Cancer lettersPub Date : 2024-11-01DOI: 10.1016/j.canlet.2024.217320
{"title":"Telaglenastat as an alternative to cisplatin as a radiosensitizer in the treatment of head and neck squamous cell carcinoma","authors":"","doi":"10.1016/j.canlet.2024.217320","DOIUrl":"10.1016/j.canlet.2024.217320","url":null,"abstract":"<div><div>The efficacy of radiation treatment (RT) of head and neck squamous cell carcinoma (HNSCC) is limited by radioresistance and the toxicity of FDA approved radiosensitizers. In extension to our previous research where we demonstrated that telaglenastat (CB839) increased efficacy of RT in <em>in vitro</em> and <em>in vivo</em> HNSCC models, here, we examine the radiosensitizing effects of telaglenastat in comparison to cisplatin's, as cisplatin is currently the standard of care for concurrent therapy. Combination of telaglenastat with RT reduced tumor volume in a HNSCC patient derived xenograft mouse model. The efficacy of telaglenastat with RT in reducing cell survival and increasing apoptosis was similar if not greater than that of cisplatin with RT in Cal27 and HN5 HNSCC cells. The addition of telaglenastat increased reactive oxygen species and reduced the antioxidant glutathione in both Cal27 and HN5 cells. Reverse Phase Protein Array analyses revealed alterations in cell death and DNA damage response proteins. This study provides the scientific underpinnings for the use of telaglenastat as a radiosensitizer in the treatment of HNSCC either as an alternative to cisplatin or in cisplatin-ineligible patients.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142567266","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Engraftment of a Surrogate Antigen onto Tumor Cell Surface via pHLIP Peptide to Universally Target CAR-T Cell Therapy to Solid Tumors.","authors":"Yan-Ting Zhang, Xinping Fu, Jane Jing Ting Lim, Shaun Xiaoliu Zhang","doi":"10.1016/j.canlet.2024.217319","DOIUrl":"https://doi.org/10.1016/j.canlet.2024.217319","url":null,"abstract":"<p><p>CAR-T cells and monoclonal antibodies (mAbs) are immunotherapeutics that have shown efficacies against certain malignancies. However, their broad application is hindered by the scarcity of tumor-associated antigens on tumor cell surfaces. Previous investigations unveiled the unique capacity of pH-low insertion peptide (pHLIP) to anchor to plasma membranes under acidic conditions. Considering that an acidic tumor microenvironment is a hallmark of solid tumors, we engineered a novel peptide, Myc-pHLIP, by tethering a surrogate epitope tag, the c-Myc-tag, to pHLIP. We evaluated the efficiency of Myc-pHLIP in inserting the artificial c-Myc-tag onto the plasma membrane of malignant cells and determined if this engraftment could convert it into a therapeutic target for CAR-T cells or mAbs. Our in vitro experiments demonstrated that incubating Myc-pHLIP with tumor cells in acidic media triggered significant killing by either Myc-targeted CAR-T cells (Myc-CAR-T), or by an anti-Myc mAb in the presence of NK cells. In vivo studies demonstrated substantial antitumor effects with sequential administration of Myc-pHLIP followed by either Myc-CAR-T or Myc-mAb. These findings establish that Myc-pHLIP has the potential to act as a universal surrogate tumor antigen capable of directing CAR-T cells or mAbs to treat any solid tumors by concurrently targeting both malignant and stromal cells.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142567254","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2024-11-01DOI: 10.1016/j.canlet.2024.217317
Guillermo Suay, Juan-Carlos Garcia-Cañaveras, Francisco Aparisi, José Garcia, Oscar Juan-Vidal, Agustín Lahoz
{"title":"Immune checkpoint inhibitors as first-line treatment for brain metastases in stage IV NSCLC patients without driver mutations.","authors":"Guillermo Suay, Juan-Carlos Garcia-Cañaveras, Francisco Aparisi, José Garcia, Oscar Juan-Vidal, Agustín Lahoz","doi":"10.1016/j.canlet.2024.217317","DOIUrl":"10.1016/j.canlet.2024.217317","url":null,"abstract":"<p><p>Immune checkpoint inhibitors (ICI) therapy with or without chemotherapy has been established as the first-line treatment for patients with non-oncogene addicted advanced Non-Small Cell Lung Cancer (NSCLC). Yet some clinical settings, such as the treatment sequence in patients with brain metastases, have barely been evidenced. Although ICIs cannot directly cross the blood-brain barrier (BBB), evidence suggests that BBB damage could allow ICIs into the central nervous system, or that they can have an indirect effect on the tumor immune microenvironment (TIME) and cause an anti-tumor response. Pivotal phase III trials have included a highly selected population but offer few data on these patients. Here we first review how ICIs can indirectly shape the brain metastases microenvironment through different mechanisms, and some possible causes of ICIs resistance. We also analyze the evidence reported in pivotal phase III trials and phase II trials focused on NSCLC brain metastases for first-line treatment, and the evidence for upfront or delayed local brain therapy. Finally, we discuss the best evidence-based approach to treat NSCLC patients with brain metastases and propose future research.