41 results on '"Magdalena J. Glogowska"'
Search Results
2. Supplementary Table 4 from p53 Family Members Regulate Phenotypic Response to Aurora Kinase A Inhibition in Triple-Negative Breast Cancer
- Author
-
Jennifer R. Diamond, S. Gail Eckhardt, Joaquin M. Espinosa, Kelly D. Sullivan, Carol A. Sartorius, Peter Kabos, Magdalena J. Glogowska, Todd M. Pitts, Timothy P. Newton, Aik Choon Tan, Anastasia A. Ionkina, and John J. Tentler
- Abstract
Supplementary Table 4: Combination Index (CI) Values. CI values calculated using the Chou and Talalay method from SRB proliferation data for varying concentrations of Nutlin and MLN8237 in combination. CI < 1 indicates synergy; CI = 1 additivity; CI > 1 antagonism
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 2 from p53 Family Members Regulate Phenotypic Response to Aurora Kinase A Inhibition in Triple-Negative Breast Cancer
- Author
-
Jennifer R. Diamond, S. Gail Eckhardt, Joaquin M. Espinosa, Kelly D. Sullivan, Carol A. Sartorius, Peter Kabos, Magdalena J. Glogowska, Todd M. Pitts, Timothy P. Newton, Aik Choon Tan, Anastasia A. Ionkina, and John J. Tentler
- Abstract
Supplementary Figure 2. Quantification of total cell counts and average cellular size at Day 15 as depicted in Fig. 4A.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure Legend from The Dual Pathway Inhibitor Rigosertib Is Effective in Direct Patient Tumor Xenografts of Head and Neck Squamous Cell Carcinomas
- Author
-
Antonio Jimeno, Aik-Choon Tan, Marileila Varella-Garcia, Barbara A. Frederick, Francois Wilhelm, Dara L. Aisner, Sarah M. Takimoto, Daniel Sehrt, Brian W. Vogler, Gregory N. Gan, Justin Eagles-Soukup, Adrian Umpierrez, Phuong Le, J. Jason Morton, David P. Astling, Magdalena J. Glogowska, Daniel W. Bowles, Stephen B. Keysar, and Ryan T. Anderson
- Abstract
PDF file - 64K
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Table 3 from p53 Family Members Regulate Phenotypic Response to Aurora Kinase A Inhibition in Triple-Negative Breast Cancer
- Author
-
Jennifer R. Diamond, S. Gail Eckhardt, Joaquin M. Espinosa, Kelly D. Sullivan, Carol A. Sartorius, Peter Kabos, Magdalena J. Glogowska, Todd M. Pitts, Timothy P. Newton, Aik Choon Tan, Anastasia A. Ionkina, and John J. Tentler
- Abstract
Supplementary Table 3: Quantification of total cell counts and average cellular size under conditions depicted in Fig. 4A.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 4 from p53 Family Members Regulate Phenotypic Response to Aurora Kinase A Inhibition in Triple-Negative Breast Cancer
- Author
-
Jennifer R. Diamond, S. Gail Eckhardt, Joaquin M. Espinosa, Kelly D. Sullivan, Carol A. Sartorius, Peter Kabos, Magdalena J. Glogowska, Todd M. Pitts, Timothy P. Newton, Aik Choon Tan, Anastasia A. Ionkina, and John J. Tentler
- Abstract
Supplementary Figure 4. Effect of MLN8237 on Cleaved Caspase-3 in TNBC xenografts in vivo.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 1 from p53 Family Members Regulate Phenotypic Response to Aurora Kinase A Inhibition in Triple-Negative Breast Cancer
- Author
-
Jennifer R. Diamond, S. Gail Eckhardt, Joaquin M. Espinosa, Kelly D. Sullivan, Carol A. Sartorius, Peter Kabos, Magdalena J. Glogowska, Todd M. Pitts, Timothy P. Newton, Aik Choon Tan, Anastasia A. Ionkina, and John J. Tentler
- Abstract
Supplementary Figure 1. Treatment with MLN8237 induces markers of cellular senescence in two additional CAL-51 p53 and p73 shRNA knockdown clones (p53-12, p73-26).
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table 2 from p53 Family Members Regulate Phenotypic Response to Aurora Kinase A Inhibition in Triple-Negative Breast Cancer
- Author
-
Jennifer R. Diamond, S. Gail Eckhardt, Joaquin M. Espinosa, Kelly D. Sullivan, Carol A. Sartorius, Peter Kabos, Magdalena J. Glogowska, Todd M. Pitts, Timothy P. Newton, Aik Choon Tan, Anastasia A. Ionkina, and John J. Tentler
- Abstract
Supplementary Table 2: Sensitivity of CAL-51 KD clones to MLN8237 in vitro.
- Published
- 2023
- Full Text
- View/download PDF
9. Data from The Dual Pathway Inhibitor Rigosertib Is Effective in Direct Patient Tumor Xenografts of Head and Neck Squamous Cell Carcinomas
- Author
-
Antonio Jimeno, Aik-Choon Tan, Marileila Varella-Garcia, Barbara A. Frederick, Francois Wilhelm, Dara L. Aisner, Sarah M. Takimoto, Daniel Sehrt, Brian W. Vogler, Gregory N. Gan, Justin Eagles-Soukup, Adrian Umpierrez, Phuong Le, J. Jason Morton, David P. Astling, Magdalena J. Glogowska, Daniel W. Bowles, Stephen B. Keysar, and Ryan T. Anderson
- Abstract
The dual pathway inhibitor rigosertib inhibits phosphoinositide 3-kinase (PI3K) pathway activation as well as polo-like kinase 1 (PLK1) activity across a broad spectrum of cancer cell lines. The importance of PIK3CA alterations in squamous cell carcinoma of the head and neck (HNSCC) has raised interest in exploring agents targeting PI3K, the product of PIK3CA. The genetic and molecular basis of rigosertib treatment response was investigated in a panel of 16 HNSCC cell lines, and direct patient tumor xenografts from eight patients with HNSCC [four HPV-serotype16 (HPV16)–positive]. HNSCC cell lines and xenografts were characterized by pathway enrichment gene expression analysis, exon sequencing, gene copy number, Western blotting, and immunohistochemistry (IHC). Rigosertib had potent antiproliferative effects on 11 of 16 HPV− HNSCC cell lines. Treatment sensitivity was confirmed in two cell lines using an orthotopic in vivo xenograft model. Growth reduction after rigosertib treatment was observed in three of eight HNSCC direct patient tumor lines. The responsive tumor lines carried a combination of a PI3KCA-activating event (amplification or mutation) and a p53-inactivating event (either HPV16- or mutation-mediated TP53 inactivation). In this study, we evaluated the in vitro and in vivo efficacy of rigosertib in both HPV+ and HPV− HNSCCs, focusing on inhibition of the PI3K pathway. Although consistent inhibition of the PI3K pathway was not evident in HNSCC, we identified a combination of PI3K/TP53 events necessary, but not sufficient, for rigosertib sensitivity. Mol Cancer Ther; 12(10); 1994–2005. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Tables 1-2 from EGFR Mediates Responses to Small-Molecule Drugs Targeting Oncogenic Fusion Kinases
- Author
-
Robert C. Doebele, Dara L. Aisner, Lynn E. Heasley, Antonio Jimeno, Eric B. Haura, Marileila Varella-Garcia, Kathryn E. Ware, Kurtis D. Davies, Natalia J. Sumi, Severine Kako, Matthew A. Smith, Magdalena J. Glogowska, Stephen B. Keysar, Anh T. Le, Lindsay A. Marek, Uwe Rix, Laura Schubert, and Aria Vaishnavi
- Abstract
Table 1: Cell lines, fusions, tyrosine kinase inhibitors and targets. Table 2: IC50 values for Figure 1C and relative fold change.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from EGFR Mediates Responses to Small-Molecule Drugs Targeting Oncogenic Fusion Kinases
- Author
-
Robert C. Doebele, Dara L. Aisner, Lynn E. Heasley, Antonio Jimeno, Eric B. Haura, Marileila Varella-Garcia, Kathryn E. Ware, Kurtis D. Davies, Natalia J. Sumi, Severine Kako, Matthew A. Smith, Magdalena J. Glogowska, Stephen B. Keysar, Anh T. Le, Lindsay A. Marek, Uwe Rix, Laura Schubert, and Aria Vaishnavi
- Abstract
Oncogenic kinase fusions of ALK, ROS1, RET, and NTRK1 act as drivers in human lung and other cancers. Residual tumor burden following treatment of ALK or ROS1+ lung cancer patients with oncogene-targeted therapy ultimately enables the emergence of drug-resistant clones, limiting the long-term effectiveness of these therapies. To determine the signaling mechanisms underlying incomplete tumor cell killing in oncogene-addicted cancer cells, we investigated the role of EGFR signaling in drug-naïve cancer cells harboring these oncogene fusions. We defined three distinct roles for EGFR in the response to oncogene-specific therapies. First, EGF-mediated activation of EGFR blunted fusion kinase inhibitor binding and restored fusion kinase signaling complexes. Second, fusion kinase inhibition shifted adaptor protein binding from the fusion oncoprotein to EGFR. Third, EGFR enabled bypass signaling to critical downstream pathways such as MAPK. While evidence of EGFR-mediated bypass signaling has been reported after ALK and ROS1 blockade, our results extended this effect to RET and NTRK1 blockade and uncovered the other additional mechanisms in gene fusion–positive lung cancer cells, mouse models, and human clinical specimens before the onset of acquired drug resistance. Collectively, our findings show how EGFR signaling can provide a critical adaptive survival mechanism that allows cancer cells to evade oncogene-specific inhibitors, providing a rationale to cotarget EGFR to reduce the risks of developing drug resistance. Cancer Res; 77(13); 3551–63. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
12. Figures S1-9 and Legends from EGFR Mediates Responses to Small-Molecule Drugs Targeting Oncogenic Fusion Kinases
- Author
-
Robert C. Doebele, Dara L. Aisner, Lynn E. Heasley, Antonio Jimeno, Eric B. Haura, Marileila Varella-Garcia, Kathryn E. Ware, Kurtis D. Davies, Natalia J. Sumi, Severine Kako, Matthew A. Smith, Magdalena J. Glogowska, Stephen B. Keysar, Anh T. Le, Lindsay A. Marek, Uwe Rix, Laura Schubert, and Aria Vaishnavi
- Abstract
Fig. S1: EGFR inhibition or stimulation affects normal and catalytically inactive fusion oncogene signaling. Fig. S2: EGFR knockdown reduces fusion kinase cancer cell proliferation. Fig. S3: Conserved re-phosphorylated activation loop tyrosines enabled signaling PLA design and representative image analyses of fusion kinase and EGFR signaling PLAs. Fig. S4: Optimization of the EGFR-GRB2 PLA and fusion kinase PLA antibody controls. Fig. S5: The GRB2 adaptor does not switch from ROS1 to MET with FKI treatment, but MET can signal through GRB2. FIg. S6: GRB2 and SHC1 signaling rewire in a RET+ cell line treated with a fusion kinase inhibitor (FKI). Fig. S7: TRK, RET, ROS1 and ALK fusion kinases interact specifically with EGFR. Fig. S8: EGFR is highly expressed, phosphorylated, and signaling in vivo in an NTRK1+ patient-derived xenograft (PDX) model. Fig. S9: Fusion kinase, and EGFR signaling complexes are present in RET, and ALK, fusion resistant patient samples.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figures 1 - 9 from Hedgehog Signaling Alters Reliance on EGF Receptor Signaling and Mediates Anti-EGFR Therapeutic Resistance in Head and Neck Cancer
- Author
-
Antonio Jimeno, Xiao-Jing Wang, John I. Song, Andrew Thorburn, M. Scott Lucia, Fred R. Hirsch, Hilary Serracino, Justin R. Eagles-Soukup, Gregory N. Gan, Daniel B. Sehrt, Sarah M. Takimoto, Magdalena J. Glogowska, Pamela Fernandez, Jackie Thorburn, Brian W. Vogler, Jeramiah J. Paylor, Daniel W. Bowles, J. Jason Morton, Ryan T. Anderson, Phuong N. Le, and Stephen B. Keysar
- Abstract
PDF file - 1255K, Confirmation of GLI1 modulation by EGFR activation (S1); Epithelial gene expression profile and morphology predicts erlotinib sensitivity in HNSCC cell lines (S2); Zeb1 transcription factor is key driver of EGF induced EMT (S3); EGFR activation induces EMT in HNSCC cell lines characterized by expression of Vimentin (S4); Inter-pathway crosstalk as well as EGF induced EMT are blunted in erlotinib resistant cells (S5); Silencing of HhP transcription factor GLI1 increases EGFR driven EMT in Tu-167 cells (S6); MEK/ERK signaling is required for EMT-like state while both MEK/ERK and PI3K/AKT are required for invasion through Matrigel (S7); Proliferative inhibition of erltotinib (1μM and 2μM) plus IPI-926 (1μM) or SHH (250ng/ml) (S8); Cetuximab and IPI-926 modulate EGFR, HhP and EMT gene expression in vivo (S9).
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure Legend from Hedgehog Signaling Alters Reliance on EGF Receptor Signaling and Mediates Anti-EGFR Therapeutic Resistance in Head and Neck Cancer
- Author
-
Antonio Jimeno, Xiao-Jing Wang, John I. Song, Andrew Thorburn, M. Scott Lucia, Fred R. Hirsch, Hilary Serracino, Justin R. Eagles-Soukup, Gregory N. Gan, Daniel B. Sehrt, Sarah M. Takimoto, Magdalena J. Glogowska, Pamela Fernandez, Jackie Thorburn, Brian W. Vogler, Jeramiah J. Paylor, Daniel W. Bowles, J. Jason Morton, Ryan T. Anderson, Phuong N. Le, and Stephen B. Keysar
- Abstract
PDF file - 113K
- Published
- 2023
- Full Text
- View/download PDF
15. Supplemental Methods from Hedgehog Signaling Drives Radioresistance and Stroma-Driven Tumor Repopulation in Head and Neck Squamous Cancers
- Author
-
Antonio Jimeno, Xiao-Jing Wang, David Raben, Barbara Frederick, J. Jason Morton, Phuong N. Le, Ryan T. Anderson, Cem Altunbas, Magdalena J. Glogowska, Guoliang Wang, Stephen B. Keysar, Justin Eagles, and Gregory N. Gan
- Abstract
Supplemental Methods. Description of additional methods and procedures used in the study.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Tables 1 - 2 from Hedgehog Signaling Alters Reliance on EGF Receptor Signaling and Mediates Anti-EGFR Therapeutic Resistance in Head and Neck Cancer
- Author
-
Antonio Jimeno, Xiao-Jing Wang, John I. Song, Andrew Thorburn, M. Scott Lucia, Fred R. Hirsch, Hilary Serracino, Justin R. Eagles-Soukup, Gregory N. Gan, Daniel B. Sehrt, Sarah M. Takimoto, Magdalena J. Glogowska, Pamela Fernandez, Jackie Thorburn, Brian W. Vogler, Jeramiah J. Paylor, Daniel W. Bowles, J. Jason Morton, Ryan T. Anderson, Phuong N. Le, and Stephen B. Keysar
- Abstract
PDF file - 29K, EGFR dependent EMT quantified as percent Vimentin-positive cells in culture (S1); Combination therapy delays or blocks tumor re-growth in sensitive xenografts (S2).
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental Tables S1-S2 from Hedgehog Signaling Drives Radioresistance and Stroma-Driven Tumor Repopulation in Head and Neck Squamous Cancers
- Author
-
Antonio Jimeno, Xiao-Jing Wang, David Raben, Barbara Frederick, J. Jason Morton, Phuong N. Le, Ryan T. Anderson, Cem Altunbas, Magdalena J. Glogowska, Guoliang Wang, Stephen B. Keysar, Justin Eagles, and Gregory N. Gan
- Abstract
Supplemental Tables S1-S2. Hirsch Score was calculated from tumors and their respective stroma from Figure 5 (S1); All potential flank implanted stromal:tumor and tumor:tumor combinations were measured bi-weekly to chart growth kinetics (S2).
- Published
- 2023
- Full Text
- View/download PDF
18. Supplemental Figures S1-S6 from Hedgehog Signaling Drives Radioresistance and Stroma-Driven Tumor Repopulation in Head and Neck Squamous Cancers
- Author
-
Antonio Jimeno, Xiao-Jing Wang, David Raben, Barbara Frederick, J. Jason Morton, Phuong N. Le, Ryan T. Anderson, Cem Altunbas, Magdalena J. Glogowska, Guoliang Wang, Stephen B. Keysar, Justin Eagles, and Gregory N. Gan
- Abstract
Supplemental Figures S1-S6. Comparison of HN11 and TU167 by RNA-Seq Expression (S1); Suppression of Radiation-induced GLI1 using shRNA Model (S2); Cyst Formation Following Radiation Treatment in Orthotopic TU167-implanted Tumors (S3); Radiation-induced Gli1 Expression and the Effect of Rapamycin Inhibition in HN11 Cells (S4); Effect of siRNA inhibition on Head Neck Cancer Cells and Gli1 Nuclear Translocation (S5); Schema and Histology Detailing Radiation-induced Tumor Repopulation (S6).
- Published
- 2023
- Full Text
- View/download PDF
19. Supplemental Table and Figure Legends from Hedgehog Signaling Drives Radioresistance and Stroma-Driven Tumor Repopulation in Head and Neck Squamous Cancers
- Author
-
Antonio Jimeno, Xiao-Jing Wang, David Raben, Barbara Frederick, J. Jason Morton, Phuong N. Le, Ryan T. Anderson, Cem Altunbas, Magdalena J. Glogowska, Guoliang Wang, Stephen B. Keysar, Justin Eagles, and Gregory N. Gan
- Abstract
Supplemental Table and Figure Legends. Legends for Supplemental Figures S1-S6 and Supplemental Tables S1-S2.
- Published
- 2023
- Full Text
- View/download PDF
20. EGFR Mediates Responses to Small-Molecule Drugs Targeting Oncogenic Fusion Kinases
- Author
-
Kathryn E. Ware, Magdalena J. Glogowska, Uwe Rix, Severine Kako, Eric B. Haura, Natalia J. Sumi, Robert C. Doebele, Matthew A. Smith, Lindsay Marek, Kurtis D. Davies, Marileila Varella-Garcia, Lynn E. Heasley, Laura Schubert, Dara L. Aisner, Aria Vaishnavi, Antonio Jimeno, Stephen B. Keysar, and Anh T. Le
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Oncogene Proteins, Fusion ,Mice, Nude ,Biology ,Bioinformatics ,Article ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,ROS1 ,Animals ,Humans ,Lung cancer ,Cell Proliferation ,Kinase ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,ErbB Receptors ,HEK293 Cells ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Cyclin-dependent kinase 8 ,Oncogene Fusion ,Signal transduction ,Signal Transduction - Abstract
Oncogenic kinase fusions of ALK, ROS1, RET, and NTRK1 act as drivers in human lung and other cancers. Residual tumor burden following treatment of ALK or ROS1+ lung cancer patients with oncogene-targeted therapy ultimately enables the emergence of drug-resistant clones, limiting the long-term effectiveness of these therapies. To determine the signaling mechanisms underlying incomplete tumor cell killing in oncogene-addicted cancer cells, we investigated the role of EGFR signaling in drug-naïve cancer cells harboring these oncogene fusions. We defined three distinct roles for EGFR in the response to oncogene-specific therapies. First, EGF-mediated activation of EGFR blunted fusion kinase inhibitor binding and restored fusion kinase signaling complexes. Second, fusion kinase inhibition shifted adaptor protein binding from the fusion oncoprotein to EGFR. Third, EGFR enabled bypass signaling to critical downstream pathways such as MAPK. While evidence of EGFR-mediated bypass signaling has been reported after ALK and ROS1 blockade, our results extended this effect to RET and NTRK1 blockade and uncovered the other additional mechanisms in gene fusion–positive lung cancer cells, mouse models, and human clinical specimens before the onset of acquired drug resistance. Collectively, our findings show how EGFR signaling can provide a critical adaptive survival mechanism that allows cancer cells to evade oncogene-specific inhibitors, providing a rationale to cotarget EGFR to reduce the risks of developing drug resistance. Cancer Res; 77(13); 3551–63. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
21. p53 Family Members Regulate Phenotypic Response to Aurora Kinase A Inhibition in Triple-Negative Breast Cancer
- Author
-
Todd M. Pitts, S. Gail Eckhardt, Peter Kabos, John J. Tentler, Kelly D. Sullivan, Carol A. Sartorius, Jennifer R. Diamond, Magdalena J. Glogowska, Joaquín M. Espinosa, Timothy P. Newton, Anastasia A. Ionkina, and Aik Choon Tan
- Subjects
Senescence ,Cancer Research ,Regulator ,Apoptosis ,Triple Negative Breast Neoplasms ,Biology ,Article ,Mice ,chemistry.chemical_compound ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Tumor Protein p73 ,Protein Kinase Inhibitors ,Triple-negative breast cancer ,Aurora Kinase A ,Cell Proliferation ,Tumor Suppressor Proteins ,Nuclear Proteins ,Azepines ,medicine.disease ,Xenograft Model Antitumor Assays ,DNA-Binding Proteins ,Pyrimidines ,Oncology ,chemistry ,Alisertib ,Immunology ,Cancer research ,Female ,Tumor Suppressor Protein p53 - Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease with a poor prognosis. Advances in the treatment of TNBC have been hampered by the lack of novel effective targeted therapies. The primary goal of this study was to evaluate the efficacy of targeting Aurora kinase A (AurA), a key regulator of mitosis, in TNBC models. A secondary objective was to determine the role of the p53 family of transcriptional regulators, commonly mutated in TNBC, in determining the phenotypic response to the AurA inhibitor alisertib (MLN8237). Alisertib exhibited potent antiproliferative and proapoptotic activity in a subset of TNBC models. The induction of apoptosis in response to alisertib exposure was dependent on p53 and p73 activity. In the absence of functional p53 or p73, there was a shift in the phenotypic response following alisertib exposure from apoptosis to cellular senescence. In addition, senescence was observed in patient-derived tumor xenografts with acquired resistance to alisertib treatment. AurA inhibitors are a promising class of novel therapeutics in TNBC. The role of p53 and p73 in mediating the phenotypic response to antimitotic agents in TNBC may be harnessed to develop an effective biomarker selection strategy in this difficult to target disease. Mol Cancer Ther; 14(5); 1117–29. ©2015 AACR.
- Published
- 2015
- Full Text
- View/download PDF
22. XactMice: humanizing mouse bone marrow enables microenvironment reconstitution in a patient-derived xenograft model of head and neck cancer
- Author
-
Antonio Jimeno, Xiao-Jing Wang, Justin R. Eagles, Nuria Padilla-Just, Patricia A. Estes, Yosef Refaeli, Brett McGettigan, John Morton, Ryan T. Anderson, Stephen B. Keysar, Phuong Le, Gregory H. Bird, Daniel W. Bowles, Dennis R. Roop, Gregory N. Gan, Magdalena J. Glogowska, Aik Choon Tan, Pamela Fernandez, Traci R. Lyons, David P. Astling, Marileila Varella-Garcia, John I. Song, and Pepper Schedin
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,Humanized mouse model ,xenograft model ,Nod ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Bone Marrow ,Cell Line, Tumor ,Tumor Microenvironment ,Genetics ,medicine ,Animals ,Humans ,Progenitor cell ,Molecular Biology ,Tumor microenvironment ,Hematopoietic Stem Cells ,Xenograft Model Antitumor Assays ,3. Good health ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Head and Neck Neoplasms ,Cell culture ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Cytokines ,head and neck cancer ,Bone marrow - Abstract
The limitations of cancer cell lines have led to the development of direct patient derived xenograft (PDX) models. However, the interplay between the implanted human cancer cells and recruited mouse stromal and immune cells alters the tumor microenvironment and limits the value of these models. To overcome these constraints, we have developed a technique to expand human hematopoietic stem and progenitor cells (HSPCs) and use them to reconstitute the radiation-depleted bone marrow of a NOD/SCID/IL2rg−/− (NSG) mouse on which a patient’s tumor is then transplanted (XactMice). The human HSPCs produce immune cells that home into the tumor and help replicate its natural microenvironment. Despite previous passage on nude mice, the expression of epithelial, stromal, and immune genes in XactMice tumors aligns more closely to that of the patient tumor than to those grown in non-humanized mice – an effect partially facilitated by human cytokines expressed by both the HSPC progeny and the tumor cells. The human immune and stromal cells produced in the XactMice can help recapitulate the microenvironment of an implanted xenograft, reverse the initial genetic drift seen after passage on non-humanized mice, and provide a more accurate tumor model to guide patient treatment.
- Published
- 2015
- Full Text
- View/download PDF
23. Hedgehog Signaling Alters Reliance on EGF Receptor Signaling and Mediates Anti-EGFR Therapeutic Resistance in Head and Neck Cancer
- Author
-
John I. Song, Daniel Sehrt, Sarah M. Takimoto, John Morton, Hilary S. Serracino, Fred R. Hirsch, Xiao-Jing Wang, Antonio Jimeno, Brian W. Vogler, Justin R. Eagles-Soukup, Phuong Le, Gregory N. Gan, Pamela Fernandez, Stephen B. Keysar, Andrew Thorburn, Ryan T. Anderson, Jackie Thorburn, Daniel W. Bowles, Magdalena J. Glogowska, Jeramiah J. Paylor, and M. Scott Lucia
- Subjects
Cancer Research ,medicine.medical_treatment ,Population ,Cetuximab ,Mice, Nude ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Article ,Targeted therapy ,Mice ,GLI1 ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Gene Silencing ,education ,neoplasms ,EGFR inhibitors ,education.field_of_study ,biology ,Squamous Cell Carcinoma of Head and Neck ,Head and neck cancer ,Veratrum Alkaloids ,Receptor Cross-Talk ,medicine.disease ,Xenograft Model Antitumor Assays ,Hedgehog signaling pathway ,ErbB Receptors ,Oncology ,Drug Resistance, Neoplasm ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,biology.protein ,Cancer research ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
The EGF receptor (EGFR)-directed monoclonal antibody cetuximab is the only targeted therapy approved for the treatment of squamous cell carcinoma of the head and neck (HNSCC) but is only effective in a minority of patients. Epithelial-to-mesenchymal transition (EMT) has been implicated as a drug resistance mechanism in multiple cancers, and the EGFR and Hedgehog pathways (HhP) are relevant to this process, but the interplay between the two pathways has not been defined in HNSCC. Here, we show that HNSCC cells that were naturally sensitive to EGFR inhibition over time developed increased expression of the HhP transcription factor GLI1 as they became resistant after long-term EGFR inhibitor exposure. This robustly correlated with an increase in vimentin expression. Conversely, the HhP negatively regulated an EGFR-dependent, EMT-like state in HNSCC cells, and pharmacologic or genetic inhibition of HhP signaling pushed cells further into an EGFR-dependent phenotype, increasing expression of ZEB1 and VIM. In vivo treatment with cetuximab resulted in tumor shrinkage in four of six HNSCC patient-derived xenografts; however, they eventually regrew. Cetuximab in combination with the HhP inhibitor IPI-926 eliminated tumors in two cases and significantly delayed regrowth in the other two cases. Expression of EMT genes TWIST and ZEB2 was increased in sensitive xenografts, suggesting a possible resistant mesenchymal population. In summary, we report that EGFR-dependent HNSCC cells can undergo both EGFR-dependent and -independent EMT and HhP signaling is a regulator in both processes. Cetuximab plus IPI-926 forces tumor cells into an EGFR-dependent state, delaying or completely blocking tumor recurrence. Cancer Res; 73(11); 3381–92. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
24. A patient tumor transplant model of squamous cell cancer identifies PI3K inhibitors as candidate therapeutics in defined molecular bins
- Author
-
Diana F. Hausman, Ted H. Leem, Jeramiah J. Paylor, Aik Choon Tan, Justin R. Eagles-Soukup, Ryan T. Anderson, William A. Robinson, Morgan A. Pittman, Wells A. Messersmith, Marileila Varella-Garcia, David Raben, David P. Astling, Antonio Jimeno, Phuong Le, John Morton, Ryan M. Helber, Sherif Said, Daniel W. Bowles, Fred R. Hirsch, Magdalena J. Glogowska, Julie A. Goddard, Daniel Sehrt, Sarah M. Takimoto, John J. Arcaroli, Stephen B. Keysar, Scott Peterson, Sarah M. Macfadden, Xiao-Jing Wang, Brian W. Vogler, Adrian Umpierrez, John I. Song, and Severine Kako
- Subjects
Cancer Research ,medicine.medical_treatment ,Blotting, Western ,Cetuximab ,Gonanes ,Alphapapillomavirus ,Biology ,Antibodies, Monoclonal, Humanized ,Targeted therapy ,Efficacy ,Mice ,Phosphatidylinositol 3-Kinases ,Genetics ,medicine ,Animals ,Humans ,Receptor, Notch2 ,Receptor, Notch1 ,neoplasms ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,EGFR inhibitors ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,Head and neck cancer ,Computational Biology ,General Medicine ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Head and neck squamous-cell carcinoma ,ErbB Receptors ,stomatognathic diseases ,Oncology ,Head and Neck Neoplasms ,Papers ,Immunology ,Carcinoma, Squamous Cell ,Cancer research ,Molecular Medicine ,Personalized medicine ,business ,medicine.drug - Abstract
Targeted therapy development in head and neck squamous cell carcinoma (HNSCC) is challenging given the rarity of activating mutations. Additionally, HNSCC incidence is increasing related to human papillomavirus (HPV). We sought to develop an in vivo model derived from patients reflecting the evolving HNSCC epidemiologic landscape, and use it to identify new therapies. Primary and relapsed tumors from HNSCC patients, both HPV+ and HPV-, were implanted on mice, giving rise to 25 strains. Resulting xenografts were characterized by detecting key mutations, measuring protein expression by IHC and gene expression/pathway analysis by mRNA-sequencing. Drug efficacy studies were run with representative xenografts using the approved drug cetuximab as well as the new PI3K inhibitor PX-866. Tumors maintained their original morphology, genetic profiles and drug susceptibilities through serial passaging. The genetic makeup of these tumors was consistent with known frequencies of TP53, PI3KCA, NOTCH1 and NOTCH2 mutations. Because the EGFR inhibitor cetuximab is a standard HNSCC therapy, we tested its efficacy and observed a wide spectrum of efficacy. Cetuximab-resistant strains had higher PI3K/Akt pathway gene expression and protein activation than cetuximab-sensitive strains. The PI3K inhibitor PX-866 had anti-tumor efficacy in HNSCC models with PIK3CA alterations. Finally, PI3K inhibition was effective in two cases with NOTCH1 inactivating mutations. In summary, we have developed an HNSCC model covering its clinical spectrum whose major genetic alterations and susceptibility to anticancer agents represent contemporary HNSCC. This model enables to prospectively test therapeutic-oriented hypotheses leading to personalized medicine.
- Published
- 2013
- Full Text
- View/download PDF
25. Regulation of Head and Neck Squamous Cancer Stem Cells by PI3K and SOX2
- Author
-
Dennis R. Roop, Justin R. Eagles, Julie Reisinger, Stephen B. Keysar, Aik Choon Tan, Jihye Kim, Nuria Padilla-Just, Phuong Le, Emily Warnock, Brian C. Jackson, Binwu Tang, Bettina Miller, Antonio Jimeno, Vasilis Vasiliou, Wells A. Messersmith, Cera Nieto, John J. Arcaroli, Hilary S. Serracino, Xiao-Jing Wang, Karina A. Gómez, Magdalena J. Glogowska, John Morton, Dexiang Gao, and Lalage M. Wakefield
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,Mice ,Tumor Cells, Cultured ,Papillomaviridae ,Phosphoinositide-3 Kinase Inhibitors ,education.field_of_study ,TOR Serine-Threonine Kinases ,virus diseases ,Cadherins ,ErbB Receptors ,Cell Transformation, Neoplastic ,Hyaluronan Receptors ,Oncology ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,Neoplastic Stem Cells ,Female ,Stem cell ,Cell Division ,Signal Transduction ,medicine.medical_specialty ,Population ,Mice, Nude ,Antineoplastic Agents ,Biology ,Aldehyde Dehydrogenase 1 Family ,Article ,03 medical and health sciences ,SOX2 ,Cancer stem cell ,Antigens, CD ,Spheroids, Cellular ,mental disorders ,medicine ,Animals ,Humans ,RNA, Messenger ,education ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sequence Analysis, RNA ,SOXB1 Transcription Factors ,CD44 ,Retinal Dehydrogenase ,Aldehyde Dehydrogenase ,medicine.disease ,Head and neck squamous-cell carcinoma ,stomatognathic diseases ,030104 developmental biology ,MRNA Sequencing ,Cancer research ,biology.protein ,Phosphatidylinositol 3-Kinase ,Transcriptome ,Neoplasm Transplantation - Abstract
Background We have an incomplete understanding of the differences between cancer stem cells (CSCs) in human papillomavirus-positive (HPV-positive) and -negative (HPV-negative) head and neck squamous cell cancer (HNSCC). The PI3K pathway has the most frequent activating genetic events in HNSCC (especially HPV-positive driven), but the differential signaling between CSCs and non-CSCs is also unknown. Methods We addressed these unresolved questions using CSCs identified from 10 HNSCC patient-derived xenografts (PDXs). Sored populations were serially passaged in nude mice to evaluate tumorigenicity and tumor recapitulation. The transcription profile of HNSCC CSCs was characterized by mRNA sequencing, and the susceptibility of CSCs to therapy was investigated using an in vivo model. SOX2 transcriptional activity was used to follow the asymmetric division of PDX-derived CSCs. All statistical tests were two-sided. Results CSCs were enriched by high aldehyde dehydrogenase (ALDH) activity and CD44 expression and were similar between HPV-positive and HPV-negative cases (percent tumor formation injecting ≤ 1x10(3) cells: ALDH(+)CD44(high) = 65.8%, ALDH(-)CD44(high) = 33.1%, ALDH(+)CD44(high) = 20.0%; and injecting 1x10(5) cells: ALDH(-)CD44(low) = 4.4%). CSCs were resistant to conventional therapy and had PI3K/mTOR pathway overexpression (GSEA pathway enrichment, P < .001), and PI3K inhibition in vivo decreased their tumorigenicity (40.0%-100.0% across cases). PI3K/mTOR directly regulated SOX2 protein levels, and SOX2 in turn activated ALDH1A1 (P < .001 013C and 067C) expression and ALDH activity (ALDH(+) [%] empty-control vs SOX2, 0.4% ± 0.4% vs 14.5% ± 9.8%, P = .03 for 013C and 1.7% ± 1.3% vs 3.6% ± 3.4%, P = .04 for 067C) in 013C and 067 cells. SOX2 enhanced sphere and tumor growth (spheres/well, 013C P < .001 and 067C P = .04) and therapy resistance. SOX2 expression prompted mesenchymal-to-epithelial transition (MET) by inducing CDH1 (013C P = .002, 067C P = .01), followed by asymmetric division and proliferation, which contributed to tumor formation. Conclusions The molecular link between PI3K activation and CSC properties found in this study provides insights into therapeutic strategies for HNSCC. Constitutive expression of SOX2 in HNSCC cells generates a CSC-like population that enables CSC studies.
- Published
- 2016
26. Mechanisms of mannose-binding lectin-associated serine proteases-1/3 activation of the alternative pathway of complement
- Author
-
Yuichi Endo, William P. Arend, Teizo Fujita, Nirmal K. Banda, Magdalena J. Glogowska, V. Michael Holers, Minoru Takahashi, Gregory L. Stahl, Stephanie Hyatt, Timothy A. Wiles, and Kazue Takahashi
- Subjects
Male ,Proteases ,medicine.drug_class ,Blotting, Western ,Complement Pathway, Alternative ,Immunology ,chemical and pharmacologic phenomena ,Inflammation ,Biology ,Monoclonal antibody ,Article ,Serine ,Mice ,Complement Factor D ,medicine ,Animals ,Molecular Biology ,Mannan-binding lectin ,Mice, Knockout ,bacterial infections and mycoses ,Arthritis, Experimental ,Molecular biology ,In vitro ,Mice, Inbred C57BL ,Biochemistry ,Mannose-Binding Protein-Associated Serine Proteases ,Alternative complement pathway ,medicine.symptom - Abstract
Mannose-binding lectin-associated serine proteases-1/3 (MASP-1/3) are essential in activating the alternative pathway (AP) of complement through cleaving pro-factor D (pro-Df) into mature Df. MASP are believed to require binding to mannose binding lectins (MBL) or ficolins (FCN) to carry out their biological activities. Murine sera have been reported to contain MBL-A, MBL-C, and FCN-A, but not FCN-B that exists endogenously in monocytes and is thought not to bind MASP-1. We examined some possible mechanisms whereby MASP-1/3 might activate the AP. Collagen antibody-induced arthritis, a murine model of inflammatory arthritis dependent on the AP, was unchanged in mice lacking MBL-A, MBL-C, and FCN-A (MBL(-/-)/FCN A(-/-) mice) in comparison to wild-type mice. The in vitro induction of the AP by adherent mAb to collagen II was intact using sera from MBL(-/-)/FCN A(-/-) mice. Furthermore, sera from MBL(-/-)/FCN A(-/-) mice lacked pro-Df and possessed only mature Df. Gel filtration of sera from MBL(-/-)/FCN A(-/-) mice showed the presence of MASP-1 protein in fractions containing proteins smaller than the migration of MBL-A and MBL-C in sera from C4(-/-) mice, suggesting possible binding of MASP-1 to an unknown protein. Lastly, we show that FCN-B was present in the sera of MBL(-/-)/FCN A(-/-) mice and that it was bound to MASP-1. We conclude that MASP-1 does not require binding to MBL-A, MBL-C, or FCN-A to activate the AP. MASP-1 may cleave pro-Df into mature Df through binding to FCN-B or to an unknown protein, or may function as an unbound soluble protein.
- Published
- 2011
- Full Text
- View/download PDF
27. Essential Role of Complement Mannose-Binding Lectin-Associated Serine Proteases-1/3 in the Murine Collagen Antibody-Induced Model of Inflammatory Arthritis
- Author
-
William P. Arend, Magdalena J. Glogowska, V. Michael Holers, Gregory L. Stahl, Nirmal K. Banda, Teizo Fujita, Kazue Takahashi, Jessica Nicholas, Brandt Levitt, and Minoru Takahashi
- Subjects
Male ,Inflammatory arthritis ,Blotting, Western ,Complement Pathway, Alternative ,Immunology ,Arthritis ,Biology ,Polymerase Chain Reaction ,Article ,Mice ,In vivo ,Complement Factor D ,medicine ,Animals ,Humans ,Immunology and Allergy ,Mannan-binding lectin ,Mice, Knockout ,medicine.disease ,Arthritis, Experimental ,Immunohistochemistry ,Molecular biology ,Complement system ,Mice, Inbred C57BL ,Mannose-Binding Protein-Associated Serine Proteases ,Alternative complement pathway ,biology.protein ,Female ,Factor D - Abstract
Gene-targeted mice deficient in the complement mannose-binding lectin-associated serine protease-1 and -3 (MASP1/3−/−) express only the zymogen of factor D (pro-factor D [pro-Df]), a necessary component of the alternative pathway (AP). We used the murine collagen Ab-induced arthritis (CAIA) model, in which the AP is unique among complement pathways in being both necessary and sufficient for disease induction, to determine whether MASP-1/3 are required in vivo for the development of tissue injury. Disease activity scores, complement C3 tissue deposition in the joint, and histopathologic injury scores were markedly decreased in MASP1/3−/− as compared with wild-type (WT) mice. MASP-1 protein was immunochemically localized to synovial cells of knees of WT mice with arthritis. Pro-Df was present in both synovial cells and chondrocytes of knees of WT and MASP1/3−/− mice without arthritis, with increased amounts present in synovial cells of WT mice with CAIA. No conversion of pro-Df to mature Df was detectable in the serum of MASP1/3−/− mice during the evolution of CAIA. C3 activation and deposition as well as C5a generation induced in vitro by adherent anti-type II collagen mAbs were absent using sera from MASP1/3−/− mice under conditions in which only the AP was active. The addition of human Df fully reconstituted in vitro C3 activation and C5a generation using sera from MASP1/3−/− mice. Our studies demonstrate for the first time, to our knowledge, the absolute requirement for the activity of MASP-1 protein in autoimmune-associated inflammatory tissue injury in vivo through activation of the AP of complement by cleavage of pro-Df to mature Df.
- Published
- 2010
- Full Text
- View/download PDF
28. A pilot study of cetuximab and the hedgehog inhibitor IPI-926 in recurrent/metastatic head and neck squamous cell carcinoma
- Author
-
Dennis R. Roop, Charles E. Ray, Phuong Le, Aik Choon Tan, Hilary S. Serracino, Jessica D. McDermott, Antonio Jimeno, Stephen B. Keysar, Magdalena J. Glogowska, Dexiang Gao, Guoliang Wang, Paul J. Rochon, Daniel W. Bowles, and Justin R. Eagles
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,Maximum Tolerated Dose ,Cetuximab ,Gene Expression ,Antineoplastic Agents ,Pilot Projects ,Loading dose ,Disease-Free Survival ,Article ,03 medical and health sciences ,0302 clinical medicine ,ErbB ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Hedgehog Proteins ,Progression-free survival ,Adverse effect ,neoplasms ,business.industry ,Middle Aged ,medicine.disease ,Rash ,Head and neck squamous-cell carcinoma ,030104 developmental biology ,Treatment Outcome ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Female ,Oral Surgery ,medicine.symptom ,Neoplasm Recurrence, Local ,business ,medicine.drug - Abstract
Summary Background This phase 1, dose-finding study determined the safety, maximum tolerated dose (MTD)/recommended phase 2 dose (RP2D), antitumor activity, and molecular correlates of IPI-926, a Hedgehog pathway (HhP) inhibitor, combined with cetuximab in patients with relapsed/metastatic squamous cell carcinoma of the head and neck. Patients and methods Cetuximab was given with a 400 mg/m 2 loading dose followed by 250 mg/m 2 weekly. IPI-926 was given daily starting two weeks after cetuximab initiation. A “3 + 3” study design was used. Prior therapy with cetuximab was allowed. Tumor biopsies occurred prior to cetuximab initiation, prior to IPI-926 initiation, and after treatment with both drugs. Results Nine patients were enrolled. The RP2D was 160 mg, the same as the single-agent IPI-926 MTD. Among 9 treated, 8 evaluable patients, the best responses were 1 partial response (12.5%), 4 stable disease (50%), and 3 disease progressions (37.5%). The median progression free survival was 77 days (95% confidence interval 39–156). Decreases in tumor size were seen in both cetuximab-naive patients (one HPV-positive, one HPV-negative). The most frequent treatment-emergent adverse events were fatigue, muscle cramps, and rash. No DLTs were observed. Tumor shrinkage and progression free survival were associated with intra-tumoral ErbB and HhP gene expression down-regulation during therapy, supporting the preclinical hypothesis. Conclusion Treatment with IPI-926 and cetuximab yielded expected toxicities with signs of anti-tumor activity. Serial tumor biopsies were feasible and revealed proof-of-concept biomarkers.
- Published
- 2015
29. Hedgehog signaling drives radioresistance and stroma-driven tumor repopulation in head and neck squamous cancers
- Author
-
Barbara Frederick, Phuong Le, Ryan T. Anderson, Magdalena J. Glogowska, Stephen B. Keysar, Justin R. Eagles, Antonio Jimeno, David Raben, Xiao-Jing Wang, Guoliang Wang, Cem Altunbas, John Morton, and Gregory N. Gan
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Cyclopamine ,Mice, Nude ,Biology ,Radiation Tolerance ,Zinc Finger Protein GLI1 ,Article ,chemistry.chemical_compound ,Mice ,GLI1 ,Radioresistance ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Epithelial–mesenchymal transition ,Hedgehog ,PI3K/AKT/mTOR pathway ,integumentary system ,Squamous Cell Carcinoma of Head and Neck ,TOR Serine-Threonine Kinases ,Veratrum Alkaloids ,Cancer ,Ribosomal Protein S6 Kinases, 70-kDa ,medicine.disease ,Hedgehog signaling pathway ,Up-Regulation ,Oncology ,chemistry ,Head and Neck Neoplasms ,Cancer research ,biology.protein ,Carcinoma, Squamous Cell ,Stromal Cells ,Signal Transduction ,Transcription Factors - Abstract
Local control and overall survival in patients with advanced head and neck squamous cell cancer (HNSCC) remains dismal. Signaling through the Hedgehog (Hh) pathway is associated with epithelial-to-mesenchymal transition, and activation of the Hh effector transcription factor Gli1 is a poor prognostic factor in this disease setting. Here, we report that increased GLI1 expression in the leading edge of HNSCC tumors is further increased by irradiation, where it contributes to therapeutic inhibition. Hh pathway blockade with cyclopamine suppressed GLI1 activation and enhanced tumor sensitivity to radiotherapy. Furthermore, radiotherapy-induced GLI1 expression was mediated in part by the mTOR/S6K1 pathway. Stroma exposed to radiotherapy promoted rapid tumor repopulation, and this effect was suppressed by Hh inhibition. Our results demonstrate that Gli1 that is upregulated at the tumor–stroma intersection in HNSCC is elevated by radiotherapy, where it contributes to stromal-mediated resistance, and that Hh inhibitors offer a rational strategy to reverse this process to sensitize HNSCC to radiotherapy. Cancer Res; 74(23); 7024–36. ©2014 AACR.
- Published
- 2014
30. The dual pathway inhibitor rigosertib is effective in direct patient tumor xenografts of head and neck squamous cell carcinomas
- Author
-
Justin R. Eagles-Soukup, Stephen B. Keysar, Aik Choon Tan, Marileila Varella-Garcia, Sarah M. Takimoto, Phuong Le, Daniel W. Bowles, Antonio Jimeno, Brian W. Vogler, John Morton, Barbara Frederick, Francois Wilhelm, Magdalena J. Glogowska, Dara L. Aisner, Gregory N. Gan, Ryan T. Anderson, Adrian Umpierrez, Daniel Sehrt, and David P. Astling
- Subjects
Cancer Research ,Class I Phosphatidylinositol 3-Kinases ,Cell ,Glycine ,Cell Cycle Proteins ,Biology ,Protein Serine-Threonine Kinases ,PLK1 ,Article ,Phosphatidylinositol 3-Kinases ,In vivo ,Proto-Oncogene Proteins ,Carcinoma ,medicine ,Humans ,Sulfones ,neoplasms ,Papillomaviridae ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Kinase ,Papillomavirus Infections ,Rigosertib ,medicine.disease ,Molecular biology ,Xenograft Model Antitumor Assays ,stomatognathic diseases ,medicine.anatomical_structure ,Oncology ,Head and Neck Neoplasms ,Mutation ,Carcinoma, Squamous Cell ,Immunohistochemistry ,Signal Transduction - Abstract
The dual pathway inhibitor rigosertib inhibits phosphoinositide 3-kinase (PI3K) pathway activation as well as polo-like kinase 1 (PLK1) activity across a broad spectrum of cancer cell lines. The importance of PIK3CA alterations in squamous cell carcinoma of the head and neck (HNSCC) has raised interest in exploring agents targeting PI3K, the product of PIK3CA. The genetic and molecular basis of rigosertib treatment response was investigated in a panel of 16 HNSCC cell lines, and direct patient tumor xenografts from eight patients with HNSCC [four HPV-serotype16 (HPV16)–positive]. HNSCC cell lines and xenografts were characterized by pathway enrichment gene expression analysis, exon sequencing, gene copy number, Western blotting, and immunohistochemistry (IHC). Rigosertib had potent antiproliferative effects on 11 of 16 HPV− HNSCC cell lines. Treatment sensitivity was confirmed in two cell lines using an orthotopic in vivo xenograft model. Growth reduction after rigosertib treatment was observed in three of eight HNSCC direct patient tumor lines. The responsive tumor lines carried a combination of a PI3KCA-activating event (amplification or mutation) and a p53-inactivating event (either HPV16- or mutation-mediated TP53 inactivation). In this study, we evaluated the in vitro and in vivo efficacy of rigosertib in both HPV+ and HPV− HNSCCs, focusing on inhibition of the PI3K pathway. Although consistent inhibition of the PI3K pathway was not evident in HNSCC, we identified a combination of PI3K/TP53 events necessary, but not sufficient, for rigosertib sensitivity. Mol Cancer Ther; 12(10); 1994–2005. ©2013 AACR.
- Published
- 2013
31. Migration of Head and Neck Squamous Cell Cancer Cells is Dependent on Tenascin-C Expression
- Author
-
Carissa M. Thomas, Stephen B. Keysar, Magdalena J. Glogowska, and Antonio Jimeno
- Subjects
Cancer Research ,Radiation ,Squamous cell cancer ,Oncology ,biology ,business.industry ,Tenascin C ,Cancer research ,biology.protein ,Medicine ,Radiology, Nuclear Medicine and imaging ,business ,Head and neck - Published
- 2016
- Full Text
- View/download PDF
32. Complement alternative pathway activation in the autologous phase of nephrotoxic serum nephritis
- Author
-
Richard J. Quigg, Sarah E. Panzer, Susan A. Boackle, Svetlana N. Tchepeleva, Magdalena J. Glogowska, V. Michael Holers, Mark Haas, and Joshua M. Thurman
- Subjects
Mice, Knockout ,Innate immune system ,Nephritis ,Physiology ,Complement Pathway, Alternative ,Kidney Glomerulus ,Glomerulonephritis ,Complement receptor ,Complement C3 ,Articles ,Biology ,medicine.disease ,Complement factor B ,Immune complex ,Complement system ,Disease Models, Animal ,Mice ,Proteinuria ,Immunology ,Alternative complement pathway ,medicine ,Animals ,Complement Factor B - Abstract
The complement cascade is an important part of the innate immune system, but pathological activation of this system causes tissue injury in several autoimmune and inflammatory diseases, including immune complex glomerulonephritis. We examined whether mice with targeted deletion of the gene for factor B ( fB−/− mice) and selective deficiency in the alternative pathway of complement are protected from injury in the nephrotoxic serum (NTS) nephritis model of antibody-mediated glomerulonephritis. When the acute affects of the anti-glomerular basement membrane antibody were assessed, fB−/− mice developed a degree of injury similar to wild-type controls. If the mice were presensitized with sheep IgG or if the mice were followed for 5 mo postinjection, however, the fB−/− mice developed milder injury than wild-type mice. The immune response of fB−/− mice exposed to sheep IgG was similar to that of wild-type mice, but the fB−/− mice had less glomerular C3 deposition and lower levels of albuminuria. These results demonstrate that fB−/− mice are not significantly protected from acute heterologous injury in NTS nephritis but are protected from autologous injury in response to a planted glomerular antigen. Thus, although the glomerulus is resistant to antibody-initiated, alternative pathway-mediated injury, inhibition of this complement pathway may be beneficial in chronic immune complex-mediated diseases.
- Published
- 2012
33. Role of C3a receptors, C5a receptors, and complement protein C6 deficiency in collagen antibody-induced arthritis in mice
- Author
-
Rick A. Wetsel, Magdalena J. Glogowska, V. Michael Holers, Nirmal K. Banda, Gregory L. Stahl, Jason T. White, Tod J. Merkel, Alexandra H. Antonioli, William P. Arend, Stacey L. Mueller-Ortiz, Stephanie Hyatt, and Kazue Takahashi
- Subjects
Neutrophils ,medicine.medical_treatment ,Immunology ,Arthritis ,Immunoglobulin G ,Article ,Mice ,Synovial Fluid ,medicine ,Immunology and Allergy ,Animals ,Receptor ,Complement Activation ,Receptor, Anaphylatoxin C5a ,Mice, Knockout ,biology ,Macrophages ,medicine.disease ,Molecular biology ,Arthritis, Experimental ,Complement system ,Complement C6 ,Receptors, Complement ,Cytokine ,Alternative complement pathway ,biology.protein ,Complement C3a ,Cytokines ,Tumor necrosis factor alpha ,Disease Susceptibility ,Complement membrane attack complex - Abstract
The complement system, especially the alternative pathway, plays essential roles in the induction of injury in collagen Ab-induced arthritis (CAIA) in mice. The goal of the current study was to directly compare the roles of receptors for C3a and C5a, as well as the membrane attack complex, as effector mechanisms in the pathogenesis of CAIA. Clinical disease activity in C3aR−/−, C5aR−/−, and C6-deficient (C6-def) mice was decreased by 52, 94, and 65%, respectively, as compared with wild-type mice. Decreases in histopathologic injury as well as in IgG and C3 deposition paralleled the clinical disease activity. A decrease in the percentage of synovial neutrophils was observed in C3aR−/−, C5aR−/−, and C6-def mice, and a decrease in macrophages was observed in C3aR−/− and C5aR−/−, but not in C6-def, mice. Synovial mRNA obtained by laser capture microdissection exhibited a decrease in TNF-α in C5aR−/− mice and in IL-1β in both C5aR−/− and C6-def mice, whereas C3aR−/− mice demonstrated no change in either cytokine. Our findings show that absent C3aR-, C5aR-, or membrane attack complex-initiated effector mechanisms each decrease susceptibility to CAIA, with clinical effects most pronounced in C5aR-deficient mice. Although the absence of C3aR, C5aR, or C6 led to differential deficiencies in effector mechanisms, decreased proximal joint IgG and C3 deposition was common to all three genotypes in comparison with wild-type mice. These data suggest the existence of positive-feedback amplification pathways downstream of all three effectors that promote additional IgG deposition and C3 activation in the joint.
- Published
- 2011
34. N-α-Benzoyl-N5-(2-Chloro-1-Iminoethyl)-L-Ornithine Amide, a Protein Arginine Deiminase Inhibitor, Reduces the Severity of Murine Collagen-Induced Arthritis
- Author
-
Corey P. Causey, Magdalena J. Glogowska, V. Michael Holers, Paul R. Thompson, Piyanka E Chandra, Kristen N. Cordova, Van C. Willis, Alison M. Gizinski, Bryan Knuckley, Nirmal K. Banda, William P. Arend, Brandt Levitt, Yuan Luo, Liudmila Kulik, and William H. Robinson
- Subjects
Male ,Ornithine ,Hydrolases ,Inflammatory arthritis ,Immunology ,Amidines ,Arthritis ,Pharmacology ,medicine.disease_cause ,Protein citrullination ,Peptides, Cyclic ,Severity of Illness Index ,Article ,Autoimmunity ,Arthritis, Rheumatoid ,Mice ,medicine ,Immunology and Allergy ,Animals ,Enzyme Inhibitors ,Collagen Type II ,Autoantibodies ,business.industry ,Synovial Membrane ,Autoantibody ,Citrullination ,Protein-arginine deiminase ,medicine.disease ,Arthritis, Experimental ,Mice, Inbred DBA ,Rheumatoid arthritis ,Protein-Arginine Deiminases ,Citrulline ,business ,Immunosuppressive Agents - Abstract
Rheumatoid arthritis is associated with the development of autoantibodies to citrullinated self-proteins. Citrullinated synovial proteins, which are generated via the actions of the protein arginine deiminases (PADs), are known to develop in the murine collagen-induced arthritis (CIA) model of inflammatory arthritis. Given these findings, we evaluated whether N-α-benzoyl-N5-(2-chloro-1-iminoethyl)-l-ornithine amide (Cl-amidine), a recently described pan-PAD inhibitor, could affect the development of arthritis and autoimmunity by treating mice in the CIA model with Cl-amidine on days 0–35. Cl-amidine treatment reduced total synovial and serum citrullination, decreased clinical disease activity by ∼50%, and significantly decreased IgG2a anti-mouse type II collagen Abs. Additionally, histopathology scores and total complement C3 deposition were significantly lower in Cl-amidine–treated mice compared with vehicle controls. Synovial microarray analyses demonstrated decreased IgG reactivity to several native and citrullinated epitopes compared with vehicle controls. Cl-amidine treatment had no ameliorative effect on collagen Ab-induced arthritis, suggesting its primary protective mechanism was not mediated through effector pathways. Reduced levels of citrullinated synovial proteins observed in mice treated with Cl-amidine are consistent with the notion that Cl-amidine derives its efficacy from its ability to inhibit the deiminating activity of PADs. In total, these results suggested that PADs are necessary participants in the autoimmune and subsequent inflammatory processes in CIA. Cl-amidine may represent a novel class of disease-modifying agents that modulate aberrant citrullination, and perhaps other immune processes, necessary for the development of inflammatory arthritis.
- Published
- 2011
35. Intrathecal pathogenic anti-aquaporin-4 antibodies in early neuromyelitis optica
- Author
-
Katherine Bautista, Sudhakar Reddy Kalluri, Magdalena J. Glogowska, David Case, Bernhard Hemmer, Chiwah Lam, Cecily Dupree, Christine Stadelmann, Philippe Saikali, Jack P. Antel, Stefan Nessler, Gregory P. Owens, Donald H. Gilden, and Jeffrey Bennett
- Subjects
Time Factors ,Population ,Molecular Sequence Data ,Plasma cell ,Spinal Puncture ,Article ,Epitope ,Cell Line ,Mice ,Fetus ,Immunopathology ,Demyelinating disease ,medicine ,Animals ,Humans ,Amino Acid Sequence ,education ,Cells, Cultured ,Autoantibodies ,Aquaporin 4 ,education.field_of_study ,Neuromyelitis optica ,biology ,business.industry ,Experimental autoimmune encephalomyelitis ,Neuromyelitis Optica ,Middle Aged ,medicine.disease ,Rats ,medicine.anatomical_structure ,Neurology ,Rats, Inbred Lew ,Immunoglobulin G ,Immunology ,biology.protein ,Female ,Neurology (clinical) ,Antibody ,business ,Biomarkers - Abstract
Objective The serum of most neuromyelitis optica (NMO) patients contains autoantibodies (NMO-IgGs) directed against the aquaporin-4 (AQP4) water channel located on astrocyte foot processes in the perivessel and subpial areas of the brain. Our objectives were to determine the source of central nervous system (CNS) NMO-IgGs and their role in disease pathogenesis. Methods Fluorescence-activated cell sorting and single-cell reverse transcriptase polymerase chain reaction were used to identify overrepresented plasma cell immunoglobulin (Ig) sequences in the cerebrospinal fluid (CSF) of an NMO patient after a first clinical attack. Monoclonal recombinant antibodies (rAbs) were generated from the paired heavy and light chain sequences and tested for target specificity and Fc effector function. The effect of CSF rAbs on CNS immunopathology was investigated by delivering single rAbs to rats with experimental autoimmune encephalomyelitis (EAE). Results Repertoire analysis revealed a dynamic, clonally expanded plasma cell population with features of an antigen-targeted response. Using multiple independent assays, 6 of 11 rAbs generated from CSF plasma cell clones specifically bound to AQP4. AQP4-specific rAbs recognized conformational epitopes and mediated both AQP4-directed antibody-dependent cellular cytotoxicity and complement-mediated lysis. When administered to rats with EAE, an AQP4-specific NMO CSF rAb induced NMO immunopathology: perivascular astrocyte depletion, myelinolysis, and complement and Ig deposition. Interpretation Molecular characterization of the CSF plasma cell repertoire in an early NMO patient demonstrates that AQP4-specfic Ig is synthesized intrathecally at disease onset and directly contributes to CNS pathology. AQP4 is now the first confirmed antigenic target in human demyelinating disease. Ann Neurol 2009;66:617–629
- Published
- 2009
36. Targeted inhibition of the complement alternative pathway with complement receptor 2 and factor H attenuates collagen antibody-induced arthritis in mice
- Author
-
Nirmal K. Banda, Magdalena J. Glogowska, V. Michael Holers, Gregory L. Stahl, William P. Arend, Joshua M. Thurman, Stephen Tomlinson, Brandt Levitt, and Kazue Takahashi
- Subjects
Male ,medicine.medical_specialty ,Complement receptor 2 ,Recombinant Fusion Proteins ,Immunology ,Complement Pathway, Alternative ,Type II collagen ,Arthritis ,chemical and pharmacologic phenomena ,Complement factor B ,Article ,Classical complement pathway ,Mice ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Collagen Type II ,Mice, Knockout ,Complement Inactivator Proteins ,Chemistry ,Antibodies, Monoclonal ,medicine.disease ,Molecular biology ,Arthritis, Experimental ,Complement system ,Mice, Inbred C57BL ,Drug Combinations ,Endocrinology ,Lectin pathway ,Complement Factor H ,Alternative complement pathway ,Cattle ,Receptors, Complement 3d - Abstract
The alternative pathway (AP) of complement is required for the induction of collagen Ab-induced arthritis (CAIA) in mice. The objective of this study was to examine the effect of a recombinant AP inhibitor containing complement receptor 2 and factor H (CR2-fH) on CAIA in mice. CR2 binds to tissue-fixed activation fragments of C3, and the linked fH is a potent local inhibitor of the AP. CAIA was induced in C57BL/6 mice by i.p. injections of 4 mAb to type II collagen (CII) on day 0 and LPS on day 3. PBS or CR2-fH (250 or 500 μg) were injected i.p. 15 min after the mAb to CII on day 0 and 15 min after LPS on day 3; the mice were sacrificed on day 10. The disease activity score (DAS) was decreased significantly (p < 0.001) in both groups receiving CR2-fH compared with the PBS. Histology scores for inflammation, pannus, bone damage, and cartilage damage decreased in parallel with the DAS. C3 deposition in the synovium and cartilage was significantly reduced (p < 0.0001) in the mice treated with CR2-fH. In vitro studies with immune complexes containing type II collagen and mAb to CII showed that CR2-fH specifically inhibited the AP with minimal effect on the classical pathway (CP) and no effect on the lectin pathway (LP). The relative potency of CR2-fH in vitro was superior to mAbs to factor B and C5. Thus, CR2-fH specifically targets and inhibits the AP of complement in vitro and is effective in CAIA in vivo.
- Published
- 2009
37. The Hedgehog Pathway Modulates Radiation Therapy Resistance in Head-and-Neck Cancer
- Author
-
David Raben, Antonio Jimeno, Ryan T. Anderson, Magdalena J. Glogowska, Phuong Le, Justin R. Eagles-Soukup, Stephen B. Keysar, John Morton, Gregory N. Gan, and C. Altunbus
- Subjects
Radiation therapy ,Cancer Research ,Radiation ,Oncology ,business.industry ,medicine.medical_treatment ,Head and neck cancer ,medicine ,Cancer research ,Radiology, Nuclear Medicine and imaging ,medicine.disease ,business ,Hedgehog signaling pathway - Published
- 2013
- Full Text
- View/download PDF
38. Independent roles for complement C3a and C5a receptors and the membrane attack complex in the pathogenesis of collagen antibody-induced arthritis in mice: Potential therapeutic implications
- Author
-
T.J. Merkel, Magdalena J. Glogowska, Gregory L. Stahl, Nirmal K. Banda, B. Lu, Kazue Takahashi, Craig Gerard, V.M. Holers, Stephanie Hyatt, William P. Arend, and Rick A. Wetsel
- Subjects
Pathogenesis ,Collagen antibody induced arthritis ,Immunology ,Biology ,Complement membrane attack complex ,Receptor ,Molecular Biology ,Complement system ,Complement (complexity) ,Cell biology - Published
- 2011
- Full Text
- View/download PDF
39. Essential role of complement MASP-1/3 in the murine collagen antibody-induced model of inflammatory arthritis 1
- Author
-
Brandt Levitt, Nirmal K. Banda, Gregory L. Stahl, Kazue Takahashi, Minoru Takahashi, Teizo Fujita, Jessica Nicholas, Magdalena J. Glogowska, V. Michael Holers, and William P. Arend
- Subjects
business.industry ,Inflammatory arthritis ,Immunology ,Medicine ,business ,medicine.disease ,Molecular Biology ,Collagen antibody ,Complement (complexity) - Published
- 2010
- Full Text
- View/download PDF
40. Targeted inhibition of the alternative pathway of complement with complement receptor 2-factor H protein attenuates collagen antibody-induced arthritis in mice
- Author
-
Nirmal K. Banda, Magdalena J. Glogowska, V. Michael Holers, Gregory L. Stahl, William P. Arend, Joshua M. Thurman, Kazue Takahashi, Brandt Levitt, and Stephen Tomlinson
- Subjects
Complement component 5 ,Complement component 2 ,Chemistry ,Complement receptor 2 ,Factor H ,Immunology ,Alternative complement pathway ,Molecular Biology ,Molecular biology ,Decay-accelerating factor ,CFHR5 ,Complement system - Published
- 2009
- Full Text
- View/download PDF
41. The alternative pathway of complement alone can mediate collagen antibody-induced arthritis in mice and generate high levels of C5a in vitro
- Author
-
Magdalena J. Glogowska, V. Michael Holers, Kazue Takahashi, Nirmal K. Banda, Brandt Levitt, William P. Arend, Gregory L. Stahl, and Allyson Wood
- Subjects
Chemistry ,Collagen antibody induced arthritis ,Immunology ,Alternative complement pathway ,Molecular Biology ,In vitro ,Complement (complexity) - 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.