1
|
Pérez-de-Oliveira ME, Wagner VP, Bingle CD, Vargas PA, Bingle L. Disruption of oncogenic pathways in mucoepidermoid carcinoma: CREB inhibitor 666.15 as a potential therapeutic agent. Oral Oncol 2024; 159:107029. [PMID: 39332274 DOI: 10.1016/j.oraloncology.2024.107029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/07/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVES Mucoepidermoid carcinoma (MEC) is the most common malignant salivary gland tumour with around 50 % of cases carrying the CRTC1-MAML2 translocation. The CREB pathway has been associated with the transforming activity of this translocation. The aim of this study was to determine the effects of CREB inhibition on MEC cell behaviour in vitro. MATERIAL AND METHODS Two translocation-positive (UM-HMC-2 and H292) and one translocation-negative (H253) MEC cell lines were treated with 666.15, a CREB inhibitor. Drug IC50 doses were determined for each cell line. Clonogenic and spheroid assays were used to assess survival, including percentage of cancer stem cells, and transwell and scratch assays evaluated invasive and migratory capacities, respectively. Immunofluorescence staining was used to determine E-cadherin expression. RESULTS CREB inhibition significantly reduced the number of surviving colonies and spheroids and delayed cell invasion in all cell lines, but this was more significant in the fusion positive, UM-HMC-2 cells. The expression of E-cadherin was significantly higher in treated UM-HMC-2 and H292 cells. CONCLUSION CREB inhibition with 666.15 impaired key MEC oncogenic behaviours associated with metastasis and drug resistance, including cell invasion and survival.
Collapse
Affiliation(s)
- Maria Eduarda Pérez-de-Oliveira
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Vivian Petersen Wagner
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom; Department of Oral Medicine, School of Dentistry, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Colin D Bingle
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil
| | - Lynne Bingle
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
2
|
Silva LC, Pérez-de-Oliveira ME, Pedroso CM, Leite AA, Santos-Silva AR, Lopes MA, Junior GD, Martins MD, Wagner VP, Kowalski LP, Squarize CH, Castilho RM, Vargas PA. Systemic therapies for salivary gland carcinomas: an overview of published clinical trials. Med Oral Patol Oral Cir Bucal 2024; 29:e280-e287. [PMID: 38150606 PMCID: PMC10945875 DOI: 10.4317/medoral.26264] [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: 07/31/2023] [Accepted: 11/20/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND There is no consensus about effective systemic therapy for salivary gland carcinomas (sgcs). Our aim was summarized the clinical trials assessing the systemic therapies (ST) on sgcs. MATERIAL AND METHODS Electronic searches were carried out through MEDLINE/pubmed, EMBASE, Scopus, Web of Science, and the Cochrane Library databases, and gray literature. RESULTS Seventeen different drugs were evaluated, and the most frequent histological subtype was adenoid cystic carcinoma (n=195, 45.5%). Stable disease, observed in 11 ST, achieved the highest rate in adenoid cystic carcinoma treated with sunitinib. The highest complete (11.1%) and partial response (30.5%) rates were seen in androgen receptor-positive tumors treated with leuprorelin acetate. CONCLUSIONS Despite all the advances in this field, there is yet no effective evidence-based regimen of ST, with all the clinical trials identified showing low rates of complete and partial responses. Further, translational studies are urgently required to characterize molecular targets and effective ST.
Collapse
Affiliation(s)
- L-C Silva
- Department of Oral Diagnosis Piracicaba Dental School, University of Campinas Av Limeira, 901, Postal code:13414-016, Piracicaba, São Paulo, Brasil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Jayawickrama SM, Ranaweera PM, Pradeep RGGR, Jayasinghe YA, Senevirathna K, Hilmi AJ, Rajapakse RMG, Kanmodi KK, Jayasinghe RD. Developments and future prospects of personalized medicine in head and neck squamous cell carcinoma diagnoses and treatments. Cancer Rep (Hoboken) 2024; 7:e2045. [PMID: 38522008 PMCID: PMC10961052 DOI: 10.1002/cnr2.2045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Precision healthcare has entered a new era because of the developments in personalized medicine, especially in the diagnosis and treatment of head and neck squamous cell carcinoma (HNSCC). This paper explores the dynamic landscape of personalized medicine as applied to HNSCC, encompassing both current developments and future prospects. RECENT FINDINGS The integration of personalized medicine strategies into HNSCC diagnosis is driven by the utilization of genetic data and biomarkers. Epigenetic biomarkers, which reflect modifications to DNA that can influence gene expression, have emerged as valuable indicators for early detection and risk assessment. Treatment approaches within the personalized medicine framework are equally promising. Immunotherapy, gene silencing, and editing techniques, including RNA interference and CRISPR/Cas9, offer innovative means to modulate gene expression and correct genetic aberrations driving HNSCC. The integration of stem cell research with personalized medicine presents opportunities for tailored regenerative approaches. The synergy between personalized medicine and technological advancements is exemplified by artificial intelligence (AI) and machine learning (ML) applications. These tools empower clinicians to analyze vast datasets, predict patient responses, and optimize treatment strategies with unprecedented accuracy. CONCLUSION The developments and prospects of personalized medicine in HNSCC diagnosis and treatment offer a transformative approach to managing this complex malignancy. By harnessing genetic insights, biomarkers, immunotherapy, gene editing, stem cell therapies, and advanced technologies like AI and ML, personalized medicine holds the key to enhancing patient outcomes and ushering in a new era of precision oncology.
Collapse
Affiliation(s)
| | | | | | | | - Kalpani Senevirathna
- Centre for Research in Oral Cancer, Faculty of Dental SciencesUniversity of PeradeniyaKandySri Lanka
| | | | | | - Kehinde Kazeem Kanmodi
- School of DentistryUniversity of RwandaKigaliRwanda
- Faculty of DentistryUniversity of PuthisastraPhnom PenhCambodia
- Cephas Health Research Initiative IncIbadanNigeria
- School of Health and Life SciencesTeesside UniversityMiddlesbroughUK
| | - Ruwan Duminda Jayasinghe
- Centre for Research in Oral Cancer, Faculty of Dental SciencesUniversity of PeradeniyaKandySri Lanka
- Faculty of DentistryUniversity of PuthisastraPhnom PenhCambodia
- School of Health and Life SciencesTeesside UniversityMiddlesbroughUK
- Department of Oral Medicine and Periodontology, Faculty of Dental SciencesUniversity of PeradeniyaKandySri Lanka
| |
Collapse
|
4
|
Almeida LO, Silva LC, Emerick C, Amorim Dos Santos J, Castilho RM, Squarize CH. Head and neck cancer stem cell maintenance relies on mTOR signaling, specifically involving the mechanistic target of rapamycin complexes 1 and 2 (mTORC1 and mTORC2). Arch Oral Biol 2024; 157:105840. [PMID: 37939517 DOI: 10.1016/j.archoralbio.2023.105840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/13/2023] [Accepted: 10/29/2023] [Indexed: 11/10/2023]
Abstract
OBJECTIVE Emerging evidence suggests that the modest response of head and neck squamous cell carcinoma (HNSCC) to treatment is associated with cancer stem cells (CSC). However, the signaling pathways that play a role in HNSCC CSC maintenance and therapy response are not well-understood. In this study, we investigate the response of CSCs to phosphatase and tensin homolog (PTEN) modulation and its potential dependency on the mammalian target of rapamycin (mTOR) signaling. DESIGN PTEN deficiency was stably induced using short hairpin RNA (shRNA). Downregulation of RPTOR/mTORC1 and RICTOR/mTORC2 was achieved using small interfering RNA (siRNA). CSCs were evaluated through tumorsphere formation and were classified into various subtypes: parasphere, merosphere, and holosphere. We investigated the effect of rapamycin on CSC properties in both control and PTEN-deficient HNSCC cells. RESULTS PTEN deficiency led to an accumulation of CSCs and enhanced a favorable response to rapamycin treatment. The viability of HNSCC CSCs was dependent on mTOR signaling. Deficiencies in both mTORC1 and mTORC2 reduced the number of CSCs. However, CSCs with PTEN deficiency had a greater reliance on mTORC1 signaling. Interestingly, when considering CSC subtypes, a deficiency in mTORC2 led to an increased number of paraspheres in both the control and PTEN-deficient groups. CONCLUSIONS Loss of PTEN signaling increased the HNSCC CSC population, which can be targeted by rapamycin. However, the mTORC2 deficiency can induce a problematic selection of paraspheres CSCs subtype.
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 Basic and Oral Biology, University of Sao Paulo School of Dentistry, Ribeirao Preto, São Paulo, Brazil
| | - Luan César Silva
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA; Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Carolina Emerick
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA; Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Juliana Amorim Dos Santos
- 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
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Silva LC, Leite AA, Borgato GB, Wagner VP, Martins MD, Loureiro FJA, Lopes MA, Santos-Silva AR, Sperandio M, de Castro Junior G, Kowalski LP, Squarize CH, Castilho RM, Vargas PA. Oral squamous cell carcinoma cancer stem cells have different drug sensitive to pharmacological NFκB and histone deacetylation inhibition. Am J Cancer Res 2023; 13:6038-6050. [PMID: 38187064 PMCID: PMC10767341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/06/2023] [Indexed: 01/09/2024] Open
Abstract
Despite many progresses in the development of new systemic therapies for oral squamous cell carcinoma (OSCC), the five-year survival rate of OSCC is low. The traditional chemotherapies approach (cisplatin - CDDP) shows some limitations like drug toxicity, limited efficacy, and drug resistance. Promising studies suggested OSCC cancer stem cells (CSC) presented resistance to CDDP. We have previously studied many targets, and we extensively showed the efficacy of the NFκB signaling and the role of histones acetylation, on different malignant tumors, including adenoid cystic carcinoma and mucoepidermoid carcinoma, but until then the effects of the NFkB inhibitor and histone deacetylase (HDAC) inhibitor on the biology of OSCC were not evaluated. Here we assessed the pharmacological inhibitor of NFκB emetine and HDAC inhibitor SAHA on the behavior of CSC derived from OSCC. Our data suggested that CDDP administration resulted in reduced viability of bulk OSCC cells and increased CSC. A single and isolated shot of emetine and SAHA were able to disrupt CSC by inhibiting the NFκB pathway and increasing the histone acetylation levels, respectively. Further, the combined administration of emetine and SAHA presented the same CSC disruption as seen in emetine alone.
Collapse
Affiliation(s)
- Luan César Silva
- Department of Oral Diagnosis, Piracicaba Dental School, University of CampinasPiracicaba, SP, Brazil
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of DentistryAnn Arbor, MI, USA
| | - Amanda Almeida Leite
- Department of Oral Diagnosis, Piracicaba Dental School, University of CampinasPiracicaba, SP, Brazil
| | | | - Vivian Petersen Wagner
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, Department of Clinical Dentistry, University of SheffieldSheffield, SY, UK
| | - Manoela Domingues Martins
- Department of Oral Diagnosis, Piracicaba Dental School, University of CampinasPiracicaba, SP, Brazil
- Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do SulPorto Alegre, Brazil
| | | | - Márcio Ajudarte Lopes
- Department of Oral Diagnosis, Piracicaba Dental School, University of CampinasPiracicaba, SP, Brazil
| | - Alan Roger Santos-Silva
- Department of Oral Diagnosis, Piracicaba Dental School, University of CampinasPiracicaba, SP, Brazil
| | - Marcelo Sperandio
- Department of Oral Pathology & Medicine, Sao Leopoldo Mandic Dental Institute and Research CenterCampinas, SP, Brazil
| | - Gilberto de Castro Junior
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São PauloSão Paulo, SP, Brazil
| | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery, University of Sao Paulo Medical School and Head and Neck Surgery and Otorhinolaryngology Department, AC Camargo Cancer CenterSão Paulo, SP, Brazil
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of DentistryAnn Arbor, MI, USA
| | - Rogerio Moraes Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of DentistryAnn Arbor, MI, USA
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, Piracicaba Dental School, University of CampinasPiracicaba, SP, Brazil
| |
Collapse
|
6
|
Joshi P, Bhattacharya K, Menon MB, Ghosh Laskar S, Joshi A, Patil V, Chaturvedi P. Malignant Minor Salivary gland neoplasms ofLarynx: Our Experience. OTOLARYNGOLOGIA POLSKA 2023; 77:1-7. [PMID: 38032326 DOI: 10.5604/01.3001.0053.4040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
<br><b>Introduction:</b> Malignant minor salivary gland tumors are rare, accounting for fewer than 1% of all laryngeal cancers.</br> <br><b>Aim:</b> This study aims to share our experiences regarding clinical, radiological, pathological profiles and their management.</br> <br><b>Materials and methods:</b> The current study reviews 11 cases of malignant minor salivary gland tumors of the larynx treated surgically at our Institute between 2005 and 2019.</br> <br><b>Results:</b> The mean age of the patients was 54 years (range 38-75 years) with six females and five males in the series (1.2:1). Subglottis and trachea were the sites of origin in 54% of the cases, and hoarseness with dyspnea were the most common presenting symptoms. There were nine Adenoid cystic and two Mucoepidermoid carcinoma patients. Surgery was the primary mode of treatment.</br> <br><b>Conclusions:</b> Most of the larynx's malignant minor salivary gland tumors are submucosal in origin. The outcome and prognosis vary considerably based on the tumor's histology, grade, and stage.</br>.
Collapse
Affiliation(s)
- Poonam Joshi
- Department of Head and Neck Surgery, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Kajari Bhattacharya
- Department of Radiology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Munita Bal Menon
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sarbani Ghosh Laskar
- Department of Radiotherapy, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Amit Joshi
- Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Vijay Patil
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Pankaj Chaturvedi
- Department of Head and Neck Surgery, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
7
|
Tan Y, Wang Z, Xu M, Li B, Huang Z, Qin S, Nice EC, Tang J, Huang C. Oral squamous cell carcinomas: state of the field and emerging directions. Int J Oral Sci 2023; 15:44. [PMID: 37736748 PMCID: PMC10517027 DOI: 10.1038/s41368-023-00249-w] [Citation(s) in RCA: 171] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/25/2023] [Accepted: 09/04/2023] [Indexed: 09/23/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) develops on the mucosal epithelium of the oral cavity. It accounts for approximately 90% of oral malignancies and impairs appearance, pronunciation, swallowing, and flavor perception. In 2020, 377,713 OSCC cases were reported globally. According to the Global Cancer Observatory (GCO), the incidence of OSCC will rise by approximately 40% by 2040, accompanied by a growth in mortality. Persistent exposure to various risk factors, including tobacco, alcohol, betel quid (BQ), and human papillomavirus (HPV), will lead to the development of oral potentially malignant disorders (OPMDs), which are oral mucosal lesions with an increased risk of developing into OSCC. Complex and multifactorial, the oncogenesis process involves genetic alteration, epigenetic modification, and a dysregulated tumor microenvironment. Although various therapeutic interventions, such as chemotherapy, radiation, immunotherapy, and nanomedicine, have been proposed to prevent or treat OSCC and OPMDs, understanding the mechanism of malignancies will facilitate the identification of therapeutic and prognostic factors, thereby improving the efficacy of treatment for OSCC patients. This review summarizes the mechanisms involved in OSCC. Moreover, the current therapeutic interventions and prognostic methods for OSCC and OPMDs are discussed to facilitate comprehension and provide several prospective outlooks for the fields.
