1
|
Jiang Q, Xiao J, Hsieh YC, Kumar NL, Han L, Zou Y, Li H. The Role of the PI3K/Akt/mTOR Axis in Head and Neck Squamous Cell Carcinoma. Biomedicines 2024; 12:1610. [PMID: 39062182 PMCID: PMC11274428 DOI: 10.3390/biomedicines12071610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignancies globally, representing a significant public health problem with a poor prognosis. The development of efficient therapeutic strategies for HNSCC prevention and treatment is urgently needed. The PI3K/AKT/mTOR (PAM) signaling pathway is a highly conserved transduction network in eukaryotic cells that promotes cell survival, growth, and cycle progression. Dysfunction in components of this pathway, such as hyperactivity of PI3K, loss of PTEN function, and gain-of-function mutations in AKT, are well-known drivers of treatment resistance and disease progression in cancer. In this review, we discuss the major mutations and dysregulations in the PAM signaling pathway in HNSCC. We highlight the results of clinical trials involving inhibitors targeting the PAM signaling pathway as a strategy for treating HNSCC. Additionally, we examine the primary mechanisms of resistance to drugs targeting the PAM pathway and potential therapeutic strategies.
Collapse
Affiliation(s)
- Qian Jiang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210093, China; (Q.J.)
- International Dentist Pathway, University of California, San Francisco, CA 94158, USA
| | - Jingyi Xiao
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210093, China; (Q.J.)
| | - Yao-Ching Hsieh
- International Dentist Pathway, University of California, San Francisco, CA 94158, USA
| | - Neha Love Kumar
- International Dentist Pathway, University of California, San Francisco, CA 94158, USA
| | - Lei Han
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210093, China; (Q.J.)
| | - Yuntao Zou
- Division of Hospital Medicine, University of California, San Francisco, CA 94158, USA
| | - Huang Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210093, China; (Q.J.)
| |
Collapse
|
2
|
Alves LB, Moura AC, Amorim Dos Santos J, Borges GA, Guerra ENS. Pharmacological PI3K inhibition in head and neck squamous cell carcinoma: A systematic review. Toxicol In Vitro 2023; 88:105558. [PMID: 36681288 DOI: 10.1016/j.tiv.2023.105558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 12/12/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
BACKGROUND This systematic review aimed to investigate the in vitro and in vivo effects of phosphatidylinositol-3-kinase (PI3K) inhibitors on head and neck squamous cell carcinoma (HNSCC). Considering the role of PI3K and its downstream effectors in cell proliferation, invasion, and survival, it is reasonable to expect that treatment with PI3K inhibitors could control HNSCC onset and progression. Thus, the research question for our review was whether pharmacological inhibition of PI3K affects HNSCC progression. METHODS In vitro and in vivo studies were selected from six databases. We collected data regarding cell viability, apoptosis, and the regulation of protein expression levels from in vitro studies. For the in vivo studies, we analyzed the reduction in tumor size or gene and protein expression. RESULTS The included studies showed reduced cell proliferation and apoptosis after treatment with PI3K inhibitors. PI3K inhibitors in combination with other drugs had an enhanced anticancer effects compared to those of single-drug treatments. CONCLUSIONS The results support the potential of PI3K inhibitors as candidates for clinical trials in HNSCC.
Collapse
Affiliation(s)
- L B Alves
- Laboratory of Oral Histopathology, School of Health Sciences, University of Brasília, Brasília, Brazil
| | - A C Moura
- Laboratory of Oral Histopathology, School of Health Sciences, University of Brasília, Brasília, Brazil
| | - J Amorim Dos Santos
- Laboratory of Oral Histopathology, School of Health Sciences, University of Brasília, Brasília, Brazil
| | - G A Borges
- Department of Oral Medicine and Periodontics, Faculty of Dentistry, University of Michigan, Ann Arbor, United States
| | - E N S Guerra
- Laboratory of Oral Histopathology, School of Health Sciences, University of Brasília, Brasília, Brazil.
| |
Collapse
|
3
|
Li Q, Tie Y, Alu A, Ma X, Shi H. Targeted therapy for head and neck cancer: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:31. [PMID: 36646686 PMCID: PMC9842704 DOI: 10.1038/s41392-022-01297-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/27/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Head and neck cancer (HNC) is malignant, genetically complex and difficult to treat and is the sixth most frequent cancer, with tobacco, alcohol and human papillomavirus being major risk factors. Based on epigenetic data, HNC is remarkably heterogeneous, and treatment remains challenging. There is a lack of significant improvement in survival and quality of life in patients with HNC. Over half of HNC patients experience locoregional recurrence or distal metastasis despite the current multiple traditional therapeutic strategies and immunotherapy. In addition, resistance to chemotherapy, radiotherapy and some targeted therapies is common. Therefore, it is urgent to explore more effective and tolerable targeted therapies to improve the clinical outcomes of HNC patients. Recent targeted therapy studies have focused on identifying promising biomarkers and developing more effective targeted therapies. A well understanding of the pathogenesis of HNC contributes to learning more about its inner association, which provides novel insight into the development of small molecule inhibitors. In this review, we summarized the vital signaling pathways and discussed the current potential therapeutic targets against critical molecules in HNC, as well as presenting preclinical animal models and ongoing or completed clinical studies about targeted therapy, which may contribute to a more favorable prognosis of HNC. Targeted therapy in combination with other therapies and its limitations were also discussed.
Collapse
Affiliation(s)
- Qingfang Li
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Tie
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Co-encapsulation of PI3-Kδ/HDAC6 dual inhibitor and Navitoclax in Quatramer™ nanoparticles for synergistic effect in ER+ breast cancer. Int J Pharm 2022; 628:122343. [DOI: 10.1016/j.ijpharm.2022.122343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 11/11/2022]
|
5
|
Histone Deacetylase Inhibitors as Multitarget-Directed Epi-Drugs in Blocking PI3K Oncogenic Signaling: A Polypharmacology Approach. Int J Mol Sci 2020; 21:ijms21218198. [PMID: 33147762 PMCID: PMC7662987 DOI: 10.3390/ijms21218198] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic mutations and aberrant epigenetic alterations are the triggers for carcinogenesis. The emergence of the drugs targeting epigenetic aberrations has provided a better outlook for cancer treatment. Histone deacetylases (HDACs) are epigenetic modifiers playing critical roles in numerous key biological functions. Inappropriate expression of HDACs and dysregulation of PI3K signaling pathway are common aberrations observed in human diseases, particularly in cancers. Histone deacetylase inhibitors (HDACIs) are a class of epigenetic small-molecular therapeutics exhibiting promising applications in the treatment of hematological and solid malignancies, and in non-neoplastic diseases. Although HDACIs as single agents exhibit synergy by inhibiting HDAC and the PI3K pathway, resistance to HDACIs is frequently encountered due to activation of compensatory survival pathway. Targeted simultaneous inhibition of both HDACs and PI3Ks with their respective inhibitors in combination displayed synergistic therapeutic efficacy and encouraged the development of a single HDAC-PI3K hybrid molecule via polypharmacology strategy. This review provides an overview of HDACs and the evolution of HDACs-based epigenetic therapeutic approaches targeting the PI3K pathway.
Collapse
|
6
|
Marques AEM, do Nascimento Filho CHV, Marinho Bezerra TM, Guerra ENS, Castilho RM, Squarize CH. Entinostat is a novel therapeutic agent to treat oral squamous cell carcinoma. J Oral Pathol Med 2020; 49:771-779. [PMID: 32450006 DOI: 10.1111/jop.13039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/18/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Alterations of the epigenome may influence cancer initiation and progression. At the cellular level, histones are key regulators of chromatin accessibility and gene transcription; thus, the inhibition of histone deacetylase enzymes (HDACs) constitutes an attractive target for therapy. In this study, we investigated the effects of the HDAC inhibitor entinostat on oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS We tested the effects of entinostat on OSCC cell lines. Cell viability and growth were analyzed using MTT assay. Cell cycle analysis, cell apoptosis, cancer stem cell (CSC) content, and the concentration of reactive oxygen species (ROS) in OSCC tumor cells were assessed using flow cytometry. The expression of histones and cell cycle regulatory proteins was examined by Western blot. RESULTS The administration of entinostat resulted in reduced proliferation of OSCC cells, followed by cell cycle arrest at the G0/G1 phase, as well as substantial tumor apoptosis. We found an increase in ROS production and significant reductions in CSCs. We also found that entinostat caused increased acetylation histone H3 and histone H4, and changes in the expression of cell cycle-associated proteins such as p21. CONCLUSION This study indicates that entinostat is a potential novel therapeutic agent for OSCC by halting tumor proliferation, inducing cytotoxicity and intracellular ROS, and attacking the CSCs.
Collapse
Affiliation(s)
- Ana Elizia M Marques
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasilia, Brasilia, Brazil
| | - Carlos Henrique V do Nascimento Filho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Thamara M Marinho Bezerra
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,School of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | - Eliete N S Guerra
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasilia, Brasilia, Brazil
| | - Rogerio M Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,The University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,The University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Yang Q, Jiang W, Hou P. Emerging role of PI3K/AKT in tumor-related epigenetic regulation. Semin Cancer Biol 2019; 59:112-124. [DOI: 10.1016/j.semcancer.2019.04.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/14/2019] [Accepted: 04/01/2019] [Indexed: 01/23/2023]
|
8
|
Schomberg J. Identification of Targetable Pathways in Oral Cancer Patients via Random Forest and Chemical Informatics. Cancer Inform 2019; 18:1176935119889911. [PMID: 31819345 PMCID: PMC6883365 DOI: 10.1177/1176935119889911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
Treatment of head and neck cancer has been slow to change with epidermal growth
factor receptor (EGFR) inhibitors, PD1 inhibitors, and
taxane-/plant-alkaloid-derived chemotherapies being the only therapies approved
by the U.S. Food and Drug Administration (FDA) in the last 10 years for the
treatment of head and neck cancers. Head and neck cancer is a relatively rare
cancer compared to breast or lung cancers. However, it is possible that existing
therapies for more common solid tumors or for the treatment of other diseases
could also prove effective against oral cancers. Many therapies have molecular
targets that could be appropriate in oral cancer as well as the cancer in which
the drug gained initial FDA approval. Also, there may be targets in oral cancer
for which existing FDA-approved drugs could be applied. This study describes
informatics methods that use machine learning to identify influential gene
targets in patients receiving platinum-based chemotherapy, non-platinum-based
chemotherapy, and genes influential in both groups of patients. This analysis
yielded 6 small molecules that had a high Tanimoto similarity (>50%) to
ligands binding genes shown to be highly influential in determining treatment
response in oral cancer patients. In addition to influencing treatment response,
these genes were also found to act as gene hubs connected to more than 100 other
genes in pathways enriched with genes determined to be influential in treatment
response by a random forest classifier with 20 000 trees trying 320 variables at
each tree node. This analysis validates the use of multiple informatics methods
to identify small molecules that have a greater likelihood of efficacy in a
given cancer of interest.
Collapse
Affiliation(s)
- John Schomberg
- CHOC Children's, Orange, CA, USA.,School of Population Health Science, University of California Irvine, Irvine, CA, USA.,Afecta Pharmaceuticals, Irvine, CA, USA
| |
Collapse
|
9
|
Wei T, Choi S, Buehler D, Anderson RA, Lambert PF. A PI3K/AKT Scaffolding Protein, IQ Motif-Containing GTPase Associating Protein 1 (IQGAP1), Promotes Head and Neck Carcinogenesis. Clin Cancer Res 2019; 26:301-311. [PMID: 31597661 DOI: 10.1158/1078-0432.ccr-19-1063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/15/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Head and neck cancer (HNC) is the sixth most common cancer worldwide with a 5-year survival rate of less than 50%. The PI3K/AKT/mTOR signaling pathway is frequently implicated in HNC. Recently, IQ motif-containing GTPase-activating protein 1 (IQGAP1) was discovered to scaffold the PI3K/AKT signaling pathway. IQGAP1 gene expression is increased in HNC, raising the hypothesis that IQGAP1 contributes to HNC. EXPERIMENTAL DESIGN We performed a combination of in vitro studies using human cancer cell lines treated with a cell-permeable peptide that interferes with IQGAP1's ability to bind to PI3K, and in vivo studies utilizing mice genetically knocked out for the Iqgap1 (Iqgap1 -/-). In vivo EGF stimulation assays were used to evaluate PI3K signaling. To study the role of IQGAP1 in HNC, we used a well-validated mouse model that drives HNC via a synthetic oral carcinogen, 4-nitroquinoline 1-oxide (4NQO). RESULTS IQGAP1 is necessary for efficient PI3K signaling in vitro and in vivo. Disruption of IQGAP1-scaffolded PI3K/AKT signaling reduced HNC cell survival. Iqgap1 -/- mice had significantly lower cancer incidences, lesser disease severity, and fewer cancer foci. IQGAP1 protein levels were increased in HNC arising in Iqgap1+/+ mice. The level of PI3K signaling in 4NQO-induced HNC arising in Iqgap1 -/- mice was significantly reduced, consistent with the hypothesis that IQGAP1 contributes to HNC at least partly through PI3K signaling. High IQGAP1 expression correlated with reduced survival, and high pS6 levels correlated with high IQGAP1 levels in patients with HNC. CONCLUSIONS These data demonstrate that IQGAP1 contributes to head and neck carcinogenesis.
Collapse
Affiliation(s)
- Tao Wei
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Suyong Choi
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Darya Buehler
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Richard A Anderson
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin. .,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
10
|
McKenna M, McGarrigle S, Pidgeon GP. The next generation of PI3K-Akt-mTOR pathway inhibitors in breast cancer cohorts. Biochim Biophys Acta Rev Cancer 2018; 1870:185-197. [PMID: 30318472 DOI: 10.1016/j.bbcan.2018.08.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/09/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
The PI3K/Akt/mTOR pathway plays a role in various oncogenic processes in breast cancer and key pathway aberrations have been identified which drive the different molecular subtypes. Early drugs developed targeting this pathway produced some clinical success but were hampered by pharmacokinetics, tolerability and efficacy problems. This created a need for new PI3K pathway-inhibiting drugs, which would produce more robust results allowing incorporation into treatment regimens for breast cancer patients. In this review, the most promising candidates from the new generation of PI3K-pathway inhibitors is explored, presenting evidence from preclinical and early clinical research, as well as ongoing trials utilising these drugs in breast cancer cohorts. The problems hindering the development of drugs targeting the PI3K pathway are examined, which have created problems for their use as monotherapies. PI3K pathway inhibitor combinations therefore remains a dynamic research area, and their role in combination with immunotherapies and epigenetic therapies is also inspected.
Collapse
Affiliation(s)
- Michael McKenna
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Sarah McGarrigle
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Graham P Pidgeon
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
11
|
Wu N, Zhu Y, Xu X, Zhu Y, Song Y, Pang L, Chen Z. The anti-tumor effects of dual PI3K/mTOR inhibitor BEZ235 and histone deacetylase inhibitor Trichostatin A on inducing autophagy in esophageal squamous cell carcinoma. J Cancer 2018; 9:987-997. [PMID: 29581778 PMCID: PMC5868166 DOI: 10.7150/jca.22861] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
The effect and regulation of autophagy-related proteins Beclin-1 and LC3 in esophageal squamous cell carcinoma have not been fully studied. The aim of this study was to assess the expression of Beclin-1 and LC3 in ESCCs, and to investigate the association between the two markers and clinicopathological characteristics as well as prognosis. Meanwhile, we explored the anti-tumor effect of the PI3K/mTOR dual inhibitor BEZ235 and the histone deacetylase inhibitor TSA on inducing autophagy in ESCC cells. Our study included 118 ESCC tumors and paired non-tumor esophageal mucosa tissues. Beclin-1 and LC3 expression were performed by immunohistochemistry. Human ESCC cells Eca-109 and TE-1 were treated with BEZ235 and TSA either alone or in combination in Vitro. The expression of both Beclin-1 and LC3 proteins were decreased significantly in ESCCs, but there was no significant relation between the expression of Beclin-1 and LC3 (P = 0.427). The negative expression of either Beclin-1 or LC3 was associated with advanced TNM stages (P = 0.006 and P<0.001, respectively). Patients with a high expression of Beclin-1 and LC3 predict better prognosis. In Vitro co-treatment with BEZ235 and TSA showed a synergistic effect on inhibition of ESCC cell viability and induction of autophagy with the increasing expressions of Beclin-1, LC3-II and the ratio of LC3-II/LC3-I. Our results demonstrated that the autophagy-related proteins Beclin-1 and LC3 were decreased in ESCCs and the low expression of the two markers predicted a worse prognosis. The co-treatment of BEZ235 and TSA significantly induced autophagy and enhanced anti-tumor activities, provided a new effective therapeutic target in ESCCs.
Collapse
Affiliation(s)
- Ning Wu
- Department of Cardio-thoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, 200040, China
| | - Yingfeng Zhu
- Department of Pathology, HuaShan Hospital of Fudan University, Shanghai, 200040, China
| | - Xiao Xu
- Central laboratory, HuaShan Hospital of Fudan University, Shanghai, 200040, China
| | - Yongjun Zhu
- Department of Cardio-thoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, 200040, China
| | - Yang Song
- Department of Cardio-thoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, 200040, China
| | - Liewen Pang
- Department of Cardio-thoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, 200040, China
| | - Zhiming Chen
- Department of Cardio-thoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, 200040, China
| |
Collapse
|
12
|
Anticancer drugs and the regulation of Hedgehog genes GLI1 and PTCH1, a comparative study in nonmelanoma skin cancer cell lines. Anticancer Drugs 2017; 28:1106-1117. [DOI: 10.1097/cad.0000000000000551] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
13
|
PI3K/mTOR dual inhibitor BEZ235 and histone deacetylase inhibitor Trichostatin A synergistically exert anti-tumor activity in breast cancer. Oncotarget 2017; 8:11937-11949. [PMID: 28060760 PMCID: PMC5355316 DOI: 10.18632/oncotarget.14442] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/27/2016] [Indexed: 01/01/2023] Open
Abstract
Molecule-targeted therapy has achieved great progress in cancer therapy. Effective drug combinations are one way to enhance the therapeutic efficacy and combat resistance. Here, we determined the effect of the PI3K/mTOR dual inhibitor BEZ235 and the histone deacetylase inhibitor Trichostatin A (TSA) on human breast cancer. We demonstrated that the combination of BEZ235 and TSA results in significant synergistic growth inhibition of multiple breast cancer cell lines. Mechanistic studies revealed that the combined therapy induced apoptosis in a caspase-dependent manner, which might be related to the further depression of the PI3K/Akt/mTOR signalling pathway. Additionally, co-treatment with BEZ235 and TSA enhanced autophagic cell death by up-regulating the expression of LC3B-II and Beclin-1. The vivo tumour modelling studies revealed that BEZ235 combined with TSA blocked tumour growth without noticeable side effects. These data suggest that the combination of BEZ235 and TSA may be a new selective strategy, which may have significant clinical application in the treatment of breast cancer patients.
