27 results on '"Bast, Jr, Robert C."'
Search Results
2. Author Correction: Tuning microtubule dynamics to enhance cancer therapy by modulating FER-mediated CRMP2 phosphorylation
- Author
-
Zheng, Yiyan, Sethi, Ritika, Mangala, Lingegowda S., Taylor, Charlotte, Goldsmith, Juliet, Wang, Ming, Masuda, Kenta, Carrami, Eli M., Mannion, David, Miranda, Fabrizio, Herrero-Gonzalez, Sandra, Hellner, Karin, Chen, Fiona, Alsaadi, Abdulkhaliq, Albukhari, Ashwag, Fotso, Donatien Chedom, Yau, Christopher, Jiang, Dahai, Pradeep, Sunila, Rodriguez-Aguayo, Cristian, Lopez-Berestein, Gabriel, Knapp, Stefan, Gray, Nathanael S., Campo, Leticia, Myers, Kevin A., Dhar, Sunanda, Ferguson, David, Bast, Jr, Robert C., Sood, Anil K., von Delft, Frank, and Ahmed, Ahmed Ashour
- Published
- 2022
- Full Text
- View/download PDF
3. IL-2-free tumor-infiltrating lymphocyte therapy with PD-1 blockade demonstrates potent efficacy in advanced gynecologic cancer.
- Author
-
Guo, Jing, Wang, Chunyan, Luo, Ning, Wu, Yuliang, Huang, Wei, Zhu, Jihui, Shi, Weihui, Ding, Jinye, Ge, Yao, Liu, Chunhong, Lu, Zhen, Bast Jr, Robert C., Ai, Guihai, Yang, Weihong, Wang, Rui, Li, Caixia, Chen, Rong, Liu, Shupeng, Jin, Huajun, and Zhao, Binghui
- Subjects
GYNECOLOGIC cancer ,TUMOR-infiltrating immune cells ,CANCER patients ,PROGRAMMED cell death 1 receptors ,T cells ,INTRAVENOUS therapy - Abstract
Background: Tumor-infiltrating lymphocyte (TIL) therapy has been restricted by intensive lymphodepletion and high-dose intravenous interleukin-2 (IL-2) administration. To address these limitations, we conducted preclinical and clinical studies to evaluate the safety, antitumor activity, and pharmacokinetics of an innovative modified regimen in patients with advanced gynecologic cancer. Methods: Patient-derived xenografts (PDX) were established from a local recurrent cervical cancer patient. TILs were expanded ex vivo from minced tumors without feeder cells in the modified TIL therapy regimen. Patients underwent low-dose cyclophosphamide lymphodepletion followed by TIL infusion without intravenous IL-2. The primary endpoint was safety; the secondary endpoints included objective response rate, duration of response, and T cell persistence. Results: In matched patient-derived xenografts (PDX) models, homologous TILs efficiently reduced tumor size (p < 0.0001) and underwent IL-2 absence in vivo. In the clinical section, all enrolled patients received TIL infusion using a modified TIL therapy regimen successfully with a manageable safety profile. Five (36%, 95% CI 16.3–61.2) out of 14 evaluable patients experienced objective responses, and three complete responses were ongoing at 19.5, 15.4, and 5.2 months, respectively. Responders had longer overall survival (OS) than non-responders (p = 0.036). Infused TILs showed continuous proliferation and long-term persistence in all patients and showed greater proliferation in responders which was indicated by the Morisita overlap index (MOI) of TCR clonotypes between infused TILs and peripheral T cells on day 14 (p = 0.004) and day 30 (p = 0.004). Higher alteration of the CD8
+ /CD4+ ratio on day 14 indicated a longer OS (p = 0.010). Conclusions: Our modified TIL therapy regimen demonstrated manageable safety, and TILs could survive and proliferate without IL-2 intravenous administration, showing potent efficacy in patients with advanced gynecologic cancer. Trial registration: NCT04766320, Jan 04, 2021. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. A phase II study of imatinib mesylate and letrozole in patients with hormone receptor-positive metastatic breast cancer expressing c-kit or PDGFR-β
- Author
-
Yam, Clinton, Murthy, Rashmi K., Rauch, Gaiane M., Murray, James L., Walters, Ronald S., Valero, Vicente, Brewster, Abenaa M., Bast, Jr, Robert C., Booser, Daniel J., Giordano, Sharon H., Esteva, Francisco J., Yang, Wei, Hortobagyi, Gabriel N., Moulder, Stacy L., and Arun, Banu
- Published
- 2018
- Full Text
- View/download PDF
5. Novel Approaches to Ovarian Cancer Screening
- Author
-
Nebgen, Denise R., Lu, Karen H., and Bast, Jr, Robert C.
- Published
- 2019
- Full Text
- View/download PDF
6. Tuning microtubule dynamics to enhance cancer therapy by modulating FER-mediated CRMP2 phosphorylation
- Author
-
Zheng, Yiyan, Sethi, Ritika, Mangala, Lingegowda S., Taylor, Charlotte, Goldsmith, Juliet, Wang, Ming, Masuda, Kenta, Carrami, Eli M., Mannion, David, Miranda, Fabrizio, Herrero-Gonzalez, Sandra, Hellner, Karin, Chen, Fiona, Alsaadi, Abdulkhaliq, Albukhari, Ashwag, Fotso, Donatien Chedom, Yau, Christopher, Jiang, Dahai, Pradeep, Sunila, Rodriguez-Aguayo, Cristian, Lopez-Berestein, Gabriel, Knapp, Stefan, Gray, Nathanael S., Campo, Leticia, Myers, Kevin A., Dhar, Sunanda, Ferguson, David, Bast, Jr., Robert C., Sood, Anil K., von Delft, Frank, and Ahmed, Ahmed Ashour
- Published
- 2018
- Full Text
- View/download PDF
7. Epac1 knockdown inhibits the proliferation of ovarian cancer cells by inactivating AKT/Cyclin D1/CDK4 pathway in vitro and in vivo
- Author
-
Gao, Meng, Ma, Yanyan, Bast, Jr., Robert C., Li, Yue, Wan, Lu, Liu, Yanping, Sun, Yingshuo, Fang, Zhenghui, Zhang, Lining, Wang, Xiaoyan, and Wei, Zengtao
- Published
- 2016
- Full Text
- View/download PDF
8. Adipocyte-like signature in ovarian cancer minimal residual disease identifies metabolic vulnerabilities of tumor initiating cells
- Author
-
Artibani, Mara, primary, Masuda, Kenta, additional, Hu, Zhiyuan, additional, Rauher, Pascal C., additional, Mallett, Garry, additional, Wietek, Nina, additional, Morotti, Matteo, additional, Chong, Kay, additional, KaramiNejadRanjbar, Mohammad, additional, Zois, Christos E., additional, Dhar, Sunanda, additional, El-Sahhar, Salma, additional, Campo, Leticia, additional, Blagden, Sarah P., additional, Damato, Stephen, additional, Pathiraja, Pubudu N., additional, Nicum, Shibani, additional, Gleeson, Fergus, additional, Laios, Alexandros, additional, Alsaadi, Abdulkhaliq, additional, Santana Gonzalez, Laura, additional, Motohara, Takeshi, additional, Albukhari, Ashwag, additional, Lu, Zhen, additional, Bast Jr., Robert C., additional, Harris, Adrian L., additional, Ejsing, Christer S., additional, Klemm, Robin W., additional, Yau, Christopher, additional, Sauka-Spengler, Tatjana, additional, and Ahmed, Ahmed A., additional
- Published
- 2021
- Full Text
- View/download PDF
9. Structure-Based Design of Stapled Peptides That Bind GABARAP and Inhibit Autophagy.
- Author
-
Brown, Hawley, Chung, Mia, Üffing, Alina, Batistatou, Nefeli, Tsang, Tiffany, Doskocil, Samantha, Mao, Weiqun, Willbold, Dieter, Bast Jr., Robert C., Lu, Zhen, Weiergräber, Oliver H., and Kritzer, Joshua A.
- Published
- 2022
- Full Text
- View/download PDF
10. SIK2 promotes ovarian cancer cell motility and metastasis by phosphorylating MYLK.
- Author
-
Xiu Shi, Xuejiao Yu, Juan Wang, Shimin Bian, Qiutong Li, Fengqing Fu, Xinwei Zou, Lin Zhang, Bast Jr., Robert C., Zhen Lu, Lingchuan Guo, Youguo Chen, and Jinhua Zhou
- Abstract
Salt-inducible kinase 2 (SIK2; also known as serine/threonine-protein kinase SIK2) is overexpressed in several cancers and has been implicated in cancer progression. However, the mechanisms by which SIK2 regulates cancer cell motility, migration and metastasis in ovarian cancer have not been fully discovered. Here, we identify that SIK2 promotes ovarian cancer cell motility, migration and metastasis in vitro and in vivo. Mechanistically, SIK2 regulated cancer cell motility and migration by myosin light chain kinase, smooth muscle (MYLK)-meditated phosphorylation of myosin light chain 2 (MYL2). SIK2 directly phosphorylated MYLK at Ser343 and activated its downstream effector MYL2, promoting ovarian cancer cell motility and metastasis. In addition, we found that adipocytes induced SIK2 phosphorylation at Ser358 and MYLK phosphorylation at Ser343, enhancing ovarian cancer cell motility. Moreover, SIK2 protein expression was positively correlated with the expression of MYLK-pS343 in ovarian cancer cell lines and tissues. The coexpression of SIK2 and MYLK-pS343 was associated with reduced median overall survival in human ovarian cancer samples. Taken together, SIK2 positively regulates ovarian cancer motility, migration and metastasis, suggesting that SIK2 is a potential candidate for ovarian cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
11. Development of a Multiprotein Classifier for the Detection of Early Stage Ovarian Cancer.
- Author
-
Boylan, Kristin L. M., Petersen, Ashley, Starr, Timothy K., Pu, Xuan, Geller, Melissa A., Bast Jr., Robert C., Lu, Karen H., Cavallaro, Ugo, Connolly, Denise C., Elias, Kevin M., Cramer, Daniel W., Pejovic, Tanja, and Skubitz, Amy P. N.
- Subjects
OVARIAN tumors ,BLOOD proteins ,EARLY detection of cancer ,GENE expression ,TUMOR markers ,TUMOR antigens ,SENSITIVITY & specificity (Statistics) - Abstract
Simple Summary: When ovarian cancer is detected early, the survival rate is high. Unfortunately, existing blood tests are neither sensitive nor specific enough to screen women for ovarian cancer. The purpose of this study was to determine the levels of 92 cancer-related proteins in the blood of women with ovarian cancer compared to healthy women in order to develop a test for ovarian cancer detection. We tested the blood of more than 400 women and identified four proteins that, when combined, successfully detected over 90% of the women with ovarian cancer. We then tested more than 700 additional blood samples and found that the combination of the four proteins successfully distinguished the majority of the blood samples from women with both early and late stages of ovarian cancer compared to healthy women. These four proteins show promise in the development of a test to detect the early stages of ovarian cancer. Background: Individual serum biomarkers are neither adequately sensitive nor specific for use in screening the general population for ovarian cancer. The purpose of this study was to develop a multiprotein classifier to detect the early stages of ovarian cancer, when it is most treatable. Methods: The Olink Proseek Multiplex Oncology II panel was used to simultaneously quantify the expression levels of 92 cancer-related proteins in sera. Results: In the discovery phase, we generated a multiprotein classifier that included CA125, HE4, ITGAV, and SEZ6L, based on an analysis of sera from 116 women with early stage ovarian cancer and 336 age-matched healthy women. CA125 alone achieved a sensitivity of 87.9% at a specificity of 95%, while the multiprotein classifier resulted in an increased sensitivity of 91.4%, while holding the specificity fixed at 95%. The performance of the multiprotein classifier was validated in a second cohort comprised of 192 women with early stage ovarian cancer and 467 age-matched healthy women. The sensitivity at 95% specificity increased from 74.5% (CA125 alone) to 79.2% with the multiprotein classifier. In addition, the multiprotein classifier had a sensitivity of 95.1% at 98% specificity for late stage ovarian cancer samples and correctly classified 80.5% of the benign samples using the 98% specificity cutpoint. Conclusions: The inclusion of the proteins HE4, ITGAV, and SEZ6L improved the sensitivity and specificity of CA125 alone for the detection of early stages of ovarian cancer in serum samples. Furthermore, we identified several proteins that may be novel biomarkers of early stage ovarian cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. Directed Evolution of PD-L1-Targeted Affibodies by mRNA Display.
- Author
-
Grindel, Brian J., Engel, Brian J., Ong, Justin N., Srinivasamani, Anupallavi, Liang, Xiaowen, Zacharias, Niki M., Bast Jr., Robert C., Curran, Michael A., Takahashi, Terry T., Roberts, Richard W., and Millward, Steven W.
- Published
- 2022
- Full Text
- View/download PDF
13. One-Pot, One-Step Synthesis of Drug-Loaded Magnetic Multimicelle Aggregates.
- Author
-
Kim, Chang Soo, Nevozhay, Dmitry, Aburto, Rebeca Romero, Pehere, Ashok, Pang, Lan, Dillard, Rebecca, Wang, Ziqiu, Smith, Clayton, Mathieu, Kelsey Boitnott, Zhang, Marie, Hazle, John D., Bast Jr., Robert C., and Sokolov, Konstantin
- Published
- 2022
- Full Text
- View/download PDF
14. Proteome Profiling Uncovers an Autoimmune Response Signature That Reflects Ovarian Cancer Pathogenesis
- Author
-
Kobayashi, Makoto, primary, Katayama, Hiroyuki, additional, Irajizad, Ehsan, additional, Vykoukal, Jody V., additional, Fahrmann, Johannes F., additional, Kundnani, Deepali L., additional, Yu, Chuan-Yih, additional, Cai, Yining, additional, Hsiao, Fu Chung, additional, Yang, Wei-Lei, additional, Lu, Zhen, additional, Celestino, Joseph, additional, Long, James P., additional, Do, Kim-Ann, additional, Lu, Karen H., additional, Ladd, Jon J., additional, Urban, Nicole, additional, Bast Jr., Robert C., additional, and Hanash, Samir M., additional
- Published
- 2020
- Full Text
- View/download PDF
15. Targeting progesterone signaling prevents metastatic ovarian cancer.
- Author
-
Kim, Olga, Eun Young Park, Sun Young Kwon, Sojin Shin, Emerson, Robert E., Yong-Hyun Shin, DeMayo, Francesco J., Lydon, John P., Coffey, Donna M., Hawkins, Shannon M., Quilliam, Lawrence A., Dong-Joo Cheon, Fernández, Facundo M., Nephew, Kenneth P., Karpf, Adam R., Widschwendter, Martin, Sood, Anil K., Bast Jr, Robert C., Godwin, Andrew K., and Miller, Kathy D.
- Subjects
OVARIAN cancer ,PROGESTERONE ,METASTASIS ,BRCA genes ,PROGESTERONE receptors - Abstract
Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (~18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
16. Biomarkers and Strategies for Early Detection of Ovarian Cancer.
- Author
-
Bast Jr, Robert C., Zhen Lu, Chae Young Han, Lu, Karen H., Anderson, Karen S., Drescher, Charles W., and Skates, Steven J.
- Abstract
Early detection of ovarian cancer remains an important unmet medical need. Effective screening could reduce mortality by 10%-30%. Used individually, neither serum CA125 nor transvaginal sonography (TVS) is sufficiently sensitive or specific. Two-stage strategies have proven more effective, where a significant rise above a woman's baseline CA125 prompts TVS and an abnormal sonogram prompts surgery. Two major screening trials have documented that this strategy has adequate specificity, but sensitivity for early-stage (I-II) disease must improve to have a greater impact on mortality. To improve the first stage, different panels of protein biomarkers have detected cases missed by CA125. Autoantibodies against TP53 have detected 20% of early-stage ovarian cancers 8 months before elevation of CA125 and 22 months before clinical diagnosis. Panels of autoantibodies and antigen-autoantibody complexes are being evaluated with the goal of detecting >90% of early-stage ovarian cancers, alone or in combination with CA125, while maintaining 98% specificity in control subjects. Other biomarkers, including micro-RNAs, ctDNA, methylated DNA, and combinations of ctDNA alterations, are being tested to provide an optimal first-stage test. New technologies are also being developed with greater sensitivity than TVS to image small volumes of tumor. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
17. The National Cancer Institute Early Detection Research Network: Two Decades of Progress.
- Author
-
Bast Jr., Robert C. and Srivastava, Sudhir
- Published
- 2020
- Full Text
- View/download PDF
18. Analysis of RAS protein interactions in living cells reveals a mechanism for pan-RAS depletion bymembrane-targeted RAS binders.
- Author
-
Yao-Cheng Li, Lytle, Nikki K., Gammon, Seth T., Luke Wang, Hayes, Tikvah K., Sutton, Margie N., Bast Jr., Robert C., Der, Channing J., Piwnica-Worms, David, McCormick, Frank, and Wahl, Geoffrey M.
- Subjects
RAS proteins ,PROTEIN-protein interactions ,ONCOGENIC proteins ,PROTEIN analysis ,CARRIER proteins - Abstract
HRAS, NRAS, and KRAS4A/KRAS4B comprise the RAS family of small GTPases that regulate signaling pathways controlling cell proliferation, differentiation, and survival. RAS pathway abnormalities cause developmental disorders and cancers. We found that KRAS4B colocalizes on the cell membrane with other RAS isoforms and a subset of prenylated small GTPase family members using a live-cell quantitative split luciferase complementation assay. RAS protein coclustering is mainly mediated by membrane association-facilitated interactions (MAFIs). Using the RAS–RBD (CRAF RAS binding domain) interaction as a model system, we showed that MAFI alone is not sufficient to induce RBD-mediated RAS inhibition. Surprisingly, we discovered that high-affinity membrane-targeted RAS binding proteins inhibit RAS activity and deplete RAS proteins through an autophagosome–lysosome-mediated degradation pathway. Our results provide a mechanism for regulating RAS activity and protein levels, a more detailed understanding of which should lead to therapeutic strategies for inhibiting and depleting oncogenic RAS proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
19. Autophagy and Cancer Chemotherapy: Inhibition or Enhancement?
- Author
-
Santiago O Farrill, Janice M, primary, Lu, Zhen, additional, and Bast Jr, Robert C, additional
- Published
- 2016
- Full Text
- View/download PDF
20. Caspase‑3 Substrates for Noninvasive Pharmacodynamic Imaging of Apoptosis by PET/CT.
- Author
-
Engel, Brian J., Gammon, Seth T., Chaudhari, Rajan, Lu, Zhen, Pisaneschi, Federica, Yang, Hailing, Ornelas, Argentina, Yan, Victoria, Kelderhouse, Lindsay, Najjar, Amer M., Tong, William P., Zhang, Shuxing, Piwnica-Worms, David, Bast Jr., Robert C., and Millward, Steven W.
- Published
- 2018
- Full Text
- View/download PDF
21. Potentially inappropriate medication use in older patients with breast and colorectal cancer.
- Author
-
Karuturi, Meghan S., Holmes, Holly M., Lei, Xiudong, Johnson, Michael, Barcenas, Carlos H., Cantor, Scott B., Gallick, Gary E., Bast, Jr, Robert C., Giordano, Sharon H., and Bast, Robert C Jr
- Subjects
DRUG utilization ,OLDER patients ,BREAST cancer ,COLON cancer ,CANCER chemotherapy - Abstract
Background: The objective of this study was to determine patient characteristics associated with potentially inappropriate medication (PIM) use and its impact on outcomes for patients with breast or colorectal cancer receiving adjuvant chemotherapy.Methods: The Surveillance, Epidemiology, and End Results database, linked to Medicare claims, was used. The cohort included patients who were 66 years old or older and were diagnosed with stage II or III breast or colorectal cancer between July 1, 2007, and December 31, 2009. The Drugs to Avoid in the Elderly (DAE) list and the Beers criteria were used to identify PIM use. Univariate/multivariate logistic regression determined the association of baseline PIMs with covariates. Event-free survival (EFS) was defined as the time from chemotherapy initiation to the first emergency room (ER) visit, hospitalization, death, or a composite until 3 months after chemotherapy. Cox proportional hazards modeling determined the association of PIMs with EFS.Results: The analysis included 1595 patients with breast cancer and 1528 patients with colorectal cancer. The baseline PIM frequencies were 22.2% (according to the DAE list) and 27.6% (according to the Beers criteria) in the breast cohort and 15.5% (according to the DAE list) and 24.8% (according to the Beers criteria) in the colorectal cohort. Among patients with breast cancer, 37.5% had at least 1 adverse outcome; associations included the use of ≥5 medications, an advanced stage, higher comorbidity, and prior ER visits/hospitalizations. Baseline PIM use according to the DAE list was associated with an increased risk of death in patients with breast cancer. Among patients with colorectal cancer, 45% had at least 1 adverse outcome, and associations included the use of ≥5 medications, older age, female sex, and higher comorbidity. A time-to-event analysis revealed no association between baseline PIM use and most outcomes.Conclusions: These findings require further prospective confirmation, but they support a correlation between polypharmacy and adverse outcomes for cancer patients and call into question the association with PIMs. Cancer 2018;124:3000-7. © 2018 American Cancer Society. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
22. Induction of autophagy by ARHI (DIRAS3) alters fundamental metabolic pathways in ovarian cancer models.
- Author
-
Ornelas, Argentina, McCullough, Christopher R., Zhen Lu, Zacharias, Niki M., Kelderhouse, Lindsay E., Gray, Joshua, Yang, Hailing, Engel, Brian J., Yan Wang, Weiqun Mao, Sutton, Margie N., Bhattacharya, Pratip K., Bast Jr., Robert C., and Millward, Steven W.
- Subjects
OVARIAN cancer treatment ,AUTOPHAGY ,TUMOR suppressor genes ,APOPTOSIS ,GENE expression ,CANCER invasiveness ,NEOPLASTIC cell transformation - Abstract
Background: Autophagy is a bulk catabolic process that modulates tumorigenesis, therapeutic resistance, and dormancy. The tumor suppressor ARHI (DIRAS3) is a potent inducer of autophagy and its expression results in necroptotic cell death in vitro and tumor dormancy in vivo. ARHI is down-regulated or lost in over 60 % of primary ovarian tumors yet is dramatically up-regulated in metastatic disease. The metabolic changes that occur during ARHI induction and their role in modulating death and dormancy are unknown. Methods: We employed Nuclear Magnetic Resonance (NMR)-based metabolomic strategies to characterize changes in key metabolic pathways in both cell culture and xenograft models of ARHI expression and autophagy. These pathways were further interrogated by cell-based immunofluorescence imaging, tracer uptake studies, targeted metabolic inhibition, and in vivo PET/CT imaging. Results: Induction of ARHI in cell culture models resulted in an autophagy-dependent increase in lactate production along with increased glucose uptake and enhanced sensitivity to glycolytic inhibitors. Increased uptake of glutamine was also dependent on autophagy and dramatically sensitized cultured ARHI-expressing ovarian cancer cell lines to glutaminase inhibition. Induction of ARHI resulted in a reduction in mitochondrial respiration, decreased mitochondrial membrane potential, and decreased Tom20 staining suggesting an ARHI-dependent loss of mitochondrial function. ARHI induction in mouse xenograft models resulted in an increase in free amino acids, a transient increase in [18F]-FDG uptake, and significantly altered choline metabolism. Conclusions: ARHI expression has previously been shown to trigger autophagy-associated necroptosis in cell culture. In this study, we have demonstrated that ARHI expression results in decreased cellular ATP/ADP, increased oxidative stress, and decreased mitochondrial function. While this bioenergetic shock is consistent with programmed necrosis, our data indicates that the accompanying up-regulation of glycolysis and glutaminolysis is autophagy-dependent and serves to support cell viability rather than facilitate necroptotic cell death. While the mechanistic basis for metabolic up-regulation following ARHI induction is unknown, our preliminary data suggest that decreased mitochondrial function and increased metabolic demand may play a role. These alterations in fundamental metabolic pathways during autophagy-associated necroptosis may provide the basis for new therapeutic strategies for the treatment of dormant ovarian tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
23. Validation of a Biomarker Panel and Longitudinal Biomarker Performance for Early Detection of Ovarian Cancer.
- Author
-
Simmons, Archana R., Clarke, Charlotte H., Badgwell, Donna B., Zhen Lu, Sokoll, Lori J., Lu, Karen H., Zhen Zhang, Bast Jr, Robert C., and Skates, Steven J.
- Abstract
Objectives: Longitudinalmultimarker combinations have the potential to improve sensitivity while maintaining the high specificity required for the early detection of ovarian cancer. The use of multiplemarkers to improve sensitivity over cancer antigen 125 (CA125) in longitudinal algorithms for early ovarian cancer detection requires the selection of markers with optimal discriminatory power and low longitudinal variance relative to disease-initiated changes. Our objectivewas to identify amultimarker panel suitable for ovarian cancer, where each individual marker has its own baseline, permitting longitudinal algorithm development. Materials and Methods: In this retrospective study, we measured CA125, human epididymis protein 4 (HE4), matrix metalloproteinase-7 (MMP-7), CA72-4, CA19-9, CA15-3, carcinoembryonic antigen, and soluble vascular cell adhesion molecule (sVCAM) concentrations using immunoassays in pretreatment sera from 142 stage I ovarian cancer cases and 5 annual samples each from 217 healthy controls. After random division into training and validation sets, all possible biomarker combinations were explored exhaustively using linear classifiers to identify the panel with the greatest sensitivity for stage I disease at a high specificity of 98%. To evaluate longitudinal performance of the individual markers, the within-person over time and the between-person coefficient of variation (CV) were estimated. Hierarchical modeling across women of log-concentrations enabled the borrowing of information across subjects to moderate variance estimates given the small number of observations per subject. Results: The 4-marker panel comprising CA125, HE4, MMP-7, and CA72-4 performed with the highest sensitivity (83.2%) at 98% specificity. The within-person CVs were lower for CA125, HE4, MMP-7, and CA72-4 (15%, 25%, 25%, and 21%, respectively) compared with their corresponding between-person CV (49%, 20%, 35%, and 84%, respectively) indicating baselines in healthy volunteers. After simple log-transformations, the withinvolunteer variation across volunteers was modeled with a normal distribution permitting parsimonious hierarchical modeling. Conclusions: The multiplex panel chosen is suitable for the early detection of ovarian cancer and the individual markers have their own baseline permitting longitudinal algorithm development. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
24. Clinical Use of Cancer Biomarkers in Epithelial Ovarian Cancer.
- Author
-
Sölétormos, György, Duffy, Michael J., Abu Hassan, Suher Othman, Verheijen, René H. M., Tholander, Bengt, Bast Jr, Robert C., Gaarenstroom, Katja N., Sturgeon, Catharine M., Bonfrer, Johannes M., Petersen, Per Hyltoft, Troonen, Hugo, CarloTorre, Gian, Kulpa, Jan Kanty, Tuxen, Malgorzata K., and Molina, Raphael
- Published
- 2016
- Full Text
- View/download PDF
25. OTX015 Epi-Drug Exerts Antitumor Effects in Ovarian Cancer Cells by Blocking GNL3-Mediated Radioresistance Mechanisms: Cellular, Molecular and Computational Evidence.
- Author
-
Megiorni, Francesca, Camero, Simona, Pontecorvi, Paola, Camicia, Lucrezia, Marampon, Francesco, Ceccarelli, Simona, Anastasiadou, Eleni, Bernabò, Nicola, Perniola, Giorgia, Pizzuti, Antonio, Benedetti Panici, Pierluigi, Tombolini, Vincenzo, Marchese, Cinzia, Rizzo, Stefania, Manganaro, Lucia, and Bast Jr., Robert C.
- Subjects
THERAPEUTIC use of antineoplastic agents ,SURVIVAL ,IN vitro studies ,OVARIAN tumors ,DNA ,PROTEIN kinase inhibitors ,NUCLEAR proteins ,APOPTOSIS ,CELL cycle ,GENE expression ,CELLULAR signal transduction ,CELL proliferation ,CELL lines ,COMBINED modality therapy ,EPIGENOMICS - Abstract
Simple Summary: The outcome for women diagnosed with ovarian cancer (OC), the most aggressive gynecological tumor worldwide, remains very poor. Encouraging therapeutic impact of epigenetic drugs has been suggested in a wide range of human solid tumors, including OC. The present study assessed the in vitro cytostatic and cytotoxic effects of OTX015, a pan Bromodomain and Extra-Terminal motif inhibitor, in human OC cells, both as single treatment and in combination with radiotherapy. Cellular, molecular and computational network analyses indicated the centrality of GNL3 downregulation in mediating the OTX015-related antitumor efficacy that blocks disease progression/maintenance and radioresistance acquisition. Our preclinical results confirm that targeted and combinatorial treatments represent effective anticancer strategies to be translated in the clinical research for improving OC patient care. Ovarian cancer (OC) is the most aggressive gynecological tumor worldwide and, notwithstanding the increment in conventional treatments, many resistance mechanisms arise, this leading to cure failure and patient death. So, the use of novel adjuvant drugs able to counteract these pathways is urgently needed to improve patient overall survival. A growing interest is focused on epigenetic drugs for cancer therapy, such as Bromodomain and Extra-Terminal motif inhibitors (BETi). Here, we investigate the antitumor effects of OTX015, a novel BETi, as a single agent or in combination with ionizing radiation (IR) in OC cellular models. OTX015 treatment significantly reduced tumor cell proliferation by triggering cell cycle arrest and apoptosis that were linked to nucleolar stress and DNA damage. OTX015 impaired migration capacity and potentiated IR effects by reducing the expression of different drivers of cancer resistance mechanisms, including GNL3 gene, whose expression was found to be significantly higher in OC biopsies than in normal ovarian tissues. Gene specific knocking down and computational network analysis confirmed the centrality of GNL3 in OTX015-mediated OC antitumor effects. Altogether, our findings suggest OTX015 as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with OC. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
26. A MYC-Driven Plasma Polyamine Signature for Early Detection of Ovarian Cancer.
- Author
-
Fahrmann, Johannes F., Irajizad, Ehsan, Kobayashi, Makoto, Vykoukal, Jody, Dennison, Jennifer B., Murage, Eunice, Wu, Ranran, Long, James P., Do, Kim-Anh, Celestino, Joseph, Lu, Karen H., Lu, Zhen, Bast Jr., Robert C., Hanash, Samir, and Dinulescu, Daniela M.
- Subjects
OVARIAN tumors ,BLOOD plasma ,ONCOGENES ,EARLY detection of cancer ,AMINES ,MASS spectrometry ,TUMOR markers ,RECEIVER operating characteristic curves ,TUMOR antigens ,DATA analysis - Abstract
Simple Summary: There is a need for additional marker(s) to detect early-stage ovarian cancer that would augment the performance of CA125. Herein, we report a polyamine signature that is detected in the blood and that has value for detecting ovarian cancers at an early stage. The polyamine signature was able to complement CA125 in identifying more ovarian cancer cases that would have been missed by CA125 alone. Our validation of a polyamine signature provides compelling evidence for the value of blood polyamine metabolites as markers for ovarian cancer detection. MYC is an oncogenic driver in the pathogenesis of ovarian cancer. We previously demonstrated that MYC regulates polyamine metabolism in triple-negative breast cancer (TNBC) and that a plasma polyamine signature is associated with TNBC development and progression. We hypothesized that a similar plasma polyamine signature may associate with ovarian cancer (OvCa) development. Using mass spectrometry, four polyamines were quantified in plasma from 116 OvCa cases and 143 controls (71 healthy controls + 72 subjects with benign pelvic masses) (Test Set). Findings were validated in an independent plasma set from 61 early-stage OvCa cases and 71 healthy controls (Validation Set). Complementarity of polyamines with CA125 was also evaluated. Receiver operating characteristic area under the curve (AUC) of individual polyamines for distinguishing cases from healthy controls ranged from 0.74–0.88. A polyamine signature consisting of diacetylspermine + N-(3-acetamidopropyl)pyrrolidin-2-one in combination with CA125 developed in the Test Set yielded improvement in sensitivity at >99% specificity relative to CA125 alone (73.7% vs 62.2%; McNemar exact test 2-sided P: 0.019) in the validation set and captured 30.4% of cases that were missed with CA125 alone. Our findings reveal a MYC-driven plasma polyamine signature associated with OvCa that complemented CA125 in detecting early-stage ovarian cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
27. A Novel Salt Inducible Kinase 2 Inhibitor, ARN-3261, Sensitizes Ovarian Cancer Cell Lines and Xenografts to Carboplatin.
- Author
-
Fan, Dengxuan, Yang, Hailing, Mao, Weiqun, Rask, Philip J., Pang, Lan, Xu, Congjian, Vankayalapat, Hariprasad, Ahmed, Ahmed A., Bast Jr., Robert C., and Lu, Zhen
- Subjects
ANIMAL experimentation ,APOPTOSIS ,BIOLOGICAL assay ,CELL lines ,DNA ,GENE expression ,MICE ,OVARIAN tumors ,PROTEINS ,SALT ,XENOGRAFTS ,PROTEIN kinase inhibitors ,CARBOPLATIN ,IMMUNOCOMPROMISED patients ,PHARMACODYNAMICS - Abstract
Simple Summary: Carboplatin and paclitaxel constitute first-line treatment for ovarian cancer, producing tumor shrinkage in 70% of patients, but curing less than 20% with advanced stage disease. Previous studies have shown that treatment with ARN-3261, a small molecule inhibitor of the enzyme salt-induced kinase 2, can improve the response to paclitaxel in human ovarian cancer cells grown in culture and in immunocompromised mice. Here we have found that ARN-3261 also increases carboplatin's ability to kill ovarian cancer cells grown in culture and in immunocompromised mice, causing additional DNA damage and decreasing levels of survivin, a protein that protects cancer cells from programmed cell death. Our studies encourage clinical evaluation of ARN-3261 and a Phase I clinical trial has been initiated to test the drug in ovarian cancer patients. Salt-induced kinase 2 (SIK2) is a serine-threonine kinase that regulates centrosome splitting, activation of PI3 kinase and phosphorylation of class IIa HDACs, affecting gene expression. Previously, we found that inhibition of SIK2 enhanced sensitivity of ovarian cancer cells to paclitaxel. Carboplatin and paclitaxel constitute first-line therapy for most patients with ovarian carcinoma, producing a 70% clinical response rate, but curing <20% of patients with advanced disease. We have asked whether inhibition of SIK2 with ARN-3261 enhances sensitivity to carboplatin in ovarian cancer cell lines and xenograft models. ARN-3261-induced DNA damage and apoptosis were measured with γ-H2AX accumulation, comet assays, and annexin V. ARN-3261 inhibited growth of eight ovarian cancer cell lines at an IC50 of 0.8 to 3.5 µM. ARN-3261 significantly enhanced sensitivity to carboplatin in seven of eight ovarian cancer cell lines and a carboplatin-resistant cell line tested. Furthermore, ARN-3261 in combination with carboplatin produced greater inhibition of tumor growth than carboplatin alone in SKOv3 and OVCAR8 ovarian cancer xenograft models. ARN-3261 enhanced DNA damage and apoptosis by downregulating expression of survivin. Thus, a SIK2 kinase inhibitor enhanced carboplatin-induced therapy in preclinical models of ovarian cancer and deserves further evaluation in clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.