Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Bioconjug Chem ; 34(3): 510-517, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36787347

RESUMEN

Cysteines are routinely used as site-specific handles to synthesize antibody-drug conjugates for targeted immunotherapy applications. Michael additions between thiols and maleimides are some of the most common methods for modifying cysteines, but these functional groups can be difficult to prepare on scale, and the resulting linkages have been shown to be reversible under some physiological conditions. Here, we show that the enzyme tyrosinase, which oxidizes conveniently accessed phenols to afford reactive ortho-quinone intermediates, can be used to attach phenolic cargo to cysteines engineered on antibody surfaces. The resulting linkages between the thiols and ortho-quinones are shown to be more resistant than maleimides to reversion under physiological conditions. Using this approach, we construct antibody conjugates bearing cytotoxic payloads, which exhibit targeted cell killing, and further demonstrate this method for the attachment of a variety of cargo to antibodies, including fluorophores and oligonucleotides.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Cisteína , Acoplamiento Oxidativo , Compuestos de Sulfhidrilo , Quinonas , Maleimidas
2.
Nature ; 527(7578): 323-8, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26536114

RESUMEN

Staphylococcus aureus is considered to be an extracellular pathogen. However, survival of S. aureus within host cells may provide a reservoir relatively protected from antibiotics, thus enabling long-term colonization of the host and explaining clinical failures and relapses after antibiotic therapy. Here we confirm that intracellular reservoirs of S. aureus in mice comprise a virulent subset of bacteria that can establish infection even in the presence of vancomycin, and we introduce a novel therapeutic that effectively kills intracellular S. aureus. This antibody-antibiotic conjugate consists of an anti-S. aureus antibody conjugated to a highly efficacious antibiotic that is activated only after it is released in the proteolytic environment of the phagolysosome. The antibody-antibiotic conjugate is superior to vancomycin for treatment of bacteraemia and provides direct evidence that intracellular S. aureus represents an important component of invasive infections.


Asunto(s)
Antibacterianos/farmacología , Bacteriemia , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Espacio Intracelular/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Vancomicina/farmacología , Animales , Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Bacteriemia/microbiología , Portador Sano/tratamiento farmacológico , Portador Sano/microbiología , Diseño de Fármacos , Femenino , Inmunoconjugados/química , Espacio Intracelular/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Pruebas de Sensibilidad Microbiana , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/microbiología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/patología , Staphylococcus aureus/patogenicidad , Vancomicina/uso terapéutico
3.
Nat Chem Biol ; 9(6): 390-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23603658

RESUMEN

The prosurvival BCL-2 family protein BCL-X(L) is often overexpressed in solid tumors and renders malignant tumor cells resistant to anticancer therapeutics. Enhancing apoptotic responses by inhibiting BCL-X(L) will most likely have widespread utility in cancer treatment and, instead of inhibiting multiple prosurvival BCL-2 family members, a BCL-X(L)-selective inhibitor would be expected to minimize the toxicity to normal tissues. We describe the use of a high-throughput screen to discover a new series of small molecules targeting BCL-X(L) and their structure-guided development by medicinal chemistry. The optimized compound, WEHI-539 (7), has high affinity (subnanomolar) and selectivity for BCL-X(L) and potently kills cells by selectively antagonizing its prosurvival activity. WEHI-539 will be an invaluable tool for distinguishing the roles of BCL-X(L) from those of its prosurvival relatives, both in normal cells and notably in malignant tumor cells, many of which may prove to rely upon BCL-X(L) for their sustained growth.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Proteína bcl-X/antagonistas & inhibidores , Proteína bcl-X/química , Animales , Apoptosis , Benzotiazoles/química , Línea Celular Tumoral , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Hidrazonas/química , Cinética , Ratones , Modelos Químicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética
4.
Drug Metab Dispos ; 41(12): 2104-13, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24041744

RESUMEN

The application of modeling and simulation techniques is increasingly common in the preclinical stages of the drug development process. GDC-0917 [(S)-1-((S)-2-cyclohexyl-2-((S)-2-(methylamino)propanamido)acetyl)-N-(2-(oxazol-2-yl)-4-phenylthiazol-5-yl)pyrrolidine-2-carboxamide] is a potent second-generation antagonist of inhibitor of apoptosis (IAP) proteins that is being developed for the treatment of various cancers. GDC-0917 has low to moderate clearance in the mouse (12.0 ml/min/kg), rat (27.0 ml/min/kg), and dog (15.3 ml/min/kg), and high clearance in the monkey (67.6 ml/min/kg). Accordingly, oral bioavailability was lowest in monkeys compared with other species. Based on our experience with a prototype molecule with similar structure, in vitro-in vivo extrapolation was used to predict a moderate clearance (11.5 ml/min/kg) in humans. The predicted human volume of distribution was estimated using simple allometry at 6.69 l/kg. Translational pharmacokinetic-pharmacodynamic (PK-PD) analysis using results from MDA-MB-231-X1.1 breast cancer xenograft studies and predicted human pharmacokinetics suggests that ED50 and ED90 targets can be achieved in humans using acceptable doses (72 mg and 660 mg, respectively) and under an acceptable time frame. The relationship between GDC-0917 concentrations and pharmacodynamic response (cIAP1 degradation) was characterized using an in vitro peripheral blood mononuclear cell immunoassay. Simulations of human GDC-0917 plasma concentration-time profile and cIAP1 degradation at the 5-mg starting dose in the phase 1 clinical trial agreed well with observations. This work shows the importance of leveraging information from prototype molecules and illustrates how modeling and simulation can be used to add value to preclinical studies in the early stages of the drug development process.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/farmacocinética , Animales , Disponibilidad Biológica , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Perros , Evaluación Preclínica de Medicamentos , Femenino , Semivida , Hepatocitos/efectos de los fármacos , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones SCID , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Cell Chem Biol ; 30(11): 1468-1477.e6, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37820725

RESUMEN

Dysregulated iron homeostasis underlies diverse pathologies, from ischemia-reperfusion injury to epithelial-mesenchymal transition and drug-tolerant "persister" cancer cell states. Here, we introduce ferrous iron-activatable luciferin-1 (FeAL-1), a small-molecule probe for bioluminescent imaging of the labile iron pool (LIP) in luciferase-expressing cells and animals. We find that FeAL-1 detects LIP fluctuations in cells after iron supplementation, depletion, or treatment with hepcidin, the master regulator of systemic iron in mammalian physiology. Utilizing FeAL-1 and a dual-luciferase reporter system, we quantify LIP in mouse liver and three different orthotopic pancreatic ductal adenocarcinoma tumors. We observed up to a 10-fold increase in FeAL-1 bioluminescent signal in xenograft tumors as compared to healthy liver, the major organ of iron storage in mammals. Treating mice with hepcidin further elevated hepatic LIP, as predicted. These studies reveal a therapeutic index between tumoral and hepatic LIP and suggest an approach to sensitize tumors toward LIP-activated therapeutics.


Asunto(s)
Hierro , Neoplasias , Humanos , Ratones , Animales , Hepcidinas , Luciferinas , Xenoinjertos , Hígado , Luciferasas , Mamíferos
6.
Bioorg Med Chem Lett ; 20(7): 2229-33, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20189383

RESUMEN

A series of IAP antagonists based on thiazole or benzothiazole amide isosteres was designed and synthesized. These compounds were tested for binding to the XIAP-BIR3 and ML-IAP BIR using a fluorescence polarization assay. The most potent of these compounds, 19a and 33b, were found to have K(i)'s of 20-30 nM against ML-IAP and 50-60 nM against XIAP-BIR3.


Asunto(s)
Amidas/química , Amidas/farmacología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Péptidos/química , Tiazoles/química , Tiazoles/farmacología , Sitios de Unión , Biomimética , Cristalografía por Rayos X , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Modelos Moleculares , Péptidos/metabolismo
7.
ACS Med Chem Lett ; 11(10): 1829-1836, 2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33062160

RESUMEN

Herein we describe the discovery of A-1331852, a first-in-class orally active BCL-XL inhibitor that selectively and potently induces apoptosis in BCL-XL-dependent tumor cells. This molecule was generated by re-engineering our previously reported BCL-XL inhibitor A-1155463 using structure-based drug design. Key design elements included rigidification of the A-1155463 pharmacophore and introduction of sp3-rich moieties capable of generating highly productive interactions within the key P4 pocket of BCL-XL. A-1331852 has since been used as a critical tool molecule for further exploring BCL-2 family protein biology, while also representing an attractive entry into a drug discovery program.

8.
J Pharm Sci ; 108(6): 1934-1943, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30639736

RESUMEN

Solubilization of new chemical entities for toxicity assessment must use excipients that do not negatively impact drug pharmacokinetics and toxicology. In this study, we investigated the tolerability of a model freebase compound, GDC-0152, solubilized by pH adjustment with succinic acid and complexation with hydroxypropyl-ß-cyclodextrin (HP-ß-CD) to enable intravenous use. Solubility, critical micelle concentration, and association constant with HP-ß-CD were determined. Blood compatibility and potential for hemolysis were assessed in vitro. Local tolerability was assessed after intravenous and subcutaneous injections in rats. A pharmacokinetic study was conducted in rats after intravenous bolus administration. GDC-0152 exhibited pH-dependent solubility that was influenced by self-association. The presence of succinic acid increased solubility in a concentration-dependent manner. HP-ß-CD alone also increased solubility, but the extent of solubility enhancement was significantly lower than succinic acid alone. Inclusion of HP-ß-CD in the solution of GDC-0152 improved blood compatibility, reduced hemolytic potential by ∼20-fold in vitro, and increased the maximum tolerated dose to 80 mg/kg.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/farmacocinética , Ciclohexanos/toxicidad , Evaluación Preclínica de Medicamentos/métodos , Excipientes/farmacocinética , Pirroles/toxicidad , Pruebas de Toxicidad Aguda/métodos , 2-Hidroxipropil-beta-Ciclodextrina/administración & dosificación , Animales , Ciclohexanos/administración & dosificación , Ciclohexanos/farmacocinética , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Excipientes/administración & dosificación , Hemólisis/efectos de los fármacos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Masculino , Dosis Máxima Tolerada , Modelos Animales , Pirroles/administración & dosificación , Pirroles/farmacocinética , Ratas , Solubilidad
9.
J Med Chem ; 61(3): 989-1000, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29227683

RESUMEN

Antibody-drug conjugates (ADCs) have become an important therapeutic modality for oncology, with three approved by the FDA and over 60 others in clinical trials. Despite the progress, improvements in ADC therapeutic index are desired. Peptide-based ADC linkers that are cleaved by lysosomal proteases have shown sufficient stability in serum and effective payload-release in targeted cells. If the linker can be preferentially hydrolyzed by tumor-specific proteases, safety margin may improve. However, the use of peptide-based linkers limits our ability to modulate protease specificity. Here we report the structure-guided discovery of novel, nonpeptidic ADC linkers. We show that a cyclobutane-1,1-dicarboxamide-containing linker is hydrolyzed predominantly by cathepsin B while the valine-citrulline dipeptide linker is not. ADCs bearing the nonpeptidic linker are as efficacious and stable in vivo as those with the dipeptide linker. Our results strongly support the application of the peptidomimetic linker and present new opportunities for improving the selectivity of ADCs.


Asunto(s)
Catepsina B/metabolismo , Descubrimiento de Drogas , Inmunoconjugados/química , Inmunoconjugados/metabolismo , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Humanos , Espacio Intracelular/metabolismo , Especificidad por Sustrato
10.
FEBS J ; 284(10): 1555-1576, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28196299

RESUMEN

The reversible post-translational modification of proteins by ubiquitin and ubiquitin-like proteins regulates almost all cellular processes, by affecting protein degradation, localization, and complex formation. Deubiquitinases (DUBs) are proteases that remove ubiquitin modifications or cleave ubiquitin chains. Most DUBs are cysteine proteases, which makes them well suited for study by activity-based probes. These DUB probes report on deubiquitinase activity by reacting covalently with the active site in an enzyme-catalyzed manner. They have proven to be important tools to study DUB selectivity and proteolytic activity in different settings, to identify novel DUBs, and to characterize deubiquitinase inhibitors. Inspired by the efficacy of activity-based probes for DUBs, several groups have recently reported probes for the ubiquitin conjugation machinery (E1, E2, and E3 enzymes). Many of these enzymes, while not proteases, also posses active site cysteine residues and can be targeted by covalent probes. In this review, we will discuss how features of the probe (cysteine-reactive group, recognition element, and reporter tag) affect reactivity and suitability for certain experimental applications. We will also review the diverse applications of the current probes, and discuss the need for new probe types to study emerging aspects of ubiquitin biology.


Asunto(s)
Ubiquitina/metabolismo , Animales , Enzimas Desubicuitinizantes/metabolismo , Humanos , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteasas Ubiquitina-Específicas/metabolismo
11.
FEBS J ; 284(10): 1540-1554, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28107776

RESUMEN

Proteasomes are multisubunit protease complexes responsible for degrading most intracellular proteins. In addition to removing damaged proteins, they regulate many important cellular processes through the controlled degradation of transcription factors, cell cycle regulators, and enzymes. Eukaryotic proteasomes have three catalytic subunits, ß1, ß2, and ß5, that each has different substrate specificities. Additionally, although we know that diverse cell types express proteasome variants with distinct activity and specificity profiles, the functions of these different pools of proteasomes are not fully understood. Covalent inhibitors of the protease activity of the proteasome have been developed as drugs for hematological malignancies and are currently under investigation for other diseases. Therefore, there is a need for tools that allow direct monitoring of proteasome activity in live cells and tissues. Activity-based probes have proven valuable for biochemical and cell biological studies of the role of individual proteasome subunits, and for evaluating the efficacy and selectivity of proteasome inhibitors. These probes react covalently with the protease active sites, and contain a reporter tag to identify the probe-labeled proteasome subunits. This review will describe the development of broad-spectrum and subunit-specific proteasome activity-based probes, and discuss how these probes have contributed to our understanding of proteasome biology, and to the development of proteasome inhibitors.


Asunto(s)
Péptido Hidrolasas/metabolismo , Inhibidores de Proteasoma/farmacología , Proteolisis , Especificidad por Sustrato
12.
Chem Sci ; 8(1): 366-370, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28451181

RESUMEN

Disulfide bonds provide a bioactivatable connection with applications in imaging and therapy. The circulation stability and intracellular release of disulfides are problematically coupled in that increasing stability causes a corresponding decrease in cleavage and payload release. However, an antibody offers the potential for a reversible stabilization. We examined this by attaching a small molecule directly to engineered cysteines in an antibody. At certain sites this unhindered disulfide was stable in circulation yet cellular internalization and antibody catabolism generated a disulfide catabolite that was rapidly reduced. We demonstrated that this stable connection and facile release is applicable to a variety of payloads. The ability to reversibly stabilize a labile functional group with an antibody may offer a way to improve targeted probes and therapeutics.

13.
J Med Chem ; 60(23): 9490-9507, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29112410

RESUMEN

Three rationally designed pyrrolobenzodiazepine (PBD) drug-linkers have been synthesized via intermediate 19 for use in antibody-drug conjugates (ADCs). They lack a cleavable trigger in the linker and consist of a maleimide for cysteine antibody conjugation, a hydrophilic spacer, and either an alkyne (6), triazole (7), or piperazine (8) link to the PBD. In vitro IC50 values were 11-48 ng/mL in HER2 3+ SK-BR-3 and KPL-4 (7 inactive) for the anti-HER2 ADCs (HER2 0 MCF7, all inactive) and 0.10-1.73 µg/mL (7 inactive) in CD22 3+ BJAB and WSU-DLCL2 for anti-CD22 ADCs (CD22 0 Jurkat, all inactive at low doses). In vivo antitumor efficacy for the anti-HER2 ADCs in Founder 5 was observed with tumor stasis at 0.5-1 mg/kg, 1 mg/kg, and 3-6 mg/kg for 6, 8, and 7, respectively. Tumor stasis at 2 mg/kg was observed for anti-CD22 6 in WSU-DLCL2. In summary, noncleavable PBD-ADCs exhibit potent activity, particularly in HER2 models.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Benzodiazepinas/química , Benzodiazepinas/uso terapéutico , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Pirroles/química , Pirroles/uso terapéutico , Animales , Antineoplásicos/farmacología , Benzodiazepinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dimerización , Femenino , Humanos , Inmunoconjugados/farmacología , Ratones , Modelos Moleculares , Pirroles/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores
14.
Mol Cancer Ther ; 16(5): 871-878, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28223423

RESUMEN

A novel disulfide linker was designed to enable a direct connection between cytotoxic pyrrolobenzodiazepine (PBD) drugs and the cysteine on a targeting antibody for use in antibody-drug conjugates (ADCs). ADCs composed of a cysteine-engineered antibody were armed with a PBD using a self-immolative disulfide linker. Both the chemical linker and the antibody site were optimized for this new bioconjugation strategy to provide a highly stable and efficacious ADC. This novel disulfide ADC was compared with a conjugate containing the same PBD drug, but attached to the antibody via a peptide linker. Both ADCs had similar efficacy in mice bearing human tumor xenografts. Safety studies in rats revealed that the disulfide-linked ADC had a higher MTD than the peptide-linked ADC. Overall, these data suggest that the novel self-immolative disulfide linker represents a valuable way to construct ADCs with equivalent efficacy and improved safety. Mol Cancer Ther; 16(5); 871-8. ©2017 AACR.


Asunto(s)
Anticuerpos/administración & dosificación , Benzodiazepinas/administración & dosificación , Inmunoconjugados/administración & dosificación , Neoplasias/tratamiento farmacológico , Pirroles/administración & dosificación , Animales , Anticuerpos/química , Anticuerpos/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/inmunología , Benzodiazepinas/química , Benzodiazepinas/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Disulfuros/química , Disulfuros/inmunología , Humanos , Inmunoconjugados/química , Ratones , Neoplasias/inmunología , Neoplasias/patología , Pirroles/química , Pirroles/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nat Chem ; 8(12): 1112-1119, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27874860

RESUMEN

The reversible attachment of a small-molecule drug to a carrier for targeted delivery can improve pharmacokinetics and the therapeutic index. Previous studies have reported the delivery of molecules that contain primary and secondary amines via an amide or carbamate bond; however, the ability to employ tertiary-amine-containing bioactive molecules has been elusive. Here we describe a bioreversible linkage based on a quaternary ammonium that can be used to connect a broad array of tertiary and heteroaryl amines to a carrier protein. Using a concise, protecting-group-free synthesis we demonstrate the chemoselective modification of 12 complex molecules that contain a range of reactive functional groups. We also show the utility of this connection with both protease-cleavable and reductively cleavable antibody-drug conjugates that were effective and stable in vitro and in vivo. Studies with a tertiary-amine-containing antibiotic show that the resulting antibody-antibiotic conjugate provided appropriate stability and release characteristics and led to an unexpected improvement in activity over the conjugates previously connected via a carbamate.


Asunto(s)
Aminas/química , Anticuerpos Monoclonales/química , Portadores de Fármacos/química , Inmunoconjugados/metabolismo , Preparaciones Farmacéuticas/química , Antibacterianos/química , Antineoplásicos/química , Catepsinas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Estabilidad de Medicamentos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Inmunoconjugados/química , Inmunoconjugados/farmacología , Preparaciones Farmacéuticas/metabolismo , Compuestos de Amonio Cuaternario/química , Solubilidad
17.
Curr Top Med Chem ; 14(3): 319-25, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24283214

RESUMEN

Partnerships between industry and academia are becoming increasingly complex and relevant in the drive to discover innovative new medicines. We describe the structure of the collaboration between the University of California - San Francisco - Small Molecule Discovery Center (UCSF-SMDC) and Genentech to develop chemical matter that inhibits the activity of caspase-6. We focus on the scientific basis for the partnership and how the orientation- and transaction-related barriers were overcome. We describe the division of labor that allowed two groups to operate as a unified team to generate multiple chemical series with distinct mechanisms of action. The successful structure of the agreement serves as a model for future collaborations at both institutions.


Asunto(s)
Biocatálisis/efectos de los fármacos , Caspasa 6/metabolismo , Inhibidores de Caspasas/farmacología , Descubrimiento de Drogas/organización & administración , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Inhibidores de Caspasas/química , Industria Farmacéutica , Activación Enzimática/efectos de los fármacos , Humanos , Enfermedades Neurodegenerativas/metabolismo , San Francisco , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Universidades
18.
J Med Chem ; 57(19): 7890-9, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25191794

RESUMEN

Antibody-drug conjugates (ADCs) have a significant impact toward the treatment of cancer, as evidenced by the clinical activity of the recently approved ADCs, brentuximab vedotin for Hodgkin lymphoma and ado-trastuzumab emtansine (trastuzumab-MCC-DM1) for metastatic HER2+ breast cancer. DM1 is an analog of the natural product maytansine, a microtubule inhibitor that by itself has limited clinical activity and high systemic toxicity. However, by conjugation of DM1 to trastuzumab, the safety was improved and clinical activity was demonstrated. Here, we report that through chemical modification of the linker-drug and antibody engineering, the therapeutic activity of trastuzumab maytansinoid ADCs can be further improved. These improvements include eliminating DM1 release in the plasma and increasing the drug load by engineering four cysteine residues into the antibody. The chemical synthesis of highly stable linker-drugs and the modification of cysteine residues of engineered site-specific antibodies resulted in a homogeneous ADC with increased therapeutic activity compared to the clinically approved ADC, trastuzumab-MCC-DM1.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/síntesis química , Inmunoconjugados/farmacología , Maitansina/análogos & derivados , Ingeniería de Proteínas , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Trastuzumab
19.
ACS Med Chem Lett ; 5(6): 662-7, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24944740

RESUMEN

Because of the promise of BCL-2 antagonists in combating chronic lymphocytic leukemia (CLL) and non-Hodgkin's lymphoma (NHL), interest in additional selective antagonists of antiapoptotic proteins has grown. Beginning with a series of selective, potent BCL-XL antagonists containing an undesirable hydrazone functionality, in silico design and X-ray crystallography were utilized to develop alternative scaffolds that retained the selectivity and potency of the starting compounds.

20.
ACS Med Chem Lett ; 5(10): 1088-93, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25313317

RESUMEN

A-1155463, a highly potent and selective BCL-XL inhibitor, was discovered through nuclear magnetic resonance (NMR) fragment screening and structure-based design. This compound is substantially more potent against BCL-XL-dependent cell lines relative to our recently reported inhibitor, WEHI-539, while possessing none of its inherent pharmaceutical liabilities. A-1155463 caused a mechanism-based and reversible thrombocytopenia in mice and inhibited H146 small cell lung cancer xenograft tumor growth in vivo following multiple doses. A-1155463 thus represents an excellent tool molecule for studying BCL-XL biology as well as a productive lead structure for further optimization.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA