{"title":"Modified hTERT promoters-driven purine nucleoside phosphorylase-gene therapy in association with chemo- and targeted therapy in the context of ovarian cancer.","authors":"Quoc Manh Nguyen, Pierre-François Dupré, Mathieu Berchel, Marylène Lévêque, Sylvie Choblet-Thery, Frédérique d'Arbonneau, Tristan Montier","doi":"10.1038/s41417-025-00932-6","DOIUrl":"https://doi.org/10.1038/s41417-025-00932-6","url":null,"abstract":"<p><p>Ovarian cancer has the highest mortality-to-incidence ratio among gynecologic cancers worldwide. E.coli Purine Nucleoside Phosphorylase-based gene-directed enzyme prodrug therapy (PNP-GDEPT) offers a promising alternative for the treatment of solid tumors. This study proposes an original ovarian cancer treatment through the use of two recently developed modified hTERT promoters: the mutated hTERT (hTERTm) and the chimeric hTERT-CMV. Four plasmids were engineered to investigate the effects of cancer-specific PNP gene expression: pCMV-PNP, phTERT-PNP, phTERTm-PNP, and phTERT-CMV-PNP. The cationic lipid formulation BSV163/DOPE was employed to transfect PNP-coding plasmids into cisplatin-sensitive ovarian cancer cells and their resistant counterparts. hTERT-driven PNP-GDEPT selectively reduced cancer cell viability while sparing primary human fibroblasts. In addition, combining PNP-GDEPT with either cisplatin or olaparib further enhanced anticancer effects on cell viability and apoptosis. However, no combined effects were observed for the concurrent use of cisplatin and olaparib, with or without PNP-GDEPT. Our results demonstrate that targeted PNP-GDEPT has the potential to enhance the efficacy of chemotherapy and targeted therapy against ovarian cancer while minimizing side effects on healthy cells. This treatment is effective irrespective of cisplatin resistance status and warrants further investigation.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":4.8,"publicationDate":"2025-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144511604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Zhihao Tu, Yuelin Chen, Zhimi Zhang, Wanrong Meng, Ling Li
{"title":"Barriers and solutions for CAR-T therapy in solid tumors.","authors":"Zhihao Tu, Yuelin Chen, Zhimi Zhang, Wanrong Meng, Ling Li","doi":"10.1038/s41417-025-00931-7","DOIUrl":"https://doi.org/10.1038/s41417-025-00931-7","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative approach for cancer treatment, particularly in hematologic malignancies. However, barriers in the development of effective CAR-T therapies for solid tumors, including antigenic escape, tumor immunosuppressive microenvironments, severe toxicities, and limitations in preclinical models, hinder its scalability and broader clinical implementation. To overcome these barriers, strategies have been developed in recent years, such as optimizing CAR designs, enhancing CAR-T cell infiltration, neutralizing immunosuppressive cells, remodeling metabolism of CAR-T cells, eliminating antigen escape, mitigating toxicities, advancing preclinical models, and in situ programming CAR-T cells. Here, we discuss current barriers and potential strategies for CAR-T cell therapy in solid tumors. Ultimately, we present perspectives on these advanced strategies for broader clinical adoption of CAR-T cell therapy.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":4.8,"publicationDate":"2025-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144511603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The dual role of GPX4 in breast cancer: mechanisms of therapeutic resistance and potential for novel targeted therapies.","authors":"Tingyu Gu, Kun Wang, Xiao Yuan, Haowen Tang, Shouchao Wang, Zhihong Zhao","doi":"10.1038/s41417-025-00927-3","DOIUrl":"https://doi.org/10.1038/s41417-025-00927-3","url":null,"abstract":"<p><p>Glutathione peroxidase 4 (GPX4) is a key intracellular antioxidant enzyme that maintains oxidative homeostasis by catalyzing the reduction of lipid peroxides and relies on glutathione-specific inhibition of iron death. In recent years, it has been found that GPX4 exhibits significant aberrant expression in breast cancer (BC) and promotes BC development by regulating tumor cell proliferation, invasion, metastasis, and stem cell properties. More importantly, GPX4 overexpression leads to decreased sensitivity of BC to chemotherapy, radiotherapy, endocrine therapy, immune checkpoint inhibitors, and targeted therapies by inhibiting iron death, attenuating oxidative damage, and activating pro-survival signaling pathways. In this paper, we systematically review the molecular characterization of GPX4 and its cancer-promoting mechanism in BC, focusing on resolving its molecular regulatory network in multimodal therapy resistance. Based on the reversal of drug resistance and synergistic anti-tumor effects demonstrated by GPX4 inhibitors in preclinical studies, iron death induction strategies targeting GPX4 or combining with existing therapies are expected to be a new direction to overcome the bottleneck of drug resistance in BC.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":4.8,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144483226","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"UBE2N inhibition abrogates cancer chemoresistance and metastasis in lung adenocarcinoma.","authors":"Zi-Mei Peng, Jin-Yong Xiong, Feng-Yi Deng, Xing Wang, Tao Wang, Chun-Xi Yang, Yan-Ru Chen, Xiao-Jian Han, Zhen Zhang","doi":"10.1038/s41417-025-00929-1","DOIUrl":"10.1038/s41417-025-00929-1","url":null,"abstract":"<p><p>Chemoresistance and metastasis remain significant challenges in cancer therapy, resulting in treatment failures for cancer patients. Therefore, there is a need to investigate the mechanisms underlying cancer chemoresistance and metastasis, identify novel drug targets, and develop innovative antitumor medications. In this study, we have shown that the ubiquitin conjugating enzyme UBE2N plays a crucial role in promoting chemoresistance and metastasis in lung adenocarcinoma (LUAD) both in vitro and in vivo. Significantly, our study revealed that the natural compound wilforine functions as a potential inhibitor of UBE2N, effectively reversing resistance to cisplatin (CDDP) and inhibiting metastasis of LUAD both in vitro and in vivo. Our findings collectively suggest that UBE2N may have a pivotal role in the development of chemoresistance and metastasis in LUAD, providing a foundation for potential therapeutic strategies in the management of advanced cancer.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":4.8,"publicationDate":"2025-06-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144339920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hee Chan Yoo, Sangkil Lee, Joong Yull Park, Eun-Ju Lee
{"title":"AAV for ovarian cancer gene therapy.","authors":"Hee Chan Yoo, Sangkil Lee, Joong Yull Park, Eun-Ju Lee","doi":"10.1038/s41417-025-00926-4","DOIUrl":"10.1038/s41417-025-00926-4","url":null,"abstract":"<p><p>Recent advancements in ovarian cancer treatment, particularly with PARP inhibitors, have markedly enhanced the recurrence-free interval, shifting the treatment paradigm and increasing treatment success in patients with BRCA mutations or HRD (homologous recombination deficiency). However, a significant proportion of cases experience relapse, resulting in poorer long-term survival rates when compared to other female cancers, such as breast cancer. This review explores the potential of adeno-associated virus (AAV) vectors for gene therapy in ovarian cancer and examines rational gene therapy strategies by categorizing them based on target cells and target genes to determine the most effective approach for ovarian cancer treatment. Specifically, it examines strategies such as anti-angiogenesis and immune modulation, highlighting the strategy of gene supplementation to hinder ovarian cancer progression. Innovations in AAV capsid design now allow for targeted delivery, focusing on ovarian cancer stem cells (CSCs) identified by specific markers. Additionally, leveraging DNA sequencing technologies enhances the identification and incorporation of therapeutic genes into AAV vectors, promising new avenues for ovarian cancer gene therapy.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":4.8,"publicationDate":"2025-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144336321","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kamran Abbasi, Parveen Ali, Virginia Barbour, Marion Birch, Inga Blum, Peter Doherty, Andy Haines, Ira Helfand, Richard Horton, Kati Juva, Jose F Lapena, Robert Mash, Olga Mironova, Arun Mitra, Carlos Monteiro, Elena N Naumova, David Onazi, Tilman Ruff, Peush Sahni, James Tumwine, Carlos Umaña, Paul Yonga, Chris Zielinski
