Experimental Hematology & Oncology最新文献

筛选
英文 中文
Efficacy of NKG2D CAR-T cells with IL-15/IL-15Rα signaling for treating Epstein-Barr virus-associated lymphoproliferative disorder. 具有IL-15/IL-15Rα信号的NKG2D CAR-T细胞治疗Epstein-Barr病毒相关淋巴组织增生性疾病的疗效。
IF 9.4 1区 医学
Experimental Hematology & Oncology Pub Date : 2024-08-19 DOI: 10.1186/s40164-024-00553-z
Qiusui Mai, Bailin He, Shikai Deng, Qing Zeng, Yanwen Xu, Cong Wang, Yunyi Pang, Sheng Zhang, Jinfeng Li, Jinfeng Zeng, Liqin Huang, Yongshui Fu, Chengyao Li, Tingting Li, Xiaojun Xu, Ling Zhang
{"title":"Efficacy of NKG2D CAR-T cells with IL-15/IL-15Rα signaling for treating Epstein-Barr virus-associated lymphoproliferative disorder.","authors":"Qiusui Mai, Bailin He, Shikai Deng, Qing Zeng, Yanwen Xu, Cong Wang, Yunyi Pang, Sheng Zhang, Jinfeng Li, Jinfeng Zeng, Liqin Huang, Yongshui Fu, Chengyao Li, Tingting Li, Xiaojun Xu, Ling Zhang","doi":"10.1186/s40164-024-00553-z","DOIUrl":"10.1186/s40164-024-00553-z","url":null,"abstract":"<p><p>Epstein-Barr virus (EBV) related post-transplant lymphoproliferative disorder (EBV-PTLD) is a life-threatening complication after hematopoietic stem cell transplantation (HSCT) or solid organ transplantation (SOT), for which no standard therapeutic means have been developed. Significant increase expression of natural killer group 2 member D ligands (NKG2DLs) was observed on B-lymphoblastoid cells of EBV-PTLD, indicating NKG2DLs as potential therapeutic targets for treatment of EBV-PTLD. In this study, the recombinant constructs of NKG2D CAR and IL-15/IL-15Rα-NKG2D CAR were generated with a retroviral vector and then transduced to human T cells to produce NKG2D CAR-T and IL-15/IL-15Rα-NKG2D CAR-T cells, respectively. B-lymphoblastoid cell lines (B-LCLs) and the xenografted mouse models were established to evaluate the efficacy of these CAR-T cells. IL-15/IL-15Rα-NKG2D CAR-T cells exhibited superior proliferation and antigen-specific cytotoxic effect compared to NKG2D CAR-T, as IL-15/IL-15Rα signaling promoted the expansion of less differentiated central memory T cells (T<sub>CM</sub>) and increased expression of CD107a and IFN-γ. Moreover, EBV DNA load was dramatically reduced, and 80% B-LCL cells were eliminated by IL-15/IL-15Rα-NKG2D CAR-T cells after co-culturing. In-vivo study confirmed that IL-15/IL-15Rα-NKG2D CAR-T cell therapy significantly enhanced antiviral efficacy in mice, as the serum load of EBV after IL-15/IL-15Rα-NKG2D CAR-T cell infusion was 1500 times lower than the untreated control (P < 0.001). The enhanced efficacy of IL-15/IL-15Rα-NKG2D CAR T cells was probably due to the IL-15/IL-15Rα signaling improved homing and persistence of NKG2D CAR-T cells in vivo, and increased the production of IFN-γ, Perforin, and Granulysin. In conclusion, NKG2D CAR-T cells co-expressing IL-15/IL-15Rα promoted the central memory CAR T cell proliferation and improved the homing and persistence of CAR T cells in vivo, resulting in enhanced anti-tumor and anti-viral effects in treating EBV-PTLD.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":"13 1","pages":"85"},"PeriodicalIF":9.4,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334566/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142003963","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
Enhanced depletion of MLL-fusion proteins in acute leukemia: potential for improved therapeutic outcomes. 加强急性白血病中 MLL 融合蛋白的清除:改善治疗效果的潜力。
IF 9.4 1区 医学
Experimental Hematology & Oncology Pub Date : 2024-08-16 DOI: 10.1186/s40164-024-00556-w
Noelia Che, Sandra Cantilena, Remi Looi-Somoye, Danesh Sundar, Kent Fung, Jasper de Boer, Owen Williams
{"title":"Enhanced depletion of MLL-fusion proteins in acute leukemia: potential for improved therapeutic outcomes.","authors":"Noelia Che, Sandra Cantilena, Remi Looi-Somoye, Danesh Sundar, Kent Fung, Jasper de Boer, Owen Williams","doi":"10.1186/s40164-024-00556-w","DOIUrl":"10.1186/s40164-024-00556-w","url":null,"abstract":"<p><p>Rearrangements of the MLL (KMT2A) locus are associated with aggressive leukaemia of both myeloid and lymphoid lineages, that present profound therapeutic challenges in pediatric and adult patient populations. MLL-fusion genes resulting from these rearrangements function as driving oncogenes and have been the focus of research aimed at understanding mechanisms underlying their leukemogenic activity and revealing novel therapeutic opportunities. Inspired by the paradigm of depleting the PML-RARA fusion protein in acute promyelocytic leukemia using all-trans retinoic acid and arsenic trioxide, we conducted a screen to identify FDA-approved drugs capable of depleting MLL-fusion protein expression in leukemia cells. Previously, we reported potent anti-leukemia effects of disulfiram (DSF), identified through this screen. In the present study, we demonstrate that another hit compound, niclosamide (NSM), is also able to deplete MLL-fusion proteins derived from a range of different MLL-fusion genes in both acute myeloid (AML) and acute lymphoid (ALL) leukemias. Loss of MLL-fusion protein appeared to result from inhibition of global protein translation by NSM. Importantly, combination of DSF with NSM enhanced MLL-fusion protein depletion. This led to more profound inhibition of downstream transcriptional leukemogenic programs regulated by MLL-fusion proteins and more effective killing of both MLL-rearranged AML and ALL cells. In contrast, DSF/NSM drug combination had little impact on normal hematopoietic progenitor cell differentiation. This study demonstrates that two FDA-approved drugs with excellent safety profiles can be combined to increase the efficacy of MLL-fusion protein depletion and elimination of MLL-rearranged leukaemia.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":"13 1","pages":"84"},"PeriodicalIF":9.4,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11328480/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141995616","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
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
Experimental Hematology & Oncology 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
{"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":"13 1","pages":"83"},"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
Experimental Hematology & Oncology 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
{"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":"13 1","pages":"81"},"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
Experimental Hematology & Oncology Pub Date : 2024-08-06 DOI: 10.1186/s40164-024-00543-1
Siwei Zheng, Wenwen Wang, Lesang Shen, Yao Yao, Wenjie Xia, Chao Ni
{"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":"13 1","pages":"80"},"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
Experimental Hematology & Oncology 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
{"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":"13 1","pages":"82"},"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
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
Experimental Hematology & Oncology 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
{"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":"13 1","pages":"77"},"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
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
Experimental Hematology & Oncology Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00549-9
Jing Li, Ping Chen, Wenxue Ma
{"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":"13 1","pages":"76"},"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
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
Experimental Hematology & Oncology 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
{"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":"13 1","pages":"74"},"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
Experimental Hematology & Oncology Pub Date : 2024-08-05 DOI: 10.1186/s40164-024-00533-3
Zhiwei He, Xiushen Li, Shiyu Chen, Kun Cai, Xiaowu Li, Hui Liu
{"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":"13 1","pages":"79"},"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信