Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33148666

RESUMO

PURPOSE: Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN: We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS: Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS: We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.


Assuntos
Gemtuzumab/uso terapêutico , Imunoconjugados/uso terapêutico , Subunidade alfa de Receptor de Interleucina-3/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/imunologia , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/uso terapêutico , Área Sob a Curva , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Gemtuzumab/imunologia , Gemtuzumab/farmacocinética , Células HL-60 , Humanos , Imunoconjugados/imunologia , Imunoconjugados/farmacocinética , Subunidade alfa de Receptor de Interleucina-3/imunologia , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/metabolismo , Macaca fascicularis , Camundongos , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
Mol Cancer Ther ; 19(10): 2068-2078, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32747418

RESUMO

The approval of ado-trastuzumab emtansine (T-DM1) in HER2+ metastatic breast cancer validated HER2 as a target for HER2-specific antibody-drug conjugates (ADC). Despite its demonstrated clinical efficacy, certain inherent properties within T-DM1 hamper this compound from achieving the full potential of targeting HER2-expressing solid tumors with ADCs. Here, we detail the discovery of PF-06804103, an anti-HER2 ADC designed to have a widened therapeutic window compared with T-DM1. We utilized an empirical conjugation site screening campaign to identify the engineered ĸkK183C and K290C residues as those that maximized in vivo ADC stability, efficacy, and safety for a four drug-antibody ratio (DAR) ADC with this linker-payload combination. PF-06804103 incorporates the following novel design elements: (i) a new auristatin payload with optimized pharmacodynamic properties, (ii) a cleavable linker for optimized payload release and enhanced antitumor efficacy, and (iii) an engineered cysteine site-specific conjugation approach that overcomes the traditional safety liabilities of conventional conjugates and generates a homogenous drug product with a DAR of 4. PF-06804103 shows (i) an enhanced efficacy against low HER2-expressing breast, gastric, and lung tumor models, (ii) overcomes in vitro- and in vivo-acquired T-DM1 resistance, and (iii) an improved safety profile by enhancing ADC stability, pharmacokinetic parameters, and reducing off-target toxicities. Herein, we showcase our platform approach in optimizing ADC design, resulting in the generation of the anti-HER2 ADC, PF-06804103. The design elements of identifying novel sites of conjugation employed in this study serve as a platform for developing optimized ADCs against other tumor-specific targets.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Imunoconjugados/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Gástricas/tratamento farmacológico , Animais , Neoplasias da Mama/patologia , Feminino , Humanos , Imunoconjugados/farmacologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Neoplasias Gástricas/patologia
3.
Mol Cancer Ther ; 14(4): 952-63, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25646013

RESUMO

Antibody-drug conjugates (ADC) are emerging as clinically effective therapy. We hypothesized that cancers treated with ADCs would acquire resistance mechanisms unique to immunoconjugate therapy and that changing ADC components may overcome resistance. Breast cancer cell lines were exposed to multiple cycles of anti-Her2 trastuzumab-maytansinoid ADC (TM-ADC) at IC80 concentrations followed by recovery. The resistant cells, 361-TM and JIMT1-TM, were characterized by cytotoxicity, proteomic, transcriptional, and other profiling. Approximately 250-fold resistance to TM-ADC developed in 361-TM cells, and cross-resistance was observed to other non-cleavable-linked ADCs. Strikingly, these 361-TM cells retained sensitivity to ADCs containing cleavable mcValCitPABC-linked auristatins. In JIMT1-TM cells, 16-fold resistance to TM-ADC developed, with cross-resistance to other trastuzumab-ADCs. Both 361-TM and JIMT1-TM cells showed minimal resistance to unconjugated mertansine (DM1) and other chemotherapeutics. Proteomics and immunoblots detected increased ABCC1 (MRP1) drug efflux protein in 361-TM cells, and decreased Her2 (ErbB2) in JIMT1-TM cells. Proteomics also showed alterations in various pathways upon chronic exposure to the drug in both cell models. Tumors derived from 361-TM cells grew in mice and were refractory to TM-ADC compared with parental cells. Hence, acquired resistance to trastuzumab-maytansinoid ADC was generated in cultured cancer cells by chronic drug treatment, and either increased ABCC1 protein or reduced Her2 antigen were primary mediators of resistance. These ADC-resistant cell models retain sensitivity to other ADCs or standard-of-care chemotherapeutics, suggesting that alternate therapies may overcome acquired ADC resistance. Mol Cancer Ther; 14(4); 952-63. ©2015 AACR.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Imunoconjugados/farmacologia , Trastuzumab/farmacologia , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Humanos , Imunoconjugados/administração & dosagem , Concentração Inibidora 50 , Camundongos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Transporte Proteico , Proteoma , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Transdução de Sinais , Transcriptoma , Trastuzumab/administração & dosagem , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nat Mater ; 13(6): 653-61, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24681647

RESUMO

Inhibitors of matrix metalloproteinases (MMPs) have been extensively explored to treat pathologies where excessive MMP activity contributes to adverse tissue remodelling. Although MMP inhibition remains a relevant therapeutic target, MMP inhibitors have not translated to clinical application owing to the dose-limiting side effects following systemic administration of the drugs. Here, we describe the synthesis of a polysaccharide-based hydrogel that can be locally injected into tissues and releases a recombinant tissue inhibitor of MMPs (rTIMP-3) in response to MMP activity. Specifically, rTIMP-3 is sequestered in the hydrogels through electrostatic interactions and is released as crosslinks are degraded by active MMPs. Targeted delivery of the hydrogel/rTIMP-3 construct to regions of MMP overexpression following a myocardial infarction significantly reduced MMP activity and attenuated adverse left ventricular remodelling in a porcine model of myocardial infarction. Our findings demonstrate that local, on-demand MMP inhibition is achievable through the use of an injectable and bioresponsive hydrogel.


Assuntos
Hidrogéis/farmacologia , Inibidores de Metaloproteinases de Matriz/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Inibidor Tecidual de Metaloproteinase-3/farmacologia , Remodelação Ventricular/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Humanos , Hidrogéis/química , Inibidores de Metaloproteinases de Matriz/química , Metaloproteinases da Matriz/metabolismo , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Suínos , Inibidor Tecidual de Metaloproteinase-3/química
5.
Biomaterials ; 33(7): 2145-53, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22177842

RESUMO

Injected therapeutics, such as cells or biological molecules, may have enhanced efficiency when delivered within a scaffold carrier. Here, we describe a dual-component Dock-and-Lock (DnL) self-assembly mechanism that can be used to construct shear-thinning, self-healing, and injectable hydrogels. One component is derived from the RIIα subunit of cAMP-dependent kinase A and is engineered as a telechelic protein with end groups that dimerize (docking step). The second component is derived from the anchoring domain of A-kinase anchoring protein (AD) and is attached to multi-arm crosslinker polymers and binds to the docked proteins (locking step). When mixed, these two DnL components form robust physical hydrogels instantaneously and under physiological conditions. Mechanical properties and erosion rates of DnL gels can be tuned through the AD peptide sequence, the concentration and ratio of each component, and the number of peptides on the cross-linking polymer. DnL gels immediately self-recover after deformation, are resistant to yield at strains as high as 400%, and completely self-heal irrespective of prior mechanical disruption. Mesenchymal stem cells mixed in DnL gels and injected through a fine needle remain highly viable (>90%) during the encapsulation and delivery process, and encapsulated large molecules are released with profiles that correspond to gel erosion. Thus, we have used molecular engineering strategies to develop cytocompatible and injectable hydrogels that have the potential to support cell and drug therapies.


Assuntos
Hidrogéis/química , Injeções , Proteínas de Ancoragem à Quinase A/química , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Sequência de Aminoácidos , Materiais Biocompatíveis/química , Terapia Baseada em Transplante de Células e Tecidos/métodos , Subunidade RIIalfa da Proteína Quinase Dependente de AMP Cíclico/química , Subunidade RIIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Composição de Medicamentos , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Dados de Sequência Molecular , Engenharia de Proteínas/métodos , Reologia
7.
J Polym Sci A Polym Chem ; 1(8): 1160-1170, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21637725

RESUMO

Elastomeric proteins are characterized by their large extensibility before rupture, reversible deformation without loss of energy, and high resilience upon stretching. Motivated by their unique mechanical properties, there has been tremendous research in understanding and manipulating elastomeric polypeptides, with most work conducted on the elastins but more recent work on an expanded set of polypeptide elastomers. Facilitated by biosynthetic strategies, it has been possible to manipulate the physical properties, conformation, and mechanical properties of these materials. Detailed understanding of the roles and organization of the natural structural proteins has permitted the design of elastomeric materials with engineered properties, and has thus expanded the scope of applications from elucidation of the mechanisms of elasticity to the development of advanced drug delivery systems and tissue engineering substrates.

8.
Soft Matter ; 5(18): 3412-3416, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20543970

RESUMO

The production of complex, yet well defined materials offers many opportunities in regenerative medicine, in which the mechanical and biological properties of the matrix must meet stringent requirements. Here we report the recombinant production of modular polypeptidic materials, based on the highly resilient protein resilin, which are equipped with multiple biologically active domains. The recombinant materials exhibit useful mechanical and cell adhesion behavior.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA