Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.689
Filtrar
1.
Biochem Pharmacol ; 225: 116335, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38824968

RESUMO

Drugs specifically targeting YKL-40, an over-expressed gene (CHI3L1) in various diseases remain developed. The current study is to create a humanized anti-YKL-40 neutralizing antibody and characterize its potentially therapeutic signature. We utilized in silico CDR-grafting bioinformatics to replace the complementarity determining regions (CDRs) of human IgG1 with mouse CDRs of our previously established anti-YKL-40 antibody (mAY). In fifteen candidates (VL1-3/VH1-5) of heavy and light chain variable region combination, one antibody L3H4 named Rosazumab demonstrated strong binding affinity with YKL-40 (KD = 4.645 × 10-8 M) and high homology with human IgG (80 %). In addition, we established different overlapping amino acid peptides of YKL-40 and found that Rosazumab specifically bound to residues K337, K342, and R344, the KR-rich functional domain of YKL-40. Rosazumab inhibited migration and tube formation of YKL-40-expressing tumor cells and induced tumor cell apoptosis. Mechanistically, Rosazumab induced interaction of N-cadherin with ß-catenin and activation of downstream MST1/RASSF1/Histone H2B axis, leading to chromosomal DNA breakage and cell apoptosis. Treatment of xenografted tumor mice with Rosazumab twice a week for 4 weeks inhibited tumor growth and angiogenesis, but induced tumor apoptosis. Rosazumab injected in mice distributed to blood, tumor, and other multiple organs, but did not impact in function or structure of liver and kidney, indicating non-detectable toxicity in vivo. Collectively, the study is the first one to demonstrate that a humanized YKL-40 neutralizing antibody offers a valuable means to block tumor development.


Assuntos
Anticorpos Monoclonais Humanizados , Proteína 1 Semelhante à Quitinase-3 , Neoplasias , Animais , Feminino , Humanos , Camundongos , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Linhagem Celular Tumoral , Proteína 1 Semelhante à Quitinase-3/antagonistas & inibidores , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
Respir Res ; 25(1): 215, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38764025

RESUMO

BACKGROUND: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of lung cancer patients with mutated EGFR. However, the efficacy of EGFR-TKIs in wild-type EGFR tumors has been shown to be marginal. Methods that can sensitize EGFR-TKIs to EGFR wild-type NSCLC remain rare. Hence, we determined whether combination treatment can maximize the therapeutic efficacy of EGFR-TKIs. METHODS: We established a focused drug screening system to investigate candidates for overcoming the intrinsic resistance of wild-type EGFR NSCLC to EGFR-TKIs. Molecular docking assays and western blotting were used to identify the binding mode and blocking effect of the candidate compounds. Proliferation assays, analyses of drug interactions, colony formation assays, flow cytometry and nude mice xenograft models were used to determine the effects and investigate the molecular mechanism of the combination treatment. RESULTS: Betulinic acid (BA) is effective at targeting EGFR and synergizes with EGFR-TKIs (gefitinib and osimertinib) preferentially against wild-type EGFR. BA showed inhibitory activity due to its interaction with the ATP-binding pocket of EGFR and dramatically enhanced the suppressive effects of EGFR-TKIs by blocking EGFR and modulating the EGFR-ATK-mTOR axis. Mechanistic studies revealed that the combination strategy activated EGFR-induced autophagic cell death and that the EGFR-AKT-mTOR signaling pathway was essential for completing autophagy and cell cycle arrest. Activation of the mTOR pathway or blockade of autophagy by specific chemical agents markedly attenuated the effect of cell cycle arrest. In vivo administration of the combination treatment caused marked tumor regression in the A549 xenografts. CONCLUSIONS: BA is a potential wild-type EGFR inhibitor that plays a critical role in sensitizing EGFR-TKI activity. BA combined with an EGFR-TKI effectively suppressed the proliferation and survival of intrinsically resistant lung cancer cells via the inhibition of EGFR as well as the induction of autophagy-related cell death, indicating that BA combined with an EGFR-TKI may be a potential therapeutic strategy for overcoming the primary resistance of wild-type EGFR-positive lung cancers.


Assuntos
Autofagia , Ácido Betulínico , Carcinoma Pulmonar de Células não Pequenas , Sinergismo Farmacológico , Receptores ErbB , Neoplasias Pulmonares , Triterpenos Pentacíclicos , Inibidores de Proteínas Quinases , Animais , Humanos , Camundongos , Células A549 , Acrilamidas/farmacologia , Compostos de Anilina/farmacologia , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Autofagia/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Receptores ErbB/antagonistas & inibidores , Gefitinibe/farmacologia , Indóis , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas , Transdução de Sinais/efeitos dos fármacos , Triterpenos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Biomed Pharmacother ; 175: 116736, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38739992

RESUMO

AIMS: The xanthone dimer 12-O-deacetyl-phomoxanthone A (12-ODPXA) was extracted from the secondary metabolites of the endophytic fungus Diaporthe goulteri. The 12-ODPXA compound exhibited anticancer properties in murine lymphoma; however, the anti-ovarian cancer (OC) mechanism has not yet been explored. Therefore, the present study evaluated whether 12-ODPXA reduces OC cell proliferation, metastasis, and invasion by downregulating pyruvate dehydrogenase kinase (PDK)4 expression. METHODS: Cell counting kit-8, colony formation, flow cytometry, wound healing, and transwell assays were performed to examine the effects of 12-ODPXA on OC cell proliferation, apoptosis, migration, and invasion. Transcriptome analysis was used to predict the changes in gene expression. Protein expression was determined using western blotting. Glucose, lactate, and adenosine triphosphate (ATP) test kits were used to measure glucose consumption and lactate and ATP production, respectively. Zebrafish xenograft models were constructed to elucidate the anti-OC effects of 12-ODPXA. RESULTS: The 12-ODPXA compound inhibited OC cell proliferation, migration, invasion, and glycolysis while inducing cell apoptosis via downregulation of PDK4. In vivo experiments showed that 12-ODPXA suppressed tumor growth and migration in zebrafish. CONCLUSION: Our data demonstrate that 12-ODPXA inhibits ovarian tumor growth and metastasis by downregulating PDK4, revealing the underlying mechanisms of action of 12-ODPXA in OC.


Assuntos
Apoptose , Movimento Celular , Proliferação de Células , Regulação para Baixo , Neoplasias Ovarianas , Piruvato Desidrogenase Quinase de Transferência de Acetil , Xantonas , Peixe-Zebra , Animais , Feminino , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Humanos , Xantonas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Piruvato Desidrogenase Quinase de Transferência de Acetil/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Metástase Neoplásica , Invasividade Neoplásica
4.
J Vis Exp ; (207)2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38801268

RESUMO

Non-small cell lung cancer (NSCLC) is a highly lethal disease with a complex and heterogeneous tumor microenvironment. Currently, common animal models based on subcutaneous inoculation of cancer cell suspensions do not recapitulate the tumor microenvironment in NSCLC. Herein we describe a murine orthotopic lung cancer xenograft model that employs the intrapulmonary inoculation of three-dimensional multicellular spheroids (MCS). Specifically, fluorescent human NSCLC cells (A549-iRFP) were cultured in low-attachment 96-well microplates with collagen for 3 weeks to form MCS, which were then inoculated intercostally into the left lung of athymic nude mice to establish the orthotopic lung cancer model. Compared with the original A549 cell line, MCS of the A549-iRFP cell line responded similarly to anticancer drugs. The long-wavelength fluorescent signal of the A549-iRFP cells correlated strongly with common markers of cancer cell growth, including spheroid volume, cell viability, and cellular protein level, thus allowing dynamic monitoring of the cancer growth in vivo by fluorescent imaging. After inoculation into mice, the A549-iRFP MCS xenograft reliably progressed through phases closely resembling the clinical stages of NSCLC, including the expansion of the primary tumor, the emergence of neighboring secondary tumors, and the metastases of cancer cells to the contralateral right lung and remote organs. Moreover, the model responded to the benchmark antilung cancer drug, cisplatin with the anticipated toxicity and slower cancer progression. Therefore, this murine orthotopic xenograft model of NSCLC would serve as a platform to recapitulate the disease's progression and facilitate the development of potential anticancer drugs.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Camundongos Nus , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/tratamento farmacológico , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Progressão da Doença , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Modelos Animais de Doenças , Células A549 , Transplante de Neoplasias
5.
Drug Metab Dispos ; 52(7): 644-653, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38670798

RESUMO

Free (unbound) drug concentration at the site of action is the key determinant of biologic activity since only unbound drugs can exert pharmacological and toxicological effects. Unbound drug concentration in tumors for solid cancers is needed to understand/explain/predict pharmacokinetics, pharmacodynamics, and efficacy relations. Fraction unbound (fu ) in tumors is usually determined across several xenografted tumors derived from various cell lines in the drug discovery stage, which is time consuming and a resource burden. In this study, we determined the fu values for a set of diverse compounds (comprising acid, base, neutral, zwitterion, and covalent drugs) across five different xenografted tumors and five commercially available mouse tissues to explore the correlation of fu between tumors and the possibility of surrogate tissue(s) for tumor fu (fu,tumor) determination. The crosstumor comparison showed that fu,tumor values across tumors are largely comparable, and systematic tissue versus tumor comparison demonstrated that only lung tissue had comparable fu to all five tumors (fu values within twofold change for >80% compounds in both comparisons). These results indicated that mouse lung tissue can be used as a surrogate matrix for a fu,tumor assay. This study will increase efficiency in fu,tumor assessment and reduce animal use (adapting the replace, reduce, and refine principle) in drug discovery. SIGNIFICANCE STATEMENT: The free drug concept is a well accepted principle in drug discovery research. Currently, tumor fraction unbound (fu,tumor) is determined in several tumors derived from different cell lines to estimate free drug concentrations of a compound. The results from this study indicated that fu,tumor across xenografted tumors is comparable, and fu,tumor can be estimated using a surrogate tissue, mouse lung. The results will increase efficiency in fu,tumor assessment and reduce animal use in drug discovery.


Assuntos
Pulmão , Animais , Camundongos , Humanos , Pulmão/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Linhagem Celular Tumoral , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/metabolismo , Feminino
6.
Methods Mol Biol ; 2806: 31-40, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676794

RESUMO

Patient-derived xenograft (PDX) modeling is a valuable tool for the study of leukemia pathogenesis, progression, and therapy response. Engraftment of human leukemia cells occurs following injection into the tail vein (or retro-orbital vein) of preconditioned immunocompromised mice. Injected mice are maintained in a sterile and supportive housing environment until leukemia engraftment is observed, at which time studies such as drug treatments or leukemia sampling can occur. Here, we outline a method for generating PDXs from Acute Myeloid Leukemia (AML) patient samples using tail vein injection; however it can also be readily applied to T- and B- Acute Lymphoblastic Leukemia (ALL) samples.


Assuntos
Modelos Animais de Doenças , Animais , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Leucemia Mieloide Aguda/patologia , Xenoenxertos , Leucemia/patologia
7.
Methods Mol Biol ; 2806: 1-8, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676791

RESUMO

Patient-derived xenografts (PDXs) represent a critical advancement in preclinical cancer research, wherein human tumor samples are implanted into animal models for evaluation of therapeutic responses. PDXs have emerged as indispensable tools in translational cancer research, facilitating investigation into tumor microenvironments and personalized medicine. This chapter elucidates the historical evolution of PDXs, from early attempts in the eighteenth century to contemporary immunocompromised host models that enhance engraftment success.


Assuntos
Hospedeiro Imunocomprometido , Pesquisa Translacional Biomédica , Humanos , Animais , Pesquisa Translacional Biomédica/métodos , Modelos Animais de Doenças , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Neoplasias/imunologia , Neoplasias/patologia , Xenoenxertos , História do Século XX , Medicina de Precisão/métodos , Microambiente Tumoral/imunologia , História do Século XXI
8.
Methods Mol Biol ; 2806: 55-74, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676796

RESUMO

Realistic and renewable laboratory models that accurately reflect the distinct clinical features of childhood cancers have enormous potential to speed research progress. These models help us to understand disease biology, develop new research methods, advance new therapies to clinical trial, and implement personalized medicine. This chapter describes methods to generate patient-derived xenograft models of neuroblastoma and rhabdomyosarcoma, two tumor types for which children with high-risk disease have abysmal survival outcomes and survivors have lifelong-debilitating effects from treatment. Further, this protocol addresses model development from diverse clinical tumor tissue samples, subcutaneous and orthotopic engraftment, and approaches to avoid model loss.


Assuntos
Neuroblastoma , Rabdomiossarcoma , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Animais , Camundongos , Neuroblastoma/patologia , Neuroblastoma/genética , Rabdomiossarcoma/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Criança , Modelos Animais de Doenças , Xenoenxertos , Medicina de Precisão/métodos , Linhagem Celular Tumoral
9.
Methods Mol Biol ; 2806: 75-90, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676797

RESUMO

The development of clinically relevant and reliable models of central nervous system tumors has been instrumental in advancing the field of Neuro-Oncology. The orthotopic intracranial injection is widely used to study the growth, invasion, and spread of tumors in a controlled environment. Orthotopic models are performed to examine tumor cells isolated from a specific region in a patient in the same site or location in an animal model. Orthotopic brain tumor models are also utilized for preclinical testing of therapeutics as they closely recapitulate the behavior of such cancer and the brain environment of patients. Below, we describe our experiences in the development of murine models of pediatric brain tumors including diffuse midline glioma (DMG), glioblastoma (GBM), and medulloblastoma. The method provides an overview of intracranial stereotactic injections in mice.


Assuntos
Neoplasias Encefálicas , Modelos Animais de Doenças , Animais , Humanos , Camundongos , Neoplasias Encefálicas/patologia , Criança , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Meduloblastoma/patologia , Glioma/patologia , Glioblastoma/patologia , Xenoenxertos
10.
Methods Mol Biol ; 2806: 101-115, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676799

RESUMO

Patient-Derived Xenografts (PDXs) are established by implanting a fragment of a patient tumor into rodents either subcutaneously or orthotopically. PDX models faithfully recapitulate the histologic and molecular profile of the donor patient's cancer and are regarded as authentic preclinical models for drug testing, understanding of tumor biology and biomarker discovery. This Chapter describes the detailed method for establishing robust PDXs for endometrial cancer and provide important notes for users of the protocol to consider during PDXs development.


Assuntos
Neoplasias do Endométrio , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias do Endométrio/patologia , Feminino , Humanos , Animais , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Modelos Animais de Doenças , Xenoenxertos
11.
Methods Mol Biol ; 2806: 41-53, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676795

RESUMO

Patient-derived orthotopic xenograft (PDOX) mouse models are considered the gold standard for evidence-based preclinical research in pediatric neuro-oncology. This protocol describes the generation of PDOX models by intracranial implantation of human pediatric brain cancer cells into immune-deficient mice, and their continued propagation to establish cohorts of animals for preclinical research.


Assuntos
Neoplasias Encefálicas , Modelos Animais de Doenças , Animais , Neoplasias Encefálicas/patologia , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Criança , Gradação de Tumores , Xenoenxertos
12.
Methods Mol Biol ; 2806: 91-100, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676798

RESUMO

Pancreatic cancer is associated with a high mortality rate, and there are still very few effective treatment options. Patient-derived xenografts have proven to be invaluable preclinical disease models to study cancer biology and facilitate testing of novel therapeutics. However, the severely immune-deficient mice used to generate standard models lack any functional immune system, thereby limiting their utility as a tool to investigate the tumor-immune cell interface. This chapter will outline a method for establishment of "humanized" patient-derived xenografts, which are reconstituted with human immune cells to imitate the immune-rich microenvironment of pancreatic cancer.


Assuntos
Modelos Animais de Doenças , Neoplasias Pancreáticas , Microambiente Tumoral , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/terapia , Animais , Humanos , Camundongos , Microambiente Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Xenoenxertos , Camundongos SCID
13.
Methods Mol Biol ; 2806: 187-196, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676803

RESUMO

Patient-derived xenograft (PDX) models play a crucial role for in vivo research. They maintain the original molecular characteristics of the human tumor and provide a more accurate tumor microenvironment, which cannot be replicated by in vitro models. This chapter describes four different transplantation methods, namely, intra-bursal, intrarenal capsule, intraperitoneal, and subcutaneous, to develop PDX models for ovarian cancer research.


Assuntos
Modelos Animais de Doenças , Neoplasias Ovarianas , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Neoplasias Ovarianas/patologia , Feminino , Animais , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Microambiente Tumoral , Xenoenxertos
14.
Methods Mol Biol ; 2806: 139-151, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676801

RESUMO

Cholangiocarcinoma (CCA) poses a substantial clinical hurdle as it is often detected at advanced metastatic stages with limited therapeutic options. To enhance our understanding of advanced CCA, it is imperative to establish preclinical models that faithfully recapitulate the disease's characteristics. Patient-derived xenograft (PDX) models have emerged as a valuable approach in cancer research, offering an avenue to reproduce and study the genomic, histologic, and molecular features of the original human tumors. By faithfully preserving the heterogeneity, microenvironmental interactions, and drug responses observed in human tumors, PDX models serve as highly relevant and predictive preclinical tools. Here, we present a comprehensive protocol that outlines the step-by-step process of generating and maintaining PDX models using biopsy samples from patients with advanced metastatic CCA. The protocol encompasses crucial aspects such as tissue processing, xenograft transplantation, and subsequent monitoring of the PDX models. By employing this protocol, we aim to establish a robust collection of PDX models that accurately reflect the genomic landscape, histologic diversity, and therapeutic responses observed in advanced CCA, thereby enabling improved translational research, drug development, and personalized treatment strategies for patients facing this challenging disease.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Ensaios Antitumorais Modelo de Xenoenxerto , Colangiocarcinoma/patologia , Colangiocarcinoma/genética , Humanos , Animais , Camundongos , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Modelos Animais de Doenças
15.
Methods Mol Biol ; 2806: 9-18, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38676792

RESUMO

Patient-derived xenografts (PDXs) have emerged as a pivotal tool in translational cancer research, addressing limitations of traditional methods and facilitating improved therapeutic interventions. These models involve engrafting human primary malignant cells or tissues into immunodeficient mice, allowing for the investigation of cancer mechanobiology, validation of therapeutic targets, and preclinical assessment of treatment strategies. This chapter provides an overview of PDXs methodology and their applications in both basic cancer research and preclinical studies. Despite current limitations, ongoing advancements in humanized xenochimeric models and autologous immune cell engraftment hold promise for enhancing PDX model accuracy and relevance. As PDX models continue to refine and extend their applications, they are poised to play a pivotal role in shaping the future of translational cancer research.


Assuntos
Neoplasias , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Animais , Neoplasias/patologia , Neoplasias/terapia , Neoplasias/imunologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Modelos Animais de Doenças , Xenoenxertos , Pesquisa Translacional Biomédica/métodos
16.
Biochem Pharmacol ; 224: 116207, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38621425

RESUMO

Osimertinib is a novel epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), acting as the first-line medicine for advanced EGFR-mutated NSCLC. Recently, the acquired resistance to osimertinib brings great challenges to the advanced treatment. Therefore, it is in urgent need to find effective strategy to overcome osimertinib acquired resistance. Here, we demonstrated that SREBP pathway-driven lipogenesis was a key mediator to promote osimertinib acquired resistance, and firstly found Tanshinone IIA (Tan IIA), a natural pharmacologically active constituent isolated from Salvia miltiorrhiza, could overcome osimertinib-acquired resistance in vitro and in vivo via inhibiting SREBP pathway-mediated lipid lipogenesis by using LC-MS based cellular lipidomics analysis, quantitative real-time PCR (qRT-PCR) analysis, western blotting analysis, flow cytometry, small interfering RNAs transfection, and membrane fluidity assay et al. The results showed that SREBP1/2-driven lipogenesis was highly activated in osimertinib acquired resistant NSCLC cells, while knockdown or inhibition of SREBP1/2 could restore the sensitivity of NSCLC to osimertinib via altered the proportion of saturated phospholipids and unsaturated phospholipids in osimertinib acquired-resistant cells. Furthermore, Tanshinone IIA (Tan IIA) could reverse the acquired resistance to osimertinib in lung cancer. Mechanically, Tan IIA inhibited SREBP signaling mediated lipogenesis, changed the profiles of saturated phospholipids and unsaturated phospholipids, and thus promoted osimertinib acquired resistant cancer cells to be attacked by oxidative stress-induced damage and reduce the cell membrane fluidity. The reversal effect of Tan IIA on osimertinib acquired resistant NSCLC cells was also confirmed in vivo, which is helpful for the development of strategies to reverse osimertinib acquired resistance.


Assuntos
Abietanos , Acrilamidas , Resistencia a Medicamentos Antineoplásicos , Lipogênese , Neoplasias Pulmonares , Camundongos Nus , Humanos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Abietanos/farmacologia , Animais , Acrilamidas/farmacologia , Lipogênese/efeitos dos fármacos , Camundongos , Compostos de Anilina/farmacologia , Antineoplásicos/farmacologia , Camundongos Endogâmicos BALB C , Linhagem Celular Tumoral , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Masculino , Feminino , Indóis , Pirimidinas
17.
Biochem Pharmacol ; 224: 116217, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38641306

RESUMO

The Hippo pathway is a key regulator of tissue growth, organ size, and tumorigenesis. Activating the Hippo pathway by gene editing or pharmaceutical intervention has been proven to be a new therapeutic strategy for treatment of the Hippo pathway-dependent cancers. To now, a number of compounds that directly target the downstream effector proteins of Hippo pathway, including YAP and TEADs, have been disclosed, but very few Hippo pathway activators are reported. Here, we discovered a new class of Hippo pathway activator, YL-602, which inhibited CTGF expression in cells irrespective of cell density and the presence of serum. Mechanistically, YL-602 activates the Hippo pathway via MST1/2, which is different from known activators of Hippo pathway. In vitro, YL-602 significantly induced tumor cell apoptosis and inhibited colony formation of tumor cells. In vivo, oral administration of YL-602 substantially suppressed the growth of cancer cells by activation of Hippo pathway. Overall, YL-602 could be a promising lead compound, and deserves further investigation for its mechanism of action and therapeutic applications.


Assuntos
Antineoplásicos , Via de Sinalização Hippo , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Antineoplásicos/farmacologia , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Camundongos Nus , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Camundongos Endogâmicos BALB C , Apoptose/efeitos dos fármacos , Feminino
18.
Biochem Pharmacol ; 224: 116220, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38641307

RESUMO

Alpha-enolase (ENO1), a multifunctional protein with carcinogenic properties, has emerged as a promising cancer biomarker because of its differential expression in cancer and normal cells. On the basis of this characteristic, we designed a cell-targeting peptide that specifically targets ENO1 and connected it with the drug doxorubicin (DOX) by aldehyde-amine condensation. A surface plasmon resonance (SPR) assay showed that the affinity for ENO1 was stronger (KD = 2.5 µM) for the resulting cell-targeting drug, DOX-P, than for DOX. Moreover, DOX-P exhibited acid-responsive capabilities, enabling precise release at the tumor site under the guidance of the homing peptide and alleviating DOX-induced cardiotoxicity. An efficacy experiment confirmed that, the targeting ability of DOX-P toward ENO1 demonstrated superior antitumor activity against colorectal cancer than that of DOX, while reducing its toxicity to cardiomyocytes. Furthermore, in vivo metabolic distribution results indicated low accumulation of DOX-P in nontumor sites, further validating its targeting ability. These results showed that the ENO1-targeted DOX-P peptide has great potential for application in targeted drug-delivery systems for colorectal cancer therapy.


Assuntos
Antibióticos Antineoplásicos , Neoplasias Colorretais , Doxorrubicina , Sistemas de Liberação de Medicamentos , Fosfopiruvato Hidratase , Proteínas Supressoras de Tumor , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Fosfopiruvato Hidratase/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Animais , Proteínas Supressoras de Tumor/metabolismo , Humanos , Camundongos , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/administração & dosagem , Camundongos Endogâmicos BALB C , Camundongos Nus , Masculino , Linhagem Celular Tumoral , Células HCT116 , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Biomarcadores Tumorais
19.
Cells ; 12(8)2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37190054

RESUMO

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


Assuntos
Antineoplásicos Imunológicos , Técnicas de Cultura de Células em Três Dimensões , Hidrogéis , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Células Estromais , Neoplasias/tratamento farmacológico , Humanos , Imunoterapia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Linhagem Celular Tumoral , Antineoplásicos Imunológicos/isolamento & purificação , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico
20.
Acta Biochim Pol ; 69(1): 165-172, 2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-35143148

RESUMO

Abnormal expression of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4) is closely related to the occurrence and development of tumors, and PFKFB4 has been shown to function as a protein kinase. However, the molecular mechanisms through which PFKFB4 functions in glioblastoma (GBM) remain poorly understood. Accordingly, in this study, we assessed the roles of PFKFB4 in GBM. Compared to in adjacent tissues, PFKFB4 was highly expressed in GBM, and its expression level was negatively correlated with the overall survival time. In addition, knockdown of PFKFB4 inhibited the proliferation and invasion of GBM cells and promoted apoptosis. In a xenograft tumor model, tumor growth was inhibited by knockdown of PFKFB4 using short hairpin RNA. Further studies demonstrated that PFKFB4 is involved in regulating the AKT signaling pathway. Thus, PFKFB4 acts as a protein kinase to regulate GBM progression by activating the AKT/forkhead box O1 pathway, which may be a potential therapeutic target in GBM.


Assuntos
Proteína Forkhead Box O1/metabolismo , Glioblastoma/metabolismo , Fosfofrutoquinase-2/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Silenciamento de Genes/métodos , Glioblastoma/genética , Glicólise , Humanos , Masculino , Camundongos , Fosfofrutoquinase-2/genética , RNA Interferente Pequeno/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...