Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Int Immunopharmacol ; 120: 110255, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37187126

RESUMO

Cluster of differentiation 47(CD47) is a transmembrane protein that is ubiquitously found on the surface of many cells in the body and uniquely overexpressed by both solid and hematologic malignant cells. CD47 interacts with signal-regulatory protein α (SIRPα), to trigger a "don't eat me" signal and thereby achieve cancer immune escape by inhibiting macrophage-mediated phagocytosis. Thus, blocking the CD47-SIRPα phagocytosis checkpoint, for release of the innate immune system, is a current research focus. Indeed, targeting the CD47-SIRPα axis as a cancer immunotherapy has shown promising efficacies in pre-clinical outcomes. Here, we first reviewed the origin, structure, and function of the CD47-SIRPα axis. Then, we reviewed its role as a target for cancer immunotherapies, as well as the factors regulating CD47-SIRPα axis-based immunotherapies. We specifically focused on the mechanism and progress of CD47-SIRPα axis-based immunotherapies and their combination with other treatment strategies. Finally, we discussed the challenges and directions for future research and identified potential CD47-SIRPα axis-based therapies that are suitable for clinical application.


Assuntos
Antígeno CD47 , Neoplasias , Humanos , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Imunoterapia , Macrófagos , Neoplasias/tratamento farmacológico , Fagocitose
2.
J Cereb Blood Flow Metab ; 43(9): 1475-1489, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37113078

RESUMO

Both monocyte-derived macrophages (MDMs) and brain resident microglia participate in hematoma resolution after intracerebral hemorrhage (ICH). Here, we utilized a transgenic mouse line with enhanced green fluorescent protein (EGFP) labeled microglia (Tmem119-EGFP mice) combined with a F4/80 immunohistochemistry (a pan-macrophage marker) to visualize changes in MDMs and microglia after ICH. A murine model of ICH was used in which autologous blood was stereotactically injected into the right basal ganglia. The autologous blood was co-injected with CD47 blocking antibodies to enhance phagocytosis or clodronate liposomes for phagocyte depletion. In addition, Tmem119-EGFP mice were injected with the blood components peroxiredoxin 2 (Prx2) or thrombin. MDMs entered the brain and formed a peri-hematoma cell layer by day 3 after ICH and giant phagocytes engulfed red blood cells were found. CD47 blocking antibody increased the number of MDMs around and inside the hematoma and extended MDM phagocytic activity to day 7. Both MDMs and microglia could be diminished by clodronate liposomes. Intracerebral injection of Prx2 but not thrombin attracted MDMs into brain parenchyma. In conclusion, MDMs play an important role in phagocytosis after ICH which can be enhanced by CD47 blocking antibody, suggesting the modulation of MDMs after ICH could be a future therapeutic target.


Assuntos
Antígeno CD47 , Microglia , Camundongos , Animais , Microglia/metabolismo , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Ácido Clodrônico/farmacologia , Ácido Clodrônico/metabolismo , Ácido Clodrônico/uso terapêutico , Lipossomos/metabolismo , Macrófagos/metabolismo , Hemorragia Cerebral/metabolismo , Camundongos Transgênicos , Hematoma/metabolismo
3.
Int J Biol Macromol ; 240: 124342, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37030459

RESUMO

Tumor immunotherapy hold great promise for eradicating tumors. However, immune escape and the immunosuppressive microenvironment of tumor usually limit the efficiency of tumor immunotherapy. Therefore, simultaneously blocking immune escape and improving immunosuppressive microenvironment are the current problems to be solved urgently. Among them, CD47 on cancer cells membrane could bind to signal regulatory protein α (SIRPα) on macrophages membrane and sent out "don't eat me" signal, which was an important pathway of immune escape. The large number of M2-type macrophages in tumor microenvironment was a significant factor contributing to the immunosuppressive microenvironment. Here, we present a drug loading system for enhancing cancer immunotherapy, comprising CD47 antibody (aCD47) and chloroquine (CQ) with bionic lipoprotein (BLP) carrier (BLP-CQ-aCD47). On the one hand, as drug delivery carrier, BLP could allow CQ to be preferentially taken up by M2-type macrophages, thereby efficiently polarized M2-type tumor-promoting cells into M1-type anti-tumor cells. On the other hand, blocking CD47 from binding to SIRPα could block the "don't eat me" signal, and improve the phagocytosis of macrophages to tumor cells. Taken together, BLP-CQ-aCD47 could block immune escape, improve immunosuppressive microenvironment of tumor, and induce a strong immune response without substantial systemic toxicity. Therefore, it provides a new idea for tumor immunotherapy.