{"title":"Ending nuclear weapons, before they end us.","authors":"Kamran Abbasi, Parveen Ali, Virginia Barbour, Marion Birch, Inga Blum, Peter Doherty, Andy Haines, Ira Helfand, Richard Horton, Kati Juva, Jose F Lapena, Robert Mash, Olga Mironova, Arun Mitra, Carlos Monteiro, Elena N Naumova, David Onazi, Tilman Ruff, Peush Sahni, James Tumwine, Carlos Umaña, Paul Yonga, Chris Zielinski","doi":"10.1038/s41417-025-00920-w","DOIUrl":"https://doi.org/10.1038/s41417-025-00920-w","url":null,"abstract":"","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":4.8,"publicationDate":"2025-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144265316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jianfen Wu, Haijian Wu, Fengqi Zhou, Yongjie Wang, Chun Wang, Jianxiong Ji
{"title":"Blood-brain barrier alterations as an initiator of brain metastasis from lung cancer: a new take on an old story.","authors":"Jianfen Wu, Haijian Wu, Fengqi Zhou, Yongjie Wang, Chun Wang, Jianxiong Ji","doi":"10.1038/s41417-025-00916-6","DOIUrl":"https://doi.org/10.1038/s41417-025-00916-6","url":null,"abstract":"<p><p>Lung cancer is the most common primary tumor associated with brain metastases. Currently, the molecular mechanisms underlying the pathogenesis of lung cancer brain metastasis (LCBM) have not been fully elucidated and effective therapeutic strategies for treating this disease are still lacking. The blood-brain barrier (BBB) is a selectively permeable membrane and is crucial in maintaining brain homeostasis. Increasing evidence from clinical and pre-clinical studies indicated that metastatic circulating tumor cells from lung cancer can affect the cellular biology of BBB components, and destruct its integrity as well as function to penetrate the BBB to clone in the brain parenchyma, therein producing malignant secondary tumors. Therefore, in this review, we aim to discuss about the involvement of BBB alterations during the initiation of LCBM and the cellular and molecular mechanisms underlying. A better understanding of these pathophysiological processes may help open avenues to develop new therapeutic strategies to prevent or treat this notorious disease.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":4.8,"publicationDate":"2025-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144207755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer gene therapyPub Date : 2025-06-01Epub Date: 2025-04-17DOI: 10.1038/s41417-025-00904-w
Yue Qiao, Shuihao Zhu, Zhenni Liu, Natalia Kelley, Zhencang Zheng, Jonathan A Fletcher, Wen-Bin Ou
{"title":"The activated tyrosine kinase ACK1 by multiple receptor tyrosine kinases promotes proliferation and invasion of mesothelioma via regulation of PI3K/AKT/mTOR and RAF/MAPK signaling pathways.","authors":"Yue Qiao, Shuihao Zhu, Zhenni Liu, Natalia Kelley, Zhencang Zheng, Jonathan A Fletcher, Wen-Bin Ou","doi":"10.1038/s41417-025-00904-w","DOIUrl":"10.1038/s41417-025-00904-w","url":null,"abstract":"<p><p>Activation of the receptor tyrosine kinases (RTKs) EGFR, MET, and AXL has been described in subsets of mesothelioma, suggesting that tyrosine kinases (TKs) might represent therapeutic targets in this chemotherapy resistant and highly lethal cancer. In the present study, activated TKs were identified in mesothelioma cells by phosphotyrosine immunoaffinity purification and tandem mass spectrometry, and biological functions were evaluated. The results showed that non-RTK activated-CDC42 kinase 1 (ACK1) was highly expressed and activated in 8 of 9 mesothelioma cell lines and 15 of 18 mesothelioma biopsies, but not in normal mesothelial cells. This ACK1 activation was in turn driven by the collective activation of EGFR, MET, and AXL. ACK1 inactivation by either a small molecule inhibitor (AIM-100) or RNAi had anti-proliferative, anti-migration, and pro-apoptotic effects in four mesothelioma cultures due to G1 arrest and xenograft model. These responses resulted from inhibition of the PI3K/AKT/mTOR and RAF/MAPK pathways, inhibition of cyclin A and cyclin D1, and up-regulation of cell cycle checkpoints TP53, CDKN1A (p21), and CDKN1B (p27). Combination treatment with AIM-100, cisplatin (CIS), and pemetrexed (PEM) had greater impact on mesothelioma response (apoptosis, proliferation arrest, and inhibition of migration and invasion) compared to administering only one or two of these agents. The current findings identify ACK1 as a single downstream target that can be inhibited to stymie these multiple receptor tyrosine kinase (EGFR, MET, and AXL) oncogenic programs in mesothelioma, and highlight that ACK1 inhibition, potentially in combination with PEM and CIS, warrants evaluation as a therapeutic strategy in mesothelioma.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":"678-689"},"PeriodicalIF":4.8,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143966944","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer gene therapyPub Date : 2025-06-01Epub Date: 2025-04-10DOI: 10.1038/s41417-025-00901-z
Tatiana V Kudling, Dmitrii Bychkov, James H A Clubb, Santeri A Pakola, Victor Arias, Elise Jirovec, Mirte van der Heijden, Nea Ojala, Dafne C A Quixabeira, Lyna Haybout, Katriina J Jalkanen, Tuomo Alanko, Riikka Havunen, Suvi Sorsa, Claudia Kistler, Anna Kanerva, Otto Hemminki, Joao M Santos, Victor Cervera-Carrascon, Akseli Hemminki
{"title":"Single-cell profiling of peripheral blood mononuclear cells from patients treated with oncolytic adenovirus TILT-123 reveals baseline immune status as a predictor of therapy outcomes.","authors":"Tatiana V Kudling, Dmitrii Bychkov, James H A Clubb, Santeri A Pakola, Victor Arias, Elise Jirovec, Mirte van der Heijden, Nea Ojala, Dafne C A Quixabeira, Lyna Haybout, Katriina J Jalkanen, Tuomo Alanko, Riikka Havunen, Suvi Sorsa, Claudia Kistler, Anna Kanerva, Otto Hemminki, Joao M Santos, Victor Cervera-Carrascon, Akseli Hemminki","doi":"10.1038/s41417-025-00901-z","DOIUrl":"10.1038/s41417-025-00901-z","url":null,"abstract":"<p><p>Oncolytic adenovirus Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123, igrelimogene litadenorepvec) shows promise as a therapeutic agent capable of causing tumor regression and activating host immunity. A phase I clinical study TUNIMO (NCT04695327) assessed its safety as monotherapy in patients with various solid tumors. Through single-cell profiling of peripheral blood, we identified distinct immunological features distinguishing responders from non-responders. Specifically, at baseline, responders demonstrated enhanced cytotoxic markers and stronger immune cell communication networks. Moreover, higher baseline CD16+ monocytes correlated with improved survival, while elevated regulatory T cells predicted poor response. T and B cell evaluation revealed contrasting patterns: responders showed higher numbers of T cells with predicted specificity to both adenovirus and tumor antigens, while elevated total memory B cells, regardless of specificity, predicted poor survival. Several T and B cell receptor segments matched those previously reported in other viral infections, suggesting possible cross-reactive immune responses. These findings emphasize that comprehensive biomarker analysis of peripheral blood should include not only cell frequencies but also transcriptional changes and distinct patterns of cellular and humoral immunity.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":"649-661"},"PeriodicalIF":4.8,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12183079/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143972771","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}