1
|
Kinney BLC, Gunti S, Kansal V, Parrish CJ, Saba NF, Teng Y, Henry MK, Su FY, Kwong GA, Schmitt NC. Rescue of NLRC5 expression restores antigen processing machinery in head and neck cancer cells lacking functional STAT1 and p53. Cancer Immunol Immunother 2024; 73:10. [PMID: 38231444 PMCID: PMC10794329 DOI: 10.1007/s00262-023-03589-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/13/2023] [Indexed: 01/18/2024]
Abstract
The antigen processing machinery (APM) components needed for a tumor cell to present an antigen to a T cell are expressed at low levels in solid tumors, constituting an important mechanism of immune escape. More than most other solid tumors, head and neck squamous cell carcinoma (HNSCC) cells tend to have low APM expression, rendering them insensitive to immune checkpoint blockade and most other forms of immunotherapy. In HNSCC, this APM deficiency is largely driven by high levels of EGFR and SHP2, leading to low expression and activation of STAT1; however, recent studies suggest that p53, which is often mutated in HNSCCs, may also play a role. In the current study, we aimed to investigate the extent to which STAT1 and p53 individually regulate APM component expression in HNSCC cells. We found that in cells lacking functional p53, APM expression could still be induced by interferon-gamma or DNA-damaging chemotherapy (cisplatin) as long as STAT1 expression remained intact; when both transcription factors were knocked down, APM component expression was abolished. When we bypassed these deficient pathways by rescuing the expression of NLRC5, APM expression was also restored. These results suggest that dual loss of functional STAT1 and p53 may render HNSCC cells incapable of processing and presenting antigens, but rescue of downstream NLRC5 expression may be an attractive strategy for restoring sensitivity to T cell-based immunotherapy.
Collapse
Affiliation(s)
- Brendan L C Kinney
- Department of Otolaryngology - Head and Neck Surgery, Head and Neck Cancer Program, Winship Cancer Institute, Emory University School of Medicine, 550 Peachtree Street NE, 11Th Floor Otolaryngology, Atlanta, GA, 30308, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sreenivasulu Gunti
- National Institute of Deafness and Communication Disorders, NIH, Bethesda, MD, USA
| | - Vikash Kansal
- Department of Otolaryngology - Head and Neck Surgery, Head and Neck Cancer Program, Winship Cancer Institute, Emory University School of Medicine, 550 Peachtree Street NE, 11Th Floor Otolaryngology, Atlanta, GA, 30308, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Nabil F Saba
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yong Teng
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | | | - Fang-Yi Su
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Gabriel A Kwong
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Nicole C Schmitt
- Department of Otolaryngology - Head and Neck Surgery, Head and Neck Cancer Program, Winship Cancer Institute, Emory University School of Medicine, 550 Peachtree Street NE, 11Th Floor Otolaryngology, Atlanta, GA, 30308, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Schnoell J, Sparr C, Al-Gboore S, Haas M, Brkic FF, Kadletz-Wanke L, Heiduschka G, Jank BJ. The ATR inhibitor berzosertib acts as a radio- and chemosensitizer in head and neck squamous cell carcinoma cell lines. Invest New Drugs 2023; 41:842-850. [PMID: 37934325 PMCID: PMC10663216 DOI: 10.1007/s10637-023-01408-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
Alterations in the DNA damage response play a crucial role in radio- and chemoresistance of neoplastic cells. Activation of the Ataxia telangiectasia and Rad3-related (ATR) pathway is an important DNA damage response mechanism in head and neck squamous cell carcinoma (HNSCC). Berzosertib, a selective ATR inhibitor, shows promising radio- and chemosensitizing effects in preclinical studies and is well tolerated in clinical studies. The aim of this study was to elucidate the effect of berzosertib treatment in combination with radiation and cisplatin in HNSCC. The HNSCC cell lines Cal-27 and FaDu were treated with berzosertib alone and in combination with radiation or cisplatin. Cell viability and clonogenic survival were evaluated. The effect of combination treatment was evaluated with the SynergyFinder or combination index. Apoptosis was assessed via measurement of caspase 3/7 activation and migration was evaluated using a wound healing assay. Berzosertib treatment decreased cell viability in a dose-dependent manner and increased apoptosis. The IC50 of berzosertib treatment after 72 h was 0.25-0.29 µM. Combination with irradiation treatment led to a synergistic increase in radiosensitivity and a synergistic or additive decrease in colony formation. The combination of berzosertib and cisplatin decreased cell viability in a synergistic manner. Additionally, berzosertib inhibited migration at high doses. Berzosertib displays a cytotoxic effect in HNSCC at clinically relevant doses. Further evaluation of combination treatment with irradiation and cisplatin is strongly recommended in HNSCC patients as it may hold the potential to overcome treatment resistance, reduce treatment doses and thus mitigate adverse events.
Collapse
Affiliation(s)
- Julia Schnoell
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Carmen Sparr
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Sega Al-Gboore
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Markus Haas
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Faris F Brkic
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Lorenz Kadletz-Wanke
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Gregor Heiduschka
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria.
| | - Bernhard J Jank
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Islam M, Anvarbatcha R, Kunnathodi F, Athar MT, Tariq M. Quinacrine enhances the efficacy of cisplatin by increasing apoptosis and modulating cancer survival proteins in a colorectal cancer cell line. J Cancer Res Ther 2023; 19:1988-1997. [PMID: 38376308 DOI: 10.4103/jcrt.jcrt_902_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 02/21/2024]
Abstract
BACKGROUND Cisplatin and platinum-based compounds have been used successfully to treat various cancers. However, their use is often restricted due to the acquired resistance by cancer cells. Over-expression of p53 and inhibition of NF-kB sensitize several cancer cells towards cisplatin-induced apoptosis. Quinacrine, a cytotoxic drug with predictable safety revealed to concurrently suppress NF-kB and activate p53, which may be an attractive adjuvant in cisplatin chemotherapy. Therefore, the objective of the present study was to establish the role of quinacrine as an adjuvant in lowering the dose of cisplatin during cancer therapy to circumvent its toxic effects. MATERIALS AND METHODS The colon cancer (HCT-8) cells were cultured and cell survival assays were performed using standard procedures. Cell cycle arrest and the extent of apoptosis were determined using a muse cell analyzer. Cancer survival proteins were analyzed using western blotting techniques. RESULTS AND CONCLUSION We demonstrated that concomitant use of quinacrine with cisplatin increased cell apoptosis, suppressed cell proliferation and inhibited colony formation in a colorectal cancer cell line. Moreover, cell cycle arrest in the G0/G1 and G2/M phases and upregulation of p53 expression were observed. There was also downregulation of NF-kB and Bcl-xL protein expressions, both of which are associated with enhanced cell apoptosis and an increase in the sensitivity of cancer cells to cisplatin, overcoming its chemoresistance. Overall, the results of the present study and available literature clearly indicate that the use of quinacrine as an adjuvant with cisplatin may enhance its anti-cancer activity and reduce chemoresistance.
Collapse
Affiliation(s)
- Mozaffarul Islam
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Riyasdeen Anvarbatcha
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Faisal Kunnathodi
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Md Tanwir Athar
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Dentistry and Pharmacy, Buraydah Colleges, Buraydah, Al-Qassim, Saudi Arabia
| | - Mohammad Tariq
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
Rodriguez CP, Kang H, Geiger JL, Burtness B, Chung CH, Pickering CR, Fakhry C, Le QT, Yom SS, Galloway TJ, Golemis E, Li A, Shoop J, Wong S, Mehra R, Skinner H, Saba NF, Flores ER, Myers JN, Ford JM, Karchin R, Ferris RL, Kunos C, Lynn JM, Malik S. Clinical Trial Development in TP53-Mutated Locally Advanced and Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma. J Natl Cancer Inst 2022; 114:1619-1627. [PMID: 36053203 PMCID: PMC9745425 DOI: 10.1093/jnci/djac163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/03/2022] [Accepted: 06/15/2022] [Indexed: 01/11/2023] Open
Abstract
TP53 mutation is the most frequent genetic event in head and neck squamous cell carcinoma (HNSCC), found in more than 80% of patients with human papillomavirus-negative disease. As mutations in the TP53 gene are associated with worse outcomes in HNSCC, novel therapeutic approaches are needed for patients with TP53-mutated tumors. The National Cancer Institute sponsored a Clinical Trials Planning Meeting to address the issues of identifying and developing clinical trials for patients with TP53 mutations. Subcommittees, or breakout groups, were tasked with developing clinical studies in both the locally advanced and recurrent and/or metastatic (R/M) disease settings as well as considering signal-seeking trial designs. A fourth breakout group was focused on identifying and standardizing biomarker integration into trial design; this information was provided to the other breakout groups prior to the meeting to aid in study development. A total of 4 concepts were prioritized to move forward for further development and implementation. This article summarizes the proceedings of the Clinical Trials Planning Meeting with the goal of developing clinical trials for patients with TP53-mutant HNSCC that can be conducted within the National Clinical Trials Network.
Collapse
Affiliation(s)
| | - Hyunseok Kang
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jessica L Geiger
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, Moffit Cancer Center, Tampa, FL, USA
| | - Curtis R Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carole Fakhry
- Division of Head and Neck Surgery, Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Quynh Thu Le
- Department of Radiation Oncology-Radiation Therapy, Stanford University, Palo Alto, CA, USA
| | - Sue S Yom
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Thomas J Galloway
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Erica Golemis
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Alice Li
- Kaiser Permanente Oakland, Oakland, CA, USA
| | | | - Stuart Wong
- Division of Neoplastic Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ranee Mehra
- Division of Hematology/Oncology, Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Heath Skinner
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Elsa R Flores
- Department of Molecular Oncology, Moffit Cancer Center, Tampa, FL, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Ford
- Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Rachel Karchin
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jean M Lynn
- National Institutes of Health, Bethesda, MD, USA
| | - Shakun Malik
- National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Xin Q, Ji Q, Zhang Y, Ma W, Tian B, Liu Y, Chen Y, Wang F, Zhang R, Wang X, Yuan J. Aberrant ROS Served as an Acquired Vulnerability of Cisplatin-Resistant Lung Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1112987. [PMID: 35770045 PMCID: PMC9236771 DOI: 10.1155/2022/1112987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 05/22/2022] [Accepted: 05/30/2022] [Indexed: 12/28/2022]
Abstract
Lung cancer has become a global health issue in recent decades. Approximately 80-85% of cases are non-small-cell lung cancer (NSCLC). Despite the high rate of resistance, cisplatin-base chemotherapy is still the main treatment for NSCLC patients. Thus, overcoming cisplatin resistance is urgently needed in NSCLC therapy. In this study, we identify NADPH metabolism and reactive oxygen species (ROS) levels as the main causes accounting for cisplatin resistance. Based on a small panel consisting of common chemotherapy drugs or compounds, APR-246 is proved to be an effective compound targeting cisplatin-resistant NSCLC cells. APR-246 specially inhibits proliferation and colony formation of cisplatin-resistant cells. In details, APR-246 can significantly cause G0/G1 accumulation and S phase arrest of cisplatin resistant cells and gives rise to severe mitochondria dysfunction as well as elevated apoptosis. Further study proves that it is the aberrant ROS levels as well as NRF2/SLC7A11/GSH axis dysfunction accounting for the specific antitumor effects of APR-246. Scavenging ROS with N-acetylcysteine (NAC) disrupts the inhibitory effect of APR-246 on cisplatin-resistant cells. Mechanistically, NRF2 is specifically degraded by the proteasome following its own ubiquitylation in APR-246-treated cisplatin-resistant cells, which in turn decreases NRF2/SLC7A11/GSH axis activity. Our study provides new insights into the biology driving cisplatin resistance of lung cancer and highlights APR-246 as a potential therapeutic reagent for overcoming cisplatin resistance.
