32 results on '"Thanh U. Barbie"'
Search Results
2. Supplementary Data from Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids
- Author
-
David A. Barbie, Cloud Peter Paweletz, David Dornan, Kwok-Kin Wong, Genevieve M. Boland, Roger D. Kamm, Keith T. Flaherty, F. Stephen Hodi, Patrick A. Ott, Jochen H. Lorch, Paul T. Kirschmeier, Gordon J. Freeman, Eliezer M. Van Allen, Levi A. Garraway, Meenhard Herlyn, Juan Miret, Charles H. Yoon, Raphael Bueno, William G. Richards, Joshua Wong, Vivek Sivathanu, Thanh U. Barbie, Benchun Miao, Rohit Thummalapalli, Shunsuke Kitajima, Brian C. Miller, James M. Cleary, Sareh Parangi, Viswanath Gunda, Guilherme Rabinowits, Nicole R. LeBoeuf, Manisha Thakuria, Benjamin Izar, Parin Shah, Mei-Ju Su, Lisa Cameron, Anat O. Stemmer-Rachamimov, Jean-Pierre Eliane, Adriano Piris, Mai P. Hoang, Wei Huang, Glenn J. Hanna, Shuai Li, Hua Zhang, Eric S. Wang, Raven Vlahos, Marc R. Hammond, Tran C. Thai, Israel Cañadas, Michal Barzily-Rokni, Zhiheng Jia, Lance Stapleton, Maria Anguiano, Lauren Keogh, Robert E. Jones, Asaf Rotem, Alicia Smart, Bart Phillips, Prafulla C. Gokhale, Andrew Portell, Joshua A. Kaplan, Chandrasekar Venkataramani, Max Quinn, Ashley Merlino, Jong Wook Kim, Hans Vitzthum, Mark Bittinger, Sangeetha Palakurthi, Zhi Wei, Chaoran Cheng, Tian Tian, Gao Zhang, William Walker, Meng Xiao He, Diana Miao, Hongye Liu, Jiehui Deng, Michaela Bowden, Chensheng W. Zhou, Susanna Stinson, Elena Ivanova, Patrick H. Lizotte, Amir R. Aref, and Russell W. Jenkins
- Abstract
Supplementary Figure Legends and Figures
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figures from Inhibition of KRAS-Driven Tumorigenicity by Interruption of an Autocrine Cytokine Circuit
- Author
-
David A. Barbie, William C. Hahn, Kwok-Kin Wong, Ryan B. Corcoran, Jeffrey A. Engelman, Michael J. Eck, Suzanne Gaudet, Nir Hacohen, Shuji Ogino, William E. Gillanders, Jill P. Mesirov, Lior Rozhansky, Mary T. Labowsky, Whitney Silkworth, Edmond M. Chan, Karolina Maciag, Asher N. Page, Zhi Rong Qian, Jason T. Godfrey, Pablo Tamayo, Jacob B. Reibel, Tran C. Thai, Anna C. Schinzel, Rhine R. Shen, Susan E. Moody, Shenghong Yang, Yu Imamura, Thanh U. Barbie, Travis J. Cohoon, Amir R. Aref, and Zehua Zhu
- Abstract
PDF file 1227K, This file contains Supplemental Figures 1-7, which provide additional supporting information for each of the main figures
- Published
- 2023
- Full Text
- View/download PDF
4. Supp. Movie 1 from Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids
- Author
-
David A. Barbie, Cloud Peter Paweletz, David Dornan, Kwok-Kin Wong, Genevieve M. Boland, Roger D. Kamm, Keith T. Flaherty, F. Stephen Hodi, Patrick A. Ott, Jochen H. Lorch, Paul T. Kirschmeier, Gordon J. Freeman, Eliezer M. Van Allen, Levi A. Garraway, Meenhard Herlyn, Juan Miret, Charles H. Yoon, Raphael Bueno, William G. Richards, Joshua Wong, Vivek Sivathanu, Thanh U. Barbie, Benchun Miao, Rohit Thummalapalli, Shunsuke Kitajima, Brian C. Miller, James M. Cleary, Sareh Parangi, Viswanath Gunda, Guilherme Rabinowits, Nicole R. LeBoeuf, Manisha Thakuria, Benjamin Izar, Parin Shah, Mei-Ju Su, Lisa Cameron, Anat O. Stemmer-Rachamimov, Jean-Pierre Eliane, Adriano Piris, Mai P. Hoang, Wei Huang, Glenn J. Hanna, Shuai Li, Hua Zhang, Eric S. Wang, Raven Vlahos, Marc R. Hammond, Tran C. Thai, Israel Cañadas, Michal Barzily-Rokni, Zhiheng Jia, Lance Stapleton, Maria Anguiano, Lauren Keogh, Robert E. Jones, Asaf Rotem, Alicia Smart, Bart Phillips, Prafulla C. Gokhale, Andrew Portell, Joshua A. Kaplan, Chandrasekar Venkataramani, Max Quinn, Ashley Merlino, Jong Wook Kim, Hans Vitzthum, Mark Bittinger, Sangeetha Palakurthi, Zhi Wei, Chaoran Cheng, Tian Tian, Gao Zhang, William Walker, Meng Xiao He, Diana Miao, Hongye Liu, Jiehui Deng, Michaela Bowden, Chensheng W. Zhou, Susanna Stinson, Elena Ivanova, Patrick H. Lizotte, Amir R. Aref, and Russell W. Jenkins
- Abstract
PDOTS Movie
- Published
- 2023
- Full Text
- View/download PDF
5. Supp. Movie 2 from Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids
- Author
-
David A. Barbie, Cloud Peter Paweletz, David Dornan, Kwok-Kin Wong, Genevieve M. Boland, Roger D. Kamm, Keith T. Flaherty, F. Stephen Hodi, Patrick A. Ott, Jochen H. Lorch, Paul T. Kirschmeier, Gordon J. Freeman, Eliezer M. Van Allen, Levi A. Garraway, Meenhard Herlyn, Juan Miret, Charles H. Yoon, Raphael Bueno, William G. Richards, Joshua Wong, Vivek Sivathanu, Thanh U. Barbie, Benchun Miao, Rohit Thummalapalli, Shunsuke Kitajima, Brian C. Miller, James M. Cleary, Sareh Parangi, Viswanath Gunda, Guilherme Rabinowits, Nicole R. LeBoeuf, Manisha Thakuria, Benjamin Izar, Parin Shah, Mei-Ju Su, Lisa Cameron, Anat O. Stemmer-Rachamimov, Jean-Pierre Eliane, Adriano Piris, Mai P. Hoang, Wei Huang, Glenn J. Hanna, Shuai Li, Hua Zhang, Eric S. Wang, Raven Vlahos, Marc R. Hammond, Tran C. Thai, Israel Cañadas, Michal Barzily-Rokni, Zhiheng Jia, Lance Stapleton, Maria Anguiano, Lauren Keogh, Robert E. Jones, Asaf Rotem, Alicia Smart, Bart Phillips, Prafulla C. Gokhale, Andrew Portell, Joshua A. Kaplan, Chandrasekar Venkataramani, Max Quinn, Ashley Merlino, Jong Wook Kim, Hans Vitzthum, Mark Bittinger, Sangeetha Palakurthi, Zhi Wei, Chaoran Cheng, Tian Tian, Gao Zhang, William Walker, Meng Xiao He, Diana Miao, Hongye Liu, Jiehui Deng, Michaela Bowden, Chensheng W. Zhou, Susanna Stinson, Elena Ivanova, Patrick H. Lizotte, Amir R. Aref, and Russell W. Jenkins
- Abstract
PDOTS Movie 2
- Published
- 2023
- Full Text
- View/download PDF
6. Table S2 from Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids
- Author
-
David A. Barbie, Cloud Peter Paweletz, David Dornan, Kwok-Kin Wong, Genevieve M. Boland, Roger D. Kamm, Keith T. Flaherty, F. Stephen Hodi, Patrick A. Ott, Jochen H. Lorch, Paul T. Kirschmeier, Gordon J. Freeman, Eliezer M. Van Allen, Levi A. Garraway, Meenhard Herlyn, Juan Miret, Charles H. Yoon, Raphael Bueno, William G. Richards, Joshua Wong, Vivek Sivathanu, Thanh U. Barbie, Benchun Miao, Rohit Thummalapalli, Shunsuke Kitajima, Brian C. Miller, James M. Cleary, Sareh Parangi, Viswanath Gunda, Guilherme Rabinowits, Nicole R. LeBoeuf, Manisha Thakuria, Benjamin Izar, Parin Shah, Mei-Ju Su, Lisa Cameron, Anat O. Stemmer-Rachamimov, Jean-Pierre Eliane, Adriano Piris, Mai P. Hoang, Wei Huang, Glenn J. Hanna, Shuai Li, Hua Zhang, Eric S. Wang, Raven Vlahos, Marc R. Hammond, Tran C. Thai, Israel Cañadas, Michal Barzily-Rokni, Zhiheng Jia, Lance Stapleton, Maria Anguiano, Lauren Keogh, Robert E. Jones, Asaf Rotem, Alicia Smart, Bart Phillips, Prafulla C. Gokhale, Andrew Portell, Joshua A. Kaplan, Chandrasekar Venkataramani, Max Quinn, Ashley Merlino, Jong Wook Kim, Hans Vitzthum, Mark Bittinger, Sangeetha Palakurthi, Zhi Wei, Chaoran Cheng, Tian Tian, Gao Zhang, William Walker, Meng Xiao He, Diana Miao, Hongye Liu, Jiehui Deng, Michaela Bowden, Chensheng W. Zhou, Susanna Stinson, Elena Ivanova, Patrick H. Lizotte, Amir R. Aref, and Russell W. Jenkins
- Abstract
In vitro kinase inhibitory activity of Compound 1
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Information from Inhibition of KRAS-Driven Tumorigenicity by Interruption of an Autocrine Cytokine Circuit
- Author
-
David A. Barbie, William C. Hahn, Kwok-Kin Wong, Ryan B. Corcoran, Jeffrey A. Engelman, Michael J. Eck, Suzanne Gaudet, Nir Hacohen, Shuji Ogino, William E. Gillanders, Jill P. Mesirov, Lior Rozhansky, Mary T. Labowsky, Whitney Silkworth, Edmond M. Chan, Karolina Maciag, Asher N. Page, Zhi Rong Qian, Jason T. Godfrey, Pablo Tamayo, Jacob B. Reibel, Tran C. Thai, Anna C. Schinzel, Rhine R. Shen, Susan E. Moody, Shenghong Yang, Yu Imamura, Thanh U. Barbie, Travis J. Cohoon, Amir R. Aref, and Zehua Zhu
- Abstract
PDF file 146K, This file contains the legends to Supplementary Figures 1-7 and Supplementary methods
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table from Inhibition of KRAS-Driven Tumorigenicity by Interruption of an Autocrine Cytokine Circuit
- Author
-
David A. Barbie, William C. Hahn, Kwok-Kin Wong, Ryan B. Corcoran, Jeffrey A. Engelman, Michael J. Eck, Suzanne Gaudet, Nir Hacohen, Shuji Ogino, William E. Gillanders, Jill P. Mesirov, Lior Rozhansky, Mary T. Labowsky, Whitney Silkworth, Edmond M. Chan, Karolina Maciag, Asher N. Page, Zhi Rong Qian, Jason T. Godfrey, Pablo Tamayo, Jacob B. Reibel, Tran C. Thai, Anna C. Schinzel, Rhine R. Shen, Susan E. Moody, Shenghong Yang, Yu Imamura, Thanh U. Barbie, Travis J. Cohoon, Amir R. Aref, and Zehua Zhu
- Abstract
S1 XLSX file 63K, This Supplementary Table contains the results of ORF screen to identify genes that can rescue the suppression of TBK1 in a KRAS-dependent cell line
- Published
- 2023
- Full Text
- View/download PDF
9. Supp. Movie 3 from Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids
- Author
-
David A. Barbie, Cloud Peter Paweletz, David Dornan, Kwok-Kin Wong, Genevieve M. Boland, Roger D. Kamm, Keith T. Flaherty, F. Stephen Hodi, Patrick A. Ott, Jochen H. Lorch, Paul T. Kirschmeier, Gordon J. Freeman, Eliezer M. Van Allen, Levi A. Garraway, Meenhard Herlyn, Juan Miret, Charles H. Yoon, Raphael Bueno, William G. Richards, Joshua Wong, Vivek Sivathanu, Thanh U. Barbie, Benchun Miao, Rohit Thummalapalli, Shunsuke Kitajima, Brian C. Miller, James M. Cleary, Sareh Parangi, Viswanath Gunda, Guilherme Rabinowits, Nicole R. LeBoeuf, Manisha Thakuria, Benjamin Izar, Parin Shah, Mei-Ju Su, Lisa Cameron, Anat O. Stemmer-Rachamimov, Jean-Pierre Eliane, Adriano Piris, Mai P. Hoang, Wei Huang, Glenn J. Hanna, Shuai Li, Hua Zhang, Eric S. Wang, Raven Vlahos, Marc R. Hammond, Tran C. Thai, Israel Cañadas, Michal Barzily-Rokni, Zhiheng Jia, Lance Stapleton, Maria Anguiano, Lauren Keogh, Robert E. Jones, Asaf Rotem, Alicia Smart, Bart Phillips, Prafulla C. Gokhale, Andrew Portell, Joshua A. Kaplan, Chandrasekar Venkataramani, Max Quinn, Ashley Merlino, Jong Wook Kim, Hans Vitzthum, Mark Bittinger, Sangeetha Palakurthi, Zhi Wei, Chaoran Cheng, Tian Tian, Gao Zhang, William Walker, Meng Xiao He, Diana Miao, Hongye Liu, Jiehui Deng, Michaela Bowden, Chensheng W. Zhou, Susanna Stinson, Elena Ivanova, Patrick H. Lizotte, Amir R. Aref, and Russell W. Jenkins
- Abstract
Device loading, media addition, media extraction
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S1 from Use of Ex Vivo Patient-Derived Tumor Organotypic Spheroids to Identify Combination Therapies for HER2 Mutant Non–Small Cell Lung Cancer
- Author
-
Pasi A. Jänne, Cloud P. Paweletz, Magda Bahcall, David A. Barbie, Amir A. Aref, Suzanne E. Dahlberg, Kwok-Kin Wong, Jacob J. Haworth, Geoffrey R. Oxnard, Thanh U. Barbie, Ting Chen, Shengwu Liu, Shuai Li, Emily S. Chambers, Jihyun Choi, Alshad S. Lalani, Irmina Diala, Luke Taus, Andrew J. Portell, Man Xu, Mari Kuraguchi, and Elena Ivanova
- Abstract
Supplemental Figure 1. Characterization of DFCI 359 and DFCI 315.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Phosphorylation of RAB7 by TBK1/IKKϵ Regulates Innate Immune Signaling in Triple-Negative Breast Cancer
- Author
-
Thanh U. Barbie, David A. Barbie, Steven A. Carr, Jacob D. Jaffe, Saemi Han, Brandon P. Piel, Erik H. Knelson, Philipp Mertins, Navin R. Mahadevan, Tran C. Thai, Zehua Zhu, and Jessica L. Ritter
- Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease enriched for mutations in PTEN and dysregulation of innate immune signaling. Here, we demonstrate that Rab7, a recently identified substrate of PTEN phosphatase activity, is also a substrate of the innate immune signaling kinases TANK-binding kinase 1 (TBK1)/IκB kinase ϵ (IKKϵ) on the same serine-72 (S72) site. An unbiased search for novel TBK1/IKKϵ substrates using stable isotope labeling with amino acids in cell culture phosphoproteomic analysis identified Rab7-S72 as a top hit. PTEN-null TNBC cells expressing a phosphomimetic version of Rab7-S72 exhibited diffuse cytosolic Rab7 localization and enhanced innate immune signaling, in contrast to a kinase-resistant version, which localized to active puncta that promote lysosomal-mediated stimulator of interferon genes (STING) degradation. Thus, convergence of PTEN loss and TBK1/IKKϵ activation on Rab7-S72 phosphorylation limited STING turnover and increased downstream production of IRF3 targets including CXCL10, CCL5, and IFNβ. Consistent with this data, PTEN-null TNBC tumors expressed higher levels of STING, and PTEN-null TNBC cell lines were hyperresponsive to STING agonists. Together, these findings begin to uncover how innate immune signaling is dysregulated downstream of TBK1/IKKϵ in a subset of TNBCs and reveals previously unrecognized cross-talk with STING recycling that may have implications for STING agonism in the clinic.Significance:These findings identify Rab7 as a substrate for TBK1 for regulation of innate immune signaling, thereby providing important insight for strategies aimed at manipulating the immune response to enhance therapeutic efficacy in TNBC.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Data from Phosphorylation of RAB7 by TBK1/IKKϵ Regulates Innate Immune Signaling in Triple-Negative Breast Cancer
- Author
-
Thanh U. Barbie, David A. Barbie, Steven A. Carr, Jacob D. Jaffe, Saemi Han, Brandon P. Piel, Erik H. Knelson, Philipp Mertins, Navin R. Mahadevan, Tran C. Thai, Zehua Zhu, and Jessica L. Ritter
- Abstract
SF1: Unaltered immunoblots for Fig 4E. SF2: Unaltered immunoblots for Fig 4F. SF3: Unaltered immunoblots for Fig 4G. SF4: Additional phosphopeptide analysis for cell based and in vitro SILAC screen. SF5: Lower magnification (40X) of basal expression of V5 and Rab7 in mutants, to correspond with Fig 4A. SF6: EGFR and STING localization following TBK1/IKKE inhibition. SF7: Schematic for Rab7S72 role in regulating STING/TBK1/IKKE signaling in TNBCs. STableS3: Primer sequence for qRT-PCR. STableS4: List of antibodies. STableS5: PTEN/STING Staining of a TNBC TMA.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table S1 from Use of Ex Vivo Patient-Derived Tumor Organotypic Spheroids to Identify Combination Therapies for HER2 Mutant Non–Small Cell Lung Cancer
- Author
-
Pasi A. Jänne, Cloud P. Paweletz, Magda Bahcall, David A. Barbie, Amir A. Aref, Suzanne E. Dahlberg, Kwok-Kin Wong, Jacob J. Haworth, Geoffrey R. Oxnard, Thanh U. Barbie, Ting Chen, Shengwu Liu, Shuai Li, Emily S. Chambers, Jihyun Choi, Alshad S. Lalani, Irmina Diala, Luke Taus, Andrew J. Portell, Man Xu, Mari Kuraguchi, and Elena Ivanova
- Abstract
Supplementary Table S1
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table 1 from Phosphorylation of RAB7 by TBK1/IKKϵ Regulates Innate Immune Signaling in Triple-Negative Breast Cancer
- Author
-
Thanh U. Barbie, David A. Barbie, Steven A. Carr, Jacob D. Jaffe, Saemi Han, Brandon P. Piel, Erik H. Knelson, Philipp Mertins, Navin R. Mahadevan, Tran C. Thai, Zehua Zhu, and Jessica L. Ritter
- Abstract
Phosphopeptides from cell based screen
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Use of Ex Vivo Patient-Derived Tumor Organotypic Spheroids to Identify Combination Therapies for HER2 Mutant Non–Small Cell Lung Cancer
- Author
-
Pasi A. Jänne, Cloud P. Paweletz, Magda Bahcall, David A. Barbie, Amir A. Aref, Suzanne E. Dahlberg, Kwok-Kin Wong, Jacob J. Haworth, Geoffrey R. Oxnard, Thanh U. Barbie, Ting Chen, Shengwu Liu, Shuai Li, Emily S. Chambers, Jihyun Choi, Alshad S. Lalani, Irmina Diala, Luke Taus, Andrew J. Portell, Man Xu, Mari Kuraguchi, and Elena Ivanova
- Abstract
Purpose:Evaluating drug responses using primary patient-derived cells ex vivo represents a potentially rapid and efficient approach to screening for new treatment approaches. Here, we sought to identify neratinib combinations in HER2 mutant non–small cell lung cancer (NSCLC) patient xenograft-derived organotypic spheroids (XDOTS) using a short-term ex vivo system.Experimental Design:We generated two HER2-mutant NSCLC PDX models [DFCI359 (HER2 exon19 755_757LREdelinsRP) and DFCI315 (HER2 exon20 V777_G778insGSP)] and used the PDX tumors to generate XDOTS. Tumor spheroids were grown in a microfluidic device and treated ex vivo with neratinib-based drug combinations. Live/dead quantification was performed by dual-labeling deconvolution fluorescence microscopy. The most efficacious ex vivo combination was subsequently validated in vivo using the DFCI359 and DFCI315 PDXs and a HER2YVMA genetically engineered mouse model.Results:Both neratinib and afatinib, but not gefitinib, induced cell death in DFCI359 XDOTS. The combinations of neratinib/trastuzumab and neratinib/temsirolimus enhanced the therapeutic benefit of neratinib alone in DFCI315 and DFCI359. The combination of neratinib and trastuzumab in vivo was more effective compared with single-agent neratinib or trastuzumab and was associated with more robust inhibition of HER2 and downstream signaling.Conclusions:The XDOTS platform can be used to evaluate therapies and therapeutic combinations ex vivo using PDX tumors. This approach may accelerate the identification and clinical development of therapies for targets with no or few existing models and/or therapies.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplemental Legend from Use of Ex Vivo Patient-Derived Tumor Organotypic Spheroids to Identify Combination Therapies for HER2 Mutant Non–Small Cell Lung Cancer
- Author
-
Pasi A. Jänne, Cloud P. Paweletz, Magda Bahcall, David A. Barbie, Amir A. Aref, Suzanne E. Dahlberg, Kwok-Kin Wong, Jacob J. Haworth, Geoffrey R. Oxnard, Thanh U. Barbie, Ting Chen, Shengwu Liu, Shuai Li, Emily S. Chambers, Jihyun Choi, Alshad S. Lalani, Irmina Diala, Luke Taus, Andrew J. Portell, Man Xu, Mari Kuraguchi, and Elena Ivanova
- Abstract
Supplemental Legend
- Published
- 2023
- Full Text
- View/download PDF
17. Omission of Surgical Axillary Lymph Node Staging in Patients with Tubular Breast Cancer
- Author
-
Mehra Golshan, Stephanie M. Wong, Thanh U. Barbie, Enver Özkurt, Esther Rhei, and Jane E. Brock
- Subjects
medicine.medical_specialty ,business.industry ,Urology ,Cancer ,Ductal carcinoma ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Breast cancer ,Oncology ,Surgical oncology ,030220 oncology & carcinogenesis ,Cohort ,Propensity score matching ,medicine ,030211 gastroenterology & hepatology ,Surgery ,business ,Lymph node ,Survival analysis - Abstract
With more effective screening and treatment strategies, there is debate over whether surgical axillary staging should be deescalated for patients with small favorable breast cancers, such as tubular carcinoma (TC). We identified patients with TC [defined as > 90% tubular tubules (angulated, not multilayered)] and known surgical axillary staging from our institutional database (2000–2018). Using the National Cancer Database (NCDB) (2004–2015), we identified patients with TC, ductal carcinoma in situ (DCIS), and pT1 estrogen receptor (ER)-positive invasive ductal carcinoma (IDC). We determined the rates of lymph node (LN) metastases, and the 5- and 10-year overall survival (OS) for patients with LN-negative versus LN-positive disease using the Kaplan–Meier method and propensity match analysis. In our institutional cohort, we identified 112 patients with T1 TC; only one (0.9%) patient had nodal involvement. In the NCDB cohort, we identified 6938 patients with T1 TC; 323 (4.7%) patients had axillary LN disease. The rate of axillary LN involvement for TC was comparable to that identified for patients with DCIS (4.2%), and much lower than that found for patients with grade I–III, T1, ER-positive IDC (20.5%), and patients with grade I, T1, ER-positive IDC (14.4%). There was no difference in 5-year (94.6% versus 95.4%, p = 0.67) and 10-year (83.9% versus 85.2%, p = 0.98) OS between TC patients with or without LN involvement. Kaplan–Meier survival curves even after propensity score matching suggest that tubular histology is independently associated with improved survival. T1 TC is an excellent starting point for deescalation of surgical axillary staging.
- Published
- 2020
- Full Text
- View/download PDF
18. Use of Ex Vivo Patient-Derived Tumor Organotypic Spheroids to Identify Combination Therapies for HER2 Mutant Non–Small Cell Lung Cancer
- Author
-
Ting Chen, Alshad S. Lalani, Amir Aref, Luke J. Taus, Emily S. Chambers, Man Xu, Cloud P. Paweletz, Geoffrey R. Oxnard, Irmina Diala, Jacob J. Haworth, Thanh U. Barbie, David A. Barbie, Pasi A. Jänne, Mari Kuraguchi, Jihyun Choi, Shengwu Liu, Elena Ivanova, Kwok-Kin Wong, Suzanne E. Dahlberg, Andrew Portell, Magda Bahcall, and Shuai Li
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Afatinib ,medicine.disease ,Temsirolimus ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Gefitinib ,Oncology ,Trastuzumab ,In vivo ,030220 oncology & carcinogenesis ,Neratinib ,medicine ,Cancer research ,skin and connective tissue diseases ,Lung cancer ,business ,neoplasms ,Ex vivo ,medicine.drug - Abstract
Purpose: Evaluating drug responses using primary patient-derived cells ex vivo represents a potentially rapid and efficient approach to screening for new treatment approaches. Here, we sought to identify neratinib combinations in HER2 mutant non–small cell lung cancer (NSCLC) patient xenograft-derived organotypic spheroids (XDOTS) using a short-term ex vivo system. Experimental Design: We generated two HER2-mutant NSCLC PDX models [DFCI359 (HER2 exon19 755_757LREdelinsRP) and DFCI315 (HER2 exon20 V777_G778insGSP)] and used the PDX tumors to generate XDOTS. Tumor spheroids were grown in a microfluidic device and treated ex vivo with neratinib-based drug combinations. Live/dead quantification was performed by dual-labeling deconvolution fluorescence microscopy. The most efficacious ex vivo combination was subsequently validated in vivo using the DFCI359 and DFCI315 PDXs and a HER2YVMA genetically engineered mouse model. Results: Both neratinib and afatinib, but not gefitinib, induced cell death in DFCI359 XDOTS. The combinations of neratinib/trastuzumab and neratinib/temsirolimus enhanced the therapeutic benefit of neratinib alone in DFCI315 and DFCI359. The combination of neratinib and trastuzumab in vivo was more effective compared with single-agent neratinib or trastuzumab and was associated with more robust inhibition of HER2 and downstream signaling. Conclusions: The XDOTS platform can be used to evaluate therapies and therapeutic combinations ex vivo using PDX tumors. This approach may accelerate the identification and clinical development of therapies for targets with no or few existing models and/or therapies.
- Published
- 2020
- Full Text
- View/download PDF
19. MPS1 inhibition primes immunogenicity of KRAS-LKB1 mutant lung cancer
- Author
-
Shunsuke Kitajima, Tetsuo Tani, Benjamin F. Springer, Marco Campisi, Tatsuya Osaki, Koji Haratani, Minyue Chen, Erik H. Knelson, Navin R. Mahadevan, Jessica Ritter, Ryohei Yoshida, Jens Köhler, Atsuko Ogino, Ryu-Suke Nozawa, Shriram K. Sundararaman, Tran C. Thai, Mizuki Homme, Brandon Piel, Sophie Kivlehan, Bonje N. Obua, Connor Purcell, Mamiko Yajima, Thanh U. Barbie, Patrick H. Lizotte, Pasi A. Jänne, Cloud P. Paweletz, Prafulla C. Gokhale, and David A. Barbie
- Subjects
Proto-Oncogene Proteins p21(ras) ,Cancer Research ,Genes, ras ,Lung Neoplasms ,Oncology ,Humans ,Decitabine ,Ligands - Abstract
KRAS-LKB1 (KL) mutant lung cancers silence STING owing to intrinsic mitochondrial dysfunction, resulting in T cell exclusion and resistance to programmed cell death (ligand) 1 (PD-[L]1) blockade. Here we discover that KL cells also minimize intracellular accumulation of 2'3'-cyclic GMP-AMP (2'3'-cGAMP) to further avoid downstream STING and STAT1 activation. An unbiased screen to co-opt this vulnerability reveals that transient MPS1 inhibition (MPS1i) potently re-engages this pathway in KL cells via micronuclei generation. This effect is markedly amplified by epigenetic de-repression of STING and only requires pulse MPS1i treatment, creating a therapeutic window compared with non-dividing cells. A single course of decitabine treatment followed by pulse MPS1i therapy restores T cell infiltration in vivo, enhances anti-PD-1 efficacy, and results in a durable response without evidence of significant toxicity.
- Published
- 2021
20. Phosphorylation of RAB7 by TBK1/IKKϵ Regulates Innate Immune Signaling in Triple-Negative Breast Cancer
- Author
-
Tran C. Thai, Saemi Han, Philipp Mertins, Erik H. Knelson, Steven A. Carr, David A. Barbie, Navin R. Mahadevan, Zehua Zhu, Jessica Leigh Ritter, Jacob D. Jaffe, Thanh U. Barbie, and Brandon Piel
- Subjects
0301 basic medicine ,Cancer Research ,Triple Negative Breast Neoplasms ,IκB kinase ,Protein Serine-Threonine Kinases ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,TANK-binding kinase 1 ,Cell Line, Tumor ,Serine ,Humans ,PTEN ,Breast ,Phosphorylation ,Innate immune system ,biology ,PTEN Phosphohydrolase ,Membrane Proteins ,rab7 GTP-Binding Proteins ,Immunity, Innate ,I-kappa B Kinase ,Sting ,HEK293 Cells ,030104 developmental biology ,Oncology ,rab GTP-Binding Proteins ,030220 oncology & carcinogenesis ,Stimulator of interferon genes ,Mutation ,Proteolysis ,Mutagenesis, Site-Directed ,biology.protein ,Cancer research ,Female ,IRF3 ,Signal Transduction - Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease enriched for mutations in PTEN and dysregulation of innate immune signaling. Here, we demonstrate that Rab7, a recently identified substrate of PTEN phosphatase activity, is also a substrate of the innate immune signaling kinases TANK-binding kinase 1 (TBK1)/IκB kinase ϵ (IKKϵ) on the same serine-72 (S72) site. An unbiased search for novel TBK1/IKKϵ substrates using stable isotope labeling with amino acids in cell culture phosphoproteomic analysis identified Rab7-S72 as a top hit. PTEN-null TNBC cells expressing a phosphomimetic version of Rab7-S72 exhibited diffuse cytosolic Rab7 localization and enhanced innate immune signaling, in contrast to a kinase-resistant version, which localized to active puncta that promote lysosomal-mediated stimulator of interferon genes (STING) degradation. Thus, convergence of PTEN loss and TBK1/IKKϵ activation on Rab7-S72 phosphorylation limited STING turnover and increased downstream production of IRF3 targets including CXCL10, CCL5, and IFNβ. Consistent with this data, PTEN-null TNBC tumors expressed higher levels of STING, and PTEN-null TNBC cell lines were hyperresponsive to STING agonists. Together, these findings begin to uncover how innate immune signaling is dysregulated downstream of TBK1/IKKϵ in a subset of TNBCs and reveals previously unrecognized cross-talk with STING recycling that may have implications for STING agonism in the clinic. Significance: These findings identify Rab7 as a substrate for TBK1 for regulation of innate immune signaling, thereby providing important insight for strategies aimed at manipulating the immune response to enhance therapeutic efficacy in TNBC.
- Published
- 2020
- Full Text
- View/download PDF
21. Omission of Surgical Axillary Lymph Node Staging in Patients with Tubular Breast Cancer
- Author
-
Enver, Özkurt, Stephanie, Wong, Esther, Rhei, Mehra, Golshan, Jane, Brock, and Thanh U, Barbie
- Subjects
Carcinoma, Intraductal, Noninfiltrating ,Axilla ,Carcinoma, Ductal, Breast ,Humans ,Breast Neoplasms ,Lymph Nodes ,Neoplasm Staging ,Retrospective Studies - Abstract
With more effective screening and treatment strategies, there is debate over whether surgical axillary staging should be deescalated for patients with small favorable breast cancers, such as tubular carcinoma (TC).We identified patients with TC [defined as 90% tubular tubules (angulated, not multilayered)] and known surgical axillary staging from our institutional database (2000-2018). Using the National Cancer Database (NCDB) (2004-2015), we identified patients with TC, ductal carcinoma in situ (DCIS), and pT1 estrogen receptor (ER)-positive invasive ductal carcinoma (IDC). We determined the rates of lymph node (LN) metastases, and the 5- and 10-year overall survival (OS) for patients with LN-negative versus LN-positive disease using the Kaplan-Meier method and propensity match analysis.In our institutional cohort, we identified 112 patients with T1 TC; only one (0.9%) patient had nodal involvement. In the NCDB cohort, we identified 6938 patients with T1 TC; 323 (4.7%) patients had axillary LN disease. The rate of axillary LN involvement for TC was comparable to that identified for patients with DCIS (4.2%), and much lower than that found for patients with grade I-III, T1, ER-positive IDC (20.5%), and patients with grade I, T1, ER-positive IDC (14.4%). There was no difference in 5-year (94.6% versus 95.4%, p = 0.67) and 10-year (83.9% versus 85.2%, p = 0.98) OS between TC patients with or without LN involvement. Kaplan-Meier survival curves even after propensity score matching suggest that tubular histology is independently associated with improved survival.T1 TC is an excellent starting point for deescalation of surgical axillary staging.
- Published
- 2020
22. Multidisciplinary Management of the Axilla in Patients with cT1-T2 N0 Breast Cancer Undergoing Primary Mastectomy: Results from a Prospective Single-Institution Series
- Author
-
Anvy Nguyen, Esther Rhei, Katharine Carter, Suniti Nimbkar, Jennifer K. Plichta, Margaret M. Duggan, Mehra Golshan, Jiani Hu, Rinaa S. Punglia, Samantha Grossmith, Julia Wong, Katherina Zabicki Calvillo, Linda Cutone, Faina Nakhlis, Jennifer R. Bellon, Tari A. King, Thanh U. Barbie, Laura S. Dominici, and William T. Barry
- Subjects
Adult ,medicine.medical_specialty ,medicine.medical_treatment ,Breast Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,medicine ,Humans ,Neoplasm Invasiveness ,Prospective Studies ,030212 general & internal medicine ,Prospective cohort study ,Mastectomy ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Univariate analysis ,Radiotherapy ,Sentinel Lymph Node Biopsy ,business.industry ,Carcinoma, Ductal, Breast ,Axillary Lymph Node Dissection ,Disease Management ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Radiation therapy ,Carcinoma, Lobular ,Axilla ,Lymphedema ,medicine.anatomical_structure ,Oncology ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Female ,Surgery ,Radiology ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies - Abstract
The after mapping of the axilla: radiotherapy or surgery (AMAROS) trial concluded that for patients with cT1-2 N0 breast cancer and one or two positive sentinel lymph nodes (SLNs), axillary radiotherapy (AxRT) provides equivalent locoregional control and a lower incidence of lymphedema compared with axillary lymph node dissection (ALND). The study prospectively assessed how often ALND could be replaced by AxRT in a consecutive cohort of patients undergoing mastectomy for cT1-2 N0 breast cancer. In November 2015, our multidisciplinary group agreed to omit routine intraoperative SLN evaluation for cT1-2 N0 patients undergoing upfront mastectomy and potentially eligible for postmastectomy radiation therapy (PMRT), including those 60 years of age or younger and those older than 60 years with high-risk features. Patients with one or two positive SLNs on final pathology were reviewed to determine whether PMRT including the full axilla was an appropriate alternative to ALND. From November 2015 to December 2016, 154 patients met the study criteria, and 114 (74%) formed the final study cohort. Intraoperative SLN evaluation was omitted for 76 patients (67%). Of these patients, 20 (26%) had one or two positive SLNs, and 14 of these patients received PMRT + AxRT as an alternative to ALND. Three patients returned for ALND, and three patients were observed. On univariate analysis, tumor size, LVI, number of positive lymph nodes, and receipt of chemotherapy were associated with receipt of PMRT. For the majority of patients with one or two positive SLNs, ALND was avoided in favor of PMRT + AxRT. With appropriate multidisciplinary strategies, intraoperative evaluation of the SLN and immediate ALND can be avoided for patients meeting the AMAROS criteria and eligible for PMRT.
- Published
- 2018
- Full Text
- View/download PDF
23. Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids
- Author
-
Mark A. Bittinger, Parin Shah, William F. Walker, Meng Xiao He, Nicole R. LeBoeuf, Chaoran Cheng, Rohit Thummalapalli, Chandrasekar Venkataramani, Glenn J. Hanna, Joshua A. Kaplan, Eliezer M. Van Allen, Jochen H. Lorch, Susanna Stinson, Raphael Bueno, Max M. Quinn, Michaela Bowden, Genevieve M. Boland, Cloud P. Paweletz, Bart Phillips, Gao Zhang, Jong Wook Kim, Adriano Piris, Patrick H. Lizotte, Raven Vlahos, Patrick A. Ott, Lance Stapleton, Levi A. Garraway, Hua Zhang, Chensheng W. Zhou, Jean Pierre Eliane, Eric S. Wang, Alicia Smart, Andrew Portell, Zhi Wei, Hongye Liu, Jiehui Deng, Meenhard Herlyn, Shuai Li, Diana Miao, Israel Cañadas, F. Stephen Hodi, Hans Vitzthum, Marc R. Hammond, Russell W. Jenkins, Prafulla C. Gokhale, Thanh U. Barbie, Michal Barzily-Rokni, David A. Barbie, Keith T. Flaherty, Tran C. Thai, Shunsuke Kitajima, Gordon J. Freeman, Roger D. Kamm, Juan J. Miret, Lauren Keogh, Elena Ivanova, Charles H. Yoon, Lisa A. Cameron, Viswanath Gunda, Ashley A. Merlino, David Dornan, Anat Stemmer-Rachamimov, Joshua A. Wong, Kwok-Kin Wong, William G. Richards, Sangeetha Palakurthi, Maria Anguiano, Wei Huang, Sareh Parangi, James M. Cleary, Benjamin Izar, Amir Reza Aref, Brian C. Miller, Vivek Sivathanu, Zhiheng Jia, Asaf Rotem, Mai P. Hoang, Paul Kirschmeier, Benchun Miao, Mei-Ju Su, Manisha Thakuria, Tian Tian, Robert E. Jones, and Guilherme Rabinowits
- Subjects
0301 basic medicine ,Tumor microenvironment ,medicine.diagnostic_test ,Cell ,Biology ,Immune checkpoint ,Blockade ,Flow cytometry ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,In vivo ,Immunology ,medicine ,Cancer research ,Ex vivo - Abstract
Ex vivo systems that incorporate features of the tumor microenvironment and model the dynamic response to immune checkpoint blockade (ICB) may facilitate efforts in precision immuno-oncology and the development of effective combination therapies. Here, we demonstrate the ability to interrogate ex vivo response to ICB using murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). MDOTS/PDOTS isolated from mouse and human tumors retain autologous lymphoid and myeloid cell populations and respond to ICB in short-term three-dimensional microfluidic culture. Response and resistance to ICB was recapitulated using MDOTS derived from established immunocompetent mouse tumor models. MDOTS profiling demonstrated that TBK1/IKKϵ inhibition enhanced response to PD-1 blockade, which effectively predicted tumor response in vivo. Systematic profiling of secreted cytokines in PDOTS captured key features associated with response and resistance to PD-1 blockade. Thus, MDOTS/PDOTS profiling represents a novel platform to evaluate ICB using established murine models as well as clinically relevant patient specimens. Significance: Resistance to PD-1 blockade remains a challenge for many patients, and biomarkers to guide treatment are lacking. Here, we demonstrate feasibility of ex vivo profiling of PD-1 blockade to interrogate the tumor immune microenvironment, develop therapeutic combinations, and facilitate precision immuno-oncology efforts. Cancer Discov; 8(2); 196–215. ©2017 AACR. See related commentary by Balko and Sosman, p. 143. See related article by Deng et al., p. 216. This article is highlighted in the In This Issue feature, p. 127
- Published
- 2018
- Full Text
- View/download PDF
24. De Novo Stage 4 Metastatic Breast Cancer: A Surgical Disease?
- Author
-
Mehra Golshan and Thanh U. Barbie
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,MEDLINE ,Breast Neoplasms ,Disease ,medicine.disease ,Metastatic breast cancer ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Surgical oncology ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Humans ,030211 gastroenterology & hepatology ,Surgery ,Neoplasm Metastasis ,Stage (cooking) ,business - Published
- 2018
- Full Text
- View/download PDF
25. Use of
- Author
-
Elena, Ivanova, Mari, Kuraguchi, Man, Xu, Andrew J, Portell, Luke, Taus, Irmina, Diala, Alshad S, Lalani, Jihyun, Choi, Emily S, Chambers, Shuai, Li, Shengwu, Liu, Ting, Chen, Thanh U, Barbie, Geoffrey R, Oxnard, Jacob J, Haworth, Kwok-Kin, Wong, Suzanne E, Dahlberg, Amir A, Aref, David A, Barbie, Magda, Bahcall, Cloud P, Paweletz, and Pasi A, Jänne
- Subjects
Lung Neoplasms ,Receptor, ErbB-2 ,Apoptosis ,Mice, SCID ,Trastuzumab ,Xenograft Model Antitumor Assays ,Article ,Mice ,Mice, Inbred NOD ,Carcinoma, Non-Small-Cell Lung ,Spheroids, Cellular ,Antineoplastic Combined Chemotherapy Protocols ,Mutation ,Quinolines ,Tumor Cells, Cultured ,Animals ,Humans ,Female ,skin and connective tissue diseases ,neoplasms ,Cell Proliferation - Abstract
PURPOSE: Evaluating drug responses using primary patient derived cells ex vivo represents a potentially rapid and efficient approach to screening for new treatment approaches. Here, we sought to identify neratinib combinations in HER2 mutant non-small cell lung cancer (NSCLC) patient xenograft derived organotypic spheroids (XDOTS) using a short term ex vivo system. METHODS: We generated two HER2 mutant NSCLC PDX models (DFCI359 (HER2 exon19 755_757LREdelinsRP) and DFCI315 (HER2 exon20 V777_G778insGSP)) and used the PDX tumors to generate XDOTS. Tumor spheroids were grown in a microfluidic device and treated ex vivo with neratinib based drug combinations. Live/dead quantification was performed by dual labeling deconvolution fluorescence microscopy. The most efficacious ex vivo combination was subsequently validated in vivo using the DFCI359 and DFCI315 PDXs and a HER2(YVMA) genetically engineered mouse model (GEMM). RESULTS: Both neratinib and afatinib, but not gefitinib, induced cell death in DFCI359 XDOTS. The combinations of neratinib/trastuzumab, and neratinib/temsirolimus enhanced the therapeutic benefit of neratinib alone in DFCI315 and DFCI359. The combination of neratinib and trastuzumab in vivo was more effective compared to single agent neratinib or trastuzumab and was associated with more robust inhibition of HER2 and downstream signaling. CONCLUSIONS: The XDOTS platform can be used to evaluate therapies and therapeutic combinations ex vivo using PDX tumors. This approach may accelerate the identification and clinical development of therapies for targets with no or few existing models and/or therapies.
- Published
- 2019
26. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses
- Author
-
Tran C. Thai, Masayuki Watanabe, Hideo Baba, Camilla L. Christensen, Hideki Terai, Russell W. Jenkins, Shunsuke Kitajima, Gao Zhang, Tian Tian, Pablo Tamayo, Jong Wook Kim, Jacob Sands, Yanxi Zhang, Brandon Piel, Ewa Sicinska, Cloud P. Paweletz, Timothy Hagan, Thanh U. Barbie, Marco Campisi, Rohit Thummalapalli, Hideo Watanabe, Yanan Kuang, Israel Cañadas, Amir Reza Aref, Evisa Gjini, Anika E. Adeni, Lynnette Marie Sholl, Diana Miao, Christine A. Lydon, Yu Imamura, David A. Barbie, Meenhard Herlyn, Debattama R. Sen, Ravindra Uppaluri, Kwok-Kin Wong, Shohei Koyama, Scott J. Rodig, and Zhi Wei
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Nude ,Endogenous retrovirus ,Medical and Health Sciences ,Mice ,Cancer immunotherapy ,Interferon ,Neoplasms ,2.1 Biological and endogenous factors ,Innate ,Aetiology ,Cancer ,Regulation of gene expression ,Tumor ,General Medicine ,Long terminal repeat ,3. Good health ,Cell biology ,Gene Expression Regulation, Neoplastic ,medicine.drug ,Immunology ,Animals ,Cell Line, Tumor ,Endogenous Retroviruses ,Humans ,Immunity, Innate ,Interferons ,Mice, Nude ,RNA, Antisense ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Vaccine Related ,03 medical and health sciences ,Immune system ,Genetics ,medicine ,Antisense ,Neoplastic ,Innate immune system ,Inflammatory and immune system ,Immunity ,030104 developmental biology ,Gene Expression Regulation ,RNA ,Immunization ,IRF3 - Abstract
Mesenchymal tumor subpopulations secrete pro-tumorigenic cytokines and promote treatment resistance1-4. This phenomenon has been implicated in chemorefractory small cell lung cancer and resistance to targeted therapies5-8, but remains incompletely defined. Here, we identify a subclass of endogenous retroviruses (ERVs) that engages innate immune signaling in these cells. Stimulated 3 prime antisense retroviral coding sequences (SPARCS) are oriented inversely in 3' untranslated regions of specific genes enriched for regulation by STAT1 and EZH2. Derepression of these loci results in double-stranded RNA generation following IFN-γ exposure due to bi-directional transcription from the STAT1-activated gene promoter and the 5' long terminal repeat of the antisense ERV. Engagement of MAVS and STING activates downstream TBK1, IRF3, and STAT1 signaling, sustaining a positive feedback loop. SPARCS induction in human tumors is tightly associated with major histocompatibility complex class 1 expression, mesenchymal markers, and downregulation of chromatin modifying enzymes, including EZH2. Analysis of cell lines with high inducible SPARCS expression reveals strong association with an AXL/MET-positive mesenchymal cell state. While SPARCS-high tumors are immune infiltrated, they also exhibit multiple features of an immune-suppressed microenviroment. Together, these data unveil a subclass of ERVs whose derepression triggers pathologic innate immune signaling in cancer, with important implications for cancer immunotherapy.
- Published
- 2018
27. Management of Premenopausal Women with Neoadjuvant Endocrine Therapy: A Single-Institution Experience
- Author
-
Cynthia X. Ma, Julie A. Margenthaler, and Thanh U. Barbie
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,Antineoplastic Agents, Hormonal ,medicine.medical_treatment ,Breast surgery ,Population ,Anastrozole ,Breast Neoplasms ,Gonadotropin-Releasing Hormone ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Nitriles ,Humans ,Medicine ,Endocrine system ,education ,Response Evaluation Criteria in Solid Tumors ,Neoadjuvant therapy ,education.field_of_study ,business.industry ,Letrozole ,Carcinoma, Ductal, Breast ,Middle Aged ,Triazoles ,medicine.disease ,Neoadjuvant Therapy ,Carcinoma, Lobular ,Tamoxifen ,Premenopause ,Receptors, Estrogen ,Chemotherapy, Adjuvant ,Female ,Surgery ,business ,medicine.drug - Abstract
In postmenopausal women with hormone receptor (HR)-positive breast cancer, neoadjuvant endocrine therapy (ET) provides effective downstaging of tumor for improved surgical outcome and offers an important advantage of assessing tumor endocrine responsiveness for individualized therapy in the adjuvant setting. Although approximately 60 % of breast cancers in premenopausal women are HR positive, the role of neoadjuvant ET in this population is not well defined.We identified 162 patients with stage I-III estrogen receptor-positive breast cancer treated with neoadjuvant ET between 2003 and 2012. Of this group, 17 patients were premenopausal. Data included patient/tumor characteristics, surgical, systemic, and radiation treatment received, and outcomes. Descriptive statistics were used for data summary.The cohort included 17 patients with a mean age of 46.2 years (range 39-53 years). Patients were treated with a combination of gonadotrophic-releasing hormone agonist with either an aromatase inhibitor (n = 14) or tamoxifen (n = 3) for 4-6 months. Among the premenopausal patients, six underwent breast-conserving therapy, with 3 of 6 (50.0 %) having positive margins. Adjuvant chemotherapy was recommended for 13 (76.5 %), and adjuvant radiotherapy was recommended for 13 (76.5 %). Of the 17 premenopausal women, 11 had a clinical response based on response evaluation criteria in solid tumors (RECIST) of a decrease in tumor size of 30 % (64.7 %); this is similar to that of postmenopausal women, where 85 of 145 (58.6 %) patients showed a clinical response.As with all neoadjuvant systemic interventions, we identified those with disease that did and did not respond to ET, emphasizing the heterogeneity of HR-positive breast cancers. The response rate of premenopausal women to neoadjuvant ET is similar to that of postmenopausal women.
- Published
- 2015
- Full Text
- View/download PDF
28. Targeting an IKBKE cytokine network impairs triple-negative breast cancer growth
- Author
-
Shunqiang Li, Yu Imamura, Thanh U. Barbie, Kwok-Kin Wong, Gabriela Alexe, Jill P. Mesirov, Matthew J. Ellis, Travis J. Cohoon, William E. Gillanders, Xiuli Zhang, Zehua Zhu, Pablo Tamayo, Ying Huang, Amir Reza Aref, William C. Hahn, Shuji Ogino, John M. Herndon, Timothy P. Fleming, Michaela Bowden, David A. Barbie, and Tran C. Thai
- Subjects
Angiogenesis ,medicine.medical_treatment ,Breast Neoplasms ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,TANK-binding kinase 1 ,medicine ,Animals ,Humans ,IKBKE ,Chemokine CCL5 ,Triple-negative breast cancer ,Janus Kinases ,030304 developmental biology ,0303 health sciences ,Interleukin-6 ,MEK inhibitor ,NF-kappa B ,JAK-STAT signaling pathway ,General Medicine ,Xenograft Model Antitumor Assays ,I-kappa B Kinase ,Neoplasm Proteins ,3. Good health ,Pyrimidines ,Cytokine ,030220 oncology & carcinogenesis ,Benzamides ,Cancer research ,Female ,Janus kinase ,Signal Transduction ,Research Article - Abstract
Triple-negative breast cancers (TNBCs) are a heterogeneous set of cancers that are defined by the absence of hormone receptor expression and HER2 amplification. Here, we found that inducible IκB kinase–related (IKK-related) kinase IKBKE expression and JAK/STAT pathway activation compose a cytokine signaling network in the immune-activated subset of TNBC. We found that treatment of cultured IKBKE-driven breast cancer cells with CYT387, a potent inhibitor of TBK1/IKBKE and JAK signaling, impairs proliferation, while inhibition of JAK alone does not. CYT387 treatment inhibited activation of both NF-κB and STAT and disrupted expression of the protumorigenic cytokines CCL5 and IL-6 in these IKBKE-driven breast cancer cells. Moreover, in 3D culture models, the addition of CCL5 and IL-6 to the media not only promoted tumor spheroid dispersal but also stimulated proliferation and migration of endothelial cells. Interruption of cytokine signaling by CYT387 in vivo impaired the growth of an IKBKE-driven TNBC cell line and patient-derived xenografts (PDXs). A combination of CYT387 therapy with a MEK inhibitor was particularly effective, abrogating tumor growth and angiogenesis in an aggressive PDX model of TNBC. Together, these findings reveal that IKBKE-associated cytokine signaling promotes tumorigenicity of immune-driven TNBC and identify a potential therapeutic strategy using clinically available compounds.
- Published
- 2014
- Full Text
- View/download PDF
29. EpCAM expression varies significantly and is differentially associated with prognosis in the luminal B HER2+, basal-like, and HER2 intrinsic subtypes of breast cancer
- Author
-
Thanh U. Barbie, Simone Muenst, Savas D. Soysal, Carsten T. Viehl, Daniel Oertli, Feng Gao, Ellen C. Obermann, Timothy P. Fleming, Gilbert Spizzo, and William E. Gillanders
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,Breast Neoplasms ,Cell Growth Processes ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Basal (phylogenetics) ,0302 clinical medicine ,Breast cancer ,breast cancer ,Antigens, Neoplasm ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Viability assay ,intrinsic subtypes ,skin and connective tissue diseases ,prognostic factor ,Molecular Diagnostics ,030304 developmental biology ,0303 health sciences ,Tissue microarray ,Cell adhesion molecule ,Epithelial cell adhesion molecule ,Middle Aged ,medicine.disease ,Epithelial Cell Adhesion Molecule ,Prognosis ,Immunohistochemistry ,3. Good health ,chemistry ,SKBR3 ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,EpCAM ,Female ,Cell Adhesion Molecules - Abstract
Background: Epithelial cell adhesion molecule (EpCAM) is frequently expressed in breast cancer, and its expression has been associated with poor prognosis. Breast cancer can be subdivided into intrinsic subtypes, differing in prognosis and response to therapy. Methods: To investigate the association between EpCAM expression and prognosis in the intrinsic subtypes of breast cancer, we performed immunohistochemical studies on a tissue microarray encompassing a total of 1365 breast cancers with detailed clinicopathological annotation and outcomes data. Results: We observed EpCAM expression in 660 out of 1365 (48%) cases. EpCAM expression varied significantly in the different intrinsic subtypes. In univariate analyses of all cases, EpCAM expression was associated with a significantly worse overall survival. In the intrinsic subtypes, EpCAM expression was associated with an unfavourable prognosis in the basal-like and luminal B HER2+ subtypes but associated with a favourable prognosis in the HER2 subtype. Consistently, specific ablation of EpCAM resulted in increased cell viability in the breast cancer cell line SKBR3 (ER−, PR−, and HER2+) but decreased viability in the breast cancer cell line MDA-MB-231 (ER−, PR−, and HER2− ). Conclusion: The differential association of EpCAM expression with prognosis in intrinsic subtypes has important implications for the development of EpCAM-targeted therapies in breast cancer.
- Published
- 2013
30. Impact of residual nodal disease burden on sentinel node mapping and accuracy of intraoperative frozen section in node positive (cN1) breast cancer patients treated with neoadjuvant chemotherapy (NAC)
- Author
-
Tari A. King, Faina Nakhlis, Jiani Hu, Anna Weiss, Laura S. Dominici, Melissa E. Hughes, Katherina Zabicki Calvillo, Alison Laws, Katharine Carter, William T. Barry, Esther Rhei, Suniti Nimbkar, Stuart J. Schnitt, Margaret M. Duggan, and Thanh U. Barbie
- Subjects
Cancer Research ,medicine.medical_specialty ,Frozen section procedure ,Chemotherapy ,medicine.diagnostic_test ,business.industry ,Node (networking) ,medicine.medical_treatment ,Sentinel node ,medicine.disease ,Nodal disease ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Oncology ,030220 oncology & carcinogenesis ,Biopsy ,Medicine ,030211 gastroenterology & hepatology ,Radiology ,business - Abstract
584Background: Recent trials have demonstrated the feasibility of SLN biopsy in cN1 patients who become cN0 after NAC. We sought to evaluate success of SLN mapping and accuracy of intraop frozen se...
- Published
- 2018
- Full Text
- View/download PDF
31. Mullerian Inhibiting Substance inhibits cervical cancer cell growth via a pathway involving p130 and p107
- Author
-
Patricia K. Donahoe, Shyamala Maheswaran, Thanh U. Barbie, David A. Barbie, and David T. MacLaughlin
- Subjects
Anti-Mullerian Hormone ,medicine.medical_specialty ,endocrine system diseases ,Mullerian Ducts ,medicine.medical_treatment ,Blotting, Western ,Uterine Cervical Neoplasms ,Cell Cycle Proteins ,Retinoblastoma-Like Protein p107 ,Cervix Uteri ,Biology ,Transfection ,Retinoblastoma Protein ,Cell Line ,Targeted therapy ,Internal medicine ,Chlorocebus aethiops ,Tumor Cells, Cultured ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Cell Line, Transformed ,Glycoproteins ,Cervical cancer ,Multidisciplinary ,Retinoblastoma-Like Protein p130 ,Retinoblastoma protein ,Nuclear Proteins ,Proteins ,Anti-Müllerian hormone ,Biological Sciences ,Phosphoproteins ,medicine.disease ,Cyclin-Dependent Kinases ,Rats ,Adaptor Proteins, Vesicular Transport ,Testicular Hormones ,Endocrinology ,Cell culture ,COS Cells ,biology.protein ,Cancer research ,Papilloma ,Female ,Signal transduction ,Cell Division ,Signal Transduction - Abstract
In addition to causing regression of the Mullerian duct in the male embryo, Mullerian Inhibiting Substance (MIS) inhibits the growth of epithelial ovarian cancer cells, which are known to be of Mullerian origin. Because the uterine cervix is derived from the same Mullerian duct precursor as the epithelium of the ovary, we tested the hypothesis that cervical cancer cells might also respond to MIS. A number of cervical cancer cell lines express the MIS type II receptor, and MIS inhibits the growth of both human papilloma virus-transformed and non-human papilloma virus-transformed cervical cell lines, with a more dramatic effect seen in the latter. As in the ovarian cancer cell line OVCAR8, suppression of growth of the C33A cervical cancer cell line by MIS is associated with induction of the p16 tumor suppressor protein. However, in contrast to OVCAR8 cells, induction of p130 and p107 appears to play an important role in the inhibition of growth of C33A cells by MIS. Finally, normal cervical tissue expresses the MIS type II receptor in vivo , supporting the idea that MIS could be a targeted therapy for cervical cancer.
- Published
- 2003
- Full Text
- View/download PDF
32. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit
- Author
-
William C. Hahn, Shenghong Yang, Susan Moody, Nir Hacohen, David A. Barbie, Ryan B. Corcoran, Mary T. Labowsky, Whitney Silkworth, Amir Reza Aref, Jason T. Godfrey, Karolina Maciag, Tran C. Thai, Lior Rozhansky, Yu Imamura, Zehua Zhu, Pablo Tamayo, Rhine R. Shen, Thanh U. Barbie, Asher N. Page, Travis J. Cohoon, Kwok-Kin Wong, Jacob B. Reibel, Jeffrey A. Engelman, Anna C. Schinzel, Zhi Rong Qian, Shuji Ogino, Jill P. Mesirov, William E. Gillanders, Suzanne Gaudet, Michael J. Eck, and Edmond M. Chan
- Subjects
MAPK/ERK pathway ,medicine.medical_treatment ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Article ,Mice ,TANK-binding kinase 1 ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Autocrine signalling ,neoplasms ,Chemokine CCL5 ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Kinase ,Interleukin-6 ,MEK inhibitor ,Neoplasms, Experimental ,digestive system diseases ,respiratory tract diseases ,Autocrine Communication ,Cytokine ,Pyrimidines ,Oncology ,Benzamides ,Cancer research ,ras Proteins ,I-kappa B Proteins ,KRAS ,Signal Transduction - Abstract
Although the roles of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling in KRAS-driven tumorigenesis are well established, KRAS activates additional pathways required for tumor maintenance, the inhibition of which are likely to be necessary for effective KRAS-directed therapy. Here, we show that the IκB kinase (IKK)–related kinases Tank-binding kinase-1 (TBK1) and IKKϵ promote KRAS-driven tumorigenesis by regulating autocrine CCL5 and interleukin (IL)-6 and identify CYT387 as a potent JAK/TBK1/IKKϵ inhibitor. CYT387 treatment ablates RAS-associated cytokine signaling and impairs Kras-driven murine lung cancer growth. Combined CYT387 treatment and MAPK pathway inhibition induces regression of aggressive murine lung adenocarcinomas driven by Kras mutation and p53 loss. These observations reveal that TBK1/IKKϵ promote tumor survival by activating CCL5 and IL-6 and identify concurrent inhibition of TBK1/IKKϵ, Janus-activated kinase (JAK), and MEK signaling as an effective approach to inhibit the actions of oncogenic KRAS. Significance: In addition to activating MAPK and PI3K, oncogenic KRAS engages cytokine signaling to promote tumorigenesis. CYT387, originally described as a selective JAK inhibitor, is also a potent TBK/IKKϵ inhibitor that uniquely disrupts a cytokine circuit involving CCL5, IL-6, and STAT3. The efficacy of CYT387-based treatment in murine Kras-driven lung cancer models uncovers a novel therapeutic approach for these refractory tumors with immediate translational implications. Cancer Discov; 4(4); 452–65. ©2014 AACR. This article is highlighted in the In This Issue feature, p. 377
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.