128 results on '"Dennison, Jennifer B"'
Search Results
2. Biomarker trajectory for earlier detection of lung cancer
- Author
-
Irajizad, Ehsan, Fahrmann, Johannes F., Toumazis, Iakovos, Vykoukal, Jody, Dennison, Jennifer B., Shen, Yu, Do, Kim-Anh, Ostrin, Edwin J., Feng, Ziding, and Hanash, Samir
- Published
- 2024
- Full Text
- View/download PDF
3. Modulating a prebiotic food source influences inflammation and immune-regulating gut microbes and metabolites: insights from the BE GONE trial
- Author
-
Zhang, Xiaotao, Irajizad, Ehsan, Hoffman, Kristi L., Fahrmann, Johannes F., Li, Fangyu, Seo, Yongwoo David, Browman, Gladys J., Dennison, Jennifer B., Vykoukal, Jody, Luna, Pamela N., Siu, Wesley, Wu, Ranran, Murage, Eunice, Ajami, Nadim J., McQuade, Jennifer L., Wargo, Jennifer A., Long, James P., Do, Kim-Anh, Lampe, Johanna W., Basen-Engquist, Karen M., Okhuysen, Pablo C., Kopetz, Scott, Hanash, Samir M., Petrosino, Joseph F., Scheet, Paul, and Daniel, Carrie R.
- Published
- 2023
- Full Text
- View/download PDF
4. A blood-based metabolomic signature predictive of risk for pancreatic cancer
- Author
-
Irajizad, Ehsan, Kenney, Ana, Tang, Tiffany, Vykoukal, Jody, Wu, Ranran, Murage, Eunice, Dennison, Jennifer B., Sans, Marta, Long, James P., Loftus, Maureen, Chabot, John A., Kluger, Michael D., Kastrinos, Fay, Brais, Lauren, Babic, Ana, Jajoo, Kunal, Lee, Linda S., Clancy, Thomas E., Ng, Kimmie, Bullock, Andrea, Genkinger, Jeanine M., Maitra, Anirban, Do, Kim-Anh, Yu, Bin, Wolpin, Brian M., Hanash, Sam, and Fahrmann, Johannes F.
- Published
- 2023
- Full Text
- View/download PDF
5. A polyamine-centric, blood-based metabolite panel predictive of poor response to CAR-T cell therapy in large B cell lymphoma
- Author
-
Fahrmann, Johannes F., Saini, Neeraj Y., Chia-Chi, Chang, Irajizad, Ehsan, Strati, Paolo, Nair, Ranjit, Fayad, Luis E., Ahmed, Sairah, Lee, Hun Ju, Iyer, Swaminathan, Steiner, Raphael, Vykoukal, Jody, Wu, Ranran, Dennison, Jennifer B., Nastoupil, Loretta, Jain, Preetesh, Wang, Michael, Green, Michael, Westin, Jason, Blumenberg, Viktoria, Davila, Marco, Champlin, Richard, Shpall, Elizabeth J., Kebriaei, Partow, Flowers, Christopher R., Jain, Michael, Jenq, Robert, Stein-Thoeringer, Christoph K., Subklewe, Marion, Neelapu, Sattva S., and Hanash, Sam
- Published
- 2022
- Full Text
- View/download PDF
6. CES2 sustains HNF4α expression to promote pancreatic adenocarcinoma progression through an epoxide hydrolase-dependent regulatory loop
- Author
-
Chen, Yihui, Capello, Michela, Rios Perez, Mayrim V., Vykoukal, Jody V., Roife, David, Kang, Ya'an, Prakash, Laura R., Katayama, Hiroyuki, Irajizad, Ehsan, Fleury, Alia, Ferri-Borgogno, Sammy, Baluya, Dodge L., Dennison, Jennifer B., Do, Kim-Anh, Fiehn, Oliver, Maitra, Anirban, Wang, Huamin, Chiao, Paul J., Katz, Matthew H.G., Fleming, Jason B., Hanash, Samir M., and Fahrmann, Johannes F.
- Published
- 2022
- Full Text
- View/download PDF
7. Lead-Time Trajectory of CA19-9 as an Anchor Marker for Pancreatic Cancer Early Detection
- Author
-
Fahrmann, Johannes F., Schmidt, C. Max, Mao, Xiangying, Irajizad, Ehsan, Loftus, Maureen, Zhang, Jinming, Patel, Nikul, Vykoukal, Jody, Dennison, Jennifer B., Long, James P., Do, Kim-Anh, Zhang, Jianjun, Chabot, John A., Kluger, Michael D., Kastrinos, Fay, Brais, Lauren, Babic, Ana, Jajoo, Kunal, Lee, Linda S., Clancy, Thomas E., Ng, Kimmie, Bullock, Andrea, Genkinger, Jeanine, Yip-Schneider, Michele T., Maitra, Anirban, Wolpin, Brian M., and Hanash, Samir
- Published
- 2021
- Full Text
- View/download PDF
8. Contribution of a Blood-Based Protein Biomarker Panel to the Classification of Indeterminate Pulmonary Nodules
- Author
-
Ostrin, Edwin J., Bantis, Leonidas E., Wilson, David O., Patel, Nikul, Wang, Renwei, Kundnani, Deepali, Adams-Haduch, Jennifer, Dennison, Jennifer B., Fahrmann, Johannes F., Chiu, Hsienchang Thomas, Gazdar, Adi, Feng, Ziding, Yuan, Jian-Min, and Hanash, Samir M.
- Published
- 2021
- Full Text
- View/download PDF
9. Validation of a Blood-Based Protein Biomarker Panel for a Risk Assessment of Lethal Lung Cancer in the Physicians' Health Study.
- Author
-
Song, Lulu, Irajizad, Ehsan, Rundle, Andrew, Sesso, Howard D., Gaziano, John Michael, Vykoukal, Jody V., Do, Kim-Anh, Dennison, Jennifer B., Ostrin, Edwin J., Fahrmann, Johannes F., Perera, Frederica, and Hanash, Samir
- Subjects
RISK assessment ,RECEIVER operating characteristic curves ,RESEARCH funding ,SMOKING ,LOGISTIC regression analysis ,TUMOR markers ,DESCRIPTIVE statistics ,LONGITUDINAL method ,LUNG tumors ,TUMOR antigens ,CONFIDENCE intervals ,DATA analysis software ,SENSITIVITY & specificity (Statistics) ,DISEASE risk factors - Abstract
Simple Summary: Improvements in lung cancer risk assessment to inform on the need for screening may be achieved through the use of biomarkers. Here, we report the findings of a validation study of a panel of four circulating protein biomarkers for the risk prediction of lung cancer in a cohort of pre-diagnostic plasmas obtained from the Physician's Health Study (PHS). We demonstrate that the protein panel can identify individuals at high risk of lung cancer up to two years prior to clinical diagnosis. This study aimed to assess a four-marker protein panel (4MP)'s performance, including the precursor form of surfactant protein B, cancer antigen 125, carcinoembryonic antigen, and cytokeratin-19, for predicting lung cancer in a cohort enriched with never- and ever-smokers. Blinded pre-diagnostic plasma samples collected within 2 years prior to a lung cancer diagnosis from 25 cases and 100 sex-, age-, and smoking-matched controls were obtained from the Physicians' Health Study (PHS). The 4MP yielded AUC performance estimates of 0.76 (95% CI: 0.61–0.92) and 0.69 (95% CI: 0.56–0.82) for predicting lung cancer within one year and within two years of diagnosis, respectively. When stratifying into ever-smokers and never-smokers, the 4MP had respective AUCs of 0.77 (95% CI: 0.63–0.92) and 0.72 (95% CI: 0.17–1.00) for a 1-year risk of lung cancer. The AUCs of the 4MP for predicting metastatic lung cancer within one year and two years of the blood draw were 0.95 (95% CI: 0.87–1.00) and 0.78 (95% CI: 0.62–0.94), respectively. Our findings indicate that a blood-based biomarker panel may be useful in identifying ever- and never-smokers at high risk of a diagnosis of lung cancer within one-to-two years. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Caveolin-1-mediated sphingolipid oncometabolism underlies a metabolic vulnerability of prostate cancer
- Author
-
Vykoukal, Jody, Fahrmann, Johannes F., Gregg, Justin R., Tang, Zhe, Basourakos, Spyridon, Irajizad, Ehsan, Park, Sanghee, Yang, Guang, Creighton, Chad J., Fleury, Alia, Mayo, Jeffrey, Paulucci-Holthauzen, Adriana, Dennison, Jennifer B., Murage, Eunice, Peterson, Christine B., Davis, John W., Kim, Jeri, Hanash, Samir, and Thompson, Timothy C.
- Published
- 2020
- Full Text
- View/download PDF
11. Contributions of the Microbiome-Derived Metabolome for Risk Assessment and Prognostication of Pancreatic Cancer.
- Author
-
León-Letelier, Ricardo A, Dou, Rongzhang, Vykoukal, Jody, Yip-Schneider, Michele T, Maitra, Anirban, Irajizad, Ehsan, Wu, Ranran, Dennison, Jennifer B, Do, Kim-An, Zhang, Jianjun, Schmidt, C Max, Hanash, Samir, and Fahrmann, Johannes F
- Published
- 2024
- Full Text
- View/download PDF
12. Blood-based Proteomic Signatures Associated With MENI-related Duodenopancreatic Neuroendocrine Tumor Progression.
- Author
-
Fahrmann, Johannes F., Wasylishen, Amanda R., Pieterman, Carolina R. C., Irajizad, Ehsan, Vykoukal, Jody, Wu, Ranran, Dennison, Jennifer B., Peterson, Christine B., Hua Zhao, Kim-Anh Do, Halperin, Daniel M., Agarwal, Sunita K., Blau, Jenny E., Jha, Smita, Del Rivero, Jaydira, Nilubol, Naris, Walter, Mary F., Welch, James M., Weinstein, Lee S., and Vriens, Menno R.
- Subjects
NEUROENDOCRINE tumors ,MASS spectrometry ,DISEASE progression - Abstract
Purpose: Patients with multiple endocrine neoplasia type 1 (MEN1) are predisposed to develop duodenopancreatic neuroendocrine tumors (dpNETs), and metastatic dpNET is the primary cause of disease-related mortality. Presently, there is a paucity of prognostic factors that can reliably identify patients with MEN1-related dpNETS who are at high risk of distant metastasis. In the current study, we aimed to establish novel circulating molecular protein signatures associated with disease progression. Experimental Design: Mass spectrometry-based proteomic profiling was conducted on plasmas procured through an international collaboration between MD Anderson Cancer Center, the National Institutes of Health, and the University Medical Center Utrecht from a cohort of 56 patients with MEN1 [14 with distant metastasis dpNETs (cases) and 42 with either indolent dpNETs or no dpNETs (controls)]. Findings were compared to proteomic profiles generated from serially collected plasmas from a mouse model of Men1-pancreatic neuroendocrine tumors (Men1
fl/fl Pdx1-CreTg ) and control mice (Men1fl/fl ). Results: A total of 187 proteins were found to be elevated in MEN1 patients with distant metastasis compared to controls, including 9 proteins previously associated with pancreatic cancer and other neuronal proteins. Analyses of mouse plasmas revealed 196 proteins enriched for transcriptional targets of oncogenic MYCN, YAP1, POU5F1, and SMAD that were associated with disease progression in Men1fl/fl Pdx1-CreTg mice. Cross-species intersection revealed 19 proteins positively associated with disease progression in both human patients and in Men1fl/fl Pdx1-CreTg mice. Conclusions: Our integrated analyses identified novel circulating protein markers associated with disease progression in MEN1-related dpNET. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
13. Mortality Benefit of a Blood-Based Biomarker Panel for Lung Cancer on the Basis of the Prostate, Lung, Colorectal, and Ovarian Cohort.
- Author
-
Irajizad, Ehsan, Fahrmann, Johannes F., Marsh, Tracey, Vykoukal, Jody, Dennison, Jennifer B., Long, James P., Do, Kim-Anh, Feng, Ziding, Hanash, Samir, and Ostrin, Edwin J.
- Published
- 2023
- Full Text
- View/download PDF
14. Chlorinated adenosine analogue induces AMPK and autophagy in chronic lymphocytic leukaemia cells during therapy
- Author
-
Stellrecht, Christine M., Chen, Lisa S., Ayres, Mary L., Dennison, Jennifer B., Shentu, Shujun, Chen, Yuling, Keating, Michael J., Wierda, William G., and Gandhi, Varsha
- Published
- 2017
- Full Text
- View/download PDF
15. Role of CPS1 in Cell Growth, Metabolism, and Prognosis in LKB1-Inactivated Lung Adenocarcinoma
- Author
-
Çeliktaş, Müge, Tanaka, Ichidai, Chandra Tripathi, Satyendra, Fahrmann, Johannes F., Aguilar-Bonavides, Clemente, Villalobos, Pamela, Delgado, Oliver, Dhillon, Dilsher, Dennison, Jennifer B., Ostrin, Edwin J., Wang, Hong, Behrens, Carmen, Do, Kim-Anh, Gazdar, Adi F., Hanash, Samir M., and Taguchi, Ayumu
- Published
- 2017
- Full Text
- View/download PDF
16. Kynureninase Upregulation Is a Prominent Feature of NFR2-Activated Cancers and Is Associated with Tumor Immunosuppression and Poor Prognosis.
- Author
-
León-Letelier, Ricardo A., Abdel Sater, Ali H., Chen, Yihui, Park, Soyoung, Wu, Ranran, Irajizad, Ehsan, Dennison, Jennifer B., Katayama, Hiroyuki, Vykoukal, Jody V., Hanash, Samir, Ostrin, Edwin J., and Fahrmann, Johannes F.
- Subjects
GENETIC mutation ,IMMUNOSUPPRESSION ,GENES ,MESSENGER RNA ,GENE expression profiling ,SURVIVAL analysis (Biometry) ,RESEARCH funding ,TUMORS ,TRANSCRIPTION factors ,PROPORTIONAL hazards models ,OVERALL survival - Abstract
Simple Summary: We previously reported a novel mechanism of NRF2 tumoral immune suppression through selective upregulation of the tryptophan-metabolizing enzyme kynureninase (KYNU) in lung adenocarcinoma. In the current study, we report that elevated tumoral KYNU gene expression is broadly associated with NRF2-activated cancers, including pancreatic adenocarcinoma (PDAC). Mechanistic studies confirmed that NRF2 regulates the expression of enzymatically functional KYNU in PDAC cells. We further demonstrated that elevated tumoral KYNU mRNA expression is associated with an immunosuppressive tumor microenvironment across several cancer types and is prognostic for poor overall survival in thymoma, acute myeloid leukemia, low-grade glioma, kidney renal papillary cell carcinoma, stomach adenocarcinoma, and PDAC. Our study supports the significance of KYNU as a tumor marker of tumor immunosuppression and as a multi-cancer prognostic marker for poor overall survival. The nuclear factor erythroid 2-related factor 2 (NRF2) pathway is frequently activated in various cancer types. Aberrant activation of NRF2 in cancer is attributed to gain-of-function mutations in the NRF2-encoding gene NFE2L2 or a loss of function of its suppressor, Kelch-like ECH-associated protein 1 (KEAP1). NRF2 activation exerts pro-tumoral effects in part by altering cancer cell metabolism. Previously, we reported a novel mechanism of NRF2 tumoral immune suppression through the selective upregulation of the tryptophan-metabolizing enzyme kynureninase (KYNU) in lung adenocarcinoma. In the current study, we explored the relevance of NRF2-mediated KYNU upregulation across multiple cancer types. Specifically, using a gene expression dataset for 9801 tumors representing 32 cancer types from The Cancer Genome Atlas (TCGA), we demonstrated that elevated KYNU parallels increased gene-based signatures of NRF2-activation and that elevated tumoral KYNU mRNA expression is strongly associated with an immunosuppressive tumor microenvironment, marked by high expression of gene-based signatures of Tregs as well as the immune checkpoint blockade-related genes CD274 (PDL-1), PDCD1 (PD-1), and CTLA4, regardless of the cancer type. Cox proportional hazard models further revealed that increased tumoral KYNU gene expression was prognostic for poor overall survival in several cancer types, including thymoma, acute myeloid leukemia, low-grade glioma, kidney renal papillary cell carcinoma, stomach adenocarcinoma, and pancreatic ductal adenocarcinoma (PDAC). Using PDAC as a model system, we confirmed that siRNA-mediated knockdown of NRF2 reduced KYNU mRNA expression, whereas activation of NFE2L2 (the coding gene for NRF2) through either small-molecule agonists or siRNA-mediated knockdown of KEAP1 upregulated KYNU in PDAC cells. Metabolomic analyses of the conditioned medium from PDAC cell lines revealed elevated levels of KYNU-derived anthranilate, confirming that KYNU was enzymatically functional. Collectively, our study highlights the activation of the NRF2–KYNU axis as a multi-cancer phenomenon and supports the relevance of tumoral KYNU as a marker of tumor immunosuppression and as a prognostic marker for poor overall survival. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. c-MYC-Driven Polyamine Metabolism in Ovarian Cancer: From Pathogenesis to Early Detection and Therapy.
- Author
-
Chen, Yihui, León-Letelier, Ricardo A., Abdel Sater, Ali Hussein, Vykoukal, Jody, Dennison, Jennifer B., Hanash, Samir, and Fahrmann, Johannes F.
- Subjects
BIOCHEMISTRY ,DISEASE progression ,OVARIAN tumors ,PHENOMENOLOGICAL biology ,CARCINOGENESIS ,EARLY detection of cancer ,AMINES ,CELLULAR signal transduction ,CELL proliferation - Abstract
Simple Summary: This review informs on how the MYC signaling is dysregulated and participates in ovarian cancer progression, and the unmet challenge to directly target MYC for ovarian cancer treatment. Therefore, we proposed to alternatively target essential downstream polyamine metabolism pathway of MYC. In this review we include the metabolism of polyamine, the regulation of polyamine metabolism by MYC signaling, the utility of polyamine as therapeutic targets and cancer biomarkers. c-MYC and its paralogues MYCN and MYCL are among the most frequently amplified and/or overexpressed oncoproteins in ovarian cancer. c-MYC plays a key role in promoting ovarian cancer initiation and progression. The polyamine pathway is a bona fide target of c-MYC signaling, and polyamine metabolism is strongly intertwined with ovarian malignancy. Targeting of the polyamine pathway via small molecule inhibitors has garnered considerable attention as a therapeutic strategy for ovarian cancer. Herein, we discuss the involvement of c-MYC signaling and that of its paralogues in promoting ovarian cancer tumorigenesis. We highlight the potential of targeting c-MYC-driven polyamine metabolism for the treatment of ovarian cancers and the utility of polyamine signatures in biofluids for early detection applications. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. 8-Aminoadenosine inhibits Akt/mTOR and Erk signaling in mantle cell lymphoma
- Author
-
Dennison, Jennifer B., Shanmugam, Mala, Ayres, Mary L., Qian, Jun, Krett, Nancy L., Medeiros, L. Jeffrey, Neelapu, Sattva S., Rosen, Steven T., and Gandhi, Varsha
- Published
- 2010
- Full Text
- View/download PDF
19. Deep Learning Models for Automated Assessment of Breast Density Using Multiple Mammographic Image Types.
- Author
-
Rigaud, Bastien, Weaver, Olena O., Dennison, Jennifer B., Awais, Muhammad, Anderson, Brian M., Chiang, Ting-Yu D., Yang, Wei T., Leung, Jessica W. T., Hanash, Samir M., and Brock, Kristy K.
- Subjects
DEEP learning ,RELIABILITY (Personality trait) ,MEDICAL information storage & retrieval systems ,BREAST physiology ,MAMMOGRAMS ,RETROSPECTIVE studies ,MEDICAL screening ,CLINICAL medicine ,QUESTIONNAIRES ,DATA analysis software - Abstract
Simple Summary: The DL model predictions in automated breast density assessment were independent of the imaging technologies, moderately or substantially agreed with the clinical reader density values, and had improved performance as compared to inclusion of commercial software values. Recently, convolutional neural network (CNN) models have been proposed to automate the assessment of breast density, breast cancer detection or risk stratification using single image modality. However, analysis of breast density using multiple mammographic types using clinical data has not been reported in the literature. In this study, we investigate pre-trained EfficientNetB0 deep learning (DL) models for automated assessment of breast density using multiple mammographic types with and without clinical information to improve reliability and versatility of reporting. 120,000 for-processing and for-presentation full-field digital mammograms (FFDM), digital breast tomosynthesis (DBT), and synthesized 2D images from 5032 women were retrospectively analyzed. Each participant underwent up to 3 screening examinations and completed a questionnaire at each screening encounter. Pre-trained EfficientNetB0 DL models with or without clinical history were optimized. The DL models were evaluated using BI-RADS (fatty, scattered fibroglandular densities, heterogeneously dense, or extremely dense) versus binary (non-dense or dense) density classification. Pre-trained EfficientNetB0 model performances were compared using inter-observer and commercial software (Volpara) variabilities. Results show that the average Fleiss' Kappa score between-observers ranged from 0.31–0.50 and 0.55–0.69 for the BI-RADS and binary classifications, respectively, showing higher uncertainty among experts. Volpara-observer agreement was 0.33 and 0.54 for BI-RADS and binary classifications, respectively, showing fair to moderate agreement. However, our proposed pre-trained EfficientNetB0 DL models-observer agreement was 0.61–0.66 and 0.70–0.75 for BI-RADS and binary classifications, respectively, showing moderate to substantial agreement. Overall results show that the best breast density estimation was achieved using for-presentation FFDM and DBT images without added clinical information. Pre-trained EfficientNetB0 model can automatically assess breast density from any images modality type, with the best results obtained from for-presentation FFDM and DBT, which are the most common image archived in clinical practice. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. Novel UHRF1-MYC Axis in Acute Lymphoblastic Leukemia.
- Author
-
Park, Soyoung, Sater, Ali H. Abdel, Fahrmann, Johannes F., Irajizad, Ehsan, Cai, Yining, Katayama, Hiroyuki, Vykoukal, Jody, Kobayashi, Makoto, Dennison, Jennifer B., Garcia-Manero, Guillermo, Mullighan, Charles G., Gu, Zhaohui, Konopleva, Marina, and Hanash, Samir
- Subjects
PROTEINS ,EXPERIMENTAL design ,B cells ,LYMPHOBLASTIC leukemia ,GENE expression ,IMMUNOBLOTTING ,CELL survival ,DNA-binding proteins ,CELL proliferation ,T cells ,CELL lines ,POLYMERASE chain reaction - Abstract
Simple Summary: We provided evidence that ubiquitin-like, containing PHD and RING finger domain, 1 (UHRF1) is overexpressed in acute lymphocytic leukemia (ALL). We further showed that UHRF1 directly interacts and regulates c-Myc expression to enable ALL cell growth through the cMYC-CDK4/6 phosphoRb-signaling axis. Ubiquitin-like, containing PHD and RING finger domain, (UHRF) family members are overexpressed putative oncogenes in several cancer types. We evaluated the protein abundance of UHRF family members in acute leukemia. A marked overexpression of UHRF1 protein was observed in ALL compared with AML. An analysis of human leukemia transcriptomic datasets revealed concordant overexpression of UHRF1 in B-Cell and T-Cell ALL compared with CLL, AML, and CML. In-vitro studies demonstrated reduced cell viability with siRNA-mediated knockdown of UHRF1 in both B-ALL and T-ALL, associated with reduced c-Myc protein expression. Mechanistic studies indicated that UHRF1 directly interacts with c-Myc, enabling ALL expansion via the CDK4/6-phosphoRb axis. Our findings highlight a previously unknown role of UHRF1 in regulating c-Myc protein expression and implicate UHRF1 as a potential therapeutic target in ALL. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. Contributions of Circulating microRNAs for Early Detection of Lung Cancer.
- Author
-
Vykoukal, Jody, Fahrmann, Johannes F., Patel, Nikul, Shimizu, Masayoshi, Ostrin, Edwin J., Dennison, Jennifer B., Ivan, Cristina, Goodman, Gary E., Thornquist, Mark D., Barnett, Matt J., Feng, Ziding, Calin, George A., and Hanash, Samir M.
- Subjects
PROTEINS ,BIOMARKERS ,CONFIDENCE intervals ,LUNG tumors ,EARLY detection of cancer ,MICRORNA ,MANN Whitney U Test ,COMPARATIVE studies ,SENSITIVITY & specificity (Statistics) ,TUMOR antigens - Abstract
Simple Summary: Blood-based cancer biomarkers are a minimally invasive means to achieve improved assessment of risk and/or earlier detection of lung cancer. Using serum samples from individuals that went on to develop lung cancer within one year following blood draw and from matched controls, we investigated 30 microRNAs for individual and combined utility to discriminate lung cancer cases from controls. We additionally assessed the contributions of top-performing microRNA candidates for improving on the performance of a previously validated four-protein marker panel for lung cancer detection. The results of this study indicate complementary performance of combining circulating microRNA and the four-protein-marker panel in assays for early detection of lung cancer. There is unmet need to develop circulating biomarkers that would enable earlier interception of lung cancer when more effective treatment options are available. Here, a set of 30 miRNAs, selected from a review of the published literature were assessed for their predictive performance in identifying lung cancer cases in the pre-diagnostic setting. The 30 miRNAs were assayed using sera collected from 102 individuals diagnosed with lung cancer within one year following blood draw and 212 controls matched for age, sex, and smoking status. The additive performance of top-performing miRNA candidates in combination with a previously validated four-protein marker panel (4MP) consisting of the precursor form of surfactant protein B (Pro-SFTPB), cancer antigen 125 (CA125), carcinoembryonic antigen (CEA) and cytokeratin-19 fragment (CYFRA21-1) was additionally assessed. Of the 30 miRNAs evaluated, five (miR-320a-3p, miR-210-3p, miR-92a-3p, miR-21-5p, and miR-140-3p) were statistically significantly (Wilcoxon rank sum test p < 0.05) elevated in case sera compared to controls, with individual AUCs ranging from 0.57–0.62. Compared to the 4MP alone, the combination of 3-miRNAs + 4MP improved sensitivity at 95% specificity by 19.1% ((95% CI of difference 0.0–28.6); two-sided p: 0.006). Our findings demonstrate utility for miRNAs for early detection of lung cancer in combination with a four-protein marker panel. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. A Comprehensive Search of Non-Canonical Proteins in Non-Small Cell Lung Cancer and Their Impact on the Immune Response.
- Author
-
Irajizad, Ehsan, Fahrmann, Johannes F., Long, James P., Vykoukal, Jody, Kobayashi, Makoto, Capello, Michela, Yu, Chuan-Yih, Cai, Yining, Hsiao, Fu Chung, Patel, Nikul, Park, Soyoung, Peng, Qian, Dennison, Jennifer B., Kato, Taketo, Tai, Mei Chee, Taguchi, Ayumu, Kadara, Humam, Wistuba, Ignacio I., Katayama, Hiroyuki, and Do, Kim-Anh
- Subjects
NON-small-cell lung carcinoma ,IMMUNE response ,AUTOANTIBODIES ,FRAMESHIFT mutation ,PROTEINS ,NON-coding RNA - Abstract
There is substantial interest in mining neoantigens for cancer applications. Non-canonical proteins resulting from frameshift mutations have been identified as neoantigens in cancer. We investigated the landscape of non-canonical proteins in non-small cell lung cancer (NSCLC) and their induced immune response in the form of autoantibodies. A database of cryptoproteins was computationally constructed and comprised all alternate open reading frames (altORFs) and ORFs identified in pseudogenes, noncoding RNAs, and untranslated regions of mRNAs that did not align with known canonical proteins. Proteomic profiles of seventeen lung adenocarcinoma (LUAD) cell lines were searched to evaluate the occurrence of cryptoproteins. To assess the immunogenicity, immunoglobulin (Ig)-bound cryptoproteins in plasmas were profiled by mass spectrometry. The specimen set consisted of plasmas from 30 newly diagnosed NSCLC cases, pre-diagnostic plasmas from 51 NSCLC cases, and 102 control plasmas. An analysis of LUAD cell lines identified 420 cryptoproteins. Plasma Ig-bound analyses revealed 90 cryptoproteins uniquely found in cases and 14 cryptoproteins that had a fold-change >2 compared to controls. In pre-diagnostic samples, 17 Ig-bound cryptoproteins yielded an odds ratio ≥2. Eight Ig-bound cryptoproteins were elevated in both pre-diagnostic and newly diagnosed cases compared to controls. Cryptoproteins represent a class of neoantigens that induce an autoantibody response in NSCLC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. Mutational Activation of the NRF2 Pathway Upregulates Kynureninase Resulting in Tumor Immunosuppression and Poor Outcome in Lung Adenocarcinoma.
- Author
-
Fahrmann, Johannes F., Tanaka, Ichidai, Irajizad, Ehsan, Mao, Xiangying, Dennison, Jennifer B., Murage, Eunice, Casabar, Julian, Mayo, Jeffrey, Peng, Qian, Celiktas, Muge, Vykoukal, Jody V., Park, Soyoung, Taguchi, Ayumu, Delgado, Oliver, Tripathi, Satyendra C., Katayama, Hiroyuki, Soto, Luisa Maren Solis, Rodriguez-Canales, Jaime, Behrens, Carmen, and Wistuba, Ignacio
- Subjects
ADENOCARCINOMA ,LUNG cancer ,EVALUATION of medical care ,GENETIC mutation ,IMMUNOSUPPRESSION ,CELLULAR signal transduction ,TUMOR markers - Abstract
Simple Summary: Activation of the Nuclear factor-erythroid factor 2-related factor 2 (NRF2) pathway through gain-of-function mutations or loss-of-function of its suppressor Kelch-like ECH-associated protein 1 (KEAP1) is frequent in lung cancer. NRF2 activation has also been reported to alter the tumor microenvironment. Proteomic profiles of 47 lung adenocarcinoma (LUAD) cell lines (11 KEAP1 mutant and 36 KEAP1 wild-type) revealed the tryptophan-kynurenine enzyme kynureninase (KYNU) as a top overexpressed protein associated with activated NRF2. Mechanistic studies demonstrated that NRF2 is a regulator of enzymatically functional KYNU in LUAD. Analysis of multiple independent gene expression datasets of human lung cancer and a LUAD tumor microarray demonstrated that elevated tumor KYNU expression was associated with immunosuppression, including potent induction of T-regulatory cells, increased levels of PD1 and PD-L1, and poorer overall survival. Our findings indicate a novel mechanism of NRF2 tumoral immunosuppression through upregulation of KYNU. Activation of the NRF2 pathway through gain-of-function mutations or loss-of-function of its suppressor KEAP1 is a frequent finding in lung cancer. NRF2 activation has been reported to alter the tumor microenvironment. Here, we demonstrated that NRF2 alters tryptophan metabolism through the kynurenine pathway that is associated with a tumor-promoting, immune suppressed microenvironment. Specifically, proteomic profiles of 47 lung adenocarcinoma (LUAD) cell lines (11 KEAP1 mutant and 36 KEAP1 wild-type) revealed the tryptophan-kynurenine enzyme kynureninase (KYNU) as a top overexpressed protein associated with activated NRF2. The siRNA-mediated knockdown of NFE2L2, the gene encoding for NRF2, or activation of the NRF2 pathway through siRNA-mediated knockdown of KEAP1 or via chemical induction with the NRF2-activator CDDO-Me confirmed that NRF2 is a regulator of KYNU expression in LUAD. Metabolomic analyses confirmed KYNU to be enzymatically functional. Analysis of multiple independent gene expression datasets of LUAD, as well as a LUAD tumor microarray demonstrated that elevated KYNU was associated with immunosuppression, including potent induction of T-regulatory cells, increased levels of PD1 and PD-L1, and resulted in poorer survival. Our findings indicate a novel mechanism of NRF2 tumoral immunosuppression through upregulation of KYNU. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. Blood-Based Biomarker Panel for Personalized Lung Cancer Risk Assessment.
- Author
-
Fahrmann, Johannes F, Marsh, Tracey, Irajizad, Ehsan, Patel, Nikul, Murage, Eunice, Vykoukal, Jody, Dennison, Jennifer B, Do, Kim-Anh, Ostrin, Edwin, Spitz, Margaret R, Lam, Stephen, Shete, Sanjay, Meza, Rafael, Tammemägi, Martin C, Feng, Ziding, and Hanash, Samir M
- Published
- 2022
- Full Text
- View/download PDF
25. Preclinical activity of 8-chloroadenosine with mantle cell lymphoma: roles of energy depletion and inhibition of DNA and RNA synthesis
- Author
-
Dennison, Jennifer B., Balakrishnan, Kumudha, and Gandhi, Varsha
- Published
- 2009
- Full Text
- View/download PDF
26. A Blood-based Polyamine Signature Associated With MEN1 Duodenopancreatic Neuroendocrine Tumor Progression.
- Author
-
Fahrmann, Johannes F., Wasylishen, Amanda R., Pieterman, Carolina R. C., Irajizad, Ehsan, Vykouka, Jody, Murage, Eunice, Wu, Ranran, Dennison, Jennifer B., Krishna, Hansini, Peterson, Christine B., Lozano, Guillermina, Hua Zhao, Kim-Anh Do, Halperin, Daniel M., Agarwa, Sunita K., Blau, Jenny E., Del Rivero, Jaydira, Nilubol, Naris, Walter, Mary F., and Welch, James M.
- Subjects
POLYAMINES ,PANCREATICODUODENECTOMY ,NEUROENDOCRINE tumors - Abstract
Context: Duodenopancreatic neuroendocrine tumors (dpNETs) frequently occur in patients with multiple endocrine neoplasia type 1 (MEN1), and metastatic dpNET is the primary cause of disease-related mortality. There is a need for biomarkers that can identify patients with MEN1-related dpNETs that are at high risk of developing distant metastasis. Polyamines have tumor-promoting roles in several cancer types.Objective: We hypothesized that MEN1-dpNET-related disease progression is associated with elevated levels of circulating polyamines.Methods: Through an international collaboration between The University of Texas MD Anderson Cancer Center, the National Institutes of Health, and the University Medical Center Utrecht, plasma polyamine levels were assessed using mass spectrometry in 84 patients with MEN1 (20 with distant metastatic dpNETs [patients] and 64 with either indolent dpNETs or no dpNETs [controls]). A mouse model of MEN1-pNET, Men1fl/flPdx1-CreTg, was used to test time-dependent changes in plasma polyamines associated with disease progression.Results: A 3-marker plasma polyamine signature (3MP: N-acetylputrescine, acetylspermidine, and diacetylspermidine) distinguished patients with metastatic dpNETs from controls in an initial set of plasmas from the 3 participating centers. The fixed 3MP yielded an area under the curve of 0.84 (95% CI, 0.62-1.00) with 66.7% sensitivity at 95% specificity for distinguishing patients from controls in an independent test set from MDACC. In Men1fl/flPdx1-CreTg mice, the 3MP was elevated early and remained high during disease progression.Conclusion: Our findings provide a basis for prospective testing of blood-based polyamines as a potential means for monitoring patients with MEN1 for harboring or developing aggressive disease. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
27. Quantification of Vincristine and its Major Metabolite in Human Plasma by High-Performance Liquid Chromatography/Tandem Mass Spectrometry
- Author
-
Dennison, Jennifer B, Renbarger, Jamie L, Walterhouse, David O, Jones, David R, and Hall, Stephen D
- Published
- 2008
- Full Text
- View/download PDF
28. Association Between Plasma Diacetylspermine and Tumor Spermine Synthase With Outcome in Triple-Negative Breast Cancer.
- Author
-
Fahrmann, Johannes F, Vykoukal, Jody, Fleury, Alia, Tripathi, Satyendra, Dennison, Jennifer B, Murage, Eunice, Wang, Peng, Yu, Chuan-Yih, Capello, Michela, Creighton, Chad J, Do, Kim-Anh, Long, James P, Irajizad, Ehsan, Peterson, Christine, Katayama, Hiroyuki, Disis, Mary L, Arun, Banu, and Hanash, Samir
- Subjects
SPERMINE ,FISHER exact test ,PROPORTIONAL hazards models ,ORNITHINE decarboxylase ,PROGESTERONE receptors ,RESEARCH ,LIQUID chromatography ,ANIMAL experimentation ,RESEARCH methodology ,PROGNOSIS ,METASTASIS ,EVALUATION research ,MEDICAL cooperation ,AMINES ,TUMOR classification ,GENE expression ,COMPARATIVE studies ,TRANSFERASES ,MASS spectrometry ,RESEARCH funding ,BREAST tumors ,MICE - Abstract
Background: MYC is an oncogenic driver of development and progression in triple-negative breast cancer (TNBC). Ornithine decarboxylase, the rate-limiting enzyme in polyamine metabolism, is a transcriptional target of MYC. We therefore hypothesized that a plasma polyamine signature may be predictive of TNBC development and progression.Methods: Using liquid chromatography mass spectrometry, polyamine levels were determined in plasma samples from newly diagnosed patients with TNBC (n = 87) and cancer-free controls (n = 115). Findings were validated in plasma samples from an independent prospective cohort of 54 TNBC, 55 estrogen receptor negative (ER-) and progesterone receptor negative (PR-) and HER2 positive (HER2+), and 73 ER+ case patients, and 30 cancer-free control subjects. Gene expression data and clinical data for 921 and 2359 breast cancer tumors were obtained from The Cancer Genome Atlas repository and the Oncomine database, respectively. Relationships between plasma diacetylspermine (DAS) and tumor spermine synthase (SMS) mRNA expression with metastasis-free survival and overall survival were determined using Cox proportional hazard models; Fisher exact tests were used to assess risk of distant metastasis in relation to tumor SMS mRNA expression.Results: An increase in plasma DAS, a catabolic product of spermine mediated through SMS, was observed in the TNBC subtype of breast cancer. Plasma levels of DAS in TNBC associated with increased risk of metastasis (plasma DAS value ≥ 1.16, hazard ratio = 3.06, 95% confidence interval [CI] = 1.15 to 8.13, two-sided P = .03). SMS mRNA expression in TNBC tumor tissue was also found to be predictive of poor overall survival (top 25th percentile hazard ratio = 2.06, 95% CI = 1.04 to 4.08, one-sided P = .04) and increased risk of distant metastasis in TNBC (comparison of lowest SMS quartile [reference] to highest SMS quartile relative risk = 1.90, 95% CI = 0.97 to 4.06, one-sided Fisher exact test P=.03).Conclusions: Metabolomic profiling identified plasma DAS as a predictive marker for TNBC progression and metastasis. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
29. CES2 Expression in Pancreatic Adenocarcinoma Is Predictive of Response to Irinotecan and Is Associated With Type 2 Diabetes.
- Author
-
Capello, Michela, Fahrmann, Johannes F., Rios Perez, Mayrim V., Vykoukal, Jody V., Irajizad, Ehsan, Tripathi, Satyendra C., Roife, David, Bantis, Leonidas E., Kang, Ya'an, Kundnani, Deepali L., Xu, Hanwen, Prakash, Laura R., Long, James P., Katayama, Hiroyuki, Fleury, Alia, Ferri-Borgogno, Sammy, Baluya, Dodge L., Dennison, Jennifer B., Aguilar-Bonavides, Clemente, and Casabar, Julian P.
- Subjects
TYPE 2 diabetes ,HEPATOCYTE nuclear factors ,WESTERN immunoblotting ,ADENOCARCINOMA ,PROTEIN expression ,PANCREATIC tumors - Abstract
PURPOSE: The combination chemotherapy of fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) has provided clinically meaningful improvement for pancreatic ductal adenocarcinoma (PDAC). We previously uncovered a role for the serine hydrolase carboxylesterase 2 (CES2) in mediating intratumoral activation of the prodrug irinotecan, a constituent of FOLFIRINOX. We aimed to further test the predictive value of CES2 for response to irinotecan using patient-derived xenograft (PDX) models and to elucidate the determinants of CES2 expression and response to FOLFIRINOX treatment among patients with PDAC. METHODS: PDXs were engrafted subcutaneously into nude mice and treated for 4 weeks with either saline control or irinotecan. CES2 and hepatocyte nuclear factor 4 alpha (HNF4A) expression in PDAC tissues was evaluated by immunohistochemical and Western blot analysis. Kaplan-Meier and Cox regression analyses were applied to assess the association between overall survival and hemoglobin A1C (HbA1C) levels in patients who underwent neoadjuvant FOLFIRINOX treatment. RESULTS: High CES2 activity in PDAC PDXs was associated with increased sensitivity to irinotecan. Integrated gene expression, proteomic analyses, and in vitro genetic experiments revealed that nuclear receptor HNF4A, which is upregulated in diabetes, is the upstream transcriptional regulator of CES2 expression. Elevated CES2 protein expression in PDAC tissues was positively associated with a history of type 2 diabetes (odds ratio, 4.84; P =.02). High HbA1C levels were associated with longer overall survival in patients who received neoadjuvant FOLFIRINOX treatment (P =.04). CONCLUSION: To our knowledge, we provide, for the first time, evidence that CES2 expression is associated with a history of type 2 diabetes in PDAC and that elevated HbA1C, by predicting tumor CES2 expression, may represent a novel marker for stratifying patients most likely to respond to FOLFIRINOX therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
30. Comprehensive Molecular Portraits of Invasive Lobular Breast Cancer
- Author
-
Gehlenborg, Nils, Mills, Gordon B., Ramirez, Nilsa C., Mungall, Andrew J., Zhang, Hailei, Schultz, Nikolaus, Ding, Li, Wang, Zhining, Sofia, Heidi J., Felau, Ina, Murray, Bradley A., Wiznerowicz, MacIej, Brooks, Denise, Yang, Liming, Shriver, Craig, Chin, Lynda, Jefferys, Stuart R., Perou, Charles M., Gibson, William J., Holbrook, Andrea, Jones, Steven J.M., Shi, Yan, Gatza, Michael L., Tarnuzzer, Roy, Roach, Jeffrey, Gabriel, Stacey B., Shaw, Kenna R.Mills, Beroukhim, Rameen, Robertson, A. Gordon, Hoyle, Alan P., Lolla, Laxmi, Holt, Robert A., Olopade, Olufunmilayo, Wu, Junyuan, Zack, Travis I., Davidsen, Tanja, Naresh, Rashi, Kim, Jaegil, Liu, Wenbin, Gastier-Foster, Julie M., Liu, Jia, Pastore, Alessandro, Wilson, Richard, Frazer, Scott, Benz, Christopher, Huang, Mei, Schein, Jacqueline E., Kandoth, Cyriac, Rhie, Suhn K., Soloway, Matthew G., Hwang, E. Shelley, Boice, Lori, Cho, Juok, Meyerson, Matthew, Benz, Stephen, Saksena, Gordon, Fieldhouse, Robert, Hu, Hai, Leraas, Kristen M., Laird, Peter W., King, Tari A., Getz, Gad, Ju, Zhenlin, Johnson, Nicole B., McAllister, Cynthia, Pihl, Todd, Marks, Jeffrey, Sun, Charlie, Wise, Lisa, Ling, Shiyun, Lichtenberg, Tara M., Barr, Thomas, Balasundaram, Miruna, Gerken, Mark, Weinstein, John N., Lester, Susan C., Weisenberger, Daniel J., Tse, Gary M.K., Zhang, Jiashan, Marra, Marco A., Shen, Hui, Sander, Chris, Chudamani, Sudha, Zmuda, Erik, Parker, Joel S., Collins, Laura C., Tiezzi, Daniel G., Hutter, Carolyn M., Zenklusen, Jean C., Sougnez, Carrie, Lawrence, Michael S., Lu, Yiling, Bowlby, Reanne, Robertson, Gordon, Moore, Richard A., Simons, Janae V., Bootwalla, Moiz S., Meng, Shaowu, Cherniack, Andrew D., Shih, Juliann, Hoadley, Katherine A., Berrios, Mario, Auman, J. Todd, Akbani, Rehan, Beck, Andrew H., McLellan, Michael, Demchok, John A., Maglinte, Dennis T., Schumacher, Steven E., Lai, Phillip H., Bodenheimer, Tom, Allison, Kimberly H., Bowen, Jay, Yau, Christina, Gao, Jian Jiong, Ferguson, Martin L., Mose, Lisle E., Sheth, Margi, Jensen, Kristin, Mardis, Elaine, Van Den Berg, David J., Voet, Doug, Heiman, David I., Hayes, D. Neil, Oesterreich, Steffi, Rathmell, W. Kimryn, Wilkerson, Matthew D., Lin, Pei, Ma, Yussanne, Hayat, Sikander, Balu, Saianand, Noble, Michael S., Shelley, Carl Simon, Chen, Yunn Yi, Factor, Rachel E., Wu, Ye, Wan, Yunhu, Ciriello, Giovanni, and Dennison, Jennifer B.
- Subjects
body regions ,skin and connective tissue diseases - Abstract
Invasive lobular carcinoma (ILC) is the second most prevalent histologic subtype of invasive breast cancer. Here, we comprehensively profiled 817 breast tumors, including 127 ILC, 490 ductal (IDC), and 88 mixed IDC/ILC. Besides E-cadherin loss, the best known ILC genetic hallmark, we identified mutations targeting PTEN, TBX3 and FOXA1 as ILC enriched features. PTEN loss associated with increased AKT phosphorylation, which was highest in ILC among all breast cancer subtypes. Spatially clustered FOXA1 mutations correlated with increased FOXA1 expression and activity. Conversely, GATA3 mutations and high expression characterized Luminal A IDC, suggesting differential modulation of ER activity in ILC and IDC. Proliferation and immune-related signatures determined three ILC transcriptional subtypes associated with survival differences. Mixed IDC/ILC cases were molecularly classified as ILC-like and IDC-like revealing no true hybrid features. This multidimensional molecular atlas sheds new light on the genetic bases of ILC and provides potential clinical options.
- Published
- 2015
- Full Text
- View/download PDF
31. A Plasma-Derived Protein-Metabolite Multiplexed Panel for Early-Stage Pancreatic Cancer.
- Author
-
Fahrmann, Johannes F, Bantis, Leonidas E, Capello, Michela, Scelo, Ghislaine, Dennison, Jennifer B, Patel, Nikul, Murage, Eunice, Vykoukal, Jody, Kundnani, Deepali L, Foretova, Lenka, Fabianova, Eleonora, Holcatova, Ivana, Janout, Vladimir, Feng, Ziding, Yip-Schneider, Michele, Zhang, Jianjun, Brand, Randall, Taguchi, Ayumu, Maitra, Anirban, and Brennan, Paul
- Subjects
PANCREATIC cysts ,PANCREATIC cancer ,METABOLITES ,CONFIDENCE intervals ,BIOMARKERS - Abstract
Background: We applied a training and testing approach to develop and validate a plasma metabolite panel for the detection of early-stage pancreatic ductal adenocarcinoma (PDAC) alone and in combination with a previously validated protein panel for early-stage PDAC.Methods: A comprehensive metabolomics platform was initially applied to plasmas collected from 20 PDAC cases and 80 controls. Candidate markers were filtered based on a second independent cohort that included nine invasive intraductal papillary mucinous neoplasm cases and 51 benign pancreatic cysts. Blinded validation of the resulting metabolite panel was performed in an independent test cohort consisting of 39 resectable PDAC cases and 82 matched healthy controls. The additive value of combining the metabolite panel with a previously validated protein panel was evaluated.Results: Five metabolites (acetylspermidine, diacetylspermine, an indole-derivative, and two lysophosphatidylcholines) were selected as a panel based on filtering criteria. A combination rule was developed for distinguishing between PDAC and healthy controls using the Training Set. In the blinded validation study with early-stage PDAC samples and controls, the five metabolites yielded areas under the curve (AUCs) ranging from 0.726 to 0.842, and the combined metabolite model yielded an AUC of 0.892 (95% confidence interval [CI] = 0.828 to 0.956). Performance was further statistically significantly improved by combining the metabolite panel with a previously validated protein marker panel consisting of CA 19-9, LRG1, and TIMP1 (AUC = 0.924, 95% CI = 0.864 to 0.983, comparison DeLong test one-sided P= .02).Conclusions: A metabolite panel in combination with CA19-9, TIMP1, and LRG1 exhibited substantially improved performance in the detection of early-stage PDAC compared with a protein panel alone. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
32. Characterization of the Role Rab25 in Energy Metabolism and Cancer Using Extracellular Flux Analysis and Material Balance.
- Author
-
Mitra, Shreya, Molina, Jennifer, Mills, Gordon B., and Dennison, Jennifer B.
- Published
- 2015
- Full Text
- View/download PDF
33. Long Noncoding RNA Ceruloplasmin Promotes Cancer Growth by Altering Glycolysis.
- Author
-
Rupaimoole, Rajesha, Lee, Jaehyuk, Haemmerle, Monika, Ling, Hui, Previs, Rebecca A., Pradeep, Sunila, Wu, Sherry Y., Ivan, Cristina, Ferracin, Manuela, Dennison, Jennifer B., Millward, Niki M. Zacharias, Nagaraja, Archana S., Gharpure, Kshipra M., McGuire, Michael, Sam, Nidhin, Armaiz-Pena, Guillermo N., Sadaoui, Nouara C., Rodriguez-Aguayo, Cristian, Calin, George A., and Drapkin, Ronny I.
- Abstract
Summary Long noncoding RNAs (lncRNAs) significantly influence the development and regulation of genome expression in cells. Here, we demonstrate the role of lncRNA ceruloplasmin (NRCP) in cancer metabolism and elucidate functional effects leading to increased tumor progression. NRCP was highly upregulated in ovarian tumors, and knockdown of NRCP resulted in significantly increased apoptosis, decreased cell proliferation, and decreased glycolysis compared with control cancer cells. In an orthotopic mouse model of ovarian cancer, siNRCP delivered via a liposomal carrier significantly reduced tumor growth compared with control treatment. We identified NRCP as an intermediate binding partner between STAT1 and RNA polymerase II, leading to increased expression of downstream target genes such as glucose-6-phosphate isomerase. Collectively, we report a previously unrecognized role of the lncRNA NRCP in modulating cancer metabolism. As demonstrated, DOPC nanoparticle-incorporated siRNA-mediated silencing of this lncRNA in vivo provides therapeutic avenue toward modulating lncRNAs in cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
34. Abortive Autophagy Induces Endoplasmic Reticulum Stress and Cell Death in Cancer Cells.
- Author
-
Claerhout, Sofie, Dutta, Bhaskar, Bossuyt, Wouter, Fan Zhang, Nguyen-Charles, Catherine, Dennison, Jennifer B., Qinghua Yu, Shuangxing Yu, Balázsi, Gábor, Yiling Lu, and Mills, Gordon B.
- Subjects
AUTOPHAGY ,ENDOPLASMIC reticulum ,CELL death ,CANCER cells ,CELL lines ,APOPTOSIS - Abstract
Autophagic cell death or abortive autophagy has been proposed to eliminate damaged as well as cancer cells, but there remains a critical gap in our knowledge in how this process is regulated. The goal of this study was to identify modulators of the autophagic cell death pathway and elucidate their effects on cellular signaling and function. The result of our siRNA library screenings show that an intact coatomer complex I (COPI) is obligatory for productive autophagy. Depletion of COPI complex members decreased cell survival and impaired productive autophagy which preceded endoplasmic reticulum stress. Further, abortive autophagy provoked by COPI depletion significantly altered growth factor signaling in multiple cancer cell lines. Finally, we show that COPI complex members are overexpressed in an array of cancer cell lines and several types of cancer tissues as compared to normal cell lines or tissues. In cancer tissues, overexpression of COPI members is associated with poor prognosis. Our results demonstrate that the coatomer complex is essential for productive autophagy and cellular survival, and thus inhibition of COPI members may promote cell death of cancer cells when apoptosis is compromised. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
35. Intracellular Succinylation of 8-Chloroadenosine and Its Effect on Fumarate Levels.
- Author
-
Dennison, Jennifer B., Ayres, Mary L., Kaluarachchi, Kumar, Plunkett, William, and Gandhi, Varsha
- Subjects
- *
ADENOSINES , *NUCLEOSIDES , *CHRONIC lymphocytic leukemia treatment , *SUCCINATE dehydrogenase , *METFORMIN , *PURINE synthesis - Abstract
8-Chloroadenosine (8-Cl-Ado) is a ribosyl nucleoside analog currently in phase I testing for the treatment of chronic lymphocytic leukemia (CLL). 8-Cl-Ado activity is dependent on adenosine kinase and requires intracellular accumulation of 8-Cl-Ado as mono-, di-, and tn-phosphates. In the current study with four mantle cell lymphoma cell lines, we report a new major metabolic pathway for 8-Cl-Ado intracellular metabolism, the formation of succinyl-8-chloro-adenosine (S-8-Cl-Ado) and its monophosphate (S-8-Cl-AMP), 8-Cl-AMP levels were highly associated with S-8-Cl-AMP levels and reached a steady-state prior to the secondary metabolites, 8-Cl-ATP and S-8-Cl-Ado. Consistent with fumarate as a required substrate for formation of succinyl-8-Cl-adenylate metabolites, the S-8-Cl-adenylate concentrations in multiple cell lines were associated with fumarate loss. The distribution of metabolites was also altered using the energy metabolism modifiers, metformin and oligomycin. The rates of succinyl-8-Cl-adenylate metabolism were enhanced by increasing the intracellular fumarate concentrations after metformin co-treatment. In addition, the S-8-Cl-AMP concentrations were increased after acute inhibition of ATP synthase by oligomycin. We conclude that 8-Cl-Ado metabolism not only affects intracellular purine metabolism; 8-Cl-Ado conversion to succinyl analogs ties its metabolism to the citric acid cycle by reduction of the fumarate pool. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
36. A gel-free MS-based quantitative proteomic approach accurately measures cytochrome P450 protein concentrations in human liver microsomes.
- Author
-
Wang, Michael Zhuo, Wu, Judy Qiju, Dennison, Jennifer B., Bridges, Arlene S., Hall, Stephen D., Kornbluth, Sally, Tidwell, Richard R., Smith, Philip C., Voyksner, Robert D., Paine, Mary F., and Hall, James Edwin
- Published
- 2008
- Full Text
- View/download PDF
37. Plasma Based Protein Signatures Associated with Small Cell Lung Cancer.
- Author
-
Fahrmann, Johannes F., Katayama, Hiroyuki, Irajizad, Ehsan, Chakraborty, Ashish, Kato, Taketo, Mao, Xiangying, Park, Soyoung, Murage, Eunice, Rusling, Leona, Yu, Chuan-Yih, Cai, Yinging, Hsiao, Fu Chung, Dennison, Jennifer B., Tran, Hai, Ostrin, Edwin, Wilson, David O., Yuan, Jian-Min, Vykoukal, Jody, and Hanash, Samir
- Subjects
LUNG cancer ,BIOMARKERS ,PROTEINS ,PROTEOMICS ,TUMOR classification ,EPITHELIAL-mesenchymal transition ,TUMOR suppressor genes ,TUMOR markers ,RECEIVER operating characteristic curves - Abstract
Simple Summary: Small-cell lung cancer (SCLC) typically presents at an advanced stage and is associated with high mortality. When diagnosed at an early stage with localized disease, long-term survival can, however, be achieved. In this study, we report a comprehensive proteomic profiling of case plasmas collected at the time of diagnosis or preceding diagnosis of SCLC with the objective of identifying blood-based markers associated with disease pathogenesis. Our study reveals the occurrence of circulating protein features centered on signatures of oncogenic MYC and YAP1 that were elevated in plasmas of cases at and before the time-of-diagnosis of SCLC. We further report several proteins, particularly inflammatory markers, that were identified as elevated in plasma several years prior to the diagnosis of SCLC and that may indicate increased risk of disease. In summary, our study identifies several novel circulating proteins associated with SCLC development that may offer utility for early detection. Small-cell-lung cancer (SCLC) is associated with overexpression of oncogenes including Myc family genes and YAP1 and inactivation of tumor suppressor genes. We performed in-depth proteomic profiling of plasmas collected from 15 individuals with newly diagnosed early stage SCLC and from 15 individuals before the diagnosis of SCLC and compared findings with plasma proteomic profiles of 30 matched controls to determine the occurrence of signatures that reflect disease pathogenesis. A total of 272 proteins were elevated (area under the receiver operating characteristic curve (AUC) ≥ 0.60) among newly diagnosed cases compared to matched controls of which 31 proteins were also elevated (AUC ≥ 0.60) in case plasmas collected within one year prior to diagnosis. Ingenuity Pathway analyses of SCLC-associated proteins revealed enrichment of signatures of oncogenic MYC and YAP1. Intersection of proteins elevated in case plasmas with proteomic profiles of conditioned medium from 17 SCLC cell lines yielded 52 overlapping proteins characterized by YAP1-associated signatures of cytoskeletal re-arrangement and epithelial-to-mesenchymal transition. Among samples collected more than one year prior to diagnosis there was a predominance of inflammatory markers. Our integrated analyses identified novel circulating protein features in early stage SCLC associated with oncogenic drivers. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
38. 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
39. Targeting metabolic vulnerabilities of cancer: Small molecule inhibitors in clinic.
- Author
-
Tripathi, Satyendra C., Fahrmann, Johannes F., Vykoukal, Jody V., Dennison, Jennifer B., and Hanash, Samir M.
- Published
- 2019
- Full Text
- View/download PDF
40. A murine preclinical syngeneic transplantation model for breast cancer precision medicine
- Author
-
Lorenzi, Philip L., Vellano, Christopher P., Liu, Shuying, Mills, Gordon B., Byers, Lauren A., Perou, Charles M., Mitra, Shreya, Wang, Jing, Zhang, Dong, Lin, Shiaw-Yih, Gagea, Mihai, Zhao, Wei, McGrail, Daniel J., Ju, Zhenlin, Federico, Lorenzo, Diao, Lixia, Jeong, Kang Jin, Cardnell, Robert, Lu, Yiling, Dennison, Jennifer B., and Chong, Zechen
- Subjects
3. Good health - Abstract
We previously demonstrated that altered activity of lysophosphatidic acid in murine mammary glands promotes tumorigenesis. We have now established and characterized a heterogeneous collection of mouse-derived syngeneic transplants (MDSTs) as preclinical platforms for the assessment of personalized pharmacological therapies. Detailed molecular and phenotypic analyses revealed that MDSTs are the most heterogeneous group of genetically engineered mouse models (GEMMs) of breast cancer yet observed. Response of MDSTs to trametinib, a mitogen-activated protein kinase (MAPK) kinase inhibitor, correlated with RAS/MAPK signaling activity, as expected from studies in xenografts and clinical trials providing validation of the utility of the model. Sensitivity of MDSTs to talazoparib, a poly(adenosine 5′-diphosphate–ribose) polymerase (PARP) inhibitor, was predicted by PARP1 protein levels and by a new PARP sensitivity predictor (PSP) score developed from integrated analysis of drug sensitivity data of human cell lines. PSP score–based classification of The Cancer Genome Atlas breast cancer suggested that a subset of patients with limited therapeutic options would be expected to benefit from PARP-targeted drugs. These results indicate that MDSTs are useful models for studies of targeted therapies, and propose novel potential biomarkers for identification of breast cancer patients likely to benefit from personalized pharmacological treatments.
41. (S027) Novel Mechanisms of Adaptive Resistance in Head and Neck Cancer.
- Author
-
Takiar, Vinita, Dennison, Jennifer B., Mitra, Shreya, Ma, Dominic, Phan, Jack, and Mills, Gordon B.
- Published
- 2015
42. The beneficial effects of a gas-permeable flask for expansion of Tumor-Infiltrating lymphocytes as reflected in their mitochondrial function and respiration capacity.
- Author
-
Forget, Marie-Andrée, Haymaker, Cara, Dennison, Jennifer B, Toth, Christopher, Maiti, Sourindra, Fulbright, Orenthial J, Cooper, Laurence J N, Hwu, Patrick, Radvanyi, Laszlo G, and Bernatchez, Chantale
- Subjects
LYMPHOCYTES ,MITOCHONDRIAL physiology ,RESPIRATION ,CELL migration ,MELANOMA treatment - Abstract
Adoptive transfer of autologousex vivoexpanded tumor-infiltrating lymphocytes (TIL) is a highly successful cell therapy approach in the treatment of late-stage melanoma. Notwithstanding the success of this therapy, only very few centers worldwide can provide it. To make this therapy broadly available, one of the major obstacles to overcome is the complexity of culturing the TIL. Recently, major efforts have been deployed to resolve this issue. The use of the Gas-permeable flask (G-Rex) during the REP has been one application that has facilitated this process. Here we show that the use of this new device is able to rescue poor TIL growth and maintain clonal diversity while supporting an improved mitochondrial function. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
43. Functional annotation of rare gene aberration drivers of pancreatic cancer.
- Author
-
Tsang, Yiu Huen, Dogruluk, Turgut, Tedeschi, Philip M., Wardwell-Ozgo, Joanna, Lu, Hengyu, Espitia, Maribel, Nair, Nikitha, Minelli, Rosalba, Chong, Zechen, Chen, Fengju, Chang, Qing Edward, Dennison, Jennifer B., Dogruluk, Armel, Li, Min, Ying, Haoqiang, Bertino, Joseph R., Gingras, Marie-Claude, Ittmann, Michael, Kerrigan, John, and Chen, Ken
- Published
- 2016
- Full Text
- View/download PDF
44. Antibiotic-induced loss of gut microbiome metabolic output correlates with clinical responses to CAR T-cell therapy.
- Author
-
Prasad R, Rehman A, Rehman L, Darbaniyan F, Blumenberg V, Schubert ML, Mor U, Zamir E, Schmidt S, Hayase T, Chia-Chi C, McDaniel LK, Flores I, Strati P, Nair R, Chihara D, Fayad LE, Ahmed S, Iyer SP, Wang ML, Jain P, Nastoupil LJ, Westin JR, Arora R, Turner JG, Khawaja F, Wu R, Dennison JB, Menges M, Hidalgo-Vargas M, Reid KM, Davila ML, Dreger P, Korell F, Schmitt A, Tanner MR, Champlin RE, Flowers CR, Shpall EJ, Hanash S, Neelapu SS, Schmitt M, Subklewe M, Fahrmann J, Stein-Thoeringer C, Elinav E, Jain MD, Hayase E, Jenq RR, and Saini NY
- Abstract
Antibiotic-induced microbiome dysbiosis is widespread in oncology, adversely affecting outcomes and side effects of various cancer treatments, including immune checkpoint inhibitors and chimeric antigen receptor T (CAR-T) cell therapies. In this study, we observed that prior exposure to broad-spectrum ABX with extended anaerobic coverage like piperacillin-tazobactam and meropenem was associated with worsened anti-CD19 CAR-T therapy survival outcomes in large B-cell lymphoma patients (n=422), compared to other ABX classes. In a discovery subset of these patients (n=67), we found that the use of these ABX was in turn associated with substantial dysbiosis of gut microbiome function, resulting in significant alterations of the gut and blood metabolome, including microbial effectors such as short-chain fatty acids (SCFAs) and other anionic metabolites, findings that were largely reproduced in an external validation cohort (n=58). Broader evaluation of circulating microbial metabolites revealed reductions in indole and cresol derivatives, as well as trimethylamine N-oxide, in patients who received ABX treatment (discovery n=40, validation n=28). These findings were recapitulated in an immune-competent CAR-T mouse model, where meropenem-induced dysbiosis led to a systemic dysmetabolome and decreased murine anti-CD19 CAR-T efficacy. Furthermore, we demonstrate that SCFAs can enhance the metabolic fitness of CAR-T cells, leading to improved tumor killing capacity. Together, these results suggest that broad-spectrum ABX deplete metabolically active commensals whose metabolites are essential for enhancing CAR-T efficacy, shedding light on the intricate relationship between ABX exposure, microbiome function and their impact on CAR-T cell efficacy. This highlights the potential for modulating the microbiome to augment CAR-T immunotherapy., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
45. Exercise Training Reduces the Inflammatory Response and Promotes Intestinal Mucosa-Associated Immunity in Lynch Syndrome.
- Author
-
Deng N, Reyes-Uribe L, Fahrmann JF, Thoman WS, Munsell MF, Dennison JB, Murage E, Wu R, Hawk ET, Thirumurthi S, Lynch PM, Dieli-Conwright CM, Lazar AJ, Jindal S, Chu K, Chelvanambi M, Basen-Engquist K, Li Y, Wargo JA, McAllister F, Allison JP, Sharma P, Sinha KM, Hanash S, Gilchrist SC, and Vilar E
- Subjects
- Female, Humans, Exercise, Gene Expression Profiling, Intestinal Mucosa pathology, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Colorectal Neoplasms, Hereditary Nonpolyposis therapy, Endometrial Neoplasms genetics
- Abstract
Purpose: Lynch syndrome (LS) is a hereditary condition with a high lifetime risk of colorectal and endometrial cancers. Exercise is a non-pharmacologic intervention to reduce cancer risk, though its impact on patients with LS has not been prospectively studied. Here, we evaluated the impact of a 12-month aerobic exercise cycling intervention in the biology of the immune system in LS carriers., Patients and Methods: To address this, we enrolled 21 patients with LS onto a non-randomized, sequential intervention assignation, clinical trial to assess the effect of a 12-month exercise program that included cycling classes 3 times weekly for 45 minutes versus usual care with a one-time exercise counseling session as control. We analyzed the effects of exercise on cardiorespiratory fitness, circulating, and colorectal-tissue biomarkers using metabolomics, gene expression by bulk mRNA sequencing, and spatial transcriptomics by NanoString GeoMx., Results: We observed a significant increase in oxygen consumption (VO2peak) as a primary outcome of the exercise and a decrease in inflammatory markers (prostaglandin E) in colon and blood as the secondary outcomes in the exercise versus usual care group. Gene expression profiling and spatial transcriptomics on available colon biopsies revealed an increase in the colonic mucosa levels of natural killer and CD8+ T cells in the exercise group that were further confirmed by IHC studies., Conclusions: Together these data have important implications for cancer interception in LS, and document for the first-time biological effects of exercise in the immune system of a target organ in patients at-risk for cancer., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
46. Integrated spatial transcriptomics and lipidomics of precursor lesions of pancreatic cancer identifies enrichment of long chain sulfatide biosynthesis as an early metabolic alteration.
- Author
-
Sans M, Chen Y, Thege FI, Dou R, Min J, Yip-Schneider M, Zhang J, Wu R, Irajizad E, Makino Y, Rajapakshe KI, Hurd MW, León-Letelier RA, Vykoukal J, Dennison JB, Do KA, Wolff RA, Guerrero PA, Kim MP, Schmidt CM, Maitra A, Hanash S, and Fahrmann JF
- Abstract
Background: The development of diverse spatial profiling technologies has provided an unprecedented insight into molecular mechanisms driving cancer pathogenesis. Here, we conducted the first integrated cross-species assessment of spatial transcriptomics and spatial metabolomics alterations associated with progression of intraductal papillary mucinous neoplasms (IPMN), bona fide cystic precursors of pancreatic ductal adenocarcinoma (PDAC)., Methods: Matrix Assisted Laster Desorption/Ionization (MALDI) mass spectrometry (MS)-based spatial imaging and Visium spatial transcriptomics (ST) (10X Genomics) was performed on human resected IPMN tissues (N= 23) as well as pancreata from a mutant Kras;Gnas mouse model of IPMN. Findings were further compared with lipidomic analyses of cystic fluid from 89 patients with histologically confirmed IPMNs, as well as single-cell and bulk transcriptomic data of PDAC and normal tissues., Results: MALDI-MS analyses of IPMN tissues revealed long-chain hydroxylated sulfatides, particularly the C24:0(OH) and C24:1(OH) species, to be selectively enriched in the IPMN and PDAC neoplastic epithelium. Integrated ST analyses confirmed that the cognate transcripts engaged in sulfatide biosynthesis, including UGT8, Gal3St1 , and FA2H , were co-localized with areas of sulfatide enrichment. Lipidomic analyses of cystic fluid identified several sulfatide species, including the C24:0(OH) and C24:1(OH) species, to be significantly elevated in patients with IPMN/PDAC compared to those with low-grade IPMN. Targeting of sulfatide metabolism via the selective galactosylceramide synthase inhibitor, UGT8-IN-1, resulted in ceramide-induced lethal mitophagy and subsequent cancer cell death in vitro , and attenuated tumor growth of mutant Kras;Gnas allografts. Transcript levels of UGT8 and FA2H were also selectively enriched in PDAC transcriptomic datasets compared to non-cancerous areas, and elevated tumoral UGT8 was prognostic for poor overall survival., Conclusion: Enhanced sulfatide metabolism is an early metabolic alteration in cystic pre-cancerous lesions of the pancreas that persists through invasive neoplasia. Targeting sulfatide biosynthesis might represent an actionable vulnerability for cancer interception.
- Published
- 2023
- Full Text
- View/download PDF
47. A Blood-Based Metabolite Panel for Distinguishing Ovarian Cancer from Benign Pelvic Masses.
- Author
-
Irajizad E, Han CY, Celestino J, Wu R, Murage E, Spencer R, Dennison JB, Vykoukal J, Long JP, Do KA, Drescher C, Lu K, Lu Z, Bast RC, Hanash S, and Fahrmann JF
- Subjects
- Female, Humans, CA-125 Antigen, WAP Four-Disulfide Core Domain Protein 2, Proteins metabolism, Carcinoma, Ovarian Epithelial, Biomarkers, Tumor, Algorithms, Neoplasms, Glandular and Epithelial, Ovarian Neoplasms pathology
- Abstract
Purpose: To assess the contributions of circulating metabolites for improving upon the performance of the risk of ovarian malignancy algorithm (ROMA) for risk prediction of ovarian cancer among women with ovarian cysts., Experimental Design: Metabolomic profiling was performed on an initial set of sera from 101 serous and nonserous ovarian cancer cases and 134 individuals with benign pelvic masses (BPM). Using a deep learning model, a panel consisting of seven cancer-related metabolites [diacetylspermine, diacetylspermidine, N-(3-acetamidopropyl)pyrrolidin-2-one, N-acetylneuraminate, N-acetyl-mannosamine, N-acetyl-lactosamine, and hydroxyisobutyric acid] was developed for distinguishing early-stage ovarian cancer from BPM. The performance of the metabolite panel was evaluated in an independent set of sera from 118 ovarian cancer cases and 56 subjects with BPM. The contributions of the panel for improving upon the performance of ROMA were further assessed., Results: A 7-marker metabolite panel (7MetP) developed in the training set yielded an AUC of 0.86 [95% confidence interval (CI): 0.76-0.95] for early-stage ovarian cancer in the independent test set. The 7MetP+ROMA model had an AUC of 0.93 (95% CI: 0.84-0.98) for early-stage ovarian cancer in the test set, which was improved compared with ROMA alone [0.91 (95% CI: 0.84-0.98); likelihood ratio test P: 0.03]. In the entire specimen set, the combined 7MetP+ROMA model yielded a higher positive predictive value (0.68 vs. 0.52; one-sided P < 0.001) with improved specificity (0.89 vs. 0.78; one-sided P < 0.001) for early-stage ovarian cancer compared with ROMA alone., Conclusions: A blood-based metabolite panel was developed that demonstrates independent predictive ability and complements ROMA for distinguishing early-stage ovarian cancer from benign disease to better inform clinical decision making., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
48. Application of Artificial Intelligence to Plasma Metabolomics Profiles to Predict Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer.
- Author
-
Irajizad E, Wu R, Vykoukal J, Murage E, Spencer R, Dennison JB, Moulder S, Ravenberg E, Lim B, Litton J, Tripathym D, Valero V, Damodaran S, Rauch GM, Adrada B, Candelaria R, White JB, Brewster A, Arun B, Long JP, Do KA, Hanash S, and Fahrmann JF
- Abstract
There is a need to identify biomarkers predictive of response to neoadjuvant chemotherapy (NACT) in triple-negative breast cancer (TNBC). We previously obtained evidence that a polyamine signature in the blood is associated with TNBC development and progression. In this study, we evaluated whether plasma polyamines and other metabolites may identify TNBC patients who are less likely to respond to NACT. Pre-treatment plasma levels of acetylated polyamines were elevated in TNBC patients that had moderate to extensive tumor burden (RCB-II/III) following NACT compared to those that achieved a complete pathological response (pCR/RCB-0) or had minimal residual disease (RCB-I). We further applied artificial intelligence to comprehensive metabolic profiles to identify additional metabolites associated with treatment response. Using a deep learning model (DLM), a metabolite panel consisting of two polyamines as well as nine additional metabolites was developed for improved prediction of RCB-II/III. The DLM has potential clinical value for identifying TNBC patients who are unlikely to respond to NACT and who may benefit from other treatment modalities., Competing Interests: Author SM declares honoraria from Novartis, Pfizer and research funding from Oncothyreon (Inst), Pfizer (Inst), Novartis (Inst), Genentech (Inst), Takeda (Inst), Bayer (Inst), EMD Serono (Inst), Genentech (Inst); travel, accommodations, expenses: Novartis, Pfizer. Author JLi declares consulting or advisory role: Pfizer, AstraZeneca, Medivation/Pfizer; speakers' bureau from Physician Education Resource, UpToDate, Med Learning, Group, Medscape; research funding from Novartis (Inst), Bristol-Myers Squibb (Inst), Genentech (Inst), Pfizer (Inst), EMD Serono (Inst), Jounce Therapeutics (Inst), GlaxoSmithKline (Inst), Medivation/Pfizer (Inst); patents, royalties, other intellectual property: UpToDate; travel, accommodations, expenses: Physician Education Resource, Med Learning Group, Medscape. Author DT declares consulting or advisory role: AstraZeneca, Genomic Health, Gilead Sciences Inc, GlaxoSmithKline, Novartis Pharma, OncoPep, Pfizer; research funding: Pfizer, Novartis Pharma. Author VV declares honoraria: Genentec; consulting or advisory role: Genentech; travel, accommodations, expenses: Genentech. Author SD declares honoraria: Novartis; consulting or advisory role: Tempus, Taiho Pharmaceutical, Pfizer; research funding: EMD Serono, Guardant Health; travel, accommodations; expenses: Phillips Gilmore Oncology Communications. Author BAd declares consulting or advisory role: Bright Pink, AbbVie; research funding: AbbVie (Inst), PharmaMar (Inst), AstraZeneca (Inst), InVitae (Inst); travel, accommodations, expenses: AstraZeneca. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Irajizad, Wu, Vykoukal, Murage, Spencer, Dennison, Moulder, Ravenberg, Lim, Litton, Tripathym, Valero, Damodaran, Rauch, Adrada, Candelaria, White, Brewster, Arun, Long, Do, Hanash and Fahrmann.)
- Published
- 2022
- Full Text
- View/download PDF
49. SRGN-Triggered Aggressive and Immunosuppressive Phenotype in a Subset of TTF-1-Negative Lung Adenocarcinomas.
- Author
-
Tanaka I, Dayde D, Tai MC, Mori H, Solis LM, Tripathi SC, Fahrmann JF, Unver N, Parhy G, Jain R, Parra ER, Murakami Y, Aguilar-Bonavides C, Mino B, Celiktas M, Dhillon D, Casabar JP, Nakatochi M, Stingo F, Baladandayuthapani V, Wang H, Katayama H, Dennison JB, Lorenzi PL, Do KA, Fujimoto J, Behrens C, Ostrin EJ, Rodriguez-Canales J, Hase T, Fukui T, Kajino T, Kato S, Yatabe Y, Hosoda W, Kawaguchi K, Yokoi K, Chen-Yoshikawa TF, Hasegawa Y, Gazdar AF, Wistuba II, Hanash S, and Taguchi A
- Subjects
- Animals, Humans, Mice, Phenotype, Proteomics, Thyroid Nuclear Factor 1 genetics, Tumor Microenvironment, Adenocarcinoma of Lung genetics, DNA-Binding Proteins, Lung Neoplasms genetics, Lung Neoplasms pathology, Proteoglycans metabolism, Transcription Factors, Vesicular Transport Proteins metabolism
- Abstract
Background: Approximately 20% of lung adenocarcinoma (LUAD) is negative for the lineage-specific oncogene Thyroid transcription factor 1 (TTF-1) and exhibits worse clinical outcome with a low frequency of actionable genomic alterations. To identify molecular features associated with TTF-1-negative LUAD, we compared the transcriptomic and proteomic profiles of LUAD cell lines. SRGN , a chondroitin sulfate proteoglycan Serglycin, was identified as a markedly overexpressed gene in TTF-1-negative LUAD. We therefore investigated the roles and regulation of SRGN in TTF-1-negative LUAD., Methods: Proteomic and metabolomic analyses of 41 LUAD cell lines were done using mass spectrometry. The function of SRGN was investigated in 3 TTF-1-negative and 4 TTF-1-positive LUAD cell lines and in a syngeneic mouse model (n = 5 to 8 mice per group). Expression of SRGN was evaluated in 94 and 105 surgically resected LUAD tumor specimens using immunohistochemistry. All statistical tests were 2-sided., Results: SRGN was markedly overexpressed at mRNA and protein levels in TTF-1-negative LUAD cell lines (P < .001 for both mRNA and protein levels). Expression of SRGN in LUAD tumor tissue was associated with poor outcome (hazard ratio = 4.22, 95% confidence interval = 1.12 to 15.86, likelihood ratio test, P = .03), and with higher expression of Programmed cell death 1 ligand 1 (PD-L1) in tumor cells and higher infiltration of Programmed cell death protein 1-positive lymphocytes. SRGN regulated expression of PD-L1 as well as proinflammatory cytokines, including Interleukin-6, Interleukin-8, and C-X-C motif chemokine 1 in LUAD cell lines; increased migratory and invasive properties of LUAD cells and fibroblasts; and enhanced angiogenesis. SRGN was induced by DNA demethylation resulting from Nicotinamide N-methyltransferase-mediated impairment of methionine metabolism., Conclusions: Our findings suggest that SRGN plays a pivotal role in tumor-stromal interaction and reprogramming into an aggressive and immunosuppressive tumor microenvironment in TTF-1-negative LUAD., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
50. A Deep Learning Approach to Re-create Raw Full-Field Digital Mammograms for Breast Density and Texture Analysis.
- Author
-
Shu H, Chiang T, Wei P, Do KA, Lesslie MD, Cohen EO, Srinivasan A, Moseley TW, Chang Sen LQ, Leung JWT, Dennison JB, Hanash SM, and Weaver OO
- Abstract
Purpose: To develop a computational approach to re-create rarely stored for-processing (raw) digital mammograms from routinely stored for-presentation (processed) mammograms., Materials and Methods: In this retrospective study, pairs of raw and processed mammograms collected in 884 women (mean age, 57 years ± 10 [standard deviation]; 3713 mammograms) from October 5, 2017, to August 1, 2018, were examined. Mammograms were split 3088 for training and 625 for testing. A deep learning approach based on a U-Net convolutional network and kernel regression was developed to estimate the raw images. The estimated raw images were compared with the originals by four image error and similarity metrics, breast density calculations, and 29 widely used texture features., Results: In the testing dataset, the estimated raw images had small normalized mean absolute error (0.022 ± 0.015), scaled mean absolute error (0.134 ± 0.078) and mean absolute percentage error (0.115 ± 0.059), and a high structural similarity index (0.986 ± 0.007) for the breast portion compared with the original raw images. The estimated and original raw images had a strong correlation in breast density percentage (Pearson r = 0.946) and a strong agreement in breast density grade (Cohen κ = 0.875). The estimated images had satisfactory correlations with the originals in 23 texture features (Pearson r ≥ 0.503 or Spearman ρ ≥ 0.705) and were well complemented by processed images for the other six features., Conclusion: This deep learning approach performed well in re-creating raw mammograms with strong agreement in four image evaluation metrics, breast density, and the majority of 29 widely used texture features. Keywords: Mammography, Breast, Supervised Learning, Convolutional Neural Network (CNN), Deep learning algorithms, Machine Learning AlgorithmsSee also the commentary by Chan in this issue. Supplemental material is available for this article. ©RSNA, 2021., Competing Interests: Disclosures of Conflicts of Interest: H.S. disclosed no relevant relationships. T.C. Activities related to the present article: institution received National Institutes of Health (NIH)/National Cancer Institute (NCI) Cancer Center Support Grant (P30 CA016672); institution supported by Little Green Book Foundation, Center for Global Early Detection at MD Anderson, and McCombs Institute at MD Anderson. Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships. P.W. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672). Activities not related to the present article: disclosed no relevant relationships. K.A.D. disclosed no relevant relationships. M.D.L. disclosed no relevant relationships. E.O.C. disclosed no relevant relationships. A.S. disclosed no relevant relationships. T.W.M. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672). Activities not related to the present article: author is paid medical consultant for Hologic and Merit Medical. Other relationships: disclosed no relevant relationships. L.C.S. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672); institution received support from Little Green Book Foundation, Center for Global Early Detection at MD Anderson, and McCombs Institute at MD Anderson. Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships. J.W.T.L. Activities related to the present article: disclosed no relevant relationships. Activities not related to the present article: author paid by Fujifilm and GE Healthcare for lectures; author has stock/stock options in Subtle Medical (start-up company, stock/stock options have no monetary value at this time). Other relationships: disclosed no relevant relationships. J.B.D. Activities related to the present article: institution supported by Little Green Book Foundation (has supported breast cancer mammography clinical research for the early detection, the MERIT program). Activities not related to the present article: employed by MD Anderson. Other relationships: disclosed no relevant relationships. S.M.H. disclosed no relevant relationships. O.O.W. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672); institution received support from Little Green Book Foundation (sponsor of the patient cohort retrospectively used in the study). Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships., (2021 by the Radiological Society of North America, Inc.)
- 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.