Collapse
Affiliation(s)
- Qian Xin
- Central Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Qinghong Ji
- Department of Obstetrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Ying Zhang
- Department of Respiratory Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Weihong Ma
- Department of Obstetrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Baoqing Tian
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yanli Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yunsong Chen
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fei Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ran Zhang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xingwu Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jupeng Yuan
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, Jinan, Shandong 250033, China
| |
Collapse
|
6
|
Badiee P, Maritz MF, Dmochowska N, Cheah E, Thierry B. Intratumoral Anti-PD-1 Nanoformulation Improves Its Biodistribution. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15881-15893. [PMID: 35357803 DOI: 10.1021/acsami.1c22479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Intratumoral administration of immune checkpoint inhibitors, such as programmed cell death-1 antibodies (aPD-1), is a promising approach toward addressing both the low patients' responses and high off-target toxicity, but good preclinical results have not translated in phase I clinical studies as significant off-target toxicities were observed. We hypothesized that the nanoformulation of aPD-1 could alter both their loco-regional and systemic distribution following intratumoral administration. To test this hypothesis, we developed an aPD-1 nanoformulation (aPD-1 NPs) and investigated its biodistribution following intratumoral injection in an orthotopic mice model of head and neck cancer. Biodistribution analysis demonstrated a significantly lower distribution in off-target organs of the nanoformulated aPD-1 compared to free antibodies. On the other hand, both aPD-1 NPs and free aPD-1 yielded a significantly higher tumor and tumor draining lymph node accumulation than the systemically administrated free aPD-1 used as the current clinical benchmark. In a set of comprehensive in vitro biological studies, aPD-1 NPs effectively inhibited PD-1 expression on T-cells to a similar extent to free aPD-1 and efficiently potentiated the cytotoxicity of T-cells against head and neck cancer cells in vitro. Further studies are warranted to assess the potential of this intratumoral administration of aPD-1 nanoformulation in alleviating the toxicity and enhancing the tumor efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Parisa Badiee
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
- UniSA Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| | - Michelle F Maritz
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Nicole Dmochowska
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Edward Cheah
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
- UniSA Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
- UniSA Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| |
Collapse
|
7
|
Wilkie MD, Anaam EA, Lau AS, Rubbi CP, Vlatkovic N, Jones TM, Boyd MT. Metabolic Plasticity and Combinatorial Radiosensitisation Strategies in Human Papillomavirus-Positive Squamous Cell Carcinoma of the Head and Neck Cell Lines. Cancers (Basel) 2021; 13:cancers13194836. [PMID: 34638320 PMCID: PMC8507998 DOI: 10.3390/cancers13194836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary A subset of head and neck cancers (SCCHN) are caused by human papillomavirus (HPV). As these tumours tend to affect younger patients and are associated with favourable survival, there is a pressing need to find ways to reduce long-term treatment toxicity while maintaining oncological efficacy. We studied utilisation of metabolic pathways in HPV-positive SCCHN cells with the aim of exploiting such for potential therapeutic benefit. We found that these tumours maintained metabolic diversity, in contrast to what we have observed in traditional SCCHN cells associated with mutations in the TP53 gene. This, in turn, correlated with susceptibility to metabolic inhibitors, insofar as a combination of these agents acting on different metabolic pathways was required to augment the effects of ionising radiation (a mainstay of treatment for SCCHN). Notionally, this may provide a means of treatment de-intensification by facilitating radiation dose reduction to minimise the impact of treatment on long-term function. Abstract Background: A major objective in the management of human papillomavirus (HPV)-positive squamous cell carcinoma of the head and neck (SCCHN) is to reduce long-term functional ramifications while maintaining oncological outcomes. This study examined the metabolic profile of HPV-positive SCCHN and the potential role of anti-metabolic therapeutics to achieve radiosensitisation as a potential means to de-escalate radiation therapy. Methods: Three established HPV-positive SCCHN cell lines were studied (UM-SCC-104, UPCI:SCC154, and VU-SCC-147), together with a typical TP53 mutant HPV-negative SCCHN cell line (UM-SCC-81B) for comparison. Metabolic profiling was performed using extracellular flux analysis during specifically designed mitochondrial and glycolytic stress tests. Sensitivity to ionising radiation (IR) was evaluated using clonogenic assays following no treatment, or treatment with: 25 mM 2-deoxy-D-glucose (glycolytic inhibitor) alone; 20 mM metformin (electron transport chain inhibitor) alone; or 25 mM 2-deoxy-D-glucose and 20 mM metformin combined. Expression levels of p53 and reporters of p53 function (MDM2, p53, Phospho-p53 [Ser15], TIGAR and p21 [CDKN1A]) were examined by western blotting. Results: HPV-positive SCCHN cell lines exhibited a diverse metabolic phenotype, displaying robust mitochondrial and glycolytic reserve capacities. This metabolic profile, in turn, correlated with IR response following administration of anti-metabolic agents, in that both 2-deoxy-D-glucose and metformin were required to significantly potentiate the effects of IR in these cell lines. Conclusions: In contrast to our recently published data on HPV-negative SCCHN cells, which display relative glycolytic dependence, HPV-positive SCCHN cells can only be sensitised to IR using a complex anti-metabolic approach targeting both mitochondrial respiration and glycolysis, reflecting their metabolically diverse phenotype. Notionally, this may provide an attractive platform for treatment de-intensification in the clinical setting by facilitating IR dose reduction to minimise the impact of treatment on long-term function.
Collapse
Affiliation(s)
- Mark D. Wilkie
- Cancer Research Centre, Department of Molecular & Clinical Cancer Medicine, The University of Liverpool, 200 London Road, Liverpool L3 9TA, UK; (E.A.A.); (A.S.L.); (C.P.R.); (N.V.); (T.M.J.); (M.T.B.)
- Department of Otorhinolaryngology–Head & Neck Surgery, University Hospital Aintree, Lower Lane, Liverpool L9 7AL, UK
- Correspondence:
| | - Emad A. Anaam
- Cancer Research Centre, Department of Molecular & Clinical Cancer Medicine, The University of Liverpool, 200 London Road, Liverpool L3 9TA, UK; (E.A.A.); (A.S.L.); (C.P.R.); (N.V.); (T.M.J.); (M.T.B.)
| | - Andrew S. Lau
- Cancer Research Centre, Department of Molecular & Clinical Cancer Medicine, The University of Liverpool, 200 London Road, Liverpool L3 9TA, UK; (E.A.A.); (A.S.L.); (C.P.R.); (N.V.); (T.M.J.); (M.T.B.)
- Department of Otorhinolaryngology–Head & Neck Surgery, University Hospital Aintree, Lower Lane, Liverpool L9 7AL, UK
| | - Carlos P. Rubbi
- Cancer Research Centre, Department of Molecular & Clinical Cancer Medicine, The University of Liverpool, 200 London Road, Liverpool L3 9TA, UK; (E.A.A.); (A.S.L.); (C.P.R.); (N.V.); (T.M.J.); (M.T.B.)
| | - Nikolina Vlatkovic
- Cancer Research Centre, Department of Molecular & Clinical Cancer Medicine, The University of Liverpool, 200 London Road, Liverpool L3 9TA, UK; (E.A.A.); (A.S.L.); (C.P.R.); (N.V.); (T.M.J.); (M.T.B.)
| | - Terence M. Jones
- Cancer Research Centre, Department of Molecular & Clinical Cancer Medicine, The University of Liverpool, 200 London Road, Liverpool L3 9TA, UK; (E.A.A.); (A.S.L.); (C.P.R.); (N.V.); (T.M.J.); (M.T.B.)
- Department of Otorhinolaryngology–Head & Neck Surgery, University Hospital Aintree, Lower Lane, Liverpool L9 7AL, UK
| | - Mark T. Boyd
- Cancer Research Centre, Department of Molecular & Clinical Cancer Medicine, The University of Liverpool, 200 London Road, Liverpool L3 9TA, UK; (E.A.A.); (A.S.L.); (C.P.R.); (N.V.); (T.M.J.); (M.T.B.)
| |
Collapse
|
8
|
Citron F, Segatto I, Musco L, Pellarin I, Rampioni Vinciguerra GL, Franchin G, Fanetti G, Miccichè F, Giacomarra V, Lupato V, Favero A, Concina I, Srinivasan S, Avanzo M, Castiglioni I, Barzan L, Sulfaro S, Petrone G, Viale A, Draetta GF, Vecchione A, Belletti B, Baldassarre G. miR-9 modulates and predicts the response to radiotherapy and EGFR inhibition in HNSCC. EMBO Mol Med 2021; 13:e12872. [PMID: 34062049 PMCID: PMC8261495 DOI: 10.15252/emmm.202012872] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy (RT) plus the anti-EGFR monoclonal antibody Cetuximab (CTX) is an effective combination therapy for a subset of head and neck squamous cell carcinoma (HNSCC) patients. However, predictive markers of efficacy are missing, resulting in many patients treated with disappointing results and unnecessary toxicities. Here, we report that activation of EGFR upregulates miR-9 expression, which sustains the aggressiveness of HNSCC cells and protects from RT-induced cell death. Mechanistically, by targeting KLF5, miR-9 regulates the expression of the transcription factor Sp1 that, in turn, stimulates tumor growth and confers resistance to RT+CTX in vitro and in vivo. Intriguingly, high miR-9 levels have no effect on the sensitivity of HNSCC cells to cisplatin. In primary HNSCC, miR-9 expression correlated with Sp1 mRNA levels and high miR-9 expression predicted poor prognosis in patients treated with RT+CTX. Overall, we have discovered a new signaling axis linking EGFR activation to Sp1 expression that dictates the response to combination treatments in HNSCC. We propose that miR-9 may represent a valuable biomarker to select which HNSCC patients might benefit from RT+CTX therapy.
Collapse
Affiliation(s)
- Francesca Citron
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
- Department of Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ilenia Segatto
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Lorena Musco
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Ilenia Pellarin
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Gian Luca Rampioni Vinciguerra
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
- Faculty of Medicine and PsychologyDepartment of Clinical and Molecular MedicineUniversity of Rome “Sapienza”Santo Andrea HospitalRomeItaly
| | - Giovanni Franchin
- Oncologic Radiotherapy UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Giuseppe Fanetti
- Oncologic Radiotherapy UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Francesco Miccichè
- Università Cattolica del Sacro CuoreFondazione Policlinico Universitario Agostino GemelliPolo Scienze Oncologiche ed EmatologicheRomeItaly
| | - Vittorio Giacomarra
- Division of OtorhinolaryngologyAzienda Ospedaliera Santa Maria degli AngeliPordenoneItaly
| | - Valentina Lupato
- Division of OtorhinolaryngologyAzienda Ospedaliera Santa Maria degli AngeliPordenoneItaly
| | - Andrea Favero
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Isabella Concina
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Sanjana Srinivasan
- Department of Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Michele Avanzo
- Medical Physics UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and PhysiologyNational Research Council (IBFM‐CNR)MilanItaly
- Department of PhysicsUniversità degli Studi di Milano‐BicoccaMilanItaly
| | - Luigi Barzan
- Division of OtorhinolaryngologyAzienda Ospedaliera Santa Maria degli AngeliPordenoneItaly
| | - Sandro Sulfaro
- Division of PathologyAzienda Ospedaliera Santa Maria degli AngeliPordenoneItaly
| | - Gianluigi Petrone
- Università Cattolica del Sacro CuoreFondazione Policlinico Universitario Agostino GemelliPolo Scienze Oncologiche ed EmatologicheRomeItaly
- Present address:
Centro Diagnostica MINERVARomeItaly
| | - Andrea Viale
- Department of Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Giulio F Draetta
- Department of Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Andrea Vecchione
- Faculty of Medicine and PsychologyDepartment of Clinical and Molecular MedicineUniversity of Rome “Sapienza”Santo Andrea HospitalRomeItaly
| | - Barbara Belletti
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| | - Gustavo Baldassarre
- Molecular Oncology UnitCentro di Riferimento Oncologico di Aviano (CRO)IRCCSNational Cancer InstituteAvianoItaly
| |
Collapse
|
9
|
Genotyping and Characterization of HPV Status, Hypoxia, and Radiosensitivity in 22 Head and Neck Cancer Cell Lines. Cancers (Basel) 2021; 13:cancers13051069. [PMID: 33802339 PMCID: PMC7959143 DOI: 10.3390/cancers13051069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 11/17/2022] Open
Abstract
To study head and neck squamous cell carcinomas (HNSCC) in vitro, a large variety of HNSCC cell lines have been developed. Here, we characterize a panel of 22 HNSCC cell lines, thereby providing a tool for research into tumor-specific treatment options in HNSCC. Both human papillomavirus (HPV) positive and HPV negative tumor cell lines were collected from commercial and collaborative sources. Short tandem repeat profiling was used to confirm or characterize the identity of the cell lines. Targeted sequencing was performed using a standard pathology single molecule Molecular Inversion Probe panel to detect mutations for 23 tumor suppressors and oncogenes. HPV status, p16 status, radiosensitivity data, and hypoxia data are summarized from all cell lines. We detected HPV transcripts in five cell lines, all of which overexpressed p16. One HPV negative cell line was also p16 positive. We detected mutations in KIT (SCCNij185), PIK3CA (SCCNij185), and CDKN2A (UT-SCC-5 and UT-SCC-38). TP53 mutations were the most frequent, occurring in 16/22 cell lines. HPV infection and TP53 mutations were almost mutually exclusive, with the exception of 93-VU-147T. The cell lines exhibited a wide range of sensitivities towards hypoxia and irradiation. Here, we provide a description of a set of frequently used HNSCC cell lines with diverse characteristics as found in HNSCC patients.
Collapse
|
10
|
MEK/ERK signaling is a critical regulator of high-risk human papillomavirus oncogene expression revealing therapeutic targets for HPV-induced tumors. PLoS Pathog 2021; 17:e1009216. [PMID: 33481911 PMCID: PMC7857559 DOI: 10.1371/journal.ppat.1009216] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 02/03/2021] [Accepted: 12/03/2020] [Indexed: 11/19/2022] Open
Abstract
Intracellular pathogens have evolved to utilize normal cellular processes to complete their replicative cycles. Pathogens that interface with proliferative cell signaling pathways risk infections that can lead to cancers, but the factors that influence malignant outcomes are incompletely understood. Human papillomaviruses (HPVs) predominantly cause benign hyperplasia in stratifying epithelial tissues. However, a subset of carcinogenic or “high-risk” HPV (hr-HPV) genotypes are etiologically linked to nearly 5% of all human cancers. Progression of hr-HPV-induced lesions to malignancies is characterized by increased expression of the E6 and E7 oncogenes and the oncogenic functions of these viral proteins have been widely studied. Yet, the mechanisms that regulate hr-HPV oncogene transcription and suppress their expression in benign lesions remain poorly understood. Here, we demonstrate that EGFR/MEK/ERK signaling, influenced by epithelial contact inhibition and tissue differentiation cues, regulates hr-HPV oncogene expression. Using monolayer cells, epithelial organotypic tissue models, and neoplastic tissue biopsy materials, we show that cell-extrinsic activation of ERK overrides cellular control to promote HPV oncogene expression and the neoplastic phenotype. Our data suggest that HPVs are adapted to use the EGFR/MEK/ERK signaling pathway to regulate their productive replicative cycles. Mechanistic studies show that EGFR/MEK/ERK signaling influences AP-1 transcription factor activity and AP-1 factor knockdown reduces oncogene transcription. Furthermore, pharmacological inhibitors of EGFR, MEK, and ERK signaling quash HPV oncogene expression and the neoplastic phenotype, revealing a potential clinical strategy to suppress uncontrolled cell proliferation, reduce oncogene expression and treat HPV neoplasia. Human papillomavirus (HPV) infections occur in differentiating squamous epithelium and induce hyperplasia during the viral replicative cycle. Although HPV oncogene expression is necessary to promote cellular proliferation for viral genome amplification in the middle epithelial layers, oncogene levels are thereafter suppressed to permit differentiation-induced late gene expression in the uppermost epithelial cells. Yet, the mechanisms responsible for controlling HPV oncogene expression are not well understood. Here, we demonstrate that EGFR/MEK/ERK signaling, which is subject to the normal cellular cues of contact inhibition and epithelial tissue differentiation, is a critical regulator of hr-HPV oncogene expression. We found that extrinsic activation of ERK overrides cellular control to promote oncogene expression and the neoplastic phenotype. Many epidemiologically defined risk factors activate the EGFR/MEK/ERK pathway, suggesting a common mechanism whereby they may promote HPV persistence and disease progression. Lastly, we show that HPV oncogene transcription and protein expression remain susceptible to MEK/ERK control in early neoplastic tissues and tumor cells and that targeted inhibition of MEK/ERK signaling might be exploited therapeutically for HPV-induced infections and tumors.
Collapse
|
11
|
Inukai D, Nishimura K, Okamoto H, Sano R, Ueda H, Ota A, Karnan S, Hosokawa Y, Yoshikawa K, Suzuki S, Ueda R, Murotani K, Bradford CR, Ogawa T. Identification of cisplatin-resistant factor by integration of transcriptomic and proteomic data using head and neck carcinoma cell lines. NAGOYA JOURNAL OF MEDICAL SCIENCE 2020; 82:519-531. [PMID: 33132436 PMCID: PMC7548249 DOI: 10.18999/nagjms.82.3.519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cisplatin is an important drug for the treatment of head and neck squamous cell carcinoma (HNSCC). Determining chemoresistant factors prior to treatment will lead to great benefits for clinicians and patients. Here, we evaluated chemoresistant factors by integrating proteomic and transcriptomic data using HNSCC cell lines to identify a more precise chemoresistant factor in HNSCC. We used four HNSCC cell lines: cisplatin-sensitive, acquired cisplatin resistance, naturally cisplatin-resistant, and acquired 5-FU resistance. Proteomic analysis was performed using iTRAQ, tandem mass spectrometry, and liquid chromatography-electrospray ionization-tandem mass spectrometry. Transcriptomic analysis was performed using microarrays. By integrating these independent data, common factors were addressed and functional analysis was performed using small interfering RNAs (siRNAs) to change the chemosensitivity. Using iTRAQ analysis, 7 proteins were identified as specific for cisplatin chemoresistance factors. Transcriptomic analysis revealed hundreds of potential candidate factors. By combining and integrating these data, S100A2 was identified as a potential cisplatin-specific chemoresistance factor. Functional analysis with siRNA revealed that the expression of S100A2 was reduced and cisplatin sensitivity recovered in the acquired and naturally cisplatin-resistant cell lines, but not in the cisplatin-sensitive cell lines. S100A2 was identified as a cisplatin-specific chemoresistance factor by integrating the transcriptomic and proteomic results obtained using HNSCC cell lines. This is a novel technique that allows for a precise identification, also known as a comprehensive analysis. Our findings indicate that these proteins could be used as biomarkers of HNSCC treatments, providing physicians with new treatment strategies for patients with HNSCC, showing chemoresistance.
Collapse
Affiliation(s)
- Daisuke Inukai
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kunihiro Nishimura
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroki Okamoto
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Rui Sano
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiromi Ueda
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kazuhiro Yoshikawa
- Research Creation Support Center, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Susumu Suzuki
- Research Creation Support Center, Aichi Medical University School of Medicine, Nagakute, Japan.,Center for Advanced Medical Research, Aichi Medical University School of Medicine, Nagakute, Japan.,Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kenta Murotani
- Biostatistics Center, Graduate School of Medicine, Kurume University, Kurume, Japan
| | - Carol R Bradford
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, USA
| | - Tetsuya Ogawa
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| |
Collapse
|
12
|
Broccoli extract increases drug-mediated cytotoxicity towards cancer stem cells of head and neck squamous cell carcinoma. Br J Cancer 2020; 123:1395-1403. [PMID: 32773768 PMCID: PMC7591858 DOI: 10.1038/s41416-020-1025-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 07/01/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
Background Head and neck squamous cell carcinomas (HNSCC) are malignant neoplasms with poor prognosis. Treatment-resistant cancer stem cell (CSC) is one reason for treatment failure. Considerable attention has been focused on sulforaphane (SF), a phytochemical from broccoli possessing anticancer properties. We investigated whether SF could enhance the chemotherapeutic effects of cisplatin (CIS) and 5-fluorouracil (5-FU) against HNSCC–CSCs, and its mechanisms of action. Methods CD44+/CD271+ FACS-isolated CSCs from SCC12 and SCC38 human cell lines were treated with SF alone or combined with CIS or 5-FU. Cell viability, colony- and sphere-forming ability, apoptosis, CSC-related gene and protein expression and in vivo tumour growth were assessed. Safety of SF was tested on non-cancerous stem cells and in vivo. Results SF reduced HNSCC–CSC viability in a time- and dose-dependent manner. Combining SF increased the cytotoxicity of CIS twofold and 5-FU tenfold, with no effects on non-cancerous stem cell viability and functions. SF-combined treatments inhibited CSC colony and sphere formation, and tumour progression in vivo. Potential mechanisms of action included the stimulation of caspase-dependent apoptotic pathway, inhibition of SHH pathway and decreased expression of SOX2 and OCT4. Conclusions Combining SF allowed lower doses of CIS or 5-FU while enhancing these drug cytotoxicities against HNSCC–CSCs, with minimal effects on healthy cells.
Collapse
|
13
|
Acquisition of Cisplatin Resistance Shifts Head and Neck Squamous Cell Carcinoma Metabolism toward Neutralization of Oxidative Stress. Cancers (Basel) 2020; 12:cancers12061670. [PMID: 32599707 PMCID: PMC7352569 DOI: 10.3390/cancers12061670] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Cisplatin (CDDP) is commonly utilized in the treatment of advanced solid tumors including head and neck squamous cell carcinoma (HNSCC). Cisplatin response remains highly variable among individual tumors and development of cisplatin resistance is common. We hypothesized that development of cisplatin resistance is partially driven by metabolic reprogramming. Methods: Using a pre-clinical HNSCC model and an integrated approach to steady state metabolomics, metabolic flux and gene expression data we characterized the interaction between cisplatin resistance and metabolic reprogramming. Results: Cisplatin toxicity in HNSCC was driven by generation of intra-cellular oxidative stress. This was validated by demonstrating that acquisition of cisplatin resistance generates cross-resistance to ferroptosis agonists despite the fact that cisplatin itself does not trigger ferroptosis. Acquisition of cisplatin resistance dysregulated the expression of genes involved in amino acid, fatty acid metabolism and central carbon catabolic pathways, enhanced glucose catabolism and serine synthesis. Acute cisplatin exposure increased intra-tumoral levels of S-methyl-5-thiadenosine (MTA) precursors and metabotoxins indicative of generalized oxidative stress. Conclusions: Acquisition of cisplatin resistance is linked to metabolic recovery from oxidative stress. Although this portends poor effectiveness for directed metabolic targeting, it supports the potential for biomarker development of cisplatin effectiveness using an integrated approach.
Collapse
|
14
|
Mortensen ACL, Spiegelberg D, Brown CJ, Lane DP, Nestor M. The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells. Front Oncol 2019; 9:923. [PMID: 31616635 PMCID: PMC6764291 DOI: 10.3389/fonc.2019.00923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/03/2019] [Indexed: 12/30/2022] Open
Abstract
The tumor suppressor p53 is a key mediator of cellular stress and DNA damage response cascades and is activated after exposure to ionizing radiation. Amplifying wild-type p53 expression by targeting negative regulators such as MDM2 in combination with external beam radiotherapy (EBRT) may result in increased therapeutic effects. The novel stapled peptide PM2 prevents MDM2 from suppressing wild-type p53, and is thus a promising agent for therapeutic combination with EBRT. Effects of PM2 and potential PM2-induced radiosensitivity were assessed in a panel of cancer cell lines using 2D cell viability assays. Western Blot and flow cytometric analyses were used to investigate the mechanisms behind the observed effects in samples treated with PM2 and EBRT. Finally, PM2-treatment combined with EBRT was evaluated in an in vitro 3D spheroid model. PM2-therapy decreased cell viability in wild-type p53, HPV-negative cell lines. Western Blotting and flow cytometry confirmed upregulation of p53, as well as initiation of p53-mediated apoptosis measured by increased cleaved caspase-3 and Noxa activity. Furthermore, 3D in vitro tumor spheroid experiments confirmed the superior effects of the combination, as the only treatment regime resulting in growth inhibition and complete spheroid disintegration. We conclude that PM2 induces antitumorigenic effects in wt p53 HPV-negative cancer cells and potentiates the effects of EBRT, ultimately resulting in tumor eradication in a 3D spheroid model. This strategy shows great potential as a new wt p53 specific tumor-targeting compound, and the combination of PM2 and EBRT could be a promising strategy to increase therapeutic effects and decrease adverse effects from radiotherapy.
Collapse
Affiliation(s)
| | - Diana Spiegelberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | - David Philip Lane
- Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Bourguignon LYW. Matrix Hyaluronan-CD44 Interaction Activates MicroRNA and LncRNA Signaling Associated With Chemoresistance, Invasion, and Tumor Progression. Front Oncol 2019; 9:492. [PMID: 31293964 PMCID: PMC6598393 DOI: 10.3389/fonc.2019.00492] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor malignancies involve cancer cell growth, issue invasion, metastasis and often drug resistance. A great deal of effort has been placed on searching for unique molecule(s) overexpressed in cancer cells that correlate(s) with tumor cell-specific behaviors. Hyaluronan (HA), one of the major ECM (extracellular matrix) components have been identified as a physiological ligand for surface CD44 isoforms which are frequently overexpressed in malignant tumor cells during cancer progression. The binding interaction between HA and CD44 isoforms often stimulates aberrant cellular signaling processes and appears to be responsible for the induction of multiple oncogenic events required for cancer-specific phenotypes and behaviors. In recent years, both microRNAs (miRNAs) (with ~20–25 nucleotides) and long non-coding RNAs (lncRNAs) (with ~200 nucleotides) have been found to be abnormally expressed in cancer cells and actively participate in numerous oncogenic signaling events needed for tumor cell-specific functions. In this review, I plan to place a special emphasis on HA/CD44-induced signaling pathways and the presence of several novel miRNAs (e.g., miR-10b/miR-302/miR-21) and lncRNAs (e.g., UCA1) together with their target functions (e.g., tumor cell migration, invasion, and chemoresistance) during cancer development and progression. I believe that important information can be obtained from these studies on HA/CD44-activated miRNAs and lncRNA that may be very valuable for the future development of innovative therapeutic drugs for the treatment of matrix HA/CD44-mediated cancers.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- Endocrine Unit (111N2), Department of Medicine, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
16
|
Mann JE, Kulkarni A, Birkeland AC, Kafelghazal J, Eisenberg J, Jewell BM, Ludwig ML, Spector ME, Jiang H, Carey TE, Brenner JC. The molecular landscape of the University of Michigan laryngeal squamous cell carcinoma cell line panel. Head Neck 2019; 41:3114-3124. [PMID: 31090975 DOI: 10.1002/hed.25803] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/11/2019] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Laryngeal squamous cell carcinomas (LSCCs) have a high risk of recurrence and poor prognosis. Patient-derived cancer cell lines remain important preclinical models for advancement of new therapeutic strategies, and comprehensive characterization of these models is vital in the precision medicine era. METHODS We performed exome and transcriptome sequencing as well as copy number analysis of a panel of LSCC-derived cell lines that were established at the University of Michigan and are used in laboratories worldwide. RESULTS We observed a complex array of alterations consistent with those reported in The Cancer Genome Atlas head and neck squamous cell carcinoma project, including aberrations in PIK3CA, EGFR, CDKN2A, TP53, and NOTCH family and FAT1 genes. A detailed analysis of FAT family genes and associated pathways showed disruptions to these genes in most cell lines. CONCLUSIONS The molecular profiles we have generated indicate that as a whole, this panel recapitulates the molecular diversity observed in patients and will serve as useful guides in selecting cell lines for preclinical modeling.
Collapse
Affiliation(s)
- Jacqueline E Mann
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Aditi Kulkarni
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew C Birkeland
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Judy Kafelghazal
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Julia Eisenberg
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Brittany M Jewell
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Megan L Ludwig
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Matthew E Spector
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Hui Jiang
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Biostatistics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Thomas E Carey
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - J Chad Brenner
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
17
|
Reid P, Marcu LG, Olver I, Moghaddasi L, Staudacher AH, Bezak E. Diversity of cancer stem cells in head and neck carcinomas: The role of HPV in cancer stem cell heterogeneity, plasticity and treatment response. Radiother Oncol 2019; 135:1-12. [PMID: 31015153 DOI: 10.1016/j.radonc.2019.02.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022]
Abstract
Head and neck squamous cell carcinomas (HNSCC) resulting from oncogenic transformations following human papillomavirus (HPV) infection consistently demonstrate better treatment outcomes than HNSCC from other aetiologies. Squamous cell carcinoma of the oropharynx (OPSCC) shows the highest prevalence of HPV involvement at around 70-80%. While strongly prognostic, HPV status alone is not sufficient to predict therapy response or any potential dose de-escalation. Cancer stem cell (CSC) populations within these tumour types represent the most therapy-resistant cells and are the source of recurrence and metastases, setting a benchmark for tumour control. This review examines clinical and preclinical evidence of differences in response to treatment by the HPV statuses of HNSCC and the role played by CSCs in treatment resistance and their repopulation from non-CSCs. Evidence was collated from literature searches of PubMed, Scopus and Ovid for differential treatment response by HPV status and contribution by critical biomarkers including CSC fractions and chemo-radiosensitivity. While HPV and CSC are yet to fulfil promise as biomarkers of treatment response, understanding how HPV positive and negative aetiologies affect CSC response to treatment and tumour plasticity will facilitate their use for greater treatment individualisation.
Collapse
Affiliation(s)
- Paul Reid
- School of Health Sciences, University of South Australia, Adelaide, Australia; Cancer Research Institute, University of South Australia, Adelaide, Australia.
| | - Loredana G Marcu
- School of Health Sciences, University of South Australia, Adelaide, Australia; Faculty of Science, University of Oradea, Romania
| | - Ian Olver
- Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Leyla Moghaddasi
- Department of Physics, University of Adelaide, Australia; Genesis Care, Department of Medical Physics, Adelaide, Australia
| | - Alexander H Staudacher
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia; School of Medicine, University of Adelaide, Australia
| | - Eva Bezak
- School of Health Sciences, University of South Australia, Adelaide, Australia; Cancer Research Institute, University of South Australia, Adelaide, Australia; Department of Physics, University of Adelaide, Australia
| |
Collapse
|
18
|
The role of p53 status on the synergistic effect of CKD-602 and cisplatin on oral squamous cell carcinoma cell lines. Mol Biol Rep 2018; 46:617-625. [DOI: 10.1007/s11033-018-4517-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022]
|
19
|
Ludwig ML, Kulkarni A, Birkeland AC, Michmerhuizen NL, Foltin SK, Mann JE, Hoesli RC, Devenport SN, Jewell BM, Shuman AG, Spector ME, Carey TE, Jiang H, Brenner JC. The genomic landscape of UM-SCC oral cavity squamous cell carcinoma cell lines. Oral Oncol 2018; 87:144-151. [PMID: 30527230 DOI: 10.1016/j.oraloncology.2018.10.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/18/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES We sought to describe the genetic complexity of 14 UM-SCC oral cavity cancer cell lines that have remained uncharacterized despite being used as model systems for decades. MATERIALS AND METHODS We performed exome sequencing on 14 oral cavity UM-SCC cell lines and denote the mutational profile of each line. We used a SNP array to profile the multiple copy number variations of each cell line and use immunoblotting to compare alterations to protein expression of commonly amplified genes (EGFR, PIK3CA, etc.). RNA sequencing was performed to characterize the expression of genes with copy number alterations. RESULTS The cell lines displayed a highly complex network of genetic aberrations that was consistent with alterations identified in the HNSCC TCGA project including PIK3CA amplification, CDKN2A deletion, as well as TP53 and CASP8 mutations, enabling genetic stratification of each cell line in the panel. Copy number FISH and spectral karyotyping analysis demonstrate that cell lines retain chromosomal heterogeneity. CONCLUSIONS Collectively, we developed an important resource for future oral cavity HNSCC cell line studies and highlight the complexity of genomic aberrations in cell lines.
Collapse
Affiliation(s)
- Megan L Ludwig
- Department of Otolaryngology - Head and Neck Surgery, United States; Program in Cellular and Molecular Biology, United States
| | - Aditi Kulkarni
- Department of Otolaryngology - Head and Neck Surgery, United States
| | | | - Nicole L Michmerhuizen
- Department of Otolaryngology - Head and Neck Surgery, United States; Department of Pharmacology, United States
| | - Susan K Foltin
- Department of Otolaryngology - Head and Neck Surgery, United States
| | - Jacqueline E Mann
- Department of Otolaryngology - Head and Neck Surgery, United States; Department of Pathology, United States
| | - Rebecca C Hoesli
- Department of Otolaryngology - Head and Neck Surgery, United States
| | - Samantha N Devenport
- Department of Otolaryngology - Head and Neck Surgery, United States; Program in Cellular and Molecular Biology, United States
| | | | - Andrew G Shuman
- Department of Otolaryngology - Head and Neck Surgery, United States; Center for Bioethics and Social Sciences in Medicine, United States; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI. University of Michigan Medical School, Ann Arbor, MI, United States
| | - Matthew E Spector
- Department of Otolaryngology - Head and Neck Surgery, United States; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI. University of Michigan Medical School, Ann Arbor, MI, United States
| | - Thomas E Carey
- Department of Otolaryngology - Head and Neck Surgery, United States; Department of Pharmacology, United States; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI. University of Michigan Medical School, Ann Arbor, MI, United States
| | - Hui Jiang
- Department of Biostatistics, United States; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI. University of Michigan Medical School, Ann Arbor, MI, United States
| | - J Chad Brenner
- Department of Otolaryngology - Head and Neck Surgery, United States; Department of Pharmacology, United States; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI. University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
20
|
Shaikh MH, Idris A, Johnson NW, Fallaha S, Clarke DTW, Martin D, Morgan IM, Gabrielli B, McMillan NAJ. Aurora kinases are a novel therapeutic target for HPV-positive head and neck cancers. Oral Oncol 2018; 86:105-112. [PMID: 30409290 DOI: 10.1016/j.oraloncology.2018.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/30/2018] [Accepted: 09/08/2018] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Human papilloma virus (HPV) is the main culprit in cancers of the cervix, penis, anus, skin, eye and head and neck. Current treatments for HPV cancers have not altered survival outcomes for 30 years and there is a significant lack of targeted therapeutic agents in the management of advanced HPV-related HNSCC. Here we show that survival and maintenance of HPV-positive HNC cells relies on the continuous expression of the major HPV oncogene, E7, and that Aurora kinases are critical for survival of high-risk HPV-positive HNC cells. MATERIALS AND METHODS To assess the role of HPV E7 on HNC cell survival, RNA interference (RNAi) of the E7 gene was initially performed. Using an Aurora kinase inhibitor, Alisertib, the role of Aurora kinases in the carcinogenesis of HPV E7 positive HNC tumour lines was then investigated. An in vivo HNC xenograft model was also utilised to assess loss of tumour volume in response to RNAi E7 gene silencing and Alisertib treatment. RESULTS RNAi silencing of the HPV E7 gene inhibited the growth of HPV-positive HNC cells and in vivo tumour load. We show that HPV E7 oncogene expression confers sensitivity to Alisertib on HNC cells where Alisertib-mediated loss in in vitro cell viability and in vivo tumour load is dependent on E7 expression. Moreover, Aurora kinase inhibition induced degradation of MCL-1 in HPV E7-expressing HNC cells. CONCLUSION Overall, we show that Aurora kinases are a novel therapeutic target for HPV-positive HNCs. It might be feasible to combine Aurora kinase and MCL-1 inhibitors for future HNC therapies.
Collapse
Affiliation(s)
- Mushfiq H Shaikh
- Menzies Health Institute Queensland, School of Dentistry and Oral Health, Griffith University, Southport, Queensland, Australia; Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Adi Idris
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia; Department of Clinical Laboratory, Kaifeng Central Hospital, Kaifeng City, China
| | - Newell W Johnson
- Menzies Health Institute Queensland, School of Dentistry and Oral Health, Griffith University, Southport, Queensland, Australia; Dental Institute, King's College London, London, United Kingdom
| | - Sora Fallaha
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Daniel T W Clarke
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - David Martin
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia; Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Iain M Morgan
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University School of Dentistry, Richmond, VA, USA
| | - Brian Gabrielli
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Nigel A J McMillan
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia.
| |
Collapse
|
21
|
Elkashty OA, Ashry R, Elghanam GA, Pham HM, Su X, Stegen C, Tran SD. Broccoli extract improves chemotherapeutic drug efficacy against head-neck squamous cell carcinomas. Med Oncol 2018; 35:124. [PMID: 30078069 DOI: 10.1007/s12032-018-1186-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/31/2018] [Indexed: 12/17/2022]
Abstract
The efficacy of cisplatin (CIS) and 5-fluorouracil (5-FU) against squamous cell carcinomas of the head and neck (SCCHN) remains restricted due to their severe toxic side effects on non-cancer (normal) tissues. Recently, the broccoli extract sulforaphane (SF) was successfully tested as a combination therapy to target cancer cells. However, the effect of lower doses of CIS or 5-FU combined with SF on SCCHN remained unknown. This study tested the chemotherapeutic efficacies of SF combined with much lower doses of CIS or 5-FU against SCCHN cells aiming to reduce cytotoxicity to normal cells. Titrations of SF standalone or in combination with CIS and 5-FU were tested on SCCHN human cell lines (SCC12 and SCC38) and non-cancerous human cells (fibroblasts, gingival, and salivary cells). Concentrations of SF tested were comparable to those found in the plasma following ingestion of fresh broccoli sprouts. The treatment effects on cell viability, proliferation, DNA damage, apoptosis, and gene expression were measured. SF reduced SCCHN cell viability in a time- and dose-dependent manner. SF-combined treatment increased the cytotoxic activity of CIS by twofolds and of 5-FU by tenfolds against SCCHN, with no effect on non-cancerous cells. SF-combined treatment inhibited SCCHN cell clonogenicity and post-treatment DNA repair. SF increased SCCHN apoptosis and this mechanism was due to a down-regulation of BCL2 and up-regulation of BAX, leading to an up-regulation of Caspase3. In conclusion, combining SF with low doses of CIS or 5-FU increased cytotoxicity against SCCHN cells, while having minimal effects on normal cells.
Collapse
Affiliation(s)
- Osama A Elkashty
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC, H3A 0C7, Canada.,Oral Pathology Department, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Ramy Ashry
- Oral Pathology Department, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Ghada Abu Elghanam
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC, H3A 0C7, Canada.,Faculty of Medicine, University of Jordan, Amman, Jordan
| | - Hieu M Pham
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC, H3A 0C7, Canada
| | - Xinyun Su
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC, H3A 0C7, Canada.,College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Camille Stegen
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Microbiome and Disease Tolerance Center, McGill University, Montreal, QC, Canada
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC, H3A 0C7, Canada.
| |
Collapse
|
22
|
Lo Nigro C, Vivenza D, Denaro N, Lattanzio L, Fortunato M, Crook T, Merlano MC. DUSP2 methylation is a candidate biomarker of outcome in head and neck cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:271. [PMID: 30094257 DOI: 10.21037/atm.2018.06.39] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Biomarkers predictive of response to chemoradiotherapy (CRT) regimens for locally advanced head and neck squamous cell carcinoma (LA-HNSCC) are urgently required to identify patients in whom this approach is likely to be effective. TP53 mutations and epidermal growth factor (EGFR) overexpression are common markers of disease. Dual-specificity-phosphatase-2 (DUSP2) has an essential role in cell proliferation, cancer and immune responses. Methods Aberrant DUSP2 methylation was investigated by pyrosequencing in 5 HNSCC cell lines, 112 LA-HNSCC tumours. EGFR was investigated by immunohistochemistry and TP53 was analysed by sequencing. Results We demonstrate methylation-dependent transcriptional silencing of DUSP2 in HNSCC cell lines. In LA-HNSCC patients, aberrant methylation in the DUSP2 CpG island was present in 51/112 cases (45.5%). LA-HNSCC cases with wild-type TP53, overexpression of EGFR and unmethylated DUSP2 had the worst overall survival (P≤0.001). Conclusions DUSP2 methylation, when combined with EGFR and TP53, is a candidate biomarker of clinical outcome in LA-HNSCC treated with CRT.
Collapse
Affiliation(s)
- Cristiana Lo Nigro
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Daniela Vivenza
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Nerina Denaro
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Laura Lattanzio
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Mirella Fortunato
- Department of Pathology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Tim Crook
- Department of Oncology, St. Lukes Cancer Centre, Guildford, UK
| | - Marco Carlo Merlano
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| |
Collapse
|
23
|
Adenovirus-mediated transfer of HPV 16 E6/E7 antisense RNA combined with cisplatin inhibits cellular growth and induces apoptosis in HPV-positive head and neck cancer cells. Cancer Gene Ther 2018; 25:274-283. [DOI: 10.1038/s41417-018-0024-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/04/2018] [Accepted: 03/21/2018] [Indexed: 11/09/2022]
|
24
|
Yu W, Chen Y, Dubrulle J, Stossi F, Putluri V, Sreekumar A, Putluri N, Baluya D, Lai SY, Sandulache VC. Cisplatin generates oxidative stress which is accompanied by rapid shifts in central carbon metabolism. Sci Rep 2018. [PMID: 29523854 PMCID: PMC5844883 DOI: 10.1038/s41598-018-22640-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cisplatin is commonly utilized in the treatment of solid tumors. Its mechanism of action is complex and multiple mechanisms of resistance have been described. We sought to determine the impact of cisplatin-generated oxidative stress on head and neck squamous cell carcinoma (HNSCC) proliferation, survival and metabolic activity in order to identify a potential metabolic signature associated with cisplatin response. DNA-bound cisplatin represents a small fraction of total intra-cellular cisplatin but generates a robust oxidative stress response. Neutralization of oxidative stress reverses cisplatin toxicity independent of the mechanism of cell death and TP53 mutational status. Cisplatin-induced oxidative stress triggers rapid shifts in carbon flux in 3 commonly utilized catabolic pathways: glycolysis, pentose phosphate pathway and citric acid cycle. Among these metabolic shifts, decreased flux from pyruvate into lactate is the only metabolic effect consistently observed across multiple HNSCC cell lines of varying genomic backgrounds and may reflect differential cisplatin sensitivity. Oxidative stress is a critical component of cisplatin cytotoxicity in HNSCC and is reflected in acute changes in carbon flux from pyruvate into lactate. This suggests that lactate may contribute to a metabolic signature of acute cisplatin toxicity, and could prove useful in optimizing cisplatin-based treatment regimens in HNSCC.
Collapse
Affiliation(s)
- Wangie Yu
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Yunyun Chen
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julien Dubrulle
- Integrated Microscopy Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX, USA
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Integrated Microscopy Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX, USA
| | - Vasanta Putluri
- Advanced Technology Core, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Arun Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Dodge Baluya
- Chemical Imaging Research Core, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vlad C Sandulache
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
25
|
Soulières D, Licitra L, Mesía R, Remenár É, Li SH, Karpenko A, Chol M, Wang YA, Solovieff N, Bourdeau L, Sellami D, Faivre S. Molecular Alterations and Buparlisib Efficacy in Patients with Squamous Cell Carcinoma of the Head and Neck: Biomarker Analysis from BERIL-1. Clin Cancer Res 2018; 24:2505-2516. [PMID: 29490986 DOI: 10.1158/1078-0432.ccr-17-2644] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/17/2018] [Accepted: 02/22/2018] [Indexed: 12/24/2022]
Abstract
Purpose: The preplanned exploratory analysis of the BERIL-1 trial presented here aimed to identify biomarkers of response to the combination of buparlisib and paclitaxel.Patients and Methods: BERIL-1 was a multicenter, randomized, double-blind, placebo-controlled phase II study. Patients with recurrent or metastatic squamous cell carcinoma of the head and neck (SCCHN) progressing on/after one previous platinum-based chemotherapy regimen in the recurrent or metastatic setting were treated with either buparlisib plus paclitaxel or placebo plus paclitaxel. Archival tumor tissue and ctDNA samples were analyzed for molecular alterations and immune infiltration using next-generation sequencing or immunohistochemistry.Results: Biomarker analyses were performed in randomized patients (n = 158) with available biomarker data. The most frequently (>5%) mutated genes were TP53, FAT1, TET2, KMT2D, PIK3CA, NOTCH1, NFE2L2, NOTCH2, CCND1, and CDKN2A Patients with SCCHN tumors (from various primary sites) having HPV-negative status (HR = 0.51), TP53 alterations (HR = 0.55) or low mutational load (HR = 0.57) derived overall survival (OS) benefit with the combination of buparlisib and paclitaxel. OS benefit with this combination was also increased in patients with presence of intratumoral TILs ≥10% (HR = 0.51), stromal TILs ≥15% (HR = 0.53), intratumoral CD8-positive cells ≥5% (HR = 0.45), stromal CD8-positive cells ≥10% (HR = 0.47), or CD8-positive cells in invasive margins >25% (HR = 0.37). A trend for improved progression-free survival with the combination of buparlisib and paclitaxel was also observed in these patients.Conclusions: The BERIL-1 biomarker analyses showed that patients with TP53 alterations, HPV-negative status, low mutational load, or high infiltration of TILs or CD8-positive cells derived survival benefit with the combination of buparlisib and paclitaxel. Clin Cancer Res; 24(11); 2505-16. ©2018 AACR.
Collapse
Affiliation(s)
- Denis Soulières
- Centre Hospitalier de l'Université de Montréal, Montréal, Canada
| | - Lisa Licitra
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and University of Milan, Milan, Italy
| | - Ricard Mesía
- Institut Català d'Oncologia-L'Hospitalet, Universitat de Barcelona, IDIBELL, Barcelona, Spain
| | - Éva Remenár
- Országos Onkológiai Intézet, Budapest, Hungary
| | - Shau-Hsuan Li
- Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Andrey Karpenko
- Leningrad Regional Oncology Dispensary, Saint Petersburg, Russian Federation
| | | | - Ying A Wang
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Nadia Solovieff
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | | | - Dalila Sellami
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Sandrine Faivre
- Hôpitaux Universitaires Paris Nord Val de Seine, Paris, France.
| |
Collapse
|
26
|
Abstract
Chromatin decondensation is a key mechanism that guarantees gene transcription and repair of the genome, regulated mainly by the acetylation of histones. Emerging evidence has pointed out to histones as a new controlling mechanism of stem cell maintenance and fate. In this chapter, we will focus on the methods used to enrich tumor cell lines for cancer stem cells, and in the methods to identify the status of the histone acetylation in cancer cells and stem cells using immunofluorescence, invasion, and adhesion assays and identification of nuclear size.
Collapse
Affiliation(s)
- Vivian Petersen Wagner
- Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Manoela Domingues Martins
- Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Rogerio Moraes Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-1078, USA.
| |
Collapse
|
27
|
Tran L, Allen CT, Xiao R, Moore E, Davis R, Park SJ, Spielbauer K, Van Waes C, Schmitt NC. Cisplatin Alters Antitumor Immunity and Synergizes with PD-1/PD-L1 Inhibition in Head and Neck Squamous Cell Carcinoma. Cancer Immunol Res 2017; 5:1141-1151. [PMID: 29097421 DOI: 10.1158/2326-6066.cir-17-0235] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/24/2017] [Accepted: 10/24/2017] [Indexed: 11/16/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) has been treated for decades with cisplatin chemotherapy, and anti-PD-1 immunotherapy has recently been approved for the treatment of this disease. However, preclinical studies of how antitumor immunity in HNSCC is affected by cisplatin alone or in combination with immunotherapies are lacking. Here, we show that sublethal doses of cisplatin may enhance antigen presentation and T-cell killing in vitro, though cisplatin also upregulates tumor cell expression of PD-L1 and may impair T-cell function at higher doses. In a syngeneic mouse model of HNSCC, concurrent use of cisplatin and anti-PD-L1/PD-1 delayed tumor growth and enhanced survival without significantly reducing the number or function of tumor-infiltrating immune cells or increasing cisplatin-induced toxicities. These results suggest that moderate doses of cisplatin may enhance antitumor immunity by mechanisms other than direct tumor cell killing, which may be further enhanced by anti-PD-L1/PD-1 therapy. Cancer Immunol Res; 5(12); 1141-51. ©2017 AACR.
Collapse
Affiliation(s)
- Linda Tran
- Office of the Clinical Director, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Clint T Allen
- Office of the Clinical Director, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland.,Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University, Bethesda, Maryland
| | - Roy Xiao
- Tumor Biology Section, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland.,Medical Research Scholars Program, National Institutes of Health, Bethesda, Maryland.,Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Ellen Moore
- Office of the Clinical Director, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Ruth Davis
- Medical Research Scholars Program, National Institutes of Health, Bethesda, Maryland.,Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - So-Jin Park
- Office of the Clinical Director, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Katie Spielbauer
- Medical Research Scholars Program, National Institutes of Health, Bethesda, Maryland.,Michigan State University College of Human Medicine.,Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Carter Van Waes
- Tumor Biology Section, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Nicole C Schmitt
- Office of the Clinical Director, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland. .,Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University, Bethesda, Maryland
| |
Collapse
|
28
|
Jo DW, Kim YK, Yun PY. The influence of p53 mutation status on the anti-cancer effect of cisplatin in oral squamous cell carcinoma cell lines. J Korean Assoc Oral Maxillofac Surg 2016; 42:337-344. [PMID: 28053903 PMCID: PMC5206238 DOI: 10.5125/jkaoms.2016.42.6.337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/05/2016] [Accepted: 10/07/2016] [Indexed: 01/08/2023] Open
Abstract
Objectives The purpose of this study was to evaluate the anti-cancer activity of cisplatin by studying its effects on cell viability and identifying the mechanisms underlying the induction of cell cycle arrest and apoptosis on oral squamous cell carcinoma (OSCC) cell lines with varying p53 mutation status. Materials and Methods Three OSCC cell lines, YD-8 (p53 point mutation), YD-9 (p53 wild type), and YD-38 (p53 deletion) were used. To determine the cytotoxic effect of cisplatin, MTS assay was performed. The cell cycle alteration and apoptosis were analyzed using flow cytometry. Western blot analysis was used to detect the expression of cell cycle alteration- or apoptosis-related proteins as well as p53. Results Cisplatin showed a time- and dose-dependent anti-proliferative effect in all cell lines. Cisplatin induced G2/M cell accumulation in the three cell lines after treatment with 0.5 and 1.0 µg/mL of cisplatin for 48 hours. The proportion of annexin V-FITC-stained cells increased following treatment with cisplatin. The apoptotic proportion was lower in the YD-38 cell line than in the YD-9 or YD-8 cell lines. Also, immunoblotting analysis indicated that p53 and p21 were detected only in YD-8 and YD-9 cell lines after cisplatin treatment. Conclusion In this study, cisplatin showed anti-cancer effects via G2/M phase arrest and apoptosis, with some difference among OSCC cell lines. The mutation status of p53 might have influenced the difference observed among cell lines. Further studies on p53 mutation status are needed to understand the biological behavior and characteristics of OSCCs and to establish appropriate treatment.
Collapse
Affiliation(s)
- Deuk-Won Jo
- Department of Prosthodontics, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young-Kyun Kim
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Pil-Young Yun
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
29
|
Gao W, Wu MH, Wang N, Ying MZ, Zhang YY, Hua J, Chuan L, Wang YJ. Mitochondrial transcription factor A contributes to cisplatin resistance in patients with estrogen receptor-positive breast cancer. Mol Med Rep 2016; 14:5304-5310. [DOI: 10.3892/mmr.2016.5881] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 09/19/2016] [Indexed: 11/05/2022] Open
|
30
|
Scheel A, Bellile E, McHugh JB, Walline HM, Prince ME, Urba S, Wolf GT, Eisbruch A, Worden F, Carey TE, Bradford C. Classification of TP53 mutations and HPV predict survival in advanced larynx cancer. Laryngoscope 2016; 126:E292-9. [PMID: 27345657 PMCID: PMC5002993 DOI: 10.1002/lary.25915] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/28/2015] [Accepted: 01/19/2016] [Indexed: 11/07/2022]
Abstract
OBJECTIVES/HYPOTHESIS Assess tumor suppressor p53 (TP53) functional mutations in the context of other biomarkers in advanced larynx cancer. STUDY DESIGN Prospective analysis of pretreatment tumor TP53, human papillomavirus (HPV), Bcl-xL, and cyclin D1 status in stage III and IV larynx cancer patients in a clinical trial. METHODS TP53 exons 4 through 9 from 58 tumors were sequenced. Mutations were grouped using three classifications based on their expected function. Each functional group was analyzed for response to induction chemotherapy, time to surgery, survival, HPV status, p16INK4a, Bcl-xl, and cyclin D1 expression. RESULTS TP53 mutations were found in 22 of 58 (37.9%) patients with advanced larynx cancer, including missense mutations in 13 of 58 (22.4%) patients, nonsense mutations in four of 58 (6.9%), and deletions in five of 58 (8.6%). High-risk HPV was found in 20 of 52 (38.5%) tumors. A classification based on Evolutionary Action score of p53 (EAp53) distinguished missense mutations with high risk for decreased survival from low-risk mutations (P = 0.0315). A model including this TP53 classification, HPV status, cyclin D1, and Bcl-xL staining significantly predicts survival (P = 0.0017). CONCLUSION EAp53 functional classification of TP53 mutants and biomarkers predict survival in advanced larynx cancer. LEVEL OF EVIDENCE NA. Laryngoscope, 126:E292-E299, 2016.
Collapse
Affiliation(s)
- Adam Scheel
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Emily Bellile
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Jonathan B McHugh
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Heather M Walline
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Mark E Prince
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Susan Urba
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Gregory T Wolf
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Avraham Eisbruch
- Department of Radiation Oncology, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Francis Worden
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Thomas E Carey
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| | - Carol Bradford
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan, U.S.A
| |
Collapse
|
31
|
Matsumoto CS, Almeida LO, Guimarães DM, Martins MD, Papagerakis P, Papagerakis S, Leopoldino AM, Castilho RM, Squarize CH. PI3K-PTEN dysregulation leads to mTOR-driven upregulation of the core clock gene BMAL1 in normal and malignant epithelial cells. Oncotarget 2016; 7:42393-42407. [PMID: 27285754 PMCID: PMC5173143 DOI: 10.18632/oncotarget.9877] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/19/2016] [Indexed: 01/23/2023] Open
Abstract
Dysfunctional clock signaling is observed in a variety of pathological conditions. Many members of the clock gene family are upregulated in tumor cells. Here, we explored the consequences of a commonly disrupted signaling pathway in head and neck cancer on the regulation of circadian clock genes. PTEN is a key molecular controller of the PI3K signaling, and loss of PTEN function is often observed in a variety of cancers. Our main goal was to determine whether PTEN regulates circadian clock signaling. We found that oxidation-driven loss of PTEN function resulted in the activation of mTOR signaling and activation of the core clock protein BMAL1 (also known as ARNTL). The PTEN-induced BMAL1 upregulation was further confirmed using small interference RNA targeting PTEN, and in vivo conditional depletion of PTEN from the epidermis. We observed that PTEN-driven accumulation of BMAL1 was mTOR-mediated and that administration of Rapamycin, a specific mTOR inhibitor, resulted in in vivo rescue of normal levels of BMAL1. Accumulation of BMAL1 by deletion of PER2, a Period family gene, was also rescued upon in vivo administration of mTOR inhibitor. Notably, BMAL1 regulation requires mTOR regulatory protein Raptor and Rictor. These findings indicate that mTORC1 and mTORC2 complex plays a critical role in controlling BMAL1, establishing a connection between PI3K signaling and the regulation of circadian rhythm, ultimately resulting in deregulated BMAL1 in tumor cells with disrupted PI3K signaling.
Collapse
Affiliation(s)
- Camila S. Matsumoto
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Department of Clinical Analysis, Toxicology and Bromatology, School of Pharmacy, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Luciana O. Almeida
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Douglas M. Guimarães
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Department of Oral Pathology, School of Dentistry, University of Sao Paulo, SP, Brazil
| | - Manoela D. Martins
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Petros Papagerakis
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Center for Organogenesis, University of Michigan, Ann Arbor, MI, USA
| | - Silvana Papagerakis
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Andreia M. Leopoldino
- Department of Clinical Analysis, Toxicology and Bromatology, School of Pharmacy, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rogerio M. Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Cristiane H. Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
32
|
Feng S, Rabii R, Liang G, Song C, Chen W, Guo M, Wei X, Messadi D, Hu S. The Expression Levels of XLF and Mutant P53 Are Inversely Correlated in Head and Neck Cancer Cells. J Cancer 2016; 7:1374-82. [PMID: 27471552 PMCID: PMC4964120 DOI: 10.7150/jca.14669] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 03/15/2016] [Indexed: 12/16/2022] Open
Abstract
XRCC4-like factor (XLF), also known as Cernunnos, is a protein encoded by the human NHEJ1 gene and an important repair factor for DNA double-strand breaks. In this study, we have found that XLF is over-expressed in HPV(+) versus HPV(-) head and neck squamous cell carcinoma (HNSCC) and significantly down-regulated in the HNSCC cell lines expressing high level of mutant p53 protein versus those cell lines harboring wild-type TP53 gene with low p53 protein expression. We have also demonstrated that Werner syndrome protein (WRN), a member of the NHEJ repair pathway, binds to both mutant p53 protein and NHEJ1 gene promoter, and siRNA knockdown of WRN leads to the inhibition of XLF expression in the HNSCC cells. Collectively, these findings suggest that WRN and p53 are involved in the regulation of XLF expression and the activity of WRN might be affected by mutant p53 protein in the HNSCC cells with aberrant TP53 gene mutations, due to the interaction of mutant p53 with WRN. As a result, the expression of XLF in these cancer cells is significantly suppressed. Our study also suggests that XLF is over-expressed in HPV(+) HNSCC with low expression of wild type p53, and might serve as a potential biomarker for HPV(+) HNSCC. Further studies are warranted to investigate the mechanisms underlying the interactive role of WRN and XLF in NHEJ repair pathway.
Collapse
Affiliation(s)
- Sizhe Feng
- 1. School of Dentistry and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA; 2. North Hospital, Shenyang, China
| | - Ramin Rabii
- 1. School of Dentistry and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | | | - Chenxi Song
- 1. School of Dentistry and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Wei Chen
- 1. School of Dentistry and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Mian Guo
- 1. School of Dentistry and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | | | - Diana Messadi
- 1. School of Dentistry and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Shen Hu
- 1. School of Dentistry and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
33
|
Chang JW, Jung SN, Kim JH, Shim GA, Park HS, Liu L, Kim JM, Park J, Koo BS. Carboxyl-Terminal Modulator Protein Positively Acts as an Oncogenic Driver in Head and Neck Squamous Cell Carcinoma via Regulating Akt phosphorylation. Sci Rep 2016; 6:28503. [PMID: 27328758 PMCID: PMC4916413 DOI: 10.1038/srep28503] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/02/2016] [Indexed: 01/10/2023] Open
Abstract
The exact regulatory mechanisms of carboxyl-terminal modulator protein (CTMP) and its downstream pathways in cancer have been controversial and are not completely understood. Here, we report a new mechanism of regulation of Akt serine/threonine kinase, one of the most important dysregulated signals in head and neck squamous cell carcinoma (HNSCC) by the CTMP pathway and its clinical implications. We find that HNSCC tumor tissues and cell lines had relatively high levels of CTMP expression. Clinical data indicate that CTMP expression was significantly associated with positive lymph node metastasis (OR = 3.8, P = 0.033) and correlated with poor prognosis in patients with HNSCC. CTMP was also positively correlated with Akt/GSK-3β phosphorylation, Snail up-regulation and E-cadherin down-regulation, which lead to increased proliferation and epithelial-to-mesenchymal transition, suggesting that CTMP expression results in enhanced tumorigenic and metastatic properties of HNSCC cells. Moreover, CTMP suppression restores sensitivity to cisplatin chemotherapy. Intriguingly, all the molecular responses to CTMP regulation are identical regardless of p53 status in HNSCC cells. We conclude that CTMP promotes Akt phosphorylation and functions as an oncogenic driver and prognostic marker in HNSCC irrespective of p53.
Collapse
Affiliation(s)
- Jae Won Chang
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University, Daejeon, Republic of Korea
| | - Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University, Daejeon, Republic of Korea
| | - Ju-Hee Kim
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University, Daejeon, Republic of Korea
| | - Geun-Ae Shim
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University, Daejeon, Republic of Korea
| | - Hee Sung Park
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University, Daejeon, Republic of Korea
| | - Lihua Liu
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jin Man Kim
- Research Institute for Medical Sciences and Pathology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, Metabolic Diseases and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Bon Seok Koo
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
34
|
Hu L, Liu J, Li Z, Wang C, Nawshad A. Transforming growth factor-β1 activates ΔNp63/c-Myc to promote oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2016; 122:460-482.e4. [PMID: 27567435 DOI: 10.1016/j.oooo.2016.05.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 01/21/2023]
Abstract
OBJECTIVE During the development of oral squamous cell carcinoma (OSCC), the transformed epithelial cells undergo increased proliferation resulting in tumor growth and invasion. Interestingly, throughout all phases of differentiation and progression to OSCC, transforming growth factor-β1 (TGF)-β1 induces cell cycle arrest or apoptosis; however, the role of TGF-β1 in promoting cancer cell proliferation has not been explored in detail. The purpose of this study was to identify the effect of TGF-β1 on OSCC cell proliferation. STUDY DESIGN Using both human OSCC samples and cell lines (UMSCC38 and UMSCC11B), we assessed protein, mRNA, gene expression, and protein-DNA interactions during OSCC progression. RESULTS Our results showed that TGF-β1 increased OSCC cell proliferation by upregulating the expression of ΔNp63 and c-Myc oncogenes. Although the basal OSCC cell proliferation is sustained by activating ΔNp63, increased induction of c-Myc causes unregulated OSCC cell proliferation. Following induction of the cell cycle by ΔNp63 and c-Myc, cancer cells that halt c-Myc activity undergo epithelial mesenchymal transition or invasion while those that continue to express ΔNp63/c-Myc undergo unlimited progression through the cell cycle. CONCLUSIONS OSCC proliferation is manifested by the induction of c-Myc in response to TGF-β1 signaling, which is essential for OSCC growth. Our data highlight the potential role of TGF-β1 in the induction of cancer progression and invasion of OSCC.
Collapse
Affiliation(s)
- Lihua Hu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA; Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong, P.R. China
| | - Jingpeng Liu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Zhi Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Chunling Wang
- Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong, P.R. China
| | - Ali Nawshad
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA.
| |
Collapse
|
35
|
Rossi MJ, Sundararaj K, Koybasi S, Phillips MS, Szulc ZM, Bielawska A, Day TA, Obeid LM, Hannun YA, Ogretmen B. Inhibition of growth and telomerase activity by novel cationic ceramide analogs with high solubility in human head and neck squamous cell carcinoma cells. Otolaryngol Head Neck Surg 2016; 132:55-62. [PMID: 15632910 DOI: 10.1016/j.otohns.2004.08.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES: Head and neck squamous cell carcinoma (HNSCC) is notoriously resistant to chemotherapy. The sphingolipid ceramide and its analogs have been demonstrated to exert antitumor activity in many cell types; however, the effectiveness of these analogs has been limited by potency and solubility. This study focuses on the effects of novel highly soluble cationic pyridinium-ceramides, alone and in combination with various chemotherapeutic agents, on cell survival, telomerase activity, and cell cycle arrest in HNSCC cell lines in vitro. METHODS: The concentration of pyridinium-ceramides and chemotherapeutic agents that inhibited cell growth by 50% (IC50) was determined by MTT cell survival assays. The cell cycle profiles were determined by flow cytometry. Telomerase activity was determined by telomerase repeat amplification protocol (TRAP) assay. RESULTS: Treatment of the human UM-SCC-22A (SCC of the hypopharynx) cells, as well as various other HNSCC cell lines, with C6-Pyr-Cer resulted in the inhibition of cell survival with an IC50 concentration of approximately 250 to 300 nM at 96 hours, whereas its IC50 was greater than 1000 nM in noncancerous Wi-38 human lung fibroblasts, and adult human epidermal keratinocytes. Moreover, treatment with C6-Pyr-Cer also resulted in cell cycle arrest in G0/G1, which correlated with a significant inhibition of telomerase activity in UM-SCC-22A cells. Additional results demonstrated that the combination of C6-Pyr-Cer with gemcitabine (GMZ) or doxorubicin (DOX), which have the lowest IC50 concentrations among various chemotherapeutic drugs in these cells, enhances the effects of these drugs in the inhibition of telomerase and cell growth. CONCLUSIONS These data suggest that the novel C6-Pyr-Cer with high solubility and bioavailability may lead to the development of new therapeutic strategies that target telomerase for the treatment of HNSCC.
Collapse
Affiliation(s)
- Michael J Rossi
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kachalaki S, Ebrahimi M, Mohamed Khosroshahi L, Mohammadinejad S, Baradaran B. Cancer chemoresistance; biochemical and molecular aspects: a brief overview. Eur J Pharm Sci 2016; 89:20-30. [PMID: 27094906 DOI: 10.1016/j.ejps.2016.03.025] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/02/2016] [Accepted: 03/31/2016] [Indexed: 01/27/2023]
Abstract
The effectiveness of chemotherapy is one of the main challenges in cancer treatment and resistance to classic drugs and traditional treatment processes is an obstacle to this goal. Drug resistance that may be inherent or adventitious can cause poor treatment outcome and tumor relapse. In most cases, resistance to a drug can lead to resistance to many other drugs structure and function of which is not necessarily similar to the first drug. This phenomenon is the main mechanism behind failure of many of metastatic cancers. There are various molecular mechanisms involved in multidrug resistance, including change in the activity of membrane transporters (such as ABC transporters), increase of drug metabolism, change of the target enzyme (such as mutations that change thymidylate synthase and topoisomerases), promotion of DNA damage repair, and escape from drug induced apoptosis. Clinical and laboratory investigations on biomarkers involved in the response to chemotherapy have characterized the key factors behind the failure of treatments. Knowing the molecular factors involved in drug resistance may help us to develop new strategies for more promising chemotherapy and reduce the rate of relapse. In this brief review, molecular mechanisms and tumor microenvironment leading to decreased drug sensitivity, and strategies of reversing drug resistance are described.
Collapse
Affiliation(s)
- Saeed Kachalaki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mina Ebrahimi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Sina Mohammadinejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
37
|
Bourguignon LYW, Wong G, Shiina M. Up-regulation of Histone Methyltransferase, DOT1L, by Matrix Hyaluronan Promotes MicroRNA-10 Expression Leading to Tumor Cell Invasion and Chemoresistance in Cancer Stem Cells from Head and Neck Squamous Cell Carcinoma. J Biol Chem 2016; 291:10571-85. [PMID: 27002147 DOI: 10.1074/jbc.m115.700021] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Indexed: 01/22/2023] Open
Abstract
Human head and neck squamous cell carcinoma is a solid tumor malignancy associated with major morbidity and mortality. In this study, we determined that human head and neck squamous cell carcinoma-derived HSC-3 cells contain a subpopulation of cancer stem cells (CSCs) characterized by a high level of CD44v3 and aldehyde dehydrogenase-1 (ALDH1) expression. Importantly, matrix hyaluronan (HA) induces the up-regulation of stem cell markers that display the hallmark CSC properties. Histone methyltransferase, DOT1L, is also up-regulated by HA in CSCs (isolated from HSC-3 cells). Further analyses indicate that the stimulation of microRNA-10b (miR-10b) expression is DOT1L-specific and HA/CD44-dependent in CSCs. This process subsequently results in the overexpression of RhoGTPases and survival proteins leading to tumor cell invasion and cisplatin resistance. Treatment of CSCs with DOT1L-specific small interfering RNAs (siRNAs) effectively blocks HA/CD44-mediated expression of DOT1L, miR-10b production, and RhoGTPase/survival protein up-regulation as well as reduces tumor cell invasion and enhances chemosensitivity. CSCs were also transfected with a specific anti-miR-10b inhibitor to silence miR-10b expression and block its target functions. Our results demonstrate that the anti-miR-10 inhibitor not only decreases RhoGTPase/survival protein expression and tumor cell invasion, but also increases chemosensitivity in HA-treated CSCs. Taken together, these findings strongly support the contention that histone methyltransferase, DOT1L-associated epigenetic changes induced by HA play pivotal roles in miR-10 production leading to up-regulation of RhoGTPase and survival proteins. All of these events are critically important for the acquisition of cancer stem cell properties, including self-renewal, tumor cell invasion, and chemotherapy resistance in HA/CD44-activated head and neck cancer.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- From the Endocrine Unit, Department of Medicine, University of California at San Francisco and San Francisco Veterans Affairs Medical Center, San Francisco, California 94121
| | - Gabriel Wong
- From the Endocrine Unit, Department of Medicine, University of California at San Francisco and San Francisco Veterans Affairs Medical Center, San Francisco, California 94121
| | - Marisa Shiina
- From the Endocrine Unit, Department of Medicine, University of California at San Francisco and San Francisco Veterans Affairs Medical Center, San Francisco, California 94121
| |
Collapse
|
38
|
Ohtomo-Oda R, Komatsu S, Mori T, Sekine S, Hirajima S, Yoshimoto S, Kanai Y, Otsuji E, Ikeda E, Tsuda H. SMYD2 overexpression is associated with tumor cell proliferation and a worse outcome in human papillomavirus–unrelated nonmultiple head and neck carcinomas. Hum Pathol 2016; 49:145-55. [DOI: 10.1016/j.humpath.2015.08.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/17/2015] [Accepted: 08/21/2015] [Indexed: 12/13/2022]
|
39
|
Combination of NTP with cetuximab inhibited invasion/migration of cetuximab-resistant OSCC cells: Involvement of NF-κB signaling. Sci Rep 2015; 5:18208. [PMID: 26655729 PMCID: PMC4677387 DOI: 10.1038/srep18208] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/19/2015] [Indexed: 01/30/2023] Open
Abstract
Although the epidermal growth factor receptor (EGFR) is an established target in head-and-neck cancer (HNC), resistance to EGFR-targeted therapy mediated by various mechanisms has been reported. Therefore, a combination strategy to overcome resistance to EGFR mono-targeted therapy is clinically required. We have previously demonstrated that non-thermal atmospheric pressure plasma (NTP) induces death of various cancer cells, including oral squamous cancer (OSCC) cells. In this study, we report for the first time that combining NTP treatment with cetuximab led to inhibition of migration and invasion in cetuximab-resistant OSCC cells, which could be a promising strategy to overcome resistance to anti-EGFR therapy. NTP induced deactivation of NF-κB in SCCQLL1 cells, but not in MSKQLL1 cells. In addition, NTP increased the expression level of E-cadherin, and decreased those of vimentin, Slug, Snail, matrix metalloproteinase (MMP)-2, -9, and activities of MMPs. Moreover, NF-κB upregulation using cDNA diminished the combination effect of NTP on invasion, migration and related signals. Taken together, these results indicate that the combination of NTP with cetuximab can decrease invasiveness in cetuximab-resistant OSCCs through a novel mechanism involving the NF-κB pathway. These findings show the therapeutic potential of treatment that combines NTP and cetuximab in OSCC.
