Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Curr Drug Deliv ; 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38591208

RESUMO

Macrophages are immune cells with high heterogeneity and plasticity, crucial for recognizing and eliminating foreign substances, including cancer cells. However, cancer cells can evade the immune system by producing signals that cause macrophages to switch to a pro-tumor phenotype, promoting tumor growth and progression. Tumor-associated macrophages, which infiltrate into tumor tissue, are important immune cells in the tumor microenvironment and can regulate cancer's growth, invasion, and metastasis by inhibiting tumor immunity. This review article highlights the characteristics of tumor-associated macrophages and their role in the occurrence and development of cancer. It outlines how reprogramming macrophages towards an anti-tumor phenotype can improve the response to cancer therapy. Explore the intricate process of engineered nanoparticles serving as carriers for immunostimulatory molecules, activating macrophages to instigate an anti-tumor response. Finally, it summarizes several studies demonstrating targeting macrophages is a potential in preclinical cancer models. Several challenges must be addressed in developing effective macrophage-targeted therapies, such as the heterogeneity of macrophage subtypes and their plasticity. Further research is needed to understand the mechanisms underlying macrophage function in the tumor microenvironment and identify novel targets for macrophage-directed therapies. Targeting macrophages is a promising and innovative approach to improving cancer therapy and patient outcomes.

2.
Cancer Res ; 84(8): 1237-1251, 2024 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-38241700

RESUMO

Epigenetic alterations at cis-regulatory elements (CRE) fine-tune transcriptional output. Epigenetic readers interact with CREs and can cooperate with other chromatin regulators to drive oncogene transcription. Here, we found that the YEATS domain-containing histone acetylation reader ENL (eleven-nineteen leukemia) acts as a key regulator of super-enhancers (SE), which are highly active distal CREs, across cancer types. ENL occupied the majority of SEs with substantially higher preference over typical enhancers, and the enrichment of ENL at SEs depended on its ability to bind acetylated histones. Rapid depletion of ENL by auxin-inducible degron tagging severely repressed the transcription of SE-controlled oncogenes, such as MYC, by inducing the decommissioning of their SEs, and restoring ENL protein expression largely reversed these effects. Additionally, ENL was indispensable for the rapid activation of SE-regulated immediate early genes in response to growth factor stimulation. Furthermore, ENL interacted with the histone chaperone FACT complex and was required for the deposition of FACT over CREs, which mediates nucleosome reorganization required for transcription initiation and elongation. Proper control of transcription by ENL and ENL-associated FACT was regulated by the histone reader BRD4. ENL was overexpressed in colorectal cancer and functionally contributed to colorectal cancer growth and metastasis. ENL degradation or inhibition synergized with BET inhibitors that target BRD4 in restraining colorectal cancer progression. These findings establish the essential role of epigenetic reader ENL in governing SE-driven oncogenic transcription and uncover the potential of ENL intervention to increase sensitivity to BET inhibition. SIGNIFICANCE: ENL plays a key role in decoding epigenetic marks at highly active oncogenic super-enhancers and can be targeted in combination with BET inhibition as a promising synergistic strategy for optimizing cancer treatment.


Assuntos
Neoplasias Colorretais , Histonas , Humanos , Histonas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Nucleares/metabolismo , Epigênese Genética , Neoplasias Colorretais/genética , Proteínas que Contêm Bromodomínio , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-38213150

RESUMO

Chimeric antigen receptor T-cells, known as CAR-T cells, represent a promising breakthrough in the realm of adoptive cell therapy. These T-cells are genetically engineered to carry chimeric antigen receptors that specifically target tumors. They have achieved notable success in the treatment of blood-related cancers, breathing new life into this field of medical research. However, numerous obstacles limit chimeric antigen receptors T-cell therapy's efficacy, such as it cannot survive in the body long. It is prone to fatigue and exhaustion, leading to difficult tumor elimination and repeated recurrence, affecting solid tumors and hematological malignancies. The challenges posed by solid tumors, especially in the context of the complex solid-tumor microenvironment, require specific strategies. This review outlines recent advancements in improving chimeric antigen receptors T-cell therapy by focusing on the chimeric antigen receptors protein, modifying T-cells, and optimizing the interaction between T-cells and other components within the tumor microenvironment. This article aims to provide an extensive summary of the latest discoveries regarding CAR-T cell therapy, encompassing its application across various types of human cancers. Moreover, it will delve into the obstacles that have emerged in recent times, offering insights into the challenges faced by this innovative approach. Finally, it highlights novel therapeutic options in treating hematological and solid malignancies with chimeric antigen receptors T-cell therapies.

4.
Curr Drug Deliv ; 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38275044

RESUMO

Pulmonary, nasal, and nose-to-brain diseases involve clinical approaches, such as bronchodilators, inhaled steroids, oxygen therapy, antibiotics, antihistamines, nasal steroids, decongestants, intranasal drug delivery, neurostimulation, and surgery to treat patients. However, systemic medicines have serious adverse effects, necessitating the development of inhaled formulations that allow precise drug delivery to the airways with minimum systemic drug exposure. Particle size, surface charge, biocompatibility, drug capacity, and mucoadhesive are unique chemical and physical features that must be considered for pulmonary and nasal delivery routes due to anatomical and permeability considerations. The traditional management of numerous chronic diseases has a variety of drawbacks. As a result, targeted medicine delivery systems that employ nanotechnology enhancer drug efficiency and optimize the overall outcome are created. The pulmonary route is one of the most essential targeted drug delivery systems because it allows the administering of drugs locally and systemically to the lungs, nasal cavity, and brain. Furthermore, the lungs' beneficial characteristics, such as their ability to inhibit first-pass metabolism and their thin epithelial layer, help treat several health complications. The potential to serve as noninvasive self-administration delivery sites of the lung and nasal routes is discussed in this script. New methods for treating respiratory and some systemic diseases with inhalation have been explored and highlight particular attention to using specialized nanocarriers for delivering various drugs via the nasal and pulmonary pathways. The design and development of inhaled nanomedicine for pulmonary, nasal, and respiratory medicine applications is a potential approach for clinical translation.

5.
Artigo em Inglês | MEDLINE | ID: mdl-38116189

RESUMO

A novel and rapid therapeutic approach is the treatment of human breast cancer by enhancing the host's immune system. In initial findings, program death one (PD-1) and program cell death ligand one (PD-L1) showed positive results towards solid tumors, but tumor relapse and drug resistance are the major concerns. Breast cancer therapy has been transformed by the advent of immune checkpoint blockades (ICBs). Triple-negative breast cancers (TNBCs) have exhibited enduring responses to clinical usage of immune checkpoint inhibitors (ICBs) like atezolizumab and pembrolizumab. Nonetheless, a notable proportion of individuals with TNBC do not experience advantages from these treatments, and there is limited comprehension of the resistance mechanisms. Another approach to overcome resistance is cancer stem cells (CSCs), as these cells are crucial for the initiation and growth of tumors in the body. Various cancer vaccines are created using stem cells (dendritic, whole cell, bacterial) and focus primarily on targeting tumor-related antigens. The ultimate objective of cancer vaccines is to immunize the patients by active artificial immunity against cancer, though. In this review, we primarily focused on existing immunotherapeutic options, immune checkpoint blockers, the latest progress in understanding the molecular mechanisms underlying resistance to immune checkpoint inhibitors (ICBs), advanced strategies to overcome resistance to ICBs, cancer stem cell antigens and molecular markers, ongoing clinical trials for BCs and cancer vaccines for breast cancer.

6.
Oncol Lett ; 26(3): 372, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37965160

RESUMO

Immunotherapy has become one of the most promising approaches in tumor therapy, and there are numerous associated clinical trials in China. As an immunosuppressive tumor, head and neck squamous cell carcinoma (HNSCC) carries a high mutation burden, making immune checkpoint inhibitors promising candidates in this field due to their unique mechanism of action. The present review outlines a comprehensive multidisciplinary cancer treatment approach and elaborates on how combining immunochemotherapy and immunoradiotherapy guidelines could enhance clinical efficacy in patients with HNSCC. Furthermore, the present review explores the immunology of HNSCC, current immunotherapeutic strategies to enhance antitumor activity, ongoing clinical trials and the future direction of the current immune landscape in HNSCC. Advanced-stage HNSCC presents with a poor prognosis, low survival rates and minimal improvement in patient survival trends over time. Understanding the potential of immunotherapy and ways to combine it with surgery, chemotherapy and radiotherapy confers good prospects for the management of human papillomavirus (HPV)-positive HNSCC, as well as other HPV-positive malignancies. Understanding the immune system and its effect on HNSCC progression and metastasis will help to uncover novel biomarkers for the selection of patients and to enhance the efficacy of treatments. Further research on why current immune checkpoint inhibitors and targeted drugs are only effective for some patients in the clinic is needed; therefore, further research is required to improve the overall survival of affected patients.

7.
Oncol Lett ; 26(1): 281, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37274466

RESUMO

Chimeric antigen receptor (CAR) T cell therapy has emerged as a new and breakthrough cancer immunotherapy. Although CAR-T cell therapy has made significant progress clinically in patients with refractory or drug-resistant hematological malignancies, there are numerous challenges in its application to solid tumor therapy, including antigen escape, severe toxic reactions, abnormal vascularization, tumor hypoxia, insufficient infiltration of CAR-T cells and immunosuppression. As a conventional mode of anti-tumor therapy, radiotherapy has shown promising effects in combination with CAR-T cell therapy by enhancing the specific immunity of endogenous target antigens, which promoted the infiltration and expansion of CAR-T cells and improved the hypoxic tumor microenvironment. This review focuses on the obstacles to the application of CAR-T technology in solid tumor therapy, the potential opportunities and challenges of combined radiotherapy and CAR-T cell therapy, and the review of recent literature to evaluate the best combination for the treatment of solid tumors.

8.
Biomed Pharmacother ; 162: 114658, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37031495

RESUMO

Cancer metabolism is how cancer cells utilize nutrients and energy to support their growth and proliferation. Unlike normal cells, cancer cells have a unique metabolic profile that allows them to generate energy and the building blocks they need for rapid growth and division. This metabolic profile is marked by an increased reliance on glucose and glutamine as energy sources and changes in how cancer cells use and make key metabolic intermediates like ATP, NADH, and NADPH. This script analyzes a comprehensive overview of the latest advances in tumor metabolism, identifying the key unresolved issues, elaborates on how tumor cells differ from normal cells in their metabolism of nutrients, and explains how tumor cells conflate growth signals and nutrients to proliferate. The metabolic interaction of tumorigenesis and lipid metabolism within the tumor microenvironment and the role of ROS as an anti-tumor agent by mediating various signaling pathways for clinical cancer therapeutic targeting are outlined. Cancer metabolism is highly dynamic and heterogeneous; thus, advanced technologies to better investigate metabolism at the unicellular level without altering tumor tissue are necessary for better research and clinical transformation. The study of cancer metabolism is an area of active research, as scientists seek to understand the underlying metabolic changes that drive cancer growth and to identify potential therapeutic targets.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Metabolismo Energético , Glicólise , Transdução de Sinais , Microambiente Tumoral
9.
Br J Pharmacol ; 180(13): 1748-1765, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36721985

RESUMO

BACKGROUND AND PURPOSE: Isoxazole 9 (ISX9) is a neurogenesis-promoting small molecule compound that can up-regulate the expression of NeuroD1 and induce differentiation of neuronal, cardiac and islet endocrine progenitors. So far, the molecular mechanisms underlying the action of ISX9 still remain elusive. EXPERIMENTAL APPROACH: To identify a novel agonist of the Wnt/ß-catenin, a cell-based SuperTOPFlash reporter system was used to screen known-compound libraries. An activation effect of ISX9 on the Wnt/ß-catenin pathway was analysed with the SuperTOPFlash or SuperFOPFlash reporter system. Effects of ISX9 on Axin1/LRP6 interaction were examined using a mammalian two-hybrid system, co-immunoprecipitation, microscale thermophoresis, emission spectra and mass spectrometry assays. The expression of Wnt target and stemmness marker genes were evaluated with real-time PCR and immunoblotting. In vivo hair regeneration abilities of ISX9 were analysed by immunohistochemical staining, real-time PCR and immunoblotting in hair regrowth model using C57BL/6J mice. KEY RESULTS: In this study, ISX9 was identified as a novel agonist of the Wnt/ß-catenin pathway. ISX9 targeted Axin1 by covalently binding to its N-terminal region and potentiated the LRP6-Axin1 interaction, thereby resulting in the stabilization of ß-catenin and up-regulation of Wnt target genes and stemmness marker genes. Moreover, the topical application of ISX9 markedly promoted hair regrowth in C57BL/6J mice and induced hair follicle transition from telogen to anagen via enhancing Wnt/ß-catenin pathway. CONCLUSIONS AND IMPLICATIONS: Taken together, our study unravelled that ISX9 could activate Wnt/ß-catenin signalling by potentiating the association between LRP6 and Axin1, and may be a promising therapeutic agent for alopecia treatment.


Assuntos
Via de Sinalização Wnt , beta Catenina , Camundongos , Animais , Proteína Axina/genética , Proteína Axina/metabolismo , Proteína Axina/farmacologia , beta Catenina/metabolismo , Camundongos Endogâmicos C57BL , Cabelo , Mamíferos/metabolismo
10.
Curr Cancer Drug Targets ; 23(4): 251-264, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36278447

RESUMO

The previous year's worldview for cancer treatment has advanced from general to more precise therapeutic approaches. Chemotherapies were first distinguished as the most reliable and brief therapy with promising outcomes in cancer patients. However, patients could also suffer from severe toxicities resulting from chemotherapeutic drug usage. An improved comprehension of cancer pathogenesis has led to new treatment choices, including tumor-targeted therapy and immunotherapy. Subsequently, cancer immunotherapy and targeted therapy give more hope to patients since their combination has tremendous therapeutic efficacy. The immune system responses are also initiated and modulated by targeted therapies and cytotoxic agents, which create the principal basis that when targeted therapies are combined with immunotherapy, the clinical outcomes are of excellent efficacy, as presented in this review. This review focuses on how immunotherapy and targeted therapy are applicable in cancer management and treatment. Also, it depicts promising therapeutic results with more extensive immunotherapy applications with targeted therapy. Further elaborate that immune system responses are also initiated and modulated by targeted therapies and cytotoxic agents, which create the principal basis that this combination therapy with immunotherapy can be of great outcome clinically.


Assuntos
Imunoterapia , Neoplasias , Humanos , Terapia Combinada , Neoplasias/tratamento farmacológico , Citotoxinas
11.
Artigo em Inglês | MEDLINE | ID: mdl-35260063

RESUMO

There has been a great amount of advancement in the early field of nano-immunotherapy and combination therapy. Persistent consideration regarding the clinical challenges and therapeutic hindrance should be tended to achieve therapeutic efficacy and potential. In this review, we will address how nanotechnology could defeat the difficulties resulting from cancer immunotherapy, how nanoparticles' utilization can enhance the efficacy of immune checkpoint blockers, and reconstituting the tumor microenvironment can promote antitumor responses. Moreover, this review discusses how nanoparticles mediate therapeutic modalities like chemotherapy, photodynamic therapy, photothermal therapy, and radiotherapy, which are used to target and destroy cancerous cells, initiate the release of tumor antigens, and can trigger anti-tumor immunity reactions. Furthermore, we analyzed the potential benefits of immunotherapy combinatorial using the nanoparticle delivery system to prevent tumor recurrence, hinder metastases, and decrease systemic toxicity of major organs and healthy cells common with uncontrolled targeting.


Assuntos
Imunoterapia , Nanotecnologia , Neoplasias , Humanos , Neoplasias/tratamento farmacológico
12.
Cancer Med ; 11(23): 4457-4468, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35522104

RESUMO

Tumors are presently a major threat to human life and health. Malignant tumors are conventionally treated through radiotherapy and chemotherapy. However, traditional therapies yield unsatisfactory results due to high toxicity to the normal cells, inability to treat deep tumor tissues, and the possibility of inducing drug resistance in the tumor cells. This has caused immunotherapy to emerge as an effective and alternate treatment strategy. To overcome the limitations of the conventional treatments as well as to avert the risk of various drug resistance and cytotoxicity, bacterial anti-tumor immunotherapy has raised the interest of researchers. This therapeutic strategy employs bacteria to specifically target and colonize the tumor tissues with preferential accumulation and proliferation. Such bacterial accumulation initiates a series of anti-tumor immune responses, effectively eliminating the tumor cells. This immunotherapy can use the bacteria alone or concomitantly with the other methods. For example, the bacteria can deliver the anti-cancer effect mediators by regulating the expression of the bacterial genes or by synthesizing the bioengineered bacterial complexes. This review will discuss the mechanism of utilizing bacteria in treating tumors, especially in terms of immune mechanisms. This could help in better integrating the bacterial method with other treatment options, thereby, providing a more effective, reliable, and unique treatment therapy for tumors.


Assuntos
Imunoterapia , Neoplasias , Humanos , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Bactérias/metabolismo , Terapia Combinada
13.
Int J Biol Macromol ; 193(Pt A): 228-237, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34688683

RESUMO

Conventional chemotherapeutic drugs may cause serious side effects such as hepatotoxicity and renal toxicity due to lack of targeting, which affects therapy outcome and the prognosis of patients. Therefore, biomimetic nanoparticles with long blood circulation and active targeting have attracted increasing attention. In this work, we fabricated a biomimetic R-RBC@GEF-NPs nano-system by encapsulating gefitinib-loaded albumin nanoparticles (GEF-NPs) inside cRGD-modified red blood cell (RBC) membranes. The complete RBC membrane structure and membrane proteins enabled the NPs to escape phagocytosis by macrophages. In addition, the cRGD moiety significantly improved tumor cell targeting and uptake. R-RBC@GEF-NPs inhibited the growth of A549 cells in vitro in a dose- and time-dependent manner by inducing apoptosis and cell cycle arrest at the G1 phase. Likewise, the R-RBC@GEF-NPs also decreased tumor weight and volume in the mice injected with A549 cells and prolonged survival time. In addition, the 99Tc-labeled R-RBC@GEF-NPs selectively accumulated in the tumor tissues in vivo, and enabled real time tumor imaging. Finally, blood and histological analyses showed that R-RBC@GEF-NPs did not cause any obvious systemic toxicity. Taken together, the biomimetic R-RBC@GEF-NPs is a promising therapeutic formulation for the treatment of lung cancer.


Assuntos
Albuminas/química , Portadores de Fármacos , Gefitinibe/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Nanopartículas/administração & dosagem , Células A549 , Animais , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Humanos , Camundongos , Camundongos Nus
14.
Front Genet ; 12: 678747, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34386037

RESUMO

BACKGROUND: Chemokines are implicated in tumor microenvironment (TME) cell infiltration. Development of ovarian cancer involves heterologous cells together with the adjacent microenvironment. Nonetheless, our understanding of the chemokine-related TME characteristics in ovarian cancer remains obscure. METHODS: In this large-scale multi-platform study of 10 microarray datasets consisting of 1,673 ovarian cancer patients, we comprehensively evaluated CXCL10 and CXCL9 expression risk classifications for predicting overall survival (OS) and TME immune characteristics. The cross-validation between a standard cohort (TCGA: The Cancer Genome Atlas) and three test cohorts (GEO: Gene-Expression Omnibus) was applied. We investigated differences in the biological functions and the underlying mechanisms between high- and low-risk classifications. RESULTS: We identified that evaluation of CXCL10 expression could predict the tumor development, immune cell infiltration, TME signature, genetic alteration, and patient prognosis in ovarian cancer. Low-risk classification was characterized by high CXCL10 expression and prolonged prognosis, which was positively associated with specific immune cell infiltration (i.e., T cells, DCs, aDC, and Th2 cells) and TME immune-relevant signatures. Meanwhile, the high-risk classification was defined by lower CXCL10/CXCL9 expression and relevant poor prognosis and immune infiltrations. The CXCL10-based low-risk classification was also linked to antitumor biological function of specific immune gene sets, such as IL2-STAT5 signaling. Additionally, a mutational pattern featured by enrichment of C > T transition was further identified to be associated with immune cell infiltration. CONCLUSIONS: This work proposed a promising biomarker for evaluating TME immune characteristics and clinical outcomes in patients with ovarian cancer. Estimation of CXCL10 risk pattern sheds a novel insight on ovarian cancer TME immune characteristics and provides strategies for ovarian cancer immunotherapy.

15.
Transl Oncol ; 14(8): 101118, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34020371

RESUMO

The history of low-dose total-body irradiation (LTBI) as a means of radiotherapy for treating malignant tumors can be traced back to the 1920s. Despite this very low total dose, LTBI can induce long-term remissions. Tumor cells are known to change and maintain their own survival and development conditions through autocrine and paracrine signaling. LTBI can change the tumor microenvironment, enhance the infiltration of activated T cells, and trigger inflammatory processes. LTBI-mediated immune response can exert systemic long-term anti-tumor effects, and can induce tumor regression at the primary site and metastatic sites. With a continuous improvement in the anti-tumor immune microenvironment in the field of tumor therapy, LTBI provides more choices to comprehensively treat of tumors. The present study aimed to explore the experimental research mechanism of LTBI and immune microenvironment, and discuss the difficulties and development prospects of applying LTBI to tumor treatment.

16.
Int J Nanomedicine ; 16: 2389-2404, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33790556

RESUMO

Recently, there has been an incredible increase in research about the abnormal growth of cells (neoplasm), focusing on the management, treatment and preventing reoccurrence. It has been understood that the natural defense system, composed of a variety of immune defensive cells, does not just limit its function in eliminating neoplastic cells, but also controls the growth and spread of tumor cells of different kinds to other parts of the body. Cancer immunotherapy, is a cancer treatment plan that educates the body's defensive system to forestall, control, and eliminate tumor cells. The effectiveness of immunotherapy is achieved, to its highest efficacy, by the use of nanoparticles (NPs) for precise and timely delivery of immunotherapies to specific targeted neoplasms, with less or no harm to the healthy cells. Immunotherapies have been affirmed in clinical trials as a cancer regimen for various types of cancers, the side effects resulting from imprecise and non-targeted conveyance is well managed with the use of nanoparticles. Nonetheless, we will concentrate on enhancing cancer immunotherapy approaches by the use of nanoparticles for the productivity of antitumor immunity. Nanoparticles will be presented and utilized as an objective immunotherapy delivery system for high exactness and are thus a promising methodology for cancer treatment.


Assuntos
Sistemas de Liberação de Medicamentos , Objetivos , Imunoterapia , Nanopartículas/química , Neoplasias/imunologia , Neoplasias/terapia , Animais , Humanos , Nanopartículas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA