{"title":"Oncolytic viruses: Narcissistic or altruistic arsonists?","authors":"Luke Kendall, Richard G Vile","doi":"10.1016/j.omto.2023.04.002","DOIUrl":"10.1016/j.omto.2023.04.002","url":null,"abstract":"","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"29 ","pages":"42-43"},"PeriodicalIF":5.7,"publicationDate":"2023-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/3e/08/main.PMC10165125.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9453374","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Upasana Sahu, Matthew P Mullarkey, Guangsheng Pei, Zhongming Zhao, Bangxing Hong, Balveen Kaur
{"title":"oHSV-P10 reduces glioma stem cell enrichment after oncolytic HSV therapy.","authors":"Upasana Sahu, Matthew P Mullarkey, Guangsheng Pei, Zhongming Zhao, Bangxing Hong, Balveen Kaur","doi":"10.1016/j.omto.2023.03.003","DOIUrl":"10.1016/j.omto.2023.03.003","url":null,"abstract":"<p><p>Longstanding evidence implicate glioma stem-like cells as the main drivers contributing toward glioblastoma (GBM) therapy resistance and tumor recurrence. Although oncolytic herpes simplex virus (oHSV) viral therapy is a promising biological therapy recently approved for melanoma (in the United States and Europe) and GBM (in Japan); however, the impact of this therapy on GBM stem-like cells (GSCs) is understudied. Here we show that post-oHSV virotherapy activated AKT signaling results in an enrichment of GSC signatures in glioma, which mimics the enrichment in GSC observed after radiation treatment. We also uncovered that a second-generation oncolytic virus armed with PTEN-L (oHSV-P10) decreases this by moderating IL6/JAK/STAT3 signaling. This ability was retained in the presence of radiation treatment and oHSV-P10-sensitized intracranial GBM to radiotherapy. Collectively, our findings uncover potential mechanisms to overcome GSC-mediated radiation resistance via oHSV-P10.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"29 ","pages":"30-41"},"PeriodicalIF":5.7,"publicationDate":"2023-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/c9/00/main.PMC10126842.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9364473","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiang Lv, Le Qin, Ruocong Zhao, Di Wu, Zhiping Wu, Diwei Zheng, Siyu Li, Mintao Luo, Qiting Wu, Youguo Long, Zhaoyang Tang, Yan-Lai Tang, Xuequn Luo, Yao Yao, Li-Hua Yang, Peng Li
{"title":"Disruption of CISH promotes the antitumor activity of human T cells and decreases PD-1 expression levels.","authors":"Jiang Lv, Le Qin, Ruocong Zhao, Di Wu, Zhiping Wu, Diwei Zheng, Siyu Li, Mintao Luo, Qiting Wu, Youguo Long, Zhaoyang Tang, Yan-Lai Tang, Xuequn Luo, Yao Yao, Li-Hua Yang, Peng Li","doi":"10.1016/j.omto.2022.12.003","DOIUrl":"https://doi.org/10.1016/j.omto.2022.12.003","url":null,"abstract":"<p><p>Tumor cells and the immunosuppressive tumor microenvironment suppress the antitumor activity of T cells through immune checkpoints, including the PD-L1/PD-1 axis. Cytokine-inducible SH2-containing protein (CISH), a member of the suppressor of cytokine signaling (SOCS) family, inhibits JAK-STAT and T cell receptor (TCR) signaling in T and natural killer (NK) cells. However, its role in the regulation of immune checkpoints in T cells remains unclear. In this study, we ablated CISH in T cells with CRISPR-Cas9 and found that the sensitivity of T cells to TCR and cytokine stimulation was increased. In addition, chimeric antigen receptor T cells with CISH deficiency exhibited longer survival and higher cytokine secretion and antitumor activity. Notably, PD-1 expression was decreased in activated CISH-deficient T cells <i>in vitro</i> and <i>in vivo</i>. The level of FBXO38, a ubiquitination-regulating protein that reduces PD-1 expression, was elevated in activated T cells after CISH ablation. Hence, this study reveals a mechanism by which CISH promotes PD-1 expression by suppressing the expression of FBXO38 and proposes a new strategy for augmenting the therapeutic effect of CAR-T cells by inhibiting CISH.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"28 ","pages":"46-58"},"PeriodicalIF":5.7,"publicationDate":"2023-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/11/b0/main.PMC9827364.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9100293","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Saru Basnet, Joao M Santos, Dafne C A Quixabeira, James H A Clubb, Susanna A M Grönberg-Vähä-Koskela, Victor Arias, Santeri Pakola, Tatiana V Kudling, Camilla Heiniö, Riikka Havunen, Victor Cervera-Carrascon, Suvi Sorsa, Marjukka Anttila, Anna Kanerva, Akseli Hemminki
