{"title":"Bile molecular landscape provides pathological insight and classifies signatures predictive of carcinoma of the gall bladder.","authors":"Nupur Sharma, Sadam H Bhat, Babu Mathew, Manisha Yadav, Gaurav Tripathi, Vasundhra Bindal, Sanju Yadav, Neha Sharma, Sushmita Pandey, Hami Hemati, Deepika Bohra, Rashmi Rana, Narendra Kumar Sharma, Sanyam Falari, Viniyendra Pamecha, Jaswinder Singh Maras","doi":"10.1016/j.omton.2024.200904","DOIUrl":"10.1016/j.omton.2024.200904","url":null,"abstract":"<p><p>Carcinoma of the gall bladder (CAGB) has a poor prognosis. Molecular analysis of bile could classify indicators of CAGB. Bile samples (<i>n</i> = 87; training cohort) were screened for proteomics and metabolomics signatures of cancer detection. In bile, CAGB showed distinct proteomic (217 upregulated, 258 downregulated) and metabolomic phenotypes (111 upregulated, 505 downregulated, <i>p</i> < 0.05, fold change > 1.5, false discovery rate <0.01) linked to significantly increased inflammation (coagulation, arachidonic acid, bile acid) and alternate energy pathways (pentose-phosphate pathway, amino acids, lipid metabolism); and decreased glycolysis, cholesterol metabolism, PPAR, RAS, and RAP1 signaling, oxidative phosphorylation, and others compared to gallstone or healthy controls (<i>p</i> < 0.05). Bile proteins/metabolites signatures showed significant correlation (<i>r</i> <sup><i>2</i></sup> > 0.5, <i>p</i> < 0.05) with clinical parameters. Metabolite/protein signature-based probability of detection for CAGB (cancer) was >90% (<i>p</i> < 0.05), with area under the receiver operating characteristic curve >0.94. Validation of the top four metabolites-toluene, 5,6-DHET, creatine, and phenylacetaldehyde-in separate cohorts (<i>n</i> = 80; bile [T1] and paired plasma [T2]) showed accuracy (99%) and sensitivity/specificity (>98%) for CAGB detection. Tissue validation showed bile 5,6-DHET positively correlated with tissue PCNA (proliferation), and caspase-3 linked to cancer development (<i>r</i> <sup><i>2</i></sup> >0.5, <i>p</i> < 0.05). In conclusion, the bile molecular landscape provides critical molecular understanding and outlines metabolomic indicator panels for early CAGB detection.</p>","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 4","pages":"200904"},"PeriodicalIF":0.0,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11617464/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142788175","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-10-28eCollection Date: 2024-12-19DOI: 10.1016/j.omton.2024.200899
Amanda Rosewell Shaw, Daisuke Morita, Caroline E Porter, Eric Tu, Greyson W Biegert, Sonia Agrawal, Nicholas Durham, Malcolm K Brenner, Masataka Suzuki
{"title":"IL-12 encoding oNDV synergizes with CAR-T cells in orthotopic models of non-small cell lung cancer.","authors":"Amanda Rosewell Shaw, Daisuke Morita, Caroline E Porter, Eric Tu, Greyson W Biegert, Sonia Agrawal, Nicholas Durham, Malcolm K Brenner, Masataka Suzuki","doi":"10.1016/j.omton.2024.200899","DOIUrl":"10.1016/j.omton.2024.200899","url":null,"abstract":"<p><p>Systemic administration of oncolytic viruses (OVs) is a promising approach for targeting metastatic solid tumors, but their anti-tumor activity is limited by pre-existing neutralizing antibodies against common human viruses. Therefore, investigators have developed OVs derived from non-human host viruses. Successful implementation of this strategy requires that the viral vector selectively infects and replicates within human cancer cells. Newcastle disease virus (NDV) is an avian paramyxovirus that, as NDV-based OVs (oNDVs), has demonstrated safety and activity against multiple human tumors in clinical trials. Their use as a single agent, however, is insufficient to cure tumors. Similarly, chimeric antigen receptor-modified T cells (CAR-T cells) enable systemic targeting of cancer cells but have limited anti-tumor effects against bulky solid tumors, in part due to the immunosuppressive tumor environment. In this study, we evaluated the anti-tumor effects of combining systemic oNDV and CAR-T cell treatments. In models of non-small cell lung carcinoma (NSCLC), we found that oNDV itself and interleukin (IL)-12 derived from oNDVs enhance HER2-directed CAR-T cell anti-tumor activity and persistence <i>in vitro</i> and <i>in vivo</i>, leading to superior control of NSCLC tumors compared with either agent alone <i>in vivo</i>. Our data indicate that oNDV enhances the anti-tumor effects of HER2.CAR-T cells, thus controlling the growth of orthotopic NSCLC tumors.</p>","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 4","pages":"200899"},"PeriodicalIF":0.0,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11609364/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142776304","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-10-10eCollection Date: 2024-12-19DOI: 10.1016/j.omton.2024.200869
Ajay Prakash, Travis Gates, Emil Lou
{"title":"Senescence-associated secretory phenotype regulation by dual-drug delivery biomimetic nanoplatform for enhanced tumor chemotherapy.","authors":"Ajay Prakash, Travis Gates, Emil Lou","doi":"10.1016/j.omton.2024.200869","DOIUrl":"https://doi.org/10.1016/j.omton.2024.200869","url":null,"abstract":"<p><p>Wang and colleagues provide a comprehensive evaluation of a biomimetic platform in which a specially designed drug delivery system, referred to in the manuscript as mPtP@Lipo, is used to increased penetration of dense stromatous tumors. This work represents a significant advancement in the treatment of desmoplastic tumors by targeting the tumor microenvironment (TME), particularly focusing on remodeling cancer-associated fibroblasts (CAFs). The study leverages previous work, which has demonstrated that platinum (Pt)-based chemotherapeutic regimens can induce a senescence-associated secretory phenotype (SASP) through persistent DNA damage, which can contribute to an immunosuppressive tumor immune microenvironment.</p>","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 4","pages":"200869"},"PeriodicalIF":0.0,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11907207/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143653024","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-09-26eCollection Date: 2024-12-19DOI: 10.1016/j.omton.2024.200886
Emily M Ringwalt, Mark A Currier, Andrea M Glaspell, Chun-Yu Chen, Matthew V Cannon, Maren Cam, Amy C Gross, Matthew Gust, Pin-Yi Wang, Louis Boon, Laura E Biederman, Emily Schwarz, Prajwal Rajappa, Dean A Lee, Elaine R Mardis, William E Carson, Ryan D Roberts, Timothy P Cripe
{"title":"Trabectedin promotes oncolytic virus antitumor efficacy, viral gene expression, and immune effector function in models of bone sarcoma.","authors":"Emily M Ringwalt, Mark A Currier, Andrea M Glaspell, Chun-Yu Chen, Matthew V Cannon, Maren Cam, Amy C Gross, Matthew Gust, Pin-Yi Wang, Louis Boon, Laura E Biederman, Emily Schwarz, Prajwal Rajappa, Dean A Lee, Elaine R Mardis, William E Carson, Ryan D Roberts, Timothy P Cripe","doi":"10.1016/j.omton.2024.200886","DOIUrl":"10.1016/j.omton.2024.200886","url":null,"abstract":"<p><p>We previously reported that the DNA alkylator and transcriptional-blocking chemotherapeutic agent trabectedin enhances oncolytic herpes simplex viroimmunotherapy in human sarcoma xenograft models, though the mechanism remained to be elucidated. Here we report trabectedin disrupts the intrinsic cellular antiviral response which increases viral transcript presence in the human tumor cells. We also extended our synergy findings to syngeneic murine sarcoma models, which are poorly susceptible to virus infection. In the absence of robust virus replication, we found trabectedin enhanced viroimmunotherapy efficacy by reducing infiltrating immunosuppressive CD4 T and myeloid cells and stimulating granzyme expression in infiltrating T and natural killer cells to cause immune-mediated tumor regressions. Thus, trabectedin enhances both the direct virus-mediated killing of tumor cells and the viral-induced activation of cytotoxic effector lymphocytes to cause tumor regressions across models. Our data provide a strong rationale for clinical translation as both mechanisms should be simultaneously active in human patients.