Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Bioorg Med Chem Lett ; 27(16): 3647-3652, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28720505

RESUMEN

Bispecific antibodies (BsAbs) are designed to engage two antigens simultaneously, thus, effectively expanding the ability of antibody-based therapeutics to target multiple pathways within the same cell, engage two separate soluble antigens, bind the same antigen with distinct paratopes, or crosslink two different cell types. Many recombinant BsAb formats have emerged, however, expression and purification of such constructs can often be challenging. To this end, we have developed a chemical strategy for generating BsAbs using native IgG2 architecture. Full-length antibodies can be conjugated via disulfide bridging with linkers bearing orthogonal groups to produce BsAbs. We report that an αHER2/EGFR BsAb was successfully generated by this approach and retained the ability to bind both antigens with no significant loss of potency.


Asunto(s)
Anticuerpos Biespecíficos/química , Disulfuros/química , Inmunoglobulina G/inmunología , Anticuerpos Biespecíficos/inmunología , Reacciones Antígeno-Anticuerpo , Sitios de Unión de Anticuerpos , Línea Celular Tumoral , Química Clic , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Humanos , Células MCF-7 , Microscopía Fluorescente , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo
2.
Bioorg Med Chem Lett ; 27(24): 5490-5495, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29126850

RESUMEN

Bioconjugate formats provide alternative strategies for antigen targeting with bispecific antibodies. Here, PSMA-targeted Fab conjugates were generated using different bispecific formats. Interchain disulfide bridging of an αCD3 Fab enabled installation of either the PSMA-targeting small molecule DUPA (SynFab) or the attachment of an αPSMA Fab (BisFab) by covalent linkage. Optimization of the reducing conditions was critical for selective interchain disulfide reduction and good bioconjugate yield. Activity of αPSMA/CD3 Fab conjugates was tested by in vitro cytotoxicity assays using prostate cancer cell lines. Both bispecific formats demonstrated excellent potency and antigen selectivity.


Asunto(s)
Anticuerpos Biespecíficos/química , Antígenos de Superficie/inmunología , Glutamato Carboxipeptidasa II/inmunología , Fragmentos Fab de Inmunoglobulinas/química , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Complejo CD3/inmunología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Química Clic , Disulfuros/química , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/farmacología , Leucocitos Mononucleares/citología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
3.
Cancer Res Commun ; 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39292169

RESUMEN

Treatment for patients with Multiple Myeloma (MM) has experienced rapid development and improvement in recent years, however, patients continue to experience relapse and MM remains largely incurable. B cell maturation antigen (BCMA) has been widely recognized as a promising target for treatment of MM due to its exclusive expression in B cell linage cells and its critical role in the growth and survival of malignant plasma cells. Here, we introduce STI-8811, a BCMA-targeting antibody drug conjugate linked to an auristatin-derived duostatin payload via an enzymatically cleavable peptide linker, using our proprietary C-lock technology. STI-8811 exhibits target specific binding activity and rapid internalization, leading to G2/M cell cycle arrest, caspase 3/7 activation and apoptosis in BCMA-expressing tumor cells in vitro. Soluble BCMA (sBCMA) is shed by MM cells into the blood and increases with disease progression, competing for ADC binding and reducing its efficacy. We report enhanced cytotoxic activity in the presence of high levels of sBCMA compared to a belantamab mafodotin biosimilar (J6M0-mcMMAF). STI-8811 demonstrated greater in vivo activity than J6M0-mcMMAF in solid and disseminated multiple myeloma models, including tumor models with low BCMA expression and/or in large solid tumors representing soft tissue plasmacytomas. In Cynomolgus monkeys, STI-8811 was well tolerated, with toxicities consistent with other BCMA targeting ADCs with auristatin payloads in clinical studies. STI-8811 has the potential to outperform current clinical candidates with lower toxicity and higher activity under conditions found in patients with advanced disease.

4.
Med ; 5(1): 42-61.e23, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38181791

RESUMEN

BACKGROUND: Oral antiviral drugs with improved antiviral potency and safety are needed to address current challenges in clinical practice for treatment of COVID-19, including the risks of rebound, drug-drug interactions, and emerging resistance. METHODS: Olgotrelvir (STI-1558) is designed as a next-generation antiviral targeting the SARS-CoV-2 main protease (Mpro), an essential enzyme for SARS-CoV-2 replication, and human cathepsin L (CTSL), a key enzyme for SARS-CoV-2 entry into host cells. FINDINGS: Olgotrelvir is a highly bioavailable oral prodrug that is converted in plasma to its active form, AC1115. The dual mechanism of action of olgotrelvir and AC1115 was confirmed by enzyme activity inhibition assays and co-crystal structures of AC1115 with SARS-CoV-2 Mpro and human CTSL. AC1115 displayed antiviral activity by inhibiting replication of all tested SARS-CoV-2 variants in cell culture systems. Olgotrelvir also inhibited viral entry into cells using SARS-CoV-2 Spike-mediated pseudotypes by inhibition of host CTSL. In the K18-hACE2 transgenic mouse model of SARS-CoV-2-mediated disease, olgotrelvir significantly reduced the virus load in the lungs, prevented body weight loss, and reduced cytokine release and lung pathologies. Olgotrelvir demonstrated potent activity against the nirmatrelvir-resistant Mpro E166 mutants. Olgotrelvir showed enhanced oral bioavailability in animal models and in humans with significant plasma exposure without ritonavir. In phase I studies (ClinicalTrials.gov: NCT05364840 and NCT05523739), olgotrelvir demonstrated a favorable safety profile and antiviral activity. CONCLUSIONS: Olgotrelvir is an oral inhibitor targeting Mpro and CTSL with high antiviral activity and plasma exposure and is a standalone treatment candidate for COVID-19. FUNDING: Funded by Sorrento Therapeutics.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Inhibidores de Proteasa de Coronavirus , SARS-CoV-2 , Animales , Humanos , Ratones , Antivirales/farmacología , Antivirales/uso terapéutico , Catepsina L/antagonistas & inhibidores , COVID-19/prevención & control , Modelos Animales de Enfermedad , Ratones Transgénicos , Inhibidores de Proteasa de Coronavirus/química , Inhibidores de Proteasa de Coronavirus/farmacología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Tratamiento Farmacológico de COVID-19/métodos
5.
Expert Opin Biol Ther ; 23(11): 1137-1149, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38078403

RESUMEN

BACKGROUND: Solid tumors are becoming prevalent affecting both old and young populations. Numerous solid tumors are associated with high cMET expression. The complexity of solid tumors combined with the highly interconnected nature of the cMET/HGF pathway with other cellular pathways make the pursuit of finding an effective treatment extremely challenging. The current standard of care for these malignancies is mostly small molecule-based chemotherapy. Antibody-based therapeutics as well as antibody drug conjugates are promising emerging classes against cMET-overexpressing solid tumors. RESEARCH DESIGN AND METHODS: In this study, we described the design, synthesis, in vitro and in vivo characterization of cMET-targeting Fab drug conjugates (FDCs) as an alternative therapeutic strategy. The format is comprised of a Fab conjugated to a potent cytotoxic drug via a cleavable linker employing lysine-based and cysteine-based conjugation chemistries. RESULTS: We found that the FDCs have potent anti-tumor efficacies in cancer cells with elevated overexpression of cMET. Moreover, they demonstrated a remarkable anti-tumor effect in a human gastric xenograft mouse model. CONCLUSIONS: The FDC format has the potential to overcome some of the challenges presented by the other classes of therapeutics. This study highlights the promise of antibody fragment-based drug conjugate formats for the treatment of solid tumors.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Neoplasias , Humanos , Animales , Ratones , Inmunoconjugados/uso terapéutico , Proteínas Proto-Oncogénicas c-met/metabolismo , Neoplasias/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Anticuerpos , Línea Celular Tumoral
6.
Front Oncol ; 12: 884196, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35664753

RESUMEN

Therapeutic blockade of the CD47/SIRPα axis by small molecules or monoclonal antibodies (mAbs) is a proven strategy to enhance macrophages-mediated anti-tumor activity. However, this strategy has been hampered by elevated on-target toxicities and rapid clearance due to the extensive CD47 expression on normal cells ("antigen sink") such as red blood cells (RBCs). To address these hurdles, we report on the development of STI-6643, an affinity-engineered fully human anti-CD47 IgG4 antibody with negligible binding to normal cells. STI-6643 exhibited no hemagglutination activity on human RBCs at concentrations up to 300 µg/mL yet specifically blocked the CD47/SIPRα interaction. Of particular interest, STI-6643 preserved T cell functionality in vitro and showed significantly lower immune cell depletion in vivo in contrast to three previously published competitor reference anti-CD47 clones Hu5F9, AO-176 and 13H3. In cynomolgus monkeys, STI-6643 was well-tolerated at the highest dose tested (300 mg/kg/week) and provided favorable clinical safety margins. Finally, STI-6643 displayed comparable anti-tumor activity to the high-affinity reference clone Hu5F9 in a RAJI-Fluc xenograft tumor model as monotherapy or in combination with anti-CD20 (rituximab) or anti-CD38 (daratumumab) mAbs. These data suggest that STI-6643 possesses the characteristics of an effective therapeutic candidate given its potent anti-tumor activity and low toxicity profile.

7.
Sci Rep ; 12(1): 15517, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36109550

RESUMEN

Coronavirus disease 2019 (COVID-19) continues to significantly impact the global population, thus countermeasure platforms that enable rapid development of therapeutics against variants of SARS-CoV-2 are essential. We report use of a phage display human antibody library approach to rapidly identify neutralizing antibodies (nAbs) against SARS-CoV-2. We demonstrate the binding and neutralization capability of two nAbs, STI-2020 and STI-5041, against the SARS-CoV-2 WA-1 strain as well as the Alpha and Beta variants. STI-2020 and STI-5041 were protective when administered intravenously or intranasally in the golden (Syrian) hamster model of COVID-19 challenged with the WA-1 strain or Beta variant. The ability to administer nAbs intravenously and intranasally may have important therapeutic implications and Phase 1 healthy subjects clinical trials are ongoing.


Asunto(s)
COVID-19 , Animales , Anticuerpos Monoclonales , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Cricetinae , Humanos , Mesocricetus , Pruebas de Neutralización , SARS-CoV-2
8.
Med ; 3(10): 705-721.e11, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36044897

RESUMEN

BACKGROUND: The continual emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern, in particular the newly emerged Omicron (B.1.1.529) variant and its BA.X lineages, has rendered ineffective a number of previously FDA emergency use authorized SARS-CoV-2 neutralizing antibody therapies. Furthermore, those approved antibodies with neutralizing activity against Omicron BA.1 are reportedly ineffective against the subset of Omicron subvariants that contain a R346K substitution, BA.1.1, and the more recently emergent BA.2, demonstrating the continued need for discovery and characterization of candidate therapeutic antibodies with the breadth and potency of neutralizing activity required to treat newly diagnosed COVID-19 linked to recently emerged variants of concern. METHODS: Following a campaign of antibody discovery based on the vaccination of Harbor H2L2 mice with defined SARS-CoV-2 spike domains, we have characterized the activity of a large collection of spike-binding antibodies and identified a lead neutralizing human IgG1 LALA antibody, STI-9167. FINDINGS: STI-9167 has potent, broad-spectrum neutralizing activity against the current SARS-COV-2 variants of concern and retained activity against each of the tested Omicron subvariants in both pseudotype and live virus neutralization assays. Furthermore, STI-9167 nAb administered intranasally or intravenously provided protection against weight loss and reduced virus lung titers to levels below the limit of quantitation in Omicron-infected K18-hACE2 transgenic mice. CONCLUSIONS: With this established activity profile, a cGMP cell line has been developed and used to produce cGMP drug product intended for intravenous or intranasal use in human clinical trials. FUNDING: Funded by CRIPT (no. 75N93021R00014), DARPA (HR0011-19-2-0020), and NCI Seronet (U54CA260560).


Asunto(s)
Anticuerpos Neutralizantes , Tratamiento Farmacológico de COVID-19 , Administración Intranasal , Animales , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Humanos , Inmunoglobulina G , Glicoproteínas de Membrana , Ratones , Pruebas de Neutralización , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Proteínas del Envoltorio Viral
9.
Antiviral Res ; 195: 105185, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34634289

RESUMEN

Monoclonal antibodies (mAbs) are emerging as safe and effective therapeutics against SARS-CoV-2. However, variant strains of SARS-CoV-2 have evolved, with early studies showing that some mAbs may not sustain their efficacy in the face of escape mutants. Also, from the onset of the COVID-19 pandemic, concern has been raised about the potential for Fcγ receptor-mediated antibody-dependent enhancement (ADE) of infection. In this study, plaque reduction neutralization assays demonstrated that mAb 1741-LALA neutralizes SARS-CoV-2 strains B.1.351, D614 and D614G. MAbs S1D2-hIgG1 and S1D2-LALA mutant (STI-1499-LALA) did not neutralize B.1.351, but did neutralize SARS-CoV-2 strains D614 and D614G. LALA mutations did not result in substantial differences in neutralizing abilities between clones S1D2-hIgG1 vs STI-1499-LALA. S1D2-hIgG1, STI-1499-LALA, and convalescent plasma showed minimal ability to induce ADE in human blood monocyte-derived macrophages. Further, no differences in pharmacokinetic clearance of S1D2-hIgG1 vs STI-1499-LALA were observed in mice expressing human FcRn. These findings confirm that SARS-CoV-2 has already escaped some mAbs, and identify a mAb candidate that may neutralize multiple SARS-CoV-2 variants. They also suggest that risk of ADE in macrophages may be low with SARS-CoV-2 D614, and LALA Fc change impacts neither viral neutralization nor Ab clearance.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Acrecentamiento Dependiente de Anticuerpo , SARS-CoV-2/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Glicoproteína de la Espiga del Coronavirus/inmunología , Células Vero
11.
Immunol Lett ; 103(1): 33-8, 2006 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-16386801

RESUMEN

We have recently discovered a reaction that all antibodies, regardless of source or antigenic specificity can catalyze, that is the reaction between singlet dioxygen ((1)O(2)(*)) and H(2)O to generate H(2)O(2). We have named this process the antibody-catalyzed water oxidation pathway (ACWOP). As part of our ongoing investigations into the possible biological role of this pathway, we have studied whether isoalloxazine-containing cofactors, that are known to be endogenous photosensitizers via Type-II pathways to generate (1)O(2)(*), such as riboflavin (RF, Vitamin B2) can trigger the ACWOP. Herein we show that regardless of the antigenic specificity or heavy and light chain composition, all antibodies and their fragments are able to intercept the (1)O(2)(*) generated by photo-oxidation of RF in the presence of oxygen (ambient aerobic conditions) to activate the ACWOP. The initial rate of HOOH generation by a panel of murine antibodies ranges from 0.218 to 0.998 microM/min. The initial rate of antibody-catalyzed HOOH production is accelerated in D(2)O and is quenched in NaN(3), highlighting the key intermediacy of (1)O(2)(*) in the process. Critically, the ACWOP is photo-activated at physiologically relevant concentrations of RF (<50 nM) suggesting that this pathway may be relevant in an in vivo setting. Finally, when activated by RF the ACWOP generates oxidants that accelerate the hemolysis of sheep RBCs hinting at a pathophysiological effect of this RF-induced photo-oxidation pathway.


Asunto(s)
Anticuerpos Catalíticos/química , Peróxido de Hidrógeno/síntesis química , Inmunoglobulina G/química , Riboflavina/química , Transducción de Señal , Agua/química , Animales , Anticuerpos Catalíticos/metabolismo , Anticuerpos Catalíticos/efectos de la radiación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Hemólisis/fisiología , Peróxido de Hidrógeno/metabolismo , Inmunoglobulina G/metabolismo , Ratones , Oxidación-Reducción/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Riboflavina/farmacología , Agua/metabolismo
12.
Biomacromolecules ; 3(2): 262-71, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11888310

RESUMEN

A N,N-dimethylacrylamide-based hydrogel (2) with the new cross-linker (ethylenedioxy) bis[2,2'-(N-acryloylamino)ethane] (1) has been prepared, and its physicochemical properties in aqueous solution were studied. Three different native proteins (lysozyme, bovine serum albumin, and rabbit IgG) were encapsulated within the polymeric matrix 2, and the kinetics of their release from the swollen hydrogel were determined. The rate of protein release exhibits a clear dependence on both the molecular weight of the protein and the amount of cross-linker utilized to prepare the hydrogel. This is reflected by the fact that the low molecular weight proteins are released at an increased rate versus higher molecular weight proteins. In addition a greater amount of protein is released from the hydrogels with a lower percentage of cross-linker. The polymerization procedure used in this study is sufficiently mild to safeguard the functional integrity of attendant biomolecules as determined by the retention of catalytic activity of encapsulated alpha-chymotrypsin and aldolase catalytic antibody 38C2. The potential utility of these hydrogels for the controlled release of bioactive agents in vivo is strengthened by both their lack of toxicity against human dermal fibroblasts and their lack of immunogenicity in mice.


Asunto(s)
Composición de Medicamentos , Hidrogeles , Proteínas/química , Acrilamidas/química , Quimotripsina , Colorimetría , Reactivos de Enlaces Cruzados , Preparaciones de Acción Retardada , Fluorescencia , Hidrogeles/química , Estructura Molecular , Muramidasa , Polímeros , Albúmina Sérica Bovina , Temperatura
13.
Hybrid Hybridomics ; 22(1): 23-31, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12713687

RESUMEN

Dendritic cells (DC) are the professional antigen-presenting cells of the immune system. Previous studies have demonstrated that targeting foreign antigens to DC leads to enhanced antigen (Ag)-specific responses in vivo. However, the utility of this strategy for the generation of MAbs has not been investigated. To address this question we immunized mice with IgG-peptide conjugates prepared with the hamster anti-murine CD11c MAb N418. Synthetic peptides corresponding to two different exposed regions of DC-specific ICAM-3 grabbing nonintegrin (DC-SIGN), a human C-type lectin, were conjugated to N418 using thiol-based chemistry. The N418 MAb served as the targeting molecule and synthetic peptides as the Ag (MAb-Ag). A rapid and peptide specific serum IgG response was produced by Day 7 when the synthetic peptides were linked to the N418 MAb, compared to peptide co-delivered with the N418 without linkage. Spleen cells from N418-peptide immunized mice were fused on Day 10, and three IgG1/k monoclonal antibodies (MAbs) were selected to one of the peptide epitopes (MID-peptide). One of the MAbs, Novik 2, bound to two forms of recombinant DC-SIGN protein in enzyme-linked immunosorbent assay (ELISA), and was specifically inhibited by the MID-peptide in solution. Two of these MAbs show specific binding to DC-SIGN expressed by cultured human primary DC. We conclude that in vivo DC targeting enhances the immunogenicity of synthetic peptides and is an effective method for the rapid generation of MAbs to predetermined epitopes.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células Dendríticas/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Antígeno CD11c/inmunología , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/inmunología , Citometría de Flujo , Humanos , Lectinas Tipo C/análisis , Lectinas Tipo C/inmunología , Ratones , Péptidos/metabolismo , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA