Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 32(1): 13-31, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37919901

RESUMO

mRNA vaccines have evolved as promising cancer therapies. These vaccines can encode tumor-allied antigens, thus enabling personalized treatment approaches. They can also target cancer-specific mutations and overcome immune evasion mechanisms. They manipulate the body's cellular functions to produce antigens, elicit immune responses, and suppress tumors by overcoming limitations associated with specific histocompatibility leukocyte antigen molecules. However, successfully delivering mRNA into target cells destroys a crucial challenge. Viral and nonviral vectors (lipid nanoparticles and cationic liposomes) have shown great capacity in protecting mRNA from deterioration and assisting in cellular uptake. Cell-penetrating peptides, hydrogels, polymer-based nanoparticles, and dendrimers have been investigated to increase the delivery efficacy and immunogenicity of mRNA. This comprehensive review explores the landscape of mRNA vaccines and their delivery platforms for cancer, addressing design considerations, diverse delivery strategies, and recent advancements. Overall, this review contributes to the progress of mRNA vaccines as an innovative strategy for effective cancer treatment.


Assuntos
Vacinas Anticâncer , Doenças Transmissíveis , Nanopartículas , Neoplasias , Vacinas , Humanos , Vacinas de mRNA , Neoplasias/genética , Neoplasias/terapia , Antígenos de Neoplasias , Nanopartículas/química , RNA Mensageiro/genética , Vacinas Anticâncer/genética
2.
Semin Cancer Biol ; 88: 67-80, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36535506

RESUMO

Pancreatic cancer is one of the most aggressive cancers worldwide due to the resistances to conventional therapies and early metastasis. Recent research has shown that cancer stem cell populations modulate invasiveness, recurrence, and drug resistance in various cancers, including pancreatic cancer. Pancreatic cancer stem cells (PaCSCs) are characterized by their high plasticity and self-renewal capacities that endow them with unique metabolic, metastatic, and chemoresistant properties. Understanding the exact molecular and signaling mechanisms that underlay malignant processes in PaCSCs is instrumental for developing novel therapeutic modalities that overcome the limitations of current therapeutic regimens. In this paper, we provide an updated review of the latest research in the field and summarize the current knowledge of PaCSCs characteristics, cellular metabolism, stemness, and drug resistance. We explore how the crosstalk between the TME and PaCSCs influences stemness. We also highlight some of the key signalling pathways involved in PaCSCs stemness and drug evasion. The aim of this review is to explore how PaCSCs develop, maintain their properties, and drive tumor relapse in PC. The last section explores some of the latest therapeutic strategies aimed at targeting PaCSCs.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias Pancreáticas , Humanos , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Células-Tronco Neoplásicas/metabolismo , Neoplasias Pancreáticas
3.
Semin Cancer Biol ; 69: 69-76, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-31954835

RESUMO

Gastric cancer represents a deadly malignancy worldwide, yet current therapeutic regimens remain ineffective. Nanoparticle (NP) -based solutions could allow the design of novel therapeutic methods to eliminate this fatal disease. NPs typically carry out a significant role in multifunctional, multimodal imaging, and drug delivery carriers. In the recent decade, they have emerged as candidate approaches for the design of novel treatment strategies. Tumor nanotherapeutics characteristically possess various distinct advantages compared to conventional anti-cancer medications, which suffer from nonspecific bio-distribution, low solubility, and poor bioavailability. In this review, we will discuss the application of NPs in diagnosis and controlled drug delivery in gastric cancer (GC). We will focus on various NPs-based strategies employed against GC.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Imagem Multimodal/métodos , Nanopartículas/administração & dosagem , Neoplasias Gástricas/tratamento farmacológico , Animais , Disponibilidade Biológica , Humanos , Nanopartículas/química , Neoplasias Gástricas/diagnóstico por imagem , Neoplasias Gástricas/patologia
4.
Semin Cancer Biol ; 69: 376-390, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-31301361

RESUMO

Female-specific cancers are the most common cancers in women worldwide. Early detection methods remain unavailable for most of these cancers, signifying that most of them are diagnosed at later stages. Furthermore, current treatment options for most female-specific cancers are surgery, radiation and chemotherapy. Although important milestones in molecularly targeted approaches have been achieved lately, current therapeutic strategies for female-specific cancers remain limited, ineffective and plagued by the emergence of chemoresistance, which aggravates prognosis. Recently, the application of nanotechnology to the medical field has allowed the development of novel nano-based approaches for the management and treatment of cancers, including female-specific cancers. These approaches promise to improve patient survival rates by reducing side effects, enabling selective delivery of drugs to tumor tissues and enhancing the uptake of therapeutic compounds, thus increasing anti-tumor activity. In this review, we focus on the application of nano-based technologies to the design of novel and innovative diagnostic and therapeutic strategies in the context of female-specific cancers, highlighting their potential uses and limitations.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Neoplasias dos Genitais Femininos/tratamento farmacológico , Nanomedicina , Nanopartículas/administração & dosagem , Animais , Feminino , Neoplasias dos Genitais Femininos/patologia , Humanos , Nanopartículas/química
5.
J Nanobiotechnology ; 20(1): 274, 2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35701781

RESUMO

The tumor microenvironment (TME) plays a key role in cancer development and emergence of drug resistance. TME modulation has recently garnered attention as a potential approach for reprogramming the TME and resensitizing resistant neoplastic niches to existing cancer therapies such as immunotherapy or chemotherapy. Nano-based solutions have important advantages over traditional platform and can be specifically targeted and delivered to desired sites. This review explores novel nano-based approaches aimed at targeting and reprogramming aberrant TME components such as macrophages, fibroblasts, tumor vasculature, hypoxia and ROS pathways. We also discuss how nanoplatforms can be combined with existing anti-tumor regimens such as radiotherapy, immunotherapy, phototherapy or chemotherapy to enhance clinical outcomes in solid tumors.


Assuntos
Nanopartículas , Neoplasias , Humanos , Fatores Imunológicos , Imunoterapia , Macrófagos , Neoplasias/tratamento farmacológico , Microambiente Tumoral
6.
Semin Cancer Biol ; 56: 149-167, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30314681

RESUMO

Pancreatic cancer (PC) is an aggressive carcinoma and the fourth cause of cancer deaths in Western countries. Although surgery is the most effective therapeutic option for PC, the management of unresectable, locally advanced disease is highly challenging. Our improved understanding of pancreatic tumor biology and associated pathways has led to the development of various treatment modalities that can control the metastatic spread of PC. This review intends to present trials of small molecule tyrosine kinase inhibitors (TKIs) in PC management and the troubles encountered due to inevitable acquired resistance to TKIs.


Assuntos
Neoplasias Pancreáticas/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais , Ensaios Clínicos como Assunto , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Terapia de Alvo Molecular , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Inibidores de Proteínas Quinases/farmacologia , Retratamento , Transdução de Sinais/efeitos dos fármacos , Padrão de Cuidado , Resultado do Tratamento
7.
Cancer ; 126(14): 3360-3371, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32383803

RESUMO

BACKGROUND: The induction of reactive oxygen species (ROS) represents a viable strategy for enhancing the activity of radiotherapy. The authors hypothesized that napabucasin would increase ROS via its ability to inhibit NAD(P)H:quinone oxidoreductase 1 and potentiate the response to chemoradiotherapy in rectal cancer via distinct mechanisms. METHOD: Proliferation studies, colony formation assays, and ROS levels were measured in HCT116 and HT29 cell lines treated with napabucasin, chemoradiation, or their combination. DNA damage (pγH2AX), activation of STAT, and downstream angiogenesis were evaluated in both untreated and treated cell lines. Finally, the effects of napabucasin, chemoradiotherapy, and their combination were assessed in vivo with subcutaneous mouse xenograft models. RESULTS: Napabucasin significantly potentiated the growth inhibition of chemoradiation in both cell lines. Napabucasin increased ROS generation. Inhibition of ROS by N-acetylcysteine decreased the growth inhibitory effect of napabucasin alone and in combination with chemoradiotherapy. Napabucasin significantly increased pγH2AX in comparison with chemoradiotherapy alone. Napabucasin reduced the levels of pSTAT3 and VEGF and inhibited angiogenesis through an ROS-mediated effect. Napabucasin significantly potentiated the inhibition of growth and blood vessel formation by chemoradiotherapy in mouse xenografts. CONCLUSION: Napabucasin is a radiosensitizer with a novel mechanism of action: increasing ROS production and inhibiting angiogenesis. Clinical trials testing the addition of napabucasin to chemoradiotherapy in rectal cancer are needed.


Assuntos
Benzofuranos/administração & dosagem , Quimiorradioterapia/métodos , Naftoquinonas/administração & dosagem , Radiossensibilizantes/administração & dosagem , Neoplasias Retais/metabolismo , Neoplasias Retais/terapia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Células HCT116 , Células HT29 , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Retais/patologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Carcinogenesis ; 39(4): 556-561, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29471417

RESUMO

Prostate cancer is one of the most common cancers in men worldwide. Currently available diagnostic and prognostic tools for this disease, such as prostate specific antigen, suffer from lack of specificity and sensitivity, resulting in over- and misdiagnosis. Hence, there is an urgent need for clinically relevant biomarkers capable of distinguishing between aggressive and nonaggressive forms of prostate cancer to aid in stratification, management and therapeutic decisions. To address this unmet need, we investigated the patterns of expression of a panel of 68 plasma-derived microRNAs (miRNAs) in a cohort of African American (AA) and European American (EA) prostate cancer patients (n = 114). miRNA qPCR results were analyzed using in-depth statistical methods, and a bioinformatics analysis was conducted to identify potential targets of the differentially expressed miRNAs. Our data demonstrate that a new previously unreported circulating miRNA signature consisting of a combination of interacting miRNAs (miR-17/miR-192) and an independent miRNA (miR-181a) are capable of segregating aggressive and nonaggressive prostate cancer in both AA and EA patients. The interacting miRNAs outperformed independent miRNAs in identifying aggressiveness. Our results suggest that these circulating miRNAs may constitute novel biomarkers of prostate cancer aggressiveness in both races and warrant further investigation.


Assuntos
Biomarcadores Tumorais/sangue , MicroRNAs/sangue , Neoplasias da Próstata/sangue , Adulto , Idoso , Biomarcadores Tumorais/genética , MicroRNA Circulante/análise , MicroRNA Circulante/genética , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
9.
Pharmacol Res ; 117: 303-327, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28087443

RESUMO

The traditional view on GPCRs held that they function as single monomeric units composed of identical subunits. This notion was overturned by the discovery that GPCRs can form homo- and hetero-oligomers, some of which are obligatory, and can further assemble into receptor mosaics consisting of three or more protomers. Oligomerisation exerts significant impacts on receptor function and physiology, offering a platform for the diversification of receptor signalling, pharmacology, regulation, crosstalk, internalization and trafficking. Given their involvement in the modulation of crucial physiological processes, heteromers could constitute important therapeutic targets for a wide range of diseases, including schizophrenia, Parkinson's disease, substance abuse or obesity. This review aims at depicting the current developments in GPCR oligomerisation research, documenting various class A, B and C GPCR heteromers detected in vitro and in vivo using biochemical and biophysical approaches, as well as recently identified higher-order oligomeric complexes. It explores the current understanding of dimerization dynamics and the possible interaction interfaces that drive oligomerisation. Most importantly, it provides an inventory of the wide range of physiological processes and pathophysiological conditions to which GPCR oligomers contribute, surveying some of the oligomers that constitute potential drug targets. Finally, it delineates the efforts to develop novel classes of ligands that specifically target and tether to receptor oligomers instead of a single monomeric entity, thus ameliorating their ability to modulate GPCR function.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Animais , Dimerização , Humanos , Ligantes , Subunidades Proteicas/metabolismo
10.
Clin Exp Pharmacol Physiol ; 44(6): 613-622, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28271563

RESUMO

The molecular diagnostics revolution has reshaped the practice of oncology by facilitating the identification of genetic, epigenetic and proteomic modifications correlated with cancer, thus delineating 'oncomaps' for various cancer types. These advances have enhanced our understanding of gastric cancer, one of the most fatal diseases worldwide, and culminated in the approval of novel molecular therapies such as trastuzumab. Gastric tumours display recurrent aberrations in key kinase oncogenes such as Her2, epidermal growth factor receptor (EGFR), PI3K, mTOR or c-Met, suggesting that these receptors are amenable to inhibition using specific drug agents. In this review, we examine the mutational landscape of gastric cancer, the use of kinase inhibitors as targeted therapies in gastric tumours and the clinical trials underway for novel inhibitors, highlighting successes, failures and future directions.


Assuntos
Terapia de Alvo Molecular/métodos , Proteínas Quinases/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/enzimologia , Ensaios Clínicos como Assunto , Humanos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Neoplasias Gástricas/patologia
11.
Biochim Biophys Acta Rev Cancer ; 1879(1): 189065, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38160899

RESUMO

Pancreatic cancer (PC) remains a leading cause of mortality worldwide due to the absence of early detection methods and the low success rates of traditional therapeutic strategies. Drug resistance in PC is driven by its desmoplastic stroma, which creates a barrier that shields cancer niches and prevents the penetration of drugs. The PC stroma comprises heterogeneous cellular populations and non-cellular components involved in aberrant ECM deposition, immunosuppression, and drug resistance. These components can influence PC development through intricate and complex crosstalk with the PC cells. Understanding how stromal components and cells interact with and influence the invasiveness and refractoriness of PC cells is thus a prerequisite for developing successful stroma-modulating strategies capable of remodeling the PC stroma to alleviate drug resistance and enhance therapeutic outcomes. In this review, we explore how non-cellular and cellular stromal components, including cancer-associated fibroblasts and tumor-associated macrophages, contribute to the immunosuppressive and tumor-promoting effects of the stroma. We also examine the signaling pathways underlying their activation, tumorigenic effects, and interactions with PC cells. Finally, we discuss recent pre-clinical and clinical work aimed at developing and testing novel stroma-modulating agents to alleviate drug resistance and improve therapeutic outcomes in PC.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Pâncreas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos Associados a Câncer/metabolismo
12.
Drug Discov Today ; 29(7): 104053, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38849028

RESUMO

Pancreatic cancer (PC), a disease with high heterogeneity and a dense stromal microenvironment, presents significant challenges and a bleak prognosis. Recent breakthroughs have illuminated the crucial interplay among RAS, epidermal growth factor receptor (EGFR), and hedgehog pathways in PC progression. Small molecular inhibitors have emerged as a potential solution with their advantages of oral administration and the ability to target intracellular and extracellular sites effectively. However, despite the US FDA approving over 100 small-molecule targeted antitumor drugs, challenges such as low response rates and drug resistance persist. This review delves into the possibility of using small molecules to treat persistent or spreading PC, highlighting the challenges and the urgent need for a diverse selection of inhibitors to develop more effective treatment strategies.


Assuntos
Antineoplásicos , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Animais , Resistencia a Medicamentos Antineoplásicos , Terapia de Alvo Molecular , Receptores ErbB/antagonistas & inibidores , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico
13.
J Control Release ; 370: 707-720, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38744346

RESUMO

Immunotherapy is gaining prominence as a promising strategy for treating triple-negative breast cancer (TNBC). Neoantigens (neoAgs) and cancer-testis antigens (CTAs) are tumor-specific targets originating from somatic mutations and epigenetic changes in cancer cells. These antigens hold great promise for personalized cancer vaccines, as supported by preclinical and early clinical evidence in TNBC. This review delves into the potential of neoAgs and CTAs as vaccine candidates, emphasizing diverse strategies and delivery approaches. It also highlights the current status of vaccination modalities undergoing clinical trials in TNBC therapy. A comprehensive understanding of neoAgs, CTAs, vaccination strategies, and innovative delivery methods is crucial for optimizing neoAg-based immunotherapies in clinical practice.


Assuntos
Antígenos de Neoplasias , Vacinas Anticâncer , Imunoterapia , Neoplasias de Mama Triplo Negativas , Humanos , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Antígenos de Neoplasias/imunologia , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/terapia , Feminino , Animais , Imunoterapia/métodos , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos/métodos
14.
Drug Discov Today ; 28(3): 103469, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36529353

RESUMO

Mitochondria are the powerhouses of cells and modulate the essential metabolic functions required for cellular survival. Various mitochondrial pathways, such as oxidative phosphorylation or production of reactive oxygen species (ROS) are dysregulated during cancer growth and development, rendering them attractive targets against cancer. Thus, the delivery of antitumor agents to mitochondria has emerged as a potential approach for treating cancer. Recent advances in nanotechnology have provided innovative solutions for overcoming the physical barriers posed by the structure of mitochondrial organelles, and have enabled the development of efficient mitochondrial nanoplatforms. In this review, we examine the importance of mitochondria during neoplastic development, explore the most recent smart designs of nano-based systems aimed at targeting mitochondria, and highlight key mitochondrial pathways in cancer cells.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Mitocôndrias/metabolismo , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo
15.
Cytokine Growth Factor Rev ; 71-72: 26-39, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37407355

RESUMO

Pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC), presents a challenging landscape due to its complex nature and the highly immunosuppressive tumor microenvironment (TME). This immunosuppression severely limits the effectiveness of immune-based therapies. Studies have revealed the critical role of immunometabolism in shaping the TME and influencing PDAC progression. Genetic alterations, lysosomal dysfunction, gut microbiome dysbiosis, and altered metabolic pathways have been shown to modulate immunometabolism in PDAC. These metabolic alterations can significantly impact immune cell functions, including T-cells, myeloid-derived suppressor cells (MDSCs), and macrophages, evading anti-tumor immunity. Advances in immunotherapy offer promising avenues for overcoming immunosuppressive TME and enhancing patient outcomes. This review highlights the challenges and opportunities for future research in this evolving field. By exploring the connections between immunometabolism, genetic alterations, and the microbiome in PDAC, it is possible to tailor novel approaches capable of improving immunotherapy outcomes and addressing the limitations posed by immunosuppressive TME. Ultimately, these insights may pave the way for improved treatment options and better outcomes for PDAC patients.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/terapia , Imunoterapia , Macrófagos/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
16.
J Immunother Cancer ; 11(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37852737

RESUMO

BACKGROUND: Preclinical studies showed metformin reduces exhaustion of tumor-infiltrating lymphocytes and potentiates programmed cell death protein-1 (PD-1) blockade. We hypothesized that metformin with nivolumab would elicit potent antitumor and immune modulatory activity in metastatic microsatellite stable (MSS) colorectal cancer (CRC). We evaluated this hypothesis in a phase II study. METHODS: Nivolumab (480 mg) was administered intravenously every 4 weeks while metformin (1000 mg) was given orally, two times per day following a 14-day metformin only lead-in phase. Patients ≥18 years of age, with previously treated, stage IV MSS CRC, and Eastern Cooperative Oncology Group 0-1, having received no prior anti-PD-1 agent were eligible. The primary endpoint was overall response rate with secondary endpoints of overall survival (OS) and progression-free survival (PFS). Correlative studies using paired pretreatment/on-treatment biopsies and peripheral blood evaluated a series of immune biomarkers in the tumor microenvironment and systemic circulation using ChipCytometry and flow cytometry. RESULTS: A total of 24 patients were enrolled, 6 patients were replaced per protocol, 18 patients had evaluable disease. Of the 18 evaluable patients, 11/18 (61%) were women and the median age was 58 (IQR 50-67). Two patients had stable disease, but no patients had objective response, hence the study was stopped for futility. Median OS and PFS was 5.2 months (95% CI (3.2 to 11.7)) and 2.3 months (95% CI (1.7 to 2.3)). Most common grade 3/4 toxicities: Anemia (n=2), diarrhea (n=2), and fever (n=2). Metformin alone failed to increase the infiltration of T-cell subsets in the tumor, but combined metformin and nivolumab increased percentages of tumor-infiltrating leukocytes (p=0.031). Dual treatment also increased Tim3+ levels in patient tissues and decreased naïve CD8+T cells (p=0.0475). CONCLUSIONS: Nivolumab and metformin were well tolerated in patients with MSS CRC but had no evidence of efficacy. Correlative studies did not reveal an appreciable degree of immune modulation from metformin alone, but showed trends in tumorous T-cell infiltration as a result of dual metformin and PD-1 blockade despite progression in a majority of patients.


Assuntos
Neoplasias Colorretais , Metformina , Humanos , Feminino , Pessoa de Meia-Idade , Masculino , Nivolumabe/efeitos adversos , Receptor de Morte Celular Programada 1 , Metformina/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Repetições de Microssatélites , Microambiente Tumoral
17.
Drug Discov Today ; 27(5): 1404-1410, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34952224

RESUMO

Pancreatic cancer (PC) is an aggressive disease characterized by high mortality. Diagnosis at advanced stage, resistance, and recurrence are major hurdles for PC therapy and contribute to poor survival rate. Mutations in tumor-promoting kinases and epigenetic dysregulation in tumor suppressor genes are hallmarks of PC and can be used for diagnosis and therapy. In this review, we highlight dysregulated genes associated with epigenetic mechanisms, including DNA methylation and histone acetylation, involved in PC progression and resistance. We also explore epigenetic drugs currently in clinical trials. Combining epigenetic drugs and targeted therapies might represent a promising approach for PC.


Assuntos
Epigenômica , Neoplasias Pancreáticas , Metilação de DNA , Epigênese Genética , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas
18.
Life Sci ; 308: 120970, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36115581

RESUMO

Pyroptosis represents an inflammatory cell death form induced by inflammasomes and performed by gasdermins. It is characterized by swelling, pore formation, release of cellular content and the activation of innate immunity leading to inflammation. Hence, pyroptosis contributes to inflammatory conditions like cancer and has emerged as a promising immuno-strategy for treating cancer. The advent of nanotechnology, which overlaps with the discovery of pyroptotic cell death, has enabled the development of nano-based pyroptosis inducing platforms aimed at overcoming resistance to apoptosis and enhancing tumor immunity. In this paper, we will describe the various molecular pathways underlying pyroptosis, such as canonical and non-canonical pyroptosis. We will then explore the advances in the field of pyroptosis-based nanotherapeutics and their future implications.


Assuntos
Neoplasias , Piroptose , Apoptose , Morte Celular , Humanos , Inflamassomos/metabolismo , Inflamação/tratamento farmacológico , Neoplasias/tratamento farmacológico , Piroptose/fisiologia
19.
World J Biol Chem ; 12(2): 15-37, 2021 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-33815682

RESUMO

The pathology and physiology of breast cancer (BC), including metastasis, and drug resistance, is driven by multiple signaling pathways in the tumor microenvironment (TME), which hamper antitumor immunity. Recently, long non-coding RNAs have been reported to mediate pathophysiological develop-ments such as metastasis as well as immune suppression within the TME. Given the complex biology of BC, novel personalized therapeutic strategies that address its diverse pathophysiologies are needed to improve clinical outcomes. In this review, we describe the advances in the biology of breast neoplasia, including cellular and molecular biology, heterogeneity, and TME. We review the role of novel molecules such as long non-coding RNAs in the pathophysiology of BC. Finally, we provide an up-to-date overview of anticancer compounds extracted from marine microorganisms, crustaceans, and fishes and their synergistic effects in combination with other anticancer drugs. Marine compounds are a new discipline of research in BC and offer a wide range of anti-cancer effects that could be harnessed to target the various pathways involved in BC development, thus assisting current therapeutic regimens.

20.
J Biomol Struct Dyn ; 39(8): 2914-2922, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32306846

RESUMO

Nuclear factor κB (NF-κB), a dimeric transcription factor, is a major regulator and an important determinant of the biological characteristics of tumour cells. Some antioxidants or protease inhibitors have been found to act against NF-κB to suppress colorectal cancer (CRC). In the current investigation, a computational study was performed to investigate the molecular interaction between NF-κB and resveratrol. Molecular docking studies revealed that, resveratrol with NF-κB are predicted to be quite effective. The application of molecular dynamics simulation (MDS) tactics has considerably supported in increasing the prediction precision of the outcomes. Further, this study revealed that NF-κB could be a potential target for various anti-cancerous drugs for cancer therapeutics. Furthermore, animal investigations are necessary to confirm the efficacy and evaluate potency of target and drugs.Communicated by Ramaswamy H. Sarma.


Assuntos
Neoplasias Colorretais , Estilbenos , Antioxidantes , Neoplasias Colorretais/tratamento farmacológico , Humanos , Simulação de Acoplamento Molecular , NF-kappa B , Resveratrol , Estilbenos/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa