{"title":"Targeting LAG-3 restores CD8+ T cell effector function through CD94/NKG2-Qa-1b signaling","authors":"Zhiqiang Wang, Mingzhu Yin, Ge Lou","doi":"10.1002/mog2.70003","DOIUrl":"https://doi.org/10.1002/mog2.70003","url":null,"abstract":"<p>A recent study published in <i>Cell</i> by Ngiow et al. elucidates the synergistic roles of programmed cell death protein 1 (PD-1) and lymphocyte-activation gene 3 (LAG-3) in CD8<sup>+</sup> T cells.<span><sup>1</sup></span> The authors demonstrate that LAG-3 influences the fate of these cells by sustaining thymocyte selection-associated high mobility group box protein (TOX) expression and modulating the CD94/NKG2-Qa-1b axis in exhausted CD8<sup>+</sup> T cells. This research enhances our understanding of the intricate mechanisms underlying immune checkpoint blockade and paves the way for novel approaches in cancer immunotherapy.</p><p>In chronic viral infections and cancers, CD8<sup>+</sup> T cells, known as the “warriors” of the immune system, frequently experience exhaustion due to persistent antigenic stimulation. This exhaustion results in impaired proliferation and effector function of exhausted CD8<sup>+</sup> T cells (Tex), ultimately leading to immune failure. The defining characteristics of Tex include a distinct TOX-driven transcriptional and epigenetic state, along with the sustained expression of various inhibitory receptors (IRs), such as PD-1, LAG-3, and cytotoxic T-lymphocyte antigen-4 (CTLA-4).<span><sup>2</sup></span> Recent advancements in immune checkpoint blockade therapies that target various IRs have significantly improved tumor treatment outcomes. However, only a subset of tumor patients exhibit sustained clinical responses to monotherapy with PD-1/programmed death-ligand 1 (PD-L1) blockade. Consequently, there is a growing interest in exploring the combination of multiple immune receptors to enhance the efficacy of immunotherapeutic approaches. A recent phase III clinical trial, RELATIVITY-047, demonstrated that combining a LAG-3 antibody (relatlimab) and a PD-1 antibody (nivolumab) exhibits enhanced clinical antitumor effects. However, the precise mechanisms by which PD-1 and LAG-3 regulate Tex function, as well as the interconnections between their signaling pathways, remain unclear.</p><p>Ngiow et al. investigated the effects of LAG-3 and PD-1 on CD8<sup>+</sup> T cells at various stages of chronic infection using the Quad transplantation model. Their findings revealed that CD8<sup>+</sup> T cells lacking PD-1 proliferated significantly faster during the early stages of infection; however, these cells became progressively less sustainable over time. In contrast, cells lacking LAG-3 demonstrated enhanced effector functions, particularly in terms of cytotoxicity and cytokine secretion. This indicates that PD-1 primarily inhibits cell proliferation, while LAG-3 restricts the multifunctionality and killing capacity of effector T cells as they transition into exhausted T cells. The researchers further investigated the mechanism by which LAG-3 influences the differentiation of Tex cells through the regulation of TOX expression. Their findings revealed that LAG-3 deficiency resulted in a significant reduction in TOX expres","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70003","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142707993","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":"Pharmacokinetics comparison of aumolertinib, osimertinib, gefitinib and their major metabolites in mouse model and NSCLC patients","authors":"Ran Ren, Jiangang Zhang, Yongpeng He, Xianghua Zeng, Yu Zhou, Luyao Shen, Yongsheng Li, Dairong Li, Huakan Zhao","doi":"10.1002/mog2.70002","DOIUrl":"https://doi.org/10.1002/mog2.70002","url":null,"abstract":"<p>The tyrosine kinase inhibitors (TKIs) of epidermal growth factor receptor (EGFR), such as osimertinib and gefitinib, aumolertinib have been widely used in EGFR mutation-positive non-small cell lung cancer (NSCLC) patients. However, the discrepancy of pharmacokinetic features and distributions in important tissues between these EGFR-TKIs remains obscure. In this study, an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method with specificity and accuracy was established. After a single equivalent dose ratio or equal dose gavage, aumolertinib displayed the shortest elimination half-life time (<i>t</i><sub>1/2</sub>), while it showed the largest area under the concentration–time curve in mouse plasma and bone marrow among these 3 EGFR-TKIs. Furthermore, at the time of reaching maximum concentration (<i>t</i><sub>max</sub>) after single equivalent dose ratio gavage, the concentrations of aumolertinib were significantly higher than that of osimertinib and gefitinib in 9 important tissues of mice. Moreover, after single oral administration, aumolertinib displayed the highest concentration in plasma samples from EGFR mutation-positive NSCLC patients. Collectively, our findings manifest that the bioavailability and tissue distribution features of aumolertinib are superior to those of osimertinib and gefitinib, providing a pharmacokinetic basis for the clinical application of aumolertinib and the development of next-generation EGFR-TKIs.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70002","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142708332","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}
Peilan Peng, Siyuan Qin, Lei Li, Zhijun He, Bowen Li, Edouard C. Nice, Li Zhou, Yunlong Lei
{"title":"Epigenetic remodeling under oxidative stress: Mechanisms driving tumor metastasis","authors":"Peilan Peng, Siyuan Qin, Lei Li, Zhijun He, Bowen Li, Edouard C. Nice, Li Zhou, Yunlong Lei","doi":"10.1002/mog2.70000","DOIUrl":"https://doi.org/10.1002/mog2.70000","url":null,"abstract":"<p>Tumor metastasis is a multistep, inefficient process orchestrated by diverse signaling pathways. Compared to primary tumor cells, disseminated tumor cells inevitably encounter higher oxidative stress in foreign environments. The levels of reactive oxygen species (ROS) fluctuate dynamically during different metastatic stages, adding complexity to the regulation of metastatic progression. Numerous studies suggest that epigenetic remodeling, a key reversible mechanism of gene regulation, plays a critical role in responding to oxidative stress and controlling gene expression profiles that drive metastasis. Despite extensive research, a comprehensive understanding of how oxidative stress impacts metastasis through epigenetic modifications remains elusive, such as DNA methylation, histone modification, ncRNAs, and m<sup>6</sup>A modification. Epigenetic therapeutic strategies, such as DNMT inhibitors, HDAC inhibitors (HDACis), and miRNA mimics, have shown promise, yet challenges related to immunogenicity, specificity, and delivery also exist. Furthermore, due to limited understanding, some drugs targeting m<sup>6</sup>A modification have yet to be explored. In this review, we provided an overview of how oxidative stress influences tumor metastatic behavior, summarized the epigenetic mechanisms involved in these processes, and reviewed the latest advancements in epigenetic-targeted therapies, which may pave the way to develop novel strategy for preventing or treating tumor metastasis.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70000","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142642294","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}
Pei Zhang, Chenyan Zhang, Xiaoying Li, Chen Chang, Cailing Gan, Tinghong Ye, Dan Cao
{"title":"Immunotherapy for gastric cancer: Advances and challenges","authors":"Pei Zhang, Chenyan Zhang, Xiaoying Li, Chen Chang, Cailing Gan, Tinghong Ye, Dan Cao","doi":"10.1002/mog2.92","DOIUrl":"https://doi.org/10.1002/mog2.92","url":null,"abstract":"<p>Gastric cancer (GC) ranks among the leading causes of cancer-related mortality globally. Often, its initial stages manifest subtly, and the infrequency of routine screenings contributes to late diagnoses in many cases. Systemic treatments for GC include chemotherapy, targeted therapy, and immunotherapy, among which immunotherapy is the first-line standard treatment for advanced GC. In recent years, immunotherapy has seen notable advancements, as evidenced by the Food and Drug Administration's approval of drugs such as nivolumab and pembrolizumab for GC treatment. Additionally, several other drugs are currently under rigorous preclinical and clinical investigation. This review aims to shed light on the recent advancements in immunotherapy for GC, particularly emphasizing the insights gained from phase 2/3 clinical trials that assess the efficacy, safety, and promise of various immunotherapeutic modalities, including immune checkpoint inhibitors, CAR-T-cell therapies, and cancer vaccines, in enhancing patient outcomes. Moreover, this review delves into the intricate immunological framework of GC, focusing on the tumor microenvironment, interactions among immune cells, and the roles of immune checkpoints such as PD-L1. We also address the hurdles and prospective paths forward in the realm of immunotherapy for GC, offering fresh viewpoints on potential therapeutic approaches in this evolving domain.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.92","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142587953","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":"Nanoparticle-encapsuled celastrol-Fe(III): “Single-edged sword” for tumor therapy featuring microenvironment-dependent toxicity","authors":"Yu Liu, Zhiyong Qian","doi":"10.1002/mog2.70001","DOIUrl":"https://doi.org/10.1002/mog2.70001","url":null,"abstract":"<p>Recently, Li et al. published a study in <i>Science Advances</i> on the use of dual-responsive celastrol (CEL) materials for cancer treatment. The study took advantage of the high adenosine triphosphate (ATP) levels in the tumor microenvironment to modify the interior and surface of CEL, reducing its toxicity to normal tissues and maximizing the efficacy of its cancer treatment which contributed significantly to drug delivery for tumor therapy.<span><sup>1</sup></span></p><p>CEL is a drug with anticancer activity isolated from traditional Chinese medicinal herbs.<span><sup>2</sup></span> It inhibits cancer cell growth and survival signaling by binding to Cdc37 and Hsp90 reaction sites, causing disruption of the Hsp90-Cdc37 binding and folding of some of the client proteins.<span><sup>3</sup></span> However, its water stability, bioavailability, and targeting are undesirable, which limited its clinical application. There are currently two main approaches to promote CEL's therapeutic potential in clinic: 1. synthesizing derivatives by surface modification which mainly focuses on C-20 carboxylic acid functionality, alteration at the A ring, and C-6-modification at the B ring, and 2. adopting nanomedicine delivery systems, where liposomes, polymeric micelles, and nanoparticles (NPs) are commonly used. The second approach, however, requires strictly selected materials. Liposomes, with its ease of aggregation and instability and leakage when packaging drugs, and polymeric micelles, with potential toxicity, unclear drug delivery mechanisms, and poor quality/specification control, demand extra considerations in application. In this study an ATP/ROS dual-responsive CEL derivative was designed for tumor therapy by mainly taking advantage of the environment in which ATP levels inside and outside tumor cells are 10<sup>3</sup> to 10<sup>4</sup> times higher than that around normal tissues.<span><sup>4</sup></span> Surface modification of CEL using metal chelation was performed to address the toxicity to normal tissues, and NPs were selected for packaging to promote the release of CEL when it reaches the tumor site. Compared with liposomes and polymeric micelles, CEL-Fe with ATP/ROS dual-response has a well-defined structural study, low drug toxicity, high stability, and ultimately the ability to respond to the specificity of the tumor microenvironment for effective drug release and treatment which improves the biosafety and biocompatibility of the therapeutic drugs. Fe(III) was chosen to coordinately bond with oxygens on C-2 and C-3 in CEL to produce the less toxic CEL-Fe, which is then coated with polymer membrane consisting of thioketal-containing polyethylene glycol-grafted polymer and polyethylene enamine-modified F127 polymer to elevate its permeability and retention effects.<span><sup>5</sup></span> The high concentration of ATP in tumor tissues disrupts the coordination of Fe(III), restoring the pharmacological toxicity of CEL. In response to high ","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142541032","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}
Jing Zuo, Han Yan, Siyuan Qin, Yonghao Yan, Shiqi Wang, Li Yang, Jinlin Yang, Jun Yang
{"title":"Extracellular vesicles in cancer drug resistance: Mechanistic insights and therapeutic implications","authors":"Jing Zuo, Han Yan, Siyuan Qin, Yonghao Yan, Shiqi Wang, Li Yang, Jinlin Yang, Jun Yang","doi":"10.1002/mog2.94","DOIUrl":"https://doi.org/10.1002/mog2.94","url":null,"abstract":"<p>Despite significant advancements in the treatment of cancer, therapeutic resistance remains a major hurdle for the achievement of full cures. Besides being typically driven by intratumoral heterogeneity, such acquired resistance also relies heavily on cell-cell communication whereby extracellular vesicles (EVs) carrying resistance-related components can be transferred from drug-resistant cells or nontumorigenic members within tumor microenvironment (TME) to their sensitive neighbors. The cargo and membrane surface proteins of EVs derived from drug-resistant cells and TME cells carry abundant biological information, transferring therapy-resistant capacity to sensitive cancer cells. Hence, a deep understanding of the roles of EVs in cancer therapy resistance will facilitate identifying biomarkers and developing new approaches to restore therapy sensitivity. In this review, we summarize our current understanding regarding the causes and effects of EV-mediated cell–cell communication in drug resistance, with a particular notice on how various kinds of cargoes derived from different cells within TME are linked to the resistant phenotype. We also discuss how this knowledge can contribute to improvements in clinical practice, that is, the opportunities and challenges of EVs in functioning as potential biomarkers in predicting therapeutic resistance and, by extension, EV-based therapy in achieving deeper and longer-lasting clinical responses.