Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
NPJ Regen Med ; 8(1): 49, 2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37696884

RESUMO

Non-healing wounds have a negative impact on quality of life and account for many cases of amputation and even early death among patients. Diabetic patients are the predominate population affected by these non-healing wounds. Despite the significant clinical demand, treatment with biologics has not broadly impacted clinical care. Interleukin-4 (IL-4) is a potent modulator of the immune system, capable of skewing macrophages towards a pro-regeneration phenotype (M2) and promoting angiogenesis, but can be toxic after frequent administration and is limited by its short half-life and low bioavailability. Here, we demonstrate the design and characterization of an engineered recombinant interleukin-4 construct. We utilize this collagen-binding, serum albumin-fused IL-4 variant (CBD-SA-IL-4) delivered in a hyaluronic acid (HA)-based gel for localized application of IL-4 to dermal wounds in a type 2 diabetic mouse model known for poor healing as proof-of-concept for improved tissue repair. Our studies indicate that CBD-SA-IL-4 is retained within the wound and can modulate the wound microenvironment through induction of M2 macrophages and angiogenesis. CBD-SA-IL-4 treatment significantly accelerated wound healing compared to native IL-4 and HA vehicle treatment without inducing systemic side effects. This CBD-SA-IL-4 construct can address the underlying immune dysfunction present in the non-healing wound, leading to more effective tissue healing in the clinic.

3.
Arthritis Rheumatol ; 73(5): 769-778, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33169522

RESUMO

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.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Articulações do Pé/efeitos dos fármacos , Interleucina-10/farmacologia , Linfonodos/imunologia , Macrófagos/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Albumina Sérica/farmacologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Artrite Experimental/metabolismo , Artrite Reumatoide/metabolismo , Modelos Animais de Doenças , , Articulações do Pé/imunologia , Articulações do Pé/metabolismo , Articulações do Pé/patologia , Membro Posterior , Antígenos de Histocompatibilidade Classe I/metabolismo , Injeções Intravenosas , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucina-6/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Camundongos , Engenharia de Proteínas , Transporte Proteico , Receptores Fc/metabolismo , Fator de Crescimento Transformador beta/efeitos dos fármacos , Fator de Crescimento Transformador beta/imunologia , Inibidores do Fator de Necrose Tumoral/farmacologia
4.
Nat Biomed Eng ; 5(5): 387-398, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33046864

RESUMO

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.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Imunossupressores/administração & dosagem , Interleucina-4/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Albumina Sérica/metabolismo , Animais , Encefalomielite Autoimune Experimental/imunologia , Feminino , Meia-Vida , Imunossupressores/farmacocinética , Imunossupressores/farmacologia , Injeções Intravenosas , Linfonodos/química , Linfonodos/imunologia , Camundongos , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Baço/química , Baço/imunologia , Células Th17/efeitos dos fármacos
6.
Nat Biomed Eng ; 4(5): 531-543, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32284554

RESUMO

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.


Assuntos
Colágeno/metabolismo , Inflamação/patologia , Interleucina-12/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Epitopos/imunologia , Feminino , Imunidade Inata/efeitos dos fármacos , Interleucina-12/farmacologia , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Indução de Remissão , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
7.
Arthritis Res Ther ; 21(1): 298, 2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31870429

RESUMO

BACKGROUND: Although disease in a majority of rheumatoid arthritis (RA) patients is often initially limited to one or a few joints, currently approved medications including anti-tumor necrosis factor-α antibody (α-TNF) are injected systemically. Given that α-TNF systemic injection typically does not cure RA and involves risk of treatment-related adverse events, one possible approach to enhance therapeutic efficacy and reduce α-TNF systemic exposure is to retain the antibodies in arthritic joints after local administration. The aim of this study was to evaluate the approach of conferring extracellular matrix (ECM) binding affinity to α-TNF antibodies in a RA model. METHODS: α-TNF was chemically conjugated with a promiscuous ECM-binding peptide derived from placenta growth factor 2 (PlGF-2123-144). The binding activity of PlGF-2123-144-conjugated α-TNF (PlGF-2123-144-α-TNF) against ECM proteins was assessed by ELISA and by immunostaining on human cartilage specimens. The effect of conjugation on antibody function was assessed as a neutralizing activity against osteoclast differentiation. Retention at the injection site and therapeutic efficacy of PlGF-2123-144-α-TNF were tested in a collagen antibody-induced arthritis (CAIA) model in the mouse. RESULTS: PlGF-2123-144 peptide conjugation conferred α-TNF with affinity to ECM proteins without impairment of antigen recognition. PlGF-2123-144-α-TNF locally injected at a paw in the CAIA model was retained for at least 96 h at the injection site, whereas unmodified α-TNF was dispersed rapidly after injection. Local treatment with unmodified α-TNF did not suppress the arthritis score relative to isotype controls. By contrast, local administration of PlGF-2123-144-α-TNF suppressed arthritis development almost completely in the treated paw even at a 1000× lower dose. CONCLUSION: These data demonstrate that retention of α-TNF in arthritic joints can suppress arthritis development and enhance therapeutic efficacy. This simple bioengineering approach of ECM-binding peptide conjugation offers a powerful and clinically translational approach to treat RA.


Assuntos
Anticorpos/imunologia , Artrite Reumatoide/imunologia , Matriz Extracelular/imunologia , Imunoconjugados/imunologia , Fator de Crescimento Placentário/imunologia , Fator de Necrose Tumoral alfa/imunologia , Sequência de Aminoácidos , Animais , Anticorpos/metabolismo , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Experimental/prevenção & controle , Artrite Reumatoide/metabolismo , Artrite Reumatoide/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Humanos , Imunoconjugados/metabolismo , Imunoconjugados/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Fator de Crescimento Placentário/química , Fator de Crescimento Placentário/metabolismo , Células RAW 264.7 , Fator de Necrose Tumoral alfa/metabolismo
8.
Sci Adv ; 5(12): eaay1357, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31844672

RESUMO

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.


Assuntos
Antígenos CD/imunologia , Quimiocina CCL4/genética , Imunoterapia , Cadeias alfa de Integrinas/imunologia , Melanoma Experimental/imunologia , Melanoma/imunologia , Animais , Antígenos CD/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/imunologia , Quimiocina CCL4/imunologia , Quimiocina CCL4/farmacologia , Colágeno/genética , Colágeno/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Humanos , Cadeias alfa de Integrinas/genética , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Melanoma/genética , Melanoma/terapia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Ligação Proteica/genética , Microambiente Tumoral/efeitos dos fármacos , Fator de von Willebrand/genética
9.
Sci Adv ; 5(11): eaay1971, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31723606

RESUMO

Enhancing the therapeutic efficacy of drugs for inflammatory diseases is of high demand. One possible approach is targeting drugs to the extracellular matrix of the inflamed area. Here, we target collagens in the matrix, which are inaccessible in most tissues yet are exposed to the bloodstream in the inflamed area because of vascular hyperpermeability. We conferred collagen affinity to anti-tumor necrosis factor-α (α-TNF) antibody by conjugating a collagen-binding peptide (CBP) derived from the sequence of decorin. CBP-α-TNF accumulated in the inflamed paw of the arthritis model, and arthritis development was significantly suppressed by treatment with CBP-α-TNF compared with the unmodified antibody. Similarly, CBP-anti-transforming growth factor-ß (α-TGF-ß) accumulated in the inflamed lung of pulmonary fibrosis model and significantly suppressed pulmonary fibrosis compared with the unmodified antibody. Together, collagen affinity enables the anticytokine antibodies to target arthritis and pulmonary fibrosis accompanied by inflammation, demonstrating a clinically translational approach to treat inflammatory diseases.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticorpos/uso terapêutico , Colágeno/antagonistas & inibidores , Inflamação/tratamento farmacológico , Pulmão/efeitos dos fármacos , Fibrose Pulmonar/tratamento farmacológico , Animais , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/metabolismo , Anticorpos/imunologia , Anticorpos/metabolismo , Colágeno/imunologia , Colágeno/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Pulmão/imunologia , Pulmão/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular/métodos , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/metabolismo , Sialoglicoproteínas/imunologia , Sialoglicoproteínas/metabolismo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
10.
Sci Adv ; 5(8): eaaw6081, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31453327

RESUMO

Serum albumin (SA) is used as a carrier to deliver cytotoxic agents to tumors via passive targeting. To further improve SA's tumor targeting capacity, we sought to develop an approach to retain SA-drug conjugates within tumors through a combination of passive and active targeting. SA was recombinantly fused with a collagen-binding domain (CBD) of von Willebrand factor to bind within the tumor stroma after extravasation due to tumor vascular permeability. Doxorubicin (Dox) was conjugated to the CBD-SA via a pH-sensitive linker. Dox-CBD-SA treatment significantly suppressed tumor growth compared to both Dox-SA and aldoxorubicin treatment in a mouse model of breast cancer. Dox-CBD-SA efficiently stimulated host antitumor immunity, resulting in the complete eradication of MC38 colon carcinoma when used in combination with anti-PD-1 checkpoint inhibitor. Dox-CBD-SA decreased adverse events compared to aldoxorubicin. Thus, engineered CBD-SA could be a versatile and clinically relevant drug conjugate carrier protein for treatment of solid tumors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Albumina Sérica/química , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/análogos & derivados , Doxorrubicina/química , Feminino , Hidrazonas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Domínios Proteicos , Fator de von Willebrand/química
11.
Blood ; 133(24): 2559-2569, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-30975637

RESUMO

During wound healing, the distribution, availability, and signaling of growth factors (GFs) are orchestrated by their binding to extracellular matrix components in the wound microenvironment. Extracellular matrix proteins have been shown to modulate angiogenesis and promote wound healing through GF binding. The hemostatic protein von Willebrand factor (VWF) released by endothelial cells (ECs) in plasma and in the subendothelial matrix has been shown to regulate angiogenesis; this function is relevant to patients in whom VWF deficiency or dysfunction is associated with vascular malformations. Here, we show that VWF deficiency in mice causes delayed wound healing accompanied by decreased angiogenesis and decreased amounts of angiogenic GFs in the wound. We show that in vitro VWF binds to several GFs, including vascular endothelial growth factor-A (VEGF-A) isoforms and platelet-derived growth factor-BB (PDGF-BB), mainly through the heparin-binding domain (HBD) within the VWF A1 domain. VWF also binds to VEGF-A and fibroblast growth factor-2 (FGF-2) in human plasma and colocalizes with VEGF-A in ECs. Incorporation of the VWF A1 HBD into fibrin matrices enables sequestration and slow release of incorporated GFs. In vivo, VWF A1 HBD-functionalized fibrin matrices increased angiogenesis and GF retention in VWF-deficient mice. Treatment of chronic skin wounds in diabetic mice with VEGF-A165 and PDGF-BB incorporated within VWF A1 HBD-functionalized fibrin matrices accelerated wound healing, with increased angiogenesis and smooth muscle cell proliferation. Therefore, the VWF A1 HBD can function as a GF reservoir, leading to effective angiogenesis and tissue regeneration.


Assuntos
Neovascularização Fisiológica/fisiologia , Cicatrização/fisiologia , Fator de von Willebrand/metabolismo , Animais , Diabetes Mellitus Experimental , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Domínios Proteicos
12.
Sci Transl Med ; 11(487)2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30971453

RESUMO

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.


Assuntos
Anticorpos Antineoplásicos/imunologia , Colágeno/metabolismo , Citocinas/imunologia , Imunoterapia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Citotoxicidade Imunológica , Modelos Animais de Doenças , Humanos , Imunidade , Injeções Intravenosas , Interleucina-2/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Domínios Proteicos , Resultado do Tratamento
13.
Mol Cancer Ther ; 17(11): 2399-2411, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30097487

RESUMO

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.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Afinidade de Anticorpos , Antígenos CD40/agonistas , Matriz Extracelular/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos B/imunologia , Antígenos CD40/imunologia , Linhagem Celular Tumoral , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Humanos , Imunidade , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Camundongos Nus , Peptídeos/química , Fator de Crescimento Placentário/metabolismo , Linfócitos T/imunologia , beta Catenina/metabolismo
14.
Nat Commun ; 9(1): 2163, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29867149

RESUMO

Laminin, as a key component of the basement membrane extracellular matrix (ECM), regulates tissue morphogenesis. Here, we show that multiple laminin isoforms promiscuously bind to growth factors (GFs) with high affinity, through their heparin-binding domains (HBDs) located in the α chain laminin-type G (LG) domains. These domains also bind to syndecan cell-surface receptors, promoting attachment of fibroblasts and endothelial cells. We explore the application of these multifunctional laminin HBDs in wound healing in the type-2 diabetic mouse. We demonstrate that covalent incorporation of laminin HBDs into fibrin matrices improves retention of GFs and significantly enhances the efficacy of vascular endothelial cell growth factor (VEGF-A165) and platelet-derived growth factor (PDGF-BB) in promoting wound healing in vivo, under conditions where the GFs alone in fibrin are inefficacious. This laminin HBD peptide may be clinically useful by improving biomaterial matrices as both GF reservoirs and cell scaffolds, leading to effective tissue regeneration.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Heparina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Laminina/metabolismo , Peptídeos/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Fibrina , Humanos , Masculino , Camundongos Endogâmicos C57BL , Ligação Proteica , Pele/lesões , Cicatrização
15.
Sci Transl Med ; 9(415)2017 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118259

RESUMO

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.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Matriz Extracelular/metabolismo , Imunoterapia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Anticorpos/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4 , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/metabolismo , Fibrinolisina/metabolismo , Engenharia Genética , Heparina/metabolismo , Imunidade , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Fator de Crescimento Placentário/química , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...