nonimmolation of LD5 did not affect the cell-killing potency of its ADC5 since immolation is not required for DNA alkylation by the center-linked pyrrolobenzodiazepine. Therefore, careful evaluation needs to be conducted when the Val-Cit-PAB linker is used to connect antibodies to a phenol-containing drug as the linker immolation, as well as payload potency and stability, affects the cell-killing activity of an ADC.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Inmunoconjugados/química , Inmunoconjugados/farmacología , Fenol/química , Fenol/farmacología , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Brentuximab Vedotina , Línea Celular Tumoral , Ciclopropanos/química , Ciclopropanos/farmacología , Humanos , Neoplasias/tratamiento farmacológico
10.
Chemistry ; 24(19): 4830-4834, 2018 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-29493023

RESUMEN

A novel strategy to attach indole-containing payloads to antibodies through a carbamate moiety and a self-immolating, disulfide-based linker is described. This new strategy was employed to connect a selective estrogen receptor down-regulator (SERD) to various antibodies in a site-selective manner. The resulting conjugates displayed potent, antigen-dependent down-regulation of estrogen receptor levels in MCF7-neo/HER2 and MCF7-hB7H4 cells. They also exhibited similar antigen-dependent modulation of the estrogen receptor in tumors when administered intravenously to mice bearing MCF7-neo/HER2 tumor xenografts. The indole-carbamate moiety present in the new linker was stable in whole blood from various species and also exhibited good in vivo stability properties in mice.


Asunto(s)
Indoles/química , Animales , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Humanos , Inmunoconjugados/administración & dosificación , Células MCF-7 , Ratones
11.
Mol Pharm ; 15(9): 3979-3996, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30040421

RESUMEN

A number of cytotoxic pyrrolobenzodiazepine (PBD) monomers containing various disulfide-based prodrugs were evaluated for their ability to undergo activation (disulfide cleavage) in vitro in the presence of either glutathione (GSH) or cysteine (Cys). A good correlation was observed between in vitro GSH stability and in vitro cytotoxicity toward tumor cell lines. The prodrug-containing compounds were typically more potent against cells with relatively high intracellular GSH levels (e.g., KPL-4 cells). Several antibody-drug conjugates (ADCs) were subsequently constructed from PBD dimers that incorporated selected disulfide-based prodrugs. Such HER2 conjugates exhibited potent antiproliferation activity against KPL-4 cells in vitro in an antigen-dependent manner. However, the disulfide prodrugs contained in the majority of such entities were surprisingly unstable toward whole blood from various species. One HER2-targeting conjugate that contained a thiophenol-derived disulfide prodrug was an exception to this stability trend. It exhibited potent activity in a KPL-4 in vivo efficacy model that was approximately three-fold weaker than that displayed by the corresponding parent ADC. The same prodrug-containing conjugate demonstrated a three-fold improvement in mouse tolerability properties in vivo relative to the parent ADC, which did not contain the prodrug.


Asunto(s)
Benzodiazepinas/química , Disulfuros/química , Inmunoconjugados/química , Profármacos/química , Pirroles/química , Línea Celular Tumoral , Cisteína/metabolismo , Glutatión/metabolismo , Humanos , Inmunoconjugados/metabolismo , Estructura Molecular
12.
Bioorg Med Chem Lett ; 27(23): 5300-5304, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29079474

RESUMEN

The ability of various pyrrolobenzodiazepine(PBD)-containing cytotoxic compounds to function as hypoxia-activated prodrugs was assessed. These molecules incorporated a 1-methyl-2-nitro-1H-imidazole hypoxia-activated trigger (present in the clinically evaluated compound TH-302) in a manner that masked a reactive imine moiety required for cytotoxic activity. Incubation of the prodrugs with cytochrome P450-reductase under normoxic and hypoxic conditions revealed that some, but not all, were efficient substrates for the enzyme. In these experiments, prodrugs derived from PBD-monomers underwent rapid conversion to the parent cytotoxic compounds under low-oxygen conditions while related PBD-dimers did not. The ability of a given prodrug to function as an efficient cytochrome P450-reductase substrate correlated with the ratio of cytotoxic potencies measured for the compound against NCI460 cells under normoxic and hypoxic conditions.


Asunto(s)
Benzodiazepinas/farmacología , Diseño de Fármacos , Hipoxia/metabolismo , Profármacos/farmacología , Pirroles/farmacología , Benzodiazepinas/síntesis química , Benzodiazepinas/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , NADPH-Ferrihemoproteína Reductasa/metabolismo , Profármacos/síntesis química , Profármacos/química , Pirroles/síntesis química , Pirroles/química , Relación Estructura-Actividad
13.
Bioorg Med Chem Lett ; 27(13): 2974-2981, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28512031

RESUMEN

A high-throughput screening (HTS) of the Genentech/Roche library identified a novel, uncharged scaffold as a KDM5A inhibitor. Lacking insight into the binding mode, initial attempts to improve inhibitor potency failed to improve potency, and synthesis of analogs was further hampered by the presence of a C-C bond between the pyrrolidine and pyridine. Replacing this with a C-N bond significantly simplified synthesis, yielding pyrazole analog 35, of which we obtained a co-crystal structure with KDM5A. Using structure-based design approach, we identified 50 with improved biochemical, cell potency and reduced MW and lower lipophilicity (LogD) compared with the original hit. Furthermore, 50 showed lower clearance than 9 in mice. In combination with its remarkably low plasma protein binding (PPB) in mice (40%), oral dosing of 50 at 5mg/kg resulted in unbound Cmax ∼2-fold of its cell potency (PC9 H3K4Me3 0.96µM), meeting our criteria for an in vivo tool compound from a new scaffold.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Pirazoles/farmacología , Proteína 2 de Unión a Retinoblastoma/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Humanos , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Pirazoles/administración & dosificación , Pirazoles/química , Ratas , Proteína 2 de Unión a Retinoblastoma/metabolismo , Relación Estructura-Actividad
14.
Drug Metab Dispos ; 44(9): 1517-23, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417182

RESUMEN

Despite recent technological advances in quantifying antibody drug conjugate (ADC) species, such as total antibody, conjugated antibody, conjugated drug, and payload drug in circulation, the correlation of their exposures with the efficacy of ADC outcomes in vivo remains challenging. Here, the chemical structures and concentrations of intratumor catabolites were investigated to better understand the drivers of ADC in vivo efficacy. Anti-CD22 disulfide-linked pyrrolobenzodiazepine (PBD-dimer) conjugates containing methyl- and cyclobutyl-substituted disulfide linkers exhibited strong efficacy in a WSU-DLCL2 xenograft mouse model, whereas an ADC derived from a cyclopropyl linker was inactive. Total ADC antibody concentrations and drug-to-antibody ratios (DAR) in circulation were similar between the cyclobutyl-containing ADC and the cyclopropyl-containing ADC; however, the former afforded the release of the PBD-dimer payload in the tumor, but the latter only generated a nonimmolating thiol-containing catabolite that did not bind to DNA. These results suggest that intratumor catabolite analysis rather than systemic pharmacokinetic analysis may be used to better explain and predict ADC in vivo efficacy. These are good examples to demonstrate that the chemical nature and concentration of intratumor catabolites depend on the linker type used for drug conjugation, and the potency of the released drug moiety ultimately determines the ADC in vivo efficacy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Benzodiazepinas/farmacocinética , Inmunoconjugados/farmacocinética , Neoplasias/metabolismo , Pirroles/farmacocinética , Animales , Anticuerpos Monoclonales Humanizados/química , Benzodiazepinas/química , Femenino , Inmunoconjugados/química , Ratones , Ratones SCID , Pirroles/química , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Drug Metab Dispos ; 44(12): 1958-1962, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27683653

RESUMEN

Pyrrolobenzodiazepine (PBD)-dimer is a DNA minor groove alkylator, and its CD22 THIOMAB antibody drug conjugate (ADC) demonstrated, through a disulfide linker, an efficacy in tumor reduction for more than 7 weeks with minimal body weight loss in xenograft mice after a single 0.5-1 mg/kg i.v. dose. The DNA alkylation was investigated here in tumors and healthy organs of mice to understand the sustained efficacy and tolerability. The experimental procedures included the collection of tumors and organ tissues of xenograft mice treated with the ADC followed by DNA isolation/hydrolysis/quantitation and payload recovery from reversible DNA alkylation. PBD-dimer formed a considerable amount of adducts with tissue DNA, representing approximately 98% (at 24 hours), and 99% (at 96 hours) of the total PBD-dimer in tumors, and 78-89% in liver and lung tissues, suggesting highly efficient covalent binding of the released PBD-dimer to tissue DNA. The amount of PBD-DNA adducts in tumor tissues was approximately 24-fold (at 24 hours) and 70-fold (at 96 hours) greater than the corresponding amount of adducts in liver and lung tissues. In addition, the DNA alkylation levels increased 3-fold to 4-fold from 24 to 96 hours in tumors [41/106 base pairs (bp) at 96 hours] but remained at the same level (1/106 bp) in livers and lungs. These results support the typical target-mediated cumulative uptake of ADC into tumors and payload release that offers an explanation for its sustained antitumor efficacy. In addition, the low level of DNA alkylation in normal tissues is consistent with the tolerability observed in mice.


Asunto(s)
Alquilación/fisiología , Anticuerpos/metabolismo , Benzodiazepinas/metabolismo , ADN/metabolismo , Pirroles/metabolismo , Animales , Aductos de ADN/metabolismo , Xenoinjertos/metabolismo , Inmunoconjugados/metabolismo , Hígado/metabolismo , Pulmón/metabolismo , Ratones , Neoplasias/metabolismo
16.
Bioorg Med Chem Lett ; 26(16): 4036-41, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27406798

RESUMEN

Starting with a lead [1,5-a]pyrimidin-7(4H)-one-containing molecule (1), we generated potent, selective and orally bioavailable KDM5 inhibitors. Using structure- and property-based approaches, we designed 48 with improved cell potency (PC9 H3K4Me3 EC50=0.34µM). Furthermore, 48 maintained suitable physiochemical properties and displayed an excellent pharmacokinetic (PK) profile in mice. When dosed orally in mice at 50mg/kg twice a day (BID), 48 showed an unbound maximal plasma concentration (Cmax) >15-fold over its cell EC50, thereby providing a robust chemical probe for studying KDM5 biological functions in vivo.


Asunto(s)
Pirazoles/química , Pirimidinonas/química , Proteína 2 de Unión a Retinoblastoma/antagonistas & inhibidores , Administración Oral , Animales , Sitios de Unión , Cristalografía por Rayos X , Femenino , Semivida , Histonas/metabolismo , Humanos , Hígado/metabolismo , Ratones , Microsomas Hepáticos/metabolismo , Simulación de Dinámica Molecular , Pirazoles/síntesis química , Pirazoles/farmacocinética , Pirimidinonas/sangre , Pirimidinonas/síntesis química , Pirimidinonas/farmacocinética , Ratas , Proteína 2 de Unión a Retinoblastoma/metabolismo , Relación Estructura-Actividad
17.
Bioorg Med Chem Lett ; 26(18): 4492-4496, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27499454

RESUMEN

Features from a high throughput screening (HTS) hit and a previously reported scaffold were combined to generate 1,7-naphthyridones as novel KDM5 enzyme inhibitors with nanomolar potencies. These molecules exhibited high selectivity over the related KDM4C and KDM2B isoforms. An X-ray co-crystal structure of a representative molecule bound to KDM5A showed that these inhibitors are competitive with the co-substrate (2-oxoglutarate or 2-OG).


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Naftiridinas/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Proteína 2 de Unión a Retinoblastoma/antagonistas & inhibidores , Animales , Cristalografía por Rayos X , Perros , Diseño de Fármacos , Humanos , Células de Riñón Canino Madin Darby , Naftiridinas/química , Relación Estructura-Actividad
18.
Bioorg Med Chem Lett ; 25(1): 75-82, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25466195

RESUMEN

Optimization of 5-(2,6-dichlorophenyl)-3-hydroxy-2-mercaptocyclohex-2-enone using structure-based design strategies resulted in inhibitors with considerable improvement in biochemical potency against human lactate dehydrogenase A (LDHA). These potent inhibitors were typically selective for LDHA over LDHB isoform (4­10 fold) and other structurally related malate dehydrogenases, MDH1 and MDH2 (>500 fold). An X-ray crystal structure of enzymatically most potent molecule bound to LDHA revealed two additional interactions associated with enhanced biochemical potency.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Animales , Cristalografía por Rayos X , Perros , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , L-Lactato Deshidrogenasa/metabolismo , Células de Riñón Canino Madin Darby
19.
Bioorg Med Chem Lett ; 25(3): 529-41, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25556090

RESUMEN

Herein we report the optimization efforts to ameliorate the potent CYP3A4 time-dependent inhibition (TDI) and low aqueous solubility exhibited by a previously identified lead compound from our NAMPT inhibitor program (1, GNE-617). Metabolite identification studies pinpointed the imidazopyridine moiety present in 1 as the likely source of the TDI signal, and replacement with other bicyclic systems was found to reduce or eliminate the TDI finding. A strategy of reducing the number of aromatic rings and/or lowering cLogD7.4 was then employed to significantly improve aqueous solubility. These efforts culminated in the discovery of 42, a compound with no evidence of TDI, improved aqueous solubility, and robust efficacy in tumor xenograft studies.


Asunto(s)
Citocromo P-450 CYP3A/química , Inhibidores Enzimáticos/química , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Animales , Sitios de Unión , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A/química , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Inhibidores del Citocromo P-450 CYP3A/toxicidad , Perros , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Femenino , Semivida , Humanos , Cinética , Células de Riñón Canino Madin Darby , Ratones , Ratones Desnudos , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Nicotinamida Fosforribosiltransferasa/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Pirimidinas/química , Pirimidinas/uso terapéutico , Pirimidinas/toxicidad , Solubilidad , Relación Estructura-Actividad , Termodinámica , Trasplante Heterólogo , Agua/química
20.
Toxicol Mech Methods ; 25(3): 201-11, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25894564

RESUMEN

Nicotinamide adenine dinucleotide (NAD) is an essential co-factor in glycolysis and is a key molecule involved in maintaining cellular energy metabolism. Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step of an important salvage pathway in which nicotinamide is recycled into NAD. NAMPT is up-regulated in many types of cancer and NAMPT inhibitors (NAMPTi) have potential therapeutic benefit in cancer by impairing tumor metabolism. Clinical trials with NAMPTi APO-866 and GMX-1778, however, failed to reach projected efficacious exposures due to dose-limiting thrombocytopenia. We evaluated preclinical models for thrombocytopenia that could be used in candidate drug selection and risk mitigation strategies for NAMPTi-related toxicity. Rats treated with a suite of structurally diverse and potent NAMPTi at maximum tolerated doses had decreased reticulocyte and lymphocyte counts, but no thrombocytopenia. We therefore evaluated and qualified a human colony forming unit-megakaryocyte (CFU-MK) as in vitro predictive model of NAMPTi-induced MK toxicity and thrombocytopenia. We further demonstrate that the MK toxicity is on-target based on the evidence that nicotinic acid (NA), which is converted to NAD via a NAMPT-independent pathway, can mitigate NAMPTi toxicity to human CFU-MK in vitro and was also protective for the hematotoxicity in rats in vivo. Finally, assessment of CFU-MK and human platelet bioenergetics and function show that NAMPTi was toxic to MK and not platelets, which is consistent with the clinically observed time-course of thrombocytopenia.


Asunto(s)
Antineoplásicos/efectos adversos , Inhibidores Enzimáticos/efectos adversos , Hematopoyesis/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Niacina/metabolismo , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Trombocitopenia/inducido químicamente , Animales , Antineoplásicos/química , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Suplementos Dietéticos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Interacciones Alimento-Droga , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Macaca fascicularis , Masculino , Megacariocitos/citología , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Estructura Molecular , Niacina/uso terapéutico , Nicotinamida Fosforribosiltransferasa/genética , Nicotinamida Fosforribosiltransferasa/metabolismo , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Ratas Sprague-Dawley , Trombocitopenia/metabolismo , Trombocitopenia/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA