Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
MAbs ; 16(1): 2381891, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39041287

RESUMO

Novel engineered IL-2 agonists strive to increase the therapeutic window of aldesleukin (human IL-2) by increasing selectivity toward effector over regulatory T cells and reducing dose-limiting toxicities. Here we describe ANV419, an IL-2/anti-IL2 antibody fusion protein designed for selective IL-2 receptor ßγ (IL-2 Rßγ) activation by sterically hindering IL-2 from binding to IL-2 Rα. The fusion protein has an IL-2 connected to the light chain complementarity-determining region (CDR) domain of a humanized antibody that binds to IL-2 at the same epitope as IL-2 Rα. Optimization of the selectivity and pharmacological properties led to the selection of ANV419. ANV419 preferentially expands CD8+ T cells and natural killer (NK) cells over Tregs and can be safely administered at doses that elicit strong pharmacodynamic effects and efficacy in mouse tumor models. Its anti-tumor efficacy was enhanced when combined with programmed cell death protein 1 (PD-1) or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) checkpoint inhibitors. ANV419 also enhances the NK cell killing capacity and increases tumor growth inhibition when used alongside trastuzumab in a Her-2+ xenograft mouse model. In cynomolgus monkeys, the estimated half-life of ANV419 is 24 h, and doses that induced sustained expansion of effector cells were well tolerated without the severe toxicities typically observed with high-dose IL-2. These data support the clinical development of ANV419 in solid tumors and hematological malignancies as monotherapy and in combination with checkpoint inhibitors or agents that induce antibody-dependent cellular cytotoxicity. ANV419 is currently in Phase 1/2 clinical development and may provide cancer patients with a wider therapeutic window than aldesleukin.


Assuntos
Linfócitos T CD8-Positivos , Interleucina-2 , Células Matadoras Naturais , Proteínas Recombinantes de Fusão , Animais , Células Matadoras Naturais/imunologia , Humanos , Interleucina-2/imunologia , Linfócitos T CD8-Positivos/imunologia , Camundongos , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/genética , Imunoterapia/métodos , Macaca fascicularis , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto , Linhagem Celular Tumoral , Feminino
2.
Mol Cancer Ther ; 20(7): 1270-1282, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33879555

RESUMO

The cell surface glycoprotein P-cadherin is highly expressed in a number of malignancies, including those arising in the epithelium of the bladder, breast, esophagus, lung, and upper aerodigestive system. PCA062 is a P-cadherin specific antibody-drug conjugate that utilizes the clinically validated SMCC-DM1 linker payload to mediate potent cytotoxicity in cell lines expressing high levels of P-cadherin in vitro, while displaying no specific activity in P-cadherin-negative cell lines. High cell surface P-cadherin is necessary, but not sufficient, to mediate PCA062 cytotoxicity. In vivo, PCA062 demonstrated high serum stability and a potent ability to induce mitotic arrest. In addition, PCA062 was efficacious in clinically relevant models of P-cadherin-expressing cancers, including breast, esophageal, and head and neck. Preclinical non-human primate toxicology studies demonstrated a favorable safety profile that supports clinical development. Genome-wide CRISPR screens reveal that expression of the multidrug-resistant gene ABCC1 and the lysosomal transporter SLC46A3 differentially impact tumor cell sensitivity to PCA062. The preclinical data presented here suggest that PCA062 may have clinical value for treating patients with multiple cancer types including basal-like breast cancer.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Biomarcadores Tumorais , Caderinas/genética , Imunoconjugados/farmacologia , Neoplasias/genética , Sequência de Aminoácidos , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/farmacocinética , Sítios de Ligação , Caderinas/química , Caderinas/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Expressão Gênica , Humanos , Imunoconjugados/química , Imunoconjugados/farmacocinética , Imuno-Histoquímica , Macaca fascicularis , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Transporte Proteico , Ratos , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Proc Natl Acad Sci U S A ; 114(47): 12448-12453, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29109273

RESUMO

The TGF-ß family ligands myostatin, GDF11, and activins are negative regulators of skeletal muscle mass, which have been reported to primarily signal via the ActRIIB receptor on skeletal muscle and thereby induce muscle wasting described as cachexia. Use of a soluble ActRIIB-Fc "trap," to block myostatin pathway signaling in normal or cachectic mice leads to hypertrophy or prevention of muscle loss, perhaps suggesting that the ActRIIB receptor is primarily responsible for muscle growth regulation. Genetic evidence demonstrates however that both ActRIIB- and ActRIIA-deficient mice display a hypertrophic phenotype. Here, we describe the mode of action of bimagrumab (BYM338), as a human dual-specific anti-ActRIIA/ActRIIB antibody, at the molecular and cellular levels. As shown by X-ray analysis, bimagrumab binds to both ActRIIA and ActRIIB ligand binding domains in a competitive manner at the critical myostatin/activin binding site, hence preventing signal transduction through either ActRII. Myostatin and the activins are capable of binding to both ActRIIA and ActRIIB, with different affinities. However, blockade of either single receptor through the use of specific anti-ActRIIA or anti-ActRIIB antibodies achieves only a partial signaling blockade upon myostatin or activin A stimulation, and this leads to only a small increase in muscle mass. Complete neutralization and maximal anabolic response are achieved only by simultaneous blockade of both receptors. These findings demonstrate the importance of ActRIIA in addition to ActRIIB in mediating myostatin and activin signaling and highlight the need for blocking both receptors to achieve a strong functional benefit.


Assuntos
Receptores de Activinas Tipo II/antagonistas & inibidores , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Hipertrofia/induzido quimicamente , Músculo Esquelético/efeitos dos fármacos , Receptores de Activinas Tipo II/metabolismo , Ativinas/metabolismo , Animais , Anticorpos Bloqueadores/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Proteínas Morfogenéticas Ósseas/metabolismo , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Fatores de Diferenciação de Crescimento/metabolismo , Células HEK293 , Humanos , Hipertrofia/patologia , Masculino , Camundongos , Camundongos SCID , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miostatina/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Síndrome de Emaciação/tratamento farmacológico , Síndrome de Emaciação/patologia
4.
ChemMedChem ; 10(11): 1884-91, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26381451

RESUMO

Farnesyl pyrophosphate synthase (FPPS) is an established target for the treatment of bone diseases, but also shows promise as an anticancer and anti-infective drug target. Currently available anti-FPPS drugs are active-site-directed bisphosphonate inhibitors, the peculiar pharmacological profile of which is inadequate for therapeutic indications beyond bone diseases. The recent discovery of an allosteric binding site has paved the way toward the development of novel non-bisphosphonate FPPS inhibitors with broader therapeutic potential, notably as immunomodulators in oncology. Herein we report the discovery, by an integrated lead finding approach, of two new chemical classes of allosteric FPPS inhibitors that belong to the salicylic acid and quinoline chemotypes. We present their synthesis, biochemical and cellular activities, structure-activity relationships, and provide X-ray structures of several representative FPPS complexes. These novel allosteric FPPS inhibitors are devoid of any affinity for bone mineral and could serve as leads to evaluate their potential in none-bone diseases.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Geraniltranstransferase/antagonistas & inibidores , Quinolinas/farmacologia , Ácido Salicílico/farmacologia , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Geraniltranstransferase/metabolismo , Humanos , Estrutura Molecular , Quinolinas/síntese química , Quinolinas/química , Ácido Salicílico/síntese química , Ácido Salicílico/química , Relação Estrutura-Atividade
5.
J Biomol Screen ; 19(6): 917-27, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24554445

RESUMO

Bioactive small molecules are an invaluable source of therapeutics and chemical probes for exploring biological pathways. Yet, significant hurdles in drug discovery often come from lacking a comprehensive view of the target(s) for both early tool molecules and even late-stage drugs. To address this challenge, a method is provided that allows for assessing the interactions of small molecules with thousands of targets without any need to modify the small molecule of interest or attach any component to a surface. We describe size-exclusion chromatography for target identification (SEC-TID), a method for accurately and reproducibly detecting ligand-macromolecular interactions for small molecules targeting nucleic acid and several protein classes. We report the use of SEC-TID, with a library consisting of approximately 1000 purified proteins derived from the protein databank (PDB), to identify the efficacy targets tankyrase 1 and 2 for the Wnt inhibitor XAV939. In addition, we report novel interactions for the tumor-vascular disrupting agent vadimezan/ASA404 (interacting with farnesyl pyrophosphate synthase) and the diuretic mefruside (interacting with carbonic anhydrase XIII). We believe this method can dramatically enhance our understanding of the mechanism of action and potential liabilities for small molecules in drug discovery pipelines through comprehensive profiling of candidate druggable targets.


Assuntos
Cromatografia em Gel/métodos , Espectrometria de Massas/métodos , Proteínas/química , Proteômica/métodos , Animais , Calorimetria , Domínio Catalítico , Bovinos , Cromatografia Líquida , Cristalografia por Raios X , Bases de Dados de Proteínas , Descoberta de Drogas , Fluorometria , Humanos , Ligantes , Substâncias Macromoleculares , Espectroscopia de Ressonância Magnética , Ácidos Nucleicos/química , Bibliotecas de Moléculas Pequenas/química , Xantonas/química
6.
Nat Chem Biol ; 6(9): 660-6, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20711197

RESUMO

Bisphosphonates are potent inhibitors of farnesyl pyrophosphate synthase (FPPS) and are highly efficacious in the treatment of bone diseases such as osteoporosis, Paget's disease and tumor-induced osteolysis. In addition, the potential for direct antitumor effects has been postulated on the basis of in vitro and in vivo studies and has recently been demonstrated clinically in early breast cancer patients treated with the potent bisphosphonate zoledronic acid. However, the high affinity of bisphosphonates for bone mineral seems suboptimal for the direct treatment of soft-tissue tumors. Here we report the discovery of the first potent non-bisphosphonate FPPS inhibitors. These new inhibitors bind to a previously unknown allosteric site on FPPS, which was identified by fragment-based approaches using NMR and X-ray crystallography. This allosteric and druggable pocket allows the development of a new generation of FPPS inhibitors that are optimized for direct antitumor effects in soft tissue.


Assuntos
Difosfonatos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Geraniltranstransferase/antagonistas & inibidores , Regulação Alostérica , Sítio Alostérico , Osso e Ossos/química , Osso e Ossos/metabolismo , Cristalografia por Raios X , Difosfonatos/análise , Difosfonatos/química , Difosfonatos/metabolismo , Difosfonatos/farmacologia , Inibidores Enzimáticos/química , Geraniltranstransferase/metabolismo , Humanos , Imidazóis/análise , Imidazóis/química , Imidazóis/farmacologia , Espectroscopia de Ressonância Magnética , Neoplasias de Tecidos Moles/tratamento farmacológico , Ácido Zoledrônico
8.
J Med Chem ; 48(16): 5175-90, 2005 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16078837

RESUMO

Molecular modeling based on the X-ray crystal structure of the Tang-Ghosh heptapeptide inhibitor 1 (OM99-2) of BACE led to the design and synthesis of a series of constrained P(1)' analogues. A cyclopentane ring was incorporated in 1 spanning the P(1)' Ala methyl group and the adjacent methylene carbon atom of the chain. Progressive truncation at the P(2)'-P(4)' sites led to a potent truncated analogue 5 with good selectivity over Cathepsin D. Using the same backbone replacement concept, a series of cyclopentane, cyclopentanone, tetrahydrofuran, pyrrolidine, and pyrrolidinone analogues were synthesized with considerable variation at the P and P' sites. The cyclopentanone and 2-pyrrolidinone analogues 45 and 57 showed low nM BACE inhibition. X-ray cocrystal structures of two analogues 5 and 45 revealed excellent convergence with the original inhibitor 1 structure while providing new insights into other interactions which could be exploited for future modifications.


Assuntos
Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ciclopentanos/síntese química , Furanos/síntese química , Peptídeos/química , Pirrolidinas/síntese química , Secretases da Proteína Precursora do Amiloide , Ácido Aspártico Endopeptidases/química , Sítios de Ligação , Catepsina D/antagonistas & inibidores , Cristalografia por Raios X , Ciclopentanos/química , Endopeptidases , Furanos/química , Humanos , Modelos Moleculares , Mimetismo Molecular , Pirrolidinas/química , Pirrolidinonas/síntese química , Pirrolidinonas/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA