Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Rep Med ; 2(5): 100279, 2021 05 18.
Article de Anglais | MEDLINE | ID: mdl-34095881

RÉSUMÉ

Aberrant NOTCH3 signaling and overexpression is oncogenic, associated with cancer stem cells and drug resistance, yet therapeutic targeting remains elusive. Here, we develop NOTCH3-targeted antibody drug conjugates (NOTCH3-ADCs) by bioconjugation of an auristatin microtubule inhibitor through a protease cleavable linker to two antibodies with differential abilities to inhibit signaling. The signaling inhibitory antibody rapidly induces ligand-independent receptor clustering and internalization through both caveolin and clathrin-mediated pathways. The non-inhibitory antibody also efficiently endocytoses via clathrin without inducing receptor clustering but with slower lysosomal co-localization kinetics. In addition, DLL4 ligand binding to the NOTCH3 receptor mediates transendocytosis of NOTCH3-ADCs into ligand-expressing cells. NOTCH3-ADCs internalize into receptor and ligand cells independent of signaling and induce cell death in both cell types representing an atypical mechanism of ADC cytotoxicity. Treatment of xenografts with NOTCH3-ADCs leads to sustained tumor regressions, outperforms standard-of-care chemotherapy, and allows targeting of tumors that overexpress NOTCH3 independent of signaling inhibition.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Immunoconjugués/pharmacologie , Récepteur Notch3/métabolisme , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Humains , Immunoconjugués/métabolisme , Oncogènes/effets des médicaments et des substances chimiques , Récepteur Notch3/immunologie , Récepteurs Notch/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Article de Anglais | MEDLINE | ID: mdl-33148666

RÉSUMÉ

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.


Sujet(s)
Gemtuzumab/usage thérapeutique , Immunoconjugués/usage thérapeutique , Sous-unité alpha du récepteur à l'interleukine-3/antagonistes et inhibiteurs , Leucémie aigüe myéloïde/traitement médicamenteux , Lectine-3 de type Ig liant l'acide sialique/antagonistes et inhibiteurs , Animaux , Antinéoplasiques immunologiques/immunologie , Antinéoplasiques immunologiques/pharmacocinétique , Antinéoplasiques immunologiques/usage thérapeutique , Aire sous la courbe , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Gemtuzumab/immunologie , Gemtuzumab/pharmacocinétique , Cellules HL-60 , Humains , Immunoconjugués/immunologie , Immunoconjugués/pharmacocinétique , Sous-unité alpha du récepteur à l'interleukine-3/immunologie , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/métabolisme , Macaca fascicularis , Souris , Lectine-3 de type Ig liant l'acide sialique/immunologie , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
3.
Mol Cancer Ther ; 19(10): 2068-2078, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32747418

RÉSUMÉ

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.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Immunoconjugués/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs de l'estomac/traitement médicamenteux , Animaux , Tumeurs du sein/anatomopathologie , Femelle , Humains , Immunoconjugués/pharmacologie , Tumeurs du poumon/anatomopathologie , Souris , Souris nude , Tumeurs de l'estomac/anatomopathologie
4.
Sci Transl Med ; 9(372)2017 01 11.
Article de Anglais | MEDLINE | ID: mdl-28077676

RÉSUMÉ

Disease relapse after treatment is common in triple-negative breast cancer (TNBC), ovarian cancer (OVCA), and non-small cell lung cancer (NSCLC). Therapies that target tumor-initiating cells (TICs) should improve patient survival by eliminating the cells that can drive tumor recurrence and metastasis. We demonstrate that protein tyrosine kinase 7 (PTK7), a highly conserved but catalytically inactive receptor tyrosine kinase in the Wnt signaling pathway, is enriched on TICs in low-passage TNBC, OVCA, and NSCLC patient-derived xenografts (PDXs). To deliver a potent anticancer drug to PTK7-expressing TICs, we generated a targeted antibody-drug conjugate (ADC) composed of a humanized anti-PTK7 monoclonal antibody, a cleavable valine-citrulline-based linker, and Aur0101, an auristatin microtubule inhibitor. The PTK7-targeted ADC induced sustained tumor regressions and outperformed standard-of-care chemotherapy. Moreover, the ADC specifically reduced the frequency of TICs, as determined by serial transplantation experiments. In addition to reducing the TIC frequency, the PTK7-targeted ADC may have additional antitumor mechanisms of action, including the inhibition of angiogenesis and the stimulation of immune cells. Together, these preclinical data demonstrate the potential for the PTK7-targeted ADC to improve the long-term survival of cancer patients.


Sujet(s)
Anticorps/usage thérapeutique , Molécules d'adhérence cellulaire/composition chimique , Immunoconjugués/usage thérapeutique , Cellules souches tumorales/effets des médicaments et des substances chimiques , Récepteurs à activité tyrosine kinase/composition chimique , Aminobenzoates/usage thérapeutique , Animaux , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/thérapie , Molécules d'adhérence cellulaire/immunologie , Lignée cellulaire tumorale , Essais cliniques comme sujet , Femelle , Humains , Immunothérapie/méthodes , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie , Macaca fascicularis , Souris , Souris de lignée NOD , Souris SCID , Microtubules/composition chimique , Récidive tumorale locale/traitement médicamenteux , Oligopeptides/usage thérapeutique , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/thérapie , Récepteurs à activité tyrosine kinase/immunologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/thérapie , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Bioconjug Chem ; 27(7): 1645-54, 2016 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-27206324

RÉSUMÉ

Antibody-drug conjugates (ADC) are currently an active area of research, focused primarily on oncology therapeutics, but also to a limited extent on other areas such as infectious disease. The success of this type of targeted drug delivery is dependent upon many factors, one of which is the performance of the linker in releasing an active drug moiety under the appropriate conditions. As a tool in the development of linker/payload chemistry, we have developed an in vitro method for the identification of payload species released from ADCs in the presence of lysosomal enzymes. This method utilizes commercially available human liver S9 fraction as the source of these enzymes, and this has certain advantages over lysosomal fractions or purified enzymes. This article describes the characterization and performance of this assay with multiple ADCs composed of known and novel linkers and payloads. Additionally, we report the observation of incomplete degradation of mAb protein chains by lysosomal enzymes in vitro, believed to be the first report of this phenomenon involving an ADC therapeutic.


Sujet(s)
Vecteurs de médicaments/composition chimique , Libération de médicament , Immunoconjugués/composition chimique , Animaux , Cathepsine B/métabolisme , Lignée cellulaire tumorale , Vecteurs de médicaments/métabolisme , Humains , Foie/cytologie , Lysosomes/enzymologie , Spectrométrie de masse , Souris , Rats
6.
Mol Cancer Ther ; 14(4): 952-63, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25646013

RÉSUMÉ

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.


Sujet(s)
Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques , Immunoconjugués/pharmacologie , Trastuzumab/pharmacologie , Animaux , Antigènes de surface/génétique , Antigènes de surface/métabolisme , Antinéoplasiques/administration et posologie , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Femelle , Analyse de profil d'expression de gènes , Humains , Immunoconjugués/administration et posologie , Concentration inhibitrice 50 , Souris , Protéines associées à la multirésistance aux médicaments/génétique , Protéines associées à la multirésistance aux médicaments/métabolisme , Transport des protéines , Protéome , Récepteur ErbB-2/antagonistes et inhibiteurs , Récepteur ErbB-2/métabolisme , Transduction du signal , Transcriptome , Trastuzumab/administration et posologie , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Bioconjug Chem ; 25(10): 1871-80, 2014 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-25216346

RÉSUMÉ

The stability of the connection between the antibody and the toxin can have a profound impact on ADC safety and efficacy. There has been increasing evidence in recent years that maleimide-based ADCs are prone to payload loss via a retro-Michael type reaction. Herein, we report a mild method for the hydrolysis of the succinimide-thioether ring which results in a "ring-opened" linker. ADCs containing this hydrolyzed succinimide linker show equivalent cytotoxicity, improved in vitro stability, improved PK exposure, and improved efficacy as compared to their nonhydrolyzed counterparts. This method offers a simple way to improve the stability, exposure, and efficacy of maleimide-based ADCs.


Sujet(s)
Immunotoxines/composition chimique , Succinimides/composition chimique , Sulfures/composition chimique , Animaux , Lignée cellulaire tumorale , Stabilité de médicament , Humains , Hydrolyse , Immunotoxines/sang , Immunotoxines/usage thérapeutique , Souris , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Stabilité protéique
8.
Nat Mater ; 13(6): 653-61, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24681647

RÉSUMÉ

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.


Sujet(s)
Hydrogels/pharmacologie , Inhibiteurs de métalloprotéinases matricielles/pharmacologie , Infarctus du myocarde/traitement médicamenteux , Inhibiteur tissulaire de métalloprotéinase-3/pharmacologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Animaux , Modèles animaux de maladie humaine , Humains , Hydrogels/composition chimique , Inhibiteurs de métalloprotéinases matricielles/composition chimique , Matrix metalloproteinases/métabolisme , Infarctus du myocarde/enzymologie , Infarctus du myocarde/métabolisme , Protéines recombinantes/composition chimique , Protéines recombinantes/pharmacologie , Suidae , Inhibiteur tissulaire de métalloprotéinase-3/composition chimique
9.
Biomaterials ; 33(7): 2145-53, 2012 Mar.
Article de Anglais | MEDLINE | ID: mdl-22177842

RÉSUMÉ

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.


Sujet(s)
Hydrogels/composition chimique , Injections , Protéines d'ancrage aux protéines kinases A/composition chimique , Protéines d'ancrage aux protéines kinases A/génétique , Protéines d'ancrage aux protéines kinases A/métabolisme , Séquence d'acides aminés , Matériaux biocompatibles/composition chimique , Thérapie cellulaire et tissulaire/méthodes , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/composition chimique , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/génétique , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/métabolisme , Préparation de médicament , Test de matériaux , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/physiologie , Données de séquences moléculaires , Ingénierie des protéines/méthodes , Rhéologie
11.
J Polym Sci A Polym Chem ; 1(8): 1160-1170, 2010 Oct.
Article de Anglais | MEDLINE | ID: mdl-21637725

RÉSUMÉ

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.

12.
Soft Matter ; 5(18): 3412-3416, 2009.
Article de Anglais | MEDLINE | ID: mdl-20543970

RÉSUMÉ

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.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...