首页 > 最新文献

Journal of Experimental & Clinical Cancer Research最新文献

英文 中文
Inhibition of the chemokine receptors CXCR1 and CXCR2 synergizes with docetaxel for effective tumor control and remodeling of the immune microenvironment of HPV-negative head and neck cancer models.
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-05 DOI: 10.1186/s13046-024-03240-3
Lucas A Horn, Hanne Lind, Kristen Fousek, Haiyan Qin, Nika Rajabian, Shantel Angstadt, Nicole Hsiao-Sanchez, Miriam M Medina-Enriquez, Marcus D Kelly, Clint T Allen, Sarah M Hammoudeh, Roberto Weigert, Dean Y Maeda, John A Zebala, Claudia Palena

Background: Relapsed head and neck squamous cell carcinoma (HNSCC) unrelated to HPV infection carries a poor prognosis. Novel approaches are needed to improve the clinical outcome and prolong survival in this patient population which has poor long-term responses to immune checkpoint blockade. This study evaluated the chemokine receptors CXCR1 and CXCR2 as potential novel targets for the treatment of HPV-negative HNSCC.

Methods: Expression of IL-8, CXCR1, and CXCR2 was investigated in HNSCC tissues and human cell line models. Inhibition of CXCR1/2 with the clinical stage, small molecule inhibitor, SX-682, was evaluated in vitro and in vivo using human xenografts and murine models of HNSCC, both as a monotherapy and in combination with the taxane chemotherapy, docetaxel.

Results: High levels of IL-8, CXCR1, and CXCR2 expression were observed in HPV-negative compared to HPV-positive HNSCC tumors or cell lines. Treatment of HPV-negative HNSCC cell lines in vitro with SX-682 sensitized the tumor cells to the cytotoxic activity of docetaxel. In vivo, treatment of HNSCC xenograft models with the combination of SX-682 plus docetaxel led to strong anti-tumor control resulting in tumor cures. This phenomenon was associated with an increase of microRNA-200c and a decreased expression of its target, tubulin beta-3, a protein involved in resistance to microtubule-targeting chemotherapies. In vivo treatment of a murine syngeneic model of HNSCC with SX-682 plus docetaxel led to potent anti-tumor efficacy through a simultaneous decrease in suppressive CXCR2+ polymorphonuclear, myeloid-derived suppressor cells and an increase in cytotoxic CD8+ T cells in the combination therapy treated tumors compared to controls.

Conclusions: This study reports, for the first time, mechanistic findings through which the combination of CXCR1/2 inhibition and docetaxel chemotherapy exhibits synergy in models of HPV-negative HNSCC. These findings provide rationale for the use of this novel combination approach to treat HPV-negative HNSCC patients and for future combination studies of CXCR1/2 inhibition, docetaxel, and immune-based therapies.

{"title":"Inhibition of the chemokine receptors CXCR1 and CXCR2 synergizes with docetaxel for effective tumor control and remodeling of the immune microenvironment of HPV-negative head and neck cancer models.","authors":"Lucas A Horn, Hanne Lind, Kristen Fousek, Haiyan Qin, Nika Rajabian, Shantel Angstadt, Nicole Hsiao-Sanchez, Miriam M Medina-Enriquez, Marcus D Kelly, Clint T Allen, Sarah M Hammoudeh, Roberto Weigert, Dean Y Maeda, John A Zebala, Claudia Palena","doi":"10.1186/s13046-024-03240-3","DOIUrl":"10.1186/s13046-024-03240-3","url":null,"abstract":"<p><strong>Background: </strong>Relapsed head and neck squamous cell carcinoma (HNSCC) unrelated to HPV infection carries a poor prognosis. Novel approaches are needed to improve the clinical outcome and prolong survival in this patient population which has poor long-term responses to immune checkpoint blockade. This study evaluated the chemokine receptors CXCR1 and CXCR2 as potential novel targets for the treatment of HPV-negative HNSCC.</p><p><strong>Methods: </strong>Expression of IL-8, CXCR1, and CXCR2 was investigated in HNSCC tissues and human cell line models. Inhibition of CXCR1/2 with the clinical stage, small molecule inhibitor, SX-682, was evaluated in vitro and in vivo using human xenografts and murine models of HNSCC, both as a monotherapy and in combination with the taxane chemotherapy, docetaxel.</p><p><strong>Results: </strong>High levels of IL-8, CXCR1, and CXCR2 expression were observed in HPV-negative compared to HPV-positive HNSCC tumors or cell lines. Treatment of HPV-negative HNSCC cell lines in vitro with SX-682 sensitized the tumor cells to the cytotoxic activity of docetaxel. In vivo, treatment of HNSCC xenograft models with the combination of SX-682 plus docetaxel led to strong anti-tumor control resulting in tumor cures. This phenomenon was associated with an increase of microRNA-200c and a decreased expression of its target, tubulin beta-3, a protein involved in resistance to microtubule-targeting chemotherapies. In vivo treatment of a murine syngeneic model of HNSCC with SX-682 plus docetaxel led to potent anti-tumor efficacy through a simultaneous decrease in suppressive CXCR2<sup>+</sup> polymorphonuclear, myeloid-derived suppressor cells and an increase in cytotoxic CD8<sup>+</sup> T cells in the combination therapy treated tumors compared to controls.</p><p><strong>Conclusions: </strong>This study reports, for the first time, mechanistic findings through which the combination of CXCR1/2 inhibition and docetaxel chemotherapy exhibits synergy in models of HPV-negative HNSCC. These findings provide rationale for the use of this novel combination approach to treat HPV-negative HNSCC patients and for future combination studies of CXCR1/2 inhibition, docetaxel, and immune-based therapies.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"318"},"PeriodicalIF":11.4,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11619435/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142787553","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
Receptor tyrosine kinase inhibition leads to regression of acral melanoma by targeting the tumor microenvironment.
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1186/s13046-024-03234-1
Eric A Smith, Rachel L Belote, Nelly M Cruz, Tarek E Moustafa, Carly A Becker, Amanda Jiang, Shukran Alizada, Anastasia Prokofyeva, Tsz Yin Chan, Tori A Seasor, Michael Balatico, Emilio Cortes-Sanchez, David H Lum, John R Hyngstrom, Hanlin Zeng, Dekker C Deacon, Allie H Grossmann, Richard M White, Thomas A Zangle, Robert L Judson-Torres

Background: Acral melanoma (AM) is an aggressive melanoma variant that arises from palmar, plantar, and nail unit melanocytes. Compared to non-acral cutaneous melanoma (CM), AM is biologically distinct, has an equal incidence across genetic ancestries, typically presents in advanced stage disease, is less responsive to therapy, and has an overall worse prognosis.

Methods: An independent analysis of published sequencing data was performed to evaluate the frequency of receptor tyrosine kinase (RTK) ligands and adapter protein gene variants and expression. To target these genetic variants, a zebrafish acral melanoma model and preclinical patient-derived xenograft (PDX) mouse models were treated with a panel of RTK inhibitors. Residual PDX tumors were evaluated for changes in proliferation, vasculature, necrosis, and ferroptosis by histology and immunohistochemistry.

Results: RTK ligands and adapter proteins are frequently amplified, translocated, and/or overexpressed in AM. Dual FGFR/VEGFR inhibitors decrease acral-analogous melanocyte proliferation and migration in zebrafish, and the potent pan-FGFR/VEGFR inhibitor, Lenvatinib, uniformly induces tumor regression in AM PDX tumors but only slows tumor growth in CM models. Unlike other multi-RTK inhibitors, Lenvatinib is not directly cytotoxic to dissociated AM PDX tumor cells and instead disrupts tumor architecture and vascular networks.

Conclusion: Considering the great difficulty in establishing AM cell culture lines, these findings suggest that AM may be more sensitive to microenvironment perturbations than CM. In conclusion, dual FGFR/VEGFR inhibition may be a viable therapeutic strategy that targets the unique biology of AM.

{"title":"Receptor tyrosine kinase inhibition leads to regression of acral melanoma by targeting the tumor microenvironment.","authors":"Eric A Smith, Rachel L Belote, Nelly M Cruz, Tarek E Moustafa, Carly A Becker, Amanda Jiang, Shukran Alizada, Anastasia Prokofyeva, Tsz Yin Chan, Tori A Seasor, Michael Balatico, Emilio Cortes-Sanchez, David H Lum, John R Hyngstrom, Hanlin Zeng, Dekker C Deacon, Allie H Grossmann, Richard M White, Thomas A Zangle, Robert L Judson-Torres","doi":"10.1186/s13046-024-03234-1","DOIUrl":"10.1186/s13046-024-03234-1","url":null,"abstract":"<p><strong>Background: </strong>Acral melanoma (AM) is an aggressive melanoma variant that arises from palmar, plantar, and nail unit melanocytes. Compared to non-acral cutaneous melanoma (CM), AM is biologically distinct, has an equal incidence across genetic ancestries, typically presents in advanced stage disease, is less responsive to therapy, and has an overall worse prognosis.</p><p><strong>Methods: </strong>An independent analysis of published sequencing data was performed to evaluate the frequency of receptor tyrosine kinase (RTK) ligands and adapter protein gene variants and expression. To target these genetic variants, a zebrafish acral melanoma model and preclinical patient-derived xenograft (PDX) mouse models were treated with a panel of RTK inhibitors. Residual PDX tumors were evaluated for changes in proliferation, vasculature, necrosis, and ferroptosis by histology and immunohistochemistry.</p><p><strong>Results: </strong>RTK ligands and adapter proteins are frequently amplified, translocated, and/or overexpressed in AM. Dual FGFR/VEGFR inhibitors decrease acral-analogous melanocyte proliferation and migration in zebrafish, and the potent pan-FGFR/VEGFR inhibitor, Lenvatinib, uniformly induces tumor regression in AM PDX tumors but only slows tumor growth in CM models. Unlike other multi-RTK inhibitors, Lenvatinib is not directly cytotoxic to dissociated AM PDX tumor cells and instead disrupts tumor architecture and vascular networks.</p><p><strong>Conclusion: </strong>Considering the great difficulty in establishing AM cell culture lines, these findings suggest that AM may be more sensitive to microenvironment perturbations than CM. In conclusion, dual FGFR/VEGFR inhibition may be a viable therapeutic strategy that targets the unique biology of AM.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"317"},"PeriodicalIF":11.4,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11613472/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142774365","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
Targeting PRMT5 through PROTAC for the treatment of triple-negative breast cancer.
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-30 DOI: 10.1186/s13046-024-03237-y
Yaxun Guo, Yuzhan Li, Zhongmei Zhou, Lei Hou, Wenjing Liu, Wenlong Ren, Dazhao Mi, Jian Sun, Xueqin Dai, Yingying Wu, Zhuo Cheng, Tingyue Wu, Qianmei Luo, Cong Tian, Fubing Li, Zhigang Yu, Yihua Chen, Ceshi Chen

Background: Triple-negative breast cancer (TNBC) is currently the most aggressive subtype of breast cancer, characterized by high heterogeneity and strong invasiveness, and currently lacks effective therapies. PRMT5, a type II protein arginine methyltransferase, is upregulated in numerous cancers, including TNBC, and plays a critical role, marked it as an attractive therapeutic target. PROTAC (Proteolysis Targeting Chimeras) is an innovative drug development technology that utilizes the ubiquitin-proteasome system (UPS) to degrade target proteins, which is characterized by higher activity, enhanced safety, lower resistance, and reduced toxicity, offering significant value for clinical translation.

Methods: This study utilizes the PROTAC technology to develop potential degraders targeting PRMT5 in vitro and in vivo.

Results: Through the design, synthesis and screening of a series of targeted compounds, we identified YZ-836P as an effective compound that exerted cytotoxic effects and reduced the protein levels of PRMT5 and its key downstream target protein KLF5 in TNBC after 48 h. Its efficacy was significantly superior to the PRMT5 PROTAC degraders that had been reported. YZ-836P induced G1 phase cell cycle arrest and significantly induced apoptosis in TNBC cells. Additionally, we demonstrated that YZ-836P promoted the ubiquitination and degradation of PRMT5 in a cereblon (CRBN)-dependent manner. Notably, YZ-836P exhibited pronounced efficacy in inhibiting the growth of TNBC patient-derived organoids and xenografts in nude mice.

Conclusions: These findings position YZ-836P as a promising candidate for advancing treatment modalities for TNBC.

Trial registration: Ethics Committee of Yunnan Cancer Hospital, KYCS2023-078. Registered 7 June 2023.

{"title":"Targeting PRMT5 through PROTAC for the treatment of triple-negative breast cancer.","authors":"Yaxun Guo, Yuzhan Li, Zhongmei Zhou, Lei Hou, Wenjing Liu, Wenlong Ren, Dazhao Mi, Jian Sun, Xueqin Dai, Yingying Wu, Zhuo Cheng, Tingyue Wu, Qianmei Luo, Cong Tian, Fubing Li, Zhigang Yu, Yihua Chen, Ceshi Chen","doi":"10.1186/s13046-024-03237-y","DOIUrl":"10.1186/s13046-024-03237-y","url":null,"abstract":"<p><strong>Background: </strong>Triple-negative breast cancer (TNBC) is currently the most aggressive subtype of breast cancer, characterized by high heterogeneity and strong invasiveness, and currently lacks effective therapies. PRMT5, a type II protein arginine methyltransferase, is upregulated in numerous cancers, including TNBC, and plays a critical role, marked it as an attractive therapeutic target. PROTAC (Proteolysis Targeting Chimeras) is an innovative drug development technology that utilizes the ubiquitin-proteasome system (UPS) to degrade target proteins, which is characterized by higher activity, enhanced safety, lower resistance, and reduced toxicity, offering significant value for clinical translation.</p><p><strong>Methods: </strong>This study utilizes the PROTAC technology to develop potential degraders targeting PRMT5 in vitro and in vivo.</p><p><strong>Results: </strong>Through the design, synthesis and screening of a series of targeted compounds, we identified YZ-836P as an effective compound that exerted cytotoxic effects and reduced the protein levels of PRMT5 and its key downstream target protein KLF5 in TNBC after 48 h. Its efficacy was significantly superior to the PRMT5 PROTAC degraders that had been reported. YZ-836P induced G1 phase cell cycle arrest and significantly induced apoptosis in TNBC cells. Additionally, we demonstrated that YZ-836P promoted the ubiquitination and degradation of PRMT5 in a cereblon (CRBN)-dependent manner. Notably, YZ-836P exhibited pronounced efficacy in inhibiting the growth of TNBC patient-derived organoids and xenografts in nude mice.</p><p><strong>Conclusions: </strong>These findings position YZ-836P as a promising candidate for advancing treatment modalities for TNBC.</p><p><strong>Trial registration: </strong>Ethics Committee of Yunnan Cancer Hospital, KYCS2023-078. Registered 7 June 2023.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"314"},"PeriodicalIF":11.4,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11607928/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142755829","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
Advances in non-small cell lung cancer mechanomedicine: deciphering the signaling networks that govern tumor-TME interactions.
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-30 DOI: 10.1186/s13046-024-03242-1
Antonios N Gargalionis, Kostas A Papavassiliou, Efthimia K Basdra, Athanasios G Papavassiliou

Cells from the tumor microenvironment (TME) interact with tumor cells in non-small cell lung cancer (NSCLC) to form a reciprocal crosstalk which influences tumor growth, proliferation, metastasis and multidrug response. This crosstalk is modulated by TME mechanical inputs, which elicit the processes of mechanosensing and mechanotransduction. Recent advances in unveiling these signaling networks establish the interdisciplinary field of mechanomedicine to exploit emerging diagnostic, predictive and therapeutic tools for more effective NSCLC treatments.

{"title":"Advances in non-small cell lung cancer mechanomedicine: deciphering the signaling networks that govern tumor-TME interactions.","authors":"Antonios N Gargalionis, Kostas A Papavassiliou, Efthimia K Basdra, Athanasios G Papavassiliou","doi":"10.1186/s13046-024-03242-1","DOIUrl":"https://doi.org/10.1186/s13046-024-03242-1","url":null,"abstract":"<p><p>Cells from the tumor microenvironment (TME) interact with tumor cells in non-small cell lung cancer (NSCLC) to form a reciprocal crosstalk which influences tumor growth, proliferation, metastasis and multidrug response. This crosstalk is modulated by TME mechanical inputs, which elicit the processes of mechanosensing and mechanotransduction. Recent advances in unveiling these signaling networks establish the interdisciplinary field of mechanomedicine to exploit emerging diagnostic, predictive and therapeutic tools for more effective NSCLC treatments.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"316"},"PeriodicalIF":11.4,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11608457/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142774354","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
Ferroptosis and the tumor microenvironment.
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-30 DOI: 10.1186/s13046-024-03235-0
Kaisa Cui, Kang Wang, Zhaohui Huang