Collapse
|
40
|
Szentkúti G, Dános K, Brauswetter D, Kiszner G, Krenács T, Csákó L, Répássy G, Tamás L. Correlations between prognosis and regional biomarker profiles in head and neck squamous cell carcinomas. Pathol Oncol Res 2015; 21:643-50. [PMID: 25547827 DOI: 10.1007/s12253-014-9869-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022]
Abstract
Head and neck squamous cell carcinomas (HNSCC) show diverse clinicopathological features and are mostly linked with poor outcome. In this study, we tested if the expression of tumor growth, cell cycle and basement membrane anchorage related biomarkers allow prognostic and clinicopathological stratification of HNSCC. Archived HNSCC samples from 226 patients included into tissue microarrays (TMA) were tested using immunohistochemistry. Histopathological evaluation and the analysis of immunostaining for EGFR, Ki67, p53, p16(ink4) and Collagen XVII proteins were carried out in digital whole slides. Statistical evaluation was carried out using Pearson's Chi-square test and Kaplan-Meier survival analysis. In the tested cohort, hypopharyngeal cancers had the least favorable, and glottic cancers had the most favorable prognosis. High Ki67 positive tumor cell fractions were associated with significantly worse prognosis and elevated rate of lymph node metastasis. Both Ki67 and EGFR expression correlated significantly with the tumor localization. Ki67 index was the highest in the hypopharyngeal region and it proved to be the lowest in the glottic region. EGFR expression was the highest in the oral cavity and the lowest in the glottic region. The survival rate of patients with p16(ink4)-negative cancer was significantly lower than of those with p16(ink4)-positive disease. A significant inverse correlation was found between histological grade and the prognosis of HNSCC. Our data support that elevated Ki67 positive proliferating cell fractions contribute to the unfavorable prognosis of hypopharyngeal cancers, while glottic cancers have the most favorable prognosis because of the lowest Ki67 expression rate.
Collapse
Affiliation(s)
- Gabriella Szentkúti
- Department of Oto-Rhino-Laryngology, Jahn Ferenc South-Pest Hospital, 1st Köves Street, 1204, Budapest, Hungary,
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Bourguignon LYW, Shiina M, Li JJ. Hyaluronan-CD44 interaction promotes oncogenic signaling, microRNA functions, chemoresistance, and radiation resistance in cancer stem cells leading to tumor progression. Adv Cancer Res 2015; 123:255-75. [PMID: 25081533 DOI: 10.1016/b978-0-12-800092-2.00010-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hyaluronan (HA), a major component of the extracellular matrix (ECM), is enriched in many types of tumors. There is good evidence linking high levels of HA production in human carcinomas to an aggressive phenotype and tumor metastasis. HA is generally bound to CD44 isoforms (so-called CD44s and CD44v3) which are ubiquitous, abundant, and functionally important cell surface receptors. This chapter describes the evidence for HA/CD44v3-mediated activation of the cytoskeleton (e.g., ankyrin and GTPases) and matrix metalloproteinase (MMP) signaling during tumor progression. A special focus is placed on the role of HA-CD44v3 interaction in cancer stem cells (CSCs). Matrix HA is known to be present in CSC niches. Since CD44v3 serves as a CSC marker, it provides an important physical linkage between matrix HA and various transcription factors that regulate tumor cell functions through distinct signaling pathways. CSCs are known to be chemoresistant and/or radiation resistant and to cause cancer relapse. The purpose of this chapter is to review the most current research on the cellular and molecular biology of CSCs. The emphasis will be placed on both CSC niche and matrix HA-induced microRNA signaling plus various CSC functions (e.g., self-renewal, differentiation, and chemoresistance) during cancer progression. Understanding the regulation of CSCs is critically important for designing CSC-specific therapeutic targets to prevent cancer development and progression.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- Department of Medicine, University of California at San Francisco & Endocrine Unit (111N), VA Medical Center, San Francisco, California, USA.
| | - Marisa Shiina
- Department of Medicine, University of California at San Francisco & Endocrine Unit (111N), VA Medical Center, San Francisco, California, USA
| | - Jian-Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| |
Collapse
|
42
|
Eytan DF, Snow GE, Carlson SG, Schiltz S, Chen Z, Van Waes C. Combination effects of SMAC mimetic birinapant with TNFα, TRAIL, and docetaxel in preclinical models of HNSCC. Laryngoscope 2015; 125:E118-24. [PMID: 25431358 PMCID: PMC4336212 DOI: 10.1002/lary.25056] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVES/HYPOTHESIS Head and neck squamous cell carcinoma (HNSCC) cells are resistant to cell death induced by tumor necrosis factor ligands such as tumor necrosis factor α (TNFα) or TNF-related apoptosis-inducing ligand (TRAIL) and cytotoxic chemotherapies. Recently, genetic alterations in cell death pathways, including inhibitor of apoptosis proteins, have been demonstrated in HNSCC. We investigated the effects of birinapant, a novel, second mitochondria-derived activator of caspases (SMAC)-mimetic that targets inhibitor of apoptosis proteins, alone and in combination with TNFα, TRAIL, or chemotherapy docetaxel. STUDY DESIGN Experimental study using human HNSCC cell lines in vitro and xenograft mouse model in vivo. METHODS A panel of HNSCC cell lines with varying genetic alterations in cell death pathway components were treated with birinapant ± TNFα, TRAIL, and docetaxel and were assessed for effects on cell density, cell cycle, and death. Synergism was determined at varying concentrations of treatments using the Chou-Talalay method. Combination studies using birinapant ± docetaxel were performed in a xenograft mouse model. RESULTS Birinapant, alone or in combination with TNFα or TRAIL, decreased cell density in cell lines, with IC50 s ranging from 0.5 nM to > 1 µM. Birinapant alone or with TNF significantly increased subG0 cell death in different lines. Docetaxel showed synergism with birinapant ± TNFα in vitro. Birinapant monotherapy-inhibited growth in a tumor xenograft model resistant to docetaxel, and combination treatment further delayed growth. CONCLUSIONS Birinapant alone or in combination with TNFα or TRAIL and docetaxel decreased cell density, increased cell death, and displayed antitumor activity in a preclinical HNSCC xenograft exhibiting aberrations in cell death pathway components and docetaxel resistance.
Collapse
Affiliation(s)
- Danielle F. Eytan
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, USA
- NIH Medical Research Scholars Program, National Institutes of Health, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Grace E. Snow
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, USA
- NIH Medical Research Scholars Program, National Institutes of Health, USA
| | - Sophie G. Carlson
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, USA
| | - Stephen Schiltz
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, USA
| | - Zhong Chen
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, USA
| | - Carter Van Waes
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, USA
| |
Collapse
|
43
|
Adams AK, Hallenbeck GE, Casper KA, Patil YJ, Wilson KM, Kimple RJ, Lambert PF, Witte DP, Xiao W, Gillison ML, Wikenheiser-Brokamp KA, Wise-Draper TM, Wells SI. DEK promotes HPV-positive and -negative head and neck cancer cell proliferation. Oncogene 2015; 34:868-77. [PMID: 24608431 PMCID: PMC4160430 DOI: 10.1038/onc.2014.15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/21/2014] [Accepted: 02/03/2014] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignancy worldwide, and patient outcomes using current treatments remain poor. Tumor development is etiologically associated with tobacco or alcohol use and/or human papillomavirus (HPV) infection. HPV-positive HNSCCs, which frequently harbor wild-type p53, carry a more favorable prognosis and are a biologically distinct subgroup when compared with their HPV-negative counterparts. HPV E7 induces expression of the human DEK gene, both in vitro and in vivo. In keratinocytes, DEK overexpression is sufficient for causing oncogenic phenotypes in the absence of E7. Conversely, DEK loss results in cell death in HPV-positive cervical cancer cells at least in part through p53 activation, and Dek knockout mice are relatively resistant to the development of chemically induced skin papillomas. Despite the established oncogenic role of DEK in HPV-associated cervical cancer cell lines and keratinocytes, a functional role of DEK has not yet been explored in HNSCC. Using an established transgenic mouse model of HPV16 E7-induced HNSCC, we demonstrate that Dek is required for optimal proliferation of E7-transgenic epidermal cells and for the growth of HNSCC tumors. Importantly, these studies also demonstrate that DEK protein is universally upregulated in both HPV-positive and -negative human HNSCC tumors relative to adjacent normal tissue. Furthermore, DEK knockdown inhibited the proliferation of HPV-positive and -negative HNSCC cells, establishing a functional role for DEK in human disease. Mechanistic studies reveal that attenuated HNSCC cell growth in response to DEK loss was associated with reduced expression of the oncogenic p53 family member, ΔNp63. Exogenous ΔNp63 expression rescued the proliferative defect in the absence of DEK, thereby establishing a functional DEK-ΔNp63 oncogenic pathway that promotes HNSCC. Taken together, our data demonstrate that DEK stimulates HNSCC cellular growth and identify ΔNp63 as a novel DEK effector.
Collapse
Affiliation(s)
- Allie K. Adams
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Grace E. Hallenbeck
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Keith A. Casper
- Department of Otolaryngology, Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Yash J. Patil
- Department of Otolaryngology, Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Keith M. Wilson
- Department of Otolaryngology, Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Randall J. Kimple
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David P. Witte
- Division of Pathology & Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Weihong Xiao
- Viral Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Maura L. Gillison
- Viral Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Kathryn A. Wikenheiser-Brokamp
- Pathology & Laboratory Medicine and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center/University of Cincinnati, Cincinnati, OH, USA
| | - Trisha M. Wise-Draper
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Hematology/Oncology, University Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - Susanne I. Wells
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
44
|
Veena MS, Wilken R, Zheng JY, Gholkar A, Venkatesan N, Vira D, Ahmed S, Basak SK, Dalgard CL, Ravichandran S, Batra RK, Kasahara N, Elashoff D, Fishbein MC, Whitelegge JP, Torres JZ, Wang MB, Srivatsan ES. p16 Protein and gigaxonin are associated with the ubiquitination of NFκB in cisplatin-induced senescence of cancer cells. J Biol Chem 2014; 289:34921-37. [PMID: 25331947 DOI: 10.1074/jbc.m114.568543] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The molecular mechanism of p16-mediated senescence in cisplatin-treated cancer cells is not fully understood. Here we show that cisplatin treatment of head and neck cancer cells results in nuclear transport of p16 leading to a molecular modification of NFκB. Chromatin immunoprecipitation assays show that this modification is associated with the inhibition of NFκB interacting with its DNA binding sequences, leading to decreased expression of NFκB-transcribed proteins. LCMS proteomic analysis of LAP-TAP-purified proteins from HeLa cells containing a tetracycline-inducible GFP-S peptide-NFκB expression system identified gigaxonin, an ubiquitin E3 ligase adaptor, as an NFκB-interacting protein. Immunoblotting and siRNA studies confirmed the NFκB-gigaxonin interaction and the dependence of this binding on p16-NFκB binding. Using gel shift assays, we have confirmed p16-NFκB and gigaxonin-NFκB interactions. Furthermore, we have observed increased NFκB ubiquitination with cisplatin treatment that is abolished in the absence of p16 and gigaxonin expression. Analysis of 103 primary tumors has shown that increased nuclear p16 expression correlates with enhanced survival of head and neck cancer patients (p < 0.0000542), indicating the importance of nuclear p16 expression in prognosis. Finally, p16 expression is associated with reduced cytokine expression and the presence of human papilloma virus in chemoradiation-sensitive basaloid tumors. However, the absence of p16 expression is associated with enhanced cytokine expression and the absence of human papilloma virus in aggressive tumors. These results clearly demonstrate that nuclear p16 and gigaxonin play an important role in chemosensitivity of head and neck cancers through ubiquitination of NFκB.
Collapse
Affiliation(s)
- Mysore S Veena
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| | - Reason Wilken
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| | - Jun-Ying Zheng
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| | - Ankur Gholkar
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095
| | - Natarajan Venkatesan
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| | - Darshni Vira
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073, Department of Head and Neck Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095
| | - Sameer Ahmed
- Department of Head and Neck Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095
| | - Saroj K Basak
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| | - Clifton L Dalgard
- Departments of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland 20814
| | - Sandhiya Ravichandran
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| | - Raj K Batra
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, Department of Medicine and Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095
| | | | | | - Michael C Fishbein
- Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Julian P Whitelegge
- Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, California 90095, and
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095
| | - Marilene B Wang
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073, Department of Head and Neck Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095
| | - Eri S Srivatsan
- From the Department of Surgery, Veterans Affairs Greater Los Angeles Healthcare System/David Geffen School of Medicine at UCLA, Los Angeles, California 90073, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095
| |
Collapse
|
45
|
Gao Y, Trivedi S, Ferris RL, Koide K. Regulation of HPV16 E6 and MCL1 by SF3B1 inhibitor in head and neck cancer cells. Sci Rep 2014; 4:6098. [PMID: 25139387 PMCID: PMC4138523 DOI: 10.1038/srep06098] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/22/2014] [Indexed: 11/16/2022] Open
Abstract
ABT-737 inhibits the anti-apoptotic proteins B-cell lymphoma 2 (BCL-2) and BCL-XL. Meayamycin B switches the splicing pattern of myeloid cell leukemia factor 1 (MCL1) pre-mRNA. Specifically, inhibition of splicing factor 3B subunit 1 (SF3B1) with meayamycin B promotes the generation of the proapoptotic, short splicing variant (MCL1-S) and diminishes the antiapoptotic, long variant (MCL1-L). This action was previously associated with the cytotoxicity of meayamycin B in non-small cell lung carcinoma cell lines. ABT-737 induced apoptosis in response to an ablation of MCL1-L by meayamycin B. In this study, we further exploited this synergistic combination in head and neck squamous cell carcinoma (HNSCC), up to 90% of which overexpress MCL1 and BCL-XL. In a panel of seven HNSCC cell lines, the combination of meayamycin B and ABT-737 rapidly triggered a Bax/Bak-mediated apoptosis that overcame the resistance from HPV16-positive HNSCC against each agent alone. Both RT-PCR and Western blotting showed that meayamycin B up-regulated MCL1-S and down-regulated MCL1-L. Significantly, we discovered that SF3B1 was involved in the splicing of oncogenic HPV16 E6 to produce non-oncogenic HPV16 E6*, indicating that SF3B1 may inhibit HPV16-induced tumorigenesis.
Collapse
Affiliation(s)
- Yang Gao
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, USA
| | - Sumita Trivedi
- University of Pittsburgh Cancer institute, Pittsburgh, Pennsylvania 15213, USA
| | - Robert L Ferris
- University of Pittsburgh Cancer institute, Pittsburgh, Pennsylvania 15213, USA
| | - Kazunori Koide
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, USA
| |
Collapse
|
46
|
Kumar B, Yadav A, Hideg K, Kuppusamy P, Teknos TN, Kumar P. A novel curcumin analog (H-4073) enhances the therapeutic efficacy of cisplatin treatment in head and neck cancer. PLoS One 2014; 9:e93208. [PMID: 24675768 PMCID: PMC3968069 DOI: 10.1371/journal.pone.0093208] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/28/2014] [Indexed: 12/27/2022] Open
Abstract
Chemotherapy constitutes the standard modality of treatment for localized head and neck squamous cell carcinomas (HNSCC). However, many patients fail to respond and relapse after this treatments due to the acquisition of chemo-resistance. Therefore, there is an urgent need to develop novel drugs that could reverse the resistant phenotype. Curcumin, the constituent of the spice turmeric has been shown to have anti-inflammatory, anti-oxidant and anti-proliferative properties in several tumor types. However, use of curcumin has been limited due to its poor bio-absorption. Recently, a novel class of curcumin analogs, based on diarylidenylpiperidones (DAP), has been developed by incorporating a piperidone link to the beta-diketone structure and fluoro substitutions on the phenyl groups. In this study, we evaluated the effectiveness of H-4073, a parafluorinated variant of DAP, using both in vitro and in vivo head and neck cancer models. Our results demonstrate that H-4073 is a potent anti-tumor agent and it significantly inhibited cell proliferation in all the HNSCC cell lines tested in a dose-dependent manner. In addition, pretreatment of cisplatin-resistant HNSCC cell lines with H-4073 significantly reversed the chemo-resistance as observed by cell viability assay (MTT), apoptosis assay (Annexin V binding) and cleaved caspase-3 (Western blot). H-4073 mediated its anti-tumor effects by inhibiting JAK/STAT3, FAK, Akt and VEGF signaling pathways that play important roles in cell proliferation, migration, survival and angiogenesis. In the SCID mouse xenograft model, H-4073 significantly enhanced the anti-tumor and anti-angiogenesis effects of cisplatin, with no added systemic toxicity. Interestingly, H-4073 inhibited tumor angiogenesis by blocking VEGF production by tumor cells as well as directly inhibiting endothelial cell function. Taken together, our results suggest that H-4073 is a potent anti-tumor agent and it can be used to overcome chemotherapy resistance in HNSCC.
Collapse
Affiliation(s)
- Bhavna Kumar
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Arti Yadav
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Kalman Hideg
- Institute of Organic and Medicinal Chemistry, University of Pécs, Pécs, Hungary
| | - Periannan Kuppusamy
- Geisel School of Medicine, Dartmouth, Hanover, New Hampshire, United States of America
| | - Theodoros N. Teknos
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (TT) (TT); (P. Kumar) (PK)
| | - Pawan Kumar
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (TT) (TT); (P. Kumar) (PK)
| |
Collapse
|
47
|
Sobral LM, Sousa LO, Coletta RD, Cabral H, Greene LJ, Tajara EH, Gutkind JS, Curti C, Leopoldino AM. Stable SET knockdown in head and neck squamous cell carcinoma promotes cell invasion and the mesenchymal-like phenotype in vitro, as well as necrosis, cisplatin sensitivity and lymph node metastasis in xenograft tumor models. Mol Cancer 2014; 13:32. [PMID: 24555657 PMCID: PMC3936887 DOI: 10.1186/1476-4598-13-32] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 02/13/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND SET/I2PP2A is a multifunctional protein that is up-regulated in head and neck squamous cell carcinoma (HNSCC). The action of SET in HNSCC tumorigenicity is unknown. METHODS Stable SET knockdown by shRNA (shSET) was established in three HNSCC cell lines: HN12, HN13, and Cal27. Protein expression and phosphorylated protein levels were determined by Western blotting and immunofluorescence, cell migration and invasion were measured by functional analysis, and PP2A activity was determined using a serine/threonine phosphatase assay. A real-time PCR array was used to quantify 84 genes associated with cell motility. Metalloproteinase (MMP) activity was assessed by zymographic and fluorometric assays. HN12shSET xenograft tumors (flank and tongue models) were established in Balb/c nude mice; the xenograft characteristics and cisplatin sensitivity were demonstrated by macroscopic, immunohistochemical, and histological analyses, as well as lymph node metastasis by histology. RESULTS The HN12shSET cells displayed reduced ERK1/2 and p53 phosphorylation compared with control. ShSET reduced HN12 cell proliferation and increased the sub-G1 population of HN12 and Cal27 cells. Increased PP2A activity was also associated with shSET. The PCR array indicated up-regulation of three mRNAs in HN12 cells: vimentin, matrix metalloproteinase-9 (MMP9) and non-muscle myosin heavy chain IIB. Reduced E-cadherin and pan-cytokeratin, as well as increased vimentin, were also demonstrated as the result of SET knockdown. These changes were accompanied by an increase in MMP-9 and MMP-2 activities, migration and invasion. The HN12shSET subcutaneous xenograft tumors presented a poorly differentiated phenotype, reduced cell proliferation, and cisplatin sensitivity. An orthotopic xenograft tumor model using the HN12shSET cells displayed increased metastatic potential. CONCLUSIONS SET accumulation has important actions in HNSCC. As an oncogene, SET promotes cell proliferation, survival, and resistance to cell death by cisplatin in vivo. As a metastasis suppressor, SET regulates invasion, the epithelial mesenchymal transition, and metastasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Andréia M Leopoldino
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Riberião Preto, University of São Paulo, Av, Café, s/n, 14040-903 Ribeirão Preto, SP, Brazil.
| |
Collapse
|
48
|
p27 and BCL2 expression predicts response to chemotherapy in head and neck squamous cell carcinomas. Oral Oncol 2014; 50:128-34. [DOI: 10.1016/j.oraloncology.2013.10.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/06/2013] [Accepted: 10/26/2013] [Indexed: 11/17/2022]
|
49
|
Tchelebi L, Ashamalla H, Graves PR. Mutant p53 and the response to chemotherapy and radiation. Subcell Biochem 2014; 85:133-59. [PMID: 25201193 DOI: 10.1007/978-94-017-9211-0_8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In addition to playing roles in the genesis and progression of cancer, mutant p53 also appears to play a significant role in the response to cancer therapy. In response to chemotherapy and radiation, two mainstays of cancer treatment, most cancer cells harboring p53 mutations show a reduced sensitivity compared to cells lacking p53 or those with wild type p53. However, there are also many instances where mutant p53 has shown no effect or enhances cellular sensitivity to chemotherapy and radiation. Similar to the in vitro cellular studies, the majority of clinical studies show a correlation between the presence of mutant p53 in patient tumors and adverse outcomes following treatment with chemotherapy agents or radiation in comparison to tumors with wild-type p53. However, it still remains unclear whether the presence of mutant p53 in tumors can serve as a reliable prognostic factor and aid in treatment planning. Thus, as genomic analysis of patient tumors becomes more cost effective, the role of mutant p53 in tumor responses from cancer therapy ultimately needs to be addressed. This chapter will discuss current mechanisms of how p53 mutations affect cellular responses to chemotherapy and radiation and discuss patient outcomes based on p53 status.
Collapse
Affiliation(s)
- Leila Tchelebi
- Department of Radiation Oncology, New York Methodist Hospital, 506 6th Street, Brooklyn, NY, 11215, USA
| | | | | |
Collapse
|
50
|
Almeida LO, Abrahao AC, Rosselli-Murai LK, Giudice FS, Zagni C, Leopoldino AM, Squarize CH, Castilho RM. NFκB mediates cisplatin resistance through histone modifications in head and neck squamous cell carcinoma (HNSCC). FEBS Open Bio 2013; 4:96-104. [PMID: 24490130 PMCID: PMC3907686 DOI: 10.1016/j.fob.2013.12.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/13/2013] [Accepted: 12/22/2013] [Indexed: 12/20/2022] Open
Abstract
Cisplatin-based chemotherapy is the standard treatment of choice for head and neck squamous cell carcinoma (HNSCC). The efficiency of platinum-based therapies is directly influenced by the development of tumor resistance. Multiple signaling pathways have been linked to tumor resistance, including activation of nuclear factor kappa B (NFκB). We explore a novel mechanism by which NFκB drives HNSCC resistance through histone modifications. Post-translational modification of histones alters chromatin structure, facilitating the binding of nuclear factors that mediate DNA repair, transcription, and other processes. We found that chemoresistant HNSCC cells with active NFκB signaling respond to chemotherapy by reducing nuclear BRCA1 levels and by promoting histone deacetylation (chromatin compaction). Activation of this molecular signature resulted in impaired DNA damage repair, prolonged accumulation of histone γH2AX and increased genomic instability. We found that pharmacological induction of histone acetylation using HDAC inhibitors prevented NFκB-induced cisplatin resistance. Furthermore, silencing NFκB in HNSCC induced acetylation of tumor histones, resulting in reduced chemoresistance and increased cytotoxicity following cisplatin treatment. Collectively, these findings suggest that epigenetic modifications of HNSCC resulting from NFκB-induced histone modifications constitute a novel molecular mechanism responsible for chemoresistance in HNSCC. Therefore, targeted inhibition of HDAC may be used as a viable therapeutic strategy for disrupting tumor resistance caused by NFκB. Chemoresistant HNSCC cells have deacetylation of histones and active NFκB signaling. Histone deacetylation reduces BRCA1 levels and enhances genomic instability. Histone deacetylase (HDAC) inhibitors sensitize HNSCC to chemotherapy. NFκB signaling drives HNSCC chemoresistance by inducing histone deacetylation. NFκB inhibition results in histone acetylation and sensitizes HNSCC to chemotherapy.
Collapse
Key Words
- BRCA1, breast cancer type 1
- BSA, bovine serum albumin
- Chemoresistance
- Chromatin remodeling
- DDR, DNA damage repair
- DMSO, dimethyl sulfoxide
- DSB, double strand breaks
- HDAC inhibitor
- HDAC, histone deacetylases
- HNSCC
- HNSCC, head and neck squamous cell carcinoma
- Histone acetylation
- IC50, half maximal inhibitory concentration
- IKKα, IκB kinase alpha
- IKKβ, IκB kinase beta
- MTS, non-radioactive cell proliferation assay
- NFκB
- NFκB, nuclear factor kappa B
- NIH, National Institutes of Health
- TSA, trichostatin A
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Luciana O Almeida
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA ; Department of Clinical Analysis, Toxicology and Bromatology, School of Pharmacy, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Aline C Abrahao
- Department of Pathology and Oral Diagnosis, Federal University of Rio de Janeiro School of Dentistry, Rio de Janeiro, RJ, Brazil
| | - Luciana K Rosselli-Murai
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Fernanda S Giudice
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Chiara Zagni
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Andreia M Leopoldino
- Department of Clinical Analysis, Toxicology and Bromatology, School of Pharmacy, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Rogerio M Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|