Current cancer drug targets最新文献

筛选
英文 中文
Cancer Immunotherapies: Navigating the Immune Landscape.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-03-06 DOI: 10.2174/0115680096346735250218110708
Rakesh Kumar, Joseph Finian Maleme, Shubham Kumar, Kalvatala Sudhakar, Darshan R Telange, Oshin Maiti, Pratibha Sahoo, Alaa A A Aljabali, Vijay Mishra, Yachana Mishra
{"title":"Cancer Immunotherapies: Navigating the Immune Landscape.","authors":"Rakesh Kumar, Joseph Finian Maleme, Shubham Kumar, Kalvatala Sudhakar, Darshan R Telange, Oshin Maiti, Pratibha Sahoo, Alaa A A Aljabali, Vijay Mishra, Yachana Mishra","doi":"10.2174/0115680096346735250218110708","DOIUrl":"https://doi.org/10.2174/0115680096346735250218110708","url":null,"abstract":"<p><p>Biotechnology has paved the way for the development of cancer therapeutics that harness biological systems. Cancer immunotherapy (CI) is a pivotal and swiftly progressing therapeutic modality, alongside surgical intervention, cytotoxic chemo-therapy, radiation therapy, and targeted therapy. Therefore, this is the fifth cornerstone of cancer management. Biotechnological pharmaceuticals are superior modalities for combat-ing neoplastic conditions when juxtaposed with conventional chemical therapeutics. Consid-ering empirical evidence, it can be posited that biotechnology exhibits a heightened level of precision in targeting malignant cells associated with cancer, thereby minimizing collateral damage compared to conventional chemotherapeutic approaches. Furthermore, this ap-proach harnesses the inherent capabilities of the immune system to impede cancer recur-rence, thereby facilitating a more proactive therapeutic intervention, as opposed to a mere deleterious remedy. Novel cancer immunotherapeutic medications, along with uncomplicat-ed methodologies that enable the quantitative evaluation of the effectiveness of compounds capable of modifying T cell-mediated, tumor antigen-specific immune responses, play a pivotal role in the assessment process. This highlights the considerable promise of immuno-therapies, monoclonal antibodies, and an array of biotechnological products in a relentless battle against cancer.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-03-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143572410","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pharmacological Evaluation of Amorphophalli rhizoma to Inhibit the Progression of Estrogen Receptor+ (ER+) Breast Cancer by Modulating the PI3K/AKT Cell Signaling Pathway.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-03-05 DOI: 10.2174/0115680096360994250106082347
Hailong Li, Qinghong Yu, Jiaqing Song, Haining Ding, Yian Chen, Ying Jin, Hongting Wu, Liaqat Hussain, Xiufei Gao
{"title":"Pharmacological Evaluation of Amorphophalli rhizoma to Inhibit the Progression of Estrogen Receptor+ (ER+) Breast Cancer by Modulating the PI3K/AKT Cell Signaling Pathway.","authors":"Hailong Li, Qinghong Yu, Jiaqing Song, Haining Ding, Yian Chen, Ying Jin, Hongting Wu, Liaqat Hussain, Xiufei Gao","doi":"10.2174/0115680096360994250106082347","DOIUrl":"https://doi.org/10.2174/0115680096360994250106082347","url":null,"abstract":"<p><strong>Introduction: </strong>Breast Cancer (BC) is one of the most prevalent malignant tumors in women. The incidence of estrogen receptor-positive (ER+) breast cancer is as high as 70%, and it is increasing. Amorphophalli rhizoma (APR) has the potential to be used in breast cancer.</p><p><strong>Aims: </strong>The objectives of the present study were to explore the impact of different APR extracts on the proliferation, migration, and invasion of ER+BC and to investigate their possible mechanism at the molecular level.</p><p><strong>Methods: </strong>Various extracts of APR were prepared in different solvents, such as petroleum ether, ethyl acetate, n-butanol, and water. ER+ T47D breast cancer cell lines were acquired and uti-lized to assess the effect of APR extracts on ER+ BC. Cell viability was assessed using the cell counting kit8 (CCk8) method, while anti-invasive and migratory effects were examined by transwell and wound healing assay. All the extracts were initially screened, and the ethyl acetate fraction (APR-EA) was found to be the most effective. Ultra High-Performance Liquid Chro-matography (UHPLC) of APR-EAE extract revealed the presence of various phytochemicals, such as succinic acid, 2-methoxy resorcinol, penicillic acid, morphine, salicylic acid, α-linolenic acid, and linolenic acid. Flow cytometry, western blot, and immunohistochemistry were used to explore molecular mechanisms.</p><p><strong>Results: </strong>APR-EA demonstrated anti-proliferative, anti-migratory, and anti-invasive effects on the ER+ T47D cell line. Thus, APR-EAE might inhibit the expression of P-PI3K/PI3K and P-Akt/Akt proteins, which subsequently represses the expression of ERα. This inhibition affects the downstream expression of the proteins CDK4 and Bcl-2, which are linked to cell growth and apoptosis.</p><p><strong>Conclusion: </strong>Additionally, APR-EA might increase the expression of P21 and Bax proteins, which are associated with cell cycle arrest and apoptosis. Overall, these effects contribute to the anti-ER+ breast cancer properties of APR-EA.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143566434","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Embracing the Circular Journey of RNA in Colorectal Cancer.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-03-03 DOI: 10.2174/0115680096324880250117062148
AmirHosein Barjasteh, Ayat Heidar Abdolamir, Abdulridha Mohammed Al-Asady, Hanieh Latifi, Amir Avan, Majid Khazaei, Mikhail Ryzhikov, Seyed Mahdi Hassanian
{"title":"Embracing the Circular Journey of RNA in Colorectal Cancer.","authors":"AmirHosein Barjasteh, Ayat Heidar Abdolamir, Abdulridha Mohammed Al-Asady, Hanieh Latifi, Amir Avan, Majid Khazaei, Mikhail Ryzhikov, Seyed Mahdi Hassanian","doi":"10.2174/0115680096324880250117062148","DOIUrl":"https://doi.org/10.2174/0115680096324880250117062148","url":null,"abstract":"<p><p>Recent investigations have indicated that circRNAs (circular RNAs), a novel member of the Noncoding RNA family, are a crucial genetic factor in Colorectal Cancer (CRC), development. These circRNAs have a range of biological functions, including regu-lating the expression of target genes, altering protein activity, and producing proteins. Addi-tionally, circRNAs significantly affect tumor cell proliferation, aggression, migration, and cell death. CircRNAs have an essential role in carcinogenesis, metastasis, and drug toler-ance. Various signaling pathways are linked by circRNAs, providing opportunities for a more tailored chemotherapy regimen as well as early diagnosis and prognosis determination. By identifying new and potent circRNAs in signaling pathways, we can aim for them with ASO or siRNAs and provide a powerful combination therapy. In this review, we strive to summarize the biological and carcinogenic functions of circRNAs and their impact on spe-cific signaling pathways like PI3K/AKT, MAPK, Notch, JAK/STAT, Hippo/YAP, and WNT/β-catenin which lead to Epithelial-mesenchymal-transition(EMT) in CRC. Further-more, we explore the clinical significance of circRNAs and important signaling proteins in treating CRC.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143540251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Using Cyclic Ketones to Synthesize New 3,5,6,7,8,9-Hexahydro-pyrazolo[1,5-a]quinoline Derivatives with Antiproliferative Activities: Morphological Studies.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-02-28 DOI: 10.2174/0115680096356497250210064209
Rafat M Mohareb, Marwa S Gamaan, Nadia Y Megally Abdo, Ibram R Mikhail
{"title":"Using Cyclic Ketones to Synthesize New 3,5,6,7,8,9-Hexahydro-pyrazolo[1,5-a]quinoline Derivatives with Antiproliferative Activities: Morphological Studies.","authors":"Rafat M Mohareb, Marwa S Gamaan, Nadia Y Megally Abdo, Ibram R Mikhail","doi":"10.2174/0115680096356497250210064209","DOIUrl":"https://doi.org/10.2174/0115680096356497250210064209","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Quinoline derivatives, often incorporating other heterocyclic structures, have shown a wide range of therapeutic potential, especially in the treatment of cancer. These compounds have demonstrated significant anticancer activity against various cell lines, including HeLa (human cervical cancer) and MDA-MB-435 (melanoma), exhibiting strong inhibitory effects.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Objective: &lt;/strong&gt;In this study, arylhydrazonopyrazole derivatives (3a-c) were employed in a series of multicomponent reactions to synthesize 3,5,6,7,8,9-hexahydropyrazolo[1,5-a]quinoline and pyran derivatives. Pyrazolo[1,5-a]quinoline derivatives, due to their structural properties, are considered valuable scaffolds for the development of novel drugs targeting similar biological pathways, with the potential for improved therapeutic efficacy. This study aimed to demonstrate the use of simple arylhydrazonopyrazole derivatives in multicomponent reactions with cyclic ketones and aromatic aldehydes. The resulting compounds were then assessed for their cytotoxic and antiproliferative activities. Following these reactions, further heterocyclization processes were conducted, incorporating the quinoline moiety into the final structures. These findings underscore the potential of pyrazolo [1,5-a]quinoline derivatives as promising candidates for drug discovery, offering new avenues for targeting diseases with related molecular mechanisms.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;The key starting compound in this study was 3,5-dimethyl-4-(2-phenylhydrazono)-4H-pyrazole, which has been utilized in numerous heterocyclization reactions. These reactions, involving various reagents, such as cyclic ketones and diketones in the presence of aromatic aldehydes, led to the formation of fused tetracyclic compounds. Arylhydrazonopyrazole derivatives (5a-c) were employed in multicomponent reactions to synthesize 3,5,6,7,8,9-hexahydropyrazolo[1,5-a]quinoline and pyran derivatives. The reactions were carried out using both conventional catalysts and ionic liquid-immobilized catalysts. Notably, the use of ionic liquid-immobilized catalysts resulted in higher yields of the desired compounds.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;In this study, new compounds were synthesized, characterized, and evaluated for their cytotoxicity against six cancer cell lines: A549, HT-29, MKN-45, U87MG, SMMC-7721, and H460. Additionally, the cytotoxic effects of the synthesized compounds were assessed against hepatocellular carcinoma (HepG2) and cervical carcinoma (HeLa) cell lines. Morphological studies of selected compounds were also conducted to further understand their effects on the cancer cells. Moreover, the cytotoxicity of the selected compounds was tested against seventeen different cancer cell lines, categorized by disease type. Morphological analyses of these selected compounds were also performed to gain deeper insights into their potential as anticancer agents.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/stron","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143540263","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Regulatory Effect of Non-Coding RNAs on Programmed Cell Death in Melanoma: Novel Therapeutic Crosstalk for Targeted Therapy.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-02-27 DOI: 10.2174/0115680096329727250129173510
Andarz Fazlollahpour-Naghibi, Seyed Reza Taha, Mahdieh Shariatzadeh, Danyal Daneshdoust, Kimia Bagheri, Tahereh Farkhondeh, Mohammad Samini, Mohammad Hossein Pourhanifeh, Saeed Samarghandian
{"title":"Regulatory Effect of Non-Coding RNAs on Programmed Cell Death in Melanoma: Novel Therapeutic Crosstalk for Targeted Therapy.","authors":"Andarz Fazlollahpour-Naghibi, Seyed Reza Taha, Mahdieh Shariatzadeh, Danyal Daneshdoust, Kimia Bagheri, Tahereh Farkhondeh, Mohammad Samini, Mohammad Hossein Pourhanifeh, Saeed Samarghandian","doi":"10.2174/0115680096329727250129173510","DOIUrl":"https://doi.org/10.2174/0115680096329727250129173510","url":null,"abstract":"<p><p>Cutaneous melanoma, one of the most fatal and aggressive types of skin cancer, is a malignant tumor created from pigment-producing cells called melanocytes. The mor-bidity and incidence rate of melanoma are increasing around the world. Considering all developments in diagnostic and therapeutic approaches, early diagnosis and more effective targeted therapies are still urgent for melanoma. Non-coding RNAs (ncRNAs), including microRNAs and long non-coding RNAs, are key role players in fundamental cellular pro-cesses and have various biological properties. It has been demonstrated that the dysregula-tion of these biomarkers contributes to melanoma pathogenesis. Furthermore, programmed cell death (PCD), as an important cellular event, has been shown to play a role in the initi-ation and progression of cutaneous melanoma. Recent investigations have reported that the interplay between ncRNAs and PCD can be considered a novel choice for targeted therapy in melanoma. Thus, this review discusses PCD-related ncRNAs in melanoma, focusing on underlying pathways.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143522875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ADH1B Gene Promotes Gastric Cancer Tumorigenesis and its Cisplatin Resistance through the EMT Pathway.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-02-26 DOI: 10.2174/0115680096351094241125111654
Zhenguo Pan, Chengcheng Gao
{"title":"ADH1B Gene Promotes Gastric Cancer Tumorigenesis and its Cisplatin Resistance through the EMT Pathway.","authors":"Zhenguo Pan, Chengcheng Gao","doi":"10.2174/0115680096351094241125111654","DOIUrl":"https://doi.org/10.2174/0115680096351094241125111654","url":null,"abstract":"<p><strong>Background: </strong>Cisplatin resistance significantly affects the outcome of gastric cancer treatment. In this study, genes associated with cisplatin resistance were investigated and discussed.</p><p><strong>Methods: </strong>We analyzed the sequencing data of GSE14208 patients from the GEO database using differential and enrichment analyses. Gastric cancer cells with high ADH1B expression and low ADH1B expression were selected by qPCR and WB to construct ADH1B overexpress and silence cells. The optimal cisplatin concentrations for treatment were determined via CCK-8 detection and WB. Furthermore, we assessed drug resistance, cellular activity, and invasion and migration capacities using IC50, CCK-8 assays, Transwell, and migration tests. Apoptosis was evaluated using flow cytometry and EDU staining. The pathway influenced by ADH1B was examined through WB and immunofluorescence. The impact of gene expression on the tumorigenic potential of gastric cancer cells was assessed by analyzing tumor size, mass, and histology in HE-stained tumor sections and Ki67 and protein pathway immuno-histochemistry in a mouse tumorigenesis model.</p><p><strong>Results: </strong>This study revealed that ADH1B may exhibit a cancer-promoting effect, according to our database analysis, and is associated with drug resistance. Silencing ADH1B in AGS cells led to a reduced IC50, as well as decreased viability, invasion, and migration capabilities while increasing apoptosis rates. Conversely, overexpressing ADH1B in MKN-45 cells reversed these effects. Western blot and immunofluorescence results indicated that the expression levels of proteins involved in the EMT, PI3K, and MAPK pathways were altered following the silencing and overexpression of ADH1B. Moreover, silencing ADH1B not only reduced tumor volume and weight but also enhanced the tumor-reducing effects of cisplatin, as evidenced by changes in tumor structure; overexpression had the opposite effects. The alterations in pathway protein expression in tumor sections mirrored those observed in cells.</p><p><strong>Conclusion: </strong>This study identified the gene ADH1B as a critical factor in the incidence and drug resistance of gastric cancer. It was demonstrated through cellular and tumorigenic assays that ADH1B promotes carcinogenesis and enhances cisplatin resistance in gastric cancer cells via the EMT signaling pathways.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143514887","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Spinal KCC2 Mediates the Modulation Effect of HDAC2 on Bone Cancer Pain in Rats.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-02-24 DOI: 10.2174/0115680096356509250117092430
Tongxuan Wang, Yalin Li, Xinran Hou, Qulian Guo, Yingqi Weng
{"title":"Spinal KCC2 Mediates the Modulation Effect of HDAC2 on Bone Cancer Pain in Rats.","authors":"Tongxuan Wang, Yalin Li, Xinran Hou, Qulian Guo, Yingqi Weng","doi":"10.2174/0115680096356509250117092430","DOIUrl":"https://doi.org/10.2174/0115680096356509250117092430","url":null,"abstract":"<p><strong>Background: </strong>Bone cancer pain is a global medical concern with limited treatment options that significantly reduce the quality of life for cancer patients. Therefore, identifying a promising therapeutic target for bone cancer pain is urgently needed.</p><p><strong>Objective: </strong>Our previous research indicated that KCC2 may be associated with the modulation of HDAC2 in a rat model of bone cancer pain. The current study aimed to investigate whether KCC2 in the lumbar spinal cord is a key downstream molecule in the modulation of HDAC2 related to bone cancer pain.</p><p><strong>Methods: </strong>In this study, we assessed the expression levels of KCC2 and HDAC2 in the lumbar spinal cord of rats with bone cancer pain using Western blotting and RT-PCR. Mechanical hyperalgesia was evaluated using Von Frey hairs, and immunofluorescence was employed to localize KCC2 in central nervous system cells.</p><p><strong>Results: </strong>The expression of KCC2 was down-regulated in a time-dependent manner in the lumbar spinal cord of rats with bone cancer pain. Furthermore, the use of an RNA-interfering lentivirus targeting HDAC2 restored KCC2 expression and alleviated mechanical hyperalgesia in these rats. Notably, the analgesic effect of the HDAC2-targeting lentivirus was completely reversed by the KCC2 inhibitor VU0240551.</p><p><strong>Conclusion: </strong>KCC2 in the lumbar spinal cord mediated the modulation of HDAC2 in rat models of bone cancer pain, suggesting that KCC2 could be a promising therapeutic target for treating bone cancer pain.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-02-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143491170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advanced Engineered Nanoplatforms to Overcome Biological Barriers for Targeting Brain Tumors.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-02-20 DOI: 10.2174/0115680096355101250120114819
Shaheen Sultana, Jyoti Gupta, Vikram Sharma, Komal Gupta, Gayatri Khosla, Darshna Mishra
{"title":"Advanced Engineered Nanoplatforms to Overcome Biological Barriers for Targeting Brain Tumors.","authors":"Shaheen Sultana, Jyoti Gupta, Vikram Sharma, Komal Gupta, Gayatri Khosla, Darshna Mishra","doi":"10.2174/0115680096355101250120114819","DOIUrl":"https://doi.org/10.2174/0115680096355101250120114819","url":null,"abstract":"<p><p>Effective drug delivery to the brain is critically hindered by the blood-brain barrier (BBB), a selective barrier that complicates treatment for central nervous system (CNS) disorders, including brain tumors. Recent innovations in pharmaceutical sciences have introduced new strategies to surmount this challenge and enhance therapeutic efficacy. This review aims to assess recent advancements in engineered nanoplatforms designed to overcome the BBB, with a focus on their application in brain tumor targeting. It seeks to evaluate different drug delivery strategies and formulations that enhance brain penetration, improve targeting precision, and minimize systemic side effects. A comprehensive review of the literature and recent studies on brain-targeting strategies was conducted. The review examined the strategies to prolong blood circulation time and analyzed particularly the PEGylation approach, lipid-based nanocarrier, albumin binding strategies and red blood cell-based delivery. It also explored various strategies (e.g., peptides, prodrug, antibodies, nanotechnology, ligand-based delivery) and subcellular targeting techniques aimed at enhancing brain drug delivery and cellular uptake. PEGylation was found to significantly improve the ability of nano carriers to penetrate brain tumors by reducing macrophage-mediated clearance. Nanotechnology-based strategies coupled with ligand-based approaches effectively enhance brain delivery. Subcellular targeting strategies facilitated endolysosomal escape, leading to better therapeutic agent retention within brain tumor cells. Advances in nanotechnology and targeting strategies offer promising solutions for overcoming the BBB and improving brain tumor treatment. These novel strategies significantly enhance brain targeting while minimizing systemic effects. Continued research is essential to optimize these methods and achieve more effective therapeutic outcomes.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143467140","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Role of Nrf2 in Glioma: Therapeutic Targeting Strategies.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-02-14 DOI: 10.2174/0115680096331620250119111525
Mohammad Sadra Harifi-Mood, Effat Alemzadeh, Danyal Barati, Amir Hossein Dehghani, Fatemeh Zahra Siroosi, Michael Aschner, Fariborz Samini, Saeed Samarghandian, Tahereh Farkhondeh
{"title":"The Role of Nrf2 in Glioma: Therapeutic Targeting Strategies.","authors":"Mohammad Sadra Harifi-Mood, Effat Alemzadeh, Danyal Barati, Amir Hossein Dehghani, Fatemeh Zahra Siroosi, Michael Aschner, Fariborz Samini, Saeed Samarghandian, Tahereh Farkhondeh","doi":"10.2174/0115680096331620250119111525","DOIUrl":"https://doi.org/10.2174/0115680096331620250119111525","url":null,"abstract":"<p><p>Cancer is one of the most challenging diseases to cure due to its complexity. Gli-oma, as a neuroepithelial cancer of the glial cells, is one of the rarest malignancies which has a low survival rate. The exact risk factors of glioma are still not clear, but allergy, ionizing radiation, and hereditary factors are reported to be associated with glioma. Nrf2 as an antiox-idant regulator has been reported to be highly expressed in malignances tissues like glioma. Nrf2 regulates the expression of various antioxidant and cytoprotective genes. In gliomas, Nrf2 activation helps tumor cells combat oxidative stress by enhancing the production of de-toxifying enzymes (e.g., glutathione peroxidase, NADPH quinone oxidoreductase). This al-lows glioma cells to survive and proliferate in toxic tumor microenvironments rich in reactive oxygen species (ROS). Although the role of Nrf2 in the apoptosis of cancerous glial cells is not clear yet, it has been shown that Nrf2 inhibition via different methods can increase the efficiency of the chemo-therapy agents to treat glioma. Elevated Nrf2 activity has been linked to drug resistance in gliomas. The activation of Nrf2 increases the expression of multidrug resistance-associated proteins (MRPs) and other detoxifying enzymes, which limit the effectiveness of chemother-apeutic agents like temozolomide (TMZ). Nrf2 inhibitors can suppress the signaling pathway of Nrf2 and decrease the expression of detoxifying enzymes like SOD, CAT, GPX, and GCL, which can increase the efficiency of chemotherapy agents. Using drugs that inhibit the Nrf2 expression in combination with classical chemotherapy agents can be a promising procedure to decrease chemoresistance and be effective in increasing the survival rate of patients with glioma. In this study, we focused on the association of glioma and Nrf2 expression and its targeting as a new therapeutic approach in glioma treatment.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143432621","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Groundbreaking mRNA Lung Cancer Vaccine Trials: A New Dawn in Cancer Treatment.
IF 2.3 4区 医学
Current cancer drug targets Pub Date : 2025-02-14 DOI: 10.2174/0115680096360059250131075456
Md Sadique Hussain, Ayesha Sultana, Ajay Singh Bisht, Gaurav Gupta
{"title":"Groundbreaking mRNA Lung Cancer Vaccine Trials: A New Dawn in Cancer Treatment.","authors":"Md Sadique Hussain, Ayesha Sultana, Ajay Singh Bisht, Gaurav Gupta","doi":"10.2174/0115680096360059250131075456","DOIUrl":"https://doi.org/10.2174/0115680096360059250131075456","url":null,"abstract":"<p><p>The advent of mRNA vaccines has heralded a transformative era in oncology, exemplified by the BNT116 mRNA lung cancer vaccine. Leveraging the same ground-breaking technology as COVID-19 vaccines, BNT116 delivers tumor-specific genetic in-structions to the immune system, targeting non-small cell lung cancer (NSCLC), the most prevalent lung cancer subtype. This approach contrasts with conventional therapies that lack precision and often damage healthy tissues. By encoding tumor antigens, BNT116 educates cytotoxic T cells to recognize and eradicate malignant cells, aligning with the principles of precision medicine. Early-phase clinical trials (e.g., NCT05142189) have demonstrated a favorable safety profile and promising antitumor activity, with ongoing re-search exploring its use in combination therapies, such as checkpoint inhibitors. Despite logistical challenges, such as mRNA instability and cold chain requirements, advances in lipid nanoparticle delivery systems are enhancing vaccine stability and efficacy. The adaptability of mRNA technology positions it as a cornerstone for personalized oncology, with potential applications extending to other cancers. Success in the BNT116 trials could redefine NSCLC treatment paradigms, offering a targeted, less cytotoxic alternative. This innovation can not only improve therapeutic outcomes, but also pave the way for preven-tive cancer vaccines, signaling a new dawn in cancer treatment.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143432617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信