{"title":"Transpiration of neurological phenomena in cancer: Long-term depression (LTD) abrogates cancer stem cell memory and sensitizes it to metabolic therapy","authors":"Gayathri Ravichandran, Aravind Kumar Rengan","doi":"10.1002/mog2.71","DOIUrl":"https://doi.org/10.1002/mog2.71","url":null,"abstract":"<p>Therapy resistance and recurrence are irrefutably ascribed to the unique attributes of the cancer stem cell (CSC) population. Therefore, discerning the underlying mechanism is imperative in arbitrating a strategic therapeutic approach. In this study, we have observed the coexistence of neuron-like cell (NLC) subpopulations in different cancer cell lines. The NLC subset was further validated by transfection studies and was found to be positive for PSD-95. As a proof of concept, we have also demonstrated the transpiration of neurological phenomena such as long-term potentiation (LTP) and long-term depression (LTD) in cancer. We have utilized PMA drug and IL-13 cytokine to study LTD and LTP, respectively, and observed a correlation between the memory marker (GLUN2B) and stemness markers (CD95 and CD58). Moreover, the transcriptional protein pCREB, which plays a crucial role in memory formation, was found to influence CD44 protein levels. These findings suggest that the instigation of LTD can impair cellular memory and sensitize CSC to the metabolic inhibitor, 3Bromopyruvate. Furthermore, this study interfaces the neurobiology of cancer and nanomedicine, wherein we have used biomimetic nanocarriers for the active targeting of drugs and demonstrate their ability to augment therapeutic efficiency.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.71","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140641900","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The biological essence of synthetic lethality: Bringing new opportunities for cancer therapy","authors":"Meiyi Ge, Jian Luo, Yi Wu, Guobo Shen, Xi Kuang","doi":"10.1002/mog2.70","DOIUrl":"https://doi.org/10.1002/mog2.70","url":null,"abstract":"<p>Synthetic lethality (SL), a genetic concept, has revolutionized the development of antitumor therapies by providing avenues to target previously “undruggable” targets with enhanced specificity for tumor cells over normal tissue. The principles of SL have expanded beyond genetic definitions to encompass biological functions, including genome stability, cell cycle regulation, cell death mechanisms, cellular metabolism, cell–cell interactions, and the tumor microenvironment (TME). Tumor cells with inactivated survival pathways are sensitive to therapeutic inhibition of compensatory mechanisms, while normal cells remain unaffected. Exploiting SL based on functional contexts has the potential to significantly improve cancer patient survival by reducing resistance to targeted therapies and enhancing antitumor efficacy when combined with other treatment modalities. This review explores the underlying mechanisms of synthetic lethality interactions (SLI) characterized by biological functions in individual cancer cells and the TME. We also provide a comprehensive summary of strategies for leveraging the dynamic nature of SLI to overcome therapeutic resistance. Additionally, we discuss various approaches and models for screening and designing potent SL agents tailored to the specific needs of cancer patients, as well as strategies for combining SL drugs in tumor treatment. This review offers valuable insights into harnessing SL as a promising avenue for precision cancer therapy.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140321799","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Insight into Janus kinases specificity: From molecular architecture to cancer therapeutics","authors":"Tian-Hua Wei, Meng-Yi Lu, Si-Hui Yao, Yu-Qi Hong, Jin Yang, Meng-Yuan Zhang, Yu-Qi Yin, Yu-Jie Han, Qing-Qing Li, Zi-Xuan Wang, Yi-Bo Wang, Zhen-Jiang Tong, Yun Zhou, Wei-Chen Dai, Yan-Cheng Yu, Shan-Liang Sun, Ye Yang, Nian-Guang Li, Zhi-Hao Shi","doi":"10.1002/mog2.69","DOIUrl":"https://doi.org/10.1002/mog2.69","url":null,"abstract":"<p>Janus Kinases (JAKs) play a crucial role as therapeutic targets for various cancers. However, the current JAK inhibitors (JAKi) available have limited therapeutic benefits due to their lack of selectivity. This review focuses on the structural analysis to elucidate the molecular determinants of JAKs specificity and the discovery and design of selective JAKi. It includes descriptions and comparison of different JAK structures and their binding sites, a comparative analysis of JAKi and their binding modes, detailed interaction fingerprints (IFPs), and an extensive structure-selectivity relationship (SSRs). Moreover, the review also explores the challenges and possibilities of using computational structure-based methods for discovering and designing selective JAKi. Other structure-based approaches, such as targeting the pseudokinase domain, as well as covalent and allosteric designs, are also covered. Based on this analysis, key determinants corresponding to JAK specificity and rational medicinal chemistry strategies are proposed to facilitate the development of highly selective JAKi. Overall, we aim to enhance the understanding of JAK specificity and explore strategies that can lead to the discovery of effective and selective JAKi in cancer therapy, thus improving the prognosis for cancer patients.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.69","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140291393","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advances and applications of nanoparticles in cancer therapy","authors":"Xianzhou Huang, Tao He, Xiuqi Liang, Zhongzheng Xiang, Chao Liu, Shiyao Zhou, Rui Luo, Liping Bai, Xiaorong Kou, Xinchao Li, Rui Wu, Xinyu Gou, Xinyue Wu, Dongxue Huang, Wangxian Fu, Yingjie Li, Ruhan Chen, Ningyue Xu, Yixi Wang, Hao Le, Tao Chen, Yangsong Xu, Yuting Tang, Changyang Gong","doi":"10.1002/mog2.67","DOIUrl":"https://doi.org/10.1002/mog2.67","url":null,"abstract":"<p>Rapid growth in nanoparticles (NPs) as delivery systems holds vast promise to promote therapeutic approaches for cancer treatment. Presently, a diverse array of NPs with unique properties have been developed to overcome different challenges and to achieve sophisticated delivery routes for enhancement of a series of therapies. Inspiring advances have been achieved in the field of cancer therapy using NPs. In this review, we aim to summarize the up-to-date progression of NPs for addressing various challenges, and expect to elicit novel and potential opportunities alternatively. We first introduce different sorts of NP technologies, illustrate their mechanisms, and present their applications. Then, the achievements made by NPs to break obstacles in delivering cargoes to specific sites through particular routes are highlighted, including long-circulation, tumor targeting, responsive release, and subcellular localization. We subsequently retrospect recent research of NPs in different cancer treatments from single therapy, like chemotherapy, to combination therapy, like chemoradiotherapy, and to integrative therapy. Finally, the challenges and perspectives of NPs in cancer therapy, and their potential impact on the field of oncology are discussed. We believe this review can offer a deeper understanding of the challenges and opportunities of NPs for cancer therapy.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-03-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.67","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140145801","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Targets of tumor microenvironment for potential drug development","authors":"Ling Zhang, Ziruoyu Wang, Kailu Liu, Yihang Liu, Shuai Wang, Wei Jiang, Fanghui Lu, Yongjun Dang","doi":"10.1002/mog2.68","DOIUrl":"https://doi.org/10.1002/mog2.68","url":null,"abstract":"<p>The tumor microenvironment (TME) is the ecosystem surrounding a tumor, which usually consists of nontumoral cells or components, and molecules they produce and release. The frequent and continuous interplay between tumor cells and the TME strongly affects tumor development, disease progression, metastasis, and responses to therapeutic interventions. As a hub of potential therapeutic targets, the TME has gained appreciable momentum in cancer research. Here we systematically review the progress in targeting the TME as a strategy to develop novel antitumor drugs from the immunological, stromal and extracellular matrix components of the TME, shedding light on its complex synergies with tumor cells. This exploration highlights the transformative potential these elements hold in refining cancer treatment approaches. This thorough examination not only accentuates the TME's multifaceted nature but also positions it as a formidable avenue for propelling forward the paradigms of cancer therapy. This review aims to foster a deeper understanding of the TME's role in oncogenesis and its potential exploitation in advancing targeted, efficacious cancer treatments, marking a significant stride in the realm of cancer research.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.68","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140015095","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The hidden architects of glioblastoma multiforme: Glioma stem cells","authors":"Om S. Sahoo, Rhiti Mitra, Naveen K. H. Nagaiah","doi":"10.1002/mog2.66","DOIUrl":"https://doi.org/10.1002/mog2.66","url":null,"abstract":"<p>Glioblastoma multiforme (GBM), a WHO grade IV diffuse glioma, is a highly aggressive brain tumor with a median survival of less than a year. Characterized by robust proliferation and invasion, recent studies spotlight glioma stem cells (GSCs) within GBM tumors, pivotal in tumor development, progression, and treatment resistance. This review aims to shed light on the critical role of GSCs in the initiation and progression of GBM, emphasizing their contribution to tumor development and resistance to existing treatments. Unlike normal stem cells, GSCs play a pivotal role in GBM pathogenesis. The review delves into the unique characteristics of GSCs, marked by heightened metabolic activities and distinct epigenetic and transcriptional programming. Recognizing the significance of GSCs in recent years, the review examines how their presence amplifies the lethal nature of GBM. The review also critically evaluates recent advancements in glioma and GBM diagnostic methods and treatment therapies, which also include targeting GSCs. Providing a concise yet comprehensive overview, the review contributes insights into GBM's intricate dynamics, offering potential directions for future research and therapeutic strategies.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.66","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139676577","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Targeting iron–sulfur cluster assembly scaffold protein as a vulnerability in KRAS-activated pancreatic ductal adenocarcinoma","authors":"Huarui Cai, Hongjuan Cui, Erhu Zhao","doi":"10.1002/mog2.65","DOIUrl":"https://doi.org/10.1002/mog2.65","url":null,"abstract":"<p>In two recent publications in Science<span><sup>1</sup></span> and Cell Discovery,<span><sup>2</sup></span> researchers have discovered that iron–sulfur cluster assembly scaffold protein (ISCU) plays crucial roles on maintaining glutathione (GSH) homeostasis, α-KG catabolism, DNA methylation and so on. These groundbreaking findings not only establish crucial connections among iron–sulfur (Fe–S) metabolism, GSH homeostasis, alpha-ketoglutarate (α-KG) catabolism, DNA methylation, and tumor growth but also emphasize the significant and integrated regulation of mitochondrial function and gene expression by ISCU (Figure 1).</p><p>Liu et al.<span><sup>1</sup></span> unveiled the vital role of ISCU in maintaining optimal mitochondrial functions by restoring the balance between iron and GSH. In addition, a previous study<span><sup>3</sup></span> has provided evidence that glutamine, a precursor of GSH, promotes tumor growth through a KRAS-regulated metabolic pathway in pancreatic ductal adenocarcinoma (PDAC), thereby suggesting that ISCU potentially influences tumor development by modulating GSH homeostasis. Ren et al.<span><sup>2</sup></span> uncovered a previously undisclosed association between Fe–S metabolism and tumor growth in kirsten rat sarcoma viral oncogene homolog (KRAS)-activated PDAC. Such findings offer a promising avenue for the treatment of KRAS-activated PDAC, as well as for interventions targeting ISCU-mediated cellular processes.</p><p>PDAC is a highly lethal disease with a mounting incidence and unfavorable prognostic outcomes, underscoring the urgent requirement for the development of efficacious therapeutic approaches. Thus, accurate biomarkers to help stratify risk would greatly improve current diagnostic and decision-making dilemmas. Fe–S clusters are ancient, ubiquitous metal cofactors that possess various physiological functions in antioxidant, iron regulation, the tricarboxylic acid (TCA) cycle, and many other metabolic reactions. Accumulated evidence suggests that abnormal Fe–S clusters assembly pathways lead to mitochondrial dysfunction and cause various diseases, particularly with an impact on tumor development.<span><sup>4</sup></span> For instance, nitrogen fixation gene 1 (NFS1) cysteine desulfurase, as an indispensable protein in Fe–S cluster biogenesis, plays a vital role in mitochondrial metabolic reprogramming. In human colorectal cancer, NFS1 can suppress PANoptosis under oxaliplatin chemotherapy, and NFS1 high expression was linked to unfavorable survival outcomes and hyposensitivity to chemotherapy in patients.<span><sup>5</sup></span> In addition, succinate dehydrogenase complex iron–sulfur subunit B (SDHB) is also an Fe–S cluster protein consisting of three Fe–S clusters. In SDHB-deficient cancer cells, succinate levels are elevated, resulting in hypermethylation of histones and DNA, and glutamine becomes the primary source of TCA cycle metabolites via reductive carboxylation.<span><sup>6</sup></span></p><p>No","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.65","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139655410","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Lysosome-targeting chimera (LYTAC): A silver bullet for targeted degradation of oncogenic membrane proteins","authors":"Qingquan Zheng, Jiawei Guo, Rui Ma, Wenchen Pu","doi":"10.1002/mog2.64","DOIUrl":"https://doi.org/10.1002/mog2.64","url":null,"abstract":"<p>Recently, the group of Prof. Carolyn Bertozzi, a laureate of the Nobel Prize in chemistry 2022, reported the detailed mechanism of lysosome-targeting chimera (LYTAC) in the journal of <i>Science</i>,<span><sup>1</sup></span> after the publication of their first LYTAC molecule in <i>Nature</i> in 2020.<span><sup>2</sup></span> The establishment of LYTAC, a subtype of targeted protein degradation technology, expands the scope of protein degradation to extracellular and membrane-associated targets, and Bertozzi group's new discovery is expected to accelerate the development of LYTAC in cancer therapy.</p><p>Cell membranes play a critical role in various cellular processes, including signaling transduction, cell adhesion, transport of biomolecules and immunity. Proteins embedded in or associated with the cell membrane are key executants of the function of cell membrane, and their dysregulation contributes to tumorigenesis and development of human cancers.<span><sup>3</sup></span> For example, epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase for epithelial growth factor (EGF) and transforming growth factor α (TGF-α), belonging to the ErbB receptor family. Activation of EGFR signaling promotes cell proliferation, survival, angiogenesis and metastasis of diverse malignancies.<span><sup>3</sup></span> Moreover, hepatocyte growth factor receptor (c-Met, HGFR) is another oncogenic receptor tyrosine kinase in diverse cancers. Upon the binding to hepatocyte growth factor (HGF), c-Met is activated via autophosphorylation, leading to the initiation of oncogenic downstream signaling cascades, such as PI3K/AKT and RAS/ERK pathways.<span><sup>3</sup></span> Given their central role in cancer-promoting processes, EGFR and c-Met has become attractive targets for cancer therapies. Small-molecule tyrosine kinase inhibitors (EGFR: gefitinib, afatinib, osimertinib, etc.; c-Met: capmatinib, tepotinib, savolitinib, etc.) and monoclonal antibodies (EGFR: cetuximab, panitumumab; EGFR/c-Met: amivantamab; c-Met: emibetuzumab), have been developed and approved for the treatment of various cancers, including lung and colorectal cancers (Figure 1A). But severe acquired resistance (e.g., via EGFR mutations) and limited therapeutic efficacy (slightly prolonged overall survival) of these treatments restrict their clinical benefit for patients. Moreover, nonenzymatic function of membrane proteins, such as protein–protein interactions, could not be interfered with by kinase inhibitors or monoclonal antibodies, calling for new strategies to control these oncogenic membrane proteins.</p><p>Targeted protein degradation (TPD) is a therapeutic approach that aims to selectively remove disease-causing or undesirable proteins from cells by inducing their degradation, with multiple therapies entering clinical trials and targeting proteins that are previously considered “undruggable.”<span><sup>4</sup></span> There are two main protein degradation mechanisms within cel","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.64","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139468301","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Epigenetic regulation in cancer therapy: From mechanisms to clinical advances","authors":"Lei Tao, Yue Zhou, Yuan Luo, Jiahao Qiu, Yuzhou Xiao, Jiao Zou, Yu Zhang, Xingchen Liu, Xinyu Yang, Kun Gou, Jing Xu, Xinqi Guan, Xiaobo Cen, Yinglan Zhao","doi":"10.1002/mog2.59","DOIUrl":"https://doi.org/10.1002/mog2.59","url":null,"abstract":"<p>Epigenetic regulation refers to the alteration of gene expression independent of changes in DNA sequence. It involves chemical modifications such as DNA methylation, histone methylation, and histone acetylation, which are regulated by a coordinated interplay of various regulators to ensure precise spatial and temporal regulation of gene expression. Epigenetic aberrations are commonly observed in cancer and are considered as hallmarks of cancer. In recent years, small molecules targeting specific epigenetic regulators have been developed and are demonstrating promising therapeutic potential in preclinical and clinical trials for cancer treatment. In this review, we summarize the essential regulatory mechanisms and dysfunctions of epigenetic regulators involved in DNA methylation, histone methylation, and histone acetylation during tumor development and progression. Moreover, we discuss the current advances and challenges in cancer epigenetic therapy that target these mechanisms in both hematologic malignancies and solid tumors. Finally, we discuss the potential of combining epigenetic drugs with other therapies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, as a promising approach for cancer treatment. Overall, we aim to enhance the understanding of epigenetic regulation in cancer therapy and explore targeted therapeutic strategies based on these mechanisms, to ultimately advance cancer therapy and improve patient prognosis.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.59","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139109890","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Joyeeta Talukdar, Tryambak P. Srivastava, Om S. Sahoo, Abhibroto Karmakar, Avdhesh K. Rai, Anupam Sarma, Gayatri Gogoi, Mohammed S. Alqahtani, Mohamed Abbas, Ruby Dhar, Subhradip Karmakar
{"title":"Cancer stem cells: Signaling pathways and therapeutic targeting","authors":"Joyeeta Talukdar, Tryambak P. Srivastava, Om S. Sahoo, Abhibroto Karmakar, Avdhesh K. Rai, Anupam Sarma, Gayatri Gogoi, Mohammed S. Alqahtani, Mohamed Abbas, Ruby Dhar, Subhradip Karmakar","doi":"10.1002/mog2.62","DOIUrl":"https://doi.org/10.1002/mog2.62","url":null,"abstract":"<p>Cancer stem cells (CSCs) constitute a minority cell population characterized by unbounded proliferative potential in both solid and hematological cancers. Despite sharing key stem cell attributes, CSCs possess unique traits, including the initiation and propagation of tumors and resistance to conventional therapies. The purpose of this review is to delve into the origins and fundamental characteristics of CSCs, emphasizing their role in tumor growth and metastasis. The focus extends to unraveling cellular signaling pathways driving oncogenic processes and understanding aberrant cellular crosstalk crucial for targeted cancer therapies. Beginning with an exploration of CSC properties and behavior, we progress to dissecting the cellular signaling network that fuels oncogenic pathways. The discussion spans the inception of CSCs, their survival strategies, and adaptation to new environments. We then transit to recent therapeutic advancements targeting CSCs, culminating in an exploration for precise therapeutic targeting. This review henceforth, underscores the vital significance of comprehending CSCs in cancer progression and treatment resistance. By unraveling the complex signaling pathways and survival mechanisms unique to CSCs, it paves the way for targeted therapeutic strategies that hold immense promise in enhancing cancer treatment efficacy while minimizing collateral damage.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-12-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.62","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139101183","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}