{"title":"Oncolytic adenovirus coding for bispecific T cell engager against human MUC-1 potentiates T cell response against solid tumors.","authors":"Saru Basnet, Joao M Santos, Dafne C A Quixabeira, James H A Clubb, Susanna A M Grönberg-Vähä-Koskela, Victor Arias, Santeri Pakola, Tatiana V Kudling, Camilla Heiniö, Riikka Havunen, Victor Cervera-Carrascon, Suvi Sorsa, Marjukka Anttila, Anna Kanerva, Akseli Hemminki","doi":"10.1016/j.omto.2022.12.007","DOIUrl":"https://doi.org/10.1016/j.omto.2022.12.007","url":null,"abstract":"<p><p>Immunotherapy with bispecific T cell engagers has shown efficacy in patients with hematologic malignancies and uveal melanoma. Antitumor effects of bispecific T cell engagers in most solid tumors are limited due to their short serum half-life and insufficient tumor concentration. We designed a novel serotype 5/3 oncolytic adenovirus encoding a human mucin1 antibody and the human CD3 receptor, Ad5/3-E2F-d24-aMUC1aCD3 (TILT-321). TILT-321 is engineered to replicate only in cancer cells, leading to a high concentration of the aMUC1aCD3 molecule in the tumor microenvironment. Infection and cell viability assays were performed to determine the oncolytic potential of the novel construct. The functionality of the virus-derived aMUC1aCD3 was evaluated <i>in vitro</i>. When TILT-321 was combined with allogeneic T cells, rapid tumor cell lysis was observed. TILT-321-infected cells secreted functional aMUC1aCD3, as shown by increased T cell activity and its binding to MUC1 and CD3. <i>In vivo</i>, TILT-321 treatment led to effective antitumor efficacy mediated by increased intratumoral T cell activity in an A549 and patient-derived ovarian cancer xenograft mouse model humanized with peripheral blood mononuclear cells (PBMC). This study provides a proof of concept for an effective strategy to overcome the key limitations of recombinant bispecific T cell engager delivery for solid tumor treatment.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"28 ","pages":"59-73"},"PeriodicalIF":5.7,"publicationDate":"2023-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/a3/00/main.PMC9842968.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9178066","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Valeria Guidolin, Yupeng Li, Foster C Jacobs, Margaret L MacMillan, Peter W Villalta, Stephen S Hecht, Silvia Balbo
{"title":"Characterization and quantitation of busulfan DNA adducts in the blood of patients receiving busulfan therapy.","authors":"Valeria Guidolin, Yupeng Li, Foster C Jacobs, Margaret L MacMillan, Peter W Villalta, Stephen S Hecht, Silvia Balbo","doi":"10.1016/j.omto.2023.01.005","DOIUrl":"https://doi.org/10.1016/j.omto.2023.01.005","url":null,"abstract":"<p><p>DNA alkylating drugs have been used as cancer chemotherapy with variable outcomes. The establishment of predictive biomarkers to identify patients who will effectively respond to treatment would allow for the development of personalized therapies. As the degree of interaction of alkylating drug with DNA plays a key role in their mechanism of action, our hypothesis is that the measurement of the DNA adducts formed by alkylating drugs could be used to inform patient stratification. Beginning with busulfan, we took advantage of our DNA adductomic approach to characterize DNA adducts formed by reacting busulfan with calf-thymus DNA. Samples collected from six patients undergoing busulfan-based chemotherapy prior to allogeneic hematopoietic cell transplantation were analyzed for the presence of busulfan-derived DNA adducts. Among the 15 adducts detected <i>in vitro</i>, 12 were observed in the patient blood confirming the presence of a large profile of DNA adducts <i>in vivo</i>. Two of the detected adducts were structurally confirmed by comparison with synthetic standards and quantified in patients. These data confirm our ability to comprehensively characterize busulfan-derived DNA damage and set the stage for the development of methods to support personalized chemotherapy.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"28 ","pages":"197-210"},"PeriodicalIF":5.7,"publicationDate":"2023-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/6e/2a/main.PMC9938526.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9493813","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Mechanisms of drug resistance in breast cancer liver metastases: Dilemmas and opportunities.","authors":"Chun-Yan Yan, Meng-Lu Zhao, Ya-Nan Wei, Xi-He Zhao","doi":"10.1016/j.omto.2023.02.001","DOIUrl":"https://doi.org/10.1016/j.omto.2023.02.001","url":null,"abstract":"<p><p>Breast cancer is the leading cause of cancer-related deaths in females worldwide, and the liver is one of the most common sites of distant metastases in breast cancer patients. Patients with breast cancer liver metastases face limited treatment options, and drug resistance is highly prevalent, leading to a poor prognosis and a short survival. Liver metastases respond extremely poorly to immunotherapy and have shown resistance to treatments such as chemotherapy and targeted therapies. Therefore, to develop and to optimize treatment strategies as well as to explore potential therapeutic approaches, it is crucial to understand the mechanisms of drug resistance in breast cancer liver metastases patients. In this review, we summarize recent advances in the research of drug resistance mechanisms in breast cancer liver metastases and discuss their therapeutic potential for improving patient prognoses and outcomes.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"28 ","pages":"212-229"},"PeriodicalIF":5.7,"publicationDate":"2023-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/80/70/main.PMC9969274.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10625881","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A cancer cell membrane coated, doxorubicin and microRNA co-encapsulated nanoplatform for colorectal cancer theranostics.","authors":"Sihao Zhu, Ziyuan Li, Dongye Zheng, Yue Yu, Jing Xiang, Xiao Ma, Dongqing Xu, Jiajun Qiu, Ziyu Yang, Zhiyi Wang, Jun Li, Hongfang Sun, Weiqiang Chen, Xiangxi Meng, Yanye Lu, Qiushi Ren","doi":"10.1016/j.omto.2022.12.002","DOIUrl":"https://doi.org/10.1016/j.omto.2022.12.002","url":null,"abstract":"<p><p>Endogenous microRNAs (miRNA) in tumors are currently under exhaustive investigation as potential therapeutic agents for cancer treatment. Nevertheless, RNase degradation, inefficient and untargeted delivery, limited biological effect, and currently unclear side effects remain unsettled issues that frustrate clinical application. To address this, a versatile targeted delivery system for multiple therapeutic and diagnostic agents should be adapted for miRNA. In this study, we developed membrane-coated PLGA-b-PEG DC-chol nanoparticles (m-PPDCNPs) co-encapsulating doxorubicin (Dox) and miRNA-190-Cy7. Such a system showed low biotoxicity, high loading efficiency, and superior targeting ability. Systematic delivery of m-PPDCNPs in mouse models showed exceptionally specific tumor accumulation. Sustained release of miR-190 inhibited tumor angiogenesis, tumor growth, and migration by regulating a large group of angiogenic effectors. Moreover, m-PPDCNPs also enhanced the sensitivity of Dox by suppressing TGF-β signal in colorectal cancer cell lines and mouse models. Together, our results demonstrate a stimulating and promising m-PPDCNPs nanoplatform for colorectal cancer theranostics.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"28 ","pages":"182-196"},"PeriodicalIF":5.7,"publicationDate":"2023-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9937835/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9317499","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ying Wei, Sonia Erfani, David Schweer, Rafael de Gouvea, Javeria Qadir, Junfeng Shi, Kai Cheng, Dabao Wu, Rolf Craven, Yadi Wu, Thibault Olivier, Lauren A Baldwin, Binhua Zhou, Ying Zhou, Weidong Zhao, Burton B Yang, Frederick R Ueland, Xiuwei H Yang
{"title":"Targeting receptor tyrosine kinases in ovarian cancer: Genomic dysregulation, clinical evaluation of inhibitors, and potential for combinatorial therapies.","authors":"Ying Wei, Sonia Erfani, David Schweer, Rafael de Gouvea, Javeria Qadir, Junfeng Shi, Kai Cheng, Dabao Wu, Rolf Craven, Yadi Wu, Thibault Olivier, Lauren A Baldwin, Binhua Zhou, Ying Zhou, Weidong Zhao, Burton B Yang, Frederick R Ueland, Xiuwei H Yang","doi":"10.1016/j.omto.2023.02.006","DOIUrl":"https://doi.org/10.1016/j.omto.2023.02.006","url":null,"abstract":"<p><p>Epithelial ovarian cancer (EOC) remains one of the leading causes of cancer-related deaths among women worldwide. Receptor tyrosine kinases (RTKs) have long been sought as therapeutic targets for EOC, as they are frequently hyperactivated in primary tumors and drive disease relapse, progression, and metastasis. More recently, these oncogenic drivers have been implicated in EOC response to poly(ADP-ribose) polymerase (PARP) inhibitors and epigenome-interfering agents. This evidence revives RTKs as promising targets for therapeutic intervention of EOC. This review summarizes recent studies on the role of RTKs in EOC malignancy and the use of their inhibitors for clinical treatment. Our focus is on the ERBB family, c-Met, and VEGFR, as they are linked to drug resistance and targetable using commercially available drugs. The importance of these RTKs and their inhibitors is highlighted by their impact on signal transduction and intratumoral heterogeneity in EOC and successful use as maintenance therapy in the clinic through suppression of the VEGF/VEGFR axis. Finally, the therapeutic potential of RTK inhibitors is discussed in the context of combinatorial targeting via co-inhibiting proliferative and anti-apoptotic pathways, epigenomic/transcriptional programs, and harnessing the efficacy of PARP inhibitors and programmed cell death 1/ligand 1 immune checkpoint therapies.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":"28 ","pages":"293-306"},"PeriodicalIF":5.7,"publicationDate":"2023-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/d1/2a/main.PMC9999170.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9775616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}