</p>","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 4","pages":"200886"},"PeriodicalIF":0.0,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11530761/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142570946","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-09-24eCollection Date: 2024-12-19DOI: 10.1016/j.omton.2024.200885
Daekee Kwon, Bo Kyung Moon, Mijung Han, Tae-Wook Lee, Jeehan Lee, Kyung-Sun Kang
{"title":"Genetically stable multi-gene edited iPSCs-derived NK cells for enhanced cancer immunotherapy.","authors":"Daekee Kwon, Bo Kyung Moon, Mijung Han, Tae-Wook Lee, Jeehan Lee, Kyung-Sun Kang","doi":"10.1016/j.omton.2024.200885","DOIUrl":"https://doi.org/10.1016/j.omton.2024.200885","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR)-T cell treatment is an innovative drug with excellent therapeutic effects against B cell blood cancer. However, multiple side effects and ultra-high treatment costs must be overcome. Off-the-shelf CAR natural killer (NK) cells can be a good alternative to patient-specific CAR-T cells. The purpose of this study was to combine cellular reprogramming, gene editing, and differentiation technologies to produce full-off-the-shelf NK cells and to verify their efficacy and safety. Genetically stable universal and potent CAR (upCAR)-induced pluripotent stem cells (iPSCs) showed biallelic insertions and deletions in the coding sequence and no off-target effects. upCAR-NK cells showed a very high differentiation yield and <i>in vitro</i> proliferation, and freezing/thawing was possible. In addition, upCAR-NK cells secrete interferon-γ when they meet cancer cells, showing cytotoxic effects <i>in vitro</i> and <i>in vivo</i>. upCAR-NK cells show no obvious toxicity <i>in vivo</i>. In conclusion, this study developed genetically stable upCAR-iPSCs and upCAR-NK cell platform technologies that are less likely to have side effects and can be more economically developed for B cell blood cancer than CAR-T cells. In the future, this technology could be useful in developing a full-off-the-shelf CAR-NK cells anti-cancer immune cell therapy with low side effects, high efficacy, and a low price.</p>","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 4","pages":"200885"},"PeriodicalIF":0.0,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11907209/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143653023","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-07-13eCollection Date: 2024-09-19DOI: 10.1016/j.omton.2024.200847
Viviana Rubino, Prasad S Adusumilli, Undrakh Ganbaatar
{"title":"CD38 deletion to preserve CAR T cell metabolism and promote functional persistence.","authors":"Viviana Rubino, Prasad S Adusumilli, Undrakh Ganbaatar","doi":"10.1016/j.omton.2024.200847","DOIUrl":"10.1016/j.omton.2024.200847","url":null,"abstract":"","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 3","pages":"200847"},"PeriodicalIF":0.0,"publicationDate":"2024-07-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11300923/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141899338","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-05-20eCollection Date: 2024-06-20DOI: 10.1016/j.omton.2024.200811
Christopher J LaRocca, Julia Davydova
{"title":"Harnessing the power of viroimmunotherapy to overcome challenges in cancer therapy.","authors":"Christopher J LaRocca, Julia Davydova","doi":"10.1016/j.omton.2024.200811","DOIUrl":"10.1016/j.omton.2024.200811","url":null,"abstract":"","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 2","pages":"200811"},"PeriodicalIF":0.0,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11140413/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-04-17eCollection Date: 2024-06-20DOI: 10.1016/j.omton.2024.200801
Natalie M Meléndez-Vázquez, Teresa T Nguyen, Xuejun Fan, Andrés R López-Rivas, Juan Fueyo, Candelaria Gomez-Manzano, Filipa Godoy-Vitorino
{"title":"Erratum: Gut microbiota composition is associated with the efficacy of Delta-24-RGDOX in malignant gliomas.","authors":"Natalie M Meléndez-Vázquez, Teresa T Nguyen, Xuejun Fan, Andrés R López-Rivas, Juan Fueyo, Candelaria Gomez-Manzano, Filipa Godoy-Vitorino","doi":"10.1016/j.omton.2024.200801","DOIUrl":"10.1016/j.omton.2024.200801","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1016/j.omton.2024.200787.].</p>","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 2","pages":"200801"},"PeriodicalIF":0.0,"publicationDate":"2024-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11047782/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140871355","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-03-08eCollection Date: 2024-03-21DOI: 10.1016/j.omton.2024.200781
Timothy P Cripe
{"title":"Reviving the lost art of meeting reports.","authors":"Timothy P Cripe","doi":"10.1016/j.omton.2024.200781","DOIUrl":"https://doi.org/10.1016/j.omton.2024.200781","url":null,"abstract":"","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 1","pages":"200781"},"PeriodicalIF":0.0,"publicationDate":"2024-03-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10943060/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140874170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular therapy. OncologyPub Date : 2024-02-28eCollection Date: 2024-03-21DOI: 10.1016/j.omton.2024.200787
Natalie M Meléndez-Vázquez, Teresa T Nguyen, Xuejun Fan, Andrés R López-Rivas, Juan Fueyo, Candelaria Gomez-Manzano, Filipa Godoy-Vitorino
{"title":"Gut microbiota composition is associated with the efficacy of Delta-24-RGDOX in malignant gliomas.","authors":"Natalie M Meléndez-Vázquez, Teresa T Nguyen, Xuejun Fan, Andrés R López-Rivas, Juan Fueyo, Candelaria Gomez-Manzano, Filipa Godoy-Vitorino","doi":"10.1016/j.omton.2024.200787","DOIUrl":"https://doi.org/10.1016/j.omton.2024.200787","url":null,"abstract":"<p><p>Glioblastoma, the most common primary brain tumor, has a 6.8% survival rate 5 years post diagnosis. Our team developed an oncolytic adenovirus with an OX-40L expression cassette named Delta-24-RGDOX. While studies have revealed the interaction between the gut microbiota and immunotherapy agents, there are no studies linking the gut microbiota with viroimmunotherapy efficacy. We hypothesize that gut bacterial signatures will be associated with oncolytic viral therapy efficacy. To test this hypothesis, we evaluated the changes in gut microbiota in two mouse cohorts: (1) GSC-005 glioblastoma-bearing mice treated orally with indoximod, an immunotherapeutic agent, or with Delta-24-RGDOX by intratumoral injection and (2) a mouse cohort harboring GL261-5 tumors used to mechanistically evaluate the importance of CD4<sup>+</sup> T cells in relation to viroimmunotherapy efficacy. Microbiota assessment indicated significant differences in the structure of the gut bacterial communities in viroimmunotherapy-treated animals with higher survival compared with control or indoximod-treated animals. Moreover, viroimmunotherapy-treated mice with prolonged survival had a higher abundance of <i>Bifidobacterium</i>. The CD4<sup>+</sup> T cell depletion was associated with gut dysbiosis, lower mouse survival, and lower antitumor efficacy of the therapy. These findings suggest that microbiota modulation along the gut-glioma axis contributes to the clinical efficacy and patient survival of viroimmunotherapy treated animals.</p>","PeriodicalId":519884,"journal":{"name":"Molecular therapy. Oncology","volume":"32 1","pages":"200787"},"PeriodicalIF":0.0,"publicationDate":"2024-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10951704/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140873506","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}