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.94","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142525118","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":"miRNA interplay: Mechanisms and therapeutic interventions in cancer","authors":"Zehua Wang, Hangxuan Wang, Shuhan Zhou, Jiasheng Mao, Zhiqing Zhan, Shiwei Duan","doi":"10.1002/mog2.93","DOIUrl":"https://doi.org/10.1002/mog2.93","url":null,"abstract":"<p>MicroRNAs (miRNAs) are key molecules that regulate gene expression. miRNAs regulate protein synthesis by binding to mRNA, influencing processes such as cell proliferation, metastasis, and apoptosis. They play a pivotal role in cancer development. Current research mainly explores miRNA mechanisms and applications, and the techniques underpinning this research are foundational to both basic science and clinical translation. However, no review has comprehensively examined miRNA mechanisms and applications from a technical perspective, creating a need for this work. Advances in RNA sequencing technology, CRISPR/Cas9 technology, and bioinformatics tools have deepened our understanding of miRNA interactions. miRNA can serve as a biomarker for cancer diagnosis and prognosis, with significant clinical potential. The development of miRNA mimics and inhibitors has brought new hope for cancer treatment, especially in reversing cancer drug resistance. This article reviews the vital role of miRNA interactions in cancer occurrence, development, diagnosis, and treatment, providing new perspectives and strategies for personalized medicine and cancer therapy.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.93","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142439000","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":"KMT2C/D deficiency promotes breast cancer metastasis by regulating KDM6A-mediated epigenetic remodeling","authors":"Zhao Huang, Wei Zhao","doi":"10.1002/mog2.95","DOIUrl":"https://doi.org/10.1002/mog2.95","url":null,"abstract":"<p>On June 26, 2024, Kornelia Polyak's team published an article in Nature Cell Biology that revealed a new mechanism by which histone-lysine N-methyltransferase 2 C (<i>KMT2C)</i> or <i>KMT2D</i> deletion promotes metalloproteinase 3 (MMP3) expression in a lysine-specific demethylase 6 A (KDM6A)-dependent manner, thereby driving brain metastasis (BMs) of triple-negative breast cancer (TNBC). It provides a new perspective for the treatment of TNBC at the epigenetic level.<span><sup>1</sup></span> Breast cancer is the most common neoplasm and the leading cause of tumor-related death in women worldwide. Breast cancer subtypes can be classified according to the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2.<span><sup>2</sup></span> TNBC is characterized by a high level of cell invasiveness and visceral metastasis to organs, usually brain, lungs, and liver, with an average survival time of 18 months. Although TNBC accounts for only about 15%–20% of all breast cancers, it is the subtype with the worst prognosis of breast cancer. Before the advent of immunotherapy, systemic chemotherapies including taxanes, anthracyclines and/or platinum were the predominant first-line treatment options for TNBC. However, the median overall survival of metastatic TNBC is only 9–12 months, and the 5-year survival rate is approximately 12%, which is a serious unmet medical need.<span><sup>3</sup></span></p><p>The most commonly mutated gene in BMs of breast cancer has been reported to be <i>KMT2C/D</i>, and the results of gene sequencing showed that <i>KMT2C</i> and <i>KMT2D</i> were significantly reduced in distal metastasis compared to the primary tumor, suggesting that functional loss of <i>KMT2C/D</i> plays a role in BMs of breast cancer. <i>KMT2C</i> and <i>KMT2D</i> catalyze the methylation of unmethylated H3K4 sites to form H3K4me1, which can be enriched in active enhancer and promoter regions. This modification recruits other histone modifying enzymes, including histone H3K27 acetyltransferases (such as P300) and demethylases (such as KDM6A), which are essential for the regulation of gene expression. As the core components of the SET1-associated protein epigenetic regulatory complex (COMPASS), <i>KMT2C</i> and <i>KMT2D</i> profoundly regulate the epigenetic landscape.<span><sup>4</sup></span> However, it is still unclear how <i>KMT2C</i>/<i>D</i> mutations affect the epigenetic and transcriptomic landscape to promote tumorigenesis.</p><p>Recently, Marco Seehawer and colleagues from Dana-Farber Cancer Institute in the United States found that the loss of <i>KMT2C</i> or <i>KMT2D</i> can lead to metastasis (especially BMs) in a nonmetastatic TNBC mouse model. Mechanistic studies revealed that the loss of <i>KMT2C</i> or <i>KMT2D</i> promotes the expression of matrix MMP3 in a KDM6A-dependent manner, thereby driving BMs of TNBC.<span><sup>1</sup></span> This study provides not only a new perspective for the tre","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.95","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142438969","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}
Shen Li, Maosen Xu, Yuanling Meng, Haozhen Sun, Tao Zhang, Hanle Yang, Yueyi Li, Xuelei Ma
{"title":"The application of the combination between artificial intelligence and endoscopy in gastrointestinal tumors","authors":"Shen Li, Maosen Xu, Yuanling Meng, Haozhen Sun, Tao Zhang, Hanle Yang, Yueyi Li, Xuelei Ma","doi":"10.1002/mog2.91","DOIUrl":"https://doi.org/10.1002/mog2.91","url":null,"abstract":"<p>Gastrointestinal (GI) tumors have always been a major type of malignant tumor and a leading cause of tumor-related deaths worldwide. The main principles of modern medicine for GI tumors are early prevention, early diagnosis, and early treatment, with early diagnosis being the most effective measure. Endoscopy, due to its ability to visualize lesions, has been one of the primary modalities for screening, diagnosing, and treating GI tumors. However, a qualified endoscopist often requires long training and extensive experience, which to some extent limits the wider use of endoscopy. With advances in data science, artificial intelligence (AI) has brought a new development direction for the endoscopy of GI tumors. AI can quickly process large quantities of data and images and improve diagnostic accuracy with some training, greatly reducing the workload of endoscopists and assisting them in early diagnosis. Therefore, this review focuses on the combined application of endoscopy and AI in GI tumors in recent years, describing the latest research progress on the main types of tumors and their performance in clinical trials, the application of multimodal AI in endoscopy, the development of endoscopy, and the potential applications of AI within it, with the aim of providing a reference for subsequent research.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.91","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142404314","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}
Yuquan Bai, He Xu, Minzhang Guo, Liang Xia, Senyi Deng
{"title":"Extensive infiltration of CD8+ T cells and M1 macrophages is beneficial for multiple cancer patients undergoing chemotherapy","authors":"Yuquan Bai, He Xu, Minzhang Guo, Liang Xia, Senyi Deng","doi":"10.1002/mog2.89","DOIUrl":"https://doi.org/10.1002/mog2.89","url":null,"abstract":"<p>Not all patients can benefit from chemotherapy due to the various of tumor type, stage, location, and the different distribution of immune cells in tumor immune microenvironment (TIME). Immune cells are widely involved in every step of cancer progression, including immune escape, metastasis, drug response, and prognosis. In this study, we explored the transcriptome data of 10 solid tumors treated with chemotherapy to identify the role of immune cells. We downloaded the transcriptome and mutation data of 10 cancers from TCGA databases, and used ESTIMATE and CIBERSORT algorithms to assess the proportion of immune cells in the TIME. According to the proportion of specific immune cell infiltration (SICI) of CD8<sup>+</sup> T cells and M1 macrophages to group the patients, we found that compared with the SICI low and medium groups, the SICI high group had a larger tumor mutation burden, more gene mutations with targeted drugs, more activation of immune checkpoints (PD-1, PD-L1, CTLA-4, LAG-3, TIM-3, and TIGIT), and immune molecules (CD8a, CD80, CD86, TLR2, HLA-A, HLA-B, and CD11a) (<i>p</i> < 0.05). Therefore, we can select an appropriate treatment for patients by clarifying the proportion of immune infiltration of CD8<sup>+</sup> T cells and M1 macrophages in the TIME.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 3","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.89","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142273242","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}