Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Biomed Eng ; 6(7): 819-829, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35534574

RESUMEN

Immune-checkpoint inhibitors have shown modest efficacy against immunologically 'cold' tumours. Interleukin-12 (IL-12)-a cytokine that promotes the recruitment of immune cells into tumours as well as immune cell activation, also in cold tumours-can cause severe immune-related adverse events in patients. Here, by exploiting the preferential overexpression of proteases in tumours, we show that fusing a domain of the IL-12 receptor to IL-12 via a linker cleavable by tumour-associated proteases largely restricts the pro-inflammatory effects of IL-12 to tumour sites. In mouse models of subcutaneous adenocarcinoma and orthotopic melanoma, masked IL-12 delivered intravenously did not cause systemic IL-12 signalling and eliminated systemic immune-related adverse events, led to potent therapeutic effects via the remodelling of the immune-suppressive microenvironment, and rendered cold tumours responsive to immune-checkpoint inhibition. We also show that masked IL-12 is activated in tumour lysates from patients. Protease-sensitive masking of potent yet toxic cytokines may facilitate their clinical translation.


Asunto(s)
Interleucina-12 , Melanoma , Animales , Citocinas , Inmunoterapia , Interleucina-12/farmacología , Ratones , Péptido Hidrolasas , Microambiente Tumoral
2.
Nat Biomed Eng ; 5(5): 387-398, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33046864

RESUMEN

Interleukin-4 (IL-4) suppresses the development of multiple sclerosis in a murine model of experimental autoimmune encephalomyelitis (EAE). Here, we show that, in mice with EAE, the accumulation and persistence in the lymph nodes and spleen of a systemically administered serum albumin (SA)-IL-4 fusion protein leads to higher efficacy in preventing disease development than the administration of wild-type IL-4 or of the clinically approved drug fingolimod. We also show that the SA-IL-4 fusion protein prevents immune-cell infiltration in the spinal cord, decreases integrin expression in antigen-specific CD4+ T cells, increases the number of granulocyte-like myeloid-derived suppressor cells (and their expression of programmed-death-ligand-1) in spinal cord-draining lymph nodes and decreases the number of T helper 17 cells, a pathogenic cell population in EAE. In mice with chronic EAE, SA-IL-4 inhibits immune-cell infiltration into the spinal cord and completely abrogates immune responses to myelin antigen in the spleen. The SA-IL-4 fusion protein may be prophylactically and therapeutically advantageous in the treatment of multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/administración & dosificación , Interleucina-4/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Albúmina Sérica/metabolismo , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Semivida , Inmunosupresores/farmacocinética , Inmunosupresores/farmacología , Inyecciones Intravenosas , Ganglios Linfáticos/química , Ganglios Linfáticos/inmunología , Ratones , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Bazo/química , Bazo/inmunología , Células Th17/efectos de los fármacos
3.
Arthritis Rheumatol ; 73(5): 769-778, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33169522

RESUMEN

OBJECTIVE: Rheumatoid arthritis (RA) is a major autoimmune disease that causes synovitis and joint damage. Although clinical trials have been performed using interleukin-10 (IL-10), an antiinflammatory cytokine, as a potential treatment of RA, the therapeutic effects of IL-10 have been limited, potentially due to insufficient residence in lymphoid organs, where antigen recognition primarily occurs. This study was undertaken to engineer an IL-10-serum albumin (SA) fusion protein and evaluate its effects in 2 murine models of RA. METHODS: SA-fused IL-10 (SA-IL-10) was recombinantly expressed. Mice with collagen antibody-induced arthritis (n = 4-7 per group) or collagen-induced arthritis (n = 9-15 per group) were injected intravenously with wild-type IL-10 or SA-IL-10, and the retention of SA-IL-10 in the lymph nodes (LNs), immune cell composition in the paws, and therapeutic effect of SA-IL-10 on mice with arthritis were assessed. RESULTS: SA fusion to IL-10 led to enhanced accumulation in the mouse LNs compared with unmodified IL-10. Intravenous SA-IL-10 treatment restored immune cell composition in the paws to a normal status, elevated the frequency of suppressive alternatively activated macrophages, reduced IL-17A levels in the paw-draining LN, and protected joint morphology. Intravenous SA-IL-10 treatment showed similar efficacy as treatment with an anti-tumor necrosis factor antibody. SA-IL-10 was equally effective when administered intravenously, locally, or subcutaneously, which is a benefit for clinical translation of this molecule. CONCLUSION: SA fusion to IL-10 is a simple but effective engineering strategy for RA therapy and has potential for clinical translation.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Articulaciones del Pie/efectos de los fármacos , Interleucina-10/farmacología , Ganglios Linfáticos/inmunología , Macrófagos/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Albúmina Sérica/farmacología , Animales , Células Presentadoras de Antígenos/metabolismo , Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Modelos Animales de Enfermedad , Pie , Articulaciones del Pie/inmunología , Articulaciones del Pie/metabolismo , Articulaciones del Pie/patología , Miembro Posterior , Antígenos de Histocompatibilidad Clase I/metabolismo , Inyecciones Intravenosas , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-6/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Ingeniería de Proteínas , Transporte de Proteínas , Receptores Fc/metabolismo , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/inmunología , Inhibidores del Factor de Necrosis Tumoral/farmacología
5.
Nat Biomed Eng ; 4(5): 531-543, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284554

RESUMEN

Checkpoint-inhibitor (CPI) immunotherapy has achieved remarkable clinical success, yet its efficacy in 'immunologically cold' tumours has been modest. Interleukin-12 (IL-12) is a powerful cytokine that activates the innate and adaptive arms of the immune system; however, the administration of IL-12 has been associated with immune-related adverse events. Here we show that, after intravenous administration of a collagen-binding domain fused to IL-12 (CBD-IL-12) in mice bearing aggressive mouse tumours, CBD-IL-12 accumulates in the tumour stroma due to exposed collagen in the disordered tumour vasculature. In comparison with the administration of unmodified IL-12, CBD-IL-12 induced sustained intratumoural levels of interferon-γ, substantially reduced its systemic levels as well as organ damage and provided superior anticancer efficacy, eliciting complete regression of CPI-unresponsive breast tumours. Furthermore, CBD-IL-12 potently synergized with CPI to eradicate large established melanomas, induced antigen-specific immunological memory and controlled tumour growth in a genetically engineered mouse model of melanoma. CBD-IL-12 may potentiate CPI immunotherapy for immunologically cold tumours.


Asunto(s)
Colágeno/metabolismo , Inflamación/patología , Interleucina-12/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epítopos/inmunología , Femenino , Inmunidad Innata/efectos de los fármacos , Interleucina-12/farmacología , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Inducción de Remisión , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
6.
Sci Adv ; 5(12): eaay1357, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31844672

RESUMEN

Although a clinical breakthrough for cancer treatment, it remains that a minority of patients respond to checkpoint inhibitor (CPI) immunotherapy. The composition of tumor-infiltrating immune cells has been identified as a key factor influencing CPI therapy success. Thus, enhancing tumor immune cell infiltration is a critical challenge. A lack of the chemokine CCL4 within the tumor microenvironment leads to the absence of CD103+ dendritic cells (DCs), a crucial cell population influencing CPI responsiveness. Here, we use a tumor stroma-targeting approach to deliver CCL4; by generating a fusion protein of CCL4 and the collagen-binding domain (CBD) of von Willebrand factor, we show that CBD fusion enhances CCL4 tumor localization. Intravenous CBD-CCL4 administration recruits CD103+ DCs and CD8+ T cells and improves the antitumor effect of CPI immunotherapy in multiple tumor models, including poor responders to CPI. Thus, CBD-CCL4 holds clinical translational potential by enhancing efficacy of CPI immunotherapy.


Asunto(s)
Antígenos CD/inmunología , Quimiocina CCL4/genética , Inmunoterapia , Cadenas alfa de Integrinas/inmunología , Melanoma Experimental/inmunología , Melanoma/inmunología , Animales , Antígenos CD/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Quimiocina CCL4/inmunología , Quimiocina CCL4/farmacología , Colágeno/genética , Colágeno/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Cadenas alfa de Integrinas/genética , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/genética , Melanoma/terapia , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Unión Proteica/genética , Microambiente Tumoral/efectos de los fármacos , Factor de von Willebrand/genética
7.
Sci Transl Med ; 11(487)2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971453

RESUMEN

Cancer immunotherapy with immune checkpoint inhibitors (CPIs) and interleukin-2 (IL-2) has demonstrated clinical efficacy but is frequently accompanied with severe adverse events caused by excessive and systemic immune system activation. Here, we addressed this need by targeting both the CPI antibodies anti-cytotoxic T lymphocyte antigen 4 antibody (αCTLA4) + anti-programmed death ligand 1 antibody (αPD-L1) and the cytokine IL-2 to tumors via conjugation (for the antibodies) or recombinant fusion (for the cytokine) to a collagen-binding domain (CBD) derived from the blood protein von Willebrand factor (VWF) A3 domain, harnessing the exposure of tumor stroma collagen to blood components due to the leakiness of the tumor vasculature. We show that intravenously administered CBD protein accumulated mainly in tumors. CBD conjugation or fusion decreases the systemic toxicity of both αCTLA4 + αPD-L1 combination therapy and IL-2, for example, eliminating hepatotoxicity with the CPI molecules and ameliorating pulmonary edema with IL-2. Both CBD-CPI and CBD-IL-2 suppressed tumor growth compared to their unmodified forms in multiple murine cancer models, and both CBD-CPI and CBD-IL-2 increased tumor-infiltrating CD8+ T cells. In an orthotopic breast cancer model, combination treatment with CPI and IL-2 eradicated tumors in 9 of 13 animals with the CBD-modified drugs, whereas it did so in only 1 of 13 animals with the unmodified drugs. Thus, the A3 domain of VWF can be used to improve safety and efficacy of systemically administered tumor drugs with high translational promise.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Colágeno/metabolismo , Citocinas/inmunología , Inmunoterapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Inmunidad , Inyecciones Intravenosas , Interleucina-2/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Dominios Proteicos , Resultado del Tratamiento
8.
Mol Cancer Ther ; 17(11): 2399-2411, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30097487

RESUMEN

CD40 is an immune costimulatory receptor expressed by antigen-presenting cells. Agonistic anti-CD40 antibodies have demonstrated considerable antitumor effects yet can also elicit serious treatment-related adverse events, such as liver toxicity, including in man. We engineered a variant that binds extracellular matrix through a super-affinity peptide derived from placenta growth factor-2 (PlGF-2123-144) to enhance anti-CD40's effects when administered locally. Peritumoral injection of PlGF-2123-144-anti-CD40 antibody showed prolonged tissue retention at the injection site and substantially decreased systemic exposure, resulting in decreased liver toxicity. In four mouse tumor models, PlGF-2123-144-anti-CD40 antibody demonstrated enhanced antitumor efficacy compared with its unmodified form and correlated with activated dendritic cells, B cells, and T cells in the tumor and in the tumor-draining lymph node. Moreover, in a genetically engineered BrafV600E ßCatSTA melanoma model that does not respond to checkpoint inhibitors, PlGF-2123-144-anti-CD40 antibody treatment enhanced T-cell infiltration into the tumors and slowed tumor growth. Together, these results demonstrate the marked therapeutic advantages of engineering matrix-binding domains onto agonistic anti-CD40 antibody as a therapeutic given by tumori-regional injection for cancer immunotherapy.Implications: Extracellular matrix-binding peptide conjugation to agonistic anti-CD40 antibody enhances antitumor efficacy and reduces treatment-related adverse events. Mol Cancer Ther; 17(11); 2399-411. ©2018 AACR.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Afinidad de Anticuerpos , Antígenos CD40/agonistas , Matriz Extracelular/metabolismo , Animales , Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/inmunología , Antígenos CD40/inmunología , Línea Celular Tumoral , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Humanos , Inmunidad , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Desnudos , Péptidos/química , Factor de Crecimiento Placentario/metabolismo , Linfocitos T/inmunología , beta Catenina/metabolismo
9.
Sci Transl Med ; 9(415)2017 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118259

RESUMEN

Immune checkpoint blockade exhibits considerable antitumor activity, but previous studies have reported instances of severe treatment-related adverse events. We sought to explore local immune checkpoint blockade, with an antibody (Ab) form that would be retained intra- or peritumorally, limiting systemic exposure. To accomplish this, we conjugated the checkpoint blockade Abs to an extracellular matrix (ECM)-super-affinity peptide derived from placenta growth factor-2 (PlGF-2123-144). We show enhanced tissue retention and lower Ab concentrations in blood plasma after PlGF-2123-144 conjugation, reducing systemic side effects such as the risk of autoimmune diabetes. Peritumoral injections of PlGF-2123-144-anti-CTLA4 (cytotoxic T lymphocyte antigen 4) and PlGF-2123-144-anti-PD-L1 (programmed death ligand 1) Abs delayed tumor growth and prolonged survival compared to the unmodified Abs in genetically engineered murine tumor models of melanoma and breast cancer. The PlGF-2123-144-Abs increased tumor-infiltrating activated CD8+ and CD4+ T cells, resulting in a delay of distant tumor growth as well. This simple and translatable approach of engineered ECM-binding Abs may present a viable and safer approach in checkpoint blockade.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Matriz Extracelular/metabolismo , Inmunoterapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Anticuerpos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4 , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Fibrinolisina/metabolismo , Ingeniería Genética , Heparina/metabolismo , Inmunidad , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Péptidos/farmacología , Péptidos/uso terapéutico , Factor de Crecimiento Placentario/química , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA