Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cells ; 12(8)2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37190054

RESUMEN

Human-relevant systems that mimic the 3D tumor microenvironment (TME), particularly the complex mechanisms of immuno-modulation in the tumor stroma, in a reproducible and scalable format are of high interest for the drug discovery industry. Here, we describe a novel 3D in vitro tumor panel comprising 30 distinct PDX models covering a range of histotypes and molecular subtypes and cocultured with fibroblasts and PBMCs in planar (flat) extracellular matrix hydrogels to reflect the three compartments of the TME-tumor, stroma, and immune cells. The panel was constructed in a 96-well plate format and assayed tumor size, tumor killing, and T-cell infiltration using high-content image analysis after 4 days of treatment. We screened the panel first against the chemotherapy drug Cisplatin to demonstrate feasibility and robustness, and subsequently assayed immuno-oncology agents Solitomab (CD3/EpCAM bispecific T-cell engager) and the immune checkpoint inhibitors (ICIs) Atezolizumab (anti-PDL1), Nivolumab (anti-PD1) and Ipilimumab (anti-CTLA4). Solitomab displayed a strong response across many PDX models in terms of tumor reduction and killing, allowing for its subsequent use as a positive control for ICIs. Interestingly, Atezolizumab and Nivolumab demonstrated a mild response compared to Ipilimumab in a subset of models from the panel. We later determined that PBMC spatial proximity in the assay setup was important for the PD1 inhibitor, hypothesizing that both duration and concentration of antigen exposure may be critical. The described 30-model panel represents a significant advancement toward screening in vitro models of the tumor microenvironment that include tumor, fibroblast, and immune cell populations in an extracellular matrix hydrogel, with robust and standardized high content image analysis in a planar hydrogel. The platform is aimed at rapidly screening various combinations and novel agents and forming a critical conduit to the clinic, thus accelerating drug discovery for the next generation of therapeutics.


Asunto(s)
Antineoplásicos Inmunológicos , Técnicas de Cultivo Tridimensional de Células , Hidrogeles , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Células del Estroma , Neoplasias/tratamiento farmacológico , Humanos , Inmunoterapia , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Línea Celular Tumoral , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico
2.
Mol Biotechnol ; 63(11): 1016-1029, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34185248

RESUMEN

Plants are promising drug-production platforms with high economic efficiency, stability, and convenience in mass production. However, studies comparing the equivalency between the original antibodies and those produced in plants are limited. Amino acid sequences that constitute the Fab region of an antibody are diverse, and the post-transcriptional modifications that occur according to these sequences in animals and plants are also highly variable. In this study, rituximab, a blockbuster antibody drug used in the treatment of non-Hodgkin's lymphoma, was produced in Nicotiana benthamiana leaves and Arabidopsis thaliana callus, and was compared to the original rituximab produced in CHO cells. Interestingly, the epitope recognition and antigen-binding abilities of rituximab from N. benthamiana leaves were almost lost. In the case of rituximab produced in A. thaliana callus, the specific binding ability and CD20 capping activity were maintained, but the binding affinity was less than 50% of that of original rituximab from CHO cells. These results suggest that different plant species exhibit different binding affinities. Accordingly, in addition to the differences in PTMs between mammals and plants, the differences between the species must also be considered in the process of producing antibodies in plants.


Asunto(s)
Antígenos CD20/metabolismo , Arabidopsis/metabolismo , Nicotiana/metabolismo , Hojas de la Planta/química , Rituximab/metabolismo , Animales , Afinidad de Anticuerpos , Antígenos CD20/química , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/metabolismo , Arabidopsis/genética , Cricetinae , Humanos , Hojas de la Planta/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Rituximab/biosíntesis , Rituximab/genética , Rituximab/aislamiento & purificación , Nicotiana/genética
3.
FEBS Lett ; 595(11): 1587-1603, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33792041

RESUMEN

OX40 is a costimulatory molecule that belongs to the tumor necrosis factor receptor (TNFR) superfamily. OX40 agonist-based combinations are emerging as promising candidates for novel cancer immunotherapy. Clinical trials have shown that OX40 agonist antibodies could lead to better results in cancer patients. Using a hybridoma platform and three different types of immunization strategies, namely recombinant protein, DNA, and overexpressing cells, we identified a chimeric anti-OX40 antibody (mAb035-hIgG1 from DNA immunization) that shows excellent binding specificity, and slightly stronger activation of human memory CD4+ T cells and similar potent antitumor activity compared with BMS 986178, an anti-OX40 antibody currently being evaluated for the treatment of solid tumors. This paper further systematically investigates the antigen-specific immune response, the number of binders, epitope bins, and functional activities of antibodies among different immunization strategies. Interestingly, we found that different immunization strategies affect the biological activity of monoclonal antibodies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Inmunización/métodos , Receptores OX40/inmunología , Proteínas Recombinantes de Fusión/farmacología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/metabolismo , Bioensayo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Células CHO , Cricetulus , Femenino , Adyuvante de Freund/administración & dosificación , Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Hibridomas/química , Hibridomas/inmunología , Fragmentos Fc de Inmunoglobulinas/biosíntesis , Fragmentos Fc de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fc de Inmunoglobulinas/farmacología , Células Jurkat , Luciferasas/genética , Luciferasas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/inmunología , Receptores OX40/antagonistas & inhibidores , Receptores OX40/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación
4.
Protein Expr Purif ; 184: 105888, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33857600

RESUMEN

Anti-EGFR nanobodies have been successfully applied as antitumor moieties in the photodynamic therapy and drug delivery systems. But the yields of nanobodies were still limited due to the volumetric capacity of the periplasmic compartments and inclusion bodies of Escherichia coli. A comparative study of Pichia pastoris and Escherichia coli was done through characterizing their products. Nanobody 7D12 and 7D12-9G8 were successfully expressed in Pichia pastoris with 6-13.6-fold higher yield. Both two types of nanobodies had internalization ability to be developed as antitumor moieties.


Asunto(s)
Antineoplásicos Inmunológicos , Escherichia coli , Proteínas de Neoplasias , Saccharomycetales , Anticuerpos de Dominio Único , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Saccharomycetales/genética , Saccharomycetales/metabolismo , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/aislamiento & purificación , Anticuerpos de Dominio Único/farmacología
5.
BMC Cancer ; 21(1): 321, 2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33765976

RESUMEN

BACKGROUND: We prepared an anti-p21Ras scFv which could specifically bind with mutant and wild-type p21Ras. However, it cannot penetrate the cell membrane, which prevents it from binding to p21Ras in the cytoplasm. Here, the RGD4C peptide was used to mediate the scFv penetration into tumor cells and produce antitumor effects. METHODS: RGD4C-EGFP and RGD4C-p21Ras-scFv recombinant expression plasmids were constructed to express fusion proteins in E. coli, then the fusion proteins were purified with HisPur Ni-NTA. RGD4C-EGFP was used as reporter to test the factors affecting RGD4C penetration into tumor cell. The immunoreactivity of RGD4C-p21Ras-scFv toward p21Ras was identified by ELISA and western blotting. The ability of RGD4C-p21Ras-scFv to penetrate SW480 cells and colocalization with Ras protein was detected by immunocytochemistry and immunofluorescence. The antitumor activity of the RGD4C-p21Ras-scFv was assessed with the MTT, TUNEL, colony formation and cell migration assays. Chloroquine (CQ) was used an endosomal escape enhancing agent to enhance endosomal escape of RGD4C-scFv. RESULTS: RGD4C-p21Ras-scFv fusion protein were successfully expressed and purified. We found that the RGD4C fusion protein could penetrate into tumor cells, but the tumor cell entry of was time and concentration dependent. Endocytosis inhibitors and a low temperature inhibited RGD4C fusion protein endocytosis into cells. The change of the cell membrane potential did not affect penetrability. RGD4C-p21Ras-scFv could penetrate SW480 cells, effectively inhibit the growth, proliferation and migration of SW480 cells and promote this cells apoptosis. In addition, chloroquine (CQ) could increase endosomal escape and improve antitumor activity of RGD4C-scFv in SW480 cells. CONCLUSION: The RGD4C peptide can mediate anti-p21Ras scFv entry into SW480 cells and produce an inhibitory effect, which indicates that RGD4C-p21Ras-scFv may be a potential therapeutic antibody for the treatment of ras-driven cancers.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Humanos , Inmunoconjugados/genética , Inmunoconjugados/aislamiento & purificación , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/uso terapéutico , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/aislamiento & purificación , Anticuerpos de Cadena Única/farmacología , Anticuerpos de Cadena Única/uso terapéutico
6.
Cancer Immunol Immunother ; 70(9): 2529-2543, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33570675

RESUMEN

Intravesical Bovis bacillus Calmette-Guérin (BCG) therapy is the most effective immunotherapy for bladder cancer, but it sometime causes serious side effects because of its inclusion of live bacteria. It is necessary to develop a more active but less toxic immunotherapeutic agent. Trehalose 6,6'-dimycolate (TDM), the most abundant hydrophobic glycolipid of the BCG cell wall, has been reported to show various immunostimulatory activities such as granulomagenesis and adjuvant activity. Here, we developed cationic liposomes incorporating TDM purified from Mycobacterium bovis BCG Connaught, and we investigated the antitumor effect of the cationic liposome TDM (Lip-TDM). Lip-TDM exerted an antitumor effect in bladder cancer, colon cancer, and melanoma-bearing mouse models that was comparable or even superior to that of BCG, with no body weight loss or granuloma formation. The antitumor effect of Lip-TDM disappeared in two types of mice: those with depletion of CD8+ T cells, and those with knockout of macrophage-inducible C-type lectin (Mincle) which recognize TDM. Lip-TDM treatment enhanced the maturation and migration of dendritic cells in the tumor microenvironment in a Mincle-dependent manner. Our results elucidate mechanisms that underlie Lip-TDM treatment and suggest that Lip-TDM has potential as a safe and effective treatment for various cancers.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Factores Cordón/administración & dosificación , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Factores Inmunológicos/administración & dosificación , Mycobacterium bovis , Adyuvantes Inmunológicos , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/aislamiento & purificación , Linfocitos T CD8-positivos/metabolismo , Fraccionamiento Químico , Factores Cordón/química , Factores Cordón/aislamiento & purificación , Citocinas/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/aislamiento & purificación , Inmunofenotipificación , Infusiones Parenterales , Liposomas , Activación de Linfocitos , Ratones , Estructura Molecular , Mycobacterium bovis/química , Solventes , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biomed Chromatogr ; 35(1): e4996, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33047346

RESUMEN

Immuno-oncology (IO) is an emerging option to treat cancer malignancies. In the last two years, IO has accounted for more than 90% of the new active drugs in various therapeutic indications of oncology drug development. Bioanalytical methods used for the quantitation of various IO small molecule drugs have been summarized in this review. The most commonly used are HPLC and LC-MS/MS methods. Determination of IO drugs from biological matrices involves drug extraction from the biological matrix, which is mostly achieved by simple protein precipitation, liquid-liquid extraction and solid-phase extraction. Subsequently, quantitation is usually achieved by LC-MS/MS, but HPLC-UV has also been employed. The bioanalytical methods reported for each drug are briefly discussed and tabulated for easy access. Our review indicates that LC-MS/MS is a versatile and reliable tool for the sensitive, rapid and robust quantitation of IO drugs.


Asunto(s)
Antineoplásicos Inmunológicos/análisis , Antineoplásicos Inmunológicos/aislamiento & purificación , Cromatografía Liquida , Espectrometría de Masas en Tándem , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Cromatografía Líquida de Alta Presión , Humanos , Extracción Líquido-Líquido , Ratones , Neoplasias/tratamiento farmacológico , Extracción en Fase Sólida
8.
Oncol Rep ; 44(5): 1949-1960, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33000243

RESUMEN

CD44 is widely expressed on the surface of most tissues and all hematopoietic cells, and regulates many genes associated with cell adhesion, migration, proliferation, differentiation, and survival. CD44 has also been studied as a therapeutic target in several cancers. Previously, an anti­CD44 monoclonal antibody (mAb), C44Mab­5 (IgG1, kappa) was established by immunizing mice with CD44­overexpressing Chinese hamster ovary (CHO)-K1 cells. C44Mab­5 recognized all CD44 isoforms, and showed high sensitivity for flow cytometry and immunohistochemical analysis in oral cancers. However, as the IgG1 subclass of C44Mab­5 lacks antibody­dependent cellular cytotoxicity (ADCC) and complement­dependent cytotoxicity (CDC), the antitumor activity of C44Mab­5 could not be determined. In the present study, we converted the mouse IgG1 subclass antibody C44Mab­5 into an IgG2a subclass antibody, 5­mG2a, and further produced a defucosylated version, 5­mG2a­f, using FUT8­deficient ExpiCHO­S (BINDS­09) cells. Defucosylation of 5­mG2a­f was confirmed using fucose­binding lectins, such as AAL and PhoSL. The dissociation constants (KD) for 5­mG2a­f against SAS and HSC­2 oral cancer cells were determined through flow cytometry to be 2.8x10­10 M and 2.6x10­9 M, respectively, indicating that 5­mG2a­f possesses extremely high binding affinity. Furthermore, immunohistochemical staining using 5­mG2a­f specifically stained the membranes of oral cancer cells. In vitro analysis demonstrated that 5­mG2a­f showed moderate ADCC and CDC activities against SAS and HSC­2 oral cancer cells. In vivo analysis revealed that 5­mG2a­f significantly reduced tumor development in SAS and HSC­2 xenografts in comparison to control mouse IgG, even after injection seven days post­tumor inoculation. Collectively, these results suggest that treatment with 5­mG2a­f may represent a useful therapy for patients with CD44­expressing oral cancers.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Receptores de Hialuranos/antagonistas & inhibidores , Neoplasias de la Boca/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/farmacología , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetulus , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Ratones , Neoplasias de la Boca/inmunología , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Chemother Pharmacol ; 86(5): 595-606, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32965540

RESUMEN

PURPOSE: To report population pharmacokinetic (PK) analysis of the phase 1 study (FPA144-001, NCT02318329) and to select a clinical dose and schedule that will achieve an empirical target trough concentration (Ctrough) for an anti-fibroblast growth factor receptor 2b antibody, bemarituzumab. METHODS: Nonlinear mixed-effect modeling was used to analyse PK data. In vitro binding affinity and receptor occupancy of bemarituzumab were determined. Simulation was conducted to estimate dose and schedule to achieve an empirical target Ctrough in a phase 2 trial (FIGHT, NCT03694522) for patients receiving first-line treatment combined with modified 5-fluourouracil, oxaliplatin and leucovorin (mFOLFOX6) for gastric and gastroesophageal junction adenocarcinoma. RESULTS: Bemarituzumab PK is best described by a two-compartment model with parallel linear and nonlinear (Michaelis-Menten) elimination from the central compartment. Albumin, gender, and body weight were identified as the covariates on the linear clearance and/or volume of distribution in the central compartment, and no dose adjustment was warranted. An empirical target of bemarituzumab Ctrough of ≥ 60 µg/mL was projected to achieve > 95% receptor occupancy based on in vitro data. Fifteen mg/kg every 2 weeks, with a single dose of 7.5 mg/kg on Cycle 1 Day 8, was projected to achieve the target Ctrough on Day 15 in 98% of patients with 96% maintaining the target at steady state, which was confirmed in the FIGHT trial. CONCLUSION: A projected dose and schedule to achieve the target Ctrough was validated in phase 1 of the FIGHT trial which supported selection of the phase 2 dose and schedule for bemarituzumab.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/farmacocinética , Antineoplásicos Inmunológicos/farmacocinética , Neoplasias Esofágicas/tratamiento farmacológico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Adenocarcinoma/sangre , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/aislamiento & purificación , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/aislamiento & purificación , Células CHO , Ensayos Clínicos Fase II como Asunto , Cricetulus , Relación Dosis-Respuesta a Droga , Neoplasias Esofágicas/sangre , Neoplasias Esofágicas/patología , Unión Esofagogástrica/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Neoplasias Gástricas/sangre , Neoplasias Gástricas/patología
10.
J Immunother ; 43(6): 175-188, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32349046

RESUMEN

B-cell maturation antigen (BCMA) is a highly plasma cell-selective protein expressed on malignant plasma cells of patients with multiple myeloma (MM), and it is a defined therapeutic target. Major histocompatibility complex class I-related chain A (MICA) is frequently expressed in lymphoproliferative malignancies including MM. MICA activates natural killer (NK) cells and costimulates T cells by interaction with its immunoreceptor NK cell receptor G2D (NKG2D). Nonetheless, during full-blown MM, tumor cells promote efficient MICA shedding, which evokes NKG2D internalization and immune suppression. To enhance the directional killing efficacy of immune cells against myeloma cells, we constructed a novel bispecific antibody 2A9-MICA and explored its potential antimyeloma activity against MM. 2A9-MICA consists of human MICA extracellular region and a single-chain antibody fragment (scFv) that targets BCMA generated by phage display technology. In vitro, 2A9-MICA activated NK cell-mediated cytotoxicity and induced NK cells to kill BCMA-positive human myeloma cells. Moreover, in BCMA-positive, MM-bearing nude mice, 2A9-MICA specifically targeted tumor tissue, where it effectively recruited immune cells and inhibited tumor tissue growth showed superior antitumor activity. Taken together, bispecific antibody 2A9-MICA provides a new approach for MM-targeting immunotherapy and has attractive potential for clinical applications.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Antígeno de Maduración de Linfocitos B/antagonistas & inhibidores , Mieloma Múltiple/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/agonistas , Anticuerpos Biespecíficos/aislamiento & purificación , Anticuerpos Biespecíficos/uso terapéutico , Afinidad de Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/uso terapéutico , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/etiología , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología
11.
Chem Biodivers ; 17(5): e2000037, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32163220

RESUMEN

The ocotillol (OCT)-type saponins have been known as a tetracyclic triterpenoid, possessing five- or six-membered epoxy ring in the side chain. Interestingly, this type saponin was mostly found in Panax vietnamensis Ha et Grushv., Araliaceae (VG), hence making VG unique from the other Panax spp. Five OCT-type saponins, majonoside R2, vina-ginsenoside R2, majonoside R1, pseudoginsenoside RT4, vina-ginsenoside R11, together with three protopanaxadiol (PPD)-type saponins and four protopanaxatriol (PPT)-type saponins from VG were evaluated for their antimelanogenic activity. All of isolates were found to be active. More importantly, the five OCT-type saponins inhibited melanin production in B16-F10 mouse melanoma cells, without showing any cytotoxicity. Besides ginsenoside Rd and ginsenoside Rg3 in PPD and notoginsenoside R1 in PPT-type saponins, majonoside R2 was the most potent melanogenesis inhibitory activity in OCT-type saponins. In this article, we highlighted antimelanogenic activity of OCT-type saponins and potential structure-activity relationship (SAR) of ginsenosides. Our results suggested that OCT-type saponins could be used as a depigmentation agent.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Ginsenósidos/farmacología , Melanoma/tratamiento farmacológico , Panax/química , Saponinas/farmacología , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/aislamiento & purificación , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Ginsenósidos/química , Ginsenósidos/aislamiento & purificación , Melaninas/antagonistas & inhibidores , Melaninas/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Conformación Molecular , Plantas Medicinales , Saponinas/química , Saponinas/aislamiento & purificación , Relación Estructura-Actividad , Células Tumorales Cultivadas
12.
Prep Biochem Biotechnol ; 50(3): 252-259, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31799894

RESUMEN

Targeting the interaction interface is an effective strategy to obtain programmed death receptor 1 (PD-1)/PD-1 ligand 1 (PD-L1) nanobody blockers. To validate this strategy, the interaction interface between PD-1 and the PD-L1 extracellular domain were analyzed using Cn3D 4.1. The peptide PD-1125-136 located at the interface of PD-1 was selected as the antigen to screen nanobodies from a humanized nanobody phage display library. Six different nanobodies were screened, with molecular weights of 12 ∼ 13 kDa, excluding a single basic protein. The nanobody with the longest CDR3 region, termed PD-1-Nb-B20, was selected for further analysis. For mass production, the C-terminal His6-tagged nanobody coding sequence was optimized and cloned into pET-21b for over-expression under the T7 promoter in Escherichia coli BL21 (DE3). PD-1-Nb-B20 was expressed and pancreatic adenocarcinoma cells BxPC-3 over-expressing PD-L1 were selected for nanobody competitive inhibition assays. The purified nanobodies significantly inhibited PD-1 binding to the surface of target cells, indicating their ability to block the PD-1/PD-L1 interaction.


Asunto(s)
Antineoplásicos Inmunológicos , Antígeno B7-H1/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Pancreáticas/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Anticuerpos de Dominio Único , Células A549 , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/aislamiento & purificación , Células HeLa , Humanos , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/aislamiento & purificación , Neoplasias Pancreáticas
13.
J Exp Clin Cancer Res ; 38(1): 332, 2019 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-31362764

RESUMEN

BACKGROUND: Immunotherapeutic approaches designed to augment T and B cell mediated killing of tumor cells has met with clinical success in recent years suggesting tremendous potential for treatment in a broad spectrum of tumor types. After complex recognition of target cells by T and B cells, delivery of the serine protease granzyme B (GrB) to tumor cells comprises the cytotoxic insult resulting in a well-characterized, multimodal apoptotic cascade. METHODS: We designed a recombinant fusion construct, GrB-Fc-4D5, composed of a humanized anti-HER2 scFv fused to active GrB for recognition of tumor cells and internal delivery of GrB, simulating T and B cell therapy. We assessed the construct's antigen-binding specificity and GrB enzymatic activity, as well as in vitro cytotoxicity and internalization into target and control cells. We also assessed pharmacokinetic and toxicology parameters in vivo. RESULTS: GrB-Fc-4D5 was highly cytotoxic to Her2 positive cells such as SKBR3, MCF7 and MDA-MB-231 with IC50 values of 56, 99 and 27 nM, respectively, and against a panel of HER2+ cell lines regardless of endogenous expression levels of the PI-9 inhibitor. Contemporaneous studies with Kadcyla demonstrated similar levels of in vitro activity against virtually all cells tested. GrB-Fc-4D5 internalized rapidly into target SKOV3 cells within 1 h of exposure rapidly delivering GrB to the cytoplasmic compartment. In keeping with its relatively high molecular weight (160 kDa), the construct demonstrated a terminal-phase serum half-life in mice of 39.2 h. Toxicity studies conducted on BALB/c mice demonstrated no statistically significant changes in SGPT, SGOT or serum LDH. Histopathologic analysis of tissues from treated mice demonstrated no drug-related changes in any tissues examined. CONCLUSION: GrB-Fc-4D5 shows excellent, specific cytotoxicity and demonstrates no significant toxicity in normal, antigen-negative murine models. This construct constitutes a novel approach against HER2-expressing tumors and is an excellent candidate for further development.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Desarrollo de Medicamentos , Terapia Molecular Dirigida , Receptor ErbB-2/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Animales , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/aislamiento & purificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Expresión Génica , Vectores Genéticos/genética , Granzimas/administración & dosificación , Granzimas/genética , Humanos , Ratones , Terapia Molecular Dirigida/métodos , Unión Proteica/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Anticuerpos de Cadena Única/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Exp Cell Res ; 383(2): 111508, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31356815

RESUMEN

OBJECTIVES: Immunotherapy and personalized medicine therapeutics are emerging as promising approaches in the management of head and neck squamous cell carcinoma (HNSCC). In spite of that, there is yet no assay that could predict individual response to immunotherapy. METHODS: We manufactured an in vitro 3D microfluidic chip to test the efficacy of immunotherapy. The assay was first tested using a tongue cancer cell line (HSC-3) embedded in a human tumour-derived matrix "Myogel/fibrin" and immune cells from three healthy donors. Next, the chips were used with freshly isolated cancer cells, patients' serum and immune cells. Chips were loaded with different immune checkpoint inhibitors, PD-L1 antibody and IDO 1 inhibitor. Migration of immune cells towards cancer cells and the cancer cell proliferation rate were evaluated. RESULTS: Immune cell migration towards HSC-3 cells was cancer cell density dependent. IDO 1 inhibitor induced immune cells to migrate towards cancer cells both in HSC-3 and in two HNSCC patient samples. Efficacy of PD-L1 antibody and IDO 1 inhibitor was patient dependent. CONCLUSION: We introduced the first humanized in vitro microfluidic chip assay to test immunotherapeutic drugs against HNSCC patient samples. This assay could be used to predict the efficacy of immunotherapeutic drugs for individual patients.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias de Cabeza y Cuello/patología , Inmunoterapia , Dispositivos Laboratorio en un Chip , Medicina de Precisión , Cultivo Primario de Células/métodos , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Anciano , Anciano de 80 o más Años , Antineoplásicos Inmunológicos/aislamiento & purificación , Biomarcadores Farmacológicos/análisis , Ensayos de Migración Celular/instrumentación , Ensayos de Migración Celular/métodos , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo/instrumentación , Técnicas de Cocultivo/métodos , Ensayos de Selección de Medicamentos Antitumorales/instrumentación , Ensayos de Selección de Medicamentos Antitumorales/métodos , Diseño de Equipo , Femenino , Neoplasias de Cabeza y Cuello/diagnóstico , Neoplasias de Cabeza y Cuello/terapia , Humanos , Factores Inmunológicos/aislamiento & purificación , Factores Inmunológicos/uso terapéutico , Inmunoterapia/instrumentación , Inmunoterapia/métodos , Masculino , Microfluídica/instrumentación , Microfluídica/métodos , Persona de Mediana Edad , Lesiones Precancerosas/diagnóstico , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/patología , Medicina de Precisión/instrumentación , Medicina de Precisión/métodos , Cultivo Primario de Células/instrumentación , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/diagnóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Andamios del Tejido , Células Tumorales Cultivadas
15.
Sci Rep ; 9(1): 8097, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31147593

RESUMEN

Described in several epithelial cancer cells, Tn- (GalNAcα1-O-Ser/Thr) and T- (Galß3GalNAcα1-O-Ser/Thr) antigens are examples of tumor-associated antigens. Increased expression of Tn- and T-antigens is associated with tumor invasion and metastasis, and patients with high concentration of anti-Tn and anti-T antibodies have a more benign evolution of pathology. Asialofetuin (ASF) and ovine submaxillary mucin (OSM) are two glycoproteins that expose T- and Tn-antigen, respectively. In this work, using ASF or OSM we affinity-purified anti-T and anti-Tn antibodies from normal human plasma and tested their ability to specifically recognize tumor human tissues. Whereas purified anti-T antibodies (purity degree increase of 127-fold, and 22% recovery) were mainly IgG, for purified anti-Tn antibodies (purity degree enhancement of 125-fold, and 26% yield) the IgM fraction was predominant over the IgG one. IgG2 subclass was significantly enriched in both purified antibody samples. Purified antibodies did not bind normal human tissue (0/42), although recognized malignant tissues from different origin such as colon carcinoma (11/77 by anti-Tn; 7/79 by anti-T), breast carcinoma (10/23 by anti-Tn; 7/23 by anti-T), and kidney carcinoma (45/51 by anti-Tn; 42/51 by anti-T). Our results suggest that purified human anti-Tn and anti-T antibodies have a potential as anti-tumor therapeutic agents; restoring their levels in human sera could positively affect the evolution of patients with epithelial tumor pathologies.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antineoplásicos Inmunológicos/farmacología , Carcinoma/tratamiento farmacológico , Inmunoglobulina G/farmacología , Inmunoglobulina M/farmacología , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/uso terapéutico , Asialoglicoproteínas/inmunología , Carcinoma/inmunología , Carcinoma/patología , Línea Celular Tumoral , Cromatografía de Afinidad/métodos , Ensayos de Selección de Medicamentos Antitumorales , Fetuínas/inmunología , Humanos , Proteínas Inmovilizadas/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/uso terapéutico , Inmunoglobulina M/inmunología , Inmunoglobulina M/aislamiento & purificación , Inmunoglobulina M/uso terapéutico , Mucinas/inmunología , Plasma/inmunología
16.
Carbohydr Polym ; 211: 272-280, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824089

RESUMEN

The biological activity of macrofungal polysaccharides (MFPS) depends on their structural features and is a topic of keen interest for researchers since long time. In this communication, we report a water soluble macrofungal heteropolysaccharide (MFPS1) with a molar weight of ˜145,000 g/mol, obtained through alkali extraction, of the wild mushroom, Marasmiellus palmivorus, with significant immunomodulatory properties. In cancer, after the induction of metastasis, the anticancer immune system becomes unresponsive. By studying cytokine secretion and immune phenotyping, it was observed that MFPS1 reactivated the anticancer immune surveillance system. MFPS1 executed T-cell maturation and activation via M1Φ; and also stimulated natural killer (NK) cell and B-cell population. The entire immune activation pathway corroborates its anticancer activity. The RP-HPLC analysis of hydrolyzed MFPS1 showed arabinose, glucose, galactose and mannose as monosaccharide units. The proposed structure of repeating unit was established from methylation analysis, 1D- and 2D NMR study, HR-MS and MALDI-TOF MS analysis.


Asunto(s)
Agaricales/química , Antineoplásicos Inmunológicos , Cuerpos Fructíferos de los Hongos/química , Polisacáridos , Células A549 , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/farmacología , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Citocinas/inmunología , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Polisacáridos/farmacología , Células RAW 264.7
17.
Med Sci (Paris) ; 35(12): 1043-1053, 2019 Dec.
Artículo en Francés | MEDLINE | ID: mdl-31903916

RESUMEN

An Antibody-Drug Conjugate (armed antibody) is a vectorized chemotherapy that results from the grafting of a cytotoxic agent on a monoclonal antibody via a judiciously designed spacer arm. ADCs have made considerable progress in 10 years. In 2009, only gemtuzumab ozogamicin (Mylotarg®) was used clinically. In 2019, 4 other ADCs have been approved and more than 80 others are in active clinical trials. The second part of this review will focus on new emerging strategies to address ADCs drawbacks and attempt to broaden their therapeutic window. Finally, combinations with conventional chemotherapy or checkpoint inhibitors will be discussed, in the pursuit to make Antibody-Drug Conjugates the embodiment of Paul Ehrlich's dream of the magic bullet.


TITLE: Les immunoconjugués en oncologie - Les nouvelles stratégies en développement. ABSTRACT: Un anticorps armé (ADC, antibody-drug conjugate en anglais) est une chimiothérapie vectorisée qui résulte du greffage d'un agent cytotoxique sur un anticorps monoclonal par l'intermédiaire d'un bras espaceur judicieusement construit. Les anticorps armés ont fait des progrès considérables en 10 ans. En 2009, seul le gemtuzumab ozogamicine (Mylotarg®) était utilisé en clinique. En 2019, 4 autres ADC ont été approuvés par la Food and Drug Administration et plus de 80 autres sont en études cliniques actives. La seconde partie de cette revue sera focalisée sur les nouvelles stratégies émergentes pour faire face aux limitations des ADC actuels et pour tenter d'élargir leur fenêtre thérapeutique. Enfin, les combinaisons avec la chimiothérapie classique ou les inhibiteurs de points de contrôles seront discutées, pour tenter de faire des anticorps armés la magic bullet dont rêvait Paul Ehrlich.


Asunto(s)
Desarrollo de Medicamentos/métodos , Inmunoconjugados , Oncología Médica , Neoplasias/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/clasificación , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/uso terapéutico , Desarrollo de Medicamentos/tendencias , Historia del Siglo XXI , Humanos , Inmunoconjugados/clasificación , Inmunoconjugados/aislamiento & purificación , Inmunoconjugados/uso terapéutico , Oncología Médica/métodos , Oncología Médica/tendencias , Neoplasias/patología
18.
Med Sci (Paris) ; 35(12): 1202-1203, 2019 Dec.
Artículo en Francés | MEDLINE | ID: mdl-31903940
19.
Drug Des Devel Ther ; 12: 2645-2654, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30214153

RESUMEN

INTRODUCTION: Interleukin-15 (IL-15) is an immunomodulatory cytokine. It can activate and expand cytotoxic CD8 T lymphocytes and natural killer cells, leading to potent antitumor effects. Various forms of IL-15 are now in different stages of development for cancer immunotherapy. One of the major issues with IL-15 or IL15-IL15Rα fusion is high toxicity due to systemic activation of immune cells. MATERIALS AND METHODS: In this study, we engineered a nanobody-cytokine fusion molecule, anti-CEA-IL15, in which an anti-CEA nanobody was linked to an IL15Rα-IL15 fusion. The nanobody-cytokine fusion exhibited multiple mechanisms to kill tumor cells, including promoting immune cell proliferation and directing antibody-dependent cytotoxicity against CEA-positive tumor cells. RESULTS: In xenograft models, anti-CEA-IL15 was localized in the tumor microenvironment and exhibited more potent antitumor activities than non-targeting IL-15, supporting potential application of this multifunctional fusion molecule in tumor immunotherapy. CONCLUSION: We generated and validated a tumortargeting fusion protein, anti-CEA-IL15, which has potent cytokine activity to activate and mobilize the immune system to fight cancer cells. Such strategies may also be applied to other cytokines and tumor-targeting molecules to increase antitumor efficacy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antineoplásicos Inmunológicos/farmacología , Antígeno Carcinoembrionario/inmunología , Interleucina-15/inmunología , Animales , Anticuerpos Monoclonales/química , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/aislamiento & purificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Inmunoterapia , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
20.
Hum Antibodies ; 26(2): 95-101, 2018 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-29036806

RESUMEN

Immunotherapy is now at the forefront of cancer therapeutic development. Gliomas are a particularly aggressive form of brain cancer for which immunotherapy may hold promise. Pritumumab (also known in the literature as CLNH11, CLN-IgG, and ACA-11) was the first monoclonal antibody tested in cancer patients. Pritumumab is a natural human monoclonal antibody developed from a B lymphocyte isolated from a regional draining lymph node of a patient with cervical carcinoma. The antibody binds ecto-domain vimentin on the surface of cancer cells. Pritumumab was originally tested in clinical trials with brain cancer patients in Japan where it demonstrated therapeutic benefit. It was reported to be a safe and effective therapy for brain cancer patients at doses 5-10 fold less than currently approved antibodies. Phase I dose escalation clinical trials are now being planned with pritumumab for the near future. Here we review data on the development and characterization of pritumumab, and review clinical trails data assessing immunotherapeutic effects of pritumumab for glioma patients.


Asunto(s)
Anticuerpos Monoclonales/aislamiento & purificación , Antineoplásicos Inmunológicos/aislamiento & purificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Inmunoglobulina G/aislamiento & purificación , Vimentina/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/metabolismo , Antineoplásicos Inmunológicos/uso terapéutico , Linfocitos B/química , Linfocitos B/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Ensayos Clínicos como Asunto , Expresión Génica , Glioma/genética , Glioma/inmunología , Glioma/mortalidad , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/uso terapéutico , Inmunoterapia/métodos , Ratones , Análisis de Supervivencia , Vimentina/antagonistas & inhibidores , Vimentina/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA