首页 > 最新文献

Experimental Hematology & Oncology最新文献

英文 中文
M2-like tumor-associated macrophage-secreted CCL2 facilitates gallbladder cancer stemness and metastasis. M2样肿瘤相关巨噬细胞分泌的CCL2有助于胆囊癌的干细胞形成和转移。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-13 DOI: 10.1186/s40164-024-00550-2
Weihong Chen, Mingyuan Chen, Lingju Hong, Abudukeremu Xiahenazi, Maotuan Huang, Nanhong Tang, Xinyue Yang, Feifei She, Yanling Chen

Background: The predominant immune cells in solid tumors are M2-like tumor-associated macrophages (M2-like TAMs), which significantly impact the promotion of epithelial-mesenchymal transition (EMT) in tumors, enhancing stemness and facilitating tumor invasion and metastasis. However, the contribution of M2-like TAMs to tumor progression in gallbladder cancer (GBC) is partially known.

Methods: Immunohistochemistry was used to evaluate the expression of M2-like TAMs and cancer stem cell (CSC) markers in 24 pairs of GBC and adjacent noncancerous tissues from patients with GBC. Subsequently, GBC cells and M2-like TAMs were co-cultured to examine the expression of CSC markers, EMT markers, and migratory behavior. Proteomics was performed on the culture supernatant of M2-like TAMs. The mechanisms underlying the induction of EMT, stemness, and metastasis in GBC by M2-like TAMs were elucidated using proteomics and transcriptomics. GBC cells were co-cultured with undifferentiated macrophages (M0) and analyzed. The therapeutic effect of gemcitabine combined with a chemokine (C-C motif) receptor 2 (CCR2) antagonist on GBC was observed in vivo.

Results: The expression levels of CD68 and CD163 in M2-like TAMs and CD44 and CD133 in gallbladder cancer stem cells (GBCSCs) were increased and positively correlated in GBC tissues compared with those in neighboring noncancerous tissues. M2-like TAMs secreted a significant amount of chemotactic cytokine ligand 2 (CCL2), which activated the MEK/extracellular regulated protein kinase (ERK) pathway and enhanced SNAIL expression after binding to the receptor CCR2 on GBC cells. Activation of the ERK pathway caused nuclear translocation of ELK1, which subsequently led to increased SNAIL expression. GBCSCs mediated the recruitment and polarization of M0 into M2-like TAMs within the GBC microenvironment via CCL2 secretion. In the murine models, the combination of a CCR2 antagonist and gemcitabine efficiently inhibited the growth of subcutaneous tumors in GBC.

Conclusions: The interaction between M2-like TAMs and GBC cells is mediated by the chemokine CCL2, which activates the MEK/ERK/ELK1/SNAIL pathway in GBC cells, promoting EMT, stemness, and metastasis. A combination of a CCR2 inhibitor and gemcitabine effectively suppressed the growth of subcutaneous tumors. Consequently, our study identified promising therapeutic targets and strategies for treating GBC.

背景:实体瘤中最主要的免疫细胞是M2样肿瘤相关巨噬细胞(M2样TAMs),它们对促进肿瘤上皮-间质转化(EMT)、增强干性、促进肿瘤侵袭和转移具有重要影响。然而,M2样TAMs对胆囊癌(GBC)肿瘤进展的贡献尚不完全清楚:免疫组化法评估了24对GBC患者的GBC和邻近非癌组织中M2样TAMs和癌症干细胞(CSC)标记物的表达。随后,将 GBC 细胞和 M2 样 TAMs 共同培养,以检测 CSC 标记、EMT 标记和迁移行为的表达。对M2样TAMs的培养上清进行了蛋白质组学研究。利用蛋白质组学和转录组学阐明了M2样TAMs诱导GBC的EMT、干性和转移的机制。将GBC细胞与未分化巨噬细胞(M0)共培养并进行分析。在体内观察了吉西他滨联合趋化因子(C-C基序)受体2(CCR2)拮抗剂对GBC的治疗效果:结果:与邻近的非癌组织相比,GBC组织中M2样TAMs的CD68和CD163以及胆囊癌干细胞(GBCSCs)的CD44和CD133的表达水平升高并呈正相关。M2样TAMs分泌大量趋化细胞因子配体2(CCL2),与GBC细胞上的受体CCR2结合后激活MEK/细胞外调节蛋白激酶(ERK)通路并增强SNAIL的表达。ERK通路的激活引起了ELK1的核转位,进而导致SNAIL的表达增加。GBCSCs通过分泌CCL2介导了GBC微环境中M0向M2样TAMs的招募和极化。在小鼠模型中,CCR2拮抗剂与吉西他滨联合使用可有效抑制GBC皮下肿瘤的生长:结论:M2样TAMs与GBC细胞之间的相互作用是由趋化因子CCL2介导的,它激活了GBC细胞中的MEK/ERK/ELK1/SNAIL通路,促进了EMT、干性和转移。CCR2抑制剂与吉西他滨联合使用可有效抑制皮下肿瘤的生长。因此,我们的研究发现了治疗GBC的有希望的治疗靶点和策略。
{"title":"M2-like tumor-associated macrophage-secreted CCL2 facilitates gallbladder cancer stemness and metastasis.","authors":"Weihong Chen, Mingyuan Chen, Lingju Hong, Abudukeremu Xiahenazi, Maotuan Huang, Nanhong Tang, Xinyue Yang, Feifei She, Yanling Chen","doi":"10.1186/s40164-024-00550-2","DOIUrl":"10.1186/s40164-024-00550-2","url":null,"abstract":"<p><strong>Background: </strong>The predominant immune cells in solid tumors are M2-like tumor-associated macrophages (M2-like TAMs), which significantly impact the promotion of epithelial-mesenchymal transition (EMT) in tumors, enhancing stemness and facilitating tumor invasion and metastasis. However, the contribution of M2-like TAMs to tumor progression in gallbladder cancer (GBC) is partially known.</p><p><strong>Methods: </strong>Immunohistochemistry was used to evaluate the expression of M2-like TAMs and cancer stem cell (CSC) markers in 24 pairs of GBC and adjacent noncancerous tissues from patients with GBC. Subsequently, GBC cells and M2-like TAMs were co-cultured to examine the expression of CSC markers, EMT markers, and migratory behavior. Proteomics was performed on the culture supernatant of M2-like TAMs. The mechanisms underlying the induction of EMT, stemness, and metastasis in GBC by M2-like TAMs were elucidated using proteomics and transcriptomics. GBC cells were co-cultured with undifferentiated macrophages (M0) and analyzed. The therapeutic effect of gemcitabine combined with a chemokine (C-C motif) receptor 2 (CCR2) antagonist on GBC was observed in vivo.</p><p><strong>Results: </strong>The expression levels of CD68 and CD163 in M2-like TAMs and CD44 and CD133 in gallbladder cancer stem cells (GBCSCs) were increased and positively correlated in GBC tissues compared with those in neighboring noncancerous tissues. M2-like TAMs secreted a significant amount of chemotactic cytokine ligand 2 (CCL2), which activated the MEK/extracellular regulated protein kinase (ERK) pathway and enhanced SNAIL expression after binding to the receptor CCR2 on GBC cells. Activation of the ERK pathway caused nuclear translocation of ELK1, which subsequently led to increased SNAIL expression. GBCSCs mediated the recruitment and polarization of M0 into M2-like TAMs within the GBC microenvironment via CCL2 secretion. In the murine models, the combination of a CCR2 antagonist and gemcitabine efficiently inhibited the growth of subcutaneous tumors in GBC.</p><p><strong>Conclusions: </strong>The interaction between M2-like TAMs and GBC cells is mediated by the chemokine CCL2, which activates the MEK/ERK/ELK1/SNAIL pathway in GBC cells, promoting EMT, stemness, and metastasis. A combination of a CCR2 inhibitor and gemcitabine effectively suppressed the growth of subcutaneous tumors. Consequently, our study identified promising therapeutic targets and strategies for treating GBC.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11320879/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141975451","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hematopoietic stem/progenitor cell transplantation recovers immune defects and prevents lymphomas in Atm-deficient mice. 造血干细胞/祖细胞移植可恢复 Atm 缺陷小鼠的免疫缺陷并预防淋巴瘤。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-06 DOI: 10.1186/s40164-024-00544-0
Bruna Sabino Pinho de Oliveira, Alessandro Giovinazzo, Sabrina Putti, Matilde Merolle, Tiziana Orsini, Giuseppe D Tocchini-Valentini, Christophe Lancrin, Fabio Naro, Manuela Pellegrini

Background: Ataxia-telangiectasia (A-T) is a rare autosomal recessive multi-system and life-shortening disease, characterized by progressive cerebellar neurodegeneration, immunodeficiency, radiation sensitivity and cancer predisposition, with high incidence of leukemia and lymphoma. A-T is caused by mutations in the gene encoding for ATM protein that has a major role in maintaining the integrity of the genome. Because there are no cures for A-T, we aimed to tackle immunodeficiency and prevent cancer onset/progression by transplantation therapy.

Methods: Enriched hematopoietic stem/progenitor cells (HSPCs), collected from bone marrow of wild-type mice, were transplanted in the caudal vein of 1 month old conditioned Atm-/- mice.

Results: Genomic analyses showed that transplanted Atm positive cells were found in lymphoid organs. B cells isolated from spleen of transplanted mice were able to undergo class switching recombination. Thymocytes were capable to correctly differentiate and consequently an increase of helper T cells and TCRβhi expressing cells was observed. Protein analysis of isolated T and B cells from transplanted mice, revealed that they expressed Atm and responded to DNA damage by initiating an Atm-dependent phosphorylation cascade. Indeed, aberrant metaphases were reduced in transplanted Atm-deficient mice. Six months after transplantation, Atm-/- mice showed signs of aging, but they maintained the rescue of T cells maturation, showed DNA damage response, and prevented thymoma.

Conclusion: We can conclude that wild-type enriched HSPCs transplantation into young Atm-deficient mice can ameliorate A-T hematopoietic phenotypes and prevent tumor of hematopoietic origin.

背景:共济失调性脊髓侧索硬化症(A-T)是一种罕见的常染色体隐性遗传多系统和缩短寿命的疾病,其特征是进行性小脑神经变性、免疫缺陷、辐射敏感性和癌症易感性,其中白血病和淋巴瘤的发病率很高。A-T是由编码ATM蛋白的基因突变引起的,ATM蛋白在维持基因组完整性方面发挥着重要作用。由于A-T无法治愈,我们的目标是通过移植疗法解决免疫缺陷问题并预防癌症的发生/发展:方法:将从野生型小鼠骨髓中采集的富集造血干细胞/祖细胞(HSPCs)移植到 1 个月大的条件Atm-/-小鼠的尾静脉中:基因组分析表明,移植的Atm阳性细胞存在于淋巴器官中。从移植小鼠脾脏中分离出的 B 细胞能够进行类重组。胸腺细胞能够正确分化,因此观察到辅助性T细胞和TCRβhi表达细胞增加。对移植小鼠分离出的 T 细胞和 B 细胞进行的蛋白质分析表明,它们表达 Atm,并通过启动 Atm 依赖性磷酸化级联对 DNA 损伤做出反应。事实上,Atm缺陷小鼠移植后的异常分裂减少了。移植6个月后,Atm-/-小鼠出现衰老迹象,但它们仍能挽救T细胞的成熟,表现出DNA损伤反应,并能预防胸腺瘤:我们可以得出结论:将野生型富集的 HSPCs 移植到年轻的 Atm 缺乏小鼠体内,可以改善 A-T 造血表型,预防造血源肿瘤。
{"title":"Hematopoietic stem/progenitor cell transplantation recovers immune defects and prevents lymphomas in Atm-deficient mice.","authors":"Bruna Sabino Pinho de Oliveira, Alessandro Giovinazzo, Sabrina Putti, Matilde Merolle, Tiziana Orsini, Giuseppe D Tocchini-Valentini, Christophe Lancrin, Fabio Naro, Manuela Pellegrini","doi":"10.1186/s40164-024-00544-0","DOIUrl":"10.1186/s40164-024-00544-0","url":null,"abstract":"<p><strong>Background: </strong>Ataxia-telangiectasia (A-T) is a rare autosomal recessive multi-system and life-shortening disease, characterized by progressive cerebellar neurodegeneration, immunodeficiency, radiation sensitivity and cancer predisposition, with high incidence of leukemia and lymphoma. A-T is caused by mutations in the gene encoding for ATM protein that has a major role in maintaining the integrity of the genome. Because there are no cures for A-T, we aimed to tackle immunodeficiency and prevent cancer onset/progression by transplantation therapy.</p><p><strong>Methods: </strong>Enriched hematopoietic stem/progenitor cells (HSPCs), collected from bone marrow of wild-type mice, were transplanted in the caudal vein of 1 month old conditioned Atm<sup>-/-</sup> mice.</p><p><strong>Results: </strong>Genomic analyses showed that transplanted Atm positive cells were found in lymphoid organs. B cells isolated from spleen of transplanted mice were able to undergo class switching recombination. Thymocytes were capable to correctly differentiate and consequently an increase of helper T cells and TCRβ<sup>hi</sup> expressing cells was observed. Protein analysis of isolated T and B cells from transplanted mice, revealed that they expressed Atm and responded to DNA damage by initiating an Atm-dependent phosphorylation cascade. Indeed, aberrant metaphases were reduced in transplanted Atm-deficient mice. Six months after transplantation, Atm<sup>-/-</sup> mice showed signs of aging, but they maintained the rescue of T cells maturation, showed DNA damage response, and prevented thymoma.</p><p><strong>Conclusion: </strong>We can conclude that wild-type enriched HSPCs transplantation into young Atm-deficient mice can ameliorate A-T hematopoietic phenotypes and prevent tumor of hematopoietic origin.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302080/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141897173","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","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 battlefield within inflamed, excluded or desert immune phenotypes: the mechanisms and strategies. 炎症、排斥或沙漠免疫表型中的肿瘤战场:机制与策略。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-06 DOI: 10.1186/s40164-024-00543-1
Siwei Zheng, Wenwen Wang, Lesang Shen, Yao Yao, Wenjie Xia, Chao Ni

The tumor microenvironment demonstrates great immunophenotypic heterogeneity, which has been leveraged in traditional immune-hot/cold tumor categorization based on the abundance of intra-tumoral immune cells. By incorporating the spatial immune contexture, the tumor immunophenotype was further elaborated into immune-inflamed, immune-excluded, and immune-desert. However, the mechanisms underlying these different immune phenotypes are yet to be comprehensively elucidated. In this review, we discuss how tumor cells and the tumor microenvironment interact collectively to shape the immune landscape from the perspectives of tumor cells, immune cells, the extracellular matrix, and cancer metabolism, and we summarize potential therapeutic options according to distinct immunophenotypes for personalized precision medicine.

肿瘤微环境表现出极大的免疫表型异质性,传统的免疫热/冷肿瘤分类法就是根据瘤内免疫细胞的丰度来进行分类的。结合空间免疫背景,肿瘤免疫表型被进一步细化为免疫炎症型、免疫排斥型和免疫荒漠型。然而,这些不同免疫表型的机制仍有待全面阐明。在这篇综述中,我们将从肿瘤细胞、免疫细胞、细胞外基质和癌症代谢的角度,讨论肿瘤细胞和肿瘤微环境如何相互作用,共同塑造免疫格局,并根据不同的免疫表型总结出个性化精准医疗的潜在治疗方案。
{"title":"Tumor battlefield within inflamed, excluded or desert immune phenotypes: the mechanisms and strategies.","authors":"Siwei Zheng, Wenwen Wang, Lesang Shen, Yao Yao, Wenjie Xia, Chao Ni","doi":"10.1186/s40164-024-00543-1","DOIUrl":"10.1186/s40164-024-00543-1","url":null,"abstract":"<p><p>The tumor microenvironment demonstrates great immunophenotypic heterogeneity, which has been leveraged in traditional immune-hot/cold tumor categorization based on the abundance of intra-tumoral immune cells. By incorporating the spatial immune contexture, the tumor immunophenotype was further elaborated into immune-inflamed, immune-excluded, and immune-desert. However, the mechanisms underlying these different immune phenotypes are yet to be comprehensively elucidated. In this review, we discuss how tumor cells and the tumor microenvironment interact collectively to shape the immune landscape from the perspectives of tumor cells, immune cells, the extracellular matrix, and cancer metabolism, and we summarize potential therapeutic options according to distinct immunophenotypes for personalized precision medicine.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11301948/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141897175","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Measurable therapeutic antibody in serum as potential predictive factor of response to anti-CD38 therapy in non-IgG-k myeloma patients. 血清中可测量的治疗抗体是非 IgG-k 骨髓瘤患者对抗 CD38 治疗反应的潜在预测因素。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-06 DOI: 10.1186/s40164-024-00547-x
Emilia Gigliotta, Federica Plano, Giusy Corsale, Anna Maria Corsale, Cristina Aquilina, Maria Speciale, Andrea Rizzuto, Enrica Antonia Martino, Dario Leotta, Antonio Giovanni Solimando, Roberto Ria, Massimo Gentile, Sergio Siragusa, Cirino Botta

Multiple myeloma (MM) is a hematologic malignancy characterized by abnormal plasma cell proliferation in the bone marrow. Recent advancements in anti-CD38 monoclonal antibody therapies, such as daratumumab and isatuximab, have significantly improved MM patient survival. However, the lack of predictive factors of response to these therapies remains a challenge. Notably, anti-CD38 antibodies can interfere with laboratory tests, complicating response assessment. We conducted a retrospective study to evaluate the association between the appearance of positive IgGk (therapeutic antibody) on immunofixation/immunosubtraction (IF) and clinical parameters in 87 non-IgGk MM patients treated with anti-CD38 therapy. Positive IgGk IF was observed in 42 patients after a median of three treatment courses. Patients with positive IgGk IF had higher rates of complete/very good partial responses (p = 0.03) and improved progression-free survival (median not reached vs. 21.83 months, p < 0.01). High BMI (p = 0.03), higher hemoglobin (p = 0.02), lower CRP (p = 0.04), and lower monoclonal protein levels (p = 0.03) were associated with positive IgGk IF. Our findings suggest that monitoring therapeutic antibody appearance on IF may predict and optimize anti-CD38 therapy in MM. Potential explanations include the impact of patient factors (e.g. BMI) on drug pharmacokinetics, the relationship between antibody levels and immune response, and the influence of tumor biology. Further research is needed to elucidate the underlying mechanisms and clinical utility of this biomarker. Nonetheless, our results highlight the importance of considering therapeutic antibody detection when interpreting laboratory tests and managing MM patients receiving anti-CD38 therapies.

多发性骨髓瘤(MM)是一种以骨髓浆细胞异常增殖为特征的血液系统恶性肿瘤。抗CD38单克隆抗体疗法(如达拉单抗和伊沙妥昔单抗)的最新进展大大改善了多发性骨髓瘤患者的生存率。然而,缺乏对这些疗法反应的预测因素仍是一项挑战。值得注意的是,抗CD38抗体会干扰实验室检测,使反应评估变得复杂。我们进行了一项回顾性研究,以评估87例接受抗CD38治疗的非IgGk MM患者免疫固定/免疫吸附(IF)阳性IgGk(治疗抗体)的出现与临床参数之间的关联。经过三个疗程的中位治疗后,有 42 例患者观察到 IgGk IF 阳性。IgGk IF阳性患者的完全/非常好部分反应率较高(p = 0.03),无进展生存期也有所改善(中位未达 21.83 个月,p = 0.05)。
{"title":"Measurable therapeutic antibody in serum as potential predictive factor of response to anti-CD38 therapy in non-IgG-k myeloma patients.","authors":"Emilia Gigliotta, Federica Plano, Giusy Corsale, Anna Maria Corsale, Cristina Aquilina, Maria Speciale, Andrea Rizzuto, Enrica Antonia Martino, Dario Leotta, Antonio Giovanni Solimando, Roberto Ria, Massimo Gentile, Sergio Siragusa, Cirino Botta","doi":"10.1186/s40164-024-00547-x","DOIUrl":"10.1186/s40164-024-00547-x","url":null,"abstract":"<p><p>Multiple myeloma (MM) is a hematologic malignancy characterized by abnormal plasma cell proliferation in the bone marrow. Recent advancements in anti-CD38 monoclonal antibody therapies, such as daratumumab and isatuximab, have significantly improved MM patient survival. However, the lack of predictive factors of response to these therapies remains a challenge. Notably, anti-CD38 antibodies can interfere with laboratory tests, complicating response assessment. We conducted a retrospective study to evaluate the association between the appearance of positive IgGk (therapeutic antibody) on immunofixation/immunosubtraction (IF) and clinical parameters in 87 non-IgGk MM patients treated with anti-CD38 therapy. Positive IgGk IF was observed in 42 patients after a median of three treatment courses. Patients with positive IgGk IF had higher rates of complete/very good partial responses (p = 0.03) and improved progression-free survival (median not reached vs. 21.83 months, p < 0.01). High BMI (p = 0.03), higher hemoglobin (p = 0.02), lower CRP (p = 0.04), and lower monoclonal protein levels (p = 0.03) were associated with positive IgGk IF. Our findings suggest that monitoring therapeutic antibody appearance on IF may predict and optimize anti-CD38 therapy in MM. Potential explanations include the impact of patient factors (e.g. BMI) on drug pharmacokinetics, the relationship between antibody levels and immune response, and the influence of tumor biology. Further research is needed to elucidate the underlying mechanisms and clinical utility of this biomarker. Nonetheless, our results highlight the importance of considering therapeutic antibody detection when interpreting laboratory tests and managing MM patients receiving anti-CD38 therapies.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302264/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141897174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The next frontier in immunotherapy: potential and challenges of CAR-macrophages. 免疫疗法的下一个前沿:CAR-巨噬细胞的潜力与挑战。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00549-9
Jing Li, Ping Chen, Wenxue Ma

Chimeric antigen receptor macrophage (CAR-MΦ) represents a significant advancement in immunotherapy, especially for treating solid tumors where traditional CAR-T therapies face limitations. CAR-MΦ offers a promising approach to target and eradicate tumor cells by utilizing macrophages' phagocytic and antigen-presenting abilities. However, challenges such as the complex tumor microenvironment (TME), variability in antigen expression, and immune suppression limit their efficacy. This review addresses these issues, exploring mechanisms of CAR-MΦ action, optimal construct designs, and interactions within the TME. It also delves into the ex vivo manufacturing challenges of CAR-MΦ, discussing autologous and allogeneic sources and the importance of stringent quality control. The potential synergies of integrating CAR-MΦ with existing cancer therapies like checkpoint inhibitors and conventional chemotherapeutics are examined to highlight possible enhanced treatment outcomes. Furthermore, regulatory pathways for CAR-MΦ therapies are scrutinized alongside established protocols for CAR-T cells, identifying unique considerations essential for clinical trials and market approval. Proposed safety monitoring frameworks aim to manage potential adverse events, such as cytokine release syndrome, crucial for patient safety. Consolidating current research and clinical insights, this review seeks to refine CAR-MΦ therapeutic applications, overcome barriers, and suggest future research directions to transition CAR-MΦ therapies from experimental platforms to standard cancer care options.

嵌合抗原受体巨噬细胞(CAR-MΦ)是免疫疗法的一大进步,尤其是在治疗实体瘤方面,传统的CAR-T疗法面临着种种限制。通过利用巨噬细胞的吞噬和抗原递呈能力,CAR-MΦ为靶向和消灭肿瘤细胞提供了一种前景广阔的方法。然而,复杂的肿瘤微环境(TME)、抗原表达的可变性以及免疫抑制等挑战限制了它们的疗效。本综述针对这些问题,探讨了CAR-MΦ的作用机制、最佳构建设计以及TME内的相互作用。它还深入探讨了 CAR-MΦ 的体外制造挑战,讨论了自体和异体来源以及严格质量控制的重要性。研究还探讨了将 CAR-MΦ 与现有癌症疗法(如检查点抑制剂和传统化疗药物)相结合的潜在协同作用,以突出可能增强的治疗效果。此外,CAR-MΦ疗法的监管途径与CAR-T细胞的既定方案一起进行了仔细研究,确定了临床试验和市场批准所必需的独特考虑因素。拟议的安全监测框架旨在管理潜在的不良事件,如细胞因子释放综合征,这对患者安全至关重要。本综述综合了当前的研究和临床见解,旨在完善CAR-MΦ疗法的应用,克服障碍,并提出未来的研究方向,将CAR-MΦ疗法从实验平台过渡到标准癌症治疗方案。
{"title":"The next frontier in immunotherapy: potential and challenges of CAR-macrophages.","authors":"Jing Li, Ping Chen, Wenxue Ma","doi":"10.1186/s40164-024-00549-9","DOIUrl":"10.1186/s40164-024-00549-9","url":null,"abstract":"<p><p>Chimeric antigen receptor macrophage (CAR-MΦ) represents a significant advancement in immunotherapy, especially for treating solid tumors where traditional CAR-T therapies face limitations. CAR-MΦ offers a promising approach to target and eradicate tumor cells by utilizing macrophages' phagocytic and antigen-presenting abilities. However, challenges such as the complex tumor microenvironment (TME), variability in antigen expression, and immune suppression limit their efficacy. This review addresses these issues, exploring mechanisms of CAR-MΦ action, optimal construct designs, and interactions within the TME. It also delves into the ex vivo manufacturing challenges of CAR-MΦ, discussing autologous and allogeneic sources and the importance of stringent quality control. The potential synergies of integrating CAR-MΦ with existing cancer therapies like checkpoint inhibitors and conventional chemotherapeutics are examined to highlight possible enhanced treatment outcomes. Furthermore, regulatory pathways for CAR-MΦ therapies are scrutinized alongside established protocols for CAR-T cells, identifying unique considerations essential for clinical trials and market approval. Proposed safety monitoring frameworks aim to manage potential adverse events, such as cytokine release syndrome, crucial for patient safety. Consolidating current research and clinical insights, this review seeks to refine CAR-MΦ therapeutic applications, overcome barriers, and suggest future research directions to transition CAR-MΦ therapies from experimental platforms to standard cancer care options.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302330/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141893282","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Overexpression of ESYT3 improves radioimmune responses through activating cGAS-STING pathway in lung adenocarcinoma. 过表达 ESYT3 可通过激活 cGAS-STING 通路改善肺腺癌的放射免疫反应。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00546-y
Zan Luo, Ying Li, Bin Xu, Tenghua Yu, Mingming Luo, PeiMeng You, Xing Niu, Junyu Li

Background: Radiotherapy can modulate systemic antitumor immunity, while immune status in the tumor microenvironment also influences the efficacy of radiotherapy, but relevant molecular mechanisms are poorly understood in lung adenocarcinoma (LUAD).

Methods: In this study, we innovatively proposed a radiotherapy response classification for LUAD, and discovered ESYT3 served as a tumor suppressor and radioimmune response sensitizer. ESYT3 expression was measured both in radioresistant and radiosensitive LUAD tissues and cells. The influence of ESYT3 on radiotherapy sensitivity and resistance was then investigated. Interaction between ESYT3 and STING was evaluated through multiple immunofluorescent staining and coimmunoprecipitation, and downstream molecules were further analyzed. In vivo models were constructed to assess the combination treatment efficacy of ESYT3 overexpression with radiotherapy.

Results: We found that radioresistant subtype presented immunosuppressive state and activation of DNA damage repair pathways than radiosensitive subtype. ESYT3 expression was remarkably attenuated both in radioresistant LUAD tissues and cells. Clinically, low ESYT3 expression was linked with radioresistance. Overexpression of ESYT3 enabled to alleviate radioresistance, and sensitize LUAD cells to DNA damage induced by irradiation. Mechanically, ESYT3 directly interacted with STING, and activated cGAS-STING signaling, subsequently increasing the generation of type I IFNs as well as downstream chemokines CCL5 and CXCL10, thus improving radioimmune responses. The combination treatment of ESYT3 overexpression with radiotherapy had a synergistic anticancer effect in vitro and in vivo.

Conclusions: In summary, low ESYT3 expression confers resistance to radiotherapy in LUAD, and its overexpression can improve radioimmune responses through activating cGAS-STING-dependent pathway, thus providing an alternative combination therapeutic strategy for LUAD patients.

背景:放疗可调节全身抗肿瘤免疫,而肿瘤微环境中的免疫状态也会影响放疗的疗效,但肺腺癌(LUAD)的相关分子机制尚不清楚:本研究创新性地提出了肺腺癌放疗反应分级,发现ESYT3是肿瘤抑制因子和放射免疫反应敏感因子。ESYT3在抗放射和对放射敏感的LUAD组织和细胞中均有表达。随后研究了ESYT3对放疗敏感性和耐药性的影响。通过多重免疫荧光染色和共沉淀评估了ESYT3和STING之间的相互作用,并进一步分析了下游分子。我们构建了体内模型,以评估ESYT3过表达与放疗的联合治疗效果:结果:与放疗敏感亚型相比,耐放疗亚型呈现免疫抑制状态,DNA损伤修复通路被激活。ESYT3 在耐放疗 LUAD 组织和细胞中的表达均显著降低。在临床上,ESYT3的低表达与放射抗性有关。ESYT3的过表达能缓解放射抗性,并使LUAD细胞对辐照诱导的DNA损伤敏感。在机制上,ESYT3与STING直接相互作用,激活了cGAS-STING信号转导,随后增加了I型IFNs以及下游趋化因子CCL5和CXCL10的生成,从而改善了放射免疫反应。ESYT3过表达与放疗联合治疗在体外和体内具有协同抗癌作用:综上所述,ESYT3的低表达会导致LUAD患者对放疗产生耐药性,而ESYT3的过表达可通过激活cGAS-STING依赖性通路改善放射免疫反应,从而为LUAD患者提供了另一种联合治疗策略。
{"title":"Overexpression of ESYT3 improves radioimmune responses through activating cGAS-STING pathway in lung adenocarcinoma.","authors":"Zan Luo, Ying Li, Bin Xu, Tenghua Yu, Mingming Luo, PeiMeng You, Xing Niu, Junyu Li","doi":"10.1186/s40164-024-00546-y","DOIUrl":"10.1186/s40164-024-00546-y","url":null,"abstract":"<p><strong>Background: </strong>Radiotherapy can modulate systemic antitumor immunity, while immune status in the tumor microenvironment also influences the efficacy of radiotherapy, but relevant molecular mechanisms are poorly understood in lung adenocarcinoma (LUAD).</p><p><strong>Methods: </strong>In this study, we innovatively proposed a radiotherapy response classification for LUAD, and discovered ESYT3 served as a tumor suppressor and radioimmune response sensitizer. ESYT3 expression was measured both in radioresistant and radiosensitive LUAD tissues and cells. The influence of ESYT3 on radiotherapy sensitivity and resistance was then investigated. Interaction between ESYT3 and STING was evaluated through multiple immunofluorescent staining and coimmunoprecipitation, and downstream molecules were further analyzed. In vivo models were constructed to assess the combination treatment efficacy of ESYT3 overexpression with radiotherapy.</p><p><strong>Results: </strong>We found that radioresistant subtype presented immunosuppressive state and activation of DNA damage repair pathways than radiosensitive subtype. ESYT3 expression was remarkably attenuated both in radioresistant LUAD tissues and cells. Clinically, low ESYT3 expression was linked with radioresistance. Overexpression of ESYT3 enabled to alleviate radioresistance, and sensitize LUAD cells to DNA damage induced by irradiation. Mechanically, ESYT3 directly interacted with STING, and activated cGAS-STING signaling, subsequently increasing the generation of type I IFNs as well as downstream chemokines CCL5 and CXCL10, thus improving radioimmune responses. The combination treatment of ESYT3 overexpression with radiotherapy had a synergistic anticancer effect in vitro and in vivo.</p><p><strong>Conclusions: </strong>In summary, low ESYT3 expression confers resistance to radiotherapy in LUAD, and its overexpression can improve radioimmune responses through activating cGAS-STING-dependent pathway, thus providing an alternative combination therapeutic strategy for LUAD patients.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302107/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141893279","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of CDK4 as prognostic biomarker in Soft Tissue Sarcoma and synergistic effect of its inhibition in dedifferentiated liposarcoma sequential treatment. CDK4作为软组织肉瘤预后生物标志物的作用以及抑制CDK4在去分化脂肪肉瘤序贯治疗中的协同效应
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00540-4
Silvia Vanni, Giacomo Miserocchi, Graziana Gallo, Valentina Fausti, Sofia Gabellone, Chiara Liverani, Chiara Spadazzi, Claudia Cocchi, Chiara Calabrese, Giovanni De Luca, Massimo Bassi, Manlio Gessaroli, Nicola Tomasetti, Angelo Campobassi, Federica Pieri, Giorgio Ercolani, Davide Cavaliere, Lorena Gurrieri, Nada Riva, Federica Recine, Toni Ibrahim, Laura Mercatali, Robin Jones, Alessandro De Vita

Soft tissue sarcomas represent an heterogeneous group of rare mesenchymal tumors comprising 1% of all solid malignancies. Among them, liposarcoma is one of the most common histotypes with atypical lipomatous tumor/well differentiated liposarcoma and dedifferentiated liposarcoma (ALT/WDLPS and DDLPS) as the major sub-entities. The unavailability of predictive, prognostic and druggable biomarkers makes the management of these lesions challenging. In recent years CDK4 and its inhibitors have emerged as potential agents for these lesions especially for ALT/WDLPS and DDLPS but the results are not conclusive and need to be elucidated. This study involved 21 ALT/WDLPS and DDLPS patients. Histological analyses of MDM2 and CDK4 were carried out. Moreover, a DDLPS patient-derived cancer model was established in vitro and in vivo assessing the efficacy of palbociclib in combination and sequential treatment. Finally, in silico analyses on CDK4 expression were carried out. The results showed a higher expression of CDK4 and MDM2 in DDLPS compared to ALT/WDLPS. Moreover, no correlation between MDM2 expression and CDK4 was observed. Next, in vitro analysis of CDK4 inhibitor palbociclib showed an antagonistic effect when combined to other chemotherapeutics, while it exhibited a significant synergy when administered in sequential schedule with lenvatinib. Next, in vivo analysis on DDLPS xenotransplanted embryos assessing the efficacy and safety profile of the in vitro tested schedules confirmed the observed data. This proof-of-concept study sheds light on the natural history of ALT/WDLPS and DDLPS and provides the rationale for the clinical applicability of sequential treatment with palbociclib in the management of DDLPS.

软组织肉瘤是一类异质性的罕见间质肿瘤,占所有实体恶性肿瘤的 1%。其中,脂肪肉瘤是最常见的组织类型之一,非典型脂肪瘤/分化良好的脂肪肉瘤和去分化脂肪肉瘤(ALT/WDLPS 和 DDLPS)是主要的亚实体。由于没有可预测、可预后和可用药的生物标志物,因此这些病变的治疗具有挑战性。近年来,CDK4 及其抑制剂已成为治疗这些病变,尤其是 ALT/WDLPS 和 DDLPS 的潜在药物,但其结果尚无定论,需要进一步阐明。本研究涉及 21 例 ALT/WDLPS 和 DDLPS 患者。对MDM2和CDK4进行了组织学分析。此外,还在体外和体内建立了DDLPS患者衍生癌症模型,评估了palbociclib联合治疗和序贯治疗的疗效。最后,还对 CDK4 的表达进行了硅学分析。结果显示,与ALT/WDLPS相比,CDK4和MDM2在DDLPS中的表达量更高。此外,未观察到 MDM2 表达与 CDK4 之间的相关性。接下来,对CDK4抑制剂帕博西尼(palbociclib)的体外分析表明,当它与其他化疗药物联用时,会产生拮抗作用,而当它与来伐替尼按顺序联用时,则会产生显著的协同作用。接下来,对DDLPS异种移植胚胎进行体内分析,评估体外测试方案的疗效和安全性,证实了观察到的数据。这项概念验证研究揭示了ALT/WDLPS和DDLPS的自然史,并为palbociclib序贯治疗DDLPS的临床应用提供了理论依据。
{"title":"Role of CDK4 as prognostic biomarker in Soft Tissue Sarcoma and synergistic effect of its inhibition in dedifferentiated liposarcoma sequential treatment.","authors":"Silvia Vanni, Giacomo Miserocchi, Graziana Gallo, Valentina Fausti, Sofia Gabellone, Chiara Liverani, Chiara Spadazzi, Claudia Cocchi, Chiara Calabrese, Giovanni De Luca, Massimo Bassi, Manlio Gessaroli, Nicola Tomasetti, Angelo Campobassi, Federica Pieri, Giorgio Ercolani, Davide Cavaliere, Lorena Gurrieri, Nada Riva, Federica Recine, Toni Ibrahim, Laura Mercatali, Robin Jones, Alessandro De Vita","doi":"10.1186/s40164-024-00540-4","DOIUrl":"10.1186/s40164-024-00540-4","url":null,"abstract":"<p><p>Soft tissue sarcomas represent an heterogeneous group of rare mesenchymal tumors comprising 1% of all solid malignancies. Among them, liposarcoma is one of the most common histotypes with atypical lipomatous tumor/well differentiated liposarcoma and dedifferentiated liposarcoma (ALT/WDLPS and DDLPS) as the major sub-entities. The unavailability of predictive, prognostic and druggable biomarkers makes the management of these lesions challenging. In recent years CDK4 and its inhibitors have emerged as potential agents for these lesions especially for ALT/WDLPS and DDLPS but the results are not conclusive and need to be elucidated. This study involved 21 ALT/WDLPS and DDLPS patients. Histological analyses of MDM2 and CDK4 were carried out. Moreover, a DDLPS patient-derived cancer model was established in vitro and in vivo assessing the efficacy of palbociclib in combination and sequential treatment. Finally, in silico analyses on CDK4 expression were carried out. The results showed a higher expression of CDK4 and MDM2 in DDLPS compared to ALT/WDLPS. Moreover, no correlation between MDM2 expression and CDK4 was observed. Next, in vitro analysis of CDK4 inhibitor palbociclib showed an antagonistic effect when combined to other chemotherapeutics, while it exhibited a significant synergy when administered in sequential schedule with lenvatinib. Next, in vivo analysis on DDLPS xenotransplanted embryos assessing the efficacy and safety profile of the in vitro tested schedules confirmed the observed data. This proof-of-concept study sheds light on the natural history of ALT/WDLPS and DDLPS and provides the rationale for the clinical applicability of sequential treatment with palbociclib in the management of DDLPS.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11299298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141893280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CD105+CAF-derived exosomes CircAMPK1 promotes pancreatic cancer progression by activating autophagy. CD105+CAF衍生的外泌体CircAMPK1通过激活自噬促进胰腺癌的进展。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00533-3
Zhiwei He, Xiushen Li, Shiyu Chen, Kun Cai, Xiaowu Li, Hui Liu

Previous studies have shown that the heterogeneity of tumor-associated fibroblasts (CAFs) in the tumor microenvironment may play a critical role in tumorigenesis; however, the biological function of CAFs in pancreatic cancer is still controversial. In this study, we found that CD105-positive (CD105+) CAF-derived exosomes significantly promoted the proliferative and invasive metastatic abilities of pancreatic cancer cells. Furthermore, RNA-seq and qRT‒PCR experiments revealed circAMPK1 as a key molecule in exosomes from CD105+ CAFs that mediates the malignant progression of pancreatic cancer. Furthermore, we demonstrated that circAMPK1 encodes a novel protein (AMPK1-360aa) in pancreatic cancer cells. This protein competes with AMPK1 to bind to the ubiquitination ligase NEDD4, which inhibits AMPK1 protein degradation and ubiquitination and thereby increases AMPK1 levels. Finally, we demonstrated that AMPK1-360aa induces cellular autophagy via NEDD4/AMPK1 to promote the proliferation and invasion of pancreatic cancer cells. In summary, circAMPK1 in CD105+ CAF-derived exosomes may mediate pancreatic cancer cell proliferation and invasive metastasis by inducing autophagy in target cells. Moreover, circAMPK1 may competitively bind to ubiquitinating enzymes through the encoded protein AMPK1-360aa, which in turn inhibits the ubiquitination-mediated degradation of AMPK1 and contributes to the upregulation of AMPK1 expression, thus inducing cellular autophagy to mediate the malignant progression of pancreatic cancer.

以往的研究表明,肿瘤微环境中肿瘤相关成纤维细胞(CAFs)的异质性可能在肿瘤发生中起着关键作用;然而,CAFs在胰腺癌中的生物学功能仍存在争议。本研究发现,CD105 阳性(CD105+)的 CAF 衍生外泌体能显著促进胰腺癌细胞的增殖和侵袭转移能力。此外,RNA-seq和qRT-PCR实验发现circAMPK1是CD105+ CAFs外泌体中的一个关键分子,它介导了胰腺癌的恶性进展。此外,我们还证明 circAMPK1 在胰腺癌细胞中编码一种新型蛋白(AMPK1-360aa)。这种蛋白与 AMPK1 竞争,与泛素化连接酶 NEDD4 结合,抑制 AMPK1 蛋白的降解和泛素化,从而提高 AMPK1 的水平。最后,我们证明了 AMPK1-360aa 通过 NEDD4/AMPK1 诱导细胞自噬,从而促进胰腺癌细胞的增殖和侵袭。综上所述,CD105+ CAF衍生外泌体中的circAMPK1可能通过诱导靶细胞自噬而介导胰腺癌细胞增殖和侵袭转移。此外,circAMPK1可能通过编码蛋白AMPK1-360aa与泛素化酶竞争性结合,进而抑制泛素化介导的AMPK1降解,促使AMPK1表达上调,从而诱导细胞自噬,介导胰腺癌的恶性进展。
{"title":"CD105+CAF-derived exosomes CircAMPK1 promotes pancreatic cancer progression by activating autophagy.","authors":"Zhiwei He, Xiushen Li, Shiyu Chen, Kun Cai, Xiaowu Li, Hui Liu","doi":"10.1186/s40164-024-00533-3","DOIUrl":"10.1186/s40164-024-00533-3","url":null,"abstract":"<p><p>Previous studies have shown that the heterogeneity of tumor-associated fibroblasts (CAFs) in the tumor microenvironment may play a critical role in tumorigenesis; however, the biological function of CAFs in pancreatic cancer is still controversial. In this study, we found that CD105-positive (CD105<sup>+</sup>) CAF-derived exosomes significantly promoted the proliferative and invasive metastatic abilities of pancreatic cancer cells. Furthermore, RNA-seq and qRT‒PCR experiments revealed circAMPK1 as a key molecule in exosomes from CD105<sup>+</sup> CAFs that mediates the malignant progression of pancreatic cancer. Furthermore, we demonstrated that circAMPK1 encodes a novel protein (AMPK1-360aa) in pancreatic cancer cells. This protein competes with AMPK1 to bind to the ubiquitination ligase NEDD4, which inhibits AMPK1 protein degradation and ubiquitination and thereby increases AMPK1 levels. Finally, we demonstrated that AMPK1-360aa induces cellular autophagy via NEDD4/AMPK1 to promote the proliferation and invasion of pancreatic cancer cells. In summary, circAMPK1 in CD105<sup>+</sup> CAF-derived exosomes may mediate pancreatic cancer cell proliferation and invasive metastasis by inducing autophagy in target cells. Moreover, circAMPK1 may competitively bind to ubiquitinating enzymes through the encoded protein AMPK1-360aa, which in turn inhibits the ubiquitination-mediated degradation of AMPK1 and contributes to the upregulation of AMPK1 expression, thus inducing cellular autophagy to mediate the malignant progression of pancreatic cancer.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11301837/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141893277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Small cell lung cancer: emerging subtypes, signaling pathways, and therapeutic vulnerabilities. 小细胞肺癌:新出现的亚型、信号通路和治疗弱点。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00548-w
Jing Zhang, Xiaoping Zeng, Qiji Guo, Zhenxin Sheng, Yan Chen, Shiyue Wan, Lele Zhang, Peng Zhang

Small cell lung cancer (SCLC) is a recalcitrant cancer characterized by early metastasis, rapid tumor growth and poor prognosis. In recent decades, the epidemiology, initiation and mutation characteristics of SCLC, as well as abnormal signaling pathways contributing to its progression, have been widely studied. Despite extensive investigation, fewer drugs have been approved for SCLC. Recent advancements in multi-omics studies have revealed diverse classifications of SCLC that are featured by distinct characteristics and therapeutic vulnerabilities. With the accumulation of SCLC samples, different subtypes of SCLC and specific treatments for these subtypes were further explored. The identification of different molecular subtypes has opened up novel avenues for the treatment of SCLC; however, the inconsistent and uncertain classification of SCLC has hindered the translation from basic research to clinical applications. Therefore, a comprehensives review is essential to conclude these emerging subtypes and related drugs targeting specific therapeutic vulnerabilities within abnormal signaling pathways. In this current review, we summarized the epidemiology, risk factors, mutation characteristics of and classification, related molecular pathways and treatments for SCLC. We hope that this review will facilitate the translation of molecular subtyping of SCLC from theory to clinical application.

小细胞肺癌(SCLC)是一种顽固性癌症,其特点是转移早、肿瘤生长快、预后差。近几十年来,人们对小细胞肺癌的流行病学、发病和突变特征以及导致其进展的异常信号通路进行了广泛研究。尽管进行了广泛的研究,但获准用于治疗 SCLC 的药物较少。多组学研究的最新进展揭示了SCLC的多种分类,这些分类具有不同的特征和治疗弱点。随着 SCLC 样本的积累,人们进一步探索了 SCLC 的不同亚型以及针对这些亚型的特定治疗方法。不同分子亚型的确定为SCLC的治疗开辟了新途径;然而,SCLC分类的不一致和不确定性阻碍了基础研究向临床应用的转化。因此,全面综述对于总结这些新出现的亚型和针对异常信号通路中特定治疗漏洞的相关药物至关重要。在本综述中,我们总结了SCLC的流行病学、危险因素、突变特征和分类、相关分子通路和治疗方法。我们希望这篇综述能促进SCLC分子亚型从理论到临床应用的转化。
{"title":"Small cell lung cancer: emerging subtypes, signaling pathways, and therapeutic vulnerabilities.","authors":"Jing Zhang, Xiaoping Zeng, Qiji Guo, Zhenxin Sheng, Yan Chen, Shiyue Wan, Lele Zhang, Peng Zhang","doi":"10.1186/s40164-024-00548-w","DOIUrl":"10.1186/s40164-024-00548-w","url":null,"abstract":"<p><p>Small cell lung cancer (SCLC) is a recalcitrant cancer characterized by early metastasis, rapid tumor growth and poor prognosis. In recent decades, the epidemiology, initiation and mutation characteristics of SCLC, as well as abnormal signaling pathways contributing to its progression, have been widely studied. Despite extensive investigation, fewer drugs have been approved for SCLC. Recent advancements in multi-omics studies have revealed diverse classifications of SCLC that are featured by distinct characteristics and therapeutic vulnerabilities. With the accumulation of SCLC samples, different subtypes of SCLC and specific treatments for these subtypes were further explored. The identification of different molecular subtypes has opened up novel avenues for the treatment of SCLC; however, the inconsistent and uncertain classification of SCLC has hindered the translation from basic research to clinical applications. Therefore, a comprehensives review is essential to conclude these emerging subtypes and related drugs targeting specific therapeutic vulnerabilities within abnormal signaling pathways. In this current review, we summarized the epidemiology, risk factors, mutation characteristics of and classification, related molecular pathways and treatments for SCLC. We hope that this review will facilitate the translation of molecular subtyping of SCLC from theory to clinical application.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11301971/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141893281","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cell membrane patches transfer CAR molecules from a cellular depot to conventional T cells for constructing innovative fused-CAR-T cells without necessitating genetic modification. 细胞膜贴片可将细胞储库中的 CAR 分子转移到传统 T 细胞中,从而构建出创新的融合 CAR-T 细胞,而无需进行基因修饰。
IF 9.4 1区 医学 Q1 HEMATOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00545-z
Jing Hu, Luyi Zhong, Yiqiu Wang, Shiyi Hu, Lijiaqi Zhang, Qingchang Tian

Chimeric antigen receptor (CAR) serves as the foundational element of CAR-T cells. Exogenous CAR molecules can exert functional effects on allogeneic T cells, leading to their activation and subsequent functional alterations. Here we show a new method based on this biological principle: the transfer of CAR molecules from exogenous cells to the membrane of receptor T cells. This process facilitates receptor T cell to recognize target antigens and induces their activation. These patches imbued normal T cells with enhanced tumor targeting capabilities and activated their inherent killing functions. This method's efficacy introduces an approach for constructing non-genetically manipulated CAR-T cells and holds potential for application to other immune cells.

嵌合抗原受体(CAR)是 CAR-T 细胞的基本要素。外源性 CAR 分子可以对异体 T 细胞产生功能效应,导致其活化和随后的功能改变。在此,我们展示了一种基于这一生物学原理的新方法:将 CAR 分子从外源细胞转移到受体 T 细胞膜上。这一过程有助于受体 T 细胞识别目标抗原并诱导其活化。这些贴片赋予正常 T 细胞更强的肿瘤靶向能力,并激活其固有的杀伤功能。这种方法的有效性为构建非基因操纵的 CAR-T 细胞提供了一种方法,并有望应用于其他免疫细胞。
{"title":"Cell membrane patches transfer CAR molecules from a cellular depot to conventional T cells for constructing innovative fused-CAR-T cells without necessitating genetic modification.","authors":"Jing Hu, Luyi Zhong, Yiqiu Wang, Shiyi Hu, Lijiaqi Zhang, Qingchang Tian","doi":"10.1186/s40164-024-00545-z","DOIUrl":"10.1186/s40164-024-00545-z","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) serves as the foundational element of CAR-T cells. Exogenous CAR molecules can exert functional effects on allogeneic T cells, leading to their activation and subsequent functional alterations. Here we show a new method based on this biological principle: the transfer of CAR molecules from exogenous cells to the membrane of receptor T cells. This process facilitates receptor T cell to recognize target antigens and induces their activation. These patches imbued normal T cells with enhanced tumor targeting capabilities and activated their inherent killing functions. This method's efficacy introduces an approach for constructing non-genetically manipulated CAR-T cells and holds potential for application to other immune cells.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302086/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141893278","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","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
Experimental Hematology & 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