OncotargetPub Date : 2025-05-29DOI: 10.18632/oncotarget.28730
Jinah Kim, Liam Il-Young Chung, Horyun Choi, Yoonhee Choi, Maria Jose Aguilera Chuchuca, Youjin Oh, Young Kwang Chae
{"title":"Durable complete response in leptomeningeal disease of EGFR mutated non-small cell lung cancer to amivantamab, an EGFR-MET receptor bispecific antibody, after progressing on osimertinib.","authors":"Jinah Kim, Liam Il-Young Chung, Horyun Choi, Yoonhee Choi, Maria Jose Aguilera Chuchuca, Youjin Oh, Young Kwang Chae","doi":"10.18632/oncotarget.28730","DOIUrl":"https://doi.org/10.18632/oncotarget.28730","url":null,"abstract":"<p><p>Patients with NSCLC harboring uncommon EGFR mutations have limited therapeutic options and often poor outcomes, especially following progression on standard EGFR tyrosine kinase inhibitors (TKIs). Amivantamab, an anti-EGFR/MET bispecific antibody, shows efficacy in EGFR-mutated NSCLC, but its role in rare EGFR alterations and CNS involvement, including leptomeningeal disease (LMD), remains insufficiently characterized. We report a 67-year-old male with stage IVB NSCLC harboring EGFR G719A and A289V mutations who developed LMD while on osimertinib. After progression on immunotherapy and chemotherapy, amivantamab monotherapy was initiated. Treatment produced a durable response over 19 months, including a 32.2% reduction in tumor size at six weeks, and complete resolution of brain metastases and LMD by six months. Circulating tumor DNA analyses showed EGFR variant allele fractions decreasing from 25.6% to undetectable. This case challenges current paradigms, demonstrating that amivantamab monotherapy can effectively target rare EGFR mutations, achieve durable extracranial and CNS disease control, and potentially overcome blood-brain barrier limitations. These findings suggest that amivantamab's utility may extend beyond established combination regimens and well-characterized EGFR mutations, offering an effective option for patients with atypical molecular profiles who lack standard therapies. Amivantamab monotherapy may represent a viable strategy for patients with uncommon EGFR mutations and CNS involvement, including LMD. Further research is warranted to elucidate underlying mechanisms, refine treatment strategies, and assess amivantamab's broader applicability in similarly challenging NSCLC scenarios.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"16 ","pages":"411-418"},"PeriodicalIF":0.0,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144174292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncotargetPub Date : 2025-05-20DOI: 10.18632/oncotarget.28724
Nawar Al Nasrallah, Bowa Lee, Benjamin M Wiese, Marie N Karam, Elizabeth A Mickler, Huaxin Zhou, Nicki Paolelli, Robert S Stearman, Mark W Geraci, Catherine R Sears
{"title":"Cigarette smoke and decreased DNA repair by Xeroderma Pigmentosum Group C use a double hit mechanism for epithelial cell lung carcinogenesis.","authors":"Nawar Al Nasrallah, Bowa Lee, Benjamin M Wiese, Marie N Karam, Elizabeth A Mickler, Huaxin Zhou, Nicki Paolelli, Robert S Stearman, Mark W Geraci, Catherine R Sears","doi":"10.18632/oncotarget.28724","DOIUrl":"10.18632/oncotarget.28724","url":null,"abstract":"<p><p>Emerging evidence suggests a complex interplay of environmental and genetic factors in non-small cell lung cancer (NSCLC) development. Among these factors, compromised DNA repair plays a critical but incompletely understood role in lung tumorigenesis and concurrent lung diseases, such as chronic obstructive lung disease (COPD). In this study, we investigated the interplay between cigarette smoke, DNA damage and repair, focusing on the Nucleotide Excision Repair (NER) protein Xeroderma Pigmentosum Group C (XPC). We found decreased XPC mRNA expression in most NSCLCs compared to subject-matched, non-cancerous lung. In non-cancerous bronchial epithelial cells, cigarette smoke decreased NER, increased total DNA damage and resultant apoptosis, each exacerbated by XPC deficiency. In contrast, lung cancer cells exhibit greater resilience to cigarette smoke, requiring higher doses to induce comparable DNA damage and apoptosis, and are less reliant on XPC expression for survival. Importantly, XPC protects against chromosomal instability in benign bronchial epithelial cells, but not in lung cancer cells. Our findings support a \"double hit\" mechanism wherein early decreased XPC expression and resultant aberrant DNA repair, when combined with cigarette smoke exposure, may lead to loss of non-malignant epithelial cells (as observed in COPD), and contributes to early NSCLC transition through altered DNA damage response.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"16 ","pages":"396-409"},"PeriodicalIF":0.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144111510","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncotargetPub Date : 2025-05-20DOI: 10.18632/oncotarget.28722
Shan Lu, Zhongyun Dong
{"title":"Targeting PCNA/AR interaction inhibits AR-mediated signaling in castration resistant prostate cancer cells.","authors":"Shan Lu, Zhongyun Dong","doi":"10.18632/oncotarget.28722","DOIUrl":"https://doi.org/10.18632/oncotarget.28722","url":null,"abstract":"<p><p>We previously showed that proliferating cell nuclear antigen (PCNA) interacts with androgen receptor (AR) through a PIP-box (PIP-box4) at the N-terminus of AR and regulates AR activity. In this study, we further investigated PCNA/AR interaction. We identified a second PIP-box (PIP-box592) in the DNA binding domain of AR and found that dihydrotestosterone enhances the binding of full-length AR (AR-FL) but not a constitutively active variant (AR-V7) to PCNA. Treatment with R9-AR-PIP, a PIP-box4-mimicking small peptide, inhibits the PCNA/AR interaction, AR occupancy at the androgen response element (ARE) in <i>PSA</i> and <i>p21</i> genes, and expression of AR target genes, and induces cytotoxicity in AR-positive castration-resistant prostate cancer (CRPC) cells. R9-AR-PIP also significantly inhibits transcriptional activity of AR-FL upon dihydrotestosterone stimulation and the constitutive activity of AR-V7. Moreover, R9-AR-PIP and PCNA-I1S, a small molecule PCNA inhibitor, inhibit the ARE occupancy by AR-FL and AR-Vs in <i>CCNA2</i> gene that encodes cyclin A2 and cyclin A2 expression. Finally, we found that cyclin A2 is overexpressed in all CRPC cells examined, suggesting that it may contribute to the development of CRPC. These data indicate that targeting PCNA/AR interaction inhibits both AR-FL- and AR-Vs-mediated signaling and implicates it could be a novel therapeutic strategy against CRPC.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"16 ","pages":"383-395"},"PeriodicalIF":0.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144111517","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncotargetPub Date : 2025-05-20DOI: 10.18632/oncotarget.28728
María Laura Polo, Marina Riggio, María May, María Jimena Rodríguez, María Cecilia Perrone, Melody Stallings-Mann, Diego Kaen, Marlene Frost, Matthew Goetz, Judy Boughey, Claudia Lanari, Derek Radisky, Virginia Novaro
{"title":"Correction: Activation of PI3K/Akt/mTOR signaling in the tumor stroma drives endocrine therapy-dependent breast tumor regression.","authors":"María Laura Polo, Marina Riggio, María May, María Jimena Rodríguez, María Cecilia Perrone, Melody Stallings-Mann, Diego Kaen, Marlene Frost, Matthew Goetz, Judy Boughey, Claudia Lanari, Derek Radisky, Virginia Novaro","doi":"10.18632/oncotarget.28728","DOIUrl":"10.18632/oncotarget.28728","url":null,"abstract":"","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"16 ","pages":"379-381"},"PeriodicalIF":0.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144111607","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advancements in bladder cancer treatment: The synergy of radiation and immunotherapy.","authors":"Nazmul Hasan, Daniel Yang, Spencer Gibson, Barbod Khaleghi, Rozhan Ziari, Arash Rezazadeh Kalebasty","doi":"10.18632/oncotarget.28723","DOIUrl":"10.18632/oncotarget.28723","url":null,"abstract":"<p><p>Different treatment strategies are required for the non-muscle-invasive, muscle-invasive, and metastatic stages of bladder cancer. Standard treatments include surgery, chemotherapy, and radiation; however, they have their limitations. New discoveries have shown that combining immunotherapy and radiation treatment may improve patient outcomes. Radiation therapy promotes immunogenic cell death, which leads to antigen release and immune cell activation, whereas immunotherapy enhances the immune system's ability to recognize and destroy cancer cells by targeting checkpoint pathways like PD-1/PD-L1 and CTLA-4. This review examines the synergistic mechanisms of diverse modalities, focusing on their capacity to alter the tumor microenvironment and elicit systemic anti-tumor responses, such as the abscopal effect. Key clinical trials, such as BTCRC-GU15-023 and ANZUP, have demonstrated the efficacy and safety of combining these medications. However, difficulties persist, such as overlapping toxicities, unpredictability in patient responses, and a lack of accurate patient selection markers. Large-scale randomized trials are needed in the future to fine-tune treatment procedures, minimize toxicity, and validate predictive biomarkers such as PD-L1 expression and tumor mutation burden. By addressing these hurdles, the combination of radiation treatment and immunotherapy has the potential to change the bladder cancer therapeutic landscape.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"16 ","pages":"337-346"},"PeriodicalIF":0.0,"publicationDate":"2025-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12088031/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144094482","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}
OncotargetPub Date : 2025-05-19DOI: 10.18632/oncotarget.28721
Leonardo Gomes da Fonseca, Federico Piñero, Margarita Anders, Carla Bermudez, Ezequiel Demirdjian, Adriana Varón, Daniela Perez, Jorge Rodriguez, Oscar Beltrán, Ezequiel Ridruejo, Pablo Caballini, Alexandre Araujo, Juan Diego Torres Florez, Juan Ignacio Marín, Marina Villa, Federico Orozco, Jaime Poniachik, Sebastián Marciano, Fernando Bessone, Manuel Mendizabal
{"title":"Immune-mediated adverse events following atezolizumab and bevacizumab in a multinational Latin American cohort of unresectable hepatocellular carcinoma.","authors":"Leonardo Gomes da Fonseca, Federico Piñero, Margarita Anders, Carla Bermudez, Ezequiel Demirdjian, Adriana Varón, Daniela Perez, Jorge Rodriguez, Oscar Beltrán, Ezequiel Ridruejo, Pablo Caballini, Alexandre Araujo, Juan Diego Torres Florez, Juan Ignacio Marín, Marina Villa, Federico Orozco, Jaime Poniachik, Sebastián Marciano, Fernando Bessone, Manuel Mendizabal","doi":"10.18632/oncotarget.28721","DOIUrl":"10.18632/oncotarget.28721","url":null,"abstract":"<p><strong>Aims: </strong>Latin America has been underrepresented in trials evaluating immunotherapy for hepatocellular carcinoma (HCC). We aimed to describe the incidence of immune-related adverse events (irAEs) and their impact on outcomes in a Latin American cohort.</p><p><strong>Methods: </strong>A multicenter prospective study was conducted in Argentina, Brazil, Chile, and Colombia, including patients who received atezolizumab plus bevacizumab. A time-covarite proportional hazard analysis evaluated the effect of irAEs.</p><p><strong>Results: </strong>99 patients were included. The median treatment duration was 6 months, with a median survival of 17.0 months (95% CI: 12.6-19.8). The irAE incidence rate was 2.1 cases per 100 persons-months (cumulative incidence 18.1% (95% CI: 11.1-27.2%)). Median time to irAE was 2.3 months (range 1.4-4.8), most frequently hepatitis (<i>n</i> = 6), thyroiditis (<i>n</i> = 5), and 8/18 required steroids. Follow-up, treatment duration, and overall survival were similar regardless of the occurrence of irAEs (HR = 1.71, 95% CI: 0.76-3.86; <i>P</i> = 0.19). Baseline alpha-feto protein ≥400 ng/ml (HR: 2.9 (95% CI: 1.1-7.6)) was independently associated with irAE.</p><p><strong>Conclusion: </strong>The incidence of irAEs in this cohort is lower than reported in controlled trials, withouut impact on survival outcomes. Education and early recognition are crucial to ensure that these events are identified and addressed.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"16 ","pages":"348-360"},"PeriodicalIF":0.0,"publicationDate":"2025-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12088043/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144093628","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}
{"title":"PRDX1 protects ATM from arsenite-induced proteotoxicity and maintains its stability during DNA damage signaling.","authors":"Reem Ali, Mashael Algethami, Amera Sheha, Shatha Alqahtani, Ahmad Altayyar, Ayat Lashen, Emad Rakha, Abdallah Alhaj Sulaiman, Srinivasan Madhusudan, Dindial Ramotar","doi":"10.18632/oncotarget.28720","DOIUrl":"10.18632/oncotarget.28720","url":null,"abstract":"<p><p>Redox regulation and DNA repair coordination are essential for genomic stability. Peroxiredoxin 1 (PRDX1) is a thiol-dependent peroxidase and a chaperone that protects proteins from excessive oxidation. ATM kinase (Ataxia-Telangiectasia Mutated) and the MRN (MRE11-RAD50-NBS1) complex are DNA damage signaling and repair proteins. We previously showed that cells lacking PRDX1 are sensitive to arsenite, a toxic metal that induces DNA single- and double-strand breaks (DSBs). Herein, we showed that PRDX1 interacts with ATM. PRDX1-deleted cells have reduced ATM, MRE11, and RAD50 protein levels, but not NBS1. In control cells treated with arsenite, we observed γH2AX foci formation due to arsenite-induced DSBs, and not from PRDX1-deleted cells. Arsenite caused profound depletion of ATM in PRDX1-deleted cells, suggesting that PRDX1 protects and stabilizes ATM required to form γH2AX foci. Importantly, arsenite pretreatment of PRDX1-deleted cells caused hypersensitivity to chemotherapeutic agents that generate DSBs. Analysis of a clinical cohort of ovarian cancers treated with platinum chemotherapy revealed that tumours with high PRDX1/high ATM or high PRDX1/high MRE11 expression manifested aggressive phenotypes and poor patient survival. The data suggest that PRDX1 can predict responses to chemotherapy, and targeting PRDX1 could be a viable strategy to improve the efficacy of platinum chemotherapy.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"16 ","pages":"362-378"},"PeriodicalIF":0.0,"publicationDate":"2025-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12088036/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144093644","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}