Assuntos
Antígeno CD47 , Neoplasias , Humanos , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Biônica , Antígenos de Diferenciação , Receptores Imunológicos/metabolismo , Fagocitose , Neoplasias/tratamento farmacológico , Lipoproteínas , Imunoterapia , Microambiente Tumoral
4.
Free Radic Biol Med ; 201: 26-40, 2023 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-36907254

RESUMO

Cold atmospheric plasma-treated liquids (PTLs) exhibit selective toxicity toward tumor cells and are provoked by a cocktail of reactive oxygen and nitrogen species in such liquids. Compared to the gaseous phase, these reactive species are more persistent in the aqueous phase. This indirect plasma treatment method has gradually gathered interest in the discipline of plasma medicine to treat cancer. PTL's motivated effect on immunosuppressive proteins and immunogenic cell death (ICD) in solid cancer cells is still not explored. In this study, we aimed to induce immunomodulation by plasma-treated Ringer's lactate (PT-RL) and phosphate-buffered saline (PT-PBS) solutions for cancer treatment. PTLs induced minimum cytotoxicity in normal lung cells and inhibited cancer cell growth. ICD is confirmed by the enhanced expression of damage-associated molecular patterns (DAMPs). We evidenced that PTLs induce intracellular nitrogen oxide species accumulation and elevate immunogenicity in cancer cells owing to the production of pro-inflammatory cytokines, DAMPs, and reduced immunosuppressive protein CD47 expression. In addition, PTLs influenced A549 cells to elevate the organelles (mitochondria and lysosomes) in macrophages. Taken together, we have developed a therapeutic approach to potentially facilitate the selection of a suitable candidate for direct clinical applications.


Assuntos
Carcinoma , Neoplasias Pulmonares , Gases em Plasma , Humanos , Argônio/uso terapêutico , Antígeno CD47/uso terapêutico , Morte Celular Imunogênica , Neoplasias Pulmonares/tratamento farmacológico , Gases em Plasma/farmacologia , Gases em Plasma/uso terapêutico , Pulmão
5.
Cancer Control ; 30: 10732748231159706, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36826231

RESUMO

Among the three primary gynecological malignancies, ovarian cancer has the lowest incidence but the worst prognosis. Because of the poor prognosis of ovarian cancer patients treated with existing treatments, immunotherapy is emerging as a potentially ideal alternative to surgery, chemotherapy, and targeted therapy. Among immunotherapies, immune checkpoint inhibitors have been the most thoroughly studied, and many drugs have been successfully used in the clinic. CD47, a novel immune checkpoint, provides insights into ovarian cancer immunotherapy. This review highlights the mechanisms of tumor immune evasion via CD47-mediated inhibition of phagocytosis and provides a comprehensive insight into the progress of the relevant targeted agents in ovarian cancer.


Assuntos
Antineoplásicos , Neoplasias , Neoplasias Ovarianas , Humanos , Feminino , Antígeno CD47/uso terapêutico , Fagocitose , Imunoterapia , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico
6.
Drug Dev Res ; 84(2): 238-252, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36598070