Ferroptosis is a type of regulated cell death characterized by its non-apoptotic, iron-dependent and oxidative nature. Since its discovery in 2012, extensive research has demonstrated its pivotal roles in tumorigenesis, metastasis and cancer therapy. The tumor microenvironment (TME) is a complex ecosystem comprising cancer cells, non-cancer cells, extracellular matrix, metabolites and cytokines. Recent studies have underscored a new paradigm in which non-cancer cells in the TME, such as immune and stromal cells, also play significant roles in regulating tumor progression and therapeutic resistance typically through complicated crosstalk with cancer cells. Notably, this crosstalk in the TME were partially mediated through ferrotopsis-related mechanisms. This review provides a comprehensive and systematic summary of the current findings concerning the roles of ferroptosis in the TME and how ferroptosis-mediated TME reprogramming impacts cancer therapeutic resistance and progression. Additionally, this review outlines various ferroptosis-related therapeutic strategies aimed at targeting the TME.

{"title":"Ferroptosis and the tumor microenvironment.","authors":"Kaisa Cui, Kang Wang, Zhaohui Huang","doi":"10.1186/s13046-024-03235-0","DOIUrl":"10.1186/s13046-024-03235-0","url":null,"abstract":"<p><p>Ferroptosis is a type of regulated cell death characterized by its non-apoptotic, iron-dependent and oxidative nature. Since its discovery in 2012, extensive research has demonstrated its pivotal roles in tumorigenesis, metastasis and cancer therapy. The tumor microenvironment (TME) is a complex ecosystem comprising cancer cells, non-cancer cells, extracellular matrix, metabolites and cytokines. Recent studies have underscored a new paradigm in which non-cancer cells in the TME, such as immune and stromal cells, also play significant roles in regulating tumor progression and therapeutic resistance typically through complicated crosstalk with cancer cells. Notably, this crosstalk in the TME were partially mediated through ferrotopsis-related mechanisms. This review provides a comprehensive and systematic summary of the current findings concerning the roles of ferroptosis in the TME and how ferroptosis-mediated TME reprogramming impacts cancer therapeutic resistance and progression. Additionally, this review outlines various ferroptosis-related therapeutic strategies aimed at targeting the TME.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"315"},"PeriodicalIF":11.4,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11607824/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142755825","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
AKT kinases as therapeutic targets.
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-29 DOI: 10.1186/s13046-024-03207-4
Dalal Hassan, Craig W Menges, Joseph R Testa, Alfonso Bellacosa

AKT, or protein kinase B, is a central node of the PI3K signaling pathway that is pivotal for a range of normal cellular physiologies that also underlie several pathological conditions, including inflammatory and autoimmune diseases, overgrowth syndromes, and neoplastic transformation. These pathologies, notably cancer, arise if either the activity of AKT or its positive or negative upstream or downstream regulators or effectors goes unchecked, superimposed on by its intersection with a slew of other pathways. Targeting the PI3K/AKT pathway is, therefore, a prudent countermeasure. AKT inhibitors have been tested in many clinical trials, primarily in combination with other drugs. While some have recently garnered attention for their favorable profile, concern over resistance and off-target effects have continued to hinder their widespread adoption in the clinic, mandating a discussion on alternative modes of targeting. In this review, we discuss isoform-centric targeting that may be more effective and less toxic than traditional pan-AKT inhibitors and its significance for disease prevention and treatment, including immunotherapy. We also touch on the emerging mutant- or allele-selective covalent allosteric AKT inhibitors (CAAIs), as well as indirect, novel AKT-targeting approaches, and end with a briefing on the ongoing quest for more reliable biomarkers predicting sensitivity and response to AKT inhibitors, and their current state of affairs.

{"title":"AKT kinases as therapeutic targets.","authors":"Dalal Hassan, Craig W Menges, Joseph R Testa, Alfonso Bellacosa","doi":"10.1186/s13046-024-03207-4","DOIUrl":"10.1186/s13046-024-03207-4","url":null,"abstract":"<p><p>AKT, or protein kinase B, is a central node of the PI3K signaling pathway that is pivotal for a range of normal cellular physiologies that also underlie several pathological conditions, including inflammatory and autoimmune diseases, overgrowth syndromes, and neoplastic transformation. These pathologies, notably cancer, arise if either the activity of AKT or its positive or negative upstream or downstream regulators or effectors goes unchecked, superimposed on by its intersection with a slew of other pathways. Targeting the PI3K/AKT pathway is, therefore, a prudent countermeasure. AKT inhibitors have been tested in many clinical trials, primarily in combination with other drugs. While some have recently garnered attention for their favorable profile, concern over resistance and off-target effects have continued to hinder their widespread adoption in the clinic, mandating a discussion on alternative modes of targeting. In this review, we discuss isoform-centric targeting that may be more effective and less toxic than traditional pan-AKT inhibitors and its significance for disease prevention and treatment, including immunotherapy. We also touch on the emerging mutant- or allele-selective covalent allosteric AKT inhibitors (CAAIs), as well as indirect, novel AKT-targeting approaches, and end with a briefing on the ongoing quest for more reliable biomarkers predicting sensitivity and response to AKT inhibitors, and their current state of affairs.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"313"},"PeriodicalIF":11.4,"publicationDate":"2024-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11606119/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142755819","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
Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer.
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-28 DOI: 10.1186/s13046-024-03218-1
Ali Mussa, Nor Hayati Ismail, Mahasin Hamid, Mohammad A I Al-Hatamleh, Anthony Bragoli, Khalid Hajissa, Noor Fatmawati Mokhtar, Rohimah Mohamud, Vuk Uskoković, Rosline Hassan

Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.

{"title":"Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer.","authors":"Ali Mussa, Nor Hayati Ismail, Mahasin Hamid, Mohammad A I Al-Hatamleh, Anthony Bragoli, Khalid Hajissa, Noor Fatmawati Mokhtar, Rohimah Mohamud, Vuk Uskoković, Rosline Hassan","doi":"10.1186/s13046-024-03218-1","DOIUrl":"10.1186/s13046-024-03218-1","url":null,"abstract":"<p><p>Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"312"},"PeriodicalIF":11.4,"publicationDate":"2024-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603874/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142752203","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
Tailoring glioblastoma treatment based on longitudinal analysis of post-surgical tumor microenvironment. 根据手术后肿瘤微环境的纵向分析,定制胶质母细胞瘤的治疗方法。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-28 DOI: 10.1186/s13046-024-03231-4
Chiara Bastiancich, Emmanuel Snacel-Fazy, Samantha Fernandez, Stéphane Robert, Roberta Stacchini, Léa Plantureux, Sébastien Boissonneau, Benoit Testud, Benjamin Guillet, Franck Debarbieux, Hervé Luche, Dominique Figarella-Branger, Marie-Anne Estève, Emeline Tabouret, Aurélie Tchoghandjian

Glioblastoma (GBM), an incurable primary brain tumor, typically requires surgical intervention followed by chemoradiation; however, recurrences remain fatal. Our previous work demonstrated that a nanomedicine hydrogel (GemC12-LNC) delays recurrence when administered post-surgery. However, tumor debulking also triggers time-dependent immune reactions that promote recurrence at the resection cavity borders. We hypothesized that combining the hydrogel with an immunomodulatory drug could enhance therapeutic outcomes. A thorough characterization of the post-surgical microenvironment (SMe) is crucial to guide combinatorial approaches.In this study, we performed cellular resolution imaging, flow cytometry and spatial hyperplexed immunofluorescence imaging to characterize the SMe in a syngeneic mouse model of tumor resection. Owing to our dynamic approach, we observed transient opening of the blood-brain barrier (BBB) during the first week after surgery. BBB permeability post-surgery was also confirmed in GBM patients. In our murine model, we also observed changes in immune cell morphology and spatial location post-surgery over time in resected animals as well as the accumulation of reactive microglia and anti-inflammatory macrophages in recurrences compared to unresected tumors since the first steps of recurrence growth. Therefore we investigated whether starting a systemic treatment with the SMAC mimetic small molecule (GDC-0152) directly after surgery would be beneficial for enhancing microglial anti-tumoral activity and decreasing the number of anti-inflammatory macrophages around the GemC12-LNC hydrogel-loaded tumor cavity. The immunomodulatory effects of this drug combination was firstly shown in patient-derived tumoroids. Its efficacy was confirmed in vivo by survival analysis and correlated with reversal of the immune profile as well as delayed tumor recurrence.This comprehensive study identified critical time frames and immune cellular targets within the SMe, aiding in the rational design of combination therapies to delay recurrence onset. Our findings suggest that post-surgical systemic injection of GDC-0152 in combination with GemC12-LNC local treatment is a promising and innovative approach for managing GBM recurrence, with potential for future translation to human patient.

胶质母细胞瘤(GBM)是一种无法治愈的原发性脑肿瘤,通常需要手术治疗,然后进行化疗;然而,复发仍然是致命的。我们之前的研究表明,纳米药物水凝胶(GemC12-LNC)在手术后给药可延缓复发。然而,肿瘤剥离也会引发时间依赖性免疫反应,促进切除腔边界的复发。我们假设,将水凝胶与免疫调节药物结合可提高治疗效果。在这项研究中,我们通过细胞分辨率成像、流式细胞术和空间超复合免疫荧光成像来描述肿瘤切除术后合成小鼠模型中的微环境(SMe)。由于我们采用的是动态方法,我们观察到血脑屏障(BBB)在术后第一周短暂开放。GBM 患者手术后血脑屏障的通透性也得到了证实。在我们的小鼠模型中,我们还观察到切除动物手术后免疫细胞形态和空间位置随时间发生的变化,以及自复发生长的第一步起,与未切除肿瘤相比,复发肿瘤中反应性小胶质细胞和抗炎巨噬细胞的积累。因此,我们研究了手术后直接开始使用 SMAC 拟效小分子(GDC-0152)进行全身治疗是否有利于增强小胶质细胞的抗肿瘤活性,并减少 GemC12-LNC 水凝胶负载瘤腔周围抗炎巨噬细胞的数量。这种药物组合的免疫调节作用首次在患者来源的肿瘤组织中显示出来。这项综合研究确定了SMe的关键时间框架和免疫细胞靶点,有助于合理设计联合疗法以推迟复发。我们的研究结果表明,手术后全身注射GDC-0152与GemC12-LNC局部治疗相结合是治疗GBM复发的一种前景广阔的创新方法,未来有可能应用于人类患者。
{"title":"Tailoring glioblastoma treatment based on longitudinal analysis of post-surgical tumor microenvironment.","authors":"Chiara Bastiancich, Emmanuel Snacel-Fazy, Samantha Fernandez, Stéphane Robert, Roberta Stacchini, Léa Plantureux, Sébastien Boissonneau, Benoit Testud, Benjamin Guillet, Franck Debarbieux, Hervé Luche, Dominique Figarella-Branger, Marie-Anne Estève, Emeline Tabouret, Aurélie Tchoghandjian","doi":"10.1186/s13046-024-03231-4","DOIUrl":"10.1186/s13046-024-03231-4","url":null,"abstract":"<p><p>Glioblastoma (GBM), an incurable primary brain tumor, typically requires surgical intervention followed by chemoradiation; however, recurrences remain fatal. Our previous work demonstrated that a nanomedicine hydrogel (GemC<sub>12</sub>-LNC) delays recurrence when administered post-surgery. However, tumor debulking also triggers time-dependent immune reactions that promote recurrence at the resection cavity borders. We hypothesized that combining the hydrogel with an immunomodulatory drug could enhance therapeutic outcomes. A thorough characterization of the post-surgical microenvironment (SMe) is crucial to guide combinatorial approaches.In this study, we performed cellular resolution imaging, flow cytometry and spatial hyperplexed immunofluorescence imaging to characterize the SMe in a syngeneic mouse model of tumor resection. Owing to our dynamic approach, we observed transient opening of the blood-brain barrier (BBB) during the first week after surgery. BBB permeability post-surgery was also confirmed in GBM patients. In our murine model, we also observed changes in immune cell morphology and spatial location post-surgery over time in resected animals as well as the accumulation of reactive microglia and anti-inflammatory macrophages in recurrences compared to unresected tumors since the first steps of recurrence growth. Therefore we investigated whether starting a systemic treatment with the SMAC mimetic small molecule (GDC-0152) directly after surgery would be beneficial for enhancing microglial anti-tumoral activity and decreasing the number of anti-inflammatory macrophages around the GemC<sub>12</sub>-LNC hydrogel-loaded tumor cavity. The immunomodulatory effects of this drug combination was firstly shown in patient-derived tumoroids. Its efficacy was confirmed in vivo by survival analysis and correlated with reversal of the immune profile as well as delayed tumor recurrence.This comprehensive study identified critical time frames and immune cellular targets within the SMe, aiding in the rational design of combination therapies to delay recurrence onset. Our findings suggest that post-surgical systemic injection of GDC-0152 in combination with GemC<sub>12</sub>-LNC local treatment is a promising and innovative approach for managing GBM recurrence, with potential for future translation to human patient.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"311"},"PeriodicalIF":11.4,"publicationDate":"2024-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603899/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142741289","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
Selective metabolic regulations by p53 mutant variants in pancreatic cancer. 胰腺癌中 p53 突变变体对代谢的选择性调节。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-26 DOI: 10.1186/s13046-024-03232-3
Sabrina Caporali, Alessio Butera, Alessia Ruzza, Carlotta Zampieri, Marina Bantula', Sandra Scharsich, Anna-Katerina Ückert, Ivana Celardo, Ian U Kouzel, Luigi Leanza, Andreas Gruber, Joan Montero, Angelo D'Alessandro, Thomas Brunner, Marcel Leist, Ivano Amelio

Background: Approximately half of all human cancers harbour mutations in the p53 gene, leading to the generation of neomorphic p53 mutant proteins. These mutants can exert gain-of-function (GOF) effects, potentially promoting tumour progression. However, the clinical significance of p53 GOF mutations, as well as the selectivity of individual variants, remains controversial and unclear.

Methods: To elucidate the metabolic regulations and molecular underpinnings associated with the specific p53R270H and p53R172H mutant variants (the mouse equivalents of human p53R273H and p53R175H, respectively), we employed a comprehensive approach. This included integrating global metabolomic analysis with epigenomic and transcriptomic profiling in mouse pancreatic cancer cells. Additionally, we assessed metabolic parameters such as oxygen consumption rate and conducted analyses of proliferation and cell-cell competition to validate the biological impact of metabolic changes on pancreatic ductal adenocarcinoma (PDAC) phenotype. Our findings were further corroborated through analysis of clinical datasets from human cancer cohorts.

Results: Our investigation revealed that the p53R270H variant, but not p53R172H, sustains mitochondrial function and energy production while also influencing cellular antioxidant capacity. Conversely, p53R172H, while not affecting mitochondrial metabolism, attenuates the activation of pro-tumorigenic metabolic pathways such as the urea cycle. Thus, the two variants selectively control different metabolic pathways in pancreatic cancer cells. Mechanistically, p53R270H induces alterations in the expression of genes associated with oxidative stress and reduction in mitochondrial respiration. In contrast, p53R172H specifically impacts the expression levels of enzymes involved in the urea metabolism. However, our analysis of cell proliferation and cell competition suggested that the expression of either p53R270H or p53R172H does not influence confer any selective advantage to this cellular model in vitro. Furthermore, assessment of mitochondrial priming indicated that the p53R270H-driven mitochondrial effect does not alter cytochrome c release or the apoptotic propensity of pancreatic cancer cells.

Conclusions: Our study elucidates the mutant-specific impact of p53R270H and p53R172H on metabolism of PDAC cancer cells, highlighting the need to shift from viewing p53 mutant variants as a homogeneous group of entities to a systematic assessment of each specific p53 mutant protein. Moreover, our finding underscores the importance of further exploring the significance of p53 mutant proteins using models that more accurately reflect tumor ecology.

背景:大约一半的人类癌症都存在 p53 基因突变,导致产生新形 p53 突变蛋白。这些突变体可产生功能增益(GOF)效应,可能会促进肿瘤的进展。然而,p53 GOF 突变的临床意义以及单个变体的选择性仍存在争议且不明确:为了阐明与特定 p53R270H 和 p53R172H 突变变体(分别相当于人类 p53R273H 和 p53R175H 的小鼠变体)相关的代谢调节和分子基础,我们采用了一种综合方法。这包括将全球代谢组分析与小鼠胰腺癌细胞的表观基因组和转录组分析相结合。此外,我们还评估了氧消耗率等代谢参数,并进行了增殖和细胞间竞争分析,以验证代谢变化对胰腺导管腺癌(PDAC)表型的生物学影响。通过分析人类癌症队列的临床数据集进一步证实了我们的发现:结果:我们的研究发现,p53R270H 变体(而非 p53R172H)在维持线粒体功能和能量产生的同时,也影响了细胞的抗氧化能力。相反,p53R172H 在不影响线粒体代谢的同时,还能减弱尿素循环等促肿瘤代谢途径的激活。因此,这两种变体选择性地控制胰腺癌细胞中不同的代谢途径。从机理上讲,p53R270H 会诱导与氧化应激和线粒体呼吸减少相关的基因表达发生变化。相比之下,p53R172H 会特别影响参与尿素代谢的酶的表达水平。然而,我们对细胞增殖和细胞竞争的分析表明,p53R270H 或 p53R172H 的表达并不影响这种细胞模型在体外的选择性优势。此外,对线粒体启动的评估表明,p53R270H 驱动的线粒体效应不会改变细胞色素 c 的释放或胰腺癌细胞的凋亡倾向:我们的研究阐明了p53R270H和p53R172H突变体对PDAC癌细胞新陈代谢的影响,强调了从将p53突变体视为同类实体转变为系统评估每种特定p53突变体蛋白的必要性。此外,我们的发现还强调了利用更准确反映肿瘤生态学的模型进一步探索 p53 突变蛋白重要性的重要性。
{"title":"Selective metabolic regulations by p53 mutant variants in pancreatic cancer.","authors":"Sabrina Caporali, Alessio Butera, Alessia Ruzza, Carlotta Zampieri, Marina Bantula', Sandra Scharsich, Anna-Katerina Ückert, Ivana Celardo, Ian U Kouzel, Luigi Leanza, Andreas Gruber, Joan Montero, Angelo D'Alessandro, Thomas Brunner, Marcel Leist, Ivano Amelio","doi":"10.1186/s13046-024-03232-3","DOIUrl":"10.1186/s13046-024-03232-3","url":null,"abstract":"<p><strong>Background: </strong>Approximately half of all human cancers harbour mutations in the p53 gene, leading to the generation of neomorphic p53 mutant proteins. These mutants can exert gain-of-function (GOF) effects, potentially promoting tumour progression. However, the clinical significance of p53 GOF mutations, as well as the selectivity of individual variants, remains controversial and unclear.</p><p><strong>Methods: </strong>To elucidate the metabolic regulations and molecular underpinnings associated with the specific p53<sup>R270H</sup> and p53<sup>R172H</sup> mutant variants (the mouse equivalents of human p53<sup>R273H</sup> and p53<sup>R175H</sup>, respectively), we employed a comprehensive approach. This included integrating global metabolomic analysis with epigenomic and transcriptomic profiling in mouse pancreatic cancer cells. Additionally, we assessed metabolic parameters such as oxygen consumption rate and conducted analyses of proliferation and cell-cell competition to validate the biological impact of metabolic changes on pancreatic ductal adenocarcinoma (PDAC) phenotype. Our findings were further corroborated through analysis of clinical datasets from human cancer cohorts.</p><p><strong>Results: </strong>Our investigation revealed that the p53<sup>R270H</sup> variant, but not p53<sup>R172H</sup>, sustains mitochondrial function and energy production while also influencing cellular antioxidant capacity. Conversely, p53<sup>R172H</sup>, while not affecting mitochondrial metabolism, attenuates the activation of pro-tumorigenic metabolic pathways such as the urea cycle. Thus, the two variants selectively control different metabolic pathways in pancreatic cancer cells. Mechanistically, p53<sup>R270H</sup> induces alterations in the expression of genes associated with oxidative stress and reduction in mitochondrial respiration. In contrast, p53<sup>R172H</sup> specifically impacts the expression levels of enzymes involved in the urea metabolism. However, our analysis of cell proliferation and cell competition suggested that the expression of either p53<sup>R270H</sup> or p53<sup>R172H</sup> does not influence confer any selective advantage to this cellular model in vitro. Furthermore, assessment of mitochondrial priming indicated that the p53<sup>R270H</sup>-driven mitochondrial effect does not alter cytochrome c release or the apoptotic propensity of pancreatic cancer cells.</p><p><strong>Conclusions: </strong>Our study elucidates the mutant-specific impact of p53<sup>R270H</sup> and p53<sup>R172H</sup> on metabolism of PDAC cancer cells, highlighting the need to shift from viewing p53 mutant variants as a homogeneous group of entities to a systematic assessment of each specific p53 mutant protein. Moreover, our finding underscores the importance of further exploring the significance of p53 mutant proteins using models that more accurately reflect tumor ecology.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"310"},"PeriodicalIF":11.4,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11590503/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142717521","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
Mechanism of efficacy of trabectedin against myxoid liposarcoma entails detachment of the FUS-DDIT3 transcription factor from its DNA binding sites. 曲贝替丁(Trabectedin)对类肌脂肪肉瘤的疗效机制是使 FUS-DDIT3 转录因子脱离其 DNA 结合位点。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-26 DOI: 10.1186/s13046-024-03228-z
Ilaria Craparotta, Laura Mannarino, Riccardo Zadro, Sara Ballabio, Sergio Marchini, Giulio Pavesi, Marta Russo, Salvatore Lorenzo Renne, Marina Meroni, Marianna Ponzo, Ezia Bello, Roberta Sanfilippo, Paolo G Casali, Maurizio D'Incalci, Roberta Frapolli

Background: The marine drug trabectedin has shown unusual effectiveness in the treatment of myxoid liposarcoma (MLPS), a liposarcoma characterized by the expression of the FUS-DDIT3 chimera. Trabectedin elicits a significant transcriptional response in MLPS resulting in cellular depletion and reactivation of adipogenesis. However, the role of the chimeric protein in the mechanism of action of the drug is not entirely understood.

Methods: FUS-DDIT3-specific binding sites were assessed through Chromatin Immunoprecipitation Sequencing (ChIP-Seq). Trabectedin-induced effects were studied on pre-established patient-derived xenograft models of MLPS, one sensitive to (ML017) and one resistant against (ML017ET) trabectedin at different time points (24 and 72 h, 15 days). Data were integrated with RNA-Seq from the same models.

Results: Through ChIP-Seq, here we demonstrate that trabectedin inhibits the binding of FUS-DDIT3 to its target genes, restoring adipocyte differentiation in a patient-derived xenograft model of MLPS sensitive to trabectedin. In addition, complementary RNA-Seq data on the same model demonstrates a two-phase effect of trabectedin, characterized by an initial FUS-DDIT3-independent cytotoxicity, followed by a transcriptionally active pro-differentiation phase due to the long-lasting detachment of the chimera from the DNA. Interestingly, in a trabectedin-resistant MLPS model, the effect of trabectedin on FUS-DDIT3 rapidly decreased over time, and prolonged treatment was no longer able to induce any transcription or post-transcriptional modifications.

Conclusions: These findings explain the unusual mechanism underlying trabectedin's effectiveness against MLPS by pinpointing the chimera's role in inducing the differentiation block responsible for MLPS pathogenesis. Additionally, the findings hint at a potential mechanism of resistance acquired in vivo.

背景:海洋药物曲贝替丁(Trabectedin)在治疗类粘液性脂肪肉瘤(MLPS)方面显示出不同寻常的疗效,MLPS是一种以表达FUS-DDIT3嵌合体为特征的脂肪肉瘤。曲贝替丁(Trabectedin)能在 MLPS 中引起明显的转录反应,导致细胞耗竭和脂肪生成的重新激活。然而,嵌合蛋白在药物作用机制中的作用还不完全清楚:方法:通过染色质免疫沉淀测序(ChIP-Seq)评估了FUS-DDIT3特异性结合位点。在不同的时间点(24 和 72 小时,15 天),对已建立的 MLPS 患者异种移植模型(一种对 ML017 敏感,一种对 ML017ET 耐药)进行了曲贝替定诱导效应的研究。数据与来自相同模型的 RNA-Seq 数据进行了整合:结果:通过 ChIP-Seq,我们证明了曲贝替定抑制了 FUS-DDIT3 与其靶基因的结合,从而恢复了对曲贝替定敏感的 MLPS 患者来源异种移植模型的脂肪细胞分化。此外,关于同一模型的互补 RNA-Seq 数据显示了曲贝替丁(trabectedin)的两阶段效应,其特点是最初的 FUS-DDIT3 细胞毒性不依赖于 FUS-DDIT3,随后由于嵌合体与 DNA 的持久分离,转录活跃的促分化阶段随之而来。有趣的是,在曲贝替定耐药的MLPS模型中,曲贝替定对FUS-DDIT3的作用随着时间的推移迅速减弱,长期治疗不再能诱导任何转录或转录后修饰:这些发现解释了曲贝替丁(Trabectedin)对MLPS有效的不寻常机制,指出了嵌合体在诱导导致MLPS发病机制的分化阻滞中的作用。此外,这些发现还暗示了体内获得耐药性的潜在机制。
{"title":"Mechanism of efficacy of trabectedin against myxoid liposarcoma entails detachment of the FUS-DDIT3 transcription factor from its DNA binding sites.","authors":"Ilaria Craparotta, Laura Mannarino, Riccardo Zadro, Sara Ballabio, Sergio Marchini, Giulio Pavesi, Marta Russo, Salvatore Lorenzo Renne, Marina Meroni, Marianna Ponzo, Ezia Bello, Roberta Sanfilippo, Paolo G Casali, Maurizio D'Incalci, Roberta Frapolli","doi":"10.1186/s13046-024-03228-z","DOIUrl":"10.1186/s13046-024-03228-z","url":null,"abstract":"<p><strong>Background: </strong>The marine drug trabectedin has shown unusual effectiveness in the treatment of myxoid liposarcoma (MLPS), a liposarcoma characterized by the expression of the FUS-DDIT3 chimera. Trabectedin elicits a significant transcriptional response in MLPS resulting in cellular depletion and reactivation of adipogenesis. However, the role of the chimeric protein in the mechanism of action of the drug is not entirely understood.</p><p><strong>Methods: </strong>FUS-DDIT3-specific binding sites were assessed through Chromatin Immunoprecipitation Sequencing (ChIP-Seq). Trabectedin-induced effects were studied on pre-established patient-derived xenograft models of MLPS, one sensitive to (ML017) and one resistant against (ML017ET) trabectedin at different time points (24 and 72 h, 15 days). Data were integrated with RNA-Seq from the same models.</p><p><strong>Results: </strong>Through ChIP-Seq, here we demonstrate that trabectedin inhibits the binding of FUS-DDIT3 to its target genes, restoring adipocyte differentiation in a patient-derived xenograft model of MLPS sensitive to trabectedin. In addition, complementary RNA-Seq data on the same model demonstrates a two-phase effect of trabectedin, characterized by an initial FUS-DDIT3-independent cytotoxicity, followed by a transcriptionally active pro-differentiation phase due to the long-lasting detachment of the chimera from the DNA. Interestingly, in a trabectedin-resistant MLPS model, the effect of trabectedin on FUS-DDIT3 rapidly decreased over time, and prolonged treatment was no longer able to induce any transcription or post-transcriptional modifications.</p><p><strong>Conclusions: </strong>These findings explain the unusual mechanism underlying trabectedin's effectiveness against MLPS by pinpointing the chimera's role in inducing the differentiation block responsible for MLPS pathogenesis. Additionally, the findings hint at a potential mechanism of resistance acquired in vivo.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"309"},"PeriodicalIF":11.4,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11590625/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142717454","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
Journal of Experimental & Clinical Cancer Research
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