48 results on '"Samanthi A. Perera"'
Search Results
2. Supplementary Data from STimulator of INterferon Genes Agonism Accelerates Antitumor Activity in Poorly Immunogenic Tumors
- Author
-
George H. Addona, David Jonathan Bennett, Brian J. Long, Sheila Ranganath, Anuradha Khilnani, Archie Tse, B. Wesley Trotter, Jared Cumming, Saso Cemerski, Ian Knemeyer, Vincenzo Pucci, Hyun Chong Woo, Sharad K. Sharma, Shuxia Zhao, Bo-Sheng Pan, Jennifer Piesvaux, Heidi M. Ferguson, Ellen C. Minnihan, Sarah Javaid, Manjiri Sathe, Jeremy Presland, Long Cui, Yiping Chen, Kalyan Chakravarthy, Jason Laskey, Eric S. Muise, Yanhong Ma, Johnny E. Kopinja, and Samanthi A. Perera
- Abstract
Supplementary Methods and Figures
- Published
- 2023
- Full Text
- View/download PDF
3. Table S1 from STimulator of INterferon Genes Agonism Accelerates Antitumor Activity in Poorly Immunogenic Tumors
- Author
-
George H. Addona, David Jonathan Bennett, Brian J. Long, Sheila Ranganath, Anuradha Khilnani, Archie Tse, B. Wesley Trotter, Jared Cumming, Saso Cemerski, Ian Knemeyer, Vincenzo Pucci, Hyun Chong Woo, Sharad K. Sharma, Shuxia Zhao, Bo-Sheng Pan, Jennifer Piesvaux, Heidi M. Ferguson, Ellen C. Minnihan, Sarah Javaid, Manjiri Sathe, Jeremy Presland, Long Cui, Yiping Chen, Kalyan Chakravarthy, Jason Laskey, Eric S. Muise, Yanhong Ma, Johnny E. Kopinja, and Samanthi A. Perera
- Abstract
Supplemental Table 1: Log2Ratio and p values for all 310 genes shown in Figure 6
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table 1 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 560K, Comparison of ganetespib and 17-AAG efficacy in NSCLC cell lines.
- Published
- 2023
- Full Text
- View/download PDF
5. Data from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
Purpose: We describe the anticancer activity of ganetespib, a novel non-geldanamycin heat shock protein 90 (HSP90) inhibitor, in non-small cell lung cancer (NSCLC) models.Experimental Design: The activity of ganetespib was compared with that of the geldanamycin 17-AAG in biochemical assays, cell lines, and xenografts, and evaluated in an ERBB2 YVMA-driven mouse lung adenocarcinoma model.Results: Ganetespib blocked the ability of HSP90 to bind to biotinylated geldanamycin and disrupted the association of HSP90 with its cochaperone, p23, more potently than 17-AAG. In genomically defined NSCLC cell lines, ganetespib caused depletion of receptor tyrosine kinases, extinguishing of downstream signaling, inhibition of proliferation and induction of apoptosis with IC50 values ranging 2 to 30 nmol/L, substantially lower than those required for 17-AAG (20–3,500 nmol/L). Ganetespib was also approximately 20-fold more potent in isogenic Ba/F3 pro-B cells rendered IL-3 independent by expression of EGFR and ERBB2 mutants. In mice bearing NCI-H1975 (EGFR L858R/T790M) xenografts, ganetespib was rapidly eliminated from plasma and normal tissues but was maintained in tumor with t1/2 58.3 hours, supporting once-weekly dosing experiments, in which ganetespib produced greater tumor growth inhibition than 17-AAG. However, after a single dose, reexpression of mutant EGFR occurred by 72 hours, correlating with reversal of antiproliferative and proapoptotic effects. Consecutive day dosing resulted in xenograft regressions, accompanied by more sustained pharmacodynamic effects. Ganetespib also showed activity against mouse lung adenocarcinomas driven by oncogenic ERBB2 YVMA.Conclusions: Ganetespib has greater potency than 17-AAG and potential efficacy against several NSCLC subsets, including those harboring EGFR or ERBB2 mutation. Clin Cancer Res; 18(18); 4973–85. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 1349K, Depletion of receptor tyrosine kinases and suppression of downstream signaling in HCC827 cells in response to 17-AAG or ganetespib-mediated HSP90 inhibition.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 3 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 365K, Activity of ganetespib in HCC827 xenografts.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 6 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 776K, Effects of 17-AAG and ganetespib on cell viability and mutant ERBB2 expression in ERBB2-dependent Ba/F3 cells.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table 2 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 136K, Median IC50's for NSCLC cell lines (with ganetespib and 17-AAG) with different genotypic subtypes.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table 3 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 361K, Comparison of EGFR TKI and HSP90 inhibitor efficacy in Ba/F3 cells ectopically expressing different EGFR mutations.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 2 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 1170K, Depletion of oncogenic drivers in NSCLC cell lines.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 4 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 719K, Comparison of once-weekly and consecutive day dosing schedules among individual mice bearing NCI-H1975 xenografts.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Methods from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 49K, Supplementary Immunohistochemical and Imaging Methods.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table 4 from Ganetespib (STA-9090), a Nongeldanamycin HSP90 Inhibitor, Has Potent Antitumor Activity in In Vitro and In Vivo Models of Non–Small Cell Lung Cancer
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, James Barsoum, Weiwen Ying, Matthew Meyerson, John-Paul Jimenez, Christa L. Borgman, Christopher D. Carey, Papiya Sinha, Yu-Chen Li, Julian Carretero, Danan Li, Liang Chen, Takayo Inoue, Scott J. Rodig, Jim Sang, Kevin P. Foley, Samanthi A. Perera, and Takeshi Shimamura
- Abstract
PDF file, 294K, Ganetespib Biodistribution and Pharmacokinetic Parameters.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 2 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Author
-
Kwok-Kin Wong, Lynda Chin, Bruce E. Johnson, Pasi A. Jänne, Levi A. Garraway, Roman K. Thomas, Roderick T. Bronson, Robert Padera, Lucian R. Chirieac, Ruqayyah Al-Hashem, Yanping Sun, Mitchell Albert, Liang Chen, Kate McNamara, Sara Zaghlul, Mei-Chih Liang, Danan Li, Samanthi A. Perera, Zhenxiong Wang, and Hongbin Ji
- Abstract
Supplementary Figure 2 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Methods, Legends for Figures 1-8 and Tables 1-3 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Matthew Meyerson, Daniel G. Tenen, Masaya Takahashi, Shigeto Kubo, Robert F. Padera, Lucian R. Chirieac, Susumu Kobayashi, Heidi Greulich, Roman K. Thomas, Sara Zaghlul, Samanthi A. Perera, Kate McNamara, Yuko Minami, April M. Lowell, Christa L. Borgman, Elizabeth Liniker, Henry J. Haringsma, Hongbin Ji, Danan Li, and Takeshi Shimamura
- Abstract
Supplementary Methods, Legends for Figures 1-8 and Tables 1-3 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Materials and Figure Legends 1-4 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Author
-
Kwok-Kin Wong, Lynda Chin, Bruce E. Johnson, Pasi A. Jänne, Levi A. Garraway, Roman K. Thomas, Roderick T. Bronson, Robert Padera, Lucian R. Chirieac, Ruqayyah Al-Hashem, Yanping Sun, Mitchell Albert, Liang Chen, Kate McNamara, Sara Zaghlul, Mei-Chih Liang, Danan Li, Samanthi A. Perera, Zhenxiong Wang, and Hongbin Ji
- Abstract
Supplementary Materials and Figure Legends 1-4 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 7 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Matthew Meyerson, Daniel G. Tenen, Masaya Takahashi, Shigeto Kubo, Robert F. Padera, Lucian R. Chirieac, Susumu Kobayashi, Heidi Greulich, Roman K. Thomas, Sara Zaghlul, Samanthi A. Perera, Kate McNamara, Yuko Minami, April M. Lowell, Christa L. Borgman, Elizabeth Liniker, Henry J. Haringsma, Hongbin Ji, Danan Li, and Takeshi Shimamura
- Abstract
Supplementary Figure 7 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 3 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Matthew Meyerson, Daniel G. Tenen, Masaya Takahashi, Shigeto Kubo, Robert F. Padera, Lucian R. Chirieac, Susumu Kobayashi, Heidi Greulich, Roman K. Thomas, Sara Zaghlul, Samanthi A. Perera, Kate McNamara, Yuko Minami, April M. Lowell, Christa L. Borgman, Elizabeth Liniker, Henry J. Haringsma, Hongbin Ji, Danan Li, and Takeshi Shimamura
- Abstract
Supplementary Figure 3 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 8 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Matthew Meyerson, Daniel G. Tenen, Masaya Takahashi, Shigeto Kubo, Robert F. Padera, Lucian R. Chirieac, Susumu Kobayashi, Heidi Greulich, Roman K. Thomas, Sara Zaghlul, Samanthi A. Perera, Kate McNamara, Yuko Minami, April M. Lowell, Christa L. Borgman, Elizabeth Liniker, Henry J. Haringsma, Hongbin Ji, Danan Li, and Takeshi Shimamura
- Abstract
Supplementary Figure 8 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Figure 5 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Matthew Meyerson, Daniel G. Tenen, Masaya Takahashi, Shigeto Kubo, Robert F. Padera, Lucian R. Chirieac, Susumu Kobayashi, Heidi Greulich, Roman K. Thomas, Sara Zaghlul, Samanthi A. Perera, Kate McNamara, Yuko Minami, April M. Lowell, Christa L. Borgman, Elizabeth Liniker, Henry J. Haringsma, Hongbin Ji, Danan Li, and Takeshi Shimamura
- Abstract
Supplementary Figure 5 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 1 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Matthew Meyerson, Daniel G. Tenen, Masaya Takahashi, Shigeto Kubo, Robert F. Padera, Lucian R. Chirieac, Susumu Kobayashi, Heidi Greulich, Roman K. Thomas, Sara Zaghlul, Samanthi A. Perera, Kate McNamara, Yuko Minami, April M. Lowell, Christa L. Borgman, Elizabeth Liniker, Henry J. Haringsma, Hongbin Ji, Danan Li, and Takeshi Shimamura
- Abstract
Supplementary Figure 1 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure 3 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Author
-
Kwok-Kin Wong, Lynda Chin, Bruce E. Johnson, Pasi A. Jänne, Levi A. Garraway, Roman K. Thomas, Roderick T. Bronson, Robert Padera, Lucian R. Chirieac, Ruqayyah Al-Hashem, Yanping Sun, Mitchell Albert, Liang Chen, Kate McNamara, Sara Zaghlul, Mei-Chih Liang, Danan Li, Samanthi A. Perera, Zhenxiong Wang, and Hongbin Ji
- Abstract
Supplementary Figure 3 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Published
- 2023
- Full Text
- View/download PDF
24. Data from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Author
-
Kwok-Kin Wong, Lynda Chin, Bruce E. Johnson, Pasi A. Jänne, Levi A. Garraway, Roman K. Thomas, Roderick T. Bronson, Robert Padera, Lucian R. Chirieac, Ruqayyah Al-Hashem, Yanping Sun, Mitchell Albert, Liang Chen, Kate McNamara, Sara Zaghlul, Mei-Chih Liang, Danan Li, Samanthi A. Perera, Zhenxiong Wang, and Hongbin Ji
- Abstract
Mutations in the BRAF and KRAS genes occur in ∼1% to 2% and 20% to 30% of non–small-cell lung cancer patients, respectively, suggesting that the mitogen-activated protein kinase (MAPK) pathway is preferentially activated in lung cancers. Here, we show that lung-specific expression of the BRAF V600E mutant induces the activation of extracellular signal–regulated kinase (ERK)-1/2 (MAPK) pathway and the development of lung adenocarcinoma with bronchioloalveolar carcinoma features in vivo. Deinduction of transgene expression led to dramatic tumor regression, paralleled by dramatic dephosphorylation of ERK1/2, implying a dependency of BRAF-mutant lung tumors on the MAPK pathway. Accordingly, in vivo pharmacologic inhibition of MAPK/ERK kinase (MEK; MAPKK) using a specific MEK inhibitor, CI-1040, induced tumor regression associated with inhibition of cell proliferation and induction of apoptosis in these de novo lung tumors. CI-1040 treatment also led to dramatic tumor shrinkage in murine lung tumors driven by a mutant KRas allele. Thus, somatic mutations in different signaling intermediates of the same pathway induce exquisite dependency on a shared downstream effector. These results unveil a potential common vulnerability of BRAF and KRas mutant lung tumors that potentially affects rational deployment of MEK targeted therapies to non–small-cell lung cancer patients. [Cancer Res 2007;67(10):4933–9]
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure 4 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Matthew Meyerson, Daniel G. Tenen, Masaya Takahashi, Shigeto Kubo, Robert F. Padera, Lucian R. Chirieac, Susumu Kobayashi, Heidi Greulich, Roman K. Thomas, Sara Zaghlul, Samanthi A. Perera, Kate McNamara, Yuko Minami, April M. Lowell, Christa L. Borgman, Elizabeth Liniker, Henry J. Haringsma, Hongbin Ji, Danan Li, and Takeshi Shimamura
- Abstract
Supplementary Figure 4 from Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figure 1 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Author
-
Kwok-Kin Wong, Lynda Chin, Bruce E. Johnson, Pasi A. Jänne, Levi A. Garraway, Roman K. Thomas, Roderick T. Bronson, Robert Padera, Lucian R. Chirieac, Ruqayyah Al-Hashem, Yanping Sun, Mitchell Albert, Liang Chen, Kate McNamara, Sara Zaghlul, Mei-Chih Liang, Danan Li, Samanthi A. Perera, Zhenxiong Wang, and Hongbin Ji
- Abstract
Supplementary Figure 1 from Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models
- Published
- 2023
- Full Text
- View/download PDF
27. Discovery of MK-1454: A Potent Cyclic Dinucleotide Stimulator of Interferon Genes Agonist for the Treatment of Cancer
- Author
-
Wonsuk Chang, Michael D. Altman, Charles A. Lesburg, Samanthi A. Perera, Jennifer A. Piesvaux, Gottfried K. Schroeder, Daniel F. Wyss, Saso Cemerski, Yiping Chen, Edward DiNunzio, Andrew M. Haidle, Thu Ho, Ilona Kariv, Ian Knemeyer, Johnny E. Kopinja, Brian M. Lacey, Jason Laskey, Jongwon Lim, Brian J. Long, Yanhong Ma, Matthew L. Maddess, Bo-Sheng Pan, Jeremy P. Presland, Edward Spooner, Dietrich Steinhuebel, Quang Truong, Zhibo Zhang, Jianmin Fu, George H. Addona, Alan B. Northrup, Emma Parmee, James R. Tata, David Jonathan Bennett, Jared N. Cumming, Tony Siu, and B. Wesley Trotter
- Subjects
Mice ,Neoplasms ,Drug Discovery ,Molecular Medicine ,Animals ,Cytokines ,Humans ,Membrane Proteins ,Immunotherapy ,Interferons - Abstract
Stereochemically and structurally complex cyclic dinucleotide-based stimulator of interferon genes (STING) agonists were designed and synthesized to access a previously unexplored chemical space. The assessment of biochemical affinity and cellular potency, along with computational, structural, and biophysical characterization, was applied to influence the design and optimization of novel STING agonists, resulting in the discovery of MK-1454 as a molecule with appropriate properties for clinical development. When administered intratumorally to immune-competent mice-bearing syngeneic tumors, MK-1454 exhibited robust tumor cytokine upregulation and effective antitumor activity. Tumor shrinkage in mouse models that are intrinsically resistant to single-agent therapy was further enhanced when treating the animals with MK-1454 in combination with a fully murinized antimouse PD-1 antibody, mDX400. These data support the development of STING agonists in combination with pembrolizumab (humanized anti-PD-1 antibody) for patients with tumors that are partially responsive or nonresponsive to single-agent anti-PD-1 therapy.
- Published
- 2022
28. STimulator of INterferon Genes Agonism Accelerates Antitumor Activity in Poorly Immunogenic Tumors
- Author
-
Saso Cemerski, Jennifer Piesvaux, Jason Laskey, Brian Long, B. Wesley Trotter, Jared N. Cumming, Shuxia Zhao, Eric S. Muise, Yanhong Ma, Ellen C. Minnihan, Samanthi A. Perera, Sheila Ranganath, Sarah Javaid, Bo-Sheng Pan, Sharad K. Sharma, Yiping Chen, Jeremy Presland, Long Cui, Manjiri Sathe, Archie Tse, Kalyan Chakravarthy, Vincenzo Pucci, Ian Knemeyer, Hyun Chong Woo, David Jonathan Bennett, Anuradha D. Khilnani, George H. Addona, Heidi Ferguson, and Johnny E. Kopinja
- Subjects
Agonist ,Cancer Research ,Innate immune system ,medicine.drug_class ,Chemistry ,medicine.medical_treatment ,Type I interferon production ,Immunity, Innate ,Sting ,Mice ,Cytokine ,Immune system ,Oncology ,Downregulation and upregulation ,Stimulator of interferon genes ,Cell Line, Tumor ,Neoplasms ,medicine ,Cancer research ,Animals ,Humans ,Female ,Immunotherapy ,Interferons - Abstract
The innate immune agonist STING (STimulator of INterferon Genes) binds its natural ligand 2′3′-cGAMP (cyclic guanosine-adenosine monophosphate) and initiates type I IFN production. This promotes systemic antigen-specific CD8+ T-cell priming that eventually provides potent antitumor activity. To exploit this mechanism, we synthesized a novel STING agonist, MSA-1, that activates both mouse and human STING with higher in vitro potency than cGAMP. Following intratumoral administration of MSA-1 to a panel of syngeneic mouse tumors on immune-competent mice, cytokine upregulation and its exposure were detected in plasma, other tissues, injected tumors, and noninjected tumors. This was accompanied by effective antitumor activity. Mechanistic studies in immune-deficient mice suggested that antitumor activity of intratumorally dosed STING agonists is in part due to necrosis and/or innate immune responses such as TNF-α activity, but development of a robust adaptive antitumor immunity is necessary for complete tumor elimination. Combination with PD-1 blockade in anti–PD-1–resistant murine models showed that MSA-1 may synergize with checkpoint inhibitors but can also provide superior tumor control as a single agent. We show for the first time that potent cyclic dinucleotides can promote a rapid and stronger induction of the same genes eventually regulated by PD-1 blockade. This may have contributed to the relatively early tumor control observed with MSA-1. Taken together, these data strongly support the development of STING agonists as therapy for patients with aggressive tumors that are partially responsive or nonresponsive to single-agent anti–PD-1 treatment by enhancing the anti–PD-1 immune profile.
- Published
- 2021
29. Corrigendum: Effective Anti-tumor Response by TIGIT Blockade Associated With FcγR Engagement and Myeloid Cell Activation
- Author
-
Thomas W. Rosahl, Mingmei Cai, Jin-Hwan Han, Wolfgang Seghezzi, Jeffery Grein, Elaine M. Pinheiro, M. Bigler, Hongmei Wang, Samanthi A. Perera, Sybil M. G. Williams, Roenna Ueda, and Drake LaFace
- Subjects
Antitumor activity ,lcsh:Immunologic diseases. Allergy ,business.industry ,TIGIT ,Immunology ,combination cancer immunotherapy ,immune checkpoint blockade ,Myeloid Cell Activation ,Blockade ,myeloid cells ,Myeloid cells ,FcγR ,Cancer research ,Immunology and Allergy ,Medicine ,costimulatory molecules ,business ,lcsh:RC581-607 - Published
- 2020
- Full Text
- View/download PDF
30. Effective Anti-tumor Response by TIGIT Blockade Associated With FcγR Engagement and Myeloid Cell Activation
- Author
-
Jin-Hwan Han, M. Bigler, Jeffery Grein, Wolfgang Seghezzi, Hongmei Wang, Mingmei Cai, Thomas W. Rosahl, Samanthi A. Perera, Sybil M. G. Williams, Drake LaFace, Roenna Ueda, and Elaine M. Pinheiro
- Subjects
0301 basic medicine ,Programmed Cell Death 1 Receptor ,combination cancer immunotherapy ,Granzymes ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,FcγR ,Tumor Microenvironment ,Immunology and Allergy ,Receptors, Immunologic ,Immune Checkpoint Inhibitors ,Original Research ,Mice, Knockout ,Mice, Inbred BALB C ,education.field_of_study ,Antibodies, Monoclonal ,Tumor Burden ,medicine.anatomical_structure ,myeloid cells ,Colonic Neoplasms ,Female ,costimulatory molecules ,Antibody ,Signal Transduction ,Pore Forming Cytotoxic Proteins ,lcsh:Immunologic diseases. Allergy ,medicine.drug_class ,TIGIT ,T cell ,Immunology ,Population ,Biology ,Monoclonal antibody ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,education ,Receptors, IgG ,Correction ,immune checkpoint blockade ,Immune checkpoint ,Blockade ,Mice, Inbred C57BL ,Granzyme B ,030104 developmental biology ,Cancer research ,biology.protein ,lcsh:RC581-607 ,030215 immunology - Abstract
The molecule "T cell immunoreceptor with immunoglobulin and ITIM domain," or TIGIT, has recently received much attention as a promising target in the treatment of various malignancies. In spite of the quick progression of anti-TIGIT antibodies into clinical testing both as monotherapy and in combination with programmed cell death-1 (PD-1)-directed immune checkpoint blockade, the molecular mechanism behind the observed therapeutic benefits remains poorly understood. Here we demonstrate, using mouse tumor models, that TIGIT blocking antibodies with functional Fc binding potential induce effective anti-tumor response. Our observations reveal that the anti-TIGIT therapeutic effect is not achieved by depletion of intratumoral regulatory T cells (Treg) or any cell population expressing TIGIT, but instead is mediated by possible "reverse activating signals" through FcγRs on myeloid cells, inducing expression of various mediators such as cytokines and chemokines. Furthermore, we discovered an induction of a robust and persistent granzyme B and perforin response, distinct from a predominantly interferon-γ (IFN-γ)-driven anti-PD-1 blockade. Our observations, for the first time, provide the basis for a mechanistic hypothesis involving the requirement of a functional Fc domain of anti-TIGIT monoclonal antibodies, of which various isotypes are currently under intense clinical investigation.
- Published
- 2020
- Full Text
- View/download PDF
31. An orally available non-nucleotide STING agonist with antitumor activity
- Author
-
Laura Price, Samanthi A. Perera, Daniel F. Wyss, Johnny E. Kopinja, Saso Cemerski, Sharad K. Sharma, Timothy J. Henderson, Serena Xu, Andrew M. Haidle, Min Lu, George H. Addona, Greg O’Donnell, Berengere Sauvagnat, Gottfried K. Schroeder, Ilona Kariv, Larissa Rakhilina, Sriram Tyagarajan, Bo-Sheng Pan, Hyun Chong Woo, Brian Long, Jared N. Cumming, Brandon Cash, Yiping Chen, Ryan D. Otte, B. Wesley Trotter, Jeremy Presland, Jennifer Piesvaux, Brian M. Lacey, Rui Liang, Peter J. Dandliker, Ellen C. Minnihan, Charles A. Lesburg, Ian Knemeyer, Yanhong Ma, Guo Feng, David Jonathan Bennett, Michael D. Altman, Alexei V. Buevich, Jason Laskey, James P. Jewell, and Wonsuk Chang
- Subjects
Agonist ,Tumor microenvironment ,Multidisciplinary ,Innate immune system ,Chemistry ,medicine.drug_class ,Administration, Oral ,Membrane Proteins ,Antineoplastic Agents ,Pharmacology ,Research Highlight ,Target validation ,nervous system diseases ,Sting ,Drug screening ,stomatognathic system ,Interferon ,Stimulator of interferon genes ,Systemic administration ,medicine ,Animals ,Humans ,Secretion ,medicine.drug - Abstract
Pharmacological activation of the STING (stimulator of interferon genes)-controlled innate immune pathway is a promising therapeutic strategy for cancer. Here we report the identification of MSA-2, an orally available non-nucleotide human STING agonist. In syngeneic mouse tumor models, subcutaneous and oral MSA-2 regimens were well tolerated and stimulated interferon-β secretion in tumors, induced tumor regression with durable antitumor immunity, and synergized with anti-PD-1 therapy. Experimental and theoretical analyses showed that MSA-2 exists as interconverting monomers and dimers in solution, but only dimers bind and activate STING. This model was validated by using synthetic covalent MSA-2 dimers, which were potent agonists. Cellular potency of MSA-2 increased upon extracellular acidification, which mimics the tumor microenvironment. These properties appear to underpin the favorable activity and tolerability profiles of effective systemic administration of MSA-2.
- Published
- 2020
- Full Text
- View/download PDF
32. Sonoporation‐Enhanced Delivery of STING Agonist Induced Robust Immune Modulation and Tumor Regression (Adv. Therap. 10/2021)
- Author
-
Brian Long, Adam Procopio, Sheila Ranganath, Sachin Mittal, Yun Wang, Clayton T. Larsen, Robert Saklatvala, Allen C. Templeton, Heidi Ferguson, Jun Xu, Mingmei Cai, Ernest G. Schutt, Ken Lin, Miller Paul, Rhodemann Li, Samanthi A. Perera, and Nicolas Solban
- Subjects
Pharmacology ,Agonist ,business.industry ,medicine.drug_class ,Biochemistry (medical) ,Pharmaceutical Science ,Medicine (miscellaneous) ,Immune modulation ,Sting ,Tumor regression ,Cancer research ,Medicine ,Pharmacology (medical) ,business ,Sonoporation ,Genetics (clinical) - Published
- 2021
- Full Text
- View/download PDF
33. Abstract A08: Combination with a novel STING agonist significantly improves efficacy of anti-PD1 therapy in mouse syngeneic tumor models
- Author
-
Ellen C. Minnihan, Hyun Chong Woo, Brian Long, Jared N. Cumming, Johnny E. Kopinja, Heidi Ferguson, Jason Laskey, Bo-Sheng Pan, Ian Knemeyer, Jeremy Presland, Saso Cemerski, George H. Addona, Samanthi A. Perera, Ilona Kariv, Archie Tse, Jennifer Piesvaux, Shuxia Zhao, Kalyan Chakravarthy, Long Cui, Yiping Chen, Yanhong Ma, B. Wesley Trotter, and Sharad K. Sharma
- Subjects
Agonist ,Cancer Research ,biology ,medicine.drug_class ,business.industry ,medicine.medical_treatment ,Immunology ,Immunotherapy ,Sting ,Cytokine ,Immune system ,Interferon ,Stimulator of interferon genes ,medicine ,Cancer research ,biology.protein ,Antibody ,business ,medicine.drug - Abstract
Activated STING (STimulator of INterferon Genes) bound to its natural ligand 2,3-cGAMP (cyclic guanosine-adenosine monophosphate), initiates type I interferon (IFN) and pro-inflammatory cytokine production. IFN upregulation is essential to promote antigen-specific CD8+ T-cell priming and leads to potent anti-tumor activity. To exploit this mechanism we synthesized a new STING agonist, MSA-1, that potently activates both mouse and human STING. Intratumoral (IT) administration of MSA-1 to MC38 syngeneic tumor-bearing mice increased tumor and plasma cytokine levels and was effective at driving complete responses (CRs) in 100% of the animals. Most surviving animals developed tumor-specific adaptive immune memory as demonstrated by robust protection against re-challenge with the same tumor type. Mechanistic studies in immune-deficient mice suggested that the initial antitumor activity is in part due to cytokine-driven cytotoxicity and/or other innate immune mechanisms, which may have contributed to some animals not developing an adaptive immune memory. Importantly, MSA-1 caused long-term tumor regressions or CRs in CT26 and B16-F10 tumor models, both of which are intrinsically resistant to single-agent therapy with a fully murinized anti-mouse PD-1 antibody (muDX400). The antitumor immune response in these models was further enhanced when treating the animals with MSA-1 in combination with muDX400. This combination restored T-cell responses in both blood and tumors of the treated mice and provided long-lived immunologic memory in a majority of the animals. Taken together, these data strongly support the development of STING agonists in combination with Keytruda (humanized anti-PD-1 antibody) for patients with tumors that are partially responsive or nonresponsive to single agent anti-PD-1 therapy. Citation Format: Samanthi A. Perera, Johnny E. Kopinja, Yanhong Ma, Jason Laskey, Kalyan Chakravarthy, Long Cui, Yiping Chen, Jeremy Presland, Sharad Sharma, Shuxia Zhao, Jennifer Piesvaux, Ellen C. Minnihan, Heidi Ferguson, Hyun Chong Woo, Ian Knemeyer, Ilona Kariv, Archie Tse, Saso Cemerski, Jared Cumming, B. Wesley Trotter, Bo-Sheng Pan, George H. Addona, Brian J. Long. Combination with a novel STING agonist significantly improves efficacy of anti-PD1 therapy in mouse syngeneic tumor models [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A08.
- Published
- 2018
- Full Text
- View/download PDF
34. Abstract 4721: Combining STING agonists with an anti-PD-1 antagonist results in marked antitumor activity in immune-excluded tumors
- Author
-
Ian Knemeyer, Jason Laskey, Samanthi A. Perera, Hyun Chong Woo, Saso Cemerski, Brian Long, George H. Addona, Johnny E. Kopinja, Jeremy Presland, Archie Tse, Shuxia Zhao, Ellen C. Minnihan, Yiping Chen, Kalyan Chakravarthy, Jennifer Piesvaux, Long Cui, Heidi Ferguson, Wesley B. Trotter, Jared N. Cumming, Bo-Sheng Pan, and Yanhong Ma
- Subjects
0301 basic medicine ,Agonist ,Cancer Research ,Innate immune system ,medicine.drug_class ,business.industry ,medicine.medical_treatment ,Acquired immune system ,Type I interferon production ,03 medical and health sciences ,Sting ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,Immune system ,Oncology ,030220 oncology & carcinogenesis ,Stimulator of interferon genes ,medicine ,Cancer research ,business - Abstract
The innate immune agonist STING (STimulator of INterferon Genes) binds its natural ligand 2'3'-cGAMP (cyclic guanosine-adenosine monophosphate) and initiates type I interferon production. This promotes systemic antigen-specific CD8+ T-cell priming that eventually provides potent anti-tumor activity. To exploit this mechanism we synthesized a novel STING agonist, MSA-1, that activates both mouse and human STING with higher in vitro potency than cGAMP. MSA-1 was administered to immune-competent mice bearing MC38 syngeneic tumors to monitor pharmacodynamics, pharmacokinetics and in vivo efficacy. Intratumoral (IT) dosed MSA-1 demonstrated robust tumor and plasma cytokine upregulation and effective anti-tumor activity. The highest tolerated doses provided complete responses (CRs) in 100% of MC38 tumors. Tumor models such as CT26 and B16-F10 that are intrinsically resistant to single-agent therapy with a fully murinized mouse anti-PD-1 antibody (mDX400) also demonstrated long-term tumor regressions or CRs. Mechanistic studies in immune-deficient mice suggested that anti-tumor activity of IT dosed STING agonists are in part due to cytotoxicity and/or innate immune responses rather than development of robust adaptive anti-tumor immunity. To enhance the adaptive immune response, we combined MSA-1 with mDX400 in mouse syngeneic tumor models previously characterized to be unresponsive to anti-PD-1 blockade. This combination restored T-cell responses in both blood and tumors of the treated mice and provided long-lived immunologic memory in a majority of the animals. Taken together, these data strongly support the development of STING agonists in combination with Keytruda for patients with tumors that are partially responsive or non-responsive to single agent anti-PD-1 therapy. Citation Format: Samanthi A. Perera, Johnny E. Kopinja, Yanhong Ma, Jason Laskey, Kalyan Chakravarthy, Yiping Chen, Long Cui, Jeremy Presland, Shuxia Zhao, Ellen Minnihan, Heidi Ferguson, Jennifer Piesvaux, Bo-Sheng Pan, Hyun Chong Woo, Ian Knemeyer, Saso Cemerski, Jared Cumming, Wesley Trotter, Archie Tse, George H. Addona, Brian J. Long. Combining STING agonists with an anti-PD-1 antagonist results in marked antitumor activity in immune-excluded tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4721.
- Published
- 2018
- Full Text
- View/download PDF
35. HER2 YVMA drives rapid development of adenosquamous lung tumors in mice that are sensitive to BIBW2992 and rapamycin combination therapy
- Author
-
Flavio Solca, Shigeto Kubo, Maria Gabriela Raso, Pilar Garin Chesa, Geoffrey I. Shapiro, Sara Zaghlul, Ulrich Guertler, Danan Li, Kwok-Kin Wong, Ignacio I. Wistuba, Christa L. Borgman, Heidi Greulich, Matthew Meyerson, Kathleyn A. Brandstetter, Roderick T. Bronson, Masaya Takahashi, Robert F. Padera, Lucian R. Chirieac, Samanthi A. Perera, Liang Chen, Hongbin Ji, and Takeshi Shimamura
- Subjects
Lung Neoplasms ,Combination therapy ,Receptor, ErbB-2 ,Afatinib ,Drug Evaluation, Preclinical ,Biology ,medicine.disease_cause ,Carcinoma, Adenosquamous ,Mice ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Phosphorylation ,skin and connective tissue diseases ,Lung cancer ,neoplasms ,Protein kinase B ,Sirolimus ,Multidisciplinary ,Biological Sciences ,medicine.disease ,Drug Resistance, Neoplasm ,Mutation ,Quinazolines ,Cancer research ,Erlotinib ,Carcinogenesis ,Signal Transduction ,medicine.drug - Abstract
Mutations in the HER2 kinase domain have been identified in human clinical lung cancer specimens. Here we demonstrate that inducible expression of the most common HER2 mutant (HER2 YVMA ) in mouse lung epithelium causes invasive adenosquamous carcinomas restricted to proximal and distal bronchioles. Continuous expression of HER2 YVMA is essential for tumor maintenance, suggesting a key role for HER2 in lung adenosquamous tumorigenesis. Preclinical studies assessing the in vivo effect of erlotinib, trastuzumab, BIBW2992, and/or rapamycin on HER2 YVMA transgenic mice or H1781 xenografts with documented tumor burden revealed that the combination of BIBW2992 and rapamycin is the most effective treatment paradigm causing significant tumor shrinkage. Immunohistochemical analysis of lung tumors treated with BIBW2992 and rapamycin combination revealed decreased phosphorylation levels for proteins in both upstream and downstream arms of MAPK and Akt/mTOR signaling axes, indicating inhibition of these pathways. Based on these findings, clinical testing of the BIBW2992/rapamycin combination in non-small cell lung cancer patients with tumors expressing HER2 mutations is warranted.
- Published
- 2009
- Full Text
- View/download PDF
36. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers
- Author
-
Kate McNamara, Robert F. Padera, Angela Lightbown, S. Michel Maira, Samanthi A. Perera, Lucian R. Chirieac, Ramneet Kaur, Ralph Weissleder, Liang Chen, Katherine Crosby, Lewis C. Cantley, Alexander R. Guimaraes, Umar Mahmood, Jessica Simendinger, Jeffrey A. Engelman, Xiaohong Tan, Youngchul Song, Kwok-Kin Wong, Rabi Upadhyay, Carlos Garcia-Echeverria, and Timothy Q. Li
- Subjects
Lung Neoplasms ,Class I Phosphatidylinositol 3-Kinases ,MAP Kinase Signaling System ,Down-Regulation ,Mice, Transgenic ,Biology ,Mitogen-activated protein kinase kinase ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Proto-Oncogene Proteins p21(ras) ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Animals ,c-Raf ,Enzyme Inhibitors ,Protein kinase A ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,030304 developmental biology ,Mitogen-Activated Protein Kinase Kinases ,0303 health sciences ,Kinase ,General Medicine ,Molecular biology ,3. Good health ,Gene Expression Regulation, Neoplastic ,Protein kinase domain ,030220 oncology & carcinogenesis ,Genetically Engineered Mouse ,Mutation ,ras Proteins ,Cancer research ,KRAS - Abstract
Somatic mutations that activate phosphoinositide 3-kinase (PI3K) have been identified in the p110-alpha catalytic subunit (encoded by PIK3CA). They are most frequently observed in two hotspots: the helical domain (E545K and E542K) and the kinase domain (H1047R). Although the p110-alpha mutants are transforming in vitro, their oncogenic potential has not been assessed in genetically engineered mouse models. Furthermore, clinical trials with PI3K inhibitors have recently been initiated, and it is unknown if their efficacy will be restricted to specific, genetically defined malignancies. In this study, we engineered a mouse model of lung adenocarcinomas initiated and maintained by expression of p110-alpha H1047R. Treatment of these tumors with NVP-BEZ235, a dual pan-PI3K and mammalian target of rapamycin (mTOR) inhibitor in clinical development, led to marked tumor regression as shown by positron emission tomography-computed tomography, magnetic resonance imaging and microscopic examination. In contrast, mouse lung cancers driven by mutant Kras did not substantially respond to single-agent NVP-BEZ235. However, when NVP-BEZ235 was combined with a mitogen-activated protein kinase kinase (MEK) inhibitor, ARRY-142886, there was marked synergy in shrinking these Kras-mutant cancers. These in vivo studies suggest that inhibitors of the PI3K-mTOR pathway may be active in cancers with PIK3CA mutations and, when combined with MEK inhibitors, may effectively treat KRAS mutated lung cancers.
- Published
- 2008
- Full Text
- View/download PDF
37. Proapoptotic BH3-Only BCL-2 Family Protein BIM Connects Death Signaling from Epidermal Growth Factor Receptor Inhibition to the Mitochondrion
- Author
-
Geoffrey I. Shapiro, Kwok-Kin Wong, Jing Deng, Anthony Letai, Nicole Carlson, Samanthi A. Perera, Dongpo Cai, and Takeshi Shimamura
- Subjects
Cancer Research ,Lung Neoplasms ,Cell Survival ,Apoptosis ,Biology ,Erlotinib Hydrochloride ,Growth factor receptor ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Humans ,Epidermal growth factor receptor ,RNA, Small Interfering ,Protein Kinase Inhibitors ,neoplasms ,EGFR inhibitors ,Bcl-2-Like Protein 11 ,Gene Expression Profiling ,Bcl-2 family ,Membrane Proteins ,Caspase 9 ,Mitochondria ,respiratory tract diseases ,ErbB Receptors ,Oncology ,Quinazolines ,Cancer research ,biology.protein ,Erlotinib ,Signal transduction ,Apoptosis Regulatory Proteins ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction ,medicine.drug - Abstract
A subset of lung cancers expresses mutant forms of epidermal growth factor receptor (EGFR) that are constitutively activated. Cancers bearing activated EGFR can be effectively targeted with EGFR inhibitors such as erlotinib. However, the death-signaling pathways engaged after EGFR inhibition are poorly understood. Here, we show that death after inhibition of EGFR uses the mitochondrial, or intrinsic, pathway of cell death controlled by the BCL-2 family of proteins. BCL-2 inhibits cell death induced by erlotinib, but BCL-2–protected cells are thus rendered BCL-2–dependent and sensitive to the BCL-2 antagonist ABT-737. BH3 profiling reveals that mitochondrial BCL-2 is primed by death signals after EGFR inhibition in these cells. As this result implies, key death-signaling proteins of the BCL-2 family, including BIM, were found to be up-regulated after erlotinib treatment and intercepted by overexpressed BCL-2. BIM is induced by lung cancer cell lines that are sensitive to erlotinib but not by those resistant. Reduction of BIM by siRNA induces resistance to erlotinib. We show that EGFR activity is inhibited by erlotinib in H1650, a lung cancer cell line that bears a sensitizing EGFR mutation, but that H1650 is not killed. We identify the block in apoptosis in this cell line, and show that a novel form of erlotinib resistance is present, a block in BIM up-regulation downstream of EGFR inhibition. This finding has clear implications for overcoming resistance to erlotinib. Resistance to EGFR inhibition can be modulated by alterations in the intrinsic apoptotic pathway controlled by the BCL-2 family of proteins. [Cancer Res 2007;67(24):11867–75]
- Published
- 2007
- Full Text
- View/download PDF
38. LKB1 modulates lung cancer differentiation and metastasis
- Author
-
Xihong Lin, Takeshi Shimamura, Diego H. Castrillon, Bruce E. Johnson, Danan Li, Matthew R. Ramsey, Lucian R. Chirieac, Jussi Koivunen, Geoffrey I. Shapiro, David J. Kwiatkowski, David C. Christiani, Lei Bao, Pasi A. Jänne, Kate McNamara, Piotr Kozlowski, Mei-Chih Liang, Janakiraman Krishnamurthy, Chad Torrice, Hongbin Ji, Matthew Meyerson, Cristina Contreras, D. Neil Hayes, Roderick T. Bronson, Neal I. Lindeman, George N. Naumov, Samanthi A. Perera, Cheng Fan, Nabeel Bardeesy, Robert F. Padera, Kwok-Kin Wong, Michael C. Wu, Dongpo Cai, Norman E. Sharpless, and Liang Chen
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Lung Neoplasms ,STK11 ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Biology ,medicine.disease_cause ,Metastasis ,Mice ,Germline mutation ,AMP-Activated Protein Kinase Kinases ,CDKN2A ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Carcinoma ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Neoplasm Metastasis ,skin and connective tissue diseases ,Lung cancer ,Multidisciplinary ,Gene Expression Profiling ,Genes, p16 ,Cancer ,Cell Differentiation ,Genes, p53 ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Genes, ras ,Cancer research ,Carcinogenesis ,Genes, Neoplasm - Abstract
Germline mutation in serine/threonine kinase 11 (STK11, also called LKB1) results in Peutz-Jeghers syndrome, characterized by intestinal hamartomas and increased incidence of epithelial cancers. Although uncommon in most sporadic cancers, inactivating somatic mutations of LKB1 have been reported in primary human lung adenocarcinomas and derivative cell lines. Here we used a somatically activatable mutant Kras-driven model of mouse lung cancer to compare the role of Lkb1 to other tumour suppressors in lung cancer. Although Kras mutation cooperated with loss of p53 or Ink4a/Arf (also known as Cdkn2a) in this system, the strongest cooperation was seen with homozygous inactivation of Lkb1. Lkb1-deficient tumours demonstrated shorter latency, an expanded histological spectrum (adeno-, squamous and large-cell carcinoma) and more frequent metastasis compared to tumours lacking p53 or Ink4a/Arf. Pulmonary tumorigenesis was also accelerated by hemizygous inactivation of Lkb1. Consistent with these findings, inactivation of LKB1 was found in 34% and 19% of 144 analysed human lung adenocarcinomas and squamous cell carcinomas, respectively. Expression profiling in human lung cancer cell lines and mouse lung tumours identified a variety of metastasis-promoting genes, such as NEDD9, VEGFC and CD24, as targets of LKB1 repression in lung cancer. These studies establish LKB1 as a critical barrier to pulmonary tumorigenesis, controlling initiation, differentiation and metastasis.
- Published
- 2007
- Full Text
- View/download PDF
39. Bronchial and Peripheral Murine Lung Carcinomas Induced by T790M-L858R Mutant EGFR Respond to HKI-272 and Rapamycin Combination Therapy
- Author
-
Mei-Chih Liang, Takeshi Shimamura, Liang Chen, Neal I. Lindeman, Robert F. Padera, Sara Zaghlul, Hongbin Ji, Matthew Meyerson, Kwok-Kin Wong, Samanthi A. Perera, Danan Li, Kate McNamara, Christa L. Borgman, Geoffrey I. Shapiro, Roman K. Thomas, Jeffrey A. Engelman, Yanping Sun, Masaya Takahashi, Henry J. Haringsma, Pasi A. Jänne, Michael J. Eck, Lucian R. Chirieac, and Shigeto Kubo
- Subjects
Cancer Research ,Lung Neoplasms ,Combination therapy ,CELLCYCLE ,Mice ,03 medical and health sciences ,T790M ,0302 clinical medicine ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,In Situ Nick-End Labeling ,Animals ,Medicine ,Lung cancer ,030304 developmental biology ,Sirolimus ,0303 health sciences ,Lung ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Bronchial Neoplasms ,Cell Biology ,Cell cycle ,respiratory system ,Resistance mutation ,medicine.disease ,Immunohistochemistry ,3. Good health ,respiratory tract diseases ,ErbB Receptors ,medicine.anatomical_structure ,CHEMBIO ,Oncology ,SIGNALING ,030220 oncology & carcinogenesis ,Mutation ,Immunology ,Quinolines ,Cancer research ,Erlotinib ,business ,medicine.drug - Abstract
SummaryThe EGFR T790M mutation has been identified in tumors from lung cancer patients that eventually develop resistance to erlotinib. In this study, we generated a mouse model with doxycycline-inducible expression of a mutant EGFR containing both L858R, an erlotinib-sensitizing mutation, and the T790M resistance mutation (EGFR TL). Expression of EGFR TL led to development of peripheral adenocarcinomas with bronchioloalveolar features in alveoli as well as papillary adenocarcinomas in bronchioles. Treatment with an irreversible EGFR tyrosine kinase inhibitor (TKI), HKI-272, shrunk only peripheral tumors but not bronchial tumors. However, the combination of HKI-272 and rapamycin resulted in significant regression of both types of lung tumors. This combination therapy may potentially benefit lung cancer patients with the EGFR T790M mutation.
- Published
- 2007
- Full Text
- View/download PDF
40. Localization of BCR-ABL to F-actin regulates cell adhesion but does not attenuate CML development
- Author
-
Warren S. Pear, Daniel A. Hammer, David Boettiger, Ruibao Ren, Jason A. Wertheim, and Samanthi A. Perera
- Subjects
Immunology ,Integrin ,Fusion Proteins, bcr-abl ,Bone Marrow Cells ,Integrin alpha4beta1 ,Biochemistry ,Mice ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Cell Adhesion ,medicine ,Animals ,Cell adhesion ,Bone Marrow Transplantation ,biology ,Cell adhesion molecule ,breakpoint cluster region ,Cell Biology ,Hematology ,Adhesion ,medicine.disease ,Actins ,Fibronectins ,Protein Structure, Tertiary ,Cell biology ,Mice, Inbred C57BL ,Fibronectin ,biology.protein ,Cancer research ,Neural cell adhesion molecule ,Integrin alpha5beta1 ,Protein Binding ,Chronic myelogenous leukemia - Abstract
We have previously found that P210BCR-ABL increases the adhesion of hematopoietic cell lines to fibronectin by a mechanism that is independent of tyrosine kinase activity. To investigate the pathway(s) by which P210BCR-ABL influences cell adhesion, we used a quantitative cell adhesion device that can discern small changes in cell adhesion to assay P210BCR-ABL with mutations in several critical domains. We expressed P210BCR-ABL mutants in 32D myeloblast cells and found that binding to fibronectin is mediated primarily by the α5β1 integrin. We performed a structure/function analysis to map domains important for cell adhesion. Increased adhesion was mediated by 3 domains: (1) the N-terminal coiled-coil domain that facilitates oligomerization and F-actin localization; (2) bcr sequences between aa 163 to 210; and (3) F-actin localization through the C-terminal actin-binding domain of c-abl. We compared our adhesion results with the ability of these mutants to cause a chronic myelogenous leukemia (CML)–like disease in a murine bone marrow transplantation assay and found that adhesion to fibronectin did not correlate with the ability of these mutants to cause CML. Together, our results suggest that F-actin localization may play a pivotal role in modulating adhesion but that it is dispensable for the development of CML.
- Published
- 2003
- Full Text
- View/download PDF
41. Ganetespib (STA-9090), a nongeldanamycin HSP90 inhibitor, has potent antitumor activity in in vitro and in vivo models of non-small cell lung cancer
- Author
-
John-Paul Jimenez, Christopher D. Carey, Danan Li, James Barsoum, Kwok-Kin Wong, Jim Sang, Weiwen Ying, Takeshi Shimamura, Liang Chen, Matthew Meyerson, Yu-Chen Li, Christa L. Borgman, Samanthi A. Perera, Takayo Inoue, Julian Carretero, Papiya Sinha, Scott J. Rodig, Geoffrey I. Shapiro, and Kevin Foley
- Subjects
Cancer Research ,Lung Neoplasms ,Lactams, Macrocyclic ,Ganetespib ,Antineoplastic Agents ,Mice, SCID ,Pharmacology ,Receptor tyrosine kinase ,Article ,Hsp90 inhibitor ,chemistry.chemical_compound ,T790M ,Mice ,In vivo ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,polycyclic compounds ,Benzoquinones ,Animals ,Humans ,HSP90 Heat-Shock Proteins ,Prostaglandin-E Synthases ,biology ,Protein Stability ,Geldanamycin ,Triazoles ,Hsp90 ,Xenograft Model Antitumor Assays ,Intramolecular Oxidoreductases ,Oncology ,chemistry ,Apoptosis ,biology.protein ,Female ,Protein Binding - Abstract
Purpose: We describe the anticancer activity of ganetespib, a novel non-geldanamycin heat shock protein 90 (HSP90) inhibitor, in non-small cell lung cancer (NSCLC) models. Experimental Design: The activity of ganetespib was compared with that of the geldanamycin 17-AAG in biochemical assays, cell lines, and xenografts, and evaluated in an ERBB2 YVMA-driven mouse lung adenocarcinoma model. Results: Ganetespib blocked the ability of HSP90 to bind to biotinylated geldanamycin and disrupted the association of HSP90 with its cochaperone, p23, more potently than 17-AAG. In genomically defined NSCLC cell lines, ganetespib caused depletion of receptor tyrosine kinases, extinguishing of downstream signaling, inhibition of proliferation and induction of apoptosis with IC50 values ranging 2 to 30 nmol/L, substantially lower than those required for 17-AAG (20–3,500 nmol/L). Ganetespib was also approximately 20-fold more potent in isogenic Ba/F3 pro-B cells rendered IL-3 independent by expression of EGFR and ERBB2 mutants. In mice bearing NCI-H1975 (EGFR L858R/T790M) xenografts, ganetespib was rapidly eliminated from plasma and normal tissues but was maintained in tumor with t1/2 58.3 hours, supporting once-weekly dosing experiments, in which ganetespib produced greater tumor growth inhibition than 17-AAG. However, after a single dose, reexpression of mutant EGFR occurred by 72 hours, correlating with reversal of antiproliferative and proapoptotic effects. Consecutive day dosing resulted in xenograft regressions, accompanied by more sustained pharmacodynamic effects. Ganetespib also showed activity against mouse lung adenocarcinomas driven by oncogenic ERBB2 YVMA. Conclusions: Ganetespib has greater potency than 17-AAG and potential efficacy against several NSCLC subsets, including those harboring EGFR or ERBB2 mutation. Clin Cancer Res; 18(18); 4973–85. ©2012 AACR.
- Published
- 2012
42. Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer
- Author
-
Samanthi A. Perera, Aviva Goel, Matthäus Getlik, Daniel Rauh, Lukas C. Heukamp, Martin L. Sos, Lucia Regales, Klaus P. Hoeflich, Katerina Politi, William Pao, Hamid Kashkar, Sascha Ansén, Lori Friedman, Christine Orr, Martin Peifer, Roman K. Thomas, Johannes M. Heuckmann, William R. Sellers, Kevan M. Shokat, Florian Fischer, Stefanie Fischer, Isabel Stückrath, Mirjam Koker, Jonathan Weiss, Kwok-Kin Wong, Rameen Beroukhim, Thomas Zander, Roland T. Ullrich, Stefanie Heynck, and Kathrin Michel
- Subjects
MAPK/ERK pathway ,Multidisciplinary ,biology ,Genotype ,MAP Kinase Signaling System ,Cancer ,Apoptosis ,Biological Sciences ,medicine.disease ,Receptor tyrosine kinase ,Cell biology ,Phosphatidylinositol 3-Kinases ,In vivo ,Cell culture ,Cell Line, Tumor ,Neoplasms ,biology.protein ,medicine ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors - Abstract
In cancer, genetically activated proto-oncogenes often induce “ upstream ” dependency on the activity of the mutant oncoprotein. Therapeutic inhibition of these activated oncoproteins can induce massive apoptosis of tumor cells, leading to sometimes dramatic tumor regressions in patients. The PI3K and MAPK signaling pathways are central regulators of oncogenic transformation and tumor maintenance. We hypothesized that upstream dependency engages either one of these pathways preferentially to induce “ downstream ” dependency. Therefore, we analyzed whether downstream pathway dependency segregates by genetic aberrations upstream in lung cancer cell lines. Here, we show by systematically linking drug response to genomic aberrations in non-small-cell lung cancer, as well as in cell lines of other tumor types and in a series of in vivo cancer models, that tumors with genetically activated receptor tyrosine kinases depend on PI3K signaling, whereas tumors with mutations in the RAS/RAF axis depend on MAPK signaling. However, efficacy of downstream pathway inhibition was limited by release of negative feedback loops on the reciprocal pathway. By contrast, combined blockade of both pathways was able to overcome the reciprocal pathway activation induced by inhibitor-mediated release of negative feedback loops and resulted in a significant increase in apoptosis and tumor shrinkage. Thus, by using a systematic chemo-genomics approach, we identify genetic lesions connected to PI3K and MAPK pathway activation and provide a rationale for combined inhibition of both pathways. Our findings may have implications for patient stratification in clinical trials.
- Published
- 2009
43. Differential Roles of Telomere Attrition in Type I and II Endometrial Carcinogenesis
- Author
-
Esra A. Akbay, Russell Broaddus, Diego H. Castrillon, Kwok-Kin Wong, James P. Sullivan, John O. Schorge, Samanthi A. Perera, Cristina Contreras, Raheela Ashfaq, and Hossein Saboorian
- Subjects
Genome instability ,Pathology ,medicine.medical_specialty ,Stromal cell ,Chromogenic in situ hybridization ,In situ hybridization ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Endometrium ,Mice ,medicine ,Animals ,Humans ,In Situ Hybridization ,Aged ,Endometrial cancer ,Telomere ,medicine.disease ,Aneuploidy ,Endometrial Neoplasms ,Serous fluid ,Cell Transformation, Neoplastic ,Female ,Carcinogenesis ,Regular Articles - Abstract
Endometrial cancer has been generally categorized into two broad groups of tumors, type I (TI) and type II (TII), with distinct epidemiological/clinical features and genetic alterations. Because telomere attrition appears to trigger genomic instability in certain cancers, we explored the role of telomere dysfunction in endometrial cancer by analyzing telomeres and other markers of telomere status in both tumor types. We describe a new method, telomere chromogenic in situ hybridization, which permitted us to detect cells with short telomeres relative to control (stromal) cells within the same tissue section. Using this method, we found that both types of tumor cells had short telomeres. However, only TII tumors were significantly associated with critical telomere shortening in adjacent, morphologically normal epithelium, suggesting that telomere shortening contributes to the initiation of TII but not TI tumors. To explore this hypothesis, we analyzed mice with critically short telomeres and documented distinctive endometrial lesions that histologically resembled the in situ precursor of TII serous carcinomas; these lesions have not been observed previously in TI mouse models of endometrial cancer. Based on this and previous studies, we propose a model in which telomere attrition contributes to the initiation of TII and progression of TI endometrial cancers.
- Published
- 2008
44. Hsp90 inhibition suppresses mutant EGFR-T790M signaling and overcomes kinase inhibitor resistance
- Author
-
Daniel G. Tenen, Masaya Takahashi, Danan Li, Yuko Minami, Kate McNamara, Susumu Kobayashi, Lucian R. Chirieac, Matthew Meyerson, Samanthi A. Perera, Geoffrey I. Shapiro, Hongbin Ji, Christa L. Borgman, Shigeto Kubo, Henry J. Haringsma, Roman K. Thomas, Heidi Greulich, Elizabeth Liniker, Kwok-Kin Wong, Sara Zaghlul, Robert F. Padera, Takeshi Shimamura, and April M. Lowell
- Subjects
Cancer Research ,Lung Neoplasms ,Antineoplastic Agents ,Apoptosis ,Biology ,Adenocarcinoma ,Receptor tyrosine kinase ,Article ,T790M ,Mice ,Gefitinib ,Cell Line, Tumor ,medicine ,Animals ,Epidermal growth factor receptor ,HSP90 Heat-Shock Proteins ,Enzyme Inhibitors ,Protein kinase B ,EGFR inhibitors ,TOR Serine-Threonine Kinases ,respiratory tract diseases ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Oncology ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,biology.protein ,Erlotinib ,Tyrosine kinase ,Protein Kinases ,medicine.drug ,Signal Transduction - Abstract
The epidermal growth factor receptor (EGFR) secondary kinase domain T790M non–small cell lung cancer (NSCLC) mutation enhances receptor catalytic activity and confers resistance to the reversible tyrosine kinase inhibitors gefitinib and erlotinib. Currently, irreversible inhibitors represent the primary approach in clinical use to circumvent resistance. We show that higher concentrations of the irreversible EGFR inhibitor CL-387,785 are required to inhibit EGFR phosphorylation in T790M-expressing cells compared with EGFR mutant NSCLC cells without T790M. Additionally, CL-387,785 does not fully suppress phosphorylation of other activated receptor tyrosine kinases (RTK) in T790M-expressing cells. These deficiencies result in residual Akt and mammalian target of rapamycin (mTOR) activities. Full suppression of EGFR-mediated signaling in T790M-expressing cells requires the combination of CL-387,785 and rapamycin. In contrast, Hsp90 inhibition overcomes these limitations in vitro and depletes cells of EGFR, other RTKs, and phospho-Akt and inhibits mTOR signaling whether or not T790M is present. EGFR-T790M–expressing cells rendered resistant to CL-387,785 by a kinase switch mechanism retain sensitivity to Hsp90 inhibition. Finally, Hsp90 inhibition causes regression in murine lung adenocarcinomas driven by mutant EGFR (L858R) with or without T790M. However, efficacy in the L858R-T790M model requires a more intense treatment schedule and responses were transient. Nonetheless, these findings suggest that Hsp90 inhibitors may be effective in T790M-expressing cells and offer an alternative therapeutic strategy for this subset of lung cancers. [Cancer Res 2008;68(14):5827–38]
- Published
- 2008
45. Mutations in BRAF and KRAS converge on activation of the mitogen-activated protein kinase pathway in lung cancer mouse models
- Author
-
Yanping Sun, Levi A. Garraway, Zhenxiong Wang, Danan Li, Mei-Chih Liang, Kate McNamara, Bruce E. Johnson, Roderick T. Bronson, Robert F. Padera, Lucian R. Chirieac, Lynda Chin, Roman K. Thomas, Kwok-Kin Wong, Ruqayyah Al-Hashem, Hongbin Ji, Sara Zaghlul, Pasi A. Jänne, Liang Chen, Mitchell S. Albert, and Samanthi A. Perera
- Subjects
MAPK/ERK pathway ,Proto-Oncogene Proteins B-raf ,Cancer Research ,Lung Neoplasms ,MAP Kinase Signaling System ,MAP Kinase Kinase 2 ,MAP Kinase Kinase 1 ,Mice, Transgenic ,Biology ,Adenocarcinoma ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,Mice ,medicine ,Animals ,Protein kinase A ,Lung cancer ,Kinase ,MEK inhibitor ,Adenocarcinoma, Bronchiolo-Alveolar ,medicine.disease ,respiratory tract diseases ,Disease Models, Animal ,Genes, ras ,Oncology ,Mitogen-activated protein kinase ,Doxycycline ,Benzamides ,Mutation ,Cancer research ,biology.protein ,KRAS ,Mitogen-Activated Protein Kinases - Abstract
Mutations in the BRAF and KRAS genes occur in ∼1% to 2% and 20% to 30% of non–small-cell lung cancer patients, respectively, suggesting that the mitogen-activated protein kinase (MAPK) pathway is preferentially activated in lung cancers. Here, we show that lung-specific expression of the BRAF V600E mutant induces the activation of extracellular signal–regulated kinase (ERK)-1/2 (MAPK) pathway and the development of lung adenocarcinoma with bronchioloalveolar carcinoma features in vivo. Deinduction of transgene expression led to dramatic tumor regression, paralleled by dramatic dephosphorylation of ERK1/2, implying a dependency of BRAF-mutant lung tumors on the MAPK pathway. Accordingly, in vivo pharmacologic inhibition of MAPK/ERK kinase (MEK; MAPKK) using a specific MEK inhibitor, CI-1040, induced tumor regression associated with inhibition of cell proliferation and induction of apoptosis in these de novo lung tumors. CI-1040 treatment also led to dramatic tumor shrinkage in murine lung tumors driven by a mutant KRas allele. Thus, somatic mutations in different signaling intermediates of the same pathway induce exquisite dependency on a shared downstream effector. These results unveil a potential common vulnerability of BRAF and KRas mutant lung tumors that potentially affects rational deployment of MEK targeted therapies to non–small-cell lung cancer patients. [Cancer Res 2007;67(10):4933–9]
- Published
- 2007
46. Molecular evolution of two vertebrate aryl hydrocarbon (dioxin) receptors (AHR1 and AHR2) and the PAS family
- Author
-
Sibel I. Karchner, Miriam A. Shapiro, Mark E. Hahn, and Samanthi A. Perera
- Subjects
Aryl hydrocarbon receptor nuclear translocator ,Molecular Sequence Data ,Biology ,Homology (biology) ,Evolution, Molecular ,Species Specificity ,Phylogenetics ,Molecular evolution ,biology.animal ,Basic Helix-Loop-Helix Transcription Factors ,Gene family ,Animals ,Humans ,Amino Acid Sequence ,Phylogeny ,Genetics ,Multidisciplinary ,Phylogenetic tree ,Sequence Homology, Amino Acid ,Fishes ,Vertebrate ,respiratory system ,Biological Sciences ,Aryl hydrocarbon receptor ,Invertebrates ,respiratory tract diseases ,Receptors, Aryl Hydrocarbon ,Multigene Family ,Vertebrates ,biology.protein ,Trans-Activators - Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor through which halogenated aromatic hydrocarbons such as 2,3,7,8-tetrachlorodibenzo -p- dioxin (TCDD) cause altered gene expression and toxicity. The AHR belongs to the basic helix–loop–helix/Per-ARNT-Sim (bHLH-PAS) family of transcriptional regulatory proteins, whose members play key roles in development, circadian rhythmicity, and environmental homeostasis; however, the normal cellular function of the AHR is not yet known. As part of a phylogenetic approach to understanding the function and evolutionary origin of the AHR, we sequenced the PAS homology domain of AHRs from several species of early vertebrates and performed phylogenetic analyses of these AHR amino acid sequences in relation to mammalian AHRs and 24 other members of the PAS family. AHR sequences were identified in a teleost (the killifish Fundulus heteroclitus ), two elasmobranch species (the skate Raja erinacea and the dogfish Mustelus canis ), and a jawless fish (the lamprey Petromyzon marinus ). Two putative AHR genes, designated AHR1 and AHR2 , were found both in Fundulus and Mustelus . Phylogenetic analyses indicate that the AHR2 genes in these two species are orthologous, suggesting that an AHR gene duplication occurred early in vertebrate evolution and that multiple AHR genes may be present in other vertebrates. Database searches and phylogenetic analyses identified four putative PAS proteins in the nematode Caenorhabditis elegans , including possible AHR and ARNT homologs. Phylogenetic analysis of the PAS gene family reveals distinct clades containing both invertebrate and vertebrate PAS family members; the latter include paralogous sequences that we propose have arisen by gene duplication early in vertebrate evolution. Overall, our analyses indicate that the AHR is a phylogenetically ancient protein present in all living vertebrate groups (with a possible invertebrate homolog), thus providing an evolutionary perspective to the study of dioxin toxicity and AHR function.
- Published
- 1997
47. Abstract 698: MCL1 dependent cells are sensitive to the CDK inhibitor Dinaciclib
- Author
-
Xianlu Qu, Samanthi A. Perera, Harold Hatch, Nathan R. Miselis, Danielle M. Greenawalt, Alwin Schuller, Mark Ayers, Robert Booher, Michael Nebozhyn, Lauren Harmonay, Ellie Im, Heather Hirsch, Thorseten Graef, Thi D.T. Nguyen, Minilik Angagaw, Andrey Loboda, Samer Al-Assaad, Brian Long, Rebecca L. Blanchard, Peter Strack, Leigh Zawel, and Brian Dolinski
- Subjects
Cancer Research ,biology ,business.industry ,Cell ,Cancer ,Cell cycle ,medicine.disease ,chemistry.chemical_compound ,medicine.anatomical_structure ,Oncology ,chemistry ,Apoptosis ,Cyclin-dependent kinase ,Immunology ,biology.protein ,Cancer research ,Medicine ,MCL1 ,Dinaciclib ,business ,CDK inhibitor - Abstract
Dinaciclib is a potent inhibitor of cyclin dependent kinases (CDKs) 1, 2, 5, and 9 and is currently in Phase 3 for the treatment of refractory chronic lymphocytic leukemia (CLL). To further understand the mechanism of action, identify predictive biomarkers, and find additional cancer types which may benefit from dinaciclib, we evaluated cell viability following 24 hours treatment across a panel of ∼500 cells lines. Hematopoietic cell lines were on average 3-times more sensitive than solid tumor lines. In agreement with previous findings, mRNA expression of the anti-apoptotic family member BCL-xL or the ratio of MCL1-to-BCL-xL continue to be the best predictor of dinaciclib sensitivity in both hematopoietic and solid tumor cell lines. MCL1 appears to be an important target of dinaciclib particularly in MCL1 amplified cell lines. Dependence on MCL1 was established in a panel of 19 breast, NSCLC and SCLC cell lines by depletion of the protein by either dinaciclib treatment or MCL1 RNAi. The NSCLC line H23 was highly dependent on MCL1, as RNAi knockdown decreased viability to 80% tumor regression. Cell lines which lacked pro-apoptotic proteins BAX / BAK or harbored a BAX mutation were insensitive to the inhibitor. Using apoptosis defective lines we demonstrate that 24 hours of dinaciclib treatment still impacted cell count by blocking cell cycle progression as measured by FACS. These data demonstrate that both cell cycle block and induction of apoptosis contribute to dinaciclib's mechanism of action. However, the observation that MCL1 and BCL-xL were top genes associated with sensitivity suggests that induction of apoptosis is the predominant mechanism of dinaciclib's anti-tumor effect and warrants further investigation of MCL1 amplification as a predictive biomarker in future clinical studies. Citation Format: Harold Hatch, Robert Booher, Samanthi Perera, Thi Nguyen, Brian Dolinski, Samer Al-Assaad, Lauren Harmonay, Alwin Schuller, Minilik Angagaw, Brian Long, Xianlu Qu, Nathan Miselis, Mark Ayers, Michael Nebozhyn, Heather Hirsch, Danielle Greenawalt, Andrey Loboda, Thorseten Graef, Ellie Im, Rebecca Blanchard, Leigh Zawel, Peter Strack. MCL1 dependent cells are sensitive to the CDK inhibitor Dinaciclib. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 698. doi:10.1158/1538-7445.AM2013-698
- Published
- 2013
- Full Text
- View/download PDF
48. Telomere dysfunction promotes genome instability and metastatic potential in a K-ras p53 mouse model of lung cancer.
- Author
-
Samanthi A. Perera, Richard S. Maser, Huili Xia, Kate McNamara, Alexei Protopopov, Liang Chen, Aram F.Hezel, Carla F. Kim, Roderick T. Bronson, Diego H. Castrillon, Lynda Chin, Nabeel Bardeesy, Ronald A. DePinho, and Kwok-Kin Wong
- Subjects
- *
TELOMERES , *GENOMES , *LABORATORY mice , *LUNG cancer - Abstract
Current mouse models of lung cancer recapitulate signature genetic lesions and some phenotypic features of human lung cancer. However, because mice have long telomeres, models to date do not recapitulate the aspects of lung carcinogenesis—telomere attrition and the genomic instability that ensues—believed to serve as key mechanisms driving lung tumor initiation and progression. To explore the contributions of telomere dysfunction to lung cancer progression, we combined a telomerase catalytic subunit (mTerc) mutation with the well-characterized K-rasG12D mouse lung cancer model. K-rasG12D mTerc−/− mice with telomere dysfunction but intact p53 exhibited increased lung epithelial apoptosis, delayed tumor formation and increased life span relative to K-rasG12D mTerc+/− mice with intact telomere function. This demonstrates that by itself, telomere dysfunction acts in a tumor-suppressive mechanism. Introduction of a heterozygous p53 mutation exerted a marked histopathological, biological and genomic impact. K-rasG12D mTerc−/− p53+/− mice developed aggressive tumors with more chromosomal instabilities and high metastatic potential, leading to decreased overall survival. Thus, we have generated a murine model that more faithfully recapitulates key aspects of the human disease. Furthermore, these findings clearly demonstrate (in an in vivo model system) the dual nature of telomere shortening as both a tumor-suppressive and tumor-promoting mechanism in lung cancer, dependent on p53 status. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.