Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 11(11)2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-38251688

RESUMO

BACKGROUND: Cell culture conditions during manufacturing can impact the clinical efficacy of chimeric antigen receptor (CAR) T cell products. Production methods have not been standardized because the optimal approach remains unknown. Separate CD4+ and CD8+ cultures offer a potential advantage but complicate manufacturing and may affect cell expansion and function. In a phase 1/2 clinical trial, we observed poor expansion of separate CD8+ cell cultures and hypothesized that coculture of CD4+ cells and CD8+ cells at a defined ratio at culture initiation would enhance CD8+ cell expansion and simplify manufacturing. METHODS: We generated CAR T cells either as separate CD4+ and CD8+ cells, or as combined cultures mixed in defined CD4:CD8 ratios at culture initiation. We assessed CAR T cell expansion, phenotype, function, gene expression, and in vivo activity of CAR T cells and compared these between separately expanded or mixed CAR T cell cultures. RESULTS: We found that the coculture of CD8+ CAR T cells with CD4+ cells markedly improves CD8+ cell expansion, and further discovered that CD8+ cells cultured in isolation exhibit a hypofunctional phenotype and transcriptional signature compared with those in mixed cultures with CD4+ cells. Cocultured CAR T cells also confer superior antitumor activity in vivo compared with separately expanded cells. The positive impact of CD4+ cells on CD8+ cells was mediated through both cytokines and direct cell contact, including CD40L-CD40 and CD70-CD27 interactions. CONCLUSIONS: Our data indicate that CD4+ cell help during cell culture maintains robust CD8+ CAR T cell function, with implications for clinical cell manufacturing.


Assuntos
Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Linfócitos T CD4-Positivos , Técnicas de Cultura de Células , Linfócitos T CD8-Positivos , Fenótipo
2.
Curr Med Chem ; 29(13): 2290-2305, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34514979

RESUMO

The unique features of the tumor microenvironment (TME) govern the biological properties of many cancers, including hematological malignancies. TME factors can trigger an invasion and protect against drug cytotoxicity by inhibiting apoptosis and activating specific signaling pathways (e.g. NF-ΚB). TME remodeling is facilitated due to the high self-renewal ability of the bone marrow. Progressing tumor cells can alter some extracellular matrix (ECM) components which act as a barrier to drug penetration in the TME. The initial progression of the cell cycle is controlled by the MAPK pathway (Raf/MEK/ERK) and Hippo pathway, while the final phase is regulated by the PI3K/Akt /mTOR and WNT pathways. This review summarizes the main signaling pathways involved in drug resistance (DR) and some mechanisms by which DR can occur in the bone marrow. The relationship between autophagy, endoplasmic reticulum stress, and cellular signaling pathways in DR and apoptosis is covered in the TME.


Assuntos
Neoplasias Hematológicas , Fosfatidilinositol 3-Quinases , Apoptose , Medula Óssea/metabolismo , Linhagem Celular Tumoral , Resistência a Medicamentos , Neoplasias Hematológicas/tratamento farmacológico , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Microambiente Tumoral
3.
Cancer Immunol Res ; 9(8): 856-861, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34344697

RESUMO

The development of chimeric antigen receptor (CAR) T-cell therapy has led to a paradigm shift in cancer treatment. However, patients often do not benefit from CAR T-cell therapy due to poor persistence of the adoptively transferred cells. Development of strategies based on the generation and maintenance of long-lasting memory T cells may expand the therapeutic effects of CAR T cells. Mitochondrial metabolic pathways play crucial roles in regulating the fate, function, and longevity of T cells. Here, we discuss how reprogramming of mitochondrial metabolic pathways influences function, persistence, and determination of CAR T-cell fate toward a memory phenotype. Moreover, we explore how mitochondrial activity determines persistence and the clinical outcome of CAR T-cell therapy. In addition, we review some strategies for manipulating CAR T-cell mitochondria to improve the survival of CAR T cells.


Assuntos
Imunoterapia Adotiva/métodos , Mitocôndrias/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Humanos , Longevidade
4.
Curr Pharm Biotechnol ; 22(9): 1149-1155, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33038909

RESUMO

On a worldwide scale, the outbreak of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has led to extensive damage to the health system as well as the global economy. Hitherto, there has been no approved drug or vaccine for this disease. Therefore, the use of general antiviral drugs is at the first line of treatment, though complicated with limited effectiveness and systemic side effects. Given the pathophysiology of the disease, researchers have proposed various strategies not only to find a more specific therapeutic way but also to reduce the side effects. One strategy to accomplish these goals is to use CRISPR/Cas13 system. Recently, a group of scientists has used the CRISPR/Cas13 system, which is highly effective in eliminating the genome of RNA viruses. Due to the RNA nature of the coronavirus genome, it seems that this system can be effective against the disease. The main challenge regarding the application of this system is to deliver it to the target cells efficiently. To solve this challenge, it seems that using virosomes with protein S on their membrane surface can be helpful. Studies have shown that protein S interacts with its specific receptor in target cells named Angiotensin-Converting Enzyme 2 (ACE2). Here, we propose if CRISPR/Cas13 gene constructs reach the infected cells efficiently using a virosomal delivery system, the virus genome will be cleaved and inactivated. Considering the pathophysiology of the disease, an important step to implement this hypothesis is to embed protein S on the membrane surface of virosomes to facilitate the delivery of gene constructs to the target cells.


Assuntos
COVID-19 , SARS-CoV-2 , Antivirais , Sistemas CRISPR-Cas , Genoma Viral , Humanos
5.
Cancer Lett ; 500: 107-118, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33290868

RESUMO

Therapeutic efficacy of chimeric antigen receptor (CAR) T cells is associated with their expansion, persistence and effector function. Although CAR T cell therapy has shown remarkable therapeutic effects in hematological malignancies, its therapeutic efficacy has been limited in some types of cancers - in particular, solid tumors - partially due to the cells' inability to persist and the acquisition of T cell dysfunction within a harsh immunosuppressive tumor microenvironment. Therefore, it would be expected that generation of CAR T cells with intrinsic properties for functional longevity, such as the cells with early-memory phenotypes, could beneficially enhance antitumor immunity. Furthermore, because the metabolic pathways of CAR T cells help determine cellular differentiation and lifespan, therapies targeting such pathways like glycolysis and oxidative phosphorylation, can alter CAR T cell fate and durability within tumors. Here we discuss how reprogramming of CAR T cell metabolism and metabolic switch to memory CAR T cells influences their antitumor activity. We also offer potential strategies for targeting these metabolic circuits in the setting of adoptive CAR T cell therapy, aiming to better unleash the potential of adoptive CAR T cell therapy in the clinic.


Assuntos
Reprogramação Celular/genética , Neoplasias Hematológicas/terapia , Imunoterapia Adotiva , Receptores de Antígenos Quiméricos/genética , Animais , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Antígenos Quiméricos/uso terapêutico , Linfócitos T/imunologia , Linfócitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
6.
Curr Pharm Des ; 25(33): 3563-3577, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31470781

RESUMO

Long noncoding RNAs (lncRNAs) constitute large portions of the mammalian transcriptome which appeared as a fundamental player, regulating various cellular mechanisms. LncRNAs do not encode proteins, have mRNA-like transcripts and frequently processed similar to the mRNAs. Many investigations have determined that lncRNAs interact with DNA, RNA molecules or proteins and play a significant regulatory function in several biological processes, such as genomic imprinting, epigenetic regulation, cell cycle regulation, apoptosis, and differentiation. LncRNAs can modulate gene expression on three levels: chromatin remodeling, transcription, and post-transcriptional processing. The majority of the identified lncRNAs seem to be transcribed by the RNA polymerase II. Recent evidence has illustrated that dysregulation of lncRNAs can lead to many human diseases, in particular, cancer. The aberrant expression of lncRNAs in malignancies contributes to the dysregulation of proliferation and differentiation process. Consequently, lncRNAs can be useful to the diagnosis, treatment, and prognosis, and have been characterized as potential cancer markers as well. In this review, we highlighted the role and molecular mechanisms of lncRNAs and their correlation with some of the cancers.


Assuntos
Epigênese Genética , Neoplasias/genética , RNA Longo não Codificante/genética , Animais , Montagem e Desmontagem da Cromatina , Humanos , Processamento de Proteína Pós-Traducional , Transcriptoma
7.
Curr Pharm Des ; 25(30): 3225-3238, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31465276

RESUMO

Macrophages are one of the crucial mediators of the immune response in different physiological and pathological conditions. These cells have critical functions in the inflammation mechanisms that are involved in the inhibition or progression of a wide range of diseases including cancer, autoimmune diseases, etc. It has been shown that macrophages are generally divided into two subtypes, M1 and M2, which are distinguished on the basis of their different gene expression patterns and phenotype. M1 macrophages are known as pro-inflammatory cells and are involved in inflammatory mechanisms, whereas M2 macrophages are known as anti-inflammatory cells that are involved in the inhibition of the inflammatory pathways. M2 macrophages help in tissue healing via producing anti-inflammatory cytokines. Increasing evidence indicated that the appearance of different macrophage subtypes is associated with the fate of diseases (progression versus suppression). Hence, polarization of macrophages can be introduced as an important venue in finding, designing and developing novel therapeutic approaches. Albeit, there are different pharmacological agents that are used for the treatment of various disorders, it has been shown that several natural compounds have the potential to regulate M1 to M2 macrophage polarization and vice versa. Herein, for the first time, we summarized new insights into the pharmacological effects of natural compounds on macrophage polarization.


Assuntos
Macrófagos/efeitos dos fármacos , Compostos Fitoquímicos/farmacologia , Plantas Medicinais/química , Citocinas , Humanos , Inflamação , Ativação de Macrófagos , Macrófagos/classificação
8.
Front Immunol ; 10: 780, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31049049

RESUMO

[This corrects the article DOI: 10.3389/fimmu.2017.01850.].

9.
J Cell Physiol ; 233(4): 2815-2823, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475219

RESUMO

Cell therapy is an emerging fields in the treatment of various diseases such as cardiovascular, pulmonary, hepatic, and neoplastic diseases. Stem cells are an integral tool for cell therapy. Multipotent stem cells are an important class of stem cells which have the ability to self-renew through dividing and developing into multiple specific cell types in a specific tissue or organ. These cells are capable to activate or inhibit a sequence of cellular and molecular pathways leading to anti-inflammatory and anti-apoptotic effects which might contribute to the treatment of various diseases. It has been showed that multipotent stem cells exert their therapeutic effects via inhibition/activation of a sequence of cellular and molecular pathways. Although the advantages of multipotent stem cells are numerous, further investigation is still necessary to clarify the biology and safety of these cells before they could be considered as a potential treatment for different types of diseases. This review summarizes different features of multipotent stem cells including isolation, differentiation, and therapeutic applications.


Assuntos
Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/transplante , Transplante de Células-Tronco , Animais , Separação Celular , Ensaios Clínicos como Assunto , Humanos , Neoplasias/terapia
10.
Front Immunol ; 8: 1850, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29312333

RESUMO

Adoptive cellular immunotherapy (ACT) employing engineered T lymphocytes expressing chimeric antigen receptors (CARs) has demonstrated promising antitumor effects in advanced hematologic cancers, such as relapsed or refractory acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma, supporting the translation of ACT to non-hematological malignancies. Although CAR T cell therapy has made remarkable strides in the treatment of patients with certain hematological cancers, in solid tumors success has been limited likely due to heterogeneous antigen expression, immunosuppressive networks in the tumor microenvironment limiting CAR T cell function and persistence, and suboptimal trafficking to solid tumors. Here, we outline specific approaches to overcome barriers to CAR T cell effectiveness in the context of the tumor microenvironment and offer our perspective on how expanding the use of CAR T cells in solid tumors may require modifications in CAR T cell design. We anticipate these modifications will further expand CAR T cell therapy in clinical practice.

11.
Curr Med Chem ; 23(5): 455-63, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26674785

RESUMO

Melanoma is a leading cause of mortality from skin cancer and has a poor prognosis. Despite rapid advances in the treatment of this tumor type, the efficacy of current chemo-/targeted-therapies is still limited owing to the lack of sufficient drug accumulation in the tumor tissue and development of chemo-resistance. Recently, the application of mesenchymal stem cells (MSCs) in cancer therapy has gained substantial attention, suggesting their potential roles as an intriguing vehicle in improving the delivery of targeted agents. MSCs are genetically modified with suicide tumor suppressor genes to inhibit cell signaling pathways associated with the progression and metastatic features of melanoma. Here we describe the clinical application of MSCs in melanoma with a particular emphasis on recent findings on the role of MSC expressing a distinct set of biologically functional chemokines and tumor suppressing agents. Accumulating data has shown the tumor- oriented homing capacity of MSCs and their applications as a vehicle (e.g., adipose derived mesenchymal stem cells expressing TRAIL, interferon-α/γ, pigment epithelium-derived factor and cytosine deaminase). Several questions regarding possible potential and intrinsic mechanisms that might induce tumorigenesis and drug resistance are yet to be addressed for tailoring MSC-nbased treatment of melanoma.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Portadores de Fármacos/metabolismo , Melanoma/terapia , Células-Tronco Mesenquimais/metabolismo , Citocinas/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patologia
12.
Gen Physiol Biophys ; 34(1): 43-50, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25367760

RESUMO

The purpose of this study was to evaluate the effect of resveratrol on cytogenetic damages of iododeoxyuridine (IUdR) and x-ray megavoltage radiation (6 MV) in spheroid model of U87MG glioblastoma cancer cell line using clonogenic and alkaline comet assay. Cells were cultured as spheroids (350 µm) that were treated with 20 µM resveratrol, 1 µM IUdR and 2 Gy of 6 MV x-ray. After treatment, viability of the cells, colony forming ability and the induced DNA damages were examined using trypan blue dye exclusion, colonogenic and alkaline comet assay, respectively. Our results showed that resveratrol could significantly reduce the colony number and induce the DNA damages of the cells treated with IUdR in combination with 6 MV x-ray radiation. That results indicated that resveratrol as an inhibitor of hypoxia inducible factor 1 alpha (HIF-1α) protein in combination with IUdR as a radiosensitizer enhanced the radiosensitization of glioblastoma spheroid cells.


Assuntos
Dano ao DNA , Glioblastoma/patologia , Idoxuridina/farmacologia , Estilbenos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Sobrevivência Celular , Ensaio Cometa , Relação Dose-Resposta a Droga , Humanos , Resveratrol , Estilbenos/administração & dosagem , Sais de Tetrazólio , Tiazóis , Azul Tripano , Raios X
13.
J Cancer Res Ther ; 8(3): 399-403, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23174722

RESUMO

AIM: The CD133 antigen has been identified as a putative stem cell marker in colorectal cancer tissues. The aim of this study was to investigate the cell cycle state of CD133(+) and CD133(-) cells, isolated from primary human colorectal tumors. MATERIALS AND METHODS: After mechanical and enzymatic dissociation of the tumor samples, CD133(+) and CD133(-) subsets were identified and separated by magnetic cell sorting. Flow cytometric analysis was performed to compare the cell cycle of both CD133(+) and CD133(-) cells isolated from primary and liver metastatic cancer cells. RESULTS: The results indicated that CD133(+) cells isolated from both primary and liver metastatic colorectal cancers were found in higher percentage in the G0/G1 phases. However, the CD133(-) cells isolated from primary colorectal cancers were predominantly found in the S and G2/M phases. Surprisingly, the CD133(-) cells isolated from liver metastatic colorectal cancers were mostly found in the G0/G1 phase. CONCLUSION: The present study provides evidence that CD133(+) cells are in a quiescent state in colorectal cancer, representing a mechanism that would at least partially explain chemotherapy resistance and tumor recurrence in post-therapy patients.


Assuntos
Antígenos CD/metabolismo , Ciclo Celular , Neoplasias Colorretais/patologia , Glicoproteínas/metabolismo , Neoplasias Hepáticas/secundário , Células-Tronco Neoplásicas , Peptídeos/metabolismo , Antígeno AC133 , Idoso , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...