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142567258","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2024-10-31DOI: 10.1016/j.canlet.2024.217310
Gayathri K Guruvayurappan, Tina Frankenbach-Désor, Markus Laubach, Alexander Klein, Michael von Bergwelt-Baildon, Monica Cusan, Attila Aszodi, Boris M Holzapfel, Wolfgang Böcker, Susanne Mayer-Wagner
{"title":"Clinical challenges in prostate cancer management: Metastatic bone-tropism and the role of circulating tumor cells.","authors":"Gayathri K Guruvayurappan, Tina Frankenbach-Désor, Markus Laubach, Alexander Klein, Michael von Bergwelt-Baildon, Monica Cusan, Attila Aszodi, Boris M Holzapfel, Wolfgang Böcker, Susanne Mayer-Wagner","doi":"10.1016/j.canlet.2024.217310","DOIUrl":"10.1016/j.canlet.2024.217310","url":null,"abstract":"<p><p>Prostate cancer (PCa) metastasis is one of the leading causes of cancer-related mortality in men worldwide, primarily due to its tendency to metastasize, with bones of axial skeleton being the favored target-site. PCa bone-metastasis (PCa-BM) presents significant clinical challenges, especially by the weakening of bone architecture, majorly due to the formation of osteoblastic lesions, leading to severe bone fractures. Another complication is that the disease predominantly affects elderly men. Further exploration is required to understand how the circulating tumor cells (CTCs) adapt to varying microenvironments and other biomechanical stresses encountered during the sequential steps in metastasis, finally resulting in colonization specifically in the bone niche, in PCa-BM. Deciphering how CTCs encounter and adapt to different biochemical, biomechanical and microenvironmental factors may improve the prospects of PCa diagnosis, development of novel therapeutics and prognosis. Moreover, the knowledge developed is expected to have broader implications for cancer research, paving the way for better therapeutic strategies and targeted therapies in the realm of metastatic cancer progression across different types of cancers. Our review begins with analyzing the challenges in PCa diagnosis, treatment and management, and delves into the formation and dynamics of CTCs, highlighting their role in PCa metastasis and bone-tropism. We further explore the pivotal role of individual factors in dictating the predisposition of tumors to metastasize to specific secondary sites, such as the noteworthy tendency of PCa bone-metastasis. Finally, we highlight the unresolved questions and potential avenues for further exploration.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142564083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Mobocertinib antagonizes multidrug resistance in ABCB1- and ABCG2-overexpressing cancer cells: in vitro and in vivo studies.","authors":"Xing-Duo Dong, Meng Zhang, Qiu-Xu Teng, Zi-Ning Lei, Chao-Yun Cai, Jing-Quan Wang, Zhuo-Xun Wu, Yuqi Yang, Xiang Chen, Huiqin Guo, Zhe-Sheng Chen","doi":"10.1016/j.canlet.2024.217309","DOIUrl":"https://doi.org/10.1016/j.canlet.2024.217309","url":null,"abstract":"<p><p>Overexpression of ATP-binding cassette (ABC) transporters, particularly ABCB1 and ABCG2, strongly correlates with multidrug resistance (MDR), rendering cancer chemotherapy ineffective. Exploration and identification of novel inhibitors targeting ABCB1 and ABCG2 are necessary to overcome the related MDR. Mobocertinib is an approved EGFR/HER2 inhibitor for non-small cell lung cancer (NSCLC) with EGFR exon 20 insertion mutations. This study demonstrates that mobocertinib can potentially reverse ABCB1- and ABCG2-mediated MDR. Our findings indicate a strong interaction between mobocertinib and these two proteins, supported by its high binding affinity with ABCB1 and ABCG2 models. Through inhibiting the drug efflux function of ABCB1 and ABCG2, mobocertinib facilitates substrate drugs accumulation, thereby re-sensitizing substrate drugs in drug-resistant cancer cells. Additionally, mobocertinib inhibited the ATPase activity of ABCB1 and ABCG2 without changing the expression levels or subcellular localization. In the tumor-bearing mouse model, mobocertinib boosted the antitumor effect of paclitaxel and topotecan, resulting in tumor regression. In summary, our study uncovers a novel potential for repurposing mobocertinib as a dual inhibitor of ABCB1 and ABCG2, and suggests the combination of mobocertinib with substrate drugs as a strategy to counteract MDR.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142557250","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2024-10-28DOI: 10.1016/j.canlet.2024.217308
{"title":"Role of semaphorins, neuropilins and plexins in cancer progression","authors":"","doi":"10.1016/j.canlet.2024.217308","DOIUrl":"10.1016/j.canlet.2024.217308","url":null,"abstract":"<div><div>Progress in understanding nervous system-cancer interconnections has emphasized the functional role of semaphorins (SEMAs) and their receptors, neuropilins (NRPs) and plexins (PLXNs), in cancer progression. SEMAs are a conserved and extensive family of broadly expressed soluble and membrane-associated proteins that were first described as regulators of axon guidance and neural and vascular development. However, recent advances have shown that they can have a dual role in cancer progression, acting either as tumor promoters or suppressors. SEMAs effects result from their interaction with specific co-receptors/receptors NRPs/PLXNs, that have also been described to play a role in cancer progression. They can influence both cancer cells and tumor microenvironment components modulating various aspects of tumorigenesis such as oncogenesis, tumor growth, invasion and metastatic spread or treatment resistance. In this review we focus on the role of these axon guidance signals and their receptors and co-receptors in various aspects of cancer. Furthermore, we also highlight their potential application as novel approaches for cancer treatment in the future.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142567261","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2024-10-23DOI: 10.1016/j.canlet.2024.217307
{"title":"Tumor-associated macrophages confer resistance to chemotherapy (Trifluridine/Tipiracil) in digestive cancers by overexpressing thymidine phosphorylase","authors":"","doi":"10.1016/j.canlet.2024.217307","DOIUrl":"10.1016/j.canlet.2024.217307","url":null,"abstract":"<div><div>Pyrimidine analogs are part of the first-line chemotherapy regimen for gastrointestinal cancers. Trifluridine combined with tipiracil, a specific thymidine phosphorylase inhibitor, in TAS-102 has recently emerged as a potential alternative in the face of primary or secondary chemoresistance to 5-fluorouracil. Despite its promise, we report that macrophage-specific overexpression of thymidine phosphorylase results in macrophage-induced chemoresistance to TAS-102 that is insensitive to tipiracil inhibition. Furthermore, we illustrate the human-specific nature of this mechanism, as mouse macrophages do not express substantial levels of thymidine phosphorylase, which constrains the applicability of mouse models. To study the importance of macrophages in chemoresistance to trifluridine, we developed a humanized mouse model with tumor-implanted human macrophages and demonstrated their important role in treatment resistance to pyrimidine analogs. Additionally, our findings revealed that macrophages represent a significant source of thymidine phosphorylase expression, comprising over 40 % of the expressing cells, in human colorectal cancer, thereby contributing to chemoresistance.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495689","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2024-10-18DOI: 10.1016/j.canlet.2024.217302
{"title":"Histone deacetylase upregulation of neuropilin-1 in osteosarcoma is essential for pulmonary metastasis","authors":"","doi":"10.1016/j.canlet.2024.217302","DOIUrl":"10.1016/j.canlet.2024.217302","url":null,"abstract":"<div><div>The lungs represent the most common site of metastasis for osteosarcoma (OS). Despite our advances in developing targeted therapies for treating solid malignancies, broad acting chemotherapies remain the first line treatment for OS. In assaying the efficacy of approved therapeutics for non-OS malignancies, we previously identified the histone deacetylase 1 and 2 (HDAC1 and 2) inhibitor, romidepsin, as effective for the treatment of established lung metastatic OS. Yet, romidepsin has noted toxicities in humans and so here we aimed to define the primary mechanisms through which HDAC1/2 mediate OS progression to identify more selective druggable targets/pathways. Microarray and proteomics analyses of romidepsin treated OS cells revealed a significant suppression of neuropilin-1 (NRP1), a known regulator of cancer cell migration and invasion. Silencing of <em>NRP1</em> significantly reduced OS proliferation, migration, invasion and adhesion <em>in vitro</em>. More strikingly, <em>in vivo</em>, reduced NRP1 expression significantly mitigated the lung metastatic potential of OS in two independent models (K7M2 and SAOS-LM7). Mechanistically, our data point to NRP1 mediating this effect via the down regulation of migration machinery, namely SRC, FAK and ROCK1 expression/activity, that is in part, related to NRP1 interaction with integrin beta 1 (ITGB1). In summary, our data indicate that romidepsin down regulation of NRP1 significantly mitigates the ability of OS cells to seed the lung and establish metastases, and that targeting NRP1 or its effectors with selective inhibitors may be a viable means with which to prevent this deadly aspect of the disease.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142458716","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}
Cancer lettersPub Date : 2024-10-18DOI: 10.1016/j.canlet.2024.217304
{"title":"The pharmacogenomic and immune landscape of snoRNAs in human cancers","authors":"","doi":"10.1016/j.canlet.2024.217304","DOIUrl":"10.1016/j.canlet.2024.217304","url":null,"abstract":"<div><div>Small nucleolar RNAs (snoRNAs) are a class of non-coding RNAs primarily known for their role in the chemical modification of other RNAs. Recent studies suggested that snoRNAs may play a broader role in anti-cancer treatments such as targeted therapies and immunotherapies. Despite these insights, the comprehensive landscape of snoRNA associations with drug response and immunotherapy outcomes remains unexplored. In this study, we identified 79,448 and 75,185 associations between snoRNAs and drug response using data from VAEN and CancerRxTissue, respectively. Additionally, we discovered 29,199 associations between snoRNAs and immune checkpoint genes and 47,194 associations between snoRNAs and immune cell infiltrations. Sixteen snoRNAs were significantly correlated with immunotherapy objective response rate (ORR), and 92 snoRNAs showed significantly differential expression between cancers with high and low ORR. Furthermore, we identified 17 snoRNAs with significantly differential expression between cancer types with high and low immune-related adverse event (irAE) reporting odds ratio (ROR). Several snoRNAs, such as SNORD92, and SNORD83B, may represent promising biomarkers or therapeutic targets that needs further investigation. To facilitate further research, we developed a user-friendly portal, Pharmacogenomic and Immune Landscape of SnoRNA (PISNO, <span><span>https://hanlaboratory.com/PISNO/</span><svg><path></path></svg></span>), enabling researchers to visualize, browse, and download multi-dimensional data. This study highlights the potential of snoRNAs as biomarkers or therapeutic targets, paving the way for more effective and personalized anti-cancer treatments.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142458718","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2024-10-17DOI: 10.1016/j.canlet.2024.217305
{"title":"CD94 deficiency or blockade unleashes the anti-tumor immunity in mice and humanized murine models","authors":"","doi":"10.1016/j.canlet.2024.217305","DOIUrl":"10.1016/j.canlet.2024.217305","url":null,"abstract":"<div><div>NKG2 family members have emerged as promising targets in tumor immunotherapy. CD94 can dimerize with both inhibitory and activating NKG2 proteins, while the overall effect and value of targeting CD94 on anti-tumor immunity are unclear. Here, it is shown that the expression of CD94 is upregulated on tumor-infiltrating natural killer (NK) cells and CD8<sup>+</sup> T cells, and is related to their exhausted characteristics. Tumor-bearing CD94 knockout (CD94-KO) mice exhibit delayed tumor growth, decreased lung metastases, and prolonged survival. Single cell RNA-seq reveals a remodeled tumor microenvironment in CD94-KO mice, with a reduction in immunosuppressive cells and an increase in anti-tumor immune cells. Moreover, NK cells and CD8<sup>+</sup> T cells become proliferative and strongly tumoricidal in CD94-KO mice, thus contributing to the tumor inhibition effect of CD94 deficiency. Treatment with a humanized anti-CD94 blocking antibody (h15C10) alone, in tumor-bearing humanized mouse, delays tumor progression, and improves the therapeutic efficacy of PD-L1 blockade through combination therapy. Our study indicates that CD94 may work as a candidate target in checkpoint immunotherapy.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142458714","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}