148 results on '"Gloria S. Huang"'
Search Results
2. Intersections of endocrine pathways and the epithelial mesenchymal transition in endometrial cancer
- Author
-
Julia H. Gelissen and Gloria S. Huang
- Subjects
EMT ,endometrial cancer ,endocrine ,estrogen ,diabetes ,glucose ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The epithelial mesenchymal transition (EMT) is the process by which cancer cells of epithelial origin, including endometrial cancer, acquire a mesenchymal phenotype with enhanced migratory and invasive capacity, to facilitate metastasis. The regulation of EMT is tissue-specific, and in endometrial cancer, endocrine signaling pathways serve as critical regulators of EMT. The intersections of endocrine signaling and EMT highlight potential avenues for therapeutic intervention to target cancer metastasis with the aim of reduced mortality.
- Published
- 2022
- Full Text
- View/download PDF
3. Sacituzumab govitecan, an antibody‐drug conjugate targeting trophoblast cell‐surface antigen 2, shows cytotoxic activity against poorly differentiated endometrial adenocarcinomas in vitro and in vivo
- Author
-
Emanuele Perrone, Paola Manara, Salvatore Lopez, Stefania Bellone, Elena Bonazzoli, Aranzazu Manzano, Luca Zammataro, Anna Bianchi, Burak Zeybek, Natalia Buza, Joan Tymon‐Rosario, Gary Altwerger, Chanhee Han, Gulden Menderes, Gloria S. Huang, Elena Ratner, Dan‐Arin Silasi, Masoud Azodi, Pei Hui, Peter E. Schwartz, Giovanni Scambia, and Alessandro D. Santin
- Subjects
antibody–drug conjugate ,endometrial carcinoma ,IMMU‐132 ,sacituzumab govitecan ,uterine cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Endometrial cancer is the most common gynecologic malignancy in developed countries. The antibody–drug conjugate (ADC) sacituzumab govitecan (SG) targets trophoblast cell‐surface antigen‐2 (Trop‐2) – a cell‐surface glycoprotein highly expressed in many epithelial tumors – and delivers the active metabolite of irinotecan SN‐38 to Trop‐2‐positive tumor cells. We evaluated Trop‐2 expression in endometrial endometrioid carcinoma (EC) tissues and the activity of SG against primary poorly differentiated EC cell lines and xenografts. Trop‐2 expression was assessed in 143 formalin‐fixed–paraffin‐embedded tumors and seven primary tumor cell lines by immunohistochemistry and flow cytometry, respectively. Cell viability of primary tumor cell lines was assessed following exposure to SG, or control antibodies. Antibody‐dependent cell cytotoxicity (ADCC) against Trop‐2‐positive and Trop‐2‐negative EC cell lines was measured in vitro using 4‐h chromium release assays. A Trop‐2‐positive EC xenograft model was used to determine the in vivo activity of SG. Moderate‐to‐strong staining was detected in 84% (120/143) of EC samples, whereas 43% (3/7) of the primary EC cell lines tested overexpressed Trop‐2. EC cell lines overexpressing Trop‐2 were significantly more sensitive to SG compared to control ADC (P = 0.014 and P = 0.005). Both SG and the unconjugated parental antibody hRS7 mediated high ADCC against Trop‐2‐positive cell lines. Moreover, SG induced significant bystander killing of Trop‐2‐negative tumors cocultured with Trop‐2‐positive tumors. In the xenograft model, intravenous administration of SG twice weekly for three weeks was well tolerated and demonstrated impressive tumor growth inhibition against poorly differentiated, chemotherapy‐resistant EC xenografts (P = 0.011). In summary, SG is a novel ADC with remarkable preclinical activity against poorly differentiated EC cell lines overexpressing Trop‐2. These findings warrant future clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
4. Population-Based Screening for Endometrial Cancer: Human vs. Machine Intelligence
- Author
-
Gregory R. Hart, Vanessa Yan, Gloria S. Huang, Ying Liang, Bradley J. Nartowt, Wazir Muhammad, and Jun Deng
- Subjects
endometrial cancer ,cancer screening ,early detection ,machine learning ,statistical biopsy ,Electronic computers. Computer science ,QA75.5-76.95 - Abstract
Incidence and mortality rates of endometrial cancer are increasing, leading to increased interest in endometrial cancer risk prediction and stratification to help in screening and prevention. Previous risk models have had moderate success with the area under the curve (AUC) ranging from 0.68 to 0.77. Here we demonstrate a population-based machine learning model for endometrial cancer screening that achieves a testing AUC of 0.96.We train seven machine learning algorithms based solely on personal health data, without any genomic, imaging, biomarkers, or invasive procedures. The data come from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO). We further compare our machine learning model with 15 gynecologic oncologists and primary care physicians in the stratification of endometrial cancer risk for 100 women.We find a random forest model that achieves a testing AUC of 0.96 and a neural network model that achieves a testing AUC of 0.91. We test both models in risk stratification against 15 practicing physicians. Our random forest model is 2.5 times better at identifying above-average risk women with a 2-fold reduction in the false positive rate. Our neural network model is 2 times better at identifying above-average risk women with a 3-fold reduction in the false positive rate.Our machine learning models provide a non-invasive and cost-effective way to identify high-risk sub-populations who may benefit from early screening of endometrial cancer, prior to disease onset. Through statistical biopsy of personal health data, we have identified a new and effective approach for early cancer detection and prevention for individual patients.
- Published
- 2020
- Full Text
- View/download PDF
5. Insulin‐like growth factor 2: a poor prognostic biomarker linked to racial disparity in women with uterine carcinosarcoma
- Author
-
Anne R. Van Arsdale, Rebecca C. Arend, Maria J. Cossio, Britt K. Erickson, Yanhua Wang, David W. Doo, Charles A. Leath, Gary L. Goldberg, and Gloria S. Huang
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract The objective of this study was to investigate the relationship of insulin‐like growth factor 2 (IGF2) expression and survival in women with uterine carcinosarcoma (UCS). Insulin‐like growth factor 2 protein expression was determined by immunohistochemical staining of tumor tissues from 103 patients with UCS. The H‐score (product of staining intensity and percentage positive cells) was quantified for the epithelial cytoplasmic (EC), epithelial nuclear (EN), and malignant stromal compartments. Multivariable Cox proportional hazard regression models were used to examine the relationship of IGF2 levels with progression‐free survival (PFS) and overall survival (OS). Adjusting for stage, race, and adjuvant therapy, PFS and OS were reduced in patients with high IGF2 (H‐score ≥ median) in the EC and EN compartments. Black race was independently associated with reduced PFS and OS in patients with early‐stage disease, and IGF2 levels in the EC were higher in black than in white patients (P = 0.02, Wilcoxon test). In a race‐stratified multivariable analysis, high IGF2 in the epithelial compartments more than doubled the risk of death in black women; HR = 2.43 (95% CI: 1.18–5.01, P = 0.02) for high IGF2 in the EC; and HR = 2.34 (95% CI: 1.25–4.39, P = 0.008) for high IGF2 in the EN. In conclusion, high tumor IGF2 expression is an independent risk factor for reduced PFS and OS in UCS. Black women have elevated tumor IGF2 compared with white women, and decreased survival associated with high IGF2. These findings identify IGF2 as a candidate biomarker for survival linked to racial disparity in women with UCS.
- Published
- 2018
- Full Text
- View/download PDF
6. Retreatment with aromatase inhibitor therapy in the management of granulosa cell tumor
- Author
-
Melissa Schwartz and Gloria S. Huang
- Subjects
Adult granulosa cell tumor of the ovary ,Aromatase inhibitors ,Letrozole ,FOXL2 ,Antineoplastic hormonal agents ,Statins ,Gynecology and obstetrics ,RG1-991 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2016
- Full Text
- View/download PDF
7. Clinical utility of chromogranin A and octerotide in large cell neuro endocrine carcinoma of the uterine corpus
- Author
-
Shohreh Shahabi, Ilenia Pellicciotta, June Hou, Sarah Graceffa, Gloria S. Huang, Robert N. Samuelson, and Gary L. Goldberg
- Subjects
Large cell neuroendocrine carcinoma, uterine corpus, chromogranin A, octerotide ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Primary neuroendocrine tumors of the female genital tract have been described in the cervix, ovaries and uterus. Large cell neuroendocrine carcinoma (LCNC) of the uterine corpus is the least common and appears to behave the most aggressively. We report a rare case of a large cell neuroendocrine tumor of the endometrium. These tumors are not well characterized, unlike neuroendocrine tumors of the uterine cervix, consequently, the optimal management remains still unclear. The treatment of our case consisted of surgery, radiotherapy, chemotherapy, and octreotide. Despite the aggressive treatment, the patient died of disease progression 12 months after the initial diagnosis. We discuss the diagnosis, prognosis, and treatment options for LCNC of the genital tract, and potential future therapeutics.
- Published
- 2011
- Full Text
- View/download PDF
8. Uterine leiomyosarcomas harboring MAP2K4 gene amplification are sensitive in vivo to PLX8725, a novel MAP2K4 inhibitor
- Author
-
Blair McNamara, Justin Harold, Diego Manavella, Stefania Bellone, Levent Mutlu, Tobias Max Philipp Hartwich, Margherita Zipponi, Yang Yang-Hartwich, Cem Demirkiran, Miguel Skyler Z. Verzosa, Kevin Yang, Jungmin Choi, Weilai Dong, Natalia Buza, Pei Hui, Gary Altwerger, Gloria S. Huang, Vaagn Andikyan, Mitchell Clark, Elena Ratner, Masoud Azodi, Peter E. Schwartz, Elizabeth A. Burton, Hiroaki Inagaki, Aaron Albers, Chao Zhang, Gideon Bollag, Joseph Schlessinger, and Alessandro D. Santin
- Subjects
Oncology ,Obstetrics and Gynecology - Published
- 2023
9. Trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody–drug conjugate with topoisomerase I inhibitor payload, shows antitumor activity in uterine and ovarian carcinosarcoma with HER2/neu expression
- Author
-
Dennis, Mauricio, Stefania, Bellone, Levent, Mutlu, Blair, McNamara, Diego D, Manavella, Cem, Demirkiran, Miguel Skyler Z, Verzosa, Natalia, Buza, Pei, Hui, Tobias Max Philipp, Hartwich, Justin, Harold, Yang, Yang-Hartwich, Margherita, Zipponi, Gary, Altwerger, Elena, Ratner, Gloria S, Huang, Mitchell, Clark, Vaagn, Andikyan, Masoud, Azodi, Peter E, Schwartz, and Alessandro D, Santin
- Subjects
Oncology ,Obstetrics and Gynecology - Abstract
Carcinosarcomas are highly aggressive gynecologic malignancies containing both carcinomatous and sarcomatous elements with heterogeneous HER2/neu expression and limited therapeutic options. We compared the efficacy of trastuzumab deruxtecan (DS-8201a), a novel HER2/neu-targeting antibody-drug conjugate (ADC) to an ADC isotype control (MAAA-9199) against primary uterine and ovarian carcinosarcomas in vitro and in vivo.Twelve primary carcinosarcoma (CS) cell lines were evaluated for HER2/neu surface expression by immunohistochemistry (IHC) and by flow cytometry, and gene amplification by fluorescence in situ hybridization (FISH) assays. The in vitro experiments included cytotoxicity and bystander killing effect assays on three cell lines of variable HER2/neu expression. In vivo activity was studied in a mouse CS xenograft model of 3+ HER2/neu uterine CS.In vitro studies showed that DS-8201a was highly effective against uterine and ovarian CS cell lines demonstrating 3+ HER2/neu expression compared to MAAA-9199 control; there was no significant improvement in the 0 HER2/neu CS cell line. However, DS-8201a induced efficient bystander killing of 0 HER2/neu tumor cells when admixed with 3+ HER2/neu cells. In vivo studies confirmed that DS-8201a was more effective than MAAA-9199 in 3+ HER2/neu-expressing CS xenografts.DS-8201a may represent a novel and highly effective ADC against HER2/neu-expressing CS.
- Published
- 2023
10. The Poly (ADP-ribose) polymerase inhibitor olaparib and pan-ErbB inhibitor neratinib are highly synergistic in HER2 overexpressing epithelial ovarian carcinoma in vitro and in vivo
- Author
-
Chanhee Han, Blair McNamara, Stefania Bellone, Justin Harold, Paola Manara, Tobias Max Philipp Hartwich, Levent Mutlu, Yang Yang-Hartwich, Margherita Zipponi, Cem Demirkiran, Miguel Skyler Z. Verzosa, Gary Altwerger, Elena Ratner, Gloria S. Huang, Mitchell Clark, Vaagn Andikyan, Masoud Azodi, Peter R. Dottino, Peter E. Schwartz, and Alessandro D. Santin
- Subjects
Oncology ,Obstetrics and Gynecology - Published
- 2023
11. Synergistic activity of neratinib in combination with olaparib in uterine serous carcinoma overexpressing HER2/neu
- Author
-
Ghanshyam Yadav, Dana M. Roque, Stefania Bellone, Diego D. Manavella, Tobias M.P. Hartwich, Margherita Zipponi, Justin Harold, Joan Tymon-Rosario, Levent Mutlu, Gary Altwerger, Gulden Menderes, Elena Ratner, Natalia Buza, Pei Hui, Gloria S. Huang, Vaagn Andikyan, Mitchell Clark, Masoud Azodi, Peter E. Schwartz, Ludmil B. Alexandrov, and Alessandro D. Santin
- Subjects
Oncology ,Receptor, ErbB-2 ,Cell Line, Tumor ,Uterine Neoplasms ,Quinolines ,Humans ,Phthalazines ,Obstetrics and Gynecology ,Female ,Poly(ADP-ribose) Polymerase Inhibitors ,Xenograft Model Antitumor Assays ,Piperazines ,Cystadenocarcinoma, Serous - Abstract
Uterine serous carcinoma (USC) is an aggressive variant of endometrial cancer with a poor prognosis. Approximately 30% of USC overexpress HER2/neu, a recognized target for trastuzumab in advanced/recurrent HER2/neu-positive USC. We evaluated the efficacy of the pan-c-erb inhibitor neratinib and the poly (ADP-ribose)-polymerase (PARP) inhibitor olaparib as single agents and in combination against USC cell lines and xenografts.In-vitro cell-viability assays with olaparib, neratinib, and olaparib/neratinib were assessed using flow-cytometry based assays against a panel of USC cell lines with high and low HER2/neu expression. Homologous recombination deficiency (HRD) signatures were evaluated as described by Alexandrov et al. (Nature;2020;578:94-101) while downstream signaling affected by neratinib/olaparib exposure was assessed with immunoblotting. Efficacy of single- versus dual-agent inhibition was evaluated in-vivo using two USC-xenografts with 3+ HER2/neu expression.Neratinib was more potent than olaparib in suppression of in-vitro growth of HER2/neu 3+ cell lines (ARK1: p = 0.0047; ARK2: p = 0.0428) while no difference was noted against HER2/neu 1+ tumors (ARK4). Importantly, the combination of olaparib with neratinib synergistically improved tumor suppression compared to either single-agent in vitro. USC cells exposed to olaparib upregulated HER2/neu expression, while neratinib treatment increased PARP activity (ARK1: p0.0001; ARK2: p0.0001). Single-agent neratinib transiently inhibited in vivo growth of USC xenografts harboring HER2/neu gene amplification (ARK1: p0.05; ARK2: p0.05). In contrast, the combination of the two inhibitors caused a stronger and durable growth inhibition in both USC xenografts (ARK1: p0.05; ARK2: p0.05).The combination of olaparib and neratinib is active and synergistic against primary HER2/neu + USC. This combination may represent a novel therapeutic option for USC patients with HER2/neu+, homologous recombination-proficient tumors resistant to chemotherapy.
- Published
- 2022
12. Homologous recombination deficiency (HRD) signature-3 in ovarian and uterine carcinosarcomas correlates with preclinical sensitivity to Olaparib, a poly (adenosine diphosphate [ADP]- ribose) polymerase (PARP) inhibitor
- Author
-
Joan R. Tymon-Rosario, Paola Manara, Diego D. Manavella, Stefania Bellone, Tobias Max Philipp Hartwich, Justin Harold, Yang Yang-Hartwich, Margherita Zipponi, Jungmin Choi, Kyungjo Jeong, Levent Mutlu, Kevin Yang, Gary Altwerger, Gulden Menderes, Elena Ratner, Gloria S. Huang, Mitchell Clark, Vaagn Andikyan, Masoud Azodi, Peter E. Schwartz, Ludmil B. Alexandrov, and Alessandro D. Santin
- Subjects
Ovarian Neoplasms ,Ribose ,Ovary ,Obstetrics and Gynecology ,Poly(ADP-ribose) Polymerase Inhibitors ,Piperazines ,Adenosine Diphosphate ,Carcinosarcoma ,Oncology ,Cell Line, Tumor ,Animals ,Humans ,Phthalazines ,Female ,Poly(ADP-ribose) Polymerases ,Homologous Recombination - Abstract
Carcinosarcoma (CS) of the ovary and uterus are highly aggressive malignancies associated with poor survival. Poly(ADP-ribose)-polymerase inhibitors (PARPi) are targeted agents impairing DNA repair via homologous-recombination-deficiency (HRD) mechanisms. We used whole-exome-sequencing (WES) data from a cohort of fresh tumor samples of ovarian (OCS) and uterine carcinosarcoma (UCS), primary cell lines and xenografts to investigate the role for olaparib in CSs.WES data from 73 CS samples (48 UCS and 25 OCS) were analyzed for HRD signatures. Olaparib activity was evaluated using cell-viability, cell-cycle, apoptosis and cytotoxicity assays against primary CS cell lines. Olaparib antitumor activity was tested in vivo against HRD CS xenografts.Signature-3 (i.e. HRD-related signature) was identified in 60% of OCS (15 of 25) vs 25% of UCS (12 of 48) (p = 0.005). CS cell lines harboring Signature-3/HRD (3 OCS/1 UCS) were significantly more sensitive to olaparib when compared to HRP cell lines (5 UCS/1 OCS) [mean ICOCS and UCS cell lines harboring HRD signature-3 were significantly more sensitive to olaparib in vitro and in vivo when compared to HRP CS. Clinical studies with PARPi in CS patients with a dominant signature 3 (HRD-related) are warranted.
- Published
- 2022
13. Supplementary Figure 1 from Downregulation of Filamin A Interacting Protein 1-Like is Associated with Promoter Methylation and Induces an Invasive Phenotype in Ovarian Cancer
- Author
-
Mijung Kwon, Steven K. Libutti, Gary L. Goldberg, Gloria S. Huang, Stephen M. Hewitt, Joon-Yong Chung, Kathleen D. Whitney, Folashade Adeshuko, Soo Jin Lee, Aneesa Gaffar, and Elizabeth R. Burton
- Abstract
PDF file - 123K
- Published
- 2023
14. Data from In Vitro and In Vivo Activity of IMGN853, an Antibody–Drug Conjugate Targeting Folate Receptor Alpha Linked to DM4, in Biologically Aggressive Endometrial Cancers
- Author
-
Alessandro D. Santin, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Jose F. Ponte, Ashley Morneault, Katherine Dugan, Ghanshyam Yadav, Chanhee Han, Luca Zammataro, Jacqueline Feinberg, Francesco Riccio, Anna Bianchi, Francesca Pettinella, Gulden Menderes, Tomomi Egawa-Takata, Stefania Bellone, Elena Bonazzoli, and Gary Altwerger
- Abstract
Grade 3 endometrioid and uterine serous carcinomas (USC) account for the vast majority of endometrial cancer deaths. The purpose of this study was to determine folic acid receptor alpha (FRα) expression in these biologically aggressive (type II) endometrial cancers and evaluate FRα as a targetable receptor for IMGN853 (mirvetuximab soravtansine). The expression of FRα was evaluated by immunohistochemistry (IHC) and flow cytometry in 90 endometrioid and USC samples. The in vitro cytotoxic activity and bystander effect were studied in primary uterine cancer cell lines expressing differential levels of FRα. In vivo antitumor efficacy of IMGN853 was evaluated in xenograft/patient-derived xenograft (PDX) models. Semiquantitative IHC analysis indicated that 41% of the USC patients overexpress FRα. Further, overexpression of FRα (i.e., 2+) was detected via flow cytometry in 22% (2/9) of primary endometrioid and in 27% (3/11) of primary USC cell lines. Increased cytotoxicity was seen with IMGN853 treatment compared with control in 2+ expressing uterine tumor cell lines. In contrast, tumor cell lines with low FRα showed no difference when exposed to IMGN853 versus control. IMGN853 induced bystander killing of FRα = 0 tumor cells. In an endometrioid xenograft model (END(K)265), harboring 2+ FRα, IMGN853 treatment showed complete resolution of tumors (P < 0.001). Treatment with IMGN853 in the USC PDX model (BIO(K)1), expressing 2+ FRα, induced twofold increase in median survival (P < 0.001). IMGN853 shows impressive antitumor activity in biologically aggressive FRα 2+ uterine cancers. These preclinical data suggest that patients with chemotherapy resistant/recurrent endometrial cancer overexpressing FRα may benefit from this treatment. Mol Cancer Ther; 17(5); 1003–11. ©2018 AACR.
- Published
- 2023
15. Supplementary Figure 3 from In Vitro and In Vivo Activity of IMGN853, an Antibody–Drug Conjugate Targeting Folate Receptor Alpha Linked to DM4, in Biologically Aggressive Endometrial Cancers
- Author
-
Alessandro D. Santin, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Jose F. Ponte, Ashley Morneault, Katherine Dugan, Ghanshyam Yadav, Chanhee Han, Luca Zammataro, Jacqueline Feinberg, Francesco Riccio, Anna Bianchi, Francesca Pettinella, Gulden Menderes, Tomomi Egawa-Takata, Stefania Bellone, Elena Bonazzoli, and Gary Altwerger
- Abstract
Determination by ICso of IMGN853 cytotoxicity compared to controls, ADC isotype and M9346A, in primary USC cell lines (ie, END(K) 149 and ARK19) with 2+ FRα expression
- Published
- 2023
16. Supplementary Table 1 from In Vitro and In Vivo Activity of IMGN853, an Antibody–Drug Conjugate Targeting Folate Receptor Alpha Linked to DM4, in Biologically Aggressive Endometrial Cancers
- Author
-
Alessandro D. Santin, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Jose F. Ponte, Ashley Morneault, Katherine Dugan, Ghanshyam Yadav, Chanhee Han, Luca Zammataro, Jacqueline Feinberg, Francesco Riccio, Anna Bianchi, Francesca Pettinella, Gulden Menderes, Tomomi Egawa-Takata, Stefania Bellone, Elena Bonazzoli, and Gary Altwerger
- Abstract
Endometrioid cell lines with demographics, stage, histologic grade, primary site of tumor, Mean fluorescence intensity (MFI) and score for FRα (folic acid alpha receptor).
- Published
- 2023
17. Supplementary Figure 2 from In Vitro and In Vivo Activity of IMGN853, an Antibody–Drug Conjugate Targeting Folate Receptor Alpha Linked to DM4, in Biologically Aggressive Endometrial Cancers
- Author
-
Alessandro D. Santin, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Jose F. Ponte, Ashley Morneault, Katherine Dugan, Ghanshyam Yadav, Chanhee Han, Luca Zammataro, Jacqueline Feinberg, Francesco Riccio, Anna Bianchi, Francesca Pettinella, Gulden Menderes, Tomomi Egawa-Takata, Stefania Bellone, Elena Bonazzoli, and Gary Altwerger
- Abstract
Immunohistochemistry (IHC) showing expression of FRα uterine serous cancer (USC) cell blocks
- Published
- 2023
18. Data from Downregulation of Filamin A Interacting Protein 1-Like is Associated with Promoter Methylation and Induces an Invasive Phenotype in Ovarian Cancer
- Author
-
Mijung Kwon, Steven K. Libutti, Gary L. Goldberg, Gloria S. Huang, Stephen M. Hewitt, Joon-Yong Chung, Kathleen D. Whitney, Folashade Adeshuko, Soo Jin Lee, Aneesa Gaffar, and Elizabeth R. Burton
- Abstract
Ovarian cancer is the most lethal gynecologic malignancy with a five-year survival rate below 25% for patients with stages III and IV disease. Identifying key mediators of ovarian cancer invasion and metastasis is critical to the development of more effective therapeutic interventions. We previously identified Filamin A interacting protein 1-like (FILIP1L) as an important mediator of cell proliferation and migration. In addition, targeted expression of FILIP1L in tumors inhibited tumor growth in vivo. In our present study, we confirmed that both mRNA and protein expression of FILIP1L were downregulated in ovarian cancer cells compared with normal ovarian epithelial cells. FILIP1L expression was inversely correlated with the invasive potential of ovarian cancer cell lines and clinical ovarian cancer specimens. We also provide evidence that DNA methylation is a mechanism by which FILIP1L is downregulated in ovarian cancer. The CpG island in the FILIP1L promoter was heavily methylated in ovarian cancer cells. Methylation status of the FILIP1L promoter was inversely correlated with FILIP1L expression in ovarian cell lines and clinical ovarian specimens. Reduced methylation in the FILIP1L promoter following treatment with a DNA demethylating agent was associated with restoration of FILIP1L expression in ovarian cancer cells. A transcription activator, cAMP-responsive element binding protein (CREB) was shown to bind to the CREB/ATF site in the CpG island of the FILIP1L promoter. Overall, these findings suggest that downregulation of FILIP1L associated with DNA methylation is related with the invasive phenotype in ovarian cancer and that modulation of FILIP1L expression has the potential to be a target for ovarian cancer therapy. Mol Cancer Res; 9(8); 1126–38. ©2011 AACR.
- Published
- 2023
19. Supplementary Figure Legends 1-3 from Downregulation of Filamin A Interacting Protein 1-Like is Associated with Promoter Methylation and Induces an Invasive Phenotype in Ovarian Cancer
- Author
-
Mijung Kwon, Steven K. Libutti, Gary L. Goldberg, Gloria S. Huang, Stephen M. Hewitt, Joon-Yong Chung, Kathleen D. Whitney, Folashade Adeshuko, Soo Jin Lee, Aneesa Gaffar, and Elizabeth R. Burton
- Abstract
PDF file - 71K
- Published
- 2023
20. Supplementary Figure 1 from In Vitro and In Vivo Activity of IMGN853, an Antibody–Drug Conjugate Targeting Folate Receptor Alpha Linked to DM4, in Biologically Aggressive Endometrial Cancers
- Author
-
Alessandro D. Santin, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Jose F. Ponte, Ashley Morneault, Katherine Dugan, Ghanshyam Yadav, Chanhee Han, Luca Zammataro, Jacqueline Feinberg, Francesco Riccio, Anna Bianchi, Francesca Pettinella, Gulden Menderes, Tomomi Egawa-Takata, Stefania Bellone, Elena Bonazzoli, and Gary Altwerger
- Abstract
Immunohistochemistry (IHC) showing expression of FRα endometrioid cancer cell blocks
- Published
- 2023
21. Supplementary Figure 3 from Downregulation of Filamin A Interacting Protein 1-Like is Associated with Promoter Methylation and Induces an Invasive Phenotype in Ovarian Cancer
- Author
-
Mijung Kwon, Steven K. Libutti, Gary L. Goldberg, Gloria S. Huang, Stephen M. Hewitt, Joon-Yong Chung, Kathleen D. Whitney, Folashade Adeshuko, Soo Jin Lee, Aneesa Gaffar, and Elizabeth R. Burton
- Abstract
PDF file - 78K
- Published
- 2023
22. Supplementary Table 2 from In Vitro and In Vivo Activity of IMGN853, an Antibody–Drug Conjugate Targeting Folate Receptor Alpha Linked to DM4, in Biologically Aggressive Endometrial Cancers
- Author
-
Alessandro D. Santin, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Jose F. Ponte, Ashley Morneault, Katherine Dugan, Ghanshyam Yadav, Chanhee Han, Luca Zammataro, Jacqueline Feinberg, Francesco Riccio, Anna Bianchi, Francesca Pettinella, Gulden Menderes, Tomomi Egawa-Takata, Stefania Bellone, Elena Bonazzoli, and Gary Altwerger
- Abstract
SUPPLEMENTAL Table 2: Uterine serous cell lines with demographics, stage, histologic grade, primary site of tumor, Mean fluorescence intensity (MFI) and score for FRα (folic acid alpha receptor).
- Published
- 2023
23. Supplementary Figure 2 from Downregulation of Filamin A Interacting Protein 1-Like is Associated with Promoter Methylation and Induces an Invasive Phenotype in Ovarian Cancer
- Author
-
Mijung Kwon, Steven K. Libutti, Gary L. Goldberg, Gloria S. Huang, Stephen M. Hewitt, Joon-Yong Chung, Kathleen D. Whitney, Folashade Adeshuko, Soo Jin Lee, Aneesa Gaffar, and Elizabeth R. Burton
- Abstract
PDF file - 159K
- Published
- 2023
24. Supplementary Table 2 from Downregulation of Filamin A Interacting Protein 1-Like is Associated with Promoter Methylation and Induces an Invasive Phenotype in Ovarian Cancer
- Author
-
Mijung Kwon, Steven K. Libutti, Gary L. Goldberg, Gloria S. Huang, Stephen M. Hewitt, Joon-Yong Chung, Kathleen D. Whitney, Folashade Adeshuko, Soo Jin Lee, Aneesa Gaffar, and Elizabeth R. Burton
- Abstract
PDF file - 152K
- Published
- 2023
25. Supplemental Fig S1 and S2 from RNA-seq Identification of RACGAP1 as a Metastatic Driver in Uterine Carcinosarcoma
- Author
-
Gloria S. Huang, Deyou Zheng, Gary L. Goldberg, Marc J. Gunter, Tiffany Hebert, David Smotkin, Jennifer Heim, Jurriaan Brouwer-Visser, Mingyan Lin, and Shijun Mi
- Abstract
Figure S1. Correlation of RACGAP1 Immunohistochemical Staining and mRNA Expression; Figure S2. Survivin Expression in CS99 Treated With YM155
- Published
- 2023
26. Data from Inhibition of BET Bromodomain Proteins with GS-5829 and GS-626510 in Uterine Serous Carcinoma, a Biologically Aggressive Variant of Endometrial Cancer
- Author
-
Alessandro D. Santin, Joseph Schlessinger, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Paola Manara, Aranzazu Manzano, Salvatore Lopez, Ghanshyam Yadav, Chanhee Han, Francesco Riccio, Francesca Pettinella, Anna Bianchi, Luca Zammataro, Gulden Menderes, Gary Altwerger, Stefania Bellone, Emiliano Cocco, Federica Predolini, and Elena Bonazzoli
- Abstract
Purpose: Uterine serous carcinoma (USC) is a rare and aggressive variant of endometrial cancer. Whole-exome sequencing (WES) studies have recently reported c-Myc gene amplification in a large number of USCs, suggesting c-Myc as a potential therapeutic target. We investigated the activity of novel BET bromodomain inhibitors (GS-5829 and GS-626510, Gilead Sciences Inc.) and JQ1 against primary USC cultures and USC xenografts.Experimental Design: We evaluated c-Myc expression by qRT-PCR in a total of 45 USCs including fresh-frozen tumor tissues and primary USC cell lines. We also performed IHC and Western blot experiments in 8 USC tumors. USC cultures were evaluated for sensitivity to GS-5829, GS-626510, and JQ1 in vitro using proliferation, viability, and apoptosis assays. Finally, the in vivo activity of GS-5829, GS-626510, and JQ1 was studied in USC-ARK1 and USC-ARK2 mouse xenografts.Results: Fresh-frozen USC and primary USC cell lines overexpressed c-Myc when compared with normal tissues (P = 0.0009 and 0.0083, respectively). High c-Myc expression was found in 7 of 8 of primary USC cell lines tested by qRT-PCR and 5 of 8 tested by IHC. In vitro experiments demonstrated high sensitivity of USC cell lines to the exposure to GS-5829, GS-626510, and JQ1 with BET inhibitors causing a dose-dependent decrease in the phosphorylated levels of c-Myc and a dose-dependent increase in caspase activation (apoptosis). In comparative in vivo experiments, GS-5829 and/or GS-626510 were found more effective than JQ1 at the concentrations/doses used in decreasing tumor growth in both USC-ARK1 and USC-ARK2 mouse xenograft models.Conclusions: GS-5829 and GS-626510 may represent novel, highly effective therapeutics agents against recurrent/chemotherapy-resistant USC-overexpressing c-Myc. Clinical studies with GS-5829 in patients with USC harboring chemotherapy-resistant disease are warranted. Clin Cancer Res; 24(19); 4845–53. ©2018 AACR.
- Published
- 2023
27. Table S1 from Inhibition of BET Bromodomain Proteins with GS-5829 and GS-626510 in Uterine Serous Carcinoma, a Biologically Aggressive Variant of Endometrial Cancer
- Author
-
Alessandro D. Santin, Joseph Schlessinger, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Paola Manara, Aranzazu Manzano, Salvatore Lopez, Ghanshyam Yadav, Chanhee Han, Francesco Riccio, Francesca Pettinella, Anna Bianchi, Luca Zammataro, Gulden Menderes, Gary Altwerger, Stefania Bellone, Emiliano Cocco, Federica Predolini, and Elena Bonazzoli
- Abstract
Cell lines characteristics and tissue source.
- Published
- 2023
28. Suppl Table S4 from RNA-seq Identification of RACGAP1 as a Metastatic Driver in Uterine Carcinosarcoma
- Author
-
Gloria S. Huang, Deyou Zheng, Gary L. Goldberg, Marc J. Gunter, Tiffany Hebert, David Smotkin, Jennifer Heim, Jurriaan Brouwer-Visser, Mingyan Lin, and Shijun Mi
- Abstract
Table S4. Short Tandem Repeat Profiling of Primary Cell Line DNA and Patient Tumor Tissue DNA.
- Published
- 2023
29. Supplementary Data from Insulin-like Growth Factor 2 Expression Modulates Taxol Resistance and Is a Candidate Biomarker for Reduced Disease-Free Survival in Ovarian Cancer
- Author
-
Susan Band Horwitz, Harriet O. Smith, Gary L. Goldberg, Eric R. Prossnitz, Clifford R. Qualls, Hugo Arias-Pulido, Juan Lin, Tiffany M. Hebert, Christine H. Kim, Marissa J. Ramirez, Jurriaan Brouwer-Visser, and Gloria S. Huang
- Abstract
Supplementary Data from Insulin-like Growth Factor 2 Expression Modulates Taxol Resistance and Is a Candidate Biomarker for Reduced Disease-Free Survival in Ovarian Cancer
- Published
- 2023
30. Data from Potentiation of Taxol Efficacy by Discodermolide in Ovarian Carcinoma Xenograft-Bearing Mice
- Author
-
Hayley M. McDaid, Susan Band Horwitz, Gary L. Goldberg, Amos B. Smith, B. Scott Freeze, Lluis Lopez-Barcons, and Gloria S. Huang
- Abstract
Purpose: To evaluate the drug combination of discodermolide and Taxol in human ovarian cancer cells and in an in vivo model of ovarian carcinoma.Experimental Design: The combination index method was used to evaluate the interaction of Taxol and discodermolide in human ovarian SKOV-3 carcinoma cells. Data were correlated with alterations in cell cycle distribution and caspase activation. In addition, SKOV-3 xenograft-bearing mice were treated with either Taxol, discodermolide, or a combination of both drugs given concurrently to evaluate the antitumor efficacy and toxicity of this combination. The Matrigel plug assay and CD31 immunohistochemistry were done to assess antiangiogenic effects.Results: Taxol and discodermolide interact synergistically over a range of concentrations and molar ratios that cause drug-induced aneuploidy in ovarian carcinoma cells. In SKOV-3 xenograft-bearing mice, the combination is significantly superior to either single agent, and induces tumor regressions without notable toxicities. Immunohistochemical analysis of CD31 and Matrigel plug analysis show decreased vessel formation in mice treated with the combination relative to either drug alone.Conclusions: The synergistic activity of Taxol and discodermolide in cells is most potent at drug concentrations that result in drug-induced aneuploidy rather than mitotic arrest. Moreover, in an animal model of ovarian carcinoma, this is a well-tolerated combination that induces tumor regressions and suppresses angiogenesis. These data confirm the potency of this combination and support the use of concurrent low doses of Taxol and discodermolide for potential use in cancer therapeutics.
- Published
- 2023
31. Supplementary Figure S1 from Potentiation of Taxol Efficacy by Discodermolide in Ovarian Carcinoma Xenograft-Bearing Mice
- Author
-
Hayley M. McDaid, Susan Band Horwitz, Gary L. Goldberg, Amos B. Smith, B. Scott Freeze, Lluis Lopez-Barcons, and Gloria S. Huang
- Abstract
Supplementary Figure S1 from Potentiation of Taxol Efficacy by Discodermolide in Ovarian Carcinoma Xenograft-Bearing Mice
- Published
- 2023
32. Supplemental Figure and Table Legends from RNA-seq Identification of RACGAP1 as a Metastatic Driver in Uterine Carcinosarcoma
- Author
-
Gloria S. Huang, Deyou Zheng, Gary L. Goldberg, Marc J. Gunter, Tiffany Hebert, David Smotkin, Jennifer Heim, Jurriaan Brouwer-Visser, Mingyan Lin, and Shijun Mi
- Abstract
Supplemental Figure and Table Legends
- Published
- 2023
33. Figure S1 from Inhibition of BET Bromodomain Proteins with GS-5829 and GS-626510 in Uterine Serous Carcinoma, a Biologically Aggressive Variant of Endometrial Cancer
- Author
-
Alessandro D. Santin, Joseph Schlessinger, Peter E. Schwartz, Masoud Azodi, Gloria S. Huang, Dan-Arin Silasi, Elena Ratner, Babak Litkouhi, Serena Wong, Pei Hui, Natalia Buza, Paola Manara, Aranzazu Manzano, Salvatore Lopez, Ghanshyam Yadav, Chanhee Han, Francesco Riccio, Francesca Pettinella, Anna Bianchi, Luca Zammataro, Gulden Menderes, Gary Altwerger, Stefania Bellone, Emiliano Cocco, Federica Predolini, and Elena Bonazzoli
- Abstract
Ex vivo c-Myc expression assessed by IHC on the ARK2 xenograft tumor samples, excised 1 hour and 6 hours after a single dose of GS-5829 at 40, 20, 10 mg/kg, GS-626510 at 10 mg/kg and vehicle for oral gavage studies.
- Published
- 2023
34. Data from Insulin-like Growth Factor 2 Expression Modulates Taxol Resistance and Is a Candidate Biomarker for Reduced Disease-Free Survival in Ovarian Cancer
- Author
-
Susan Band Horwitz, Harriet O. Smith, Gary L. Goldberg, Eric R. Prossnitz, Clifford R. Qualls, Hugo Arias-Pulido, Juan Lin, Tiffany M. Hebert, Christine H. Kim, Marissa J. Ramirez, Jurriaan Brouwer-Visser, and Gloria S. Huang
- Abstract
Purpose: This study was undertaken to examine the role of the insulin-like growth factor (IGF) signaling pathway in the response of ovarian cancer cells to Taxol and to evaluate the significance of this pathway in human epithelial ovarian tumors.Experimental Design: The effect of Taxol treatment on AKT activation in A2780 ovarian carcinoma cells was evaluated using antibodies specific for phospho-AKT. To study the drug-resistant phenotype, we developed a Taxol-resistant cell line, HEY-T30, derived from HEY ovarian carcinoma cells. IGF2 expression was measured by real-time PCR. A type 1 IGF receptor (IGF1R) inhibitor, NVP-AEW541, and IGF2 small interfering RNA were used to evaluate the effect of IGF pathway inhibition on proliferation and Taxol sensitivity. IGF2 protein expression was evaluated by immunohistochemistry in 115 epithelial ovarian tumors and analyzed in relation to clinical/pathologic factors using the χ2 or Fisher's exact tests. The influence of IGF2 expression on survival was studied with Cox regression.Results: Taxol-induced AKT phosphorylation required IGF1R tyrosine kinase activity and was associated with upregulation of IGF2. Resistant cells had higher IGF2 expression compared with sensitive cells, and IGF pathway inhibition restored sensitivity to Taxol. High IGF2 tumor expression correlated with advanced stage (P < 0.001) and tumor grade (P < 0.01) and reduced disease-free survival (P < 0.05).Conclusions: IGF2 modulates Taxol resistance, and tumor IGF2 expression is a candidate prognostic biomarker in epithelial ovarian tumors. IGF pathway inhibition sensitizes drug-resistant ovarian carcinoma cells to Taxol. Such novel findings suggest that IGF2 represents a therapeutic target in ovarian cancer, particularly in the setting of Taxol resistance. Clin Cancer Res; 16(11); 2999–3010. ©2010 AACR.
- Published
- 2023
35. Data from RNA-seq Identification of RACGAP1 as a Metastatic Driver in Uterine Carcinosarcoma
- Author
-
Gloria S. Huang, Deyou Zheng, Gary L. Goldberg, Marc J. Gunter, Tiffany Hebert, David Smotkin, Jennifer Heim, Jurriaan Brouwer-Visser, Mingyan Lin, and Shijun Mi
- Abstract
Purpose: Uterine carcinosarcoma is a rare aggressive malignancy frequently presenting at advanced stage of disease with extrauterine metastases. Median survival is less than 2 years due to high relapse rates after surgery and poor response to chemotherapy or radiotherapy. The goal of this study was to identify novel therapeutic targets.Experimental Design: We applied RNA-seq analysis to prospectively collected uterine carcinosarcoma tumor samples from patients undergoing primary surgical resection and for comparison, normal endometrial tissues from postmenopausal women undergoing hysterectomy for benign indications. Functional assays were done in primary carcinosarcoma cell lines developed from patients and in established cell lines, as well as a cell line–derived xenograft model. Validation was done by analysis of an independent cohort of patients with uterine carcinosarcoma from The Cancer Genome Atlas (TCGA).Results: Rac GTPase–activating protein 1 (RACGAP1) was identified to be highly upregulated in uterine carcinosarcoma. Functional assays showed that RACGAP1 mediates motility and invasion via regulation of STAT3 phosphorylation and survivin expression. RACGAP1 depletion or survivin inhibition abrogated motility and invasiveness of carcinosarcoma cells, while RACGAP1 overexpression conferred invasiveness to endometrial adenocarcinoma cells. In the TCGA cohort, RACGAP1 expression correlated with survivin expression and extrauterine spread of disease.Conclusions: The RACGAP1–STAT3–survivin signaling pathway is required for the invasive phenotype of uterine carcinosarcoma and is a newly identified therapeutic target in this lethal disease. Clin Cancer Res; 22(18); 4676–86. ©2016 AACR.
- Published
- 2023
36. Sex Hormones, Insulin, and Insulin-like Growth Factors in Recurrence of High-Stage Endometrial Cancer
- Author
-
David Miller, Alan D. Hutson, Nilsa C. Ramirez, Gloria S. Huang, Louise A. Brinton, Michael J. Birrer, Herbert Yu, Mark H. Einstein, Heather A. Lankes, Melissa A. Merritt, Howard D. Strickler, Mark E. Sherman, Xiaonan Xue, Thomas E. Rohan, and Marc J. Gunter
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Epidemiology ,medicine.drug_class ,medicine.medical_treatment ,Estrone ,Adenocarcinoma ,Gastroenterology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Sex hormone-binding globulin ,Somatomedins ,Internal medicine ,Biomarkers, Tumor ,Humans ,Insulin ,Medicine ,Prospective Studies ,Gonadal Steroid Hormones ,Prospective cohort study ,Aged ,Neoplasm Staging ,biology ,business.industry ,Gene Expression Profiling ,Endometrial cancer ,Middle Aged ,medicine.disease ,Endometrial Neoplasms ,Insulin receptor ,030104 developmental biology ,Oncology ,chemistry ,Estrogen ,030220 oncology & carcinogenesis ,Disease Progression ,biology.protein ,Female ,Neoplasm Recurrence, Local ,business ,Hormone - Abstract
Background: The influence of sex hormone and insulin/insulin-like growth factor (IGF) axis signaling on endometrial cancer recurrence is unknown. We evaluated these pathways in a prospective cohort of Gynecologic Oncology Group (GOG)0210 trial endometrial adenocarcinoma patients. Methods: Stage II–IV patients (N = 816) were included in this study. Pretreatment specimens were tested for tumor mRNA and protein expression of IGF1, IGF2, IGF-binding proteins (IGFBP)-1 and -3, insulin (IR) and IGF-I receptors (IGF1R), phosphorylated IR/IGF1R (pIGF1R/pIR), and estrogen (ER) and progesterone receptors (PR) using qPCR and IHC. Serum concentrations of insulin, IGF-I, IGFBP-3, estradiol, estrone, and sex hormone binding globulin were measured. HRs and 95% confidence intervals (CI) for progression-free survival were calculated from Cox models adjusting for age, stage, and grade. Results: Recurrence occurred in 280 (34%) cases during a median of 4.6 years of follow-up. ER positivity (HR, 0.67; 95% CI, 0.47–0.95), IR positivity (HR, 0.53; 95% CI, 0.29–0.98), and circulating IGF-I (highest vs. lowest quartile: HR, 0.66; 95% CI, 0.47–0.92) were inversely associated with recurrence risk. Circulating estradiol (highest vs. lowest tertile: HR, 1.55; 95% CI, 1.02–2.36) and pIGF1R/pIR positivity (HR, 1.40; 95% CI, 1.02–1.92) were associated with increased recurrence risk. Conclusions: Circulating estradiol and tumor tissue phosphorylated (activated) IGR1R/IR were independently associated with higher risk of recurrence in patients with endometrial cancer. Impact: This study may inform future clinical trials of endocrine-targeted adjuvant therapies in patients with endometrial cancer that could include baseline assessment of serum and tissue biomarkers of estradiol and insulin signaling pathways.
- Published
- 2021
37. Derangements in HUWE1/c-MYC pathway confer sensitivity to the BET bromodomain inhibitor GS-626510 in uterine cervical carcinoma
- Author
-
Joan Tymon-Rosario, Gloria S. Huang, Burak Zeybek, Adele Guglielmi, Stefania Bellone, Peter E. Schwartz, Paola Manara, Elena Bonazzoli, Barbara Gnutti, Nupur Nagarkatti, Vaagn Andikyan, Elena Ratner, Alessandro D. Santin, Gary Altwerger, Silvia Pelligra, Dan-Arin Silasi, Luca Zammataro, Chanhee Han, Masoud Azodi, and Gulden Menderes
- Subjects
Adult ,0301 basic medicine ,Ubiquitin-Protein Ligases ,medicine.medical_treatment ,Uterine Cervical Neoplasms ,Article ,Proto-Oncogene Proteins c-myc ,Loss of heterozygosity ,BET inhibitor ,Mice ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,Cervical carcinoma ,Animals ,Humans ,Medicine ,Gene silencing ,In Situ Hybridization, Fluorescence ,Aged ,Chemotherapy ,business.industry ,Tumor Suppressor Proteins ,Imidazoles ,Proteins ,Obstetrics and Gynecology ,Isoxazoles ,Middle Aged ,Xenograft Model Antitumor Assays ,Bromodomain ,030104 developmental biology ,Real-time polymerase chain reaction ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business ,Signal Transduction - Abstract
Objective Whole-exome-sequencing (WES) studies reported c-MYC gene-amplification and HUWE1 gene deletion/mutations in a significant number of cervical-cancer-patients (CC) suggesting HUWE1/c-MYC pathway as potential therapeutic target. We investigated HUWE1/c-MYC expression in fresh-frozen-CC and the activity of the novel BET inhibitor GS-626510 (Gilead-Science-Inc) against primary WES CC-cultures and CC-xenografts. Methods HUWE1 and c-MYC expression were evaluated by qRT-PCR in 23 CC including 12 fresh-frozen-tumor-tissues and 11 primary-cell-lines. c-Myc expression was also evaluated by Western-Blot (WB) and fluorescence-in-situ-hybridization (FISH) in all 11 fully sequenced primary-CC-cell-lines. Primary tumors were evaluated for sensitivity to GS-626510 in-vitro using proliferation and viability-assays. siRNA experiments were used to evaluate the effect of HUWE1 silencing on primary-CC-cell-line growth and sensitivity to GS-626510. Finally, the in-vivo activity of GS-626510 was studied in CC-CVX8-mouse-xenografts. Results Fresh-frozen-CC and primary-CC-cell-lines overexpressed c-MYC when compared to normal tissues (p = .01). FISH demonstrated amplification of c-MYC in 9/11 (82%) of the primary-CC-cell-lines. Cell-lines with derangements in HUWE1/c-MYC pathway were highly sensitive to GS-626510, with a dose-response decrease in cell proliferation and viability. siRNA silencing of HUWE1 significantly increased c-MYC expression and CC cell-proliferation and enhanced the in-vitro sensitivity to GS-626510. Twice-daily oral doses of GS-626510 were well tolerated in-vivo and highly effective in decreasing tumor-growth (p = .004) and increasing survival (p = .004) of CC-CVX8 xenografts. Conclusions Downregulation/inactivation of HUWE1 may increase c-MYC expression and proliferation in primary-CC-cell-lines. GS-626510 may represent a novel, potentially highly effective therapeutic agent against CC overexpressing c-MYC and/or harboring HUWE1 mutations. Clinical studies with BET inhibitor in CC-patients harboring radiation/chemotherapy-resistant disease are warranted.
- Published
- 2020
38. DHES0815A, a novel antibody-drug conjugate targeting HER2/neu, is highly active against uterine serous carcinomas in vitro and in vivo
- Author
-
Pei Hui, Elena Ratner, Peter E. Schwartz, Masoud Azodi, Natalia Buza, Stefania Bellone, Paola Manara, Burak Zeybek, Mitchell Clark, Aranzazu Manzano, Gary Altwerger, Silvia Pelligra, Elena Bonazzoli, Gulden Menderes, Luca Zammataro, Dennis Mauricio, Gloria S. Huang, Barbara Gnutti, Justin Harold, Vaagn Andikyan, Nupur Nagarkatti, Alessandro D. Santin, Adele Guglielmi, Dan-Arin Silasi, and Joan Tymon-Rosario
- Subjects
Adult ,Antibody-drug conjugate ,Immunoconjugates ,Receptor, ErbB-2 ,Primary Cell Culture ,Antibodies, Monoclonal, Humanized ,HER2/neu ,Article ,Uterine serous carcinoma ,Benzodiazepines ,Trastuzumab ,In vivo ,Cell Line, Tumor ,Medicine ,Humans ,skin and connective tissue diseases ,neoplasms ,Aged ,Aged, 80 and over ,biology ,business.industry ,Obstetrics and Gynecology ,Bystander Effect ,Middle Aged ,medicine.disease ,Xenograft Model Antitumor Assays ,Cystadenocarcinoma, Serous ,Serous fluid ,Oncology ,Drug Resistance, Neoplasm ,Uterine Neoplasms ,Cancer research ,biology.protein ,Immunohistochemistry ,Female ,Pertuzumab ,business ,medicine.drug - Abstract
OBJECTIVE: Uterine serous carcinoma (USC) is an aggressive histologic variant of endometrial cancer which portends a poor prognosis. DHES0815A is a novel antibody-drug-conjugate (ADC) which binds specifically to HER2 overexpressing tumors at a distinct epitope from that bound by trastuzumab and pertuzumab after which it delivers the toxic payload, PBD-MA, a DNA mono-alkylating agent. The objective of this study was to evaluate the preclinical activity of DHES0815A against primary USC cell lines and xenografts. METHODS: Twelve primary USC cell lines were assessed by immunohistochemistry (IHC) for HER2 protein expression and for C-erbB2 gene amplification using fluorescent in situ hybridization (FISH) analysis. Cell viability and bystander killing in USC cell lines after exposure to DHES0815A, the non-targeted ADC, and the unconjugated antibody (i.e. MHES0488A) were evaluated using flow cytometry-based-assays. In vivo activity of DHES0815A was tested against HER2/neu overexpressing USC xenografts. RESULTS: High HER2/neu protein expression was seen in 25% (3/12) of the primary USC cell lines. USC cell lines overexpressing HER2/neu were significantly more sensitive to DHES0815A when compared to the non-targeted control ADC (p
- Published
- 2021
39. Trastuzumab tolerability in the treatment of advanced (stage III-IV) or recurrent uterine serous carcinomas that overexpress HER2/neu
- Author
-
Gulden Menderes, Peter E. Schwartz, Burak Zeybek, Alessandro D. Santin, Dennis Mauricio, Naomi N Adjei, Amanda N. Fader, Gary Altwerger, Masoud Azodi, Stefania Bellone, Joan Tymon-Rosario, Justin Harold, Vaagn Andikyan, Eric R. Siegel, Gloria S. Huang, Mitchell Clark, and Elena Ratner
- Subjects
medicine.medical_specialty ,Receptor, ErbB-2 ,medicine.medical_treatment ,Gastroenterology ,HER2/neu ,Article ,Uterine serous carcinoma ,chemistry.chemical_compound ,Maintenance therapy ,Trastuzumab ,Internal medicine ,medicine ,Humans ,Aged ,Neoplasm Staging ,Chemotherapy ,biology ,business.industry ,Endometrial cancer ,Obstetrics and Gynecology ,Middle Aged ,medicine.disease ,Carboplatin ,Cystadenocarcinoma, Serous ,Oncology ,chemistry ,Tolerability ,Uterine Neoplasms ,biology.protein ,Female ,Neoplasm Recurrence, Local ,business ,medicine.drug - Abstract
Objective Due to previously reported trastuzumab safety concerns and the scant data available in endometrial cancer patients, we sought to assess the safety, tolerability and toxicity profile of trastuzumab in patients with advanced/recurrent uterine serous carcinoma (USC) that overexpress HER2/neu in our multicenter randomized phase II trial. Methods Patients were randomized 1:1 to receive carboplatin/paclitaxel (C/P) for 6 cycles ± trastuzumab (T) with the experimental arm continuing to receive single agent trastuzumab maintenance treatment until disease progression/toxicity. Progression-free-survival was the primary endpoint; overall-survival and toxicity were secondary endpoints. Adverse events (AEs) were compared between treatment arms. Results There were 28 patients in the C/P arm and 32 patients in the experimental (C/P + T) arm. Fifty-eight patients (97%) experienced 977 treatment-related AEs of which 875 (89.6%) were low-grade (grade 1–2) and 102 (10.4%) were high-grade (grade 3–5). The mean ± standard deviation of AEs per patient was 15.5 ± 16.3 in the C/P arm and 17.0 ± 16.0 in the C/P + T arm. Gastrointestinal AEs were the most common in both arms (n = 155, 15.7%) of which 94.2% were low-grade (n = 146). Importantly, no significant difference between treatment arms was detected in any system-organ class of AE including cardiac AE. Five (17%) of 29 patients who received prolonged trastuzumab maintenance therapy had no sign of cumulative toxicity after an average (range) of 5.1 (4.2–6.3) years. Conclusions Trastuzumab appears to be safe and has a manageable toxicity profile both when used in combination with chemotherapy and when used for single agent maintenance in patients with HER2/neu positive USC. This safety profile is reassuring given the proven efficacy of trastuzumab in advanced/recurrent HER2/neu positive USC.
- Published
- 2021
40. A phase 2 evaluation of pembrolizumab for recurrent Lynch-like versus sporadic endometrial cancers with microsatellite instability
- Author
-
Stefania Bellone, Dana M. Roque, Eric R. Siegel, Natalia Buza, Pei Hui, Elena Bonazzoli, Adele Guglielmi, Luca Zammataro, Nupur Nagarkatti, Samir Zaidi, Jungsoo Lee, Dan‐Arin Silasi, Gloria S. Huang, Vaagn Andikyan, Shari Damast, Mitchell Clark, Masoud Azodi, Peter E. Schwartz, Joan R. Tymon‐Rosario, Justin A. Harold, Dennis Mauricio, Burak Zeybek, Gulden Menderes, Gary Altwerger, Elena Ratner, Ludmil B. Alexandrov, Akiko Iwasaki, Yong Kong, Eric Song, Weilai Dong, Julia A. Elvin, Jungmin Choi, and Alessandro D. Santin
- Subjects
Cancer Research ,Oncology ,Humans ,Female ,Microsatellite Instability ,Pilot Projects ,Prospective Studies ,Neoplasm Recurrence, Local ,Antibodies, Monoclonal, Humanized ,DNA Mismatch Repair ,Endometrial Neoplasms - Abstract
Microsatellite instability-high (MSI-H)/mismatch repair deficiency (dMMR) is a biomarker for responses to immune checkpoint inhibitors (ICIs). Whether mechanisms underlying microsatellite instability alter responses to ICIs is unclear. This article reports data from a prospective phase 2 pilot study of pembrolizumab in patients with recurrent MSI-H endometrial cancer (EC) analyzed by whole exome sequencing (WES) and potential mechanisms of primary/secondary ICI resistance (NCT02899793).Patients with measurable MSI-H/dMMR EC confirmed by polymerase chain reaction/immunohistochemistry were evaluated by WES and received 200 mg of pembrolizumab every 3 weeks for ≤2 years. The primary end point was the objective response rate (ORR). Secondary end points included progression-free survival (PFS) and overall survival (OS).Twenty-five patients (24 evaluable) were treated. Six patients (25%) harbored Lynch/Lynch-like tumors, whereas 18 (75%) had sporadic EC. The tumor mutation burden was higher in Lynch-like tumors (median, 2939 mutations/megabase [Mut/Mb]; interquartile range [IQR], 867-5108 Mut/Mb) than sporadic tumors (median, 604 Mut/Mb; IQR, 411-798 Mut/Mb; P = .0076). The ORR was 100% in Lynch/Lynch-like patients but only 44% in sporadic patients (P = .024). The 3-year PFS and OS proportions were 100% versus 30% (P = .017) and 100% versus 43% (P = .043), respectively.This study suggests prognostic significance of Lynch-like cancers versus sporadic MSI-H/dMMR ECs for ORR, PFS, and OS when patients are treated with pembrolizumab. Larger confirmatory studies in ECs and other MSI-H/dMMR tumors are necessary. Defective antigen processing/presentation and deranged induction in interferon responses serve as mechanisms of resistance in sporadic MSI-H ECs. Oligoprogression in MSI-H/dMMR patients appears salvageable with surgical resection and/or local treatment and the continuation of pembrolizumab off study. Clinical studies evaluating separate MSI-H/dMMR EC subtypes treated with ICIs are warranted.
- Published
- 2021
41. Prognostic impact of mismatch repair deficiency in high- and low-intermediate-risk, early-stage endometrial cancer following vaginal brachytherapy
- Author
-
Henry Park, Jessie Y. Li, Elena Ratner, Melissa R. Young, Shari Damast, Gloria S. Huang, and Alessandro D. Santin
- Subjects
Oncology ,Prognostic variable ,medicine.medical_specialty ,Brachytherapy ,DNA Mismatch Repair ,Disease-Free Survival ,Continuous variable ,Internal medicine ,medicine ,Humans ,Stage (cooking) ,Aged ,Neoplasm Staging ,Retrospective Studies ,business.industry ,Proportional hazards model ,Endometrial cancer ,Obstetrics and Gynecology ,Middle Aged ,medicine.disease ,Prognosis ,Endometrial Neoplasms ,Survival Rate ,Vaginal brachytherapy ,MISMATCH REPAIR DEFICIENCY ,Female ,Neoplasm Recurrence, Local ,business ,Intermediate risk ,Carcinoma, Endometrioid - Abstract
Objectives To examine the impact of mismatch repair (MMR) status on prognosis among patients with high- and low-intermediate-risk endometrioid endometrial cancer (EEC) treated with vaginal brachytherapy (VBT). Materials/methods 198 stage I-II EEC patients with known MMR status treated with adjuvant VBT were identified. Both low-intermediate (LIR) and high-intermediate-risk (HIR) patients were included. Clinical characteristics were compared between patients with proficient and deficient mismatch repair (pMMR and dMMR) using Fisher's exact tests for categorical variables and t-tests for continuous variables. Recurrence-free survival (RFS) and overall survival (OS) were analyzed with Kaplan-Meier estimates, the log-rank test, and Cox proportional hazards regression. Results Patients with dMMR compared to pMMR were more likely to have grade 2–3 tumors (75% vs. 57%, p = 0.006), lympho-vascular invasion (40% vs. 25%, p = 0.034), and HIR classification (65% vs. 49%, p = 0.011). Three-year RFS was inferior for dMMR compared to pMMR patients (75% vs. 96%, p = 0.001). dMMR patients compared to pMMR had similarly reduced 3-year RFS within the LIR (74% vs. 100%, p = 0.026) and HIR (75% vs. 91%, p = 0.038) subgroups. Three-year OS was not different between dMMR/pMMR patients (98% vs. 97%, p = 0.653) or HIR/LIR patients (97% vs. 97%, p = 0.999). On multivariable Cox regression, dMMR status was a significant prognostic variable for RFS (HR 3.774, CI 1.495–9.526, p = 0.005), though it was not significant for OS. Conclusion Following VBT, patients with dMMR have poorer RFS compared to pMMR patients regardless of HIR/LIR risk classification. The prognosis of intermediate-risk EEC patients may lie more on a continuum dependent on molecular features rather than distinct clinicopathologic risk categories.
- Published
- 2021
42. Randomised phase II trial of weekly ixabepilone ± biweekly bevacizumab for platinum-resistant or refractory ovarian/fallopian tube/primary peritoneal cancer
- Author
-
Dana M. Roque, Eric R. Siegel, Natalia Buza, Stefania Bellone, Dan-Arin Silasi, Gloria S. Huang, Vaagn Andikyan, Mitchell Clark, Masoud Azodi, Peter E. Schwartz, Gautam G. Rao, Jocelyn C. Reader, Pei Hui, Joan R. Tymon-Rosario, Justin Harold, Dennis Mauricio, Burak Zeybek, Gulden Menderes, Gary Altwerger, Elena Ratner, and Alessandro D. Santin
- Subjects
Bevacizumab ,Ovarian Neoplasms ,Cancer Research ,Oncology ,Epothilones ,Antineoplastic Combined Chemotherapy Protocols ,Fallopian Tube Neoplasms ,Humans ,Female ,Carcinoma, Ovarian Epithelial ,Fallopian Tubes ,Peritoneal Neoplasms ,Platinum - Abstract
This multi-center RP2 study assessed activity/safety of ixabepilone + bevacizumab compared to ixabepilone in platinum-resistant/refractory ovarian/fallopian tube/primary peritoneal cancer. Additional objectives were to examine the role of prior bevacizumab and taxanes, and explore class III-ß-tubulin (TUBB3) as a predictive biomarker.Participants were randomised to receive ixabepilone 20 mg/mAmong 76 evaluable patients who received IXA + BEV (n = 39) compared to IXA (n = 37), the ORR was 33% (n = 13) versus 8% (n = 3)(P = 0.004), durable at 6 months in 37% (n = 14) and 3% (n = 1) (P 0.001). BEV significantly improved PFS (median:5.5 vs 2.2 months, HR = 0.33, 95%CI 0.19-0.55, P 0.001) and OS (median:10.0 vs 6.0 months, HR = 0.52, 95%CI 0.31-0.87, P = 0.006). Both regimens were well-tolerated. TUBB3 expression did not predict response. Subgroup analyses revealed minimal effect of prior BEV or taxane resistant/refractory status on response to IXA + BEV.IXA + BEV is a well-tolerated, effective combination for platinum/taxane-resistant ovarian cancer that extends PFS and likely OS relative to IXA monotherapy. Prior receipt of BEV should not preclude the use of IXA + BEV. TUBB3 is not a predictive biomarker.NCT3093155.
- Published
- 2021
43. Financial toxicity in patients with gynecologic malignancies: a cross sectional study
- Author
-
Masoud Azodi, Peter E. Schwartz, Natalia Pogosian, Mitchell Clark, Gary Altwerger, Joan Tymon-Rosario, Gloria S. Huang, Gulden Menderes, Emily Webster, Alessandro D. Santin, Elena Ratner, Vaagn Andikyan, Alan Balch, and Burak Zeybek
- Subjects
Cross-sectional study ,Genital Neoplasms, Female ,Comprehensive Score for Financial Toxicity ,Financial Stress ,Gynecologic oncology ,Malignancy ,Uterine Cancer ,Cost of Illness ,Uterine cancer ,Cervix Cancer ,Surveys and Questionnaires ,medicine ,Humans ,Rank correlation ,Finance ,Vagina Cancer ,business.industry ,Obstetrics and Gynecology ,General Medicine ,medicine.disease ,Ovarian Cancer ,Clinical trial ,Cross-Sectional Studies ,Oncology ,Vulva Cancer ,Toxicity ,Quality of Life ,Original Article ,Female ,business - Abstract
Objective To evaluate financial toxicity and assess its risk factors among patients with gynecologic cancers. Methods This is a cross sectional study that included 2 survey tools, as well as patient demographics, disease characteristics, and treatment regimen. Financial toxicity is measured by validated Comprehensive Score for Financial Toxicity (COST) tool. Participants were also asked to complete a 55-question-survey on attitudes and perspectives surrounding cost of care. Descriptive statistics was used to report patient demographics. Spearman's rank correlation was calculated to assess the relation between financial toxicity and patient/disease related variables. Graphpad Prism Software Version 8.0 was used for analyses. Results A total of 50 patients with various gynecologic malignancies were enrolled. Median COST score was 20.5 (range, 1–33). Sixty-five percent of the patients reported being in debt due to their cancer care and 4% filed bankruptcy. Correlation analysis showed that COST score was correlated with age (r=−0.3, p=0.028), malignancy type (r=0.3, p=0.039) and income (r=0.3, p=0.047). Ovarian cancer patients had significantly less financial toxicity (median COST score=23) when compared to patients with other gynecologic malignancies (median COST score=17, p=0.043). When scores were dichotomized into low (score ≥22) and high toxicity (score, Synopsis Financial toxicity is a growing problem, however, data is very limited. This is a cross sectional study that included two survey tools, as well as patient demographics, disease characteristics, and treatment regimen. Sixty-five percent of the patients reported being in debt due to their cancer care and 4% filed bankruptcy. Correlation analysis showed that COST score was correlated with age, malignancy type and income.
- Published
- 2021
44. Whole-exome sequencing of cervical carcinomas identifies activating ERBB2 and PIK3CA mutations as targets for combination therapy
- Author
-
Gloria S. Huang, Roberto Angioli, Titus J. Boggon, Weilai Dong, Ludmil B. Alexandrov, Gulden Menderes, Elena Bonazzoli, Francesco Raspagliesi, Richard P. Lifton, Chanhee Han, Aranzazu Manzano, Franco Odicino, Peter E. Schwartz, Antonella Ravaggi, Kaya Bilguvar, Emanuele Perrone, Laura Zanotti, Elena Ratner, Salvatore Lopez, Laura Ardighieri, Joseph Schlessinger, Valentina Pirazzoli, Jungmin Choi, Gary Altwerger, Francesca Pettinella, Siming Zhao, Alessandro D. Santin, Masoud Azodi, Carla Donzelli, Luca Zammataro, Natalia Buza, Paola Todeschini, Burak Zeybek, Dan-Arin Silasi, Eliana Bignotti, Stefania Bellone, Sergio Pecorelli, Pei Hui, Chiara Romani, Serena Wong, Amy L. Stiegler, Emiliano Cocco, Paola Manara, Giovanni Scambia, and Anna Bianchi
- Subjects
copanlisib ,DNA Copy Number Variations ,cervical cancer ,Class I Phosphatidylinositol 3-Kinases ,Receptor, ErbB-2 ,Afatinib ,HER2/neu ,PIK3CA ,neratinib ,Animals ,Cell Line, Tumor ,Combined Modality Therapy ,Female ,Heterografts ,Humans ,Polymorphism, Single Nucleotide ,Uterine Cervical Neoplasms ,Mutation ,Whole Exome Sequencing ,Biology ,Cell Line ,chemistry.chemical_compound ,ErbB-2 ,Exome Sequencing ,medicine ,Polymorphism ,neoplasms ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Exome sequencing ,Copanlisib ,Tumor ,Multidisciplinary ,Single Nucleotide ,Biological Sciences ,medicine.disease ,chemistry ,Neratinib ,biology.protein ,Cancer research ,Adenocarcinoma ,Receptor ,medicine.drug - Abstract
The prognosis of advanced/recurrent cervical cancer patients remains poor. We analyzed 54 fresh-frozen and 15 primary cervical cancer cell lines, along with matched-normal DNA, by whole-exome sequencing (WES), most of which harboring Human-Papillomavirus-type-16/18. We found recurrent somatic missense mutations in 22 genes (including PIK3CA, ERBB2, and GNAS) and a widespread APOBEC cytidine deaminase mutagenesis pattern (TCW motif) in both adenocarcinoma (ACC) and squamous cell carcinomas (SCCs). Somatic copy number variants (CNVs) identified 12 copy number gains and 40 losses, occurring more often than expected by chance, with the most frequent events in pathways similar to those found from analysis of single nucleotide variants (SNVs), including the ERBB2/PI3K/AKT/mTOR, apoptosis, chromatin remodeling, and cell cycle. To validate specific SNVs as targets, we took advantage of primary cervical tumor cell lines and xenografts to preclinically evaluate the activity of pan-HER (afatinib and neratinib) and PIK3CA (copanlisib) inhibitors, alone and in combination, against tumors harboring alterations in the ERBB2/PI3K/AKT/mTOR pathway (71%). Tumors harboring ERBB2 (5.8%) domain mutations were significantly more sensitive to single agents afatinib or neratinib when compared to wild-type tumors in preclinical in vitro and in vivo models (P = 0.001). In contrast, pan-HER and PIK3CA inhibitors demonstrated limited in vitro activity and were only transiently effective in controlling in vivo growth of PIK3CA-mutated cervical cancer xenografts. Importantly, combinations of copanlisib and neratinib were highly synergistic, inducing long-lasting regression of tumors harboring alterations in the ERBB2/PI3K/AKT/mTOR pathway. These findings define the genetic landscape of cervical cancer, suggesting that a large subset of cervical tumors might benefit from existing ERBB2/PIK3CA/AKT/mTOR-targeted drugs.
- Published
- 2019
45. PARP-1 activity (PAR) determines the sensitivity of cervical cancer to olaparib
- Author
-
Luca Zammataro, Garrett M. Frampton, Paola Manara, Elena Bonazzoli, Alessandro D. Santin, Anna Bianchi, Aranzazu Manzano, Dan-Arin Silasi, Masoud Azodi, Stefania Bellone, Gary Altwerger, Salvatore Lopez, Peter E. Schwartz, Chanhee Han, Julia A. Elvin, Gulden Menderes, Gloria S. Huang, Justin Newberg, Burak Zeybek, Emanuele Perrone, Elena Ratner, and Dean Pavlick
- Subjects
Adult ,0301 basic medicine ,Poly ADP ribose polymerase ,Poly (ADP-Ribose) Polymerase-1 ,Uterine Cervical Neoplasms ,Apoptosis ,Cell Growth Processes ,Mice, SCID ,Poly(ADP-ribose) Polymerase Inhibitors ,Piperazines ,Article ,Olaparib ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cytotoxicity ,Dose-Response Relationship, Drug ,business.industry ,Obstetrics and Gynecology ,Middle Aged ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,G2 Phase Cell Cycle Checkpoints ,030104 developmental biology ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,M Phase Cell Cycle Checkpoints ,Phthalazines ,Female ,Ovarian cancer ,business - Abstract
OBJECTIVES. Cervical cancer (CC) remains a major health problem worldwide. Poly (adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitors (PARPi) have emerged as a promising class of chemotherapeutics in ovarian cancer. We explored the preclinical in vitro and in vivo activity of olaparib against multiple primary whole exome sequenced (WES) CC cells lines and xenografts. METHODS. Olaparib cell-cycle, apoptosis, homologous-recombination-deficiency (HRD), PARP trapping and cytotoxicity activity was evaluated against 9 primary CC cell lines in vitro. PARP and PAR expression were analyzed by western blot assays. Finally, olaparib in vivo antitumor activity was tested against CC xenografts. RESULTS. While none of the cell lines demonstrated HRD, three out of 9 (33.3%) primary CC cell lines showed strong PARylation activity and demonstrated high sensitivity to olaparib in vitro treatment (cutoff IC(50) values < 2μM, p=0.0012). Olaparib suppressed CC cell growth through cell cycle arrest in the G2/M phase and caused apoptosis (p
- Published
- 2019
46. Neutrophilia and mortality in women with uterine carcinosarcoma
- Author
-
Brandon M. Roane, Rebecca C. Arend, Charles A. Leath, Gloria S. Huang, Gary L. Goldberg, Anne Van Arsdale, David W. Doo, and Anar Gojayev
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Neutrophils ,Gastroenterology ,Leukocyte Count ,03 medical and health sciences ,0302 clinical medicine ,Carcinosarcoma ,Uterine cancer ,Internal medicine ,medicine ,Humans ,Aged ,Neoplasm Staging ,Proportional Hazards Models ,Retrospective Studies ,business.industry ,Obstetrics and Gynecology ,Retrospective cohort study ,Middle Aged ,Prognosis ,medicine.disease ,Neutrophilia ,Log-rank test ,Clinical trial ,030104 developmental biology ,Oncology ,Quartile ,030220 oncology & carcinogenesis ,Uterine Neoplasms ,Alabama ,Absolute neutrophil count ,Female ,medicine.symptom ,business ,Leukocyte Disorders - Abstract
ObjectiveThe objective of this study was to investigate the relationship between pre-treatment absolute neutrophil count and clinical outcomes in patients with uterine carcinosarcoma.MethodsIn an Institutional Review Board approved, retrospective cohort study of 103 patients with uterine carcinosarcoma, the pre-treatment absolute neutrophil count data were obtained from the medical records, along with clinical, pathologic, treatment, and outcome data. Kaplan–Meier survival estimates were calculated and compared by the log rank test. Univariable and multivariable Cox proportional hazard regression models were used to examine the relationship of pre-treatment absolute neutrophil count with progression-free survival and overall survival.ResultsUterine carcinosarcoma patients in the highest quartile of pre-treatment absolute neutrophil count had significantly reduced progression-free survival (pConclusionsHigh pre-treatment absolute neutrophil count is an independent poor prognostic factor in patients with uterine carcinosarcoma and may be useful as a potential biomarker in clinical trials. The mechanistic relationship of neutrophilia and uterine carcinosarcoma progression merits further investigation.
- Published
- 2019
47. PI3K oncogenic mutations mediate resistance to afatinib in HER2/neu overexpressing gynecological cancers
- Author
-
Elena Bonazzoli, Alessandro D. Santin, Kaitlin Haines, Emiliano Cocco, Paola Manara, Anna Bianchi, Gloria S. Huang, Salvatore Lopez, Aranzazu Manzano, Dan-Arin Silasi, Ghanshyam Yadav, Stefania Bellone, Elena Ratner, Burak Zeybek, Mariana Espinal, Peter E. Schwartz, Masoud Azodi, Gary Altwerger, Emanuele Perrone, Luca Zammataro, Gulden Menderes, Babak Litkouhi, Chanhee Han, and Katherine Dugan
- Subjects
Adult ,0301 basic medicine ,Class I Phosphatidylinositol 3-Kinases ,Genital Neoplasms, Female ,Receptor, ErbB-2 ,Afatinib ,Antineoplastic Agents ,Mice, SCID ,Transfection ,Article ,HER2/neu ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,PIK3R1 ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,neoplasms ,Gene ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Aged ,biology ,business.industry ,Obstetrics and Gynecology ,Middle Aged ,Xenograft Model Antitumor Assays ,Class Ia Phosphatidylinositol 3-Kinase ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Female ,business ,medicine.drug - Abstract
Objective Aberrant expression of HER2/neu and PIK3CA gene products secondary to amplification/mutations are common in high-grade-serous-endometrial (USC) and ovarian-cancers (HGSOC). Because scant information is currently available in the literature on the potential negative effect of PIK3CA mutations on the activity of afatinib, in this study we evaluate for the first time the role of oncogenic PIK3CA mutations as a potential mechanism of resistance to afatinib in HGSOC and USC overexpressing HER2/neu. Methods We used six whole-exome-sequenced primary HGSOC/USC cell-lines and three xenografts overexpressing HER2/neu and harboring mutated or wild-type PIK3CA/PIK3R1 genes to evaluate the role of PI3K-mutations as potential mechanism of resistance to afatinib, an FDA-approved pan-c-erb-inhibitor in clinical trials in USC. Primary-USC harboring wild-type-PIK3CA gene was transfected with plasmids encoding oncogenic PIK3CA-mutations (H1047R/E545K). The effect of afatinib on HER2/PI3K/AKT/mTOR pathway was evaluated by immunoblotting. Results We found PI3K wild-type cell-lines to be significantly more sensitive (lower IC50) than PI3K-mutated cell-lines p = 0.004). In vivo, xenografts of primary cell-line USC-ARK2, transfected with the PIK3CA-H1047R or E545K hotspot-mutations, exhibited significantly more rapid tumor growth when treated with afatinib, compared to mice harboring ARK2-tumors transfected with wild-type-PIK3CA (p = 0.041 and 0.001, respectively). By western-blot, afatinib effectively reduced total and phospho-HER2 proteins in all cell-lines. However, H1047R/E545K-PIK3CA-transfected-ARK2-cells demonstrated a greater compensatory increase in phosphorylated-AKT proteins after afatinib exposure when compared to controls ARK2. Conclusions Oncogenic PI3K mutations may represent a major mechanism of resistance to afatinib. Combinations of c-erb with PIK3CA, AKT or mTOR inhibitors may be necessary to more efficiently block the PIK3CA/AKT/mTOR pathway.
- Published
- 2019
48. Integrated mutational landscape analysis of uterine leiomyosarcomas
- Author
-
Franco Odicino, Samir Zaidi, Nupur Nagarkatti, Gary Altwerger, Laura Zanotti, Alessandro D. Santin, Elena Bonazzoli, Kyungjo Jeong, Gloria S. Huang, Dan-Arin Silasi, Adele Guglielmi, Burak Zeybek, Richard P. Lifton, Luca Zammataro, Dennis Mauricio, Aranzazu Manzano, Gulden Menderes, Barbara Gnutti, Justin Harold, Siming Zhao, Laura Ardighieri, Jungmin Choi, Vaagn Andikyan, Elena Ratner, Chiara Romani, Pei Hui, Xiaotong Yao, Eliana Bignotti, Antonella Ravaggi, Masoud Azodi, Stefania Bellone, Joseph Schlessinger, Joan Tymon-Rosario, Natalia Buza, Ludmil B. Alexandrov, Weilai Dong, Paola Todeschini, Marcin Imielinski, Kaya Bilguvar, Mitchell Clark, Peter E. Schwartz, Charles M. Quick, Sergio Pecorelli, and Aditya Deshpande
- Subjects
Leiomyosarcoma ,Genotype ,education ,mutational landscape ,uterine leiomyosarcomas ,whole-exome sequencing ,whole-genome sequencing ,Animals ,Antineoplastic Agents ,Female ,Humans ,Metabolic Networks and Pathways ,Mice ,Mice, Inbred C57BL ,Molecular Targeted Therapy ,Phthalazines ,Piperazines ,Pyrimidines ,Quinazolines ,Uterine Neoplasms ,Mutation ,Oncogene Fusion ,Inbred C57BL ,Olaparib ,chemistry.chemical_compound ,medicine ,PTEN ,Copy-number variation ,Exome sequencing ,ATRX ,Multidisciplinary ,Chromothripsis ,biology ,Microsatellite instability ,Chromoplexy ,Biological Sciences ,medicine.disease ,chemistry ,Cancer research ,biology.protein - Abstract
Uterine leiomyosarcomas (uLMS) are aggressive tumors arising from the smooth muscle layer of the uterus. We analyzed 83 uLMS sample genetics, including 56 from Yale and 27 from The Cancer Genome Atlas (TCGA). Among them, a total of 55 Yale samples including two patient-derived xenografts (PDXs) and 27 TCGA samples have whole-exome sequencing (WES) data; 10 Yale and 27 TCGA samples have RNA-sequencing (RNA-Seq) data; and 11 Yale and 10 TCGA samples have whole-genome sequencing (WGS) data. We found recurrent somatic mutations in TP53, MED12, and PTEN genes. Top somatic mutated genes included TP53, ATRX, PTEN, and MEN1 genes. Somatic copy number variation (CNV) analysis identified 8 copy-number gains, including 5p15.33 (TERT), 8q24.21 (C-MYC), and 17p11.2 (MYOCD, MAP2K4) amplifications and 29 copy-number losses. Fusions involving tumor suppressors or oncogenes were deetected, with most fusions disrupting RB1, TP53, and ATRX/DAXX, and one fusion (ACTG2-ALK) being potentially targetable. WGS results demonstrated that 76% (16 of 21) of the samples harbored chromoplexy and/or chromothripsis. Clinically actionable mutational signatures of homologous-recombination DNA-repair deficiency (HRD) and microsatellite instability (MSI) were identified in 25% (12 of 48) and 2% (1 of 48) of fresh frozen uLMS, respectively. Finally, we found olaparib (PARPi; P = 0.002), GS-626510 (C-MYC/BETi; P < 0.000001 and P = 0.0005), and copanlisib (PIK3CAi; P = 0.0001) monotherapy to significantly inhibit uLMS-PDXs harboring derangements in C-MYC and PTEN/PIK3CA/AKT genes (LEY11) and/or HRD signatures (LEY16) compared to vehicle-treated mice. These findings define the genetic landscape of uLMS and suggest that a subset of uLMS may benefit from existing PARP-, PIK3CA-, and C-MYC/BET-targeted drugs.
- Published
- 2021
49. Population-Based Screening for Endometrial Cancer: Human vs. Machine Intelligence
- Author
-
Vanessa Yan, Gloria S. Huang, Gregory R. Hart, Jun Deng, Ying Liang, Wazir Muhammad, and Bradley J. Nartowt
- Subjects
Oncology ,medicine.medical_specialty ,statistical biopsy ,Population ,lcsh:QA75.5-76.95 ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Cancer screening ,medicine ,030212 general & internal medicine ,early detection ,education ,Original Research ,education.field_of_study ,business.industry ,Mortality rate ,Incidence (epidemiology) ,Endometrial cancer ,Area under the curve ,medicine.disease ,Random forest ,machine learning ,cancer screening ,030220 oncology & carcinogenesis ,endometrial cancer ,lcsh:Electronic computers. Computer science ,False positive rate ,business - Abstract
Incidence and mortality rates of endometrial cancer are increasing, leading to increased interest in endometrial cancer risk prediction and stratification to help in screening and prevention. Previous risk models have had moderate success with the area under the curve (AUC) ranging from 0.68 to 0.77. Here we demonstrate a population-based machine learning model for endometrial cancer screening that achieves a testing AUC of 0.96.We train seven machine learning algorithms based solely on personal health data, without any genomic, imaging, biomarkers, or invasive procedures. The data come from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO). We further compare our machine learning model with 15 gynecologic oncologists and primary care physicians in the stratification of endometrial cancer risk for 100 women.We find a random forest model that achieves a testing AUC of 0.96 and a neural network model that achieves a testing AUC of 0.91. We test both models in risk stratification against 15 practicing physicians. Our random forest model is 2.5 times better at identifying above-average risk women with a 2-fold reduction in the false positive rate. Our neural network model is 2 times better at identifying above-average risk women with a 3-fold reduction in the false positive rate.Our machine learning models provide a non-invasive and cost-effective way to identify high-risk sub-populations who may benefit from early screening of endometrial cancer, prior to disease onset. Through statistical biopsy of personal health data, we have identified a new and effective approach for early cancer detection and prevention for individual patients.
- Published
- 2020
50. Vulnerability of ARID1A deficient cancer cells to pyrimidine synthesis blockade
- Author
-
Zhigui Li, Pei Hui, Chia Ping H. Yang, Melissa Schwartz, Oloruntoba Ismail Osagie, Shijun Mi, Gloria S. Huang, and Jing Ji
- Subjects
Aspartate carbamoyltransferase ,chemistry.chemical_compound ,chemistry ,DNA damage ,Pyrimidine metabolism ,Cancer cell ,Teriflunomide ,Pyrimidine Synthesis Inhibition ,Cancer research ,medicine ,Cancer ,medicine.disease ,Blockade - Abstract
Here we report the discovery and preclinical validation of a novel precision medicine strategy for ARID1A-mutated cancer. Unbiased proteomics reveals for the first time that ARID1A protein (BAF250a) binds aspartate transcarbamoylase (ATCase), a key regulatory enzyme of the de novo pyrimidine synthesis pathway. Using isogenic paired ARID1A proficient/deficient cancer cell lines, we show that ARID1A protein deficiency (as occurs in ARID1A mutant cancers) leads to metabolic reprogramming and pyrimidine synthesis dependency. Pyrimidine synthesis blockade using the FDA-approved drug teriflunomide (a DHODH inhibitor) suppresses tumor growth and selectively induces DNA damage in ARID1A-deficient tumor models. Combining pyrimidine synthesis inhibition with DNA damage repair blockade, using teriflunomide and AZD6738 (an ATR inhibitor), achieves potent synergy and induces sustained tumor regression in ARID1A-mutant ovarian cancer patient-derived xenografts (PDX). These compelling preclinical data support the evaluation of this novel combination treatment in patients with ARID1A-mutated cancers.SIGNIFICANCEWe identified that ARID1A-deficient cells are selectively vulnerable to pyrimidine synthesis blockade. Preclinical studies demonstrate the in vivo efficacy of a synergistic drug combination that concurrently inhibits the de novo pyrimidine synthesis pathway and DNA damage repair to induce regression in patient-derived xenograft models of ARID1A-mutated cancer.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.