首页 > 最新文献

MedComm – Oncology最新文献

英文 中文
Macrophage-Mediated Myelin Recycling Promotes Malignant Development of Glioblastoma
Pub Date : 2025-02-24 DOI: 10.1002/mog2.70014
Huanhuan Wang, Long Zhang, Feng Xie
<p>A recent study published in the journal <i>Cell</i> [<span>1</span>] revealed the critical role of macrophages in the malignant development of brain cancers, particularly glioblastoma. Through a series of in-vivo and in-vitro experiments, the study researched how macrophages provide essential lipids and nutrients to brain cancer cells by mediating the recycling and reuse of myelin, the insulating layer in the nervous system, which in turn promotes growth and invasion of tumor. They explored the heterogeneity of tumor-associated macrophages (TAM) in glioblastoma tumor microenvironment using single-cell and multi-omics analyses and revealed their specific interactions with different glioblastoma subtypes. The study reported the dynamic contexture of the glioblastoma tumor microenvironment at single-cell levels during primary or recurrent tumor progression, revealed the colocated diversity of niche-specific interactions between TAMs and glioblastoma subtypes at spatial transcriptomic levels, evaluated the chromatin landscape changes and immuno-suppressive features associated with the lipid-laden phenotype using multi-omics sequences, addressed the transfer route of lipid flux from myelin to macrophages at last to mesenchymal-like (MES-like) glioblastoma cells, demonstrated the intrinsic lipid traffic in macrophages and the altered metabolic manner in glioblastoma cells using lipidomics analysis and experiments, presented the protumorigenic functions of lipid-laden macrophages (LLMs) in glioblastoma and their relevance to clinical survival or immunotherapeutic response.</p><p>The study found that in the brain tumor microenvironment, macrophages are able to take up and accumulate myelin debris in large quantities. These myelin fragments are converted by macrophages into cholesterol and other lipids, which are then delivered to brain cancer cells to support their growth and malignant transformation. They also found that specific types of macrophages, such as TAMs with high glycoprotein nonmetastatic melanoma protein B (GPNMB) expression, are closely associated with areas of high myelin debris accumulation and exhibit unique patterns of lipid metabolism and inflammatory activity. The study further demonstrated that macrophage-mediated lipid delivery not only provides an energy source for brain cancer cells, but also promotes the invasion and metastasis ability of cancer cells. By interfering with the lipid metabolism pathway of macrophages, the progress of brain cancer can be significantly inhibited. When macrophages take in myelin fragments, their inflammatory activity is suppressed and they shift to an “anti-inflammatory” state. This anti-inflammatory state may help maintain the stability of the tumor microenvironment, thus providing favorable conditions for tumor cell growth. The study was also verified using patient sample data and found similar patterns of macrophage activity in the tumor microenvironment of glioma patients, which suggests that
{"title":"Macrophage-Mediated Myelin Recycling Promotes Malignant Development of Glioblastoma","authors":"Huanhuan Wang,&nbsp;Long Zhang,&nbsp;Feng Xie","doi":"10.1002/mog2.70014","DOIUrl":"https://doi.org/10.1002/mog2.70014","url":null,"abstract":"&lt;p&gt;A recent study published in the journal &lt;i&gt;Cell&lt;/i&gt; [&lt;span&gt;1&lt;/span&gt;] revealed the critical role of macrophages in the malignant development of brain cancers, particularly glioblastoma. Through a series of in-vivo and in-vitro experiments, the study researched how macrophages provide essential lipids and nutrients to brain cancer cells by mediating the recycling and reuse of myelin, the insulating layer in the nervous system, which in turn promotes growth and invasion of tumor. They explored the heterogeneity of tumor-associated macrophages (TAM) in glioblastoma tumor microenvironment using single-cell and multi-omics analyses and revealed their specific interactions with different glioblastoma subtypes. The study reported the dynamic contexture of the glioblastoma tumor microenvironment at single-cell levels during primary or recurrent tumor progression, revealed the colocated diversity of niche-specific interactions between TAMs and glioblastoma subtypes at spatial transcriptomic levels, evaluated the chromatin landscape changes and immuno-suppressive features associated with the lipid-laden phenotype using multi-omics sequences, addressed the transfer route of lipid flux from myelin to macrophages at last to mesenchymal-like (MES-like) glioblastoma cells, demonstrated the intrinsic lipid traffic in macrophages and the altered metabolic manner in glioblastoma cells using lipidomics analysis and experiments, presented the protumorigenic functions of lipid-laden macrophages (LLMs) in glioblastoma and their relevance to clinical survival or immunotherapeutic response.&lt;/p&gt;&lt;p&gt;The study found that in the brain tumor microenvironment, macrophages are able to take up and accumulate myelin debris in large quantities. These myelin fragments are converted by macrophages into cholesterol and other lipids, which are then delivered to brain cancer cells to support their growth and malignant transformation. They also found that specific types of macrophages, such as TAMs with high glycoprotein nonmetastatic melanoma protein B (GPNMB) expression, are closely associated with areas of high myelin debris accumulation and exhibit unique patterns of lipid metabolism and inflammatory activity. The study further demonstrated that macrophage-mediated lipid delivery not only provides an energy source for brain cancer cells, but also promotes the invasion and metastasis ability of cancer cells. By interfering with the lipid metabolism pathway of macrophages, the progress of brain cancer can be significantly inhibited. When macrophages take in myelin fragments, their inflammatory activity is suppressed and they shift to an “anti-inflammatory” state. This anti-inflammatory state may help maintain the stability of the tumor microenvironment, thus providing favorable conditions for tumor cell growth. The study was also verified using patient sample data and found similar patterns of macrophage activity in the tumor microenvironment of glioma patients, which suggests that ","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70014","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143481329","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}
引用次数: 0
Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention
Pub Date : 2025-02-17 DOI: 10.1002/mog2.70012
Lin Tang, Shao-Cong Peng, Xiao-Wan Zhuang, Yan He, Yu-Xiang Song, Hao Nie, Can-Can Zheng, Zhen-Yu Pan, Alfred King-Yin Lam, Ming-Liang He, Xing-Yuan Shi, Bin Li, Wen Wen Xu

Metastasis remains a leading cause of cancer-related deaths, defined by a complex, multi-step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post-translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.

{"title":"Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention","authors":"Lin Tang,&nbsp;Shao-Cong Peng,&nbsp;Xiao-Wan Zhuang,&nbsp;Yan He,&nbsp;Yu-Xiang Song,&nbsp;Hao Nie,&nbsp;Can-Can Zheng,&nbsp;Zhen-Yu Pan,&nbsp;Alfred King-Yin Lam,&nbsp;Ming-Liang He,&nbsp;Xing-Yuan Shi,&nbsp;Bin Li,&nbsp;Wen Wen Xu","doi":"10.1002/mog2.70012","DOIUrl":"https://doi.org/10.1002/mog2.70012","url":null,"abstract":"<p>Metastasis remains a leading cause of cancer-related deaths, defined by a complex, multi-step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post-translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70012","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143423918","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}
引用次数: 0
Lomitapide: Targeting METTL3 to Overcome Osimertinib Resistance in NSCLC Through Autophagy Activation
Pub Date : 2025-01-28 DOI: 10.1002/mog2.70011
Xiaohui Du, Congcong Zhang, Ying Li, Peipei He, Jian Wang, Xuena Chen, Han Wang, Qi Wang

Osimertinib resistance remains a significant challenge in the treatment of non-small cell lung cancer (NSCLC). N6-methyladenosine (m6A) modifications are closely linked to various mechanisms of anticancer resistance and autophagy, offering new avenues for targeted therapies. However, the role of m6A-mediated autophagy in osimertinib-resistant NSCLC is still unclear. In this study, we utilized multi-omics sequencing analysis and found that overexpression of the m6A methyltransferase METTL3 contributes to osimertinib resistance in NSCLC. Importantly, we identified that METTL3 positively regulates the expression of the autophagy-related gene ubiquinone-cytochrome C reductase complex assembly factor 2 (UQCC2) through an m6A-dependent mechanism. Further, we confirmed that METTL3 knockdown leads to UQCC2 downregulation and triggers autophagy activation. Interestingly, lomitapide, a cholesterol-lowering drug, was repurposed to enhance the sensitivity of cancer cells to therapy by inhibiting METTL3, which in turn activated autophagy-associated cell death pathways, reversing osimertinib resistance. This study emphasizes the critical role of the METTL3/UQCC2 axis in autophagy-mediated drug resistance and positions lomitapide as a promising METTL3 inhibitor and autophagy inducer with potential therapeutic effects, either alone or in combination with other anticancer agents, in patients with osimertinib-resistant NSCLC.

{"title":"Lomitapide: Targeting METTL3 to Overcome Osimertinib Resistance in NSCLC Through Autophagy Activation","authors":"Xiaohui Du,&nbsp;Congcong Zhang,&nbsp;Ying Li,&nbsp;Peipei He,&nbsp;Jian Wang,&nbsp;Xuena Chen,&nbsp;Han Wang,&nbsp;Qi Wang","doi":"10.1002/mog2.70011","DOIUrl":"https://doi.org/10.1002/mog2.70011","url":null,"abstract":"<p>Osimertinib resistance remains a significant challenge in the treatment of non-small cell lung cancer (NSCLC). <i>N</i><sup>6</sup>-methyladenosine (m<sup>6</sup>A) modifications are closely linked to various mechanisms of anticancer resistance and autophagy, offering new avenues for targeted therapies. However, the role of m<sup>6</sup>A-mediated autophagy in osimertinib-resistant NSCLC is still unclear. In this study, we utilized multi-omics sequencing analysis and found that overexpression of the m<sup>6</sup>A methyltransferase METTL3 contributes to osimertinib resistance in NSCLC. Importantly, we identified that METTL3 positively regulates the expression of the autophagy-related gene ubiquinone-cytochrome C reductase complex assembly factor 2 (<i>UQCC2</i>) through an m<sup>6</sup>A-dependent mechanism. Further, we confirmed that <i>METTL3</i> knockdown leads to <i>UQCC2</i> downregulation and triggers autophagy activation. Interestingly, lomitapide, a cholesterol-lowering drug, was repurposed to enhance the sensitivity of cancer cells to therapy by inhibiting METTL3, which in turn activated autophagy-associated cell death pathways, reversing osimertinib resistance. This study emphasizes the critical role of the METTL3/UQCC2 axis in autophagy-mediated drug resistance and positions lomitapide as a promising METTL3 inhibitor and autophagy inducer with potential therapeutic effects, either alone or in combination with other anticancer agents, in patients with osimertinib-resistant NSCLC.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70011","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143120497","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}
引用次数: 0
Ephrin A1 ligand-based CAR-T cells for immunotherapy of EphA2-positive cancer
Pub Date : 2025-01-24 DOI: 10.1002/mog2.70010
Nan Liu, Wenwen Wei, Kexing Ren, Dandan Liang, Dong Yang, Weishan Zhang, Beibei Yang, Bin Sun, Jincheng Zhao, Dan Cao, Liqun Zou, Xudong Zhao

Chimeric antigen receptor (CAR) T cells have demonstrated promising results in hematological malignancies; however, challenges remain in treating solid tumors. New CARs with more effectiveness and lower side effects are needed. Ephrin type-A receptor 2 (EphA2) belongs to the Ephrin family of receptor tyrosine kinases, which is overexpressed in several solid malignancies. Compared with some single-chain variable fragment (ScFv) CARs that exhibit excessively high affinity for their targets, natural receptor/ligand-based CARs maintain inherent affinity for their binding partners, potentially balancing cytotoxicity and side effects to better meet clinical needs. Here, we designed a CAR targeting EphA2-positive cancer cells by exploiting the extracellular domain of its natural ligand Ephrin A1 (EFNA1). EFNA1 CAR-T cells exhibited specific cytotoxicity against various cancer cells and cancer stem-like cells in vitro, and significantly suppressed tumor growth in a pancreatic cancer xenograft mouse model. Moreover, although these CAR-T cells specifically targeted mouse EphA2 and killed mouse tumor cell lines in vitro, they did not induce obvious side effects in mice. Additionally, it also showed good safety in rhesus macaques. Collectively, these results validate the therapeutic effectiveness and safety of EFNA1 CAR-T cells for treating solid tumors.

{"title":"Ephrin A1 ligand-based CAR-T cells for immunotherapy of EphA2-positive cancer","authors":"Nan Liu,&nbsp;Wenwen Wei,&nbsp;Kexing Ren,&nbsp;Dandan Liang,&nbsp;Dong Yang,&nbsp;Weishan Zhang,&nbsp;Beibei Yang,&nbsp;Bin Sun,&nbsp;Jincheng Zhao,&nbsp;Dan Cao,&nbsp;Liqun Zou,&nbsp;Xudong Zhao","doi":"10.1002/mog2.70010","DOIUrl":"https://doi.org/10.1002/mog2.70010","url":null,"abstract":"<p>Chimeric antigen receptor (CAR) T cells have demonstrated promising results in hematological malignancies; however, challenges remain in treating solid tumors. New CARs with more effectiveness and lower side effects are needed. Ephrin type-A receptor 2 (EphA2) belongs to the Ephrin family of receptor tyrosine kinases, which is overexpressed in several solid malignancies. Compared with some single-chain variable fragment (ScFv) CARs that exhibit excessively high affinity for their targets, natural receptor/ligand-based CARs maintain inherent affinity for their binding partners, potentially balancing cytotoxicity and side effects to better meet clinical needs. Here, we designed a CAR targeting EphA2-positive cancer cells by exploiting the extracellular domain of its natural ligand Ephrin A1 (EFNA1). EFNA1 CAR-T cells exhibited specific cytotoxicity against various cancer cells and cancer stem-like cells in vitro, and significantly suppressed tumor growth in a pancreatic cancer xenograft mouse model. Moreover, although these CAR-T cells specifically targeted mouse EphA2 and killed mouse tumor cell lines in vitro, they did not induce obvious side effects in mice. Additionally, it also showed good safety in <i>rhesus macaques</i>. Collectively, these results validate the therapeutic effectiveness and safety of EFNA1 CAR-T cells for treating solid tumors.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70010","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143118772","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}
引用次数: 0
Analysis of reoperational reason of patients with thyroid cancer and strategies for its diagnosis and treatment: A 6-year single-center retrospective study
Pub Date : 2025-01-14 DOI: 10.1002/mog2.70008
Rongli Xie, Yawei Feng, Jiankang Shen, Guohui Xiao, Dan Tan
<p>The incidence of thyroid tumors has been increasing in recent years, and the vast majority of new malignant cases are papillary thyroid micro-carcinoma (PTMC).<span><sup>1</sup></span> For patients with PTMC, comprehensive diagnosis and treatment, led by surgery, is the key to clinical cure. Although the lethality of papillary thyroid carcinoma is very low, tumor proliferation mechanism and surgical method after recurrence are still a hot topic of debate.<span><sup>2-4</sup></span> In this paper, thyroid patients admitted from January 2015 to December 2020 were collected. The inclusion criteria were as follows: (1) age from 18 to 80 years old; (2) the first and subsequent surgeries for thyroid were performed in one single center; (3) the postoperative paraffin pathology confirmed thyroid tumor; and (4) the patient's clinical data were complete. In this study, the gender, age, surgical methods, tumor types, maximum diameter of tumor, metastasis of central neck group and lateral lymph nodes, postoperative complications, and length of hospital stay were collected and recorded. For patients with multiple surgeries, the reason for subsequent surgeries and the interval time between surgeries should be additionally recorded.</p><p>A total of 58 patients undergoing thyroid surgeries (1 patient with laparoscopic surgery was excluded) were collected in this study, as shown in Table 1. Fifteen patients in the observation group underwent central lymph node dissection and 26 patients in the lateral lymph node dissection, while only 1 patient in the control group underwent cervical lateral lymph node dissection, and there was a statistically significant difference in the overall surgical method between the two groups (<i>p</i> < 0.01).</p><p>In the control group, 4 cases of benign tumors and 21 cases of papillary carcinoma were confirmed by pathology after the surgery. However, 55 cases of postoperative pathology confirmed malignant tumors in the observation group, including 52 cases of papillary carcinoma, 2 cases of follicular carcinoma, and 1 case of medullary carcinoma. In patients with confirmed papillary carcinoma, the mean tumor size was 1.33 ± 0.14 cm (<i>p</i> < 0.001). The rate of lymph node metastasis in the central group was 27 out of 118 (<i>p</i> < 0.0001) and the rate of lateral lymph node metastasis was 162 out of 241 (<i>p</i> < 0.01), as shown in Table 1.</p><p>The average length of hospital stay in the observation group was 5.64 ± 0.30 days (<i>p</i> < 0.05). There were five cases of adverse reactions (<i>p</i> > 0.05) after the operation, including four cases of hoarseness, one case of choking cough (with hoarseness), and one case of hemorrhage. The average length of hospitalization in the control group was 4.44 ± 0.38 days, and no patients had obvious adverse reactions (Table S1).</p><p>Out of 58 patients who underwent multiple surgeries, 51 patients underwent two surgeries, and 7 patients underwent three surgeries. Am
{"title":"Analysis of reoperational reason of patients with thyroid cancer and strategies for its diagnosis and treatment: A 6-year single-center retrospective study","authors":"Rongli Xie,&nbsp;Yawei Feng,&nbsp;Jiankang Shen,&nbsp;Guohui Xiao,&nbsp;Dan Tan","doi":"10.1002/mog2.70008","DOIUrl":"https://doi.org/10.1002/mog2.70008","url":null,"abstract":"&lt;p&gt;The incidence of thyroid tumors has been increasing in recent years, and the vast majority of new malignant cases are papillary thyroid micro-carcinoma (PTMC).&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt; For patients with PTMC, comprehensive diagnosis and treatment, led by surgery, is the key to clinical cure. Although the lethality of papillary thyroid carcinoma is very low, tumor proliferation mechanism and surgical method after recurrence are still a hot topic of debate.&lt;span&gt;&lt;sup&gt;2-4&lt;/sup&gt;&lt;/span&gt; In this paper, thyroid patients admitted from January 2015 to December 2020 were collected. The inclusion criteria were as follows: (1) age from 18 to 80 years old; (2) the first and subsequent surgeries for thyroid were performed in one single center; (3) the postoperative paraffin pathology confirmed thyroid tumor; and (4) the patient's clinical data were complete. In this study, the gender, age, surgical methods, tumor types, maximum diameter of tumor, metastasis of central neck group and lateral lymph nodes, postoperative complications, and length of hospital stay were collected and recorded. For patients with multiple surgeries, the reason for subsequent surgeries and the interval time between surgeries should be additionally recorded.&lt;/p&gt;&lt;p&gt;A total of 58 patients undergoing thyroid surgeries (1 patient with laparoscopic surgery was excluded) were collected in this study, as shown in Table 1. Fifteen patients in the observation group underwent central lymph node dissection and 26 patients in the lateral lymph node dissection, while only 1 patient in the control group underwent cervical lateral lymph node dissection, and there was a statistically significant difference in the overall surgical method between the two groups (&lt;i&gt;p&lt;/i&gt; &lt; 0.01).&lt;/p&gt;&lt;p&gt;In the control group, 4 cases of benign tumors and 21 cases of papillary carcinoma were confirmed by pathology after the surgery. However, 55 cases of postoperative pathology confirmed malignant tumors in the observation group, including 52 cases of papillary carcinoma, 2 cases of follicular carcinoma, and 1 case of medullary carcinoma. In patients with confirmed papillary carcinoma, the mean tumor size was 1.33 ± 0.14 cm (&lt;i&gt;p&lt;/i&gt; &lt; 0.001). The rate of lymph node metastasis in the central group was 27 out of 118 (&lt;i&gt;p&lt;/i&gt; &lt; 0.0001) and the rate of lateral lymph node metastasis was 162 out of 241 (&lt;i&gt;p&lt;/i&gt; &lt; 0.01), as shown in Table 1.&lt;/p&gt;&lt;p&gt;The average length of hospital stay in the observation group was 5.64 ± 0.30 days (&lt;i&gt;p&lt;/i&gt; &lt; 0.05). There were five cases of adverse reactions (&lt;i&gt;p&lt;/i&gt; &gt; 0.05) after the operation, including four cases of hoarseness, one case of choking cough (with hoarseness), and one case of hemorrhage. The average length of hospitalization in the control group was 4.44 ± 0.38 days, and no patients had obvious adverse reactions (Table S1).&lt;/p&gt;&lt;p&gt;Out of 58 patients who underwent multiple surgeries, 51 patients underwent two surgeries, and 7 patients underwent three surgeries. Am","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70008","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143114800","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}
引用次数: 0
Unraveling the underlying mechanisms of cancer stem cells in therapeutic resistance for optimizing treatment strategies
Pub Date : 2025-01-10 DOI: 10.1002/mog2.70009
Yunhan Tan, Siyuan Qin, Zhe Zhang, Yongen Liu, Li Zhou, Bowen Li, Edouard C. Nice, Yuanyuan Zhang, Jing Jing

The success of cancer therapy has been significantly hampered by various mechanisms of therapeutic resistance. Chief among these mechanisms is the presence of clonal heterogeneity within an individual tumor mass. The introduction of the concept of cancer stem cells (CSCs)—a rare and immature subpopulation with tumorigenic potential that contributes to intratumoral heterogeneity—has deepened our understanding of drug resistance. Given the characteristics of CSCs, such as increased drug-efflux activity, enhanced DNA-repair capacity, high metabolic plasticity, adaptability to oxidative stress, and/or upregulated detoxifying aldehyde dehydrogenase (ALDH) enzymes, CSCs have been recognized as a theoretical reservoir for resistant diseases. Implicit in this recognition is the possibility that CSC-targeted therapeutic strategies might offer a breakthrough in overcoming drug resistance in cancer patients. Herein, we summarize the generation of CSCs and our current understanding of the mechanisms underlying CSC-mediated therapeutic resistance. This extended knowledge has progressively been translated into novel anticancer therapeutic strategies and significantly enriched the available options for combination treatments, all of which are anticipated to improve clinical outcomes for patients experiencing CSC-related relapse.

{"title":"Unraveling the underlying mechanisms of cancer stem cells in therapeutic resistance for optimizing treatment strategies","authors":"Yunhan Tan,&nbsp;Siyuan Qin,&nbsp;Zhe Zhang,&nbsp;Yongen Liu,&nbsp;Li Zhou,&nbsp;Bowen Li,&nbsp;Edouard C. Nice,&nbsp;Yuanyuan Zhang,&nbsp;Jing Jing","doi":"10.1002/mog2.70009","DOIUrl":"https://doi.org/10.1002/mog2.70009","url":null,"abstract":"<p>The success of cancer therapy has been significantly hampered by various mechanisms of therapeutic resistance. Chief among these mechanisms is the presence of clonal heterogeneity within an individual tumor mass. The introduction of the concept of cancer stem cells (CSCs)—a rare and immature subpopulation with tumorigenic potential that contributes to intratumoral heterogeneity—has deepened our understanding of drug resistance. Given the characteristics of CSCs, such as increased drug-efflux activity, enhanced DNA-repair capacity, high metabolic plasticity, adaptability to oxidative stress, and/or upregulated detoxifying aldehyde dehydrogenase (ALDH) enzymes, CSCs have been recognized as a theoretical reservoir for resistant diseases. Implicit in this recognition is the possibility that CSC-targeted therapeutic strategies might offer a breakthrough in overcoming drug resistance in cancer patients. Herein, we summarize the generation of CSCs and our current understanding of the mechanisms underlying CSC-mediated therapeutic resistance. This extended knowledge has progressively been translated into novel anticancer therapeutic strategies and significantly enriched the available options for combination treatments, all of which are anticipated to improve clinical outcomes for patients experiencing CSC-related relapse.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70009","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143113766","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}
引用次数: 0
Oxidative stress and cellular senescence: Roles in tumor progression and therapeutic opportunities
Pub Date : 2024-12-25 DOI: 10.1002/mog2.70007
Ping Jin, Xu-Dong Feng, Cheng-Shuang Huang, Jia Li, Hui Wang, Xian-Mei Wang, Lei Li, Lan-Qing Ma

Oxidative stress results from an imbalance between the production and neutralization of reactive oxygen species. It induces oxidative damage to cellular components including proteins, lipids, nucleic acids, and membranes, therefore intrinsically linking to aging-related diseases such as cancer, cardiovascular disease, and neurological disorders. Emerging evidence suggests that oxidative stress may promote tumor development by influencing various aspects of cellular senescence, such as its onset, pro-inflammatory secretion, and alteration of cellular function and structure. Modulating oxidative stress to target cellular senescence offers a novel strategy for cancer prevention and treatment. However, a thorough grasp of the specific mechanisms at play is lacking. This review will present the association between oxidative stress and cellular senescence and their regulatory role in tumor progression and treatment, with emphasis on senescence-associated secretory phenotype, immunosenescence and therapy-induced senescence. Current agents and strategies that remove side effects of cellular senescence via killing senescent cancer cells or modulating oxidative stress to improve antitumor efficacy will be summarized. This review will help readers better understand the complex relationship between oxidative stress and senescence in cancer, and will also provide a basis for further research in this area.

{"title":"Oxidative stress and cellular senescence: Roles in tumor progression and therapeutic opportunities","authors":"Ping Jin,&nbsp;Xu-Dong Feng,&nbsp;Cheng-Shuang Huang,&nbsp;Jia Li,&nbsp;Hui Wang,&nbsp;Xian-Mei Wang,&nbsp;Lei Li,&nbsp;Lan-Qing Ma","doi":"10.1002/mog2.70007","DOIUrl":"https://doi.org/10.1002/mog2.70007","url":null,"abstract":"<p>Oxidative stress results from an imbalance between the production and neutralization of reactive oxygen species. It induces oxidative damage to cellular components including proteins, lipids, nucleic acids, and membranes, therefore intrinsically linking to aging-related diseases such as cancer, cardiovascular disease, and neurological disorders. Emerging evidence suggests that oxidative stress may promote tumor development by influencing various aspects of cellular senescence, such as its onset, pro-inflammatory secretion, and alteration of cellular function and structure. Modulating oxidative stress to target cellular senescence offers a novel strategy for cancer prevention and treatment. However, a thorough grasp of the specific mechanisms at play is lacking. This review will present the association between oxidative stress and cellular senescence and their regulatory role in tumor progression and treatment, with emphasis on senescence-associated secretory phenotype, immunosenescence and therapy-induced senescence. Current agents and strategies that remove side effects of cellular senescence via killing senescent cancer cells or modulating oxidative stress to improve antitumor efficacy will be summarized. This review will help readers better understand the complex relationship between oxidative stress and senescence in cancer, and will also provide a basis for further research in this area.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70007","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143119011","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}
引用次数: 0
Redox regulation of cancer stem cells: Biology and therapeutic implications 癌症干细胞的氧化还原调节:生物学和治疗学意义
Pub Date : 2024-12-14 DOI: 10.1002/mog2.70005
Min Du, Jian Zhang, Max S. Wicha, Ming Luo

Cancer stem cells (CSCs) are a small group of tumor cells with the capacity to undergo self-renewal and differentiation. These cells not only initiate and maintain tumor growth, but also confer resistance to current cancer therapies. CSCs display a high degree of plasticity and can be generated under therapeutic stress via dedifferentiation from non-stem-like tumor cells, suggesting the necessity simultaneously targeting CSCs and bulk tumor cells to achieve the best therapeutic effect. Despite the findings that therapeutic stress induces CSC plasticity, the mechanisms underpinning CSC formation and therapeutic resistance are not fully defined. Tumor cells display elevated levels of reactive oxygen species (ROS), contributed by rapid proliferation, enhanced metabolic demands and oncogenic signaling. CSCs achieve redox homeostasis partly by regulating redox-sensitive transcription factors (TFs), including NRF2, HIF-1α, BACH1, NF-kB, FOXOs, AP-1, and others. This review aims to summarize the roles and underlying mechanisms of these TFs in regulation of CSCs and tumor progression from the perspectives of stem cell maintenance, metabolic reprogramming, epithelial–mesenchymal transition (EMT) and angiogenesis. We also discuss the potentials of utilizing specific inhibitors for these TFs in suppressing drug resistance and metastasis by repressing CSC activity, an approach that may provide new targeted therapies for advanced cancer and improve patient outcome.

肿瘤干细胞(CSCs)是一小群具有自我更新和分化能力的肿瘤细胞。这些细胞不仅启动和维持肿瘤生长,而且还赋予对当前癌症治疗的抵抗力。CSCs具有高度的可塑性,可在治疗应激条件下由非干样肿瘤细胞去分化而产生,提示需同时靶向CSCs和大块肿瘤细胞以达到最佳治疗效果。尽管研究发现治疗性应激可诱导CSC的可塑性,但支持CSC形成和治疗抗性的机制尚未完全确定。肿瘤细胞表现出活性氧(ROS)水平升高,这是由快速增殖、代谢需求增强和致癌信号传导所导致的。CSCs部分通过调节氧化还原敏感转录因子(TFs)实现氧化还原稳态,包括NRF2、HIF-1α、BACH1、NF-kB、FOXOs、AP-1等。本文旨在从干细胞维持、代谢重编程、上皮-间充质转化(epithelial-mesenchymal transition, EMT)和血管生成等方面综述这些tgf在CSCs和肿瘤进展调控中的作用及其机制。我们还讨论了利用这些tf的特异性抑制剂通过抑制CSC活性来抑制耐药和转移的潜力,这种方法可能为晚期癌症提供新的靶向治疗并改善患者预后。
{"title":"Redox regulation of cancer stem cells: Biology and therapeutic implications","authors":"Min Du,&nbsp;Jian Zhang,&nbsp;Max S. Wicha,&nbsp;Ming Luo","doi":"10.1002/mog2.70005","DOIUrl":"https://doi.org/10.1002/mog2.70005","url":null,"abstract":"<p>Cancer stem cells (CSCs) are a small group of tumor cells with the capacity to undergo self-renewal and differentiation. These cells not only initiate and maintain tumor growth, but also confer resistance to current cancer therapies. CSCs display a high degree of plasticity and can be generated under therapeutic stress via dedifferentiation from non-stem-like tumor cells, suggesting the necessity simultaneously targeting CSCs and bulk tumor cells to achieve the best therapeutic effect. Despite the findings that therapeutic stress induces CSC plasticity, the mechanisms underpinning CSC formation and therapeutic resistance are not fully defined. Tumor cells display elevated levels of reactive oxygen species (ROS), contributed by rapid proliferation, enhanced metabolic demands and oncogenic signaling. CSCs achieve redox homeostasis partly by regulating redox-sensitive transcription factors (TFs), including NRF2, HIF-1α, BACH1, NF-kB, FOXOs, AP-1, and others. This review aims to summarize the roles and underlying mechanisms of these TFs in regulation of CSCs and tumor progression from the perspectives of stem cell maintenance, metabolic reprogramming, epithelial–mesenchymal transition (EMT) and angiogenesis. We also discuss the potentials of utilizing specific inhibitors for these TFs in suppressing drug resistance and metastasis by repressing CSC activity, an approach that may provide new targeted therapies for advanced cancer and improve patient outcome.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70005","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142861114","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}
引用次数: 0
CSF@E-Hn: A bone marrow-targeted nanosystem for advanced treatment of hematological malignancies CSF@E-Hn:骨髓靶向纳米系统用于血液系统恶性肿瘤的高级治疗
Pub Date : 2024-12-14 DOI: 10.1002/mog2.70006
Yinyan Jiang, Jianqiao Shentu, Yalu Chen
<p>In a recent study published in <i>Nature Nanotechnology</i>, the research teams of Professor Siwen Li and Professor Yueqing Gu introduced a bispecific bone marrow-targeted nanosystem, CSF@E-Hn, based on hematopoietic stem cell (HSC) nanovesicles (Hn).<span><sup>1</sup></span> The system uses Hn vesicles, decorated with natural killer (NK) cell-activating ligands (aNKG2D) and tumor-targeting antibodies (aPD-L1), encapsulating colony-stimulating factor (CSF) to treat hematological malignancies. Experimental results confirmed the system's therapeutic efficacy in mouse models of acute myelogenous leukemia (AML) and multiple myeloma (MM) and demonstrated its ability to prevent tumor recurrence long-term.</p><p>Most malignant hematological tumors arise from uncontrolled clonal expansion of tumor cells within the bone marrow, leading to high mortality and recurrence rates. Although current treatments, including chemotherapy, immunotherapy, and cell therapy, have improved overall survival in patients with hematological malignancies, these strategies still face significant challenges. Targeting bone marrow tumor cells specifically, reducing toxic side effects, and preventing recurrence remain major hurdles due to the lack of effective bone marrow-targeting technologies and the difficulty in reversing the diseased bone marrow microenvironment.<span><sup>2, 3</sup></span> The unique physiological structure of the bone marrow also acts as a formidable barrier, severely limiting the development of in vivo targeting technologies. In this context, Hn shows great potential for overcoming these challenges. As an ideal carrier, Hn has several advantages: it mirrors the characteristics of parent cells, features a drug-loaded bilayer structure, has a small size, and exhibits low immunogenicity.<span><sup>4, 5</sup></span> However, how to translate these significant advantages into clinical treatment remains a critical issue in current research.</p><p>To address these challenges, the bone marrow-targeted nanosystem CSF@E-Hn, developed by the team of Professor Li and Professor Gu, innovatively fuses nanotechnology with immunotherapy. This system accomplishes precise bone marrow-targeted treatment by capitalizing on the natural bone marrow hematopoietic stem cells and the generation of memory T cells, thus remodeling the bone marrow microenvironment and augmenting the immune response. Additionally, the system possesses excellent biocompatibility and sustained release properties, guaranteeing favorable safety (Figure 1).</p><p>The research team conducted a comprehensive evaluation of CSF@E-Hn's performance and efficacy. In vitro experiments demonstrated that the nanosystem, after antibody modification of HSC cells, maintained stable size distribution in phosphate-buffered saline (PBS) and serum. CSF@E-Hn remained stable for up to 14 days under low-temperature storage and thawing at −80°C. When NK cells were isolated from the bone marrow of C57BL/6J mice, CSF@E-Hn eff
在最近发表在《自然纳米技术》杂志上的一项研究中,李思文教授和顾月清教授的研究团队介绍了一种基于造血干细胞(HSC)纳米囊泡(Hn)的双特异性骨髓靶向纳米系统CSF@E-Hn该系统使用Hn囊泡,用自然杀伤(NK)细胞激活配体(aNKG2D)和肿瘤靶向抗体(aPD-L1)装饰,包封集落刺激因子(CSF)来治疗血液系统恶性肿瘤。实验结果证实了该系统对急性髓性白血病(AML)和多发性骨髓瘤(MM)小鼠模型的治疗效果,并证明了其长期预防肿瘤复发的能力。大多数恶性血液学肿瘤起源于骨髓内肿瘤细胞不受控制的克隆扩增,导致高死亡率和复发率。尽管目前的治疗方法,包括化疗、免疫治疗和细胞治疗,已经提高了血液系统恶性肿瘤患者的总体生存率,但这些策略仍然面临着重大挑战。由于缺乏有效的骨髓靶向技术和难以逆转病变骨髓微环境,特异性靶向骨髓肿瘤细胞、减少毒副作用和防止复发仍然是主要障碍。2,3骨髓独特的生理结构也是一个强大的屏障,严重限制了体内靶向技术的发展。在这种情况下,Hn显示出克服这些挑战的巨大潜力。作为一种理想的载体,Hn具有以下优点:反映亲本细胞的特性,具有载药双层结构,体积小,免疫原性低。然而,如何将这些显著优势转化为临床治疗仍然是当前研究的关键问题。为了应对这些挑战,由李教授和顾教授团队开发的骨髓靶向纳米系统CSF@E-Hn将纳米技术与免疫疗法创新地融合在一起。该系统利用天然骨髓造血干细胞和记忆T细胞的生成,实现骨髓靶向治疗的精准化,从而重塑骨髓微环境,增强免疫应答。此外,该系统具有良好的生物相容性和缓释性能,保证了良好的安全性(图1)。课题组对CSF@E-Hn的性能和功效进行了综合评价。体外实验表明,经抗体修饰后的纳米系统在磷酸盐缓冲盐水(PBS)和血清中保持稳定的大小分布。CSF@E-Hn在低温储存和- 80°C解冻下保持稳定长达14天。当从C57BL/6J小鼠骨髓中分离NK细胞时,CSF@E-Hn有效地将NK细胞和C1498细胞结合在混合细胞培养基中。通过Förster共振能量转移和透射电子显微镜验证了这种相互作用,证实了纳米系统捕获这些细胞的能力。该研究还表明,CSF@E-Hn激活NK细胞,抑制肿瘤细胞增殖,而不影响健康细胞。进一步的研究评估了CSF@E-Hn的骨髓归巢能力。结果显示,该体系注射后小鼠血液循环延长,苏木精-伊红(H&amp;E)染色未见明显病理改变。虽然肝脏和脾脏的荧光信号较强,但安全性评估显示无明显毒性。急性毒性试验进一步证实了其安全性。在C1498荷瘤小鼠模型中,CSF@E-Hn治疗显著根除肿瘤细胞,80天后存活率达到87.5%。同时,CSF@E-Hn治疗组的白细胞计数和体重均优于其他治疗组。脾脏形态正常,造血细胞、树突状细胞、巨噬细胞显著增加,表明CSF@E-Hn有效调节骨髓免疫环境,恢复造血功能。在MM模型中,CSF@E-Hn减少破骨细胞和增加成骨细胞,进一步强调其治疗血液系统恶性肿瘤的潜力。这些综合评价结果表明,CSF@E-Hn纳米系统不仅在体外表现出良好的稳定性和细胞捕获能力,而且在体内也表现出显著的治疗效果和令人满意的生物安全性,从而为治疗血液系统恶性肿瘤提供了一种有希望的新策略。尽管取得了令人鼓舞的结果,但有关CSF@E-Hn结论的几个关键问题仍然存在。虽然CSF@E-Hn具有捕获NK细胞和肿瘤细胞、延长循环时间、抑制肿瘤增殖的功效,但其在更广泛的临床应用中的表现仍不确定。 动物模型的结果并不总是能预测人类临床结果,需要进一步的研究来确定CSF@E-Hn在临床试验中的可行性。鉴于原文小鼠模型为同源肿瘤移植模型,未来可逐步开展人类肿瘤异种移植模型或基因人源化小鼠模型的研究,最终启动小规模的I期临床试验。此外,长期的安全问题需要引起注意。虽然初步评估显示没有明显的毒性,但长期使用的潜在延迟副作用或意想不到的生物反应仍然未知。生物分布和组织积累可能对器官功能产生不可预见的影响,这需要进一步探索。另一个问题是CSF@E-Hn在治疗不同肿瘤或血液疾病方面的多功能性。虽然目前的研究主要集中在AML和MM,但其对其他血液系统恶性肿瘤或实体瘤的疗效仍有待评估。此外,个体患者的可变性和系统在不同人口统计学中的有效性需要通过系统的临床试验来证实。另一个重要的考虑因素是CSF@E-Hn的生产和运营成本。虽然该系统在实验环境中表现良好,但确保其经济可行性和持续生产将是临床应用的关键挑战。高效、安全和具有成本效益的制造工艺将直接影响CSF@E-Hn的临床采用和广泛使用。虽然可以通过超离心获得高纯度的Hn,但设备昂贵,操作耗时长。因此,可以考虑采用更具成本效益和效率的替代方法,如超滤、基于电荷中和的聚合物沉淀、粒径排除色谱和微流体。此外,在抗体偶联过程中,使用了大量的生物素化抗体(如aPD-L1和aNKG2D),导致抗体成本较高。为了降低成本,可以采用抗体片段化或位点特异性偶联技术来减少抗体的使用量,同时确保偶联效率和功能。此外,抗体与纳米囊泡的比例可以优化,以减少不必要的浪费。总之,CSF@E-Hn作为一种新的血液恶性肿瘤治疗策略显示出巨大的希望。随着进一步的临床试验和长期安全性评估,我们预计CSF@E-Hn可能成为治疗AML和MM的有价值的工具。如果在其他肿瘤类型中得到验证,它有可能在广泛的癌症治疗中发挥重要作用。持续的研究和创新将进一步揭示CSF@E-Hn的综合价值,从而产生更精确和有效的治疗策略。蒋银燕:概念化(主持);可视化(平等);写作-原稿(同等)。申图简桥:概念化(平等);可视化(平等);写作——原稿(引子)。陈亚璐:融资获取(等额);写作-审查和编辑(主导)。所有作者都已阅读并同意稿件的出版版本。作者声明无利益冲突。不适用。
{"title":"CSF@E-Hn: A bone marrow-targeted nanosystem for advanced treatment of hematological malignancies","authors":"Yinyan Jiang,&nbsp;Jianqiao Shentu,&nbsp;Yalu Chen","doi":"10.1002/mog2.70006","DOIUrl":"https://doi.org/10.1002/mog2.70006","url":null,"abstract":"&lt;p&gt;In a recent study published in &lt;i&gt;Nature Nanotechnology&lt;/i&gt;, the research teams of Professor Siwen Li and Professor Yueqing Gu introduced a bispecific bone marrow-targeted nanosystem, CSF@E-Hn, based on hematopoietic stem cell (HSC) nanovesicles (Hn).&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt; The system uses Hn vesicles, decorated with natural killer (NK) cell-activating ligands (aNKG2D) and tumor-targeting antibodies (aPD-L1), encapsulating colony-stimulating factor (CSF) to treat hematological malignancies. Experimental results confirmed the system's therapeutic efficacy in mouse models of acute myelogenous leukemia (AML) and multiple myeloma (MM) and demonstrated its ability to prevent tumor recurrence long-term.&lt;/p&gt;&lt;p&gt;Most malignant hematological tumors arise from uncontrolled clonal expansion of tumor cells within the bone marrow, leading to high mortality and recurrence rates. Although current treatments, including chemotherapy, immunotherapy, and cell therapy, have improved overall survival in patients with hematological malignancies, these strategies still face significant challenges. Targeting bone marrow tumor cells specifically, reducing toxic side effects, and preventing recurrence remain major hurdles due to the lack of effective bone marrow-targeting technologies and the difficulty in reversing the diseased bone marrow microenvironment.&lt;span&gt;&lt;sup&gt;2, 3&lt;/sup&gt;&lt;/span&gt; The unique physiological structure of the bone marrow also acts as a formidable barrier, severely limiting the development of in vivo targeting technologies. In this context, Hn shows great potential for overcoming these challenges. As an ideal carrier, Hn has several advantages: it mirrors the characteristics of parent cells, features a drug-loaded bilayer structure, has a small size, and exhibits low immunogenicity.&lt;span&gt;&lt;sup&gt;4, 5&lt;/sup&gt;&lt;/span&gt; However, how to translate these significant advantages into clinical treatment remains a critical issue in current research.&lt;/p&gt;&lt;p&gt;To address these challenges, the bone marrow-targeted nanosystem CSF@E-Hn, developed by the team of Professor Li and Professor Gu, innovatively fuses nanotechnology with immunotherapy. This system accomplishes precise bone marrow-targeted treatment by capitalizing on the natural bone marrow hematopoietic stem cells and the generation of memory T cells, thus remodeling the bone marrow microenvironment and augmenting the immune response. Additionally, the system possesses excellent biocompatibility and sustained release properties, guaranteeing favorable safety (Figure 1).&lt;/p&gt;&lt;p&gt;The research team conducted a comprehensive evaluation of CSF@E-Hn's performance and efficacy. In vitro experiments demonstrated that the nanosystem, after antibody modification of HSC cells, maintained stable size distribution in phosphate-buffered saline (PBS) and serum. CSF@E-Hn remained stable for up to 14 days under low-temperature storage and thawing at −80°C. When NK cells were isolated from the bone marrow of C57BL/6J mice, CSF@E-Hn eff","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70006","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142861158","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}
引用次数: 0
WNT2 blockade augments antitumor immunity by attenuating myeloid-derived suppressor cells in colorectal cancer WNT2阻断通过减弱结直肠癌骨髓源性抑制细胞增强抗肿瘤免疫
Pub Date : 2024-12-04 DOI: 10.1002/mog2.70004
Cheng Cui, Tian-Tian Zhang, Qian Lin, Tu-Xiong Huang, En-Yu Rao, Ji-Hui Du, Li Fu

Colorectal cancer (CRC) ranks as one of the most common malignancies worldwide. Myeloid-derived suppressor cells (MDSCs) represent an immunosuppressive heterogeneous population of immature monocytes and granulocytes constituting a major obstacle for CRC therapy. Previous studies demonstrated that WNT2 is enriched in tumor microenvironment (TME), promoting CRC progression. However, the role of WNT2 in regulating MDSCs to facilitate CRC progression remains largely unexplored. Our analysis of The Cancer Genome Atlas (TCGA) database and blood samples from 50 primary and recurrent CRC patients revealed a positive correlation between WNT2 expression and MDSCs abundance. Treatment with recombinant WNT2 protein significantly enhanced the accumulation and immunosuppressive function of MDSCs in vitro. Conversely, anti-WNT2 monoclonal antibody remarkably reduced the percentage and functional activity of MDSCs in CRC tumor-bearing mice. Mechanistic analyses further demonstrated that WNT2 mediates MDSCs activities through the p38 MAPK/Akt pathway. Collectively, our findings not only highlight the pivotal role of WNT2 in CRC progression by enhancing MDSCs activities within the TME, but also provide evidence that WNT2 levels and MDSCs abundance in peripheral blood could serve as predictive biomarkers for early diagnosis and prognosis of CRC patients.

结直肠癌(CRC)是世界上最常见的恶性肿瘤之一。髓源性抑制细胞(MDSCs)是一种免疫抑制异质性的未成熟单核细胞和粒细胞群体,是CRC治疗的主要障碍。既往研究表明,WNT2在肿瘤微环境(tumor microenvironment, TME)中富集,促进结直肠癌的进展。然而,WNT2在调节MDSCs促进结直肠癌进展中的作用在很大程度上仍未被探索。我们对癌症基因组图谱(TCGA)数据库和50例原发性和复发性CRC患者的血液样本进行分析,发现WNT2表达与MDSCs丰度呈正相关。重组WNT2蛋白处理可显著增强MDSCs的体外积累和免疫抑制功能。相反,抗wnt2单克隆抗体显著降低CRC荷瘤小鼠中MDSCs的百分比和功能活性。机制分析进一步表明,WNT2通过p38 MAPK/Akt通路介导MDSCs的活性。总的来说,我们的研究结果不仅强调了WNT2通过增强TME内MDSCs的活性在CRC进展中的关键作用,而且还提供了外周血中WNT2水平和MDSCs丰度可以作为CRC患者早期诊断和预后的预测性生物标志物的证据。
{"title":"WNT2 blockade augments antitumor immunity by attenuating myeloid-derived suppressor cells in colorectal cancer","authors":"Cheng Cui,&nbsp;Tian-Tian Zhang,&nbsp;Qian Lin,&nbsp;Tu-Xiong Huang,&nbsp;En-Yu Rao,&nbsp;Ji-Hui Du,&nbsp;Li Fu","doi":"10.1002/mog2.70004","DOIUrl":"https://doi.org/10.1002/mog2.70004","url":null,"abstract":"<p>Colorectal cancer (CRC) ranks as one of the most common malignancies worldwide. Myeloid-derived suppressor cells (MDSCs) represent an immunosuppressive heterogeneous population of immature monocytes and granulocytes constituting a major obstacle for CRC therapy. Previous studies demonstrated that WNT2 is enriched in tumor microenvironment (TME), promoting CRC progression. However, the role of WNT2 in regulating MDSCs to facilitate CRC progression remains largely unexplored. Our analysis of The Cancer Genome Atlas (TCGA) database and blood samples from 50 primary and recurrent CRC patients revealed a positive correlation between WNT2 expression and MDSCs abundance. Treatment with recombinant WNT2 protein significantly enhanced the accumulation and immunosuppressive function of MDSCs in vitro. Conversely, anti-WNT2 monoclonal antibody remarkably reduced the percentage and functional activity of MDSCs in CRC tumor-bearing mice. Mechanistic analyses further demonstrated that WNT2 mediates MDSCs activities through the p38 MAPK/Akt pathway. Collectively, our findings not only highlight the pivotal role of WNT2 in CRC progression by enhancing MDSCs activities within the TME, but also provide evidence that WNT2 levels and MDSCs abundance in peripheral blood could serve as predictive biomarkers for early diagnosis and prognosis of CRC patients.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70004","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142859967","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}
引用次数: 0
期刊
MedComm – Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1