RESUMO

Non-small cell lung cancer (NSCLC) is one of the most common malignancies with high morbidity and mortality. PKHB1, a serum-stable Thrombospondin-1 (TSP-1) mimic peptide, has shown some effective ability in triggering cell death against several cancers. Here, we aimed to study the potential biological function of PKHB1 and its molecular mechanism in NSCLC. Our results revealed that PKHB1 significantly suppressed NSCLC cell proliferation, cell migration, and induced apoptosis in a dose-dependent manner. Additionally, we found that PKHB1 treatment resulted in mitochondrial transmembrane potential depolarization, Ca2+ overloading as well as the upregulation of proapoptotic proteins. Mechanistically, PKHB1 induced NSCLC cells apoptosis in a CD47-independent manner. Further study revealed that PKHB1 provoked endoplasmic reticulum (ER) stress principally through the activation of CHOP and JNK signaling, which could be alleviated in the presence of 4-PBA, an ER stress inhibitor. Furthermore, xenograft tumor models showed that PKHB1 treatment could notably inhibit NSCLC tumor growth in vivo. In conclusion, these findings suggested that PKHB1 exerted antitumor efficacy in NSCLC via triggering ER stress-mediated but CD47-independent apoptosis, potentially functioned as a promising peptide-based therapeutic agent for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Trombospondina 1/farmacologia , Trombospondina 1/uso terapêutico , Antígeno CD47/uso terapêutico , Apoptose , Estresse do Retículo Endoplasmático , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Linhagem Celular Tumoral
7.
Orthop Surg ; 15(3): 829-838, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36519392

RESUMO

OBJECTIVES: The fact that studies on anti-programmed cell death 1 (PD-1) or its relevant ligand 1 (PD-L1) have yielded such few responses greatly decreases the confidence in immunotherapy with checkpoint inhibitors for advanced osteosarcoma. We intended to characterize the expression of various checkpoint molecules with immunohistochemistry in osteosarcoma specimens and analyzed the relationship of the expression of these checkpoint molecules with patients' clinical courses. METHODS: This study was a retrospective non-intervention study from August 1st 2017 to March 1st 2020. Immunohistochemistry for B7-H3 (CD276, Cluster of Differentiation 276), CD47 (Cluster of Differentiation 47), PD-L1 (programmed cell death ligand 1), TIM3 (mucin-domain containing-3), TGF-ß (TransformingGrowth Factor ß), CXCR 4 (Chemokine Receptor 4), CD27 (Cluster of Differentiation 27), IDO1 (Indoleamine 2,3-dioxygenase 1), KIRs (Killer cell Immunoglobulin-like Receptors), and SDF-1 (Stromal cell-Derived Factor-1) was performed on 35 resected osteosarcoma specimens. Patients progressed upon first-line chemotherapy with evaluable lesions were qualified for this study, and their specimens previously stored in the pathological department repository would be retrieved for analysis. Associations between the immunohischemistry markers and clinicopathological variables and survival were evaluated by the χ2 displayed by cross-table, Cox proportional hazards regression model, and Kaplan-Meier plots. RESULTS: The positive rates of B7-H3, CD47, PD-L1, TIM3, and TGF-ß expression in this sample of 35 heavily treated osteosarcomas were 29% (10/35), 15% (5/35), 9% (3/35), 6% (2/35), and 6% (2/35), respectively, and diverse staining intensities were observed. Among these advanced patients, 15/35 (43%) had positive checkpoint expression, of which 33% (5/15) showed evidence of the co-expression of more than one checkpoint molecule. We did not find any obvious correlation with clinicopathological characteristics and the positive expression of these molecules. CONCLUSIONS: The present study highlights that only a small subset of progressive osteosarcomas, which had been heavily-treated, expressed tumor immune-associated checkpoint molecules, of which B7-H3 was the most positively expressed checkpoint and might be a promising target for further osteosarcoma investigation.


Assuntos
Neoplasias Ósseas , Neoplasias Pulmonares , Osteossarcoma , Humanos , Neoplasias Pulmonares/patologia , Antígeno B7-H1/análise , Antígeno B7-H1/metabolismo , Antígeno CD47/uso terapêutico , Estudos Retrospectivos , Relevância Clínica , Receptor Celular 2 do Vírus da Hepatite A/uso terapêutico , Osteossarcoma/patologia , Neoplasias Ósseas/patologia , Imunoterapia , Antígenos B7
8.
Front Med ; 17(1): 105-118, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36414917

RESUMO

The third-generation epidermal growth factor receptor (EGFR) inhibitor osimertinib (OSI) has been approved as the first-line treatment for EGFR-mutant non-small cell lung cancer (NSCLC). This study aims to explore a rational combination strategy for enhancing the OSI efficacy. In this study, OSI induced higher CD47 expression, an important anti-phagocytic immune checkpoint, via the NF-κB pathway in EGFR-mutant NSCLC HCC827 and NCI-H1975 cells. The combination treatment of OSI and the anti-CD47 antibody exhibited dramatically increasing phagocytosis in HCC827 and NCI-H1975 cells, which highly relied on the antibody-dependent cellular phagocytosis effect. Consistently, the enhanced phagocytosis index from combination treatment was reversed in CD47 knockout HCC827 cells. Meanwhile, combining the anti-CD47 antibody significantly augmented the anticancer effect of OSI in HCC827 xenograft mice model. Notably, OSI induced the surface exposure of "eat me" signal calreticulin and reduced the expression of immune-inhibitory receptor PD-L1 in cancer cells, which might contribute to the increased phagocytosis on cancer cells pretreated with OSI. In summary, these findings suggest the multidimensional regulation by OSI and encourage the further exploration of combining anti-CD47 antibody with OSI as a new strategy to enhance the anticancer efficacy in EGFR-mutant NSCLC with CD47 activation induced by OSI.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Camundongos , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Acrilamidas/farmacologia , Receptores ErbB/metabolismo , Linhagem Celular Tumoral , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico
9.
Curr Pharm Des ; 28(46): 3720-3728, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36201266

RESUMO

Metabolic disorders include a cluster of conditions that result from hyperglycemia, hyperlipidemia, insulin resistance, obesity, and hepatic steatosis, which cause the dysfunction of immune cells and innate cells, such as macrophages, natural killer cells, vascular endothelial cells, hepatocytes, and human kidney tubular epithelial cells. Besides targeting the derangements in lipid metabolism, therapeutic modulations to regulate abnormal responses in the immune system and innate cell dysfunctions may prove to be promising strategies in the management of metabolic diseases. In recent years, several targets have been explored for the CD47 molecule (CD47), a glycosylated protein, which was originally reported to transmit an anti-phagocytic signal known as "don't eat me" in the atherosclerotic environment, hindering the efferocytosis of immune cells and promoting arterial plaque accumulation. Subsequently, the role of CD47 has been explored in obesity, fatty liver, and lipotoxic nephropathy, and its utility as a therapeutic target has been investigated using anti-CD47 antibodies or inhibitors of the THBS1/CD47 axis and the CD47/SIRPα signaling pathway. This review summarizes the mechanisms of action of CD47 in different cell types during metabolic diseases and the clinical research progress to date, providing a reference for the comprehensive targeting of CD47 to treat metabolic diseases and the devising of potential improvements to possible side effects.


Assuntos
Células Endoteliais , Neoplasias , Humanos , Células Endoteliais/metabolismo , Fagocitose , Macrófagos/metabolismo , Imunoterapia , Neoplasias/tratamento farmacológico , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico
10.
Thorac Cancer ; 13(19): 2723-2731, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36054073

RESUMO

A cluster of differentiation 47 (CD47) and immune-modulatory protein for myeloid cells has been implicated in cisplatin (CDDP) resistance. Exosome delivery of drugs has shown great potential for targeted drug delivery in the treatment of various diseases. In the current study, we explored the approach of co-delivering CDDP and CD47 antibody with MDA-MB-231 cell-derived exosome 231-exo (CaCE) and assessed the phagocytosis activity of bone marrow flow cytometry derived macrophages (BMDM) against co-cultured A549 cells. CD8+ T-cell proliferation was examined with flow cytometry analysis. In vivo, we used the Lewis lung carcinoma (LLC) tumor-bearing mouse model and assessed survival rate, tumor weight, phagocytosis, and T-cell proliferation, as well as cytokine levels in tumors analyzed by enzyme-linked immunoassay (ELISA). Although co-administration of CDDP with anti-CD47 (CDDP and aCD47) showed a significant antitumor effect, CaCE had an even more dramatic anticancer effect in survival rate and tumor weight. We observed increased phagocytosis activity selectively against lung tumor cells in vivo and in vitro with exosome CaCE treatment. CaCE treatment also increased T-cell proliferation compared to the vehicle treatment and co-administration groups. Furthermore, immunostimulatory interleukin (IL)-12p and interferon (IFN)-γ were increased, whereas transforming growth factor ß (TGF-ß) were decreased, indicating the improved CDDP anticancer effect is related to a tumor microenvironmental change. Our study demonstrates a dramatically improved anticancer effect of CDDP when administered by exosome co-delivery with anti-CD47.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Exossomos , Neoplasias Pulmonares , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Citocinas , Exossomos/metabolismo , Humanos , Interferons/uso terapêutico , Pulmão/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Fator de Crescimento Transformador beta
11.
Expert Opin Pharmacother ; 23(13): 1535-1543, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35938317

RESUMO

INTRODUCTION: With advances in molecular medicine and precision approaches, there has been significant improvement in the treatment of acute myeloid leukemia (AML) in recent years. This reflects better understanding of molecular and metabolic pathways in leukemia cells, including BCL2 upregulation that prevents apoptosis, FLT3 tyrosine kinase activating mutations that allow uncontrolled proliferation, and IDH mutations that result in differentiation block. AREAS COVERED: We performed a compressive review of important pre-clinical studies in AML that involve major molecular and metabolic pathways in AML, and we discussed standard therapeutic modalities and ongoing clinical trials for patients with AML, as well as an overall update of recent efforts in this area. EXPERT OPINION: Targeting these pathways has resulted in improvement in the overall survival of some groups of AML patients. Secondary AML and TP53 mutated AML remain challenging subtypes of AML with limited treatment options and represent areas of unmet research need. Ongoing work with menin inhibitors in MLL rearranged leukemia, which comprise a large portion of secondary AML cases, the development of CAR T cell products and targeting the CD47 receptor on macrophages in myeloid neoplasms including in TP53 mutated AML have provided hope for these challenging subtypes of AML.


Assuntos
Antígeno CD47 , Leucemia Mieloide Aguda , Adulto , Antígeno CD47/genética , Antígeno CD47/uso terapêutico , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Mutação , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases , Proteínas Proto-Oncogênicas c-bcl-2/genética
12.
Eur J Pharmacol ; 931: 175178, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35948163

RESUMO

Glutaminyl cyclases (QC) catalyze the cyclization of proteins and turn N-terminal glutamine or glutamic acid into N-terminal pyroglutamate, resulting in protection of proteins from aminopeptidases and an increase of their stabilities. The aberrant N-terminal pyroglutamate has been found in various diseases, including Alzheimer's disease (AD), Huntington's disease (HD) and cancer. Two kinds of human QC, the secretory sQC and the Golgi resident gQC, are identified to date. Several substrates of sQC involving beta amyloid (Aß), Huntington (HTT) protein and certain inflammatory mediators such as CCL2 and CX3CL1 have been observed to associate with neurodegenerative diseases and cancers. The Golgi resident gQC can modify N-terminus of CD47 that directly influences the interaction of CD47 and SIRPα resulting in the modulations of the immunological surveillance related mechanisms in cancer. Additionally, inflammatory chemokines CCL2 and CX3CL1 can also be modified by gQC. Several QC inhibitors with differential scaffold structures have been developed and investigated. Among these QC inhibitors, PQ912, a benzimidazole-based inhibitor, has been studied in a phase II clinical trial to treat AD. In this review, we will summarize the current knowledge about QCs' tissue expression patterns, their potential cellular substrates in the context of cancers, AD and HD. After introducing QCs' molecular structures and catalysis mechanisms, the structures and efficacies of the currently reported QCs' inhibitors will also be summarized.


Assuntos
Doença de Alzheimer , Aminoaciltransferases , Neoplasias , Doenças Neurodegenerativas , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Aminoaciltransferases/química , Aminoaciltransferases/metabolismo , Aminoaciltransferases/uso terapêutico , Peptídeos beta-Amiloides , Antígeno CD47/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Ácido Pirrolidonocarboxílico/metabolismo , Ácido Pirrolidonocarboxílico/uso terapêutico
13.
Clin Transl Med ; 12(8): e943, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35908284

RESUMO

BACKGROUND: The CD47-signal regulatory protein alpha (SIRPα) 'don't eat me' signalling axis is perhaps the most prominent innate immune checkpoint to date. However, from initial clinical trials, it is evident that monotherapy with CD47-SIRPα blocking has a limited therapeutic effect at the maximum tolerated dose. Furthermore, treatment is associated with severe side effects, most notably anaemia, that are attributable to the ubiquitous expression of CD47. Nevertheless, promising clinical responses have been reported upon combination with the tumour-targeting antibody rituximab or azacytidine, although toxicity issues still hamper clinical application. MAIN BODY: Here, we discuss the current state of CD47-SIRPα blocking therapy with a focus on limitations of current strategies, such as depletion of red blood cells. Subsequently, we focus on innovations designed to overcome these limitations. These include novel antibody formats designed to selectively target CD47 on tumour cells as well as tumour-targeted bispecific antibodies with improved selectivity. In addition, the rationale and outcome of combinatorial approaches to improve the therapeutic effect of CD47 blockade are discussed. Such combinations include those with tumour-targeted opsonizing antibodies, systemic therapy, epigenetic drugs, other immunomodulatory T-cell-targeted therapeutics or dual immunomodulatory CD47 bispecific antibodies. CONCLUSION: With these advances in the design of CD47-SIRPα-targeting therapeutic strategies and increasing insight into the mechanism of action of this innate checkpoint, including the role of adaptive immunity, further advances in the clinical application of this checkpoint can be anticipated.


Assuntos
Anticorpos Biespecíficos , Antígenos de Diferenciação/metabolismo , Neoplasias , Receptores Imunológicos/metabolismo , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Antígenos de Diferenciação/farmacologia , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Imunoterapia , Neoplasias/tratamento farmacológico , Fagocitose , Receptores Imunológicos/uso terapêutico
14.
Colloids Surf B Biointerfaces ; 217: 112609, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35667200

RESUMO

Acute myeloid leukemia (AML), a malignant disorder of Hematopoietic stem cells, can escape immunosurveillance by over expression of the cluster of differentiation 47 (CD47) marker, which functions as an inhibitory signal, suppressing phagocytosis by binding to signal regulatory protein α (SIRPα) on macrophages. AML is treated mainly by chemotherapy, which has drastic side effects and poor outcomes for the patients. Most AML patients develop drug resistance, so other methods to treat AML are highly required. Small interfering RNA (siRNA) is considered as an antitumor therapeutic due to its ability to silence genes associated with the overexpressed cancer markers and subsequently re-sensitize cancer cells. However, delivering siRNA into cells faces challenges, and the development of an effective delivery system is desired for successful silencing at the gene level. Herein, we report the usage of different formulations of graphene oxide (GO) as carriers for the delivery of CD47_siRNA (siRNA against CD47) into AML cells in vitro. The polyethylene glycol (PEG) and dendrimers (PAMAM) modified GO with small flake sizes achieved the highest silencing efficiency of the anti-phagocytosis marker CD47 gene, resulted CD47 protein down-regulation in AML cells. Moreover, the concentration at which the GO-based formulations was used has shown no cytotoxicity in AML cells or normal blood cells, which could be used to screen potential drugs for targeted gene therapy in AML.


Assuntos
Antígeno CD47 , Leucemia Mieloide Aguda , Antígenos CD , Antígenos de Diferenciação/farmacologia , Antígenos de Diferenciação/uso terapêutico , Antígeno CD47/genética , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Fagocitose , RNA Interferente Pequeno
15.
Biofactors ; 48(5): 1137-1144, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35533068

RESUMO

Colorectal cancer is one of the major concerns in both developed and developing societies. Because of the serious side effects of the current treatments, novel therapy agents have been developed that target immune checkpoint and immunomodulatory molecules in the tumor environment. Therefore, this study investigates the effect of docosahexaenoic acid (DHA) fatty acid on the expression of immune checkpoint molecule, PD-L1, and immunomodulatory molecules, CD47 and CD39, and their controlling miRNAs in the colorectal cancer cell lines. Human colorectal cell lines HT-29 and Caco-2 were treated with 100 µM DHA and 50 µM LA for 24 h under the normoxic and hypoxic conditions. Total RNA was extracted and the qRT-PCR was performed to analyze the expression of the studied genes and miRNAs. The western blotting technique was also used for validation. The qRT-PCR results showed that DHA treatment decreased the expression of the PD-L1, CD47, and CD39 genes, but decreases these genes controlling miRNAs, mir-424, mir-133a, and mir-142, respectively. Western blotting analysis demonstrated that PD-L1 protein expression decreased after DHA treatment. LA administration had no inhibitory effect on the studied genes. This study showed that DHA may have anti-cancer properties by downregulation of proteins involved in the immune evasion of colorectal tumors. DHA could be used as a potential immune checkpoint inhibitor for the treatment of colorectal cancers.


Assuntos
Neoplasias Colorretais , MicroRNAs , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Antígeno B7-H1/uso terapêutico , Antígeno CD47/genética , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Células CACO-2 , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Ácidos Docosa-Hexaenoicos/farmacologia , Ácidos Docosa-Hexaenoicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Inibidores de Checkpoint Imunológico , Proteínas de Checkpoint Imunológico , Evasão da Resposta Imune , Fatores Imunológicos/uso terapêutico , MicroRNAs/genética , MicroRNAs/metabolismo
16.
Urol Oncol ; 40(4): 166.e15-166.e25, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34998671

RESUMO

BACKGROUND: CD47 has been identified as a phagocytosis checkpoint conferring poor clinical outcomes in various cancer types. A flurry of clinical trials designed to evaluate agents that block CD47 have been initiated. We aimed to explore the clinical significance of CD47 and its correlation with immune infiltration and molecular features in clear cell renal cell carcinoma (ccRCC). METHODS: 235 tumor tissue microarray specimens of ccRCC patients from Zhongshan Hospital, 530 ccRCC patients from The Cancer Genome Atlas and 726 ccRCC patients from JAVELIN Renal 101 study were analyzed. CD47 expression and immune contexture were examined by immunohistochemistry and CIBERSORT algorithm. Survival analyses were conducted through Kaplan-Meier curves and Cox regression model. RESULTS: We demonstrated that ccRCC patients with high CD47 expression exhibited inferior overall survival and recurrence-free survival. CD47 expression associated with heavily immune infiltrated but immunosuppressed microenvironment. CD8+ T cells infiltration had discordant prognostic value based on CD47 expression, where high CD8+ T cell infiltration was associated with worse clinical outcome in CD47hi patients and with favorable prognosis in CD47lo patients. Patients with mutated PBRM1 and SETD2 correlated with decreased CD47 mRNA expression. Patients with higher CD47 expression possessed improved PFS in ICI + VEGFR TKI combination therapy. CONCLUSIONS: CD47 expression was an independent prognosticator of clinical outcome for ccRCC patients. CD47 expression correlated with ccRCC molecular classification and response to combination therapy. The phagocytosis checkpoint CD47 could be applied as an attractive candidate for immunotherapeutic approach in ccRCC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Biomarcadores Tumorais/metabolismo , Antígeno CD47/genética , Antígeno CD47/uso terapêutico , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Feminino , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Masculino , Prognóstico , Microambiente Tumoral
17.
Cell Oncol (Dordr) ; 43(1): 19-30, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31485984

RESUMO

BACKGROUND: CD47 is a widely expressed cellular receptor well known for its immunoregulatory functions. By interacting with its ligands, including thrombospondin-1 (TSP-1), signal regulatory protein α (SIRPα), integrins, and SH2-domain bearing protein tyrosine phosphatase substrate-1 (SHPS-1), it modulates cellular phagocytosis by macrophages, transmigration of neutrophils and activation of dendritic cells, T cells and B cells. Ample studies have shown that various types of cancer express high levels of CD47 to escape from the immune system. Based on this observation, CD47 is currently considered as a prominent target in cancer therapy. CONCLUSIONS: Here, we review the role of CD47 in the maintenance of immune system homeostasis. We also depict three emerging CD47-targeting strategies for cancer therapy, including the use of mimicry peptides, antibodies, and gene silencing strategies. Among these approaches, the most advanced one is the use of anti-CD47 antibodies, which enhances cancer cell phagocytosis via inhibition of the CD47-SIRPα axis. These antibodies can also achieve higher anti-cancer efficacies when combined with chemotherapy and immunotherapy and hold promise for improving the survival of patients with cancer.


Assuntos
Linfócitos B/imunologia , Antígeno CD47/fisiologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Neoplasias/imunologia , Fagocitose/imunologia , Linfócitos T/imunologia , Animais , Antígenos de Diferenciação/metabolismo , Antígeno CD47/metabolismo , Antígeno CD47/uso terapêutico , Células Dendríticas/metabolismo , Humanos , Integrinas/metabolismo , Macrófagos/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Neutrófilos/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia
19.
Nanomedicine (Lond) ; 12(6): 597-613, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28186432

RESUMO

AIM: To develop a novel anti-CD47 single-chain variable fragment (scFv) functionalized magnetic nanoparticles (MNPs) for targeting bladder cell lines and its applicability in thermotherapy. MATERIAL & METHODS: An immunized murine antibody phage display library was constructed and screened to isolate anti-CD47 binders. A scFv was selected and conjugated to MNPs which was then utilized to discriminate CD47+ bladder cells along with assessing its efficacy in thermotherapy. RESULTS: An scFv with high affinity to bladder cells was efficiently conjugated to MNPs. Following a hyperthermia treatment, the function of scFv-MNP conjugates led to a considerable reduction in cell viability. CONCLUSION: The anti-CD47 scFv-MNP conjugate was an effective cancer cell thermotherapy tool that might pave the way for development of bionano-based targeting techniques in both early detection and treatment of cancer.


Assuntos
Antígeno CD47/imunologia , Nanopartículas de Magnetita/administração & dosagem , Anticorpos de Cadeia Única/imunologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Antígeno CD47/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Nanopartículas de Magnetita/química , Camundongos , Anticorpos de Cadeia Única/uso terapêutico , Neoplasias da Bexiga Urinária/imunologia
20.
Yao Xue Xue Bao ; 51(3): 455-61, 2016 03.
Artigo em Chinês | MEDLINE | ID: mdl-29859029

RESUMO

To investigate the effect that folic acid-modified polyrotaxanes(FPP) transfered siRNA CD47 to inhibit melanoma proliferation, the expression of CD47 in clinical melanoma patients was tested by Western blot and RT-PCR, respectively. Physical performance of FPP(siRNA-CD47: CD47) nanoparticles was tested by Malvern particle size instrument and scanning electron microscope. The clone formation experiment demonstrated that FPP(CD47) nanoparticles inhibited the growth of clones. Invasion assay revealed that FPP(CD47) inhibited migration of B16F10 cells. Tumor bearing mice were used in the experiment to test the efficacy of FPP(CD47) treatment. Compared with the control group, high expression of CD47 was observed in the clinical melanoma patients. FPP(CD47) nanoparticle size at 80 nm exhibited a potential of 10 m V; compared with FPP(Con), fluorescence intensity was significantly reduced to 4.2% and B16F10 cell clone formation was decreased by 91% in the FPP(CD47) treatment. Tumor volume of tumor-burdened mice was decreased by 90% with FPP(CD47) treatment. FPP(CD47) lowered CD47 protein and m RNA expression in the tumor. This study suggests that FPP may transfer siRNA CD47 into the cancer cells to inhibit melanoma growth effectively.


Assuntos
Antígeno CD47/uso terapêutico , Ácido Fólico/química , Melanoma/terapia , RNA Interferente Pequeno/uso terapêutico , Rotaxanos/química , Animais , Linhagem Celular Tumoral , Proliferação de Células , Vetores Genéticos , Humanos , Melanoma Experimental , Camundongos , Nanopartículas , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA