87 results on '"Christian D. Young"'
Search Results
2. Cancer-Associated Fibroblasts Facilitate Squamous Cell Carcinoma Lung Metastasis in Mice by Providing TGFβ-Mediated Cancer Stem Cell Niche
- Author
-
Xueke Shi, Jingjing Luo, Kelsey J. Weigel, Spencer C. Hall, Danfeng Du, Fanglong Wu, Michael C. Rudolph, Hongmei Zhou, Christian D. Young, and Xiao-Jing Wang
- Subjects
squamous cell carcinoma ,transforming growth factor-beta ,cancer associated fibroblast ,lung metastasis ,cancer stem cell ,Biology (General) ,QH301-705.5 - Abstract
Cancer-associated fibroblasts (CAFs) have been shown to enhance squamous cell carcinoma (SCC) growth, but it is unclear whether they promote SCC lung metastasis. We generated CAFs from K15.KrasG12D.Smad4–/– mouse SCCs. RNA expression analyses demonstrated that CAFs had enriched transforming growth factor-beta (TGFβ) signaling compared to normal tissue-associated fibroblasts (NAFs), therefore we assessed how TGFβ-enriched CAFs impact SCC metastasis. We co-injected SCC cells with CAFs to the skin, tail vein, or the lung to mimic sequential steps of lung metastasis. CAFs increased SCC volume only in lung co-transplantations, characterized with increased proliferation and angiogenesis and decreased apoptosis compared to NAF co-transplanted SCCs. These CAF effects were attenuated by a clinically relevant TGFβ receptor inhibitor, suggesting that CAFs facilitated TGFβ-dependent SCC cell seeding and survival in the lung. CAFs also increased tumor volume when co-transplanted to the lung with limiting numbers of SCC cancer stem cells (CSCs). In vitro, CSC sphere formation and invasion were increased either with co-cultured CAFs or with CAF conditioned media (which contains the highest TGFβ1 concentration) and these CAF effects were blocked by TGFβ inhibition. Further, TGFβ activation was higher in primary human oral SCCs with lung metastasis than SCCs without lung metastasis. Similarly, TGFβ activation was detected in the lungs of mice with micrometastasis. Our data suggest that TGFβ-enriched CAFs play a causal role in CSC seeding and expansion in the lung during SCC metastasis, providing a prognostic marker and therapeutic target for SCC lung metastasis.
- Published
- 2021
- Full Text
- View/download PDF
3. Two distinct mTORC2-dependent pathways converge on Rac1 to drive breast cancer metastasis
- Author
-
Meghan Morrison Joly, Michelle M. Williams, Donna J. Hicks, Bayley Jones, Violeta Sanchez, Christian D. Young, Dos D. Sarbassov, William J. Muller, Dana Brantley-Sieders, and Rebecca S. Cook
- Subjects
mTOR ,Rictor ,Akt ,Rac ,Metastasis ,Breast cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The importance of the mTOR complex 2 (mTORC2) signaling complex in tumor progression is becoming increasingly recognized. HER2-amplified breast cancers use Rictor/mTORC2 signaling to drive tumor formation, tumor cell survival and resistance to human epidermal growth factor receptor 2 (HER2)-targeted therapy. Cell motility, a key step in the metastatic process, can be activated by mTORC2 in luminal and triple negative breast cancer cell lines, but its role in promoting metastases from HER2-amplified breast cancers is not yet clear. Methods Because Rictor is an obligate cofactor of mTORC2, we genetically engineered Rictor ablation or overexpression in mouse and human HER2-amplified breast cancer models for modulation of mTORC2 activity. Signaling through mTORC2-dependent pathways was also manipulated using pharmacological inhibitors of mTOR, Akt, and Rac. Signaling was assessed by western analysis and biochemical pull-down assays specific for Rac-GTP and for active Rac guanine nucleotide exchange factors (GEFs). Metastases were assessed from spontaneous tumors and from intravenously delivered tumor cells. Motility and invasion of cells was assessed using Matrigel-coated transwell assays. Results We found that Rictor ablation potently impaired, while Rictor overexpression increased, metastasis in spontaneous and intravenously seeded models of HER2-overexpressing breast cancers. Additionally, migration and invasion of HER2-amplified human breast cancer cells was diminished in the absence of Rictor, or upon pharmacological mTOR kinase inhibition. Active Rac1 was required for Rictor-dependent invasion and motility, which rescued invasion/motility in Rictor depleted cells. Rictor/mTORC2-dependent dampening of the endogenous Rac1 inhibitor RhoGDI2, a factor that correlated directly with increased overall survival in HER2-amplified breast cancer patients, promoted Rac1 activity and tumor cell invasion/migration. The mTORC2 substrate Akt did not affect RhoGDI2 dampening, but partially increased Rac1 activity through the Rac-GEF Tiam1, thus partially rescuing cell invasion/motility. The mTORC2 effector protein kinase C (PKC)α did rescue Rictor-mediated RhoGDI2 downregulation, partially rescuing Rac-guanosine triphosphate (GTP) and migration/motility. Conclusion These findings suggest that mTORC2 uses two coordinated pathways to activate cell invasion/motility, both of which converge on Rac1. Akt signaling activates Rac1 through the Rac-GEF Tiam1, while PKC signaling dampens expression of the endogenous Rac1 inhibitor, RhoGDI2.
- Published
- 2017
- Full Text
- View/download PDF
4. Transforming Growth Factor-β Signaling in Fibrotic Diseases and Cancer-Associated Fibroblasts
- Author
-
Xueke Shi, Christian D. Young, Hongmei Zhou, and Xiao-Jing Wang
- Subjects
TGF-β signaling ,fibroblasts ,fibrotic diseases ,cancer ,cancer-associated fibroblast ,anti-fibrosis/cancer therapy ,Microbiology ,QR1-502 - Abstract
Transforming growth factor-β (TGF-β) signaling is essential in embryo development and maintaining normal homeostasis. Extensive evidence shows that TGF-β activation acts on several cell types, including epithelial cells, fibroblasts, and immune cells, to form a pro-fibrotic environment, ultimately leading to fibrotic diseases. TGF-β is stored in the matrix in a latent form; once activated, it promotes a fibroblast to myofibroblast transition and regulates extracellular matrix (ECM) formation and remodeling in fibrosis. TGF-β signaling can also promote cancer progression through its effects on the tumor microenvironment. In cancer, TGF-β contributes to the generation of cancer-associated fibroblasts (CAFs) that have different molecular and cellular properties from activated or fibrotic fibroblasts. CAFs promote tumor progression and chronic tumor fibrosis via TGF-β signaling. Fibrosis and CAF-mediated cancer progression share several common traits and are closely related. In this review, we consider how TGF-β promotes fibrosis and CAF-mediated cancer progression. We also discuss recent evidence suggesting TGF-β inhibition as a defense against fibrotic disorders or CAF-mediated cancer progression to highlight the potential implications of TGF-β-targeted therapies for fibrosis and cancer.
- Published
- 2020
- Full Text
- View/download PDF
5. Revisiting laminin and extracellular matrix remodeling in metastatic squamous cell carcinoma: What have we learned after more than four decades of research?
- Author
-
John D. Aleman, Christian D. Young, Sana D. Karam, and Xiao‐Jing Wang
- Subjects
Cancer Research ,Molecular Biology - Abstract
Patients with squamous cell carcinoma (SCC) have significantly lower survival upon the development of distant metastases. The extracellular matrix (ECM) is a consistent yet dynamic influence on the metastatic capacity of SCCs. The ECM encompasses a milieu of structural proteins, signaling molecules, and enzymes. Just over 40 years ago, the fibrous ECM glycoprotein laminin was identified. Roughly four decades of research have revealed a pivotal role of laminins in metastasis. However, trends in ECM alterations in some cancers have been applied broadly to all metastatic diseases, despite evidence that these characteristics vary by tumor type. We will summarize how laminins influence the SCC metastatic process exclusively. Enhanced laminin protein deposition occurs at the invasive edge of SCC tumors, which correlates with elevated levels of laminin-binding β1 integrins on SCC cells, increased MMP-3 presence, worse prognosis, and lymphatic dissemination. Although these findings are significant, gaps in knowledge of the formation of a premetastatic niche, the processes of intra- and extravasation, and the contributions of the ECM to SCC metastatic cell dormancy persist. Bridging these gaps requires novel in vitro systems and animal models that reproduce tumor-stromal interactions and spontaneous metastasis seen in the clinic. These advances will allow accurate assessment of laminins to predict responders to transforming growth factor-β inhibitors and immunotherapy, as well as potential combinatorial therapies with the standard of care. Such clinical interventions may drastically improve quality of life and patient survival by explicitly targeting SCC metastasis.
- Published
- 2022
- Full Text
- View/download PDF
6. Epidermal growth factor receptor signaling in precancerous keratinocytes promotes neighboring head and neck cancer squamous cell carcinoma cancer stem cell‐like properties and phosphoinositide 3‐kinase inhibitor insensitivity
- Author
-
Khoa A. Nguyen, Madison J. Keith, Stephen B. Keysar, Spencer C. Hall, Anamol Bimali, Antonio Jimeno, Xiao‐Jing Wang, and Christian D. Young
- Subjects
Keratinocytes ,Cancer Research ,Squamous Cell Carcinoma of Head and Neck ,Receptors, Fibroblast Growth Factor ,Article ,ErbB Receptors ,Phosphatidylinositol 3-Kinases ,Head and Neck Neoplasms ,Cell Line, Tumor ,Carcinoma, Squamous Cell ,Neoplastic Stem Cells ,Tumor Microenvironment ,Humans ,Phosphatidylinositol 3-Kinase ,Precancerous Conditions ,Molecular Biology ,Phosphoinositide-3 Kinase Inhibitors - Abstract
Head and neck squamous cell carcinoma (HNSCC) is commonly associated with tobacco and alcohol consumption that induce a "precancerous field," with phosphoinositide 3-kinase (PI3K) signaling being a common driver. However, the preclinical effectiveness of PI3K inhibitors has not necessarily translated to remarkable benefit in HNSCC patients. Thus, we sought to determine how precancerous keratinocytes influence HNSCC proliferation, cancer stem cell (CSC) maintenance, and response to PI3K inhibitors. We used the NOK keratinocyte cell line as a model of preneoplastic keratinocytes because it harbors two frequent genetic events in HNSCC, CDKN2A promoter methylation and TP53 mutation, but does not form tumors. NOK cell coculture or NOK cell-conditioned media promoted HNSCC proliferation, PI3K inhibitor resistance, and CSC phenotypes. SOMAscan-targeted proteomics determined the relative levels of1300 analytes in the media conditioned by NOK cells and HNSCC cells ± PI3K inhibitor. These results demonstrated that NOK cells release abundant levels of ligands that activate epidermal growth factor receptor (EGFR) and fibroblast growth factor receptor (FGFR), two receptor tyrosine kinases with oncogenic activity. Inhibition of EGFR, but not FGFR, blunted PI3K inhibitor resistance and CSC phenotypes induced by NOK cells. Our results demonstrate that precancerous keratinocytes can directly support neighboring HNSCC by activating EGFR. Importantly, PI3K inhibitor sensitivity was not necessarily a cancer cell-intrinsic property, and the tumor microenvironment impacts therapeutic response and supports CSCs. Additionally, combined inhibition of EGFR with PI3K inhibitor diminished EGFR activation induced by PI3K inhibitor and potently inhibited cancer cell proliferation and CSC maintenance.
- Published
- 2022
- Full Text
- View/download PDF
7. IL22RA2 is a Smad7 target mediating alleviation of dermatitis and psoriatic phenotypes in mice
- Author
-
Yao Ke, Ben-Zheng Li, Khoa Nguyen, Donna Wang, Suyan Wang, Christian D. Young, and Xiao-Jing Wang
- Subjects
Cell Biology ,Dermatology ,Molecular Biology ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Data Figure 5 from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
PDF file 62K, MCL1 overexpression enhances docetaxel resistance in non-amplified TNBC cell lines
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Data Figure 3 from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
PDF file 77K, Association of gene alterations with gene expression
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Data Tables from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
XLSX file 706K, This file contains 8 supplementary data tables which include patient demographics, genes and alterations assessed by NGS, Nanostring gene expression data, and bioinformatics analyses
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Data Figure 1 from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
PDF file 45K, Association of high Ki67 with basal-like status
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Data Figure 2 from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
PDF file 26K, Association of node status with RFS and OS, stratified by menopausal status
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Data Figure 7 from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
PDF 118K, TSC1 truncation in the RD of a TNBC after NAC
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Data Figure 6 from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
PDF file 1474K, MEK activity is required for MYC-induced soft agar colony formation
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Data Figure Legends from Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Carlos L. Arteaga, Phillip J. Stephens, Vincent A. Miller, Maureen Cronin, Roman Yelensky, Gary A. Palmer, Jeffrey S. Ross, Jennifer A. Pietenpol, Joshua A. Bauer, Brian D. Lehmann, Andrei Goga, Dai Horiuchi, Henry Gómez, Franco D. Doimi, Joseph A. Pinto, Preston D. Moore, Richard Kurupi, Violeta Sánchez, Maria G. Kuba, Melinda E. Sanders, Phillip Owens, Rebecca S. Cook, Christian D. Young, Luis J. Schwarz, Kai Wang, Jennifer M. Giltnane, and Justin M. Balko
- Abstract
PDF file 64K, Legends for all supplementary data figures (1-7)
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figure S2 from PARP Inhibition Enhances Radiotherapy of SMAD4-Deficient Human Head and Neck Squamous Cell Carcinomas in Experimental Models
- Author
-
Xiao-Jing Wang, David Raben, Sana D. Karam, Ayman J. Oweida, Kenneth L. Jones, Michael C. Rudolph, G. Devon Trahan, Guanting He, Gangwen Han, Barbara Frederick, Kyle Nolan, Kelsey Weigel, Li Bian, Christian D. Young, and Ariel L. Hernandez
- Abstract
Supplementary Figure S2 demonstrates the enrichment of Fanoni Anemia pathway genes as a function of SMAD4 levels (in TCGA analyses)
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Data from Cancer Cell CD44 Mediates Macrophage/Monocyte-Driven Regulation of Head and Neck Cancer Stem Cells
- Author
-
Antonio Jimeno, Xiao-Jing Wang, Hilary L. Somerset, Hongmei Zhou, Christian D. Young, Traci R. Lyons, Anit Tyagi, Farshad N. Chowdhury, Cera Nieto, Phuong N. Le, Tugs-Saikhan Chimed, Bettina Miller, J. Jason Morton, Stephen B. Keysar, FangLong Wu, and Karina E. Gomez
- Abstract
Supplemental methods, figures, and table.
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Smad7 Promotes Healing of Radiotherapy-Induced Oral Mucositis without Compromising Oral Cancer Therapy in a Xenograft Mouse Model
- Author
-
Xiao-Jing Wang, Christian D. Young, Hongmei Zhou, David Raben, Rui Zhao, Barry Holwerda, Fanglong Wu, Danfeng Du, Chao Liang, Dongyan Wang, Melanie A. Blevins, Li Bian, and Jingjing Luo
- Abstract
Purpose:We previously reported preventive and therapeutic effects of Smad7, a multifunctional protein, on radiotherapy (RT)-induced mucositis in mice without promoting human oral cancer cell survival or migration in vitro. The current study aims to determine whether a Smad7-based biologic can treat existing oral mucositis during radiotherapy for oral cancer and whether this treatment compromises RT-induced cancer cell killing in neighboring oral cancer.Experimental Design: We transplanted human oral cancer cells into the tongues of mice and applied craniofacial irradiation to simultaneously kill tumor cells and induce oral mucositis, thus modeling RT and mucositis in oral cancer patients. We topically applied a recombinant human Smad7 protein fused with the cell-penetrating Tat tag (Tat-Smad7) to the oral mucosa of tumor-bearing mice post RT when oral mucositis began to develop.Results:Topically applied Tat-Smad7 penetrated cells in both the oral mucosa and oral cancer, attenuating TGFβ and NF-κB signaling as well as inflammation at both sites. Tat-Smad7 treatment alleviated oral mucositis with reductions in DNA damage and apoptosis in keratinocytes, but increased keratinocyte proliferation compared with vehicle-treated mucositis lesions. In contrast, adjacent oral cancer exposed to Tat-Smad7 did not show alterations in proliferation or direct DNA damage, but showed increased oxidative stress damage and apoptosis compared with tumors treated with vehicle.Conclusions:Our results suggest that short-course Tat-Smad7 application to oral mucositis promotes its healing but does not compromise the cytotoxic effect of RT on oral cancer and has context-specific effects on oral mucosa versus oral cancer.
- Published
- 2023
- Full Text
- View/download PDF
19. Data from PARP Inhibition Enhances Radiotherapy of SMAD4-Deficient Human Head and Neck Squamous Cell Carcinomas in Experimental Models
- Author
-
Xiao-Jing Wang, David Raben, Sana D. Karam, Ayman J. Oweida, Kenneth L. Jones, Michael C. Rudolph, G. Devon Trahan, Guanting He, Gangwen Han, Barbara Frederick, Kyle Nolan, Kelsey Weigel, Li Bian, Christian D. Young, and Ariel L. Hernandez
- Abstract
Purpose:SMAD4 loss causes genomic instability and the initiation/progression of head and neck squamous cell carcinoma (HNSCC). Here, we study whether SMAD4 loss sensitizes HNSCCs to olaparib (PARP inhibitor) in combination with radiotherapy (RT).Experimental Design:We analyzed HNSCC The Cancer Genome Atlas data for SMAD4 expression in association with FANC/BRCA family gene expression. Human HNSCC cell lines were screened for sensitivity to olaparib. Isogenic HNSCC cell lines were generated to restore or reduce SMAD4 expression and treated with olaparib, radiation, or the combination. HNSCC pretreatment specimens from a phase I trial investigating olaparib were analyzed.Results:SMAD4 levels correlated with levels of FANC/BRCA genes in HNSCC. HNSCC cell lines with SMAD4 homozygous deletion were sensitive to olaparib. In vivo, olaparib or RT monotherapy reduced tumor volumes in SMAD4-mutant but not SMAD4-positive tumors. Olaparib with RT dual therapy sustained tumor volume reduction in SMAD4-deficient (mutant or knockdown) xenografts, which exhibited increased DNA damage and cell death compared with vehicle-treated tumors. In vitro, olaparib alone or in combination with radiation caused lower clonogenic survival, more DNA damage–associated cell death, and less proliferation in SMAD4-deficient cells than in SMAD4-positive (endogenous SMAD4 or transduced SMAD4) cells. Applicable to clinic, 5 out of 6 SMAD4-negative HNSCCs and 4 out of 8 SMAD4-positive HNSCCs responded to a standard treatment plus olaparib in a phase I clinical trial, and SMAD4 protein levels inversely correlated with DNA damage.Conclusions:SMAD4 levels are causal in determining sensitivity to PARP inhibition in combination with RT in HNSCCs.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Table S1 from PARP Inhibition Enhances Radiotherapy of SMAD4-Deficient Human Head and Neck Squamous Cell Carcinomas in Experimental Models
- Author
-
Xiao-Jing Wang, David Raben, Sana D. Karam, Ayman J. Oweida, Kenneth L. Jones, Michael C. Rudolph, G. Devon Trahan, Guanting He, Gangwen Han, Barbara Frederick, Kyle Nolan, Kelsey Weigel, Li Bian, Christian D. Young, and Ariel L. Hernandez
- Abstract
Supplementary Table S1 lists reagents used
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Information from Conditional Loss of ErbB3 Delays Mammary Gland Hyperplasia Induced by Mutant PIK3CA without Affecting Mammary Tumor Latency, Gene Expression, or Signaling
- Author
-
Carlos L. Arteaga, Rebecca S. Cook, Jean J. Zhao, Charles M. Perou, Hailing Cheng, Violeta Sánchez, Justin M. Balko, Brent N. Rexer, María G. Kuba, Philip Owens, Adam D. Pfefferle, and Christian D. Young
- Abstract
Supplementary information - PDF file 2943K, Supplementary Figure 1: Dox-induced iPIK3.iCre transgenic mouse model of PIK3CA mutant breast cancer. Supplementary Figure 2: Loss of ErbB3 delays mammary gland hyperplasia at 12 weeks. Supplementary Figure 3: IVIS Bioluminescent imaging. Supplementary Figure 4: Histology of PIK3CA-induced mammary tumors remains diverse when ErbB3 is lost. Supplementary Figure 5: Mutant PI3K binds to IRS-1, Gab1 and other tyrosinephosphorylated proteins in tumors treated with lapatinib and/or imatinib. Supplementary Figure 6: Combination of BYL719 and lapatinib delays tumor growth without causing liver toxicity
- Published
- 2023
- Full Text
- View/download PDF
22. Therapeutic Intervention Using a Smad7-Based Tat Protein to Treat Radiation-Induced Oral Mucositis
- Author
-
Erin Trageser, Daniel P. Regan, Amber R. Prebble, Donna D. Wang, Li Bian, Ber-In Lee, Spencer C. Hall, Lyndah Chow, Christian D. Young, Steven W. Dow, Barry Holwerda, Tiffany Martin, David Raben, Sana D. Karam, Yao Ke, Mary-Keara Boss, Lauren G. Harrison, Suyan Wang, and Xiao-Jing Wang
- Subjects
Oncology ,Mucositis ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Inflammation ,Article ,Proinflammatory cytokine ,Smad7 Protein ,Mice ,Dogs ,Transforming Growth Factor beta ,Internal medicine ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Oral mucosa ,Radiation Injuries ,Stomatitis ,Radiation ,integumentary system ,business.industry ,Therapeutic effect ,Cancer ,medicine.disease ,Radiation therapy ,medicine.anatomical_structure ,Cancer cell ,Gene Products, tat ,medicine.symptom ,business - Abstract
Purpose Recent studies reported therapeutic effects of Smad7 on oral mucositis in mice without compromising radiation therapy–induced cancer cell killing in neighboring oral cancer. This study aims to assess whether a Smad7-based biologic can treat oral mucositis in a clinically relevant setting by establishing an oral mucositis model in dogs and analyzing molecular targets. Methods and Materials We created a truncated human Smad7 protein fused with the cell-penetrating Tat tag (Tat-PYC-Smad7). We used intensity modulated radiation therapy to induce oral mucositis in dogs and applied Tat-PYC-Smad7 to the oral mucosa in dose-finding studies after intensity modulated radiation therapy. Clinical outcomes were evaluated. Molecular targets were analyzed in biopsies and serum samples. Results Tat-PYC-Smad7 treatment significantly shortened the duration of grade 3 oral mucositis based on double-blinded Veterinary Radiation Therapy Oncology Group scores and histopathology evaluations. Topically applied Tat-PYC-Smad7 primarily penetrated epithelial cells and was undetectable in serum. NanoString nCounter Canine IO Panel identified that, compared to the vehicle samples, top molecular changes in Tat-PYC-Smad7 treated samples include reductions in inflammation and cell death and increases in cell growth and DNA repair. Consistently, immunostaining shows that Tat-PYC-Smad7 reduced DNA damage and neutrophil infiltration with attenuated TGF-β and NFκB signaling. Furthermore, IL-1β and TNF-α were lower in Tat-PYC-Smad7 treated mucosa and serum samples compared to those in vehicle controls. Conclusions Topical Tat-PYC-Smad7 application demonstrated therapeutic effects on oral mucositis induced by intensity modulated radiation therapy in dogs. The local effects of Tat-PYC-Smad7 targeted molecules involved in oral mucositis pathogenesis as well as reduced systemic inflammatory cytokines.
- Published
- 2021
23. Lessons learned fromSMAD4loss in squamous cell carcinomas
- Author
-
Jing Wang, Ariel L. Hernandez, Xiao-Jing Wang, and Christian D. Young
- Subjects
0301 basic medicine ,Genome instability ,Cancer Research ,animal structures ,DNA Repair ,DNA repair ,Cell ,Tumor initiation ,Biology ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Biomarkers, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,neoplasms ,Molecular Biology ,Smad4 Protein ,Tumor microenvironment ,Epithelial Cells ,digestive system diseases ,stomatognathic diseases ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,030220 oncology & carcinogenesis ,embryonic structures ,Carcinoma, Squamous Cell ,Cancer research ,Signal Transduction - Abstract
SMAD4 is a potent tumor suppressor and a central mediator of the TGFß signaling pathway. SMAD4 genetic loss is frequent in squamous cell carcinomas (SCCs). Reports of SMAD4 expression in SCCs vary significantly possibly due to inter-tumor heterogeneity or technical reasons. SMAD4 loss is an initiation event for SCCs. In tumor epithelial cells, SMAD4 loss causes increased proliferation, decreased apoptosis, and "Brca-like" genomic instability associated with DNA repair defects. SMAD4 loss also plays a role in the expansion of cancer stem cells. Epithelial SMAD4 loss causes overexpression of TGFß that is released into the tumor microenvironment and contributes to SCC progression through proinflammatory and immune evasive mechanisms. SMAD4 loss, while not a direct therapeutic target, is associated with multiple targetable pathways that require further therapeutic studies. Altogether, SMAD4 loss is a potential biomarker in SCCs that should be further studied for its values in prognostic and therapeutic predictions. Such information will potentially guide future biomarker-driven clinical trial designs and improve SCC patient outcomes.
- Published
- 2019
- Full Text
- View/download PDF
24. Smad7 Ameliorates TGF-β–Mediated Skin Inflammation and Associated Wound Healing Defects but Not Susceptibility to Experimental Skin Carcinogenesis
- Author
-
Gangwen Han, Shunsuke Iriyama, Bin Fan, Zhe Jian, Li Bian, Dongyan D. Wang, Christian D. Young, Fulun Li, Xiao-Jing Wang, and Jingjing Luo
- Subjects
Keratinocytes ,0301 basic medicine ,Skin Neoplasms ,Carcinogenesis ,Transgene ,Guinea Pigs ,Mice, Transgenic ,Inflammation ,Dermatology ,medicine.disease_cause ,Biochemistry ,Article ,Smad7 Protein ,Mice ,03 medical and health sciences ,Subcutaneous injection ,0302 clinical medicine ,Transforming Growth Factor beta ,Fibrosis ,Animals ,Humans ,Medicine ,RNA, Neoplasm ,Molecular Biology ,Skin ,Wound Healing ,integumentary system ,business.industry ,Neoplasms, Experimental ,Cell Biology ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Phenotype ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,medicine.symptom ,business ,Wound healing ,Infiltration (medical) ,Transforming growth factor - Abstract
We assessed the roles of Smad7 in skin inflammation and wound healing using genetic and pharmacological approaches. In K5.TGFβ1/K5.Smad7 bigenic (double transgenic) mice, Smad7 transgene expression reversed transforming growth factor (TGF)-β1 transgene-induced inflammation, fibrosis, and subsequent epidermal hyperplasia and molecularly abolished TGF-β and NF-κB activation. Next, we produced recombinant human Smad7 protein with a Tat-tag (Tat-Smad7) that rapidly enters cells. Subcutaneous injection of Tat-Smad7 attenuated infiltration of F4/80+ and CD11b+ leukocytes and α-smooth muscle actin+ fibroblasts before attenuating epidermal hyperplasia in K5.TGFβ1 skin. Furthermore, topically applied Tat-Smad7 on K5.TGFβ1 skin wounds accelerated wound closure, with improved re-epithelialization and reductions in inflammation and fibrotic response. A short treatment with Tat-Smad7 was also sufficient to reduce TGF-β and NF-κB signaling in K5.TGFβ1 skin and wounds. Relevant to the clinic, we found that human diabetic wounds had elevated TGF-β and NF-κB signaling compared with normal skin. To assess the oncogenic risk of a potential Smad7-based therapy, we exposed K5.Smad7 skin to chemical carcinogenesis and found reduced myeloid leukocyte infiltration in tumors but not accelerated carcinogenesis compared with wild-type littermates. Our study suggests the feasibility of using exogenous Smad7 below an oncogenic level to alleviate skin inflammation and wound healing defects associated with excessive activation of TGF-β and NF-κB.
- Published
- 2019
- Full Text
- View/download PDF
25. Abstract 3834: Assessing the role of extracellular matrix-integrins in metastatic squamous cell carcinoma
- Author
-
John Aleman, Khoa A. Nguyen, Yao Ke, Christian D. Young, and Xiao-Jing Wang
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Patients with head and neck squamous cell carcinoma (HNSCC) have a dismal survival rate. The effects of the extracellular matrix (ECM) on cancer progression have been long studied, but the roles of specific integrins in the process of HNSCC metastasis remain to be dissected. This study aims to determine how HNSCC cells affect the production of laminin-binding integrins, and how these integrins participate in the ECM interactions necessary for a metastatic phenotype. Experimental design: Our laboratory has produced syngeneic mouse SCC cell lines, P029 and A223, derived from Keratin15+ stem cells with Smad4 loss and KrasG12D mutation. In syngeneic recipients, SCCs derived from P029 cells transplanted to the flank mouse skin produced spontaneous metastases to the lung while SCCs derived from A223 cells did not form metastases. Having the same genotype, these cell lines serve as models to examine cancer cell interactions with the ECM and resulting effects on invasion and metastasis. Bulk RNA sequencing (RNAseq) was performed to compare cultured metastatic P029 cells versus non-metastatic A223 cells and identify differentially expressed genes that regulate SCC cells and ECM interactions. Immunoassays and functional invasion assays were performed to evaluate ECM-cancer cell signaling and influence on cancer cell invasion in these two cell lines. Results: RNAseq analysis revealed that, relative to A223 cells, P029 cells have increased levels of integrins, the main mechanoreceptors for numerous ECM ligands and matrix proteins. Laminin-binding integrins, including integrins α4 and β6, were greater at the RNA and protein levels. Additionally, P029 cells expressed higher levels of fibronectin and laminin coding genes compared to non-metastatic A223 cells. Correlatively, high expression of these matrix proteins is associated with worse patient survival. Additionally, P029 SCC cells displayed differential gene expression of tight and apical junctions, PI3K signaling components, and regulators of cytoskeletal remodeling. Furthermore, ELISA and western blot analysis revealed that P029 has aberrant TGFβ-Smad signaling as indicated by the elevated release of TGFβ-1 protein and higher levels of phosphorylated Smad2 and Smad3 relative to non-metastatic A223. Treating P029 cells with TGFβ-1 significantly increased their motility and invasion. Conversely, migration and motility of P029 cells were radically reduced by the TGFβ inhibitor galunisertib. Additionally, the mouse cytokine array revealed that mouse plasma bearing P029 tumors had a greater circulation of CXCL16, MMP-9, proliferin, and serpin E1. These proteins are associated with ECM remodeling and metastasis in HNSCC. Conclusions: SCC cells with metastatic properties upregulate integrins which bind to non-collagen matrix proteins and have elevated invasive and migratory capacity contributed from activated TGFβ signaling. Citation Format: John Aleman, Khoa A. Nguyen, Yao Ke, Christian D. Young, Xiao-Jing Wang. Assessing the role of extracellular matrix-integrins in metastatic squamous cell carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3834.
- Published
- 2022
- Full Text
- View/download PDF
26. Transforming Growth Factor-β Signaling in Fibrotic Diseases and Cancer-Associated Fibroblasts
- Author
-
Xiao-Jing Wang, Xueke Shi, Hongmei Zhou, and Christian D. Young
- Subjects
0301 basic medicine ,cancer-associated fibroblast ,lcsh:QR1-502 ,Review ,Biology ,Biochemistry ,TGF-β signaling ,lcsh:Microbiology ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Fibrosis ,Transforming Growth Factor beta ,fibroblasts ,medicine ,cancer ,Animals ,Humans ,Molecular Targeted Therapy ,Molecular Biology ,Tumor microenvironment ,Cancer ,anti-fibrosis/cancer therapy ,medicine.disease ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,Disease Progression ,Myofibroblast ,Transforming growth factor ,fibrotic diseases ,Signal Transduction - Abstract
Transforming growth factor-β (TGF-β) signaling is essential in embryo development and maintaining normal homeostasis. Extensive evidence shows that TGF-β activation acts on several cell types, including epithelial cells, fibroblasts, and immune cells, to form a pro-fibrotic environment, ultimately leading to fibrotic diseases. TGF-β is stored in the matrix in a latent form; once activated, it promotes a fibroblast to myofibroblast transition and regulates extracellular matrix (ECM) formation and remodeling in fibrosis. TGF-β signaling can also promote cancer progression through its effects on the tumor microenvironment. In cancer, TGF-β contributes to the generation of cancer-associated fibroblasts (CAFs) that have different molecular and cellular properties from activated or fibrotic fibroblasts. CAFs promote tumor progression and chronic tumor fibrosis via TGF-β signaling. Fibrosis and CAF-mediated cancer progression share several common traits and are closely related. In this review, we consider how TGF-β promotes fibrosis and CAF-mediated cancer progression. We also discuss recent evidence suggesting TGF-β inhibition as a defense against fibrotic disorders or CAF-mediated cancer progression to highlight the potential implications of TGF-β-targeted therapies for fibrosis and cancer.
- Published
- 2020
27. Deletion of p53 and Hyper-Activation of PIK3CA in Keratin-15+ Stem Cells Lead to the Development of Spontaneous Squamous Cell Carcinoma
- Author
-
Christian D. Young, Jing Wang, Alexandra L Krinsky, Bian Li, Rachel A. Woolaver, Zhangguo Chen, Xiaoguang Wang, Tonya Brunetti, Samantha M. Y. Chen, Dexiang Gao, Xiao-Jing Wang, and Andrew Nicklawsky
- Subjects
Class I Phosphatidylinositol 3-Kinases ,medicine.medical_treatment ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Catalysis ,Article ,head and neck cancers ,Inorganic Chemistry ,lcsh:Chemistry ,Immune system ,Lymphocytes, Tumor-Infiltrating ,Cancer immunotherapy ,Genetic model ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,neoplasms ,Spectroscopy ,Tumor microenvironment ,cancer immunotherapy ,Tumor-infiltrating lymphocytes ,Keratin-15 ,Organic Chemistry ,General Medicine ,Neoplasms, Experimental ,medicine.disease ,Genes, p53 ,Computer Science Applications ,lcsh:Biology (General) ,lcsh:QD1-999 ,tumor infiltrating lymphocytes ,Head and Neck Neoplasms ,Cancer research ,Carcinoma, Squamous Cell ,Skin cancer ,Stem cell ,Carcinogenesis - Abstract
Squamous cell carcinoma (SCC) is the second commonest type of skin cancer, and SCCs make up about 90% of head and neck cancers (HNSCCs). HNSCCs harbor two frequent molecular alterations, namely, gain-of-function alterations of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and loss-of-function mutations of tumor protein p53 (TP53). However, it remains poorly understood whether HNSCCs harboring different genetic alterations exhibit differential immune tumor microenvironments (TME). It also remains unknown whether PIK3CA hyperactivation and TP53 deletion can lead to SCC development spontaneously. Here, we analyzed the Cancer Genome Atlas (TCGA) datasets of HNSCCs and found that patients with both PIK3CA and TP53 alterations exhibited worse survival, significantly lower CD8 tumor infiltrating lymphocytes (TILs) and higher M0 macrophages than other controls. To better model human tumorigenesis, we deleted TP53 and constitutively activated PIK3CA in mouse keratin-15-expressing stem cells, which leads to the spontaneous development of multilineage tumors including SCCs, termed Keratin-15-p53-PIK3CA (KPPA) tumors. KPPA tumors were heavily infiltrated with myeloid-derived suppressor cells (MDSCs), with a drastically increased ratio of polymorphonuclear-MDSC (PMN-MDSC) versus monocytic-MDSC (M-MDSC). CD8 TILs expressed more PD-1 and reduced their polyfunctionality. Overall, we established a genetic model to mimic human HNSCC pathogenesis, manifested with an immunosuppressive TME, which may help further elucidate immune evasion mechanisms and develop more effective immunotherapies for HNSCCs.
- Published
- 2020
28. PARP Inhibition Enhances Radiotherapy of SMAD4-Deficient Human Head and Neck Squamous Cell Carcinomas in Experimental Models
- Author
-
Michael C. Rudolph, Ayman Oweida, David Raben, Kelsey J. Weigel, Xiao-Jing Wang, Ariel L. Hernandez, Kyle Nolan, Li Bian, Christian D. Young, Kenneth L. Jones, Gangwen Han, Sana D. Karam, G Devon Trahan, Guanting He, and Barbara Frederick
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Cell ,Nude ,Cetuximab ,Apoptosis ,Piperazines ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Cells, Cultured ,Smad4 Protein ,Cancer ,Gene knockdown ,Cultured ,integumentary system ,Prognosis ,Tumor Cells ,medicine.anatomical_structure ,Oncology ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,embryonic structures ,PARP inhibitor ,Female ,Programmed cell death ,animal structures ,DNA damage ,Oncology and Carcinogenesis ,Mice, Nude ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,Olaparib ,03 medical and health sciences ,Rare Diseases ,medicine ,Genetics ,Animals ,Humans ,Oncology & Carcinogenesis ,Dental/Oral and Craniofacial Disease ,neoplasms ,Cell Proliferation ,business.industry ,Animal ,Squamous Cell Carcinoma of Head and Neck ,Human Genome ,medicine.disease ,Head and neck squamous-cell carcinoma ,Xenograft Model Antitumor Assays ,digestive system diseases ,Radiation therapy ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Disease Models ,Cancer research ,Phthalazines ,business - Abstract
Purpose: SMAD4 loss causes genomic instability and the initiation/progression of head and neck squamous cell carcinoma (HNSCC). Here, we study whether SMAD4 loss sensitizes HNSCCs to olaparib (PARP inhibitor) in combination with radiotherapy (RT). Experimental Design: We analyzed HNSCC The Cancer Genome Atlas data for SMAD4 expression in association with FANC/BRCA family gene expression. Human HNSCC cell lines were screened for sensitivity to olaparib. Isogenic HNSCC cell lines were generated to restore or reduce SMAD4 expression and treated with olaparib, radiation, or the combination. HNSCC pretreatment specimens from a phase I trial investigating olaparib were analyzed. Results: SMAD4 levels correlated with levels of FANC/BRCA genes in HNSCC. HNSCC cell lines with SMAD4 homozygous deletion were sensitive to olaparib. In vivo, olaparib or RT monotherapy reduced tumor volumes in SMAD4-mutant but not SMAD4-positive tumors. Olaparib with RT dual therapy sustained tumor volume reduction in SMAD4-deficient (mutant or knockdown) xenografts, which exhibited increased DNA damage and cell death compared with vehicle-treated tumors. In vitro, olaparib alone or in combination with radiation caused lower clonogenic survival, more DNA damage–associated cell death, and less proliferation in SMAD4-deficient cells than in SMAD4-positive (endogenous SMAD4 or transduced SMAD4) cells. Applicable to clinic, 5 out of 6 SMAD4-negative HNSCCs and 4 out of 8 SMAD4-positive HNSCCs responded to a standard treatment plus olaparib in a phase I clinical trial, and SMAD4 protein levels inversely correlated with DNA damage. Conclusions: SMAD4 levels are causal in determining sensitivity to PARP inhibition in combination with RT in HNSCCs.
- Published
- 2020
29. Final Report of a Phase I Trial of Olaparib with Cetuximab and Radiation for Heavy Smoker Patients with Locally Advanced Head and Neck Cancer
- Author
-
Timothy V. Waxweiler, Aik Choo Tan, Patrick J. Blatchford, Xiao-Jing Wang, Dara L. Aisner, Christian D. Young, Antonio Jimeno, Carrie Marshall, David Raben, Ayman Oweida, Krishna Reddy, Daniel W. Bowles, Hilary Somerset, Sana D. Karam, Alicia M. DeLouize, Kurtis D. Davies, and Madeleine A. Kane
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_treatment ,DNA Mutational Analysis ,Cetuximab ,Piperazines ,chemistry.chemical_compound ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Neoplasm Metastasis ,education.field_of_study ,Smokers ,Middle Aged ,Prognosis ,Combined Modality Therapy ,Treatment Outcome ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,PARP inhibitor ,Female ,medicine.drug ,medicine.medical_specialty ,Population ,Article ,Olaparib ,03 medical and health sciences ,Internal medicine ,Biomarkers, Tumor ,medicine ,Mucositis ,Humans ,education ,Aged ,Neoplasm Staging ,business.industry ,Head and neck cancer ,Cancer ,medicine.disease ,Radiation therapy ,030104 developmental biology ,chemistry ,Phthalazines ,Radiotherapy, Intensity-Modulated ,Transcriptome ,business ,Radiotherapy, Image-Guided - Abstract
Purpose: Our goal was to evaluate the safety and toxicity of combining a PARP inhibitor, olaparib, with cetuximab and fractionated intensity-modulated radiotherapy for patients with locally advanced head and neck cancer and heavy smoking histories. Patients and Methods: Patients with ≥10 packs/year history of smoking were treated with olaparib at doses ranging from 25–200 mg orally twice daily beginning approximately 10 days prior to initiation of and with concurrent radiation (69.3 Gy in 33 fractions) using a time-to-event continual reassessment method model. Cetuximab was administered starting approximately 5 days prior to radiation per standard of care. Results: A total of 16 patients were entered onto the study, with 15 evaluable for acute toxicity. The most common treatment-related grade 3–4 side effects were radiation dermatitis and mucositis (38% and 69%, respectively). The MTD was determined to be 50 mg orally twice daily, but the recommended phase II dose was deemed to be 25 mg orally twice daily. At a median follow-up of 26 months, the actuarial median overall survival was 37 months, but was not reached for other endpoints. Two-year overall survival, progression-free survival, local control, and distant control rates were 72%, 63%, 72%, and 79%, respectively. Patients who continued to smoke during therapy experienced higher recurrence rates. MYC and KMT2A were identified as potential correlatives of response on gene amplification and mutational analysis. Conclusions: Olaparib at 25 mg orally twice daily with concurrent cetuximab and radiation was well tolerated with reduced dermatitis within the radiation field. Response rates were promising for this high-risk population. Clin Cancer Res; 24(20); 4949–59. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
30. Role of EphB3 Receptor in Mediating Head and Neck Tumor Growth, Cell Migration, and Response to PI3K Inhibitor
- Author
-
Shelby Lennon, Benjamin Van Court, Shilpa Bhatia, Christina Rohmer, Lynn E. Heasley, Jaspreet Sharma, Anastacia Griego, Christian D. Young, Sanjana Bukkapatnam, Sana D. Karam, Nomin Uyanga, Ayman Oweida, and David Raben
- Subjects
0301 basic medicine ,Cancer Research ,Class I Phosphatidylinositol 3-Kinases ,Morpholines ,Receptor, EphB3 ,Aminopyridines ,Mice, Nude ,Kaplan-Meier Estimate ,Biology ,Article ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptor ,PI3K/AKT/mTOR pathway ,Gene knockdown ,Cell growth ,Erythropoietin-producing hepatocellular (Eph) receptor ,Cell migration ,medicine.disease ,Xenograft Model Antitumor Assays ,Head and neck squamous-cell carcinoma ,Tumor Burden ,030104 developmental biology ,Oncology ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Cancer research ,biology.protein ,Female ,RNA Interference ,Signal Transduction - Abstract
Eph proteins have emerged as critical drivers affecting tumor growth and progression in human malignancies. Our The Cancer Genome Atlas (TCGA) data analysis showed that EphB3, a receptor tyrosine kinase, is frequently coamplified with PIK3CA in head and neck squamous cell carcinoma (HNSCC). We therefore hypothesized that EphB3 amplification plays a protumorigenic role in HNSCC and that EphB3 and PIK3CA are cooperating oncogenes that contribute toward its pathogenesis. This hypothesis was not experimentally supported, because EphB3 knockdown failed to alter HNSCC tumor cell growth in vitro or in vivo with an orthotopic model. However, responsiveness of EphB3 knockdown tumors to the PI3K inhibitor, BKM120, was significantly decreased in terms of both tumor growth delay and survival. This is correlated with an increase in prosurvival proteins, S6 and BcL-XL, in the EphB3 shRNA tumors treated with BKM120 compared with controls. We further observed that EphB3 knockdown resulted in increased migration in vitro and increased EMT gene signature in vivo. To explain these results, we examined EphB3 phosphorylation levels in HNSCC at baseline. Although total EphB3 levels were high, we found low phospho-EphB3 levels in HNSCCs. Forced EphB3 phosphorylation with an ephrin-B2–Fc fusion protein resulted in decreased HNSCC migration and cell growth, and enhanced response to BKM120 in vitro. These data collectively indicate that progression of HNSCC selects for low/inhibited EphB3 activity to enhance their survival and migratory abilities and decrease response to PI3K signaling. Therefore, strategies focused on activating EphB3 might be helpful to inhibit tumor growth and enhance sensitivity to PI3K inhibitors in HNSCC. Mol Cancer Ther; 17(9); 2049–59. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
31. Macrophages Promote Growth of Squamous Cancer Independent of T cells
- Author
-
Hongmei Zhou, Christian D. Young, A.A. Strait, K.A. Nguyen, Li Bian, Fanglong Wu, Kyle Nolan, Xiao-Jing Wang, Jing Wang, and Antonio Jimeno
- Subjects
0301 basic medicine ,Angiogenesis ,T-Lymphocytes ,Cell ,Mice, Nude ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immunity ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Squamous cancer ,General Dentistry ,business.industry ,Macrophages ,Research Reports ,medicine.disease ,Mice, Inbred C57BL ,stomatognathic diseases ,030104 developmental biology ,medicine.anatomical_structure ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,Carcinoma, Squamous Cell ,Oral Cancers ,Clodronic acid ,Female ,Clodronic Acid ,business ,medicine.drug - Abstract
Oral cancers, primarily squamous cell carcinomas (SCCs), progress either slowly or aggressively. Here we assessed the role of macrophages in SCC behavior. We used mouse SCC cells derived from tumors harboring a KrasG12D activation mutation and Smad4 deletion in keratin 15–positive stem cells and a human oral SCC cell line, FaDu, which has NRAS amplification and SMAD4 deletion. SCC cells were transplanted into immune-compromised or immune-competent (syngeneic) recipients. After tumors were established, we used clodronate liposomes to ablate macrophages. We found that the number of tumor-associated macrophages (TAMs) was not affected by the presence of T cells but differed considerably among tumors derived from different SCC lines. Clodronate significantly reduced TAMs and splenic macrophages, resulting in reduced SCC volumes. Tumors with clodronate treatment did not show decreased proliferation but did exhibit increased apoptosis and reduced vascular density. FLIP (Fas-associated via death domain-like interleukin 1β–converting enzyme inhibitory protein), an apoptosis inhibitor abundantly produced in tumor cells and TAMs, was reduced in tumor cells of clodronate-treated mice. Reduced FLIP levels correlated with reductions in phosphorylated nuclear NFκB p65 and NFκB inhibitor attenuated FLIP protein levels in SCC cells. Furthermore, TGFβ1 serum levels and pSmad3 were reduced in clodronate-treated mice, but their reductions were insufficient to reverse epithelial-mesenchymal transition or TGFβ-mediated angiogenesis in endothelial cells. Consequently, metastasis was not significantly reduced by macrophage reduction. However, reduced pSmad3 correlated with reduction of its transcriptional target, vascular endothelial growth factor A, in clodronate-treated tumor cells, which correlated with reduced vascular density in clodronate-treated tumors. Taken together, our study revealed that macrophages contribute to SCC expansion through interactions with tumor cells but are dispensable for SCC metastasis. Our study provides novel insights into understanding the contributions and limitations of TAMs in SCC progression.
- Published
- 2019
32. Abstract P6-12-09: Pan-HER, an antibody mixture with antitumor activity against drug-resistant HER2-overexpressing breast cancers with high ERBB ligand expression
- Author
-
Monica Red-Brewer, Luis J. Schwarz, CL Arteaga, Teresa C. Dugger, Monica V. Estrada, AL Guerrero, Melinda E. Sanders, Christian D. Young, L Formisano, Michael Kragh, Katherine E. Hutchinson, Mikkel Winther Pedersen, Ivan D. Horak, and Johan Lantto
- Subjects
Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,Cancer ,Lapatinib ,medicine.disease ,chemistry.chemical_compound ,ErbB Receptors ,Endocrinology ,Oncology ,chemistry ,ErbB ,Trastuzumab ,Internal medicine ,biology.protein ,medicine ,Cancer research ,Pertuzumab ,Antibody ,Growth inhibition ,skin and connective tissue diseases ,business ,medicine.drug - Abstract
Background: Amplification/overexpression of ERBB receptors and/or ligands has been associated with resistance to anti-HER2 therapies. Pan-HER is a mixture of six antibodies targeting each of the ERBB receptors, EGFR, HER2 and HER3, with synergistic pairs of antibodies. Each pair of antibodies simultaneously blocks ligand binding and/or induces target degradation, thus preventing compensatory mechanisms to anti-ERBB therapies. We examined the antitumor activity of Pan-HER against drug-sensitive and -resistant HER2+ breast cancer cells and xenografts. Results: Pan-HER exhibited potent growth inhibitory activity against a panel of HER2+ breast cancer cells (BT474, MDA-453, MDA-361, SUM190, HCC1954, UACC893 and SKBR3). Growth inhibition was associated with internalization and degradation of EGFR, HER2 and HER3. Pan-HER was superior to the combination of trastuzumab/pertuzumab (TP) against HER2+/PIK3CA mutant MDA-361, HCC1954, UACC893 and MDA-453 cells. We next compared the effect of Pan-HER against BT474, HCC1954 and MDA-361 xenografts established in nude mice to that of trastuzumab/lapatinib (TL), TP and T-DM1. All treatments were effective across the panel of xenografts. In mice with MDA-361 tumors, Pan-HER and TP were superior to TL. Immunoblot analysis showed significant downregulation of EGFR, HER2 and HER3 only in tumors treated with Pan-HER. After a complete response, treatment was discontinued. Among mice with BT474 xenografts treated with TP, TL and T-DM1, 25-50% of mice exhibited a tumor recurrence within 50 weeks of follow-up, while no recurrences were registered in mice treated with Pan-HER. Tumors recurring after TP and T-DM1 expressed significantly higher HER3 and P-HER3 protein levels and NRG1 mRNA levels. HCC1954 xenografts recurring after T-DM1 also overexpressed NRG1 mRNA compared to tumors before therapy. We next examined the effect of Pan-HER against trastuzumab-resistant HR6 (BT474) cells (Ritter et al. CCR 2007) and HCC1954 and UACC893 cells with acquired resistance to T-DM1 (TDR; IC50 >5-, >6- and 600-fold in HR6, UACC893-TDR and HCC1954-TDR cells, respectively, vs. parental cells). All T-DM1-resistant cells expressed significantly higher HER3 and P-HER3 protein levels and NRG1 mRNA and protein levels. Treatment with the HER3 neutralizing antibody LJM716 resensitized HR6 and HCC1954-TDR cells to T-DM1, suggesting a causal association between the NRG1-HER3 axis and drug resistance. Mice with HR6 tumors were treated with Pan-HER, TL, TP and T-DM1. Only Pan-HER arrested HR6 tumor growth and downregulated EGFR, HER2, HER3, P-HER3 and P-AKT. Finally, HCC1954-TDR tumors rapidly grew in vivo despite treatment with T-DM1. Administration of Pan-HER to mice bearing HCC1954-TDR xenografts growing in the presence of T-DM1, induced rapid tumor regressions. Conclusions: These data suggest that multitarget therapeutic interventions, such as Pan-HER, which simultaneously remove and/or block all ERBB receptors and ligands, are a feasible and effective approach against HER2-overexpressing cancers both sensitive and resistant to anti-HER2 therapies. Citation Format: Schwarz LJ, Hutchinson KE, Estrada MV, Sanders ME, Dugger TC, Formisano L, Guerrero AL, Red-Brewer M, Young CD, Lantto J, Pedersen MW, Kragh M, Horak ID, Arteaga CL. Pan-HER, an antibody mixture with antitumor activity against drug-resistant HER2-overexpressing breast cancers with high ERBB ligand expression [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P6-12-09.
- Published
- 2017
- Full Text
- View/download PDF
33. Rictor/mTORC2 Drives Progression and Therapeutic Resistance of HER2-Amplified Breast Cancers
- Author
-
Monica V. Estrada, Dana M. Brantley-Sieders, Bayley A. Jones, Meghan Morrison Joly, Rebecca S. Cook, Violeta Sanchez, Michelle M. Williams, William J. Muller, Donna J. Hicks, Christian D. Young, Dos D. Sarbassov, and Brent N. Rexer
- Subjects
0301 basic medicine ,Cancer Research ,Receptor, ErbB-2 ,Blotting, Western ,Mice, Nude ,Breast Neoplasms ,Kaplan-Meier Estimate ,Mechanistic Target of Rapamycin Complex 2 ,mTORC1 ,Biology ,Lapatinib ,mTORC2 ,Article ,Mice ,03 medical and health sciences ,Breast cancer ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Mice, Inbred BALB C ,TOR Serine-Threonine Kinases ,Cancer ,medicine.disease ,Rapamycin-Insensitive Companion of mTOR Protein ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Tissue Array Analysis ,Tumor progression ,Multiprotein Complexes ,Disease Progression ,Cancer research ,Heterografts ,Female ,Carrier Proteins ,Signal Transduction ,medicine.drug - Abstract
HER2 overexpression drives Akt signaling and cell survival and HER2-enriched breast tumors have a poor outcome when Akt is upregulated. Akt is activated by phosphorylation at T308 via PI3K and S473 via mTORC2. The importance of PI3K-activated Akt signaling is well documented in HER2-amplified breast cancer models, but the significance of mTORC2-activated Akt signaling in this setting remains uncertain. We report here that the mTORC2 obligate cofactor Rictor is enriched in HER2-amplified samples, correlating with increased phosphorylation at S473 on Akt. In invasive breast cancer specimens, Rictor expression was upregulated significantly compared with nonmalignant tissues. In a HER2/Neu mouse model of breast cancer, genetic ablation of Rictor decreased cell survival and phosphorylation at S473 on Akt, delaying tumor latency, penetrance, and burden. In HER2-amplified cells, exposure to an mTORC1/2 dual kinase inhibitor decreased Akt-dependent cell survival, including in cells resistant to lapatinib, where cytotoxicity could be restored. We replicated these findings by silencing Rictor in breast cancer cell lines, but not silencing the mTORC1 cofactor Raptor (RPTOR). Taken together, our findings establish that Rictor/mTORC2 signaling drives Akt-dependent tumor progression in HER2-amplified breast cancers, rationalizing clinical investigation of dual mTORC1/2 kinase inhibitors and developing mTORC2-specific inhibitors for use in this setting. Cancer Res; 76(16); 4752–64. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
34. Abstract 6161: PARP inhibition enhances radiotherapy of SMAD4 deficient human head and neck squamous cell carcinomas in experimental models
- Author
-
David Raben, Sana D. Karam, Ariel L. Hernandez, Li Bian, Christian D. Young, and Xiao-Jing Wang
- Subjects
Cancer Research ,Gene knockdown ,animal structures ,integumentary system ,business.industry ,DNA damage ,medicine.medical_treatment ,Cell ,Cancer ,medicine.disease ,Head and neck squamous-cell carcinoma ,digestive system diseases ,Olaparib ,Radiation therapy ,chemistry.chemical_compound ,medicine.anatomical_structure ,Oncology ,chemistry ,embryonic structures ,PARP inhibitor ,Cancer research ,medicine ,business ,neoplasms - Abstract
Purpose: SMAD4 loss causes genomic instability and the initiation/progression of head and neck squamous cell carcinoma (HNSCC). Here, we study if SMAD4 loss sensitizes HNSCCs to olaparib (PARP inhibitor) in combination with radiotherapy (RT). Experimental Design: We analyzed HNSCC TCGA data for SMAD4 expression in association with FANC/BRCA family gene expression. Human HNSCC cell lines were screened for sensitivity to olaparib. Isogenic HNSCC cell lines were generated to restore or reduce SMAD4 expression and treated with olaparib, radiation, or the combination. HNSCC pretreatment specimens from a Phase I trial investigating olaparib were analyzed. Results: SMAD4 levels correlated with levels of FANC/BRCA genes in HNSCC. HNSCC cell lines with SMAD4 homozygous deletion were sensitive to olaparib. In vivo, olaparib or RT monotherapy reduced tumor volumes in SMAD4 mutant but not SMAD4 positive tumors. Olaparib with RT dual therapy sustained tumor volume reduction in SMAD4 deficient (mutant or knockdown) xenografts, which exhibited increased DNA damage and cell death compared to vehicle treated tumors. In vitro, olaparib or in combination with radiation caused lower clonogenic survival, more DNA damage-associated cell death and less proliferation in SMAD4 deficient cells than in SMAD4-positive (endogenous SMAD4 or transduced SMAD4) cells. Applicable to clinic, 5 out of 6 SMAD4-negative HNSCCs and 4 out of 8 SMAD4-positive HNSCCs responded to a standard treatment plus olaparib in a Phase I clinical trial, and SMAD4 protein levels inversely correlated with DNA damage. Conclusion: SMAD4 levels are causal in determining sensitivity to PARP inhibition in combination with RT in HNSCCs. Citation Format: Christian D. Young, Ariel L. Hernandez, Li Bian, Sana D. Karam, David Raben, Xiao-Jing Wang. PARP inhibition enhances radiotherapy of SMAD4 deficient human head and neck squamous cell carcinomas in experimental models [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6161.
- Published
- 2020
- Full Text
- View/download PDF
35. Erratum for the Research Article: 'Genomic profiling of ER + breast cancers after short-term estrogen suppression reveals alterations associated with endocrine resistance' by J. M. Giltnane, K. E. Hutchinson, T. P. Stricker, L. Formisano, C. D. Young, M. V. Estrada, M. J. Nixon, L. Du, V. Sanchez, P. G. Ericsson, M. G. Kuba, M. E. Sanders, X. J. Mu, E. M. Van Allen, N. Wagle, I. A. Mayer, V. Abramson, H. Gόmez, M. Rizzo, W. Toy, S. Chandarlapaty, E. L. Mayer, J. Christiansen, D. Murphy, K. Fitzgerald, K. Wang, J. S. Ross, V. A. Miller, P. J. Stephens, R. Yelensky, L. Garraway, Y. Shyr, I. Meszoely, J. M. Balko, C. L. Arteaga
- Author
-
Ingrid M. Meszoely, Melinda E. Sanders, Christian D. Young, Violeta Sanchez, Yu Shyr, Liping Du, Katherine E. Hutchinson, Ingrid A. Mayer, Phillip J. Stephens, CL Arteaga, Paula Gonzalez Ericsson, L. A. Garraway, Maria G. Kuba, Vandana G. Abramson, Monica V. Estrada, Thomas Stricker, Vincent A. Miller, Jeffrey S. Ross, Danielle Murphy, Sarat Chandarlapaty, Monica Rizzo, Weiyi Toy, E. M. Van Allen, Kerry Fitzgerald, Roman Yelensky, N Wagle, Kai Wang, Jason Christiansen, Xinmeng Jasmine Mu, Erica L. Mayer, Justin M. Balko, Mellissa J. Nixon, Jennifer M. Giltnane, Luigi Formisano, and H. Gόmez
- Subjects
business.industry ,Estrogen ,medicine.drug_class ,Endocrine resistance ,Translational medicine ,Medicine ,Research article ,General Medicine ,business ,Bioinformatics ,Term (time) - Published
- 2019
- Full Text
- View/download PDF
36. Smad7 Promotes Healing of Radiotherapy-Induced Oral Mucositis without Compromising Oral Cancer Therapy in a Xenograft Mouse Model
- Author
-
Danfeng Du, Melanie A. Blevins, David Raben, Li Bian, Jingjing Luo, Dongyan Wang, Christian D. Young, Chao Liang, Hongmei Zhou, Rui Zhao, Fanglong Wu, Barry Holwerda, and Xiao-Jing Wang
- Subjects
0301 basic medicine ,Mucositis ,Cancer Research ,medicine.medical_treatment ,Fluorescent Antibody Technique ,Inflammation ,Models, Biological ,Article ,Smad7 Protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Medicine ,Cytotoxic T cell ,Animals ,Humans ,Oral mucosa ,Stomatitis ,Wound Healing ,integumentary system ,business.industry ,Cancer ,medicine.disease ,Immunohistochemistry ,Radiation therapy ,stomatognathic diseases ,Disease Models, Animal ,Radiation Injuries, Experimental ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Heterografts ,Mouth Neoplasms ,medicine.symptom ,business ,Biomarkers ,DNA Damage ,Signal Transduction - Abstract
Purpose: We previously reported preventive and therapeutic effects of Smad7, a multifunctional protein, on radiotherapy (RT)-induced mucositis in mice without promoting human oral cancer cell survival or migration in vitro. The current study aims to determine whether a Smad7-based biologic can treat existing oral mucositis during radiotherapy for oral cancer and whether this treatment compromises RT-induced cancer cell killing in neighboring oral cancer. Experimental Design: We transplanted human oral cancer cells into the tongues of mice and applied craniofacial irradiation to simultaneously kill tumor cells and induce oral mucositis, thus modeling RT and mucositis in oral cancer patients. We topically applied a recombinant human Smad7 protein fused with the cell-penetrating Tat tag (Tat-Smad7) to the oral mucosa of tumor-bearing mice post RT when oral mucositis began to develop. Results: Topically applied Tat-Smad7 penetrated cells in both the oral mucosa and oral cancer, attenuating TGFβ and NF-κB signaling as well as inflammation at both sites. Tat-Smad7 treatment alleviated oral mucositis with reductions in DNA damage and apoptosis in keratinocytes, but increased keratinocyte proliferation compared with vehicle-treated mucositis lesions. In contrast, adjacent oral cancer exposed to Tat-Smad7 did not show alterations in proliferation or direct DNA damage, but showed increased oxidative stress damage and apoptosis compared with tumors treated with vehicle. Conclusions: Our results suggest that short-course Tat-Smad7 application to oral mucositis promotes its healing but does not compromise the cytotoxic effect of RT on oral cancer and has context-specific effects on oral mucosa versus oral cancer.
- Published
- 2018
37. Mouse Models for Studying Oral Cancer: Impact in the Era of Cancer Immunotherapy
- Author
-
Christian D. Young, Jingjing Luo, Xiao-Jing Wang, and Hongmei Zhou
- Subjects
0301 basic medicine ,Stromal cell ,medicine.medical_treatment ,Reviews ,medicine.disease_cause ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Immunity ,medicine ,Animals ,General Dentistry ,Mouth neoplasm ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,stomatognathic diseases ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Mouth Neoplasms ,Carcinogenesis ,business - Abstract
Model systems for oral cancer research have progressed from tumor epithelial cell cultures to in vivo systems that mimic oral cancer genetics, pathological characteristics, and tumor-stroma interactions of oral cancer patients. In the era of cancer immunotherapy, it is imperative to use model systems to test oral cancer prevention and therapeutic interventions in the presence of an immune system and to discover mechanisms of stromal contributions to oral cancer carcinogenesis. Here, we review in vivo mouse model systems commonly used for studying oral cancer and discuss the impact these models are having in advancing basic mechanisms, chemoprevention, and therapeutic intervention of oral cancer while highlighting recent discoveries concerning the role of immune cells in oral cancer. Improvements to in vivo model systems that highly recapitulate human oral cancer hold the key to identifying features of oral cancer initiation, progression, and invasion as well as molecular and cellular targets for prevention, therapeutic response, and immunotherapy development.
- Published
- 2018
38. Adenoviral vectors transduce alveolar macrophages in lung cancer models
- Author
-
Gregory Verzosa, Stephen P. Malkoski, Howard Li, James DeGregori, Raphael A. Nemenoff, Christian D. Young, Kyle Nolan, Jing Huang, Hannah Scarborough, Joanna M. Poczobutt, and Darinee Tippimanchai
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Immunology ,Cre recombinase ,Tumor initiation ,Biology ,lcsh:RC254-282 ,Viral vector ,Green fluorescent protein ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,Immune system ,medicine ,tumor microenvironment ,Immunology and Allergy ,Lung cancer ,Original Research ,Tumor microenvironment ,alveolar macrophages ,adenovirus ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,3. Good health ,lung cancer ,Editorial ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:RC581-607 - Abstract
Adenoviral vectors expressing Cre recombinase are commonly used to initiate tumor formation in murine lung cancer models. While these vectors are designed to target genetic recombination to lung epithelial cells, adenoviruses can infect additional cell types that potentially influence tumor development. Our goal was to explore the consequences of adenoviral-mediated alveolar macrophage (AM) transduction in a Kras-initiated lung tumor model. As expected, treatment of animals harboring the KrasLSL-G12D allele and an inducible green fluorescence protein (GFP) tracking allele with an adenoviral vector expressing Cre recombinase under the control of the cytomegalovirus (CMV) promoter (Ad5-CMV-Cre), caused GFP-positive lung adenocarcinomas. Surprisingly, however, up to 70% of the total GFP+ cells were AM, and GFP+ AM could be detected 6 months after tumor initiation, and transduced AM demonstrated Kras activation and increased proliferation. In contrast, recombination was not detected in other immune cell populations and AM recombination could be eliminated by tumor initiation with an adenovirus expressing Cre recombinase under the control of the surfactant protein C (SPC) promoter. In addition, AM isolated from KrasLSL-G12D animals and transduced by Ad5-CMV-Cre ex vivo displayed prolonged survival in vitro and increased the growth of murine lung adenocarcinoma CMT/167 cells when co-injected in an orthotopic flank model. Given the importance of the immune system in tumor development and progression, inadvertent AM transduction by Ad5-CMV-Cre merits careful consideration during lung cancer model selection particularly if studies evaluating the tumor-immune interactions are planned.
- Published
- 2018
- Full Text
- View/download PDF
39. Small molecule inhibitor of the bone morphogenetic protein pathway DMH1 reduces ovarian cancer cell growth
- Author
-
Laura D. Hover, Philip Owens, Andrew J. Wilson, Christian D. Young, Harold L. Moses, Neil E. Bhola, Dineo Khabele, and Charles C. Hong
- Subjects
Cancer Research ,animal structures ,endocrine system diseases ,Receptor expression ,Antineoplastic Agents ,Bone morphogenetic protein ,Article ,Disease-Free Survival ,Ovarian tumor ,Spheroids, Cellular ,Tumor Cells, Cultured ,medicine ,Humans ,Bone morphogenetic protein receptor ,Cell Proliferation ,Ovarian Neoplasms ,biology ,Cancer ,Bone Morphogenetic Protein Receptors ,Transforming growth factor beta ,medicine.disease ,BMPR2 ,Gene Expression Regulation, Neoplastic ,Oncology ,Drug Resistance, Neoplasm ,Bone Morphogenetic Proteins ,embryonic structures ,Quinolines ,Cancer research ,biology.protein ,Pyrazoles ,Female ,Cisplatin ,Ovarian cancer ,Signal Transduction - Abstract
The bone morphogenetic protein (BMP) pathway belonging to the Transforming Growth Factor beta (TGFβ) family of secreted cytokines/growth factors is an important regulator of cancer. BMP ligands have been shown to play both tumor suppressive and promoting roles in human cancers. We have found that BMP ligands are amplified in human ovarian cancers and that BMP receptor expression correlates with poor progression-free-survival (PFS). Furthermore, active BMP signaling has been observed in human ovarian cancer tissue. We also determined that ovarian cancer cell lines have active BMP signaling in a cell autonomous fashion. Inhibition of BMP signaling with a small molecule receptor kinase antagonist is effective at reducing ovarian tumor sphere growth. Furthermore, BMP inhibition can enhance sensitivity to Cisplatin treatment and regulates gene expression involved in platinum resistance in ovarian cancer. Overall, these studies suggest targeting the BMP pathway as a novel source to enhance chemo-sensitivity in ovarian cancer.
- Published
- 2015
- Full Text
- View/download PDF
40. Abstract PD5-8: HER2/PIK3CAH1047R transgenic mammary tumors develop acquired resistance to triple therapy with trastuzumab, pertuzumab and PI3K inhibitors via multiple mechanisms
- Author
-
Carlos L. Arteaga, Thomas Stricker, Rebecca S. Cook, Kirsten M Farrar, Monica Red Brewer, Benjamin Bulen, Christian D. Young, and Ariella B. Hanker
- Subjects
Genetically modified mouse ,Cancer Research ,medicine.medical_specialty ,business.industry ,Transgene ,Cancer ,P110α ,medicine.disease ,Breast cancer ,Endocrinology ,Oncology ,Trastuzumab ,Internal medicine ,Cancer research ,Medicine ,Pertuzumab ,skin and connective tissue diseases ,business ,neoplasms ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
HER2 amplification and activating mutations in PIK3CA, the gene encoding the p110α subunit of PI3K, often co-occur in breast cancer. We generated a transgenic mouse model of HER2-overexpressing (HER2+), PIK3CAH1047R-mutant breast cancer. In these mice, PIK3CAH1047R accelerates HER2-mediated mammary epithelial transformation and metastatic progression, confers stem cell-like properties to HER2-overexpressing cancers and generates resistance to the combination of trastuzumab and pertuzumab (Hanker et al. PNAS 2013). HER2+/PIK3CA tumor growth was inhibited by treatment with the HER2 antibodies trastuzumab and pertuzumab in combination with the pan-PI3K inhibitor BKM120 (TPB). We sought to discover mechanisms of acquired resistance to the triple therapy by long-term treatment of established HER2+/PIK3CA tumors. We used tumor transplants derived from two HER2+/PIK3CA transgenic mice, #564 and #635. Tumor transplants from model 564 were initially growth inhibited by TPB, but did not regress. A subset of 564 transplants (3/11) resumed growth in the presence of continuous TPB therapy. All transplants (n=9) from model 635 regressed to a volume of 5 months) in the presence of the three drugs. Similar to the TPB-resistant tumors, P-S6 was no longer inhibited following TPB treatment in the resistant cell lines. Treatment with the TORC1/2 inhibitor MLN0128 abolished levels of P-S6 in HER2+/PIK3CAH1047R tumors. Combined treatment with MLN0128 and TPB inhibited growth of the drug-resistant tumors. Interestingly, Both TPB-resistant HER2+/PIK3CAH1047R tumor lines displayed resistance to the antibody-drug conjugate trastuzumab-DM1 (T-DM1) in vitro and in vivo, despite maintenance of HER2 overexpression. In addition, HCC1954 cells selected for resistance to TPB in culture were 66-fold less sensitive to T-DM1 than parental cells, despite maintaining equal levels of HER2 by western blot. These data suggest that multiple mechanisms may contribute to resistance to dual HER2 and PI3K blockade, including re-activation of mTOR signaling. We speculate that a similar heterogeneity of resistance mechanisms may occur in HER2+/PIK3CA-mutant metastases in patients. Citation Format: Ariella B Hanker, Benjamin Bulen, Monica Red Brewer, Christian D Young, Kirsten M Farrar, Rebecca S Cook, Thomas P Stricker, Carlos L Arteaga. HER2/PIK3CAH1047R transgenic mammary tumors develop acquired resistance to triple therapy with trastuzumab, pertuzumab and PI3K inhibitors via multiple mechanisms [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr PD5-8.
- Published
- 2015
- Full Text
- View/download PDF
41. Genomic profiling of ER+breast cancers after short-term estrogen suppression reveals alterations associated with endocrine resistance
- Author
-
Nikhil Wagle, Weiyi Toy, Monica Rizzo, Eliezer M. Van Allen, Thomas Stricker, Jason Christiansen, Vincent A. Miller, Violeta Sanchez, Maria G. Kuba, Xinmeng J. Mu, Carlos L. Arteaga, Jennifer M. Giltnane, Erica L. Mayer, Ingrid M. Meszoely, Liping Du, Henry Gόmez, Kai Wang, Phillip J. Stephens, Monica V. Estrada, Paula Gonzalez Ericsson, Vandana G. Abramson, Kerry Fitzgerald, Danielle Murphy, Levi A. Garraway, Justin M. Balko, Mellissa J. Nixon, Luigi Formisano, Roman Yelensky, Yu Shyr, Ingrid A. Mayer, Melinda E. Sanders, Christian D. Young, Katherine E. Hutchinson, Jeffrey S. Ross, Sarat Chandarlapaty, Giltnane, Jennifer M., Hutchinson, Katherine E., Stricker, Thomas P., Formisano, Luigi, Young, Christian D., Estrada, Monica V., Nixon, Mellissa J., Du, Liping, Sanchez, Violeta, Ericsson, Paula Gonzalez, Kuba, Maria G., Sanders, Melinda E., Mu, Xinmeng J., Van Allen, Eliezer M., Wagle, Nikhil, Mayer, Ingrid A., Abramson, Vandana, Gómez, Henry, Rizzo, Monica, Toy, Weiyi, Chandarlapaty, Sarat, Mayer, Erica L., Christiansen, Jason, Murphy, Danielle, Fitzgerald, Kerry, Wang, Kai, Ross, Jeffrey S., Miller, Vincent A., Stephens, Phillip J., Yelensky, Roman, Garraway, Levi, Shyr, Yu, Meszoely, Ingrid, Balko, Justin M., and Arteaga, Carlos L.
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Aromatase inhibitor ,biology ,Cyclin-dependent kinase 4 ,medicine.drug_class ,Letrozole ,Oncology, drug resistance, Estrogen Receptor, Breast Cancer ,General Medicine ,Cell cycle ,03 medical and health sciences ,030104 developmental biology ,Cyclin D1 ,Endocrinology ,Estrogen ,Internal medicine ,biology.protein ,medicine ,Cancer research ,Cyclin-dependent kinase 6 ,Estrogen receptor alpha ,medicine.drug - Abstract
Inhibition of proliferation in estrogen receptor-positive (ER+) breast cancers after short-term antiestrogen therapy correlates with long-term patient outcome. We profiled 155 ER+/human epidermal growth factor receptor 2-negative (HER2-) early breast cancers from 143 patients treated with the aromatase inhibitor letrozole for 10 to 21 days before surgery. Twenty-one percent of tumors remained highly proliferative, suggesting that these tumors harbor alterations associated with intrinsic endocrine therapy resistance. Whole-exome sequencing revealed a correlation between 8p11-12 and 11q13 gene amplifications, including FGFR1 and CCND1, respectively, and high Ki67. We corroborated these findings in a separate cohort of serial pretreatment, postneoadjuvant chemotherapy, and recurrent ER+ tumors. Combined inhibition of FGFR1 and CDK4/6 reversed antiestrogen resistance in ER+FGFR1/CCND1 coamplified CAMA1 breast cancer cells. RNA sequencing of letrozole-treated tumors revealed the existence of intrachromosomal ESR1 fusion transcripts and increased expression of gene signatures indicative of enhanced E2F-mediated transcription and cell cycle processes in cancers with high Ki67. These data suggest that short-term preoperative estrogen deprivation followed by genomic profiling can be used to identify druggable alterations that may cause intrinsic endocrine therapy resistance.
- Published
- 2017
- Full Text
- View/download PDF
42. Two distinct mTORC2-dependent pathways converge on Rac1 to drive breast cancer metastasis
- Author
-
Dana M. Brantley-Sieders, Christian D. Young, Dos D. Sarbassov, Meghan Morrison Joly, Rebecca S. Cook, Violeta Sanchez, William J. Muller, Donna J. Hicks, Michelle M. Williams, and Bayley A. Jones
- Subjects
0301 basic medicine ,Oncology ,rac1 GTP-Binding Protein ,Receptor, ErbB-2 ,mTORC2 ,Metastasis ,RhoGDI2 ,Mice ,Breast cancer ,rho Guanine Nucleotide Dissociation Inhibitor beta ,Cell Movement ,Neoplasm Metastasis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Prognosis ,mTOR ,Conditional knockout ,Heterografts ,Female ,Signal Transduction ,Research Article ,medicine.medical_specialty ,Motility ,RAC1 ,Breast Neoplasms ,Mice, Transgenic ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,lcsh:RC254-282 ,Rictor ,03 medical and health sciences ,Protein kinase C ,Internal medicine ,Cell Line, Tumor ,HER2 ,medicine ,Animals ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Neoplasm Staging ,Akt ,Gene Amplification ,medicine.disease ,Rac ,Disease Models, Animal ,030104 developmental biology ,Rapamycin-Insensitive Companion of mTOR Protein ,Mouse mammary tumor ,Tumor progression ,Cancer cell ,Cancer research ,Proto-Oncogene Proteins c-akt - Abstract
Background The importance of the mTOR complex 2 (mTORC2) signaling complex in tumor progression is becoming increasingly recognized. HER2-amplified breast cancers use Rictor/mTORC2 signaling to drive tumor formation, tumor cell survival and resistance to human epidermal growth factor receptor 2 (HER2)-targeted therapy. Cell motility, a key step in the metastatic process, can be activated by mTORC2 in luminal and triple negative breast cancer cell lines, but its role in promoting metastases from HER2-amplified breast cancers is not yet clear. Methods Because Rictor is an obligate cofactor of mTORC2, we genetically engineered Rictor ablation or overexpression in mouse and human HER2-amplified breast cancer models for modulation of mTORC2 activity. Signaling through mTORC2-dependent pathways was also manipulated using pharmacological inhibitors of mTOR, Akt, and Rac. Signaling was assessed by western analysis and biochemical pull-down assays specific for Rac-GTP and for active Rac guanine nucleotide exchange factors (GEFs). Metastases were assessed from spontaneous tumors and from intravenously delivered tumor cells. Motility and invasion of cells was assessed using Matrigel-coated transwell assays. Results We found that Rictor ablation potently impaired, while Rictor overexpression increased, metastasis in spontaneous and intravenously seeded models of HER2-overexpressing breast cancers. Additionally, migration and invasion of HER2-amplified human breast cancer cells was diminished in the absence of Rictor, or upon pharmacological mTOR kinase inhibition. Active Rac1 was required for Rictor-dependent invasion and motility, which rescued invasion/motility in Rictor depleted cells. Rictor/mTORC2-dependent dampening of the endogenous Rac1 inhibitor RhoGDI2, a factor that correlated directly with increased overall survival in HER2-amplified breast cancer patients, promoted Rac1 activity and tumor cell invasion/migration. The mTORC2 substrate Akt did not affect RhoGDI2 dampening, but partially increased Rac1 activity through the Rac-GEF Tiam1, thus partially rescuing cell invasion/motility. The mTORC2 effector protein kinase C (PKC)α did rescue Rictor-mediated RhoGDI2 downregulation, partially rescuing Rac-guanosine triphosphate (GTP) and migration/motility. Conclusion These findings suggest that mTORC2 uses two coordinated pathways to activate cell invasion/motility, both of which converge on Rac1. Akt signaling activates Rac1 through the Rac-GEF Tiam1, while PKC signaling dampens expression of the endogenous Rac1 inhibitor, RhoGDI2. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0868-8) contains supplementary material, which is available to authorized users.
- Published
- 2017
43. Efferocytosis produces a prometastatic landscape during postpartum mammary gland involution
- Author
-
Donna J. Hicks, Andrew Williams, Rebecca S. Cook, Meghan M. Morrison, Jiyeon Lim, Michelle M. Williams, Christian D. Young, Jamie C. Stanford, Justin M. Balko, David B. Vaught, Philip Owens, Debra A. Tonetti, Dana M. Brantley-Sieders, and H. Shelton Earp
- Subjects
Male ,Lung Neoplasms ,Gene Expression ,Apoptosis ,Mice, Transgenic ,Biology ,Metastasis ,Mammary Glands, Animal ,Phagocytosis ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Involution (medicine) ,Efferocytosis ,Mammary gland involution ,Tumor microenvironment ,Mammary tumor ,c-Mer Tyrosine Kinase ,Postpartum Period ,Mammary Neoplasms, Experimental ,Receptor Protein-Tyrosine Kinases ,General Medicine ,MERTK ,medicine.disease ,Coculture Techniques ,Tumor Burden ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,MCF-7 Cells ,Cancer research ,Cytokines ,Female ,Neoplasm Transplantation ,Postpartum period ,Signal Transduction ,Research Article - Abstract
Breast cancers that occur in women 2–5 years postpartum are more frequently diagnosed at metastatic stages and correlate with poorer outcomes compared with breast cancers diagnosed in young, premenopausal women. The molecular mechanisms underlying the malignant severity associated with postpartum breast cancers (ppBCs) are unclear but relate to stromal wound-healing events during postpartum involution, a dynamic process characterized by widespread cell death in milk-producing mammary epithelial cells (MECs). Using both spontaneous and allografted mammary tumors in fully immune–competent mice, we discovered that postpartum involution increases mammary tumor metastasis. Cell death was widespread, not only occurring in MECs but also in tumor epithelium. Dying tumor cells were cleared through receptor tyrosine kinase MerTK–dependent efferocytosis, which robustly induced the transcription of genes encoding wound-healing cytokines, including IL-4, IL-10, IL-13, and TGF-β. Animals lacking MerTK and animals treated with a MerTK inhibitor exhibited impaired efferocytosis in postpartum tumors, a reduction of M2-like macrophages but no change in total macrophage levels, decreased TGF-β expression, and a reduction of postpartum tumor metastasis that was similar to the metastasis frequencies observed in nulliparous mice. Moreover, TGF-β blockade reduced postpartum tumor metastasis. These data suggest that widespread cell death during postpartum involution triggers efferocytosis-induced wound-healing cytokines in the tumor microenvironment that promote metastatic tumor progression.
- Published
- 2014
- Full Text
- View/download PDF
44. Response to Letter to the Editor: 'Macrophages Promote Growth of Squamous Cancer Independent of T Cells'
- Author
-
Xiao-Jing Wang, Fanglong Wu, Christian D. Young, and Hongmei Zhou
- Subjects
Mouth neoplasm ,Text mining ,Letter to the editor ,business.industry ,Carcinoma ,Cancer research ,MEDLINE ,Medicine ,Squamous cancer ,business ,medicine.disease ,General Dentistry - Published
- 2019
- Full Text
- View/download PDF
45. Abstract LB-313: Precancerous keratinocytes promote neighboring HNSCC cancer stem cell-like properties and PI3K inhibitor insensitivity via EGFR
- Author
-
Khoa A. Nguyen, Christian D. Young, Madison J. Keith, and Xiao-Jing Wang
- Subjects
Cancer Research ,Tumor microenvironment ,biology ,Cell ,Cancer ,medicine.disease ,Head and neck squamous-cell carcinoma ,Receptor tyrosine kinase ,medicine.anatomical_structure ,Oncology ,Cancer stem cell ,CDKN2A ,Cancer research ,biology.protein ,medicine ,PI3K/AKT/mTOR pathway - Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer in the world and HNSCC patients have relatively few effective treatment options. Genomic analyses of HNSCCs have demonstrated that genes encoding components of the phosphoinositide 3-kinase (PI3K) signaling pathway are amongst the most frequently mutated and amplified. Because PI3K drives cancer cell proliferation and survival, PI3K inhibitors are being evaluated as anti-cancer therapeutic agents. However, the preclinical effectiveness of PI3K inhibitors has not necessarily translated to remarkable benefit in cancer patients. A large proportion of HNSCC is associated with tobacco and alcohol consumption that induce genetic lesions in a field of mucosa, the “precancerous field”. Carcinomas often arise from this precancerous field. Thus, we sought to determine how precancerous keratinocytes influence HNSCC proliferation, cancer stem cell (CSC) maintenance and response to PI3K inhibitors. The keratinocyte cell line NOK harbors CDKN2A promoter methylation and TP53 mutation (two frequent genetic events in HNSCC), but does not form tumors. Thus, we used NOK cells as a model of precancerous keratinocytes and examined their effect on two different HNSCC cell lines, CAL27 and UMSCC1. NOK cells promoted HNSCC proliferation and PI3K inhibitor resistance in co-culture assays. Similarly, NOK cell conditioned media applied to recipient HNSCC cells promoted proliferation, PI3K inhibitor resistance and CSC phenotypes. We utilized SOMAscan, a targeted proteomics platform, to determine the relative levels of >1,300 analytes in the media conditioned by NOK cells and control cells in the presence and absence of PI3K inhibitor. These results demonstrated that NOK cells release abundant levels of ligands that activate EGFR and FGFR, two receptor tyrosine kinases with oncogenic activity. Inhibition of EGFR, but not FGFR, blunted PI3K inhibitor resistance and CSC phenotypes induced by NOK cells. Our results demonstrate that precancerous keratinocytes directly support neighboring HNSCC by activating EGFR. Importantly, PI3K inhibitor sensitivity is not necessarily a cancer cell-intrinsic property and the tumor microenvironment impacts therapeutic response and supports CSCs. Additionally, combined inhibition of EGFR with PI3K inhibitor diminishes EGFR activation induced by PI3K inhibitor and potently inhibits cancer cell proliferation and CSC maintenance. We are currently establishing mouse models of the oral precancerous field and testing its influence on HNSCC and immune cell function. Citation Format: Khoa A. Nguyen, Madison J. Keith, Xiao-Jing Wang, Christian D. Young. Precancerous keratinocytes promote neighboring HNSCC cancer stem cell-like properties and PI3K inhibitor insensitivity via EGFR [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-313.
- Published
- 2019
- Full Text
- View/download PDF
46. Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets
- Author
-
Maureen T. Cronin, Maria G. Kuba, Carlos L. Arteaga, Joseph A. Pinto, Roman Yelensky, Jeffrey S. Ross, Phillip J. Stephens, Justin M. Balko, Henry L. Gomez, Luis J. Schwarz, Brian D. Lehmann, Jennifer A. Pietenpol, Jennifer M. Giltnane, Philip Owens, Kai Wang, Vincent A. Miller, Gary A. Palmer, Rebecca S. Cook, Violeta Sanchez, Franco Doimi, Melinda E. Sanders, Christian D. Young, Andrei Goga, Dai Horiuchi, Richard Kurupi, Joshua A. Bauer, and Preston D. Moore
- Subjects
Oncology ,Neoplasm, Residual ,medicine.medical_treatment ,Genes, myc ,Drug Resistance ,Triple Negative Breast Neoplasms ,Bioinformatics ,Targeted therapy ,Antineoplastic Combined Chemotherapy Protocols ,Gene duplication ,Cluster Analysis ,2.1 Biological and endogenous factors ,Neoplasm ,Aetiology ,Neoadjuvant therapy ,Cancer ,Tumor ,myc ,Prognosis ,Neoadjuvant Therapy ,Treatment Outcome ,Residual ,Female ,Adjuvant ,Biotechnology ,medicine.medical_specialty ,DNA Copy Number Variations ,Oncology and Carcinogenesis ,Biology ,Article ,Cell Line ,Clinical Research ,Cell Line, Tumor ,Internal medicine ,Breast Cancer ,Genetics ,medicine ,Humans ,Chemotherapy ,Gene Expression Profiling ,Human Genome ,Gene Amplification ,medicine.disease ,Gene expression profiling ,Ki-67 Antigen ,Good Health and Well Being ,Genes ,Drug Resistance, Neoplasm ,Myeloid Cell Leukemia Sequence 1 Protein - Abstract
Neoadjuvant chemotherapy (NAC) induces a pathologic complete response (pCR) in approximately 30% of patients with triple-negative breast cancers (TNBC). In patients lacking a pCR, NAC selects a subpopulation of chemotherapy-resistant tumor cells. To understand the molecular underpinnings driving treatment-resistant TNBCs, we performed comprehensive molecular analyses on the residual disease of 74 clinically defined TNBCs after NAC, including next-generation sequencing (NGS) on 20 matched pretreatment biopsies. Combined NGS and digital RNA expression analysis identified diverse molecular lesions and pathway activation in drug-resistant tumor cells. Ninety percent of the tumors contained a genetic alteration potentially treatable with a currently available targeted therapy. Thus, profiling residual TNBCs after NAC identifies targetable molecular lesions in the chemotherapy-resistant component of the tumor, which may mirror micrometastases destined to recur clinically. These data can guide biomarker-driven adjuvant studies targeting these micrometastases to improve the outcome of patients with TNBC who do not respond completely to NAC. Significance: This study demonstrates the spectrum of genomic alterations present in residual TNBC after NAC. Because TNBCs that do not achieve a CR after NAC are likely to recur as metastatic disease at variable times after surgery, these alterations may guide the selection of targeted therapies immediately after mastectomy before these metastases become evident. Cancer Discov; 4(2); 232–45. ©2013 AACR. This article is highlighted in the In This Issue feature, p. 131
- Published
- 2014
- Full Text
- View/download PDF
47. Mutant PIK3CA accelerates HER2-driven transgenic mammary tumors and induces resistance to combinations of anti-HER2 therapies
- Author
-
Maria G. Kuba, Carlos L. Arteaga, Cammie R. Sutton, Ariella B. Hanker, Hailing Cheng, Justin M. Balko, Rebecca S. Cook, Violeta Sanchez, Charles M. Perou, Jean J. Zhao, Adam D. Pfefferle, and Christian D. Young
- Subjects
Pathology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,Class I Phosphatidylinositol 3-Kinases ,Mice, Transgenic ,Biology ,Lapatinib ,Mice ,Phosphatidylinositol 3-Kinases ,Breast cancer ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Epithelial–mesenchymal transition ,skin and connective tissue diseases ,neoplasms ,PI3K/AKT/mTOR pathway ,Multidisciplinary ,Oncogene ,Mammary Neoplasms, Experimental ,Biological Sciences ,Genes, erbB-2 ,medicine.disease ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,Pertuzumab ,Stem cell ,medicine.drug - Abstract
Human epidermal growth factor receptor 2 ( HER2 ; ERBB2 ) amplification and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha ( PIK3CA ) mutations often co-occur in breast cancer. Aberrant activation of the phosphatidylinositol 3-kinase (PI3K) pathway has been shown to correlate with a diminished response to HER2-directed therapies. We generated a mouse model of HER2-overexpressing (HER2 + ), PIK3CA H1047R -mutant breast cancer. Mice expressing both human HER2 and mutant PIK3CA in the mammary epithelium developed tumors with shorter latencies compared with mice expressing either oncogene alone. HER2 and mutant PIK3CA also cooperated to promote lung metastases. By microarray analysis, HER2-driven tumors clustered with luminal breast cancers, whereas mutant PIK3CA tumors were associated with claudin-low breast cancers. PIK3CA and HER2 + / PIK3CA tumors expressed elevated transcripts encoding markers of epithelial-to-mesenchymal transition and stem cells. Cells from HER2 + / PIK3CA tumors more efficiently formed mammospheres and lung metastases. Finally, HER2 + / PIK3CA tumors were resistant to trastuzumab alone and in combination with lapatinib or pertuzumab. Both drug resistance and enhanced mammosphere formation were reversed by treatment with a PI3K inhibitor. In sum, PIK3CA H1047R accelerates HER2-mediated breast epithelial transformation and metastatic progression, alters the intrinsic phenotype of HER2-overexpressing cancers, and generates resistance to approved combinations of anti-HER2 therapies.
- Published
- 2013
- Full Text
- View/download PDF
48. Association of FGFR1 with ERαmaintains ligand-independent ER transcription and mediates resistance to estrogen deprivation in ER+ breast cancer
- Author
-
Valerie M. Jansen, Yao Lu, Stefano Cairo, Thomas Stricker, Carlos L. Arteaga, Luis J. Schwarz, Justin M. Balko, Jennifer M. Giltnane, Angel Guerrero-Zotano, Monica M. Estrada, Roberto Bianco, Neil E. Bhola, Jean Gabriel Judde, Kimberly M. Stauffer, Ingrid A. Mayer, Melinda E. Sanders, Christian D. Young, Luigi Formisano, Olivier Deas, Katherine E. Hutchinson, Teresa C. Dugger, Formisano, Luigi, Stauffer, Kimberly Mae, Young, Christian D, Bhola, Neil E, Guerrero Zotano, Angel L, Jansen, Valerie M, Estrada, Monica M, Hutchinson, Katherine E, Giltnane, Jennifer M, Schwarz, Luis J, Lu, Yao, Balko, Justin M, Deas, Olivier, Cairo, Stefano, Judde, Jean Gabriel, Mayer, Ingrid A, Sanders, Melinda, Dugger, Teresa C, Bianco, Roberto, Stricker, Thoma, and Arteaga, Carlos L.
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Transcription, Genetic ,medicine.drug_class ,Cell ,Breast Neoplasms ,Biology ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Estrogen Receptor Modulators ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Receptor, Fibroblast Growth Factor, Type 1 ,E2F ,Protein Kinase Inhibitors ,Neoplasm Staging ,Fulvestrant ,Cell growth ,Letrozole ,Estrogen Receptor alpha ,Gene Amplification ,Fibroblast Growth Factors ,Gene Expression Regulation, Neoplastic ,stomatognathic diseases ,Disease Models, Animal ,Protein Transport ,FGFR1, breast cancer, estrogen resistance ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Estrogen ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Estrogen receptor alpha ,medicine.drug ,Signal Transduction - Abstract
Purpose: FGFR1 amplification occurs in approximately 15% of estrogen receptor–positive (ER+) human breast cancers. We investigated mechanisms by which FGFR1 amplification confers antiestrogen resistance to ER+ breast cancer.Experimental Design: ER+ tumors from patients treated with letrozole before surgery were subjected to Ki67 IHC, FGFR1 FISH, and RNA sequencing (RNA-seq). ER+/FGFR1–amplified breast cancer cells, and patient-derived xenografts (PDX) were treated with FGFR1 siRNA or the FGFR tyrosine kinase inhibitor lucitanib. Endpoints were cell/xenograft growth, FGFR1/ERα association by coimmunoprecipitation and proximity ligation, ER genomic activity by ChIP sequencing, and gene expression by RT-PCR.Results: ER+/FGFR1–amplified tumors in patients treated with letrozole maintained cell proliferation (Ki67). Estrogen deprivation increased total and nuclear FGFR1 and FGF ligands expression in ER+/FGFR1–amplified primary tumors and breast cancer cells. In estrogen-free conditions, FGFR1 associated with ERα in tumor cell nuclei and regulated the transcription of ER-dependent genes. This association was inhibited by a kinase-dead FGFR1 mutant and by treatment with lucitanib. ChIP-seq analysis of estrogen-deprived ER+/FGFR1–amplified cells showed binding of FGFR1 and ERα to DNA. Treatment with fulvestrant and/or lucitanib reduced FGFR1 and ERα binding to DNA. RNA-seq data from FGFR1-amplified patients' tumors treated with letrozole showed enrichment of estrogen response and E2F target genes. Finally, growth of ER+/FGFR1–amplified cells and PDXs was more potently inhibited by fulvestrant and lucitanib combined than each drug alone.Conclusions: These data suggest the ERα pathway remains active in estrogen-deprived ER+/FGFR1–amplified breast cancers. Therefore, these tumors are endocrine resistant and should be candidates for treatment with combinations of ER and FGFR antagonists. Clin Cancer Res; 23(20); 6138–50. ©2017 AACR.
- Published
- 2017
49. An ERBB1-3 Neutralizing Antibody Mixture With High Activity Against Drug-Resistant HER2+ Breast Cancers With ERBB Ligand Overexpression
- Author
-
Monica V. Estrada, Melinda E. Sanders, Christian D. Young, Monica Red-Brewer, Michael Kragh, Katherine E. Hutchinson, Carlos L. Arteaga, Teresa C. Dugger, Mikkel Wandahl Pedersen, Brent N. Rexer, Paula Gonzalez Ericsson, Angel Guerrero-Zotano, Luis J. Schwarz, Johan Lantto, Ivan D Horak, Luigi Formisano, Schwarz, Lj, Hutchinson, Ke, Rexer, Bn, Estrada, Mv, Gonzalez Ericsson, Pi, Sanders, Me, Dugger, Tc, Formisano, L, Guerrero-Zotano, A, Red-Brewer, M, Young, Cd, Lantto, J, Pedersen, Mw, Kragh, M, Horak, Id, and Arteaga, Cl.
- Subjects
0301 basic medicine ,Cancer Research ,Receptor, ErbB-3 ,Receptor, ErbB-2 ,Mice, Nude ,Antineoplastic Agents ,Breast Neoplasms ,Ado-Trastuzumab Emtansine ,Antibodies, Monoclonal, Humanized ,Ligands ,Lapatinib ,Mice ,03 medical and health sciences ,ErbB Receptors ,0302 clinical medicine ,ErbB ,Trastuzumab ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Maytansine ,Epidermal growth factor receptor ,skin and connective tissue diseases ,biology ,Cancer ,Articles ,medicine.disease ,Antibodies, Neutralizing ,Xenograft Model Antitumor Assays ,Molecular biology ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Quinazolines ,Cancer research ,biology.protein ,Immunohistochemistry ,Female ,Pertuzumab ,medicine.drug - Abstract
BACKGROUND: Plasticity of the ERBB receptor network has been suggested to cause acquired resistance to anti–human epidermal growth factor receptor 2 (HER2) therapies. Thus, we studied whether a novel approach using an ERBB1-3-neutralizing antibody mixture can block these compensatory mechanisms of resistance. METHODS: HER2+ cell lines and xenografts (n ≥ 6 mice per group) were treated with the ERBB1-3 antibody mixture Pan-HER, trastuzumab/lapatinib (TL), trastuzumab/pertuzumab (TP), or T-DM1. Downregulation of ERBB receptors was assessed by immunoblot analysis and immunohistochemistry. Paired pre- and post-T-DM1 tumor biopsies from patients (n = 11) with HER2-amplified breast cancer were evaluated for HER2 and P-HER3 expression by immunohistochemistry and/or fluorescence in situ hybridization. ERBB ligands were measured by quantitative reverse transcription polymerase chain reaction. Drug-resistant cells were generated by chronic treatment with T-DM1. All statistical tests were two-sided. RESULTS: Treatment with Pan-HER inhibited growth and promoted degradation of ERBB1-3 receptors in a panel of HER2+ breast cancer cells. Compared with TL, TP, and T-DM1, Pan-HER induced a similar antitumor effect against established BT474 and HCC1954 tumors, but was superior to TL against MDA-361 xenografts (TL mean = 2026 mm(3), SD = 924 mm(3), vs Pan-HER mean = 565 mm(3), SD = 499 mm(3), P = .04). Pan-HER-treated BT474 xenografts did not recur after treatment discontinuation, whereas tumors treated with TL, TP, and T-DM1 did. Post-TP and post-T-DM1 recurrent tumors expressed higher levels of neuregulin-1 (NRG1), HER3 and P-HER3 (all P < .05). Higher levels of P-HER3 protein and NRG1 mRNA were also observed in HER2+ breast cancers progressing after T-DM1 and trastuzumab (NRG1 transcript fold change ± SD; pretreatment = 2, SD = 1.9, vs post-treatment = 11.4, SD = 10.3, P = .04). The HER3-neutralizing antibody LJM716 resensitized the drug-resistant cells to T-DM1, suggesting a causal association between the NRG1-HER3 axis and drug resistance. Finally, Pan-HER treatment inhibited growth of HR6 trastuzumab- and T-DM1-resistant xenografts. CONCLUSIONS: These data suggest that upregulation of a NRG1-HER3 axis can mediate escape from anti-HER2 therapies. Further, multitargeted antibody mixtures, such as Pan-HER, can simultaneously remove and/or block targeted ERBB receptor and ligands, thus representing an effective approach against drug-sensitive and -resistant HER2+ cancers.
- Published
- 2017
50. Genomic profiling of ER
- Author
-
Jennifer M, Giltnane, Katherine E, Hutchinson, Thomas P, Stricker, Luigi, Formisano, Christian D, Young, Monica V, Estrada, Mellissa J, Nixon, Liping, Du, Violeta, Sanchez, Paula Gonzalez, Ericsson, Maria G, Kuba, Melinda E, Sanders, Xinmeng J, Mu, Eliezer M, Van Allen, Nikhil, Wagle, Ingrid A, Mayer, Vandana, Abramson, Henry, Gόmez, Monica, Rizzo, Weiyi, Toy, Sarat, Chandarlapaty, Erica L, Mayer, Jason, Christiansen, Danielle, Murphy, Kerry, Fitzgerald, Kai, Wang, Jeffrey S, Ross, Vincent A, Miller, Phillip J, Stephens, Roman, Yelensky, Levi, Garraway, Yu, Shyr, Ingrid, Meszoely, Justin M, Balko, and Carlos L, Arteaga
- Subjects
Receptors, Estrogen ,Receptor, ErbB-2 ,Cell Line, Tumor ,Cyclin-Dependent Kinase 4 ,Humans ,Breast Neoplasms ,Cyclin D1 ,Female ,Cyclin-Dependent Kinase 6 ,Receptor, Fibroblast Growth Factor, Type 1 ,In Vitro Techniques - Abstract
Inhibition of proliferation in estrogen receptor-positive (ER
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.