Collapse
|
14
|
Enzenhofer E, Kadletz L, Stanisz I, Kotowski U, Seemann R, Schmid R, Thurnher D, Heiduschka G. Effect of the histone deacetylase inhibitor resminostat on head and neck squamous cell carcinoma cell lines. Head Neck 2017; 39:900-907. [PMID: 28170128 DOI: 10.1002/hed.24699] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/11/2016] [Accepted: 12/06/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Carcinogenesis is determined by various epigenetic events, such as histone deacetylation. The purpose of this study was to investigate the effect of the new histone deacetylase inhibitor resminostat on head and neck squamous cell carcinoma (HNSCC) cell lines. METHODS The cytotoxicity of resminostat and cisplatin on HNSCC cell lines SCC25, CAL27, and FaDu was determined using CCK-8 cell proliferation assay and combination index analysis. Cells were irradiated with 2 to 8 Gray. Apoptosis was measured using flow cytometry and expression of Mcl-1, p-AKT, and survivin was investigated. RESULTS Treatment with resminostat showed a decrease of cell proliferation of HNSCC cell lines. In addition, a synergistic effect with cisplatin as well as with radiation treatment could be observed. Induction of cell death and dose-dependent downregulation of survivin was evident in all cell lines. CONCLUSION Resminostat is a promising treatment of HNSCC because of its antiproliferative, chemosensitizing, and radiosensitizing effects. © 2017 Wiley Periodicals, Inc. Head Neck 39: 900-907, 2017.
Collapse
Affiliation(s)
- Elisabeth Enzenhofer
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Lorenz Kadletz
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Isabella Stanisz
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Ulana Kotowski
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Rudolf Seemann
- Department of Oral and Maxillofacial Surgery, Medical University of Vienna, Vienna, Austria
| | - Rainer Schmid
- Department of Radiotherapy, Medical University of Vienna, Vienna, Austria
| | - Dietmar Thurnher
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria.,Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Graz, Graz, Austria
| | - Gregor Heiduschka
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Dai D, Chen H, Tang J, Tang Y. Inhibition of mTOR/eIF4E by anti-viral drug ribavirin effectively enhances the effects of paclitaxel in oral tongue squamous cell carcinoma. Biochem Biophys Res Commun 2016; 482:1259-1264. [PMID: 27932243 DOI: 10.1016/j.bbrc.2016.12.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 12/03/2016] [Indexed: 01/03/2023]
Abstract
Upregulation of eIF4E is associated with poor clinical outcome in many human cancers and represents a potential therapeutic target. However, the function of eIF4E remains unknown in oral tongue squamous cell carcinoma (OTSCC). In this work, we show that ribavirin, an anti-viral drug, effectively augments sensitivity of OTSCC cells to paclitaxel via inhibiting mTOR/eIF4E signaling pathway. Ribavirin dose-dependently inhibits proliferation and induces apoptosis in SCC-9 and CAL27 cells. Combination of ribavirin and paclitaxel are more effective in inhibiting proliferation and inducing apoptosis in OTSCC cells. Importantly, the in vivo efficacy of ribavirin and its synergism with paclitaxel is confirmed by two independent OTSCC xenograft mouse models. Mechanistically, ribavirin significantly decreases mTOR/eIF4E signaling pathway in OTSCC cells via suppressing phosphorylation of Akt, mTOR, 4EBP1 and eIF4E. Overexpression of the phosphor-mimetic form of eIF4E (eIF4E S209D) but not the nonphosphorylatable form (eIF4E S209A) reverses the effects of ribavirin, confirming that eIF4E inhibition is the mechanism of action of ribavirin in OTSCC cells. In addition, eIF4E depletion significantly enhances the anti-proliferative and pro-apoptotic effects of paclitaxel, demonstrating the critical role of eIF4E in OTSCC cell response to paclitaxel. Our work is the first to demonstrate the efficacy of ribavirin as a single agent and synergism as combination with paclitaxel in OTSCC in vitro and in vivo. Our findings also demonstrate the therapeutic value of inhibiting eIF4E in OTSCC treatment.
Collapse
Affiliation(s)
- Dehua Dai
- Department of Stomatology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, People's Republic of China
| | - Hujie Chen
- Department of Stomatology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, People's Republic of China
| | - Jing Tang
- Department of Stomatology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, People's Republic of China
| | - Yi Tang
- Department of Stomatology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, People's Republic of China.
| |
Collapse
|
16
|
Ferreira ACDS, de-Freitas-Junior JCM, Morgado-Díaz JA, Ridley AJ, Klumb CE. Dual inhibition of histone deacetylases and phosphoinositide 3-kinases: effects on Burkitt lymphoma cell growth and migration. J Leukoc Biol 2016; 99:569-78. [PMID: 26561567 DOI: 10.1189/jlb.2a0415-162r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 10/19/2015] [Indexed: 02/04/2023] Open
Abstract
Burkitt lymphoma is a highly aggressive non-Hodgkin lymphoma that is characterized by MYC deregulation. Recently, the PI3K pathway has emerged as a cooperative prosurvival mechanism in Burkitt lymphoma. Despite the highly successful results of treatment that use high-dose chemotherapy regimens in pediatric Burkitt lymphoma patients, the survival rate of pediatric patients with progressive or recurrent disease is low. PI3Ks are also known to regulate cell migration, and abnormal cell migration may contribute to cancer progression and dissemination in Burkitt lymphoma. Little is known about Burkitt lymphoma cell migration, but the cooperation between MYC and PI3K in Burkitt lymphoma pathogenesis suggests that a drug combination could be used to target the different steps involved in Burkitt lymphoma cell dissemination and disease progression. The aim of this study was to investigate the effects of the histone deacetylase inhibitor suberoylanilide hydroxamic acid combined with the PI3K inhibitor LY294002 on Burkitt lymphoma cell growth and migration. The combination enhanced the cell growth inhibition and cell-cycle arrest induced by the PI3K inhibitor or histone deacetylase inhibitor individually. Moreover, histone deacetylase inhibitor/PI3K inhibitor cotreatment suppressed Burkitt lymphoma cell migration and decreased cell polarization, Akt and ERK1/2 phosphorylation, and leads to RhoB induction. In summary, the histone deacetylase inhibitor/PI3Ki combination inhibits cell proliferation and migration via alterations in PI3K signaling and histone deacetylase activity, which is involved in the acetylation of α-tubulin and the regulation of RhoB expression.
Collapse
Affiliation(s)
- Ana Carolina dos Santos Ferreira
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Julio Cesar Madureira de-Freitas-Junior
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Jose Andres Morgado-Díaz
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Anne J Ridley
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Claudete Esteves Klumb
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| |
Collapse
|
17
|
Zhang P, Zhu X, Wu Y, Hu R, Li D, Du J, Jiao X, He X. Histone deacetylase inhibitors reduce WB-F344 oval cell viability and migration capability by suppressing AKT/mTOR signaling in vitro. Arch Biochem Biophys 2015; 590:1-9. [PMID: 26558695 DOI: 10.1016/j.abb.2015.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/12/2022]
Abstract
Histone deacetylase (HDAC) can blockDNA replication and transcription and altered HDAC expression was associated with tumorigenesis. This study investigated the effects of HDAC inhibitors on hepatic oval cells and aimed to delineate the underlying molecular events. Hepatic oval cells were treated with two different HDAC inhibitors, suberoylanilidehydroxamic acid (SAHA) and trichostatin-A (TSA). Cells were subjected to cell morphology, cell viability, cell cycle, and wound healing assays. The expression of proteins related to both apoptosis and the cell cycle, and proteins of the AKT/mammalian target of rapamycin (mTOR) signaling pathway were analyzed by Western blot. The data showed that HDAC inhibitors reduced oval cell viability and migration capability, and arrested oval cells at the G0/G1 and S phases of the cell cycle, in a dose- and time-dependent manner. HDAC inhibitors altered cell morphology and reduced oval cell viability, and downregulated the expression of PCNA, cyclinD1, c-Myc and Bmi1 proteins, while also suppressing AKT/mTOR and its downstream target activity. In conclusion, this study demonstrates that HDAC inhibitors affect oval cells by suppressing AKT/mTOR signaling.
Collapse
Affiliation(s)
- Peng Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaofeng Zhu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Wu
- Department of Biostatistics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ronglin Hu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dongming Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xingyuan Jiao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
|
19
|
Abstract
In spite of a rapidly expanding understanding of head and neck tumor biology and optimization of radiation, chemotherapy, and surgical treatment modalities, head and neck squamous cell carcinoma (HNSCC) remains a major cause of cancer-related morbidity and mortality. Although our biologic understanding of these tumors had largely been limited to pathways driving proliferation, survival, and differentiation, the identification of HPV as a major driver of HNSCC and genomic sequencing analyses has dramatically influenced our understanding of tumor biology and approach to therapy. Here, we summarize molecular aspects of HNSCC biology and identify promising areas for potential diagnostic and therapeutic agents.
Collapse
Affiliation(s)
- Sidharth V Puram
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA 02114, USA; Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA
| | - James W Rocco
- Department of Otolaryngology-Head and Neck Surgery, Wexner Medical Center, James Cancer Hospital, Solove Research Institute, The Ohio State University, 320 West 10th Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
20
|
Zhang P, Guo Z, Wu Y, Hu R, Du J, He X, Jiao X, Zhu X. Histone Deacetylase Inhibitors Inhibit the Proliferation of Gallbladder Carcinoma Cells by Suppressing AKT/mTOR Signaling. PLoS One 2015; 10:e0136193. [PMID: 26287365 PMCID: PMC4542213 DOI: 10.1371/journal.pone.0136193] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/31/2015] [Indexed: 12/18/2022] Open
Abstract
Gallbladder carcinoma is an aggressive malignancy with high mortality mainly due to the limited potential for curative resection and its resistance to chemotherapeutic agents. Here, we show that the histone deacetylase inhibitors (HDACIs) trichostatin-A (TSA) and suberoylanilide hydroxamic acid (SAHA) reduce the proliferation and induce apoptosis of gallbladder carcinoma cells by suppressing the AKT/mammalian target of rapamycin (mTOR) signaling. Gallbladder carcinoma SGC-996 cells were treated with different concentrations of TSA and SAHA for different lengths of time. Cell proliferation and morphology were assessed with MTT assay and microscopy, respectively. Cell cycle distribution and cell apoptosis were analyzed with flow cytometry. Western blotting was used to detect the proteins related to apoptosis, cell cycle, and the AKT/mTOR signaling pathway. Our data showed that TSA and SAHA reduced SGC-996 cell viability and arrested cell cycle at the G1 phase in a dose- and time-dependent manner. TSA and SAHA promoted apoptosis of SGC-996 cells, down-regulated the expression of cyclin D1, c-Myc and Bmi1, and decreased the phosphorylation of AKT, mTOR p70S6K1, S6 and 4E-BP1. Additionally, the mTOR inhibitor rapamycin further reduced the cell viability of TSA- and SAHA-treated SGC-996 cells and the phosphorylation of mTOR, whereas the mTOR activator 1,2-dioctanoyl-sn-glycero-3-phosphate (C8-PA) exerted the opposite influence. Our results demonstrate that histone deacetylase inhibitors (HDACIs) suppress the proliferation of gallbladder carcinoma cell via inhibition of AKT/mTOR signaling. These findings offer a mechanistic rationale for the application of HDACIs in gallbladder carcinoma treatment.
Collapse
Affiliation(s)
- Peng Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiyong Guo
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Wu
- Department of Biostatistics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ronglin Hu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xingyuan Jiao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XJ); (XZ)
| | - Xiaofeng Zhu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XJ); (XZ)
| |
Collapse
|
21
|
Hazar-Rethinam M, de Long LM, Gannon OM, Boros S, Vargas AC, Dzienis M, Mukhopadhyay P, Saenz-Ponce N, Dantzic DDE, Simpson F, Saunders NA. RacGAP1 Is a Novel Downstream Effector of E2F7-Dependent Resistance to Doxorubicin and Is Prognostic for Overall Survival in Squamous Cell Carcinoma. Mol Cancer Ther 2015; 14:1939-50. [PMID: 26018753 DOI: 10.1158/1535-7163.mct-15-0076] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/15/2015] [Indexed: 11/16/2022]
Abstract
We have previously shown that E2F7 contributes to drug resistance in head and neck squamous cell carcinoma (HNSCC) cells. Considering that dysregulation of responses to chemotherapy-induced cytotoxicity is one of the major reasons for treatment failure in HNSCC, identifying the downstream effectors that regulate E2F7-dependent sensitivity to chemotherapeutic agents may have direct clinical impact. We used transcriptomic profiling to identify candidate pathways that contribute to E2F7-dependent resistance to doxorubicin. We then manipulated the expression of the candidate pathway using overexpression and knockdown in in vitro and in vivo models of SCC to demonstrate causality. In addition, we examined the expression of E2F7 and RacGAP1 in a custom tissue microarray (TMA) generated from HNSCC patient samples. Transcriptomic profiling identified RacGAP1 as a potential mediator of E2F7-dependent drug resistance. We validated E2F7-dependent upregulation of RacGAP1 in doxorubicin-insensitive SCC25 cells. Extending this, we found that selective upregulation of RacGAP1 induced doxorubicin resistance in previously sensitive KJDSV40. Similarly, stable knockdown of RacGAP1 in insensitive SCC25 cells induced sensitivity to doxorubicin in vitro and in vivo. RacGAP1 expression was validated in a TMA, and we showed that HNSCCs that overexpress RacGAP1 are associated with a poorer patient overall survival. Furthermore, E2F7-induced doxorubicin resistance was mediated via RacGAP1-dependent activation of AKT. Finally, we show that SCC cells deficient in RacGAP1 grow slower and are sensitized to the cytotoxic actions of doxorubicin in vivo. These findings identify RacGAP1 overexpression as a novel prognostic marker of survival and a potential target to sensitize SCC to doxorubicin.
Collapse
Affiliation(s)
- Mehlika Hazar-Rethinam
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Lilia Merida de Long
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Orla M Gannon
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Samuel Boros
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Ana Cristina Vargas
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Marcin Dzienis
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Pamela Mukhopadhyay
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Natalia Saenz-Ponce
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Daniel D E Dantzic
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nicholas A Saunders
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
22
|
Simpson DR, Mell LK, Cohen EEW. Targeting the PI3K/AKT/mTOR pathway in squamous cell carcinoma of the head and neck. Oral Oncol 2014; 51:291-8. [PMID: 25532816 DOI: 10.1016/j.oraloncology.2014.11.012] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 10/27/2014] [Accepted: 11/19/2014] [Indexed: 02/08/2023]
Abstract
Despite recent advances in novel therapies, the prognosis for patients with squamous cell carcinoma of the head and neck (SCCHN) remains poor. Progress in understanding the biology of cancer has led to the development of personalized therapy targeted at blocking defective signaling pathways of cancer cells. These drugs aim to act selectively to reduce the adverse effects associated with systemic therapy. Cetuximab (Erbitux®), an anti-epidermal growth factor receptor gene (EGFR)-targeted agent, is the only approved targeted therapy for patients with SCCHN. However, resistance to EGFR therapy remains a major obstacle to achieving a positive clinical outcome with cetuximab. Other therapies that offer better clinical outcomes in patients with advanced SCCHN are urgently needed. The phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin pathway, which is downstream of EGFR, has also been implicated in SCCHN development and progression, and therefore, targeting this pathway offers another rational treatment approach. This review discusses the potential role of PI3K pathway inhibitors in the treatment of patients with advanced SCCHN, both alone and in combination with other therapies.
Collapse
Affiliation(s)
- Daniel R Simpson
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Loren K Mell
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Ezra E W Cohen
- Department of Internal Medicine, Division of Hematology-Oncology, University of California, San Diego, La Jolla, CA, United States.
| |
Collapse
|
23
|
Hazar-Rethinam M, de Long LM, Gannon OM, Topkas E, Boros S, Vargas AC, Dzienis M, Mukhopadhyay P, Simpson F, Endo-Munoz L, Saunders NA. A novel E2F/sphingosine kinase 1 axis regulates anthracycline response in squamous cell carcinoma. Clin Cancer Res 2014; 21:417-27. [PMID: 25411162 DOI: 10.1158/1078-0432.ccr-14-1962] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Head and neck squamous cell carcinomas (HNSCC) are frequently drug resistant and have a mortality rate of 45%. We have previously shown that E2F7 may contribute to drug resistance in SCC cells. However, the mechanism and pathways involved remain unknown. EXPERIMENTAL DESIGN We used transcriptomic profiling to identify candidate pathways that may contribute to E2F7-dependent resistance to anthracyclines. We then manipulated the activity/expression of the candidate pathway using overexpression, knockdown, and pharmacological inhibitors in in vitro and in vivo models of SCC to demonstrate causality. In addition, we examined the expression of E2F7 and a downstream effector in a tissue microarray (TMA) generated from HNSCC patient samples. RESULTS E2F7-deficient keratinocytes were selectively sensitive to doxorubicin and this was reversed by overexpressing E2F7. Transcriptomic profiling identified Sphingosine kinase 1 (Sphk1) as a potential mediator of E2F7-dependent drug resistance. Knockdown and overexpression studies revealed that Sphk1 was a downstream target of E2F7. TMA studies showed that E2F7 overexpression correlated with Sphk1 overexpression in human HNSCC. Moreover, inhibition of Sphk1 by shRNA or the Sphk1-specific inhibitor, SK1-I (BML-EI411), enhanced the sensitivity of SCC cells to doxorubicin in vitro and in vivo. Furthermore, E2F7-induced doxorubicin resistance was mediated via Sphk1-dependent activation of AKT in vitro and in vivo. CONCLUSION We identify a novel drugable pathway in which E2F7 directly increases the transcription and activity of the Sphk1/S1P axis resulting in activation of AKT and subsequent drug resistance. Collectively, this novel combinatorial therapy can potentially be trialed in humans using existing agents.
Collapse
Affiliation(s)
- Mehlika Hazar-Rethinam
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Lilia Merida de Long
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Orla M Gannon
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Eleni Topkas
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Samuel Boros
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Ana Cristina Vargas
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Marcin Dzienis
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Pamela Mukhopadhyay
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Liliana Endo-Munoz
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nicholas A Saunders
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
24
|
Liang M, Lv J, Chu H, Wang J, Chen X, Zhu X, Xue Y, Guan M, Zou H. Vertical inhibition of PI3K/Akt/mTOR signaling demonstrates in vitro and in vivo anti-fibrotic activity. J Dermatol Sci 2014; 76:104-11. [PMID: 25258031 DOI: 10.1016/j.jdermsci.2014.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 07/01/2014] [Accepted: 08/05/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR) regulates cellular activity in many diseases, but the complex interplay with PI3K/Akt pathway may hampers its function. OBJECTIVE This study was undertaken to determine the activity of PI3K/Akt/mTOR signaling in the fibroblasts from systemic sclerosis (SSc) patients, and compare the effects of vertical inhibiting PI3K/Akt/mTOR by BEZ235 and inhibiting mTOR alone by rapamycin in fibroblast activation and in two complementary established mouse model of SSc. METHODS Pharmaceutical specific inhibitors BEZ235 and rapamycin were used to vertical inhibit PI3K/Akt/mTOR signaling and mTOR signaling alone in cultured fibroblasts and in mice. SSc mouse model was established by daily injecting bleomycin subcutaneously or by overexpression of constitutively active type I TGF-β receptor (TβRI(ca)). To delineate the mechanisms underlying the antifibrotic effects of BEZ235 and rapamycin, activity of PI3K/Akt/mTOR signaling was analyzed by determining the expressions of phosphorylated Akt, GSK-3β, mTOR and S6 ribosomal protein (S6). RESULTS Primary dermal fibroblasts demonstrated hyperactivity of PI3K/Akt and mTOR signaling. mTOR inhibitor rapamycin failed to inhibit dermal fibrosis in an established SSc mouse model. However, administration of a dual inhibitor for PI3K/Akt and mTOR signaling BEZ235 attenuated dermal fibrosis by reversing increased dermal thickness and collagen deposition in two SSc mouse models. Furthermore, BEZ235 showed superior inhibitory effect on fibroblast activation relative to rapamycin in vitro. Also both BEZ235 and rapamycin could prevent the phosphorylation of mTOR and S6 completely. BEZ235 also blocked the activation of Akt and GSK-3β dramatically, whereas rapamycin has been shown to increase further upregulation of phosphorylated Akt on Ser473 both in vitro and in vivo. CONCLUSION These data show that blocking PI3K/Akt/mTOR with BEZ235 leads to superior inhibitory effect for dermal fibrosis, suggesting that vertical inhibition of PI3K/Akt/mTOR signaling may have therapeutic potential for SSc.
Collapse
Affiliation(s)
- Minrui Liang
- Division of Rheumatology, Huashan Hospital, Shanghai 200040, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China
| | - Jiaoyan Lv
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Haiyan Chu
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China; Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Jiucun Wang
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China; Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xiangjun Chen
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China; Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaoxia Zhu
- Division of Rheumatology, Huashan Hospital, Shanghai 200040, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China
| | - Yu Xue
- Division of Rheumatology, Huashan Hospital, Shanghai 200040, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China
| | - Ming Guan
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China; Department of Clinical Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital, Shanghai 200040, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai 200040, China.
| |
Collapse
|
25
|
Mazumdar T, Byers LA, Ng PKS, Mills GB, Peng S, Diao L, Fan YH, Stemke-Hale K, Heymach JV, Myers JN, Glisson BS, Johnson FM. A comprehensive evaluation of biomarkers predictive of response to PI3K inhibitors and of resistance mechanisms in head and neck squamous cell carcinoma. Mol Cancer Ther 2014; 13:2738-50. [PMID: 25193510 DOI: 10.1158/1535-7163.mct-13-1090] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The PI3K/AKT/mTOR pathway is frequently activated in head and neck squamous cell carcinoma (HNSCC), but pathway inhibition has variable efficacy. Identification of predictive biomarkers and mechanisms of resistance would allow selection of patients most likely to respond and novel therapeutic combinations. The purpose of this study was to extend recent discoveries regarding the PI3K/AKT/mTOR pathway in HNSCC by more broadly examining potential biomarkers of response, by examining pathway inhibitors with a diverse range of targets, and by defining mechanisms of resistance and potential combination therapies. We used reverse-phase protein arrays (RPPA) to simultaneously evaluate expression of 195 proteins; SNP array to estimate gene copy number; and mass array to identify mutations. We examined altered signaling at baseline and after pathway inhibition. Likewise, we examined the activation of the PI3K/AKT/mTOR pathway in HNSCC tumors by RPPA. Cell lines with PIK3CA mutations were sensitive to pathway inhibitors, whereas amplification status did not predict sensitivity. While we identified a set of individual candidate biomarkers of response to pathway inhibitors, proteomic pathway scores did not correlate with amplification or mutation and did not predict response. Several receptor tyrosine kinases, including EGFR and ERK, were activated following PI3K inhibition in resistant cells; dual pathway inhibition of PI3K and EGFR or MEK demonstrated synergy. Combined MEK and PI3K inhibition was markedly synergistic in HRAS-mutant cell lines. Our findings indicate that clinical trials of single-agent PI3K/AKT/mTOR pathway inhibitors in selected populations and of PI3K/EGFR or PI3K/MEK inhibitor combinations are warranted; we plan to conduct such trials.
Collapse
Affiliation(s)
- Tuhina Mazumdar
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren A Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Patrick Kwok Shing Ng
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas. Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shaohua Peng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - You-Hong Fan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katherine Stemke-Hale
- Department of Translational Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Jeffrey N Myers
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas. Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bonnie S Glisson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Faye M Johnson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences, Houston, Texas.
| |
Collapse
|
26
|
Zhao X, Tan Q, Zhang Z, Zhao Y. 1,3,5-Triazine inhibitors of histone deacetylases: synthesis and biological activity. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1084-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
27
|
Papagerakis S, Pannone G, Zheng L, About I, Taqi N, Nguyen NPT, Matossian M, McAlpin B, Santoro A, McHugh J, Prince ME, Papagerakis P. Oral epithelial stem cells - implications in normal development and cancer metastasis. Exp Cell Res 2014; 325:111-29. [PMID: 24803391 DOI: 10.1016/j.yexcr.2014.04.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 12/18/2022]
Abstract
Oral mucosa is continuously exposed to environmental forces and has to be constantly renewed. Accordingly, the oral mucosa epithelium contains a large reservoir of epithelial stem cells necessary for tissue homeostasis. Despite considerable scientific advances in stem cell behavior in a number of tissues, fewer studies have been devoted to the stem cells in the oral epithelium. Most of oral mucosa stem cells studies are focused on identifying cancer stem cells (CSC) in oral squamous cell carcinomas (OSCCs) among other head and neck cancers. OSCCs are the most prevalent epithelial tumors of the head and neck region, marked by their aggressiveness and invasiveness. Due to their highly tumorigenic properties, it has been suggested that CSC may be the critical population of cancer cells in the development of OSCC metastasis. This review presents a brief overview of epithelium stem cells with implications in oral health, and the clinical implications of the CSC concept in OSCC metastatic dissemination.
Collapse
Affiliation(s)
- Silvana Papagerakis
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA; Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Li Zheng
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA; Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Imad About
- Aix-Marseille Université, CNRS, ISM UMR 7287, 13288, Marseille cedex 09, France
| | - Nawar Taqi
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Nghia P T Nguyen
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Margarite Matossian
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Blake McAlpin
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA; Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Angela Santoro
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Jonathan McHugh
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Mark E Prince
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Petros Papagerakis
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Center for Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, MI, USA; Center for Organogenesis, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
28
|
Overcoming inherent resistance to histone deacetylase inhibitors in multiple myeloma cells by targeting pathways integral to the actin cytoskeleton. Cell Death Dis 2014; 5:e1134. [PMID: 24651437 PMCID: PMC3973216 DOI: 10.1038/cddis.2014.98] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/22/2014] [Accepted: 02/10/2014] [Indexed: 01/05/2023]
Abstract
Histone deacetylase inhibitors (HDACi) are novel chemotherapeutics undergoing evaluation in clinical trials for the potential treatment of patients with multiple myeloma (MM). Although HDACi have demonstrable synergy when combined with proteasome inhibitors (PIs), recent evidence indicates that combination of HDACi and PI is beneficial only in a subset of patients with advanced MM, clearly indicating that other rational combinations should be explored. In this context we hypothesized that understanding the molecular signature associated with inherent resistance to HDACi would provide a basis for the identification of therapeutic combinations with improved clinical efficacy. Using human myeloma cell lines (HMCL) categorized as sensitive, intermediate or resistant to HDACi, gene expression profiling (GEP) and gene ontology enrichment analyses were performed to determine if a genetic signature associated with inherent resistance to HDACi-resistance could be identified. Correlation of GEP to increasing or decreasing sensitivity to HDACi indicated a unique 35-gene signature that was significantly enriched for two pathways – regulation of actin cytoskeleton and protein processing in endoplasmic reticulum. When HMCL and primary MM samples were treated with a combination of HDACi and agents targeting the signaling pathways integral to the actin cytoskeleton, synergistic cell death was observed in all instances, thus providing a rationale for combining these agents with HDACi for the treatment of MM to overcome resistance. This report validates a molecular approach for the identification of HDACi partner drugs and provides an experimental framework for the identification of novel therapeutic combinations for anti-MM treatment.
Collapse
|
29
|
Wanitchakool P, Wolf L, Koehl GE, Sirianant L, Schreiber R, Kulkarni S, Duvvuri U, Kunzelmann K. Role of anoctamins in cancer and apoptosis. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130096. [PMID: 24493744 PMCID: PMC3917350 DOI: 10.1098/rstb.2013.0096] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Anoctamin 1 (TMEM16A, Ano1) is a recently identified Ca(2+)-activated chloride channel and a member of a large protein family comprising 10 paralogues. Before Ano1 was identified as a chloride channel protein, it was known as the cancer marker DOG1. DOG1/Ano1 is expressed in gastrointestinal stromal tumours (GIST) and particularly in head and neck squamous cell carcinoma, at very high levels never detected in other tissues. It is now emerging that Ano1 is part of the 11q13 locus, amplified in several types of tumour, where it is thought to augment cell proliferation, cell migration and metastasis. Notably, Ano1 is upregulated through histone deacetylase (HDAC), corresponding to the known role of HDAC in HNSCC. As Ano1 does not enhance proliferation in every cell type, its function is perhaps modulated by cell-specific factors, or by the abundance of other anoctamins. Thus Ano6, by regulating Ca(2+)-induced membrane phospholipid scrambling and annexin V binding, supports cellular apoptosis rather than proliferation. Current findings implicate other cellular functions of anoctamins, apart from their role as Ca(2+)-activated Cl(-) channels.
Collapse
Affiliation(s)
- Podchanart Wanitchakool
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, Regensburg 93053, Germany
| | - Luisa Wolf
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, Regensburg 93053, Germany
| | - Gudrun E. Koehl
- Department of Surgery, University Medical Center Regensburg, University of Regensburg, Regensburg, Germany
| | - Lalida Sirianant
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, Regensburg 93053, Germany
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, Regensburg 93053, Germany
| | - Sucheta Kulkarni
- Ear & Eye Institute, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Umamaheswar Duvvuri
- Ear & Eye Institute, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, Regensburg 93053, Germany
| |
Collapse
|
30
|
Shah AT, Demory Beckler M, Walsh AJ, Jones WP, Pohlmann PR, Skala MC. Optical metabolic imaging of treatment response in human head and neck squamous cell carcinoma. PLoS One 2014; 9:e90746. [PMID: 24595244 PMCID: PMC3942493 DOI: 10.1371/journal.pone.0090746] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 02/05/2014] [Indexed: 11/26/2022] Open
Abstract
Optical metabolic imaging measures fluorescence intensity and lifetimes from metabolic cofactors nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD). These molecular level measurements provide unique biomarkers for early cellular responses to cancer treatments. Head and neck squamous cell carcinoma (HNSCC) is an attractive target for optical imaging because of easy access to the site using fiber optic probes. Two HNSCC cell lines, SCC25 and SCC61, were treated with Cetuximab (anti-EGFR antibody), BGT226 (PI3K/mTOR inhibitor), or cisplatin (chemotherapy) for 24 hours. Results show increased redox ratio, NADH α1 (contribution from free NADH), and FAD α1 (contribution from protein-bound FAD) for malignant cells compared with the nonmalignant cell line OKF6 (p<0.05). In SCC25 and SCC61 cells, the redox ratio is unaffected by cetuximab treatment and decreases with BGT226 and cisplatin treatment (p<0.05), and these results agree with standard measurements of proliferation rates after treatment. For SCC25, NADH α1 is reduced with BGT226 and cisplatin treatment. For SCC61, NADH α1 is reduced with cetuximab, BGT226, and cisplatin treatment. Trends in NADH α1 are statistically similar to changes in standard measurements of glycolytic rates after treatment. FAD α1 is reduced with cisplatin treatment (p<0.05). These shifts in optical endpoints reflect early metabolic changes induced by drug treatment. Overall, these results indicate that optical metabolic imaging has potential to detect early response to cancer treatment in HNSCC, enabling optimal treatment regimens and improved patient outcomes.
Collapse
Affiliation(s)
- Amy T. Shah
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Michelle Demory Beckler
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Radiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alex J. Walsh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - William P. Jones
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Paula R. Pohlmann
- Department of Medicine, Georgetown University Medical Center, Washington, District of Columbia, United States of America
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
31
|
Wilson-Edell KA, Yevtushenko MA, Rothschild DE, Rogers AN, Benz CC. mTORC1/C2 and pan-HDAC inhibitors synergistically impair breast cancer growth by convergent AKT and polysome inhibiting mechanisms. Breast Cancer Res Treat 2014; 144:287-298. [PMID: 24562770 DOI: 10.1007/s10549-014-2877-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 02/08/2014] [Indexed: 02/06/2023]
Abstract
Resistance of breast cancers to targeted hormone receptor (HR) or human epidermal growth factor receptor 2 (HER2) inhibitors often occurs through dysregulation of the phosphoinositide 3-kinase, protein kinase B/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway. Presently, no targeted therapies exist for breast cancers lacking HR and HER2 overexpression, many of which also exhibit PI3K/AKT/mTOR hyper-activation. Resistance of breast cancers to current therapeutics also results, in part, from aberrant epigenetic modifications including protein acetylation regulated by histone deacetylases (HDACs). We show that the investigational drug MLN0128, which inhibits both complexes of mTOR (mTORC1 and mTORC2), and the hydroxamic acid pan-HDAC inhibitor TSA synergistically inhibit the viability of a phenotypically diverse panel of five breast cancer cell lines (HR-/+, HER2-/+). The combination of MLN0128 and TSA induces apoptosis in most breast cancer cell lines tested, but not in the non-malignant MCF-10A mammary epithelial cells. In parallel, the MLN0128/TSA combination reduces phosphorylation of AKT at S473 more than single agents alone and more so in the 5 malignant breast cancer cell lines than in the non-malignant mammary epithelial cells. Examining polysome profiles from one of the most sensitive breast cancer cell lines (SKBR3), we demonstrate that this MLN0128/TSA treatment combination synergistically impairs polysome assembly in conjunction with enhanced inhibition of 4eBP1 phosphorylation at S65. Taken together, these data indicate that the synergistic growth inhibiting consequence of combining a mTORC1/C2 inhibitor like MLN0128 with a pan-HDAC inhibitor like TSA results from their mechanistic convergence onto the PI3K/AKT/mTOR pathway, profoundly inhibiting both AKT S473 and 4eBP1 S65 phosphorylation, reducing polysome formation and cancer cell viability.
Collapse
Affiliation(s)
| | - Mariya A Yevtushenko
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA; Master of Science in Biology Program, Dominican University, San Rafael, CA, USA
| | - Daniel E Rothschild
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Aric N Rogers
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Christopher C Benz
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA,
| |
Collapse
|
32
|
Tan Q, Zhang Z, Hui J, Zhao Y, Zhu L. Synthesis and anticancer activities of thieno[3,2-d]pyrimidines as novel HDAC inhibitors. Bioorg Med Chem 2014; 22:358-65. [DOI: 10.1016/j.bmc.2013.11.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 11/06/2013] [Accepted: 11/08/2013] [Indexed: 10/26/2022]
|
33
|
PIK3CA, HRAS and PTEN in human papillomavirus positive oropharyngeal squamous cell carcinoma. BMC Cancer 2013; 13:602. [PMID: 24341335 PMCID: PMC3878565 DOI: 10.1186/1471-2407-13-602] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 12/06/2013] [Indexed: 12/11/2022] Open
Abstract
Background Recent genomic evidence suggests frequent phosphatidylinositide 3-kinase (PI3K) pathway activation in human papillomavirus (HPV) positive oropharyngeal squamous cell carcinoma. Mutations/amplification of the gene encoding p110α catalytic subunit of phosphoinositide 3-kinase (PIK3CA), loss of phosphatase and tensin homolog (PTEN) and HRAS mutations are known to activate PI3K pathway. Methods and results PIK3CA mutations were identified by Sanger sequencing in 23 of 75 (31%) HPV-positive oropharyngeal carcinomas, including exon 9 (p.E545K [n = 10] and p.E542K [n = 5]) or exon 20 (p.H1047Y, n = 2) mutations. Five rare and one novel (p.R537Q) PIK3CA mutations were identified. HRAS mutation (p.Q61L) was detected in 1 of 62 tested cases. PIK3CA amplification by fluorescence in situ hybridization (FISH) was identified in 4 cases (4/21, 20%), while PTEN loss was seen in 7 (7/21, 33%) cases (chromosome 10 monosomy [n = 4], homozygous deletion [n = 3]). Conclusions Overall, genetic alterations that likely lead to PI3K pathway activation were identified in 34 of 75 cases (45%) and did not correlate with disease specific survival. These findings offer a molecular rationale for therapeutic targeting of PI3K pathway in patients with HPV-positive oropharyngeal carcinoma.
Collapse
|
34
|
Human papillomavirus-associated adenocarcinoma of the base of tongue: potentially actionable genetic changes. Head Neck Pathol 2013; 8:151-6. [PMID: 24258618 PMCID: PMC4022938 DOI: 10.1007/s12105-013-0508-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 11/13/2013] [Indexed: 01/30/2023]
Abstract
While human papillomavirus (HPV)-positive squamous and adenosquamous carcinomas of the oropharynx have been well characterized, HPV-associated pure adenocarcinomas are exceptionally rare. Herein we report the clinicopathologic features of one such HPV-associated adenocarcinoma of the base of tongue (BOT). A 70 year-old male presented with a 2.8 cm base of tongue mass and lymphadenopathy. Immunohistochemically, the adenocarcinoma was p63 negative and p16 positive. HPV positivity was shown by in situ hybridization. Features of salivary type tumor or metastasis from a distant primary were absent. IonTorrent™ semiconductor sequencing analysis for 739 cancer-associated mutations in 46 actionable cancer genes was performed and PIK3CA exon 9 (p.E545K) and MET exon 2 (p.E168D) mutations were identified. No PIK3CA or MET amplification was identified by fluorescence in situ hybridization. A re-review of archival HPV-positive oropharyngeal squamous cell carcinomas (n = 89, 1983-2013) showed no additional cases of adenocarcinoma. The clinical follow-up for the three previously reported cases of HPV-associated adenocarcinoma of the BOT was updated. All previously reported cases were tested and were negative for PIK3CA exon 9 and 20 and MET exon 2 mutations. These findings offer a molecular basis for potential therapeutic use of PIK3CA inhibitors in a subset of patients with HPV-associated adenocarcinoma of BOT.
Collapse
|
35
|
Anne M, Sammartino D, Barginear MF, Budman D. Profile of panobinostat and its potential for treatment in solid tumors: an update. Onco Targets Ther 2013; 6:1613-24. [PMID: 24265556 PMCID: PMC3833618 DOI: 10.2147/ott.s30773] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The histone deacetylase (HDAC) inhibitors have emerged as novel therapies for cancer. Panobinostat (LBH 589, Novartis Pharmaceuticals) is a pan-deacetylase inhibitor that is being evaluated in both intravenous and oral formulations across multiple tumor types. Comparable to the other HDACs, panobinostat leads to hyperacetylation of histones and other intracellular proteins, allowing for the expression of otherwise repressed genes, leading to inhibition of cellular proliferation and induction of apoptosis in malignant cells. Panobinostat, analogous to other HDAC inhibitors, also induces apoptosis by directly activating cellular death receptor pathways. Preclinical data suggests that panobinostat has inhibitory activity at nanomolar concentrations and appears to be the most potent clinically available HDAC inhibitor. Here we review the current status of panobinostat and discuss its role in the treatment of solid tumors.
Collapse
Affiliation(s)
- Madhurima Anne
- Monter Cancer Center, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| | - Daniel Sammartino
- Department of Medicine, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| | - Myra F Barginear
- Monter Cancer Center, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| | - Daniel Budman
- Monter Cancer Center, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| |
Collapse
|
36
|
Herzog A, Bian Y, Broek RV, Hall B, Coupar J, Cheng H, Sowers AL, Cook JD, Mitchell JB, Chen Z, Kulkarni AB, VanWaes C. PI3K/mTOR inhibitor PF-04691502 antitumor activity is enhanced with induction of wild-type TP53 in human xenograft and murine knockout models of head and neck cancer. Clin Cancer Res 2013; 19:3808-19. [PMID: 23640975 PMCID: PMC3715575 DOI: 10.1158/1078-0432.ccr-12-2716] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway activation is often associated with altered expression or mutations of PIK3CA, TP53/p73, PTEN, and TGF-β receptors (TGFBR) in head and neck squamous cell carcinomas (HNSCC). However, little is known about how these alterations affect response to PI3K/mTOR-targeted agents. EXPERIMENTAL DESIGN In this preclinical study, PI3K/Akt/mTOR signaling was characterized in nine HNSCC (UM-SCC) cell lines and human oral keratinocytes. We investigated the molecular and anticancer effects of dual PI3K/mTOR inhibitor PF-04691502(PF-502) in UM-SCC expressing PIK3CA with decreased wild-type TP53, mutant TP53-/+ mutantTGFBR2, and in HNSCC of a conditional Pten/Tgfbr1 double knockout mouse model displaying PI3K/Akt/mTOR activation. RESULTS UM-SCC showed increased PIK3CA expression and Akt/mTOR activation, and PF-502 inhibited PI3K/mTORC1/2 targets. In human HNSCC expressing PIK3CA and decreased wtTP53 and p73, PF-502 reciprocally enhanced TP53/p73 expression and growth inhibition, which was partially reversible by p53 inhibitor pifithrin-α. Most UM-SCC with wtTP53 exhibited a lower IC50 than those with mtTP53 status. PF-502 blocked growth in G0-G1 and increased apoptotic sub-G0 DNA. PF-502 suppressed tumorigenesis and showed combinatorial activity with radiation in a wild-type TP53 UM-SCC xenograft model. PF-502 also significantly delayed HNSCC tumorigenesis and prolonged survival of Pten/Tgfbr1-deficient mice. Significant inhibition of p-Akt, p-4EBP1, p-S6, and Ki67, as well as increased p53 and TUNEL were observed in tumor specimens. CONCLUSIONS PI3K-mTOR inhibition can enhance TP53/p73 expression and significantly inhibit tumor growth alone or when combined with radiation in HNSCC with wild-type TP53. PIK3CA, TP53/p73, PTEN, and TGF-β alterations are potential modifiers of response and merit investigation in future clinical trials with PI3K-mTOR inhibitors.
Collapse
Affiliation(s)
- Amanda Herzog
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH
- HHMI-NIH Research Scholars Program/NIH Medical Research Scholars Program
| | - Yansong Bian
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH
| | - Robert Vander Broek
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH
- HHMI-NIH Research Scholars Program/NIH Medical Research Scholars Program
| | - Bradford Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, Bethesda, MD
| | - Jamie Coupar
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH
| | - Hui Cheng
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH
| | | | - John D. Cook
- Radiation Biology Branch, National Cancer Institute, Bethesda, MD, USA
| | - James B. Mitchell
- Radiation Biology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH
| | - Ashok B. Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, Bethesda, MD
| | - Carter VanWaes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH
| |
Collapse
|
37
|
Thomas S, Thurn KT, Raha P, Chen S, Munster PN. Efficacy of histone deacetylase and estrogen receptor inhibition in breast cancer cells due to concerted down regulation of Akt. PLoS One 2013; 8:e68973. [PMID: 23874830 PMCID: PMC3711340 DOI: 10.1371/journal.pone.0068973] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/03/2013] [Indexed: 12/22/2022] Open
Abstract
Hormonal therapy resistance remains a considerable barrier in the treatment of
breast cancer. Activation of the Akt-PI3K-mTOR pathway plays an important role
in hormonal therapy resistance. Our recent preclinical and clinical studies
showed that the addition of a histone deacetylase inhibitor re-sensitized
hormonal therapy resistant breast cancer to tamoxifen. As histone deacetylases
are key regulators of Akt, we evaluated the effect of combined treatment with
the histone deacetylase inhibitor PCI-24781 and tamoxifen on Akt in breast
cancer cells. We demonstrate that while both histone deacetylase and estrogen
receptor inhibition down regulate AKT mRNA and protein, their concerted effort
results in down regulation of AKT activity with induction of cell death. Histone
deacetylase inhibition exerts its effect on AKT mRNA through an estrogen
receptor-dependent mechanism, primarily down regulating the most abundant
isoform AKT1. Although siRNA depletion of AKT modestly induces cell death, when
combined with an anti-estrogen, cytotoxicity is significantly enhanced. Thus,
histone deacetylase regulation of AKT mRNA is a key mediator of this therapeutic
combination and may represent a novel biomarker for predicting response to this
regimen.
Collapse
Affiliation(s)
- Scott Thomas
- Division of Hematology and Oncology, Department of Medicine,
University of California San Francisco, San Francisco, California, United States
of America
| | - K. Ted Thurn
- Division of Hematology and Oncology, Department of Medicine,
University of California San Francisco, San Francisco, California, United States
of America
| | - Paromita Raha
- Division of Hematology and Oncology, Department of Medicine,
University of California San Francisco, San Francisco, California, United States
of America
| | - Stephanie Chen
- Division of Hematology and Oncology, Department of Medicine,
University of California San Francisco, San Francisco, California, United States
of America
| | - Pamela N. Munster
- Division of Hematology and Oncology, Department of Medicine,
University of California San Francisco, San Francisco, California, United States
of America
- * E-mail:
| |
Collapse
|
38
|
Abstract
In recent years, histone deacetylase inhibitors (HDACis), a novel class of agents that targets mechanistic abnormalities in cancers, have shown promising anti-cancer activity in both hematological and solid cancers. Among them, vorinostat was approved by FDA to treat cutaneous T-cell lymphoma and is being evaluated in other cancer types. Although initially designed to target histone deacetylase, vorinostat were found to have additional effects on other epigenetic machineries, for example acetylation of non-HDAC, methylation and microRNA (miRNA) expression. In this review, we examined all known mechanisms of action for vorinostat. We also summarized the current findings on the `crosstalk' between different epigenetic machineries. These findings suggest that improved understanding of epigenetic regulatory role of vorinostat and/or other HDACis will provide novel insights in improving utilization of this class of novel agents.
Collapse
Affiliation(s)
- Jean Lee
- Department of Medicine, University of Chicago, USA
| | | |
Collapse
|
39
|
Wilson PM, Labonte MJ, Martin SC, Kuwahara ST, El-Khoueiry A, Lenz HJ, Ladner RD. Sustained inhibition of deacetylases is required for the antitumor activity of the histone deactylase inhibitors panobinostat and vorinostat in models of colorectal cancer. Invest New Drugs 2013; 31:845-57. [PMID: 23299388 DOI: 10.1007/s10637-012-9914-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 12/06/2012] [Indexed: 12/20/2022]
Abstract
Despite compelling preclinical data in colorectal cancer (CRC), the efficacy of HDACIs has been disappointing in the clinic. The goal of this study was to evaluate the effectiveness of vorinostat and panobinostat in a dose- and exposure-dependent manner in order to better understand the dynamics of drug action and antitumor efficacy. In a standard 72 h drug exposure MTS assay, notable concentration-dependent antiproliferative effects were observed in the IC50 range of 1.2-2.8 μmol/L for vorinostat and 5.1-17.5 nmol/L for panobinostat. However, shorter clinically relevant exposures of 3 or 6 h failed to elicit any significant growth inhibition and in most cases a >24 h exposure to vorinostat or panobinostat was required to induce a sigmoidal dose-response. Similar results were observed in colony formation assays where ≥ 24 h of exposure was required to effectively reduce colony formation. Induction of acetyl-H3, acetyl-H4 and p21 by vorinostat were transient and rapidly reversed within 12 h of drug removal. In contrast, panobinostat-induced acetyl-H3, acetyl-H4, and p21 persisted for 48 h after an initial 3 h exposure. Treatment of HCT116 xenografts with panobinostat induced significant increases in acetyl-H3 and downregulation of thymidylate synthase after treatment. Although HDACIs exert both potent growth inhibition and cytotoxic effects when CRC cells were exposed to drug for ≥ 24 h, these cells demonstrate an inherent ability to survive HDACI concentrations and exposure times that exceed those clinically achievable. Continued efforts to develop novel HDACIs with improved pharmacokinetics/phamacodynamics, enhanced intratumoral delivery and class/isoform-specificity are needed to improve the therapeutic potential of HDACIs and HDACI-based combination regimens in solid tumors.
Collapse
Affiliation(s)
- Peter M Wilson
- Department of Pathology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Epigenetic modifications constitute the next frontier in tumor biology research. Post-translation modification of histones dynamically influences gene expression independent of alterations to the DNA sequence. These mechanisms are often mediated by histone linkers or by proteins associated with the recruitment of DNA-binding proteins, HDAC I and II interacting proteins and transcriptional activators, coactivators or corepressors. Early evidence suggested that histones and their modifiers are involved in sophisticated processes that modulate tumor behavior and cellular phenotype. In this review, we discuss how recent discoveries about chromatin modifications, particularly histone acetylation, are shaping our knowledge of cell biology and our understanding of the molecular circuitry governing tumor progression and consider whether recent insights may extend to novel therapeutic approaches. Furthermore, we discuss the latest oncogenomic findings in Head and Neck Squamous Cell Carcinoma (HNSCC) from studies using Next Generation Sequencing (NGS) technology and highlight the impact of mutations identified in histones and their modifiers.
Collapse
Affiliation(s)
- Manoela D Martins
- Department of Oral Pathology, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rogerio M Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
41
|
Vorinostat, an HDAC inhibitor attenuates epidermoid squamous cell carcinoma growth by dampening mTOR signaling pathway in a human xenograft murine model. Toxicol Appl Pharmacol 2012; 266:233-44. [PMID: 23147569 DOI: 10.1016/j.taap.2012.11.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/01/2012] [Accepted: 11/02/2012] [Indexed: 01/16/2023]
Abstract
Histone deacetylase (HDAC) inhibitors are potent anticancer agents and show efficacy against various human neoplasms. Vorinostat is a potent HDAC inhibitor and has shown potential to inhibit growth of human xenograft tumors. However, its effect on the growth of skin neoplasm remains undefined. In this study, we show that vorinostat (2 μM) reduced expression of HDAC1, 2, 3, and 7 in epidermoid carcinoma A431 cells. Consistently, it increased acetylation of histone H3 and p53. Vorinostat (100mg/kg body weight, IP) treatment reduced human xenograft tumor growth in highly immunosuppressed nu/nu mice. Histologically, the vorinostat-treated tumor showed features of well-differentiation with large necrotic areas. Based on proliferating cell nuclear antigen (PCNA) staining and expression of cyclins D1, D2, E, and A, vorinostat seems to impair proliferation by down-regulating the expression of these proteins. However, it also induced apoptosis. The mechanism by which vorinostat blocks proliferation and makes tumor cells prone to apoptosis, involved inhibition of mTOR signaling which was accompanied by reduction in cell survival AKT and extracellular-signal regulated kinase (ERK) signaling pathways. Our data provide a novel mechanism-based therapeutic intervention for cutaneous squamous cell carcinoma (SCC). Vorinostat may be utilized to cure skin neoplasms in organ transplant recipient (OTR). These patients have high morbidity and surgical removal of these lesions which frequently develop in these patients, is difficult.
Collapse
|
42
|
Cameron S, de Long LM, Hazar-Rethinam M, Topkas E, Endo-Munoz L, Cumming A, Gannon O, Guminski A, Saunders N. Focal overexpression of CEACAM6 contributes to enhanced tumourigenesis in head and neck cancer via suppression of apoptosis. Mol Cancer 2012; 11:74. [PMID: 23021083 PMCID: PMC3515475 DOI: 10.1186/1476-4598-11-74] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/18/2012] [Indexed: 12/22/2022] Open
Abstract
Background Overexpression of CEACAM6 has been reported for a number of malignancies. However, the mechanism of how CEACAM6 contributes to cancer formation and its role in head and neck squamous cell carcinoma (HNSCC) remains unclear. Therefore, we examined the role of CEACAM6 in head and neck squamous cell carcinoma (HNSCC). Methods CEACAM6 expression was examined in normal squamous epithelia as well as a number of patient HNSCC samples and tumours derived from HNSCC cell lines injected into NOD/SCID mice. CEACAM6 expression was manipulated in HNSCC cell lines by shRNA-mediated CEACAM6 knockdown or virally-delivered overexpression of CEACAM6. The role of CEACAM6 in tumour growth and chemotherapeutic sensitivity was then assessed in vivo and in vitro respectively. Results CEACAM6 expression was significantly increased in highly tumourigenic HNSCC cell lines when compared to poorly tumourigenic HNSCC cell lines. Moreover, HNSCC patient tumours demonstrated focal expression of CEACAM6. Functional investigation of CEACAM6, involving over-expression and knock down studies, demonstrated that CEACAM6 over-expression could enhance tumour initiating activity and tumour growth via activation of AKT and suppression of caspase-3 mediated cell death. Conclusion We report that CEACAM6 is focally overexpressed in a large fraction of human HNSCCs in situ. We also show that over-expression of CEACAM6 increases tumour growth and tumour initiating activity by suppressing PI3K/AKT-dependent apoptosis of HNSCC in a xenotransplant model of HNSCC. Finally, our studies indicate that foci of CEACAM6 expressing cells are selectively ablated by treatment of xenotransplant tumours with pharmacological inhibitors of PI3K/AKT in vivo.
Collapse
Affiliation(s)
- Sarina Cameron
- University of Queensland Diamantina Institute, Epithelial Pathobiology Group, Princess Alexandra Hospital, Queensland, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Baiz D, Pinder TA, Hassan S, Karpova Y, Salsbury F, Welker ME, Kulik G. Synthesis and characterization of a novel prostate cancer-targeted phosphatidylinositol-3-kinase inhibitor prodrug. J Med Chem 2012; 55:8038-46. [PMID: 22924393 DOI: 10.1021/jm300881a] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The phosphatidylinositol-3-kinase/Akt (PI3K/Akt) pathway is constitutively activated in a substantial proportion of prostate tumors and is considered a key mechanism supporting progression toward an androgen-independent status, for which no effective therapy is available. Therefore, PI3K inhibitors, alone or in combination with other cytotoxic drugs, could potentially be used to treat cancer with a constitutive activated PI3K/Akt pathway. To selectively target advanced prostate tumors with a constitutive activated PI3K/Akt pathway, a prostate cancer-specific PI3K inhibitor was generated by coupling the chemically modified form of the quercetin analogue LY294002 (HO-CH(2)-LY294002, compound 8) with the peptide Mu-LEHSSKLQL, in which the internal sequence HSSKLQ is a substrate for the prostate-specific antigen (PSA) protease. The result is a water-soluble and latent PI3K inhibitor prodrug (compound 11), its activation being dependent on PSA cleavage. Once activated, the L-O-CH(2)-LY294002 (compound 10) can specifically inhibit PI3K in PSA-secreting prostate cancer cells and induce apoptosis with a potency comparable to that of the original LY294002 compound.
Collapse
Affiliation(s)
- Daniele Baiz
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Saunders NA, Simpson F, Thompson EW, Hill MM, Endo-Munoz L, Leggatt G, Minchin RF, Guminski A. Role of intratumoural heterogeneity in cancer drug resistance: molecular and clinical perspectives. EMBO Mol Med 2012; 4:675-84. [PMID: 22733553 PMCID: PMC3494067 DOI: 10.1002/emmm.201101131] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 04/22/2012] [Accepted: 04/30/2012] [Indexed: 12/15/2022] Open
Abstract
Drug resistance continues to be a major barrier to the delivery of curative therapies in cancer. Historically, drug resistance has been associated with over-expression of drug transporters, changes in drug kinetics or amplification of drug targets. However, the emergence of resistance in patients treated with new-targeted therapies has provided new insight into the complexities underlying cancer drug resistance. Recent data now implicate intratumoural heterogeneity as a major driver of drug resistance. Single cell sequencing studies that identified multiple genetically distinct variants within human tumours clearly demonstrate the heterogeneous nature of human tumours. The major contributors to intratumoural heterogeneity are (i) genetic variation, (ii) stochastic processes, (iii) the microenvironment and (iv) cell and tissue plasticity. Each of these factors impacts on drug sensitivity. To deliver curative therapies to patients, modification of current therapeutic strategies to include methods that estimate intratumoural heterogeneity and plasticity will be essential.
Collapse
Affiliation(s)
- Nicholas A Saunders
- Epithelial Cancer Program, University of Queensland, Diamantine Institute, Princess Alexandra Hospital, Queensland, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Specenier P, Vermorken JB. Biologic therapy in head and neck cancer: a road with hurdles. ISRN ONCOLOGY 2012; 2012:163752. [PMID: 22745915 PMCID: PMC3382358 DOI: 10.5402/2012/163752] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 04/11/2012] [Indexed: 12/28/2022]
Abstract
The epidermal growth factor receptor (EGFR) is overexpressed in the vast majority of cases of squamous cell carcinoma of the head and neck (SCCHN). A high EGFR expression is associated with an unfavorable prognosis. Cetuximab is a chimeric human/murine IgG1 antibody which binds with high affinity to the EGFR. It is the only targeted agent which got approval for the treatment of SCCHN from the regulatory agencies of Europe and the United States, both in locoregionally advanced disease, in association with radiation, and in recurrent/metastatic disease. The outcome of trials involving other EGFR-directed monoclonal antibodies, that is, zalutumumab and panitumumab, was consistent with the results with cetuximab. However these trials failed to meet their primary endpoint. The results with EGFR-directed tyrosine kinase inhibitors have been disappointing. Other potential targets for treatment in SCCHN include the entire ErbB family, the vascular endothelial growth factor (VEGF) and its receptor (VEGFR), the insulin-like growth factor 1 receptor (IGF-1R), the insulin receptor (IR), histone deacetylases (HDAC), the mammalian target of rapamycin (mTOR), the platelet-derived growth factor receptor (PDGFR), heat-shock protein 90 (HSP90), nuclear factor-kappa B (NF-κB), aurora A or B, and phosphatidylinositol 3-kinase (PIK3CA).
Collapse
Affiliation(s)
- Pol Specenier
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Jan B. Vermorken
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| |
Collapse
|
46
|
Abstract
Histone deacetylase inhibitors (HDACIs) are epigenetically acting agents that modify chromatin structure and by extension, gene expression. However, they may influence the behavior and survival of transformed cells by diverse mechanisms, including promoting expression of death- or differentiation-inducing genes while downregulating the expression of prosurvival genes; acting directly to increase oxidative injury and DNA damage; acetylating and disrupting the function of multiple proteins, including DNA repair and chaperone proteins; and interfering with the function of corepressor complexes. Notably, HDACIs have been shown in preclinical studies to target transformed cells selectively, and these agents have been approved in the treatment of certain hematologic malignancies, for example, cutaneous T-cell lymphoma and peripheral T-cell lymphoma. However, attempts to extend the spectrum of HDACI activity to other malignancies, for example, solid tumors, have been challenging. This has led to the perception that HDACIs may have limited activity as single agents. Because of the pleiotropic actions of HDACIs, combinations with other antineoplastic drugs, particularly other targeted agents, represent a particularly promising avenue of investigation. It is likely that emerging insights into mechanism(s) of HDACI activity will allow optimization of this approach, and hopefully, will expand HDACI approvals to additional malignancies in the future.
Collapse
Affiliation(s)
- Steven Grant
- Division of Hematology/Oncology, Virginia Commonwealth University Health Sciences Center, Richmond, Virginia, USA.
| | | |
Collapse
|