Collapse
Affiliation(s)
- Yunhan Tan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Mengtong Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jing Tang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| |
Collapse
|
8
|
Lu Y, Yang J, Zhu J, Shu Y, Zou X, Ruan Q, Luo S, Wang Y, Wen J. Advances in the Histone Acetylation Modification in the Oral Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2023; 2023:4616682. [PMID: 39282225 PMCID: PMC11401686 DOI: 10.1155/2023/4616682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/23/2022] [Accepted: 11/29/2022] [Indexed: 09/18/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is one of the common malignant tumors in the head and neck, characterized by high malignancy, rapid growth and metastasis, high invasive ability, and high mortality. In recent years, surgery combined with chemotherapy or radiotherapy remains the preferred clinical treatment for OSCC, despite considerable advances in diagnostic and therapeutic techniques. Hence, new targeted therapy is urgently needed. Histone modification affects the function of massive cells through histone acetyltransferase and histone deacetylase. Accompanied by the progress of some diseases, especially tumors, these proteins often show abnormal functions, and by reversing these abnormalities with drugs or gene therapy, the cancer phenotype can even be restored to normal. As a result, they are potential drug targets. This article reviewed the role of the histone dynamic process of acetylation modifications and their associated active modifying enzymes in the pathogenesis and progress of OSCC. Moreover, we explored the value of histone acetylation modification as a potential therapeutic target and the new progress of related drugs in clinical treatment.
Collapse
Affiliation(s)
- Ying Lu
- School of Stomatology, Southern Medical University, Guangzhou 510515, China
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Jinjin Yang
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Junwen Zhu
- Harbin Medical University Cancer Hospital, Harbin, Helongjiang 150081, China
| | - Yao Shu
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Xuan Zou
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Qiao Ruan
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuyuan Luo
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Yong Wang
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Jun Wen
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
9
|
Shahoumi LA. Oral Cancer Stem Cells: Therapeutic Implications and Challenges. FRONTIERS IN ORAL HEALTH 2022; 2:685236. [PMID: 35048028 PMCID: PMC8757826 DOI: 10.3389/froh.2021.685236] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is currently one of the 10 most common malignancies worldwide, characterized by a biologically highly diverse group of tumors with non-specific biomarkers and poor prognosis. The incidence rate of HNSCC varies widely throughout the world, with an evident prevalence in developing countries such as those in Southeast Asia and Southern Africa. Tumor relapse and metastasis following traditional treatment remain major clinical problems in oral cancer management. Current evidence suggests that therapeutic resistance and metastasis of cancer are mainly driven by a unique subpopulation of tumor cells, termed cancer stem cells (CSCs), or cancer-initiating cells (CICs), which are characterized by their capacity for self-renewal, maintenance of stemness and increased tumorigenicity. Thus, more understanding of the molecular mechanisms of CSCs and their behavior may help in developing effective therapeutic interventions that inhibit tumor growth and progression. This review provides an overview of the main signaling cascades in CSCs that drive tumor repropagation and metastasis in oral cancer, with a focus on squamous cell carcinoma. Other oral non-SCC tumors, including melanoma and malignant salivary gland tumors, will also be considered. In addition, this review discusses some of the CSC-targeted therapeutic strategies that have been employed to combat disease progression, and the challenges of targeting CSCs, with the aim of improving the clinical outcomes for patients with oral malignancies. Targeting of CSCs in head and neck cancer (HNC) represents a promising approach to improve disease outcome. Some CSC-targeted therapies have already been proven to be successful in pre-clinical studies and they are now being tested in clinical trials, mainly in combination with conventional treatment regimens. However, some studies revealed that CSCs may not be the only players that control disease relapse and progression of HNC. Further, clinical research studying a combination of therapies targeted against head and neck CSCs may provide significant advances.
Collapse
Affiliation(s)
- Linah A Shahoumi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
10
|
Silva LC, Borgato GB, Wagner VP, Martins MD, Rocha GZ, Lopes MA, Santos-Silva AR, de Castro Júnior G, Kowalski LP, Nor JE, Squarize CH, Castilho RM, Vargas PA. Cephaeline is an inductor of histone H3 acetylation and inhibitor of mucoepidermoid carcinoma cancer stem cells. J Oral Pathol Med 2021; 51:553-562. [PMID: 34661317 DOI: 10.1111/jop.13252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/21/2021] [Accepted: 09/08/2021] [Indexed: 01/02/2023]
Abstract
AIM To evaluate the potential use of Cephaeline as a therapeutic strategy to manage mucoepidermoid carcinomas (MEC) of the salivary glands. MATERIAL AND METHODS UM-HMC-1, UM-HMC-2, and UM-HMC-3A MEC cell lines were used to establish the effects of Cephaeline over tumor viability determined by MTT assay. In vitro wound healing scratch assays were performed to address cellular migration while immunofluorescence staining for histone H3 lysine 9 (H3k9ac) was used to identify the acetylation status of tumor cells upon Cephaeline administration. The presence of cancer stem cells was evaluated by the identification of ALDH enzymatic activity by flow cytometry and through functional assays using in vitro tumorsphere formation. RESULTS A single administration of Cephaeline resulted in reduced viability of MEC cells along with the halt on tumor growth and cellular migration potential. Administration of Cephaeline resulted in chromatin histone acetylation as judged by the increased levels of H3K9ac and disruption of tumorspheres formation. Interestingly, ALDH levels were increased in UM-HMC-1 and UM-HMC-3A cell lines, while UM-HMC-2 showed a reduced enzymatic activity. CONCLUSION Cephaeline has shown anti-cancer properties in all MEC cell lines tested by regulating tumor cells' viability, migration, proliferation, and disrupting the ability of cancer cells to generate tumorspheres.
Collapse
Affiliation(s)
- Luan César Silva
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | | | - Vivian Petersen Wagner
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, Department of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Manoela Domingues Martins
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, São Paulo, Brazil.,Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Zweig Rocha
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Márcio Ajudarte Lopes
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Alan Roger Santos-Silva
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Gilberto de Castro Júnior
- Clinical Oncology Service, São Paulo State Cancer Institute (ICESP), School of Medicine of the University of São Paulo, São Paulo, Brazil
| | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery, Faculty of Medicine, Head and Neck Surgery and Otorhinolaryngology Department, A C Camargo Cancer Center, Universidade de São Paulo, São Paulo, Brazil
| | - Jacques E Nor
- Department of Cariology, Restorative Sciences, Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Rogerio Moraes Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| |
Collapse
|
11
|
Picon H, Guddati AK. Cancer stem cells in head and neck cancer. AMERICAN JOURNAL OF STEM CELLS 2021; 10:28-35. [PMID: 34552815 PMCID: PMC8449141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/15/2021] [Indexed: 06/13/2023]
Abstract
Cancer stem cells (CSCs) are a unique population of cells found within tumors that are able to self-renew, restore the original heterogeneity of a tumor following treatment, and show increased tumorigenic potential when compared to other cancer cells. It is thought that they are responsible for the recurrence of tumors as well as the resistance to treatment that is seen clinically. CSCs are known to be involved in head and neck cancer (HNCs) specifically, as evidence for their existence can be found in head and neck squamous cell carcinoma (HNSCC), mucoepidermoid carcinoma (MEC), and adenoid cystic carcinoma (ACC), among others. Here, findings from various approaches to identifying and targeting CSCs and their downstream effectors in HNC are summarized, with an emphasis on recent advancements. Prognostic and therapeutic markers are discussed for each specific type of HNC, and novel treatment strategies and current clinical trials involving CSCs are detailed as well. The information provided here is intended to further the research on this important topic and lead to clinical impact in the battle against HNC.
Collapse
Affiliation(s)
- Hector Picon
- Medical College of Georgia, Augusta UniversityAugusta 30909, GA, USA
| | - Achuta Kumar Guddati
- Division of Hematology/Oncology, Georgia Cancer Center, Augusta UniversityAugusta 30912, GA, USA
| |
Collapse
|
12
|
Moura JMBDO, Gonzaga AKG, Queiroz SIML, Martins MD, Pinto LP, Souza LBD. Immunohistochemical expression of OCT4 and CD44 in major and minor salivary gland neoplasms. Braz Oral Res 2021; 35:e073. [PMID: 34161412 DOI: 10.1590/1807-3107bor-2021.vol35.0073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/22/2020] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to identify tumor parenchyma cells exhibiting immunohistochemical profile of stem cells by evaluating the immunoreactivity of OCT4 and CD44 in a number of cases of salivary gland neoplasms. The sample consisted of 20 pleomorphic adenomas, 20 mucoepidermoid carcinomas, and 20 adenoid cystic carcinomas located in major and minor salivary glands. The expression of OCT4 and CD44 was evaluated by the percentage of positive cells and the intensity of expression. All studied cases showed positive expression of OCT4 and CD44 and higher values than the control groups. For OCT4, luminal and non-luminal cells were immunostained in the case of pleomorphic adenomas and adenoid cystic carcinomas. Moreover, the immunoreactivity of CD44 was particularly evident in the non-luminal cells of these lesions. In mucoepidermoid carcinomas, there was immunoreactivity for both markers in squamous and intermediate cells and absence of staining in mucous cells. For both markers, a significantly higher immunostaining was verified in neoplasms located in the major salivary glands compared with lesions in minor salivary glands (p<0.001). In the total sample and in minor salivary glands, malignant neoplasms exhibited higher immunoreactivity for OCT4 than pleomorphic adenoma. A significant moderate positive correlation (r = 0.444 and p ≤ 0.001) was found between OCT4 and CD44 immunoexpression in the total sample. The high expression of OCT4 and CD44 may indicate that these proteins play an important role in identifying tumor stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Leão Pereira Pinto
- Universidade Federal do Rio Grande do Norte - UFRN, Department of Dentistry, Natal, RN, Brazil
| | - Lélia Batista de Souza
- Universidade Federal do Rio Grande do Norte - UFRN, Department of Dentistry, Natal, RN, Brazil
| |
Collapse
|
13
|
Parag-Sharma K, Tasoulas J, Musicant AM, do Nascimento-Filho CHV, Zhu Z, Twomey C, Liu P, Castilho RM, Amelio AL. Synergistic efficacy of combined EGFR and HDAC inhibitors overcomes tolerance to EGFR monotherapy in salivary mucoepidermoid carcinoma. Oral Oncol 2021; 115:105166. [PMID: 33581505 PMCID: PMC8026571 DOI: 10.1016/j.oraloncology.2020.105166] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/16/2020] [Accepted: 12/25/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Mucoepidermoid carcinoma (MEC) is the most common type of salivary gland malignancy. Advanced or high-grade MECs are refractory to chemotherapy, often leading to tumor recurrence/metastasis and abysmal ~35% 5-year survival. Causal links have been established between Epithelial Growth Factor Receptor (EGFR) activation and poor outcome. Herein we investigated the therapeutic efficacy of EGFR inhibition against MEC using in vitro pre-clinical models. MATERIALS AND METHODS Five human MEC cell lines were used in cell viability, cytotoxicity, apoptosis, cell cycle, 2D-clonogenicity, and 3D-spheroid formation assays following treatment with Erlotinib (EGFR inhibitor), SAHA (Histone Deacetylase inhibitor; HDAC) and CUDC-101 (dual EGFR-HDAC inhibitor). Effects on MEC cancer stem cells were evaluated using flow cytometry. Gene expression and pathway regulation were evaluated via qPCR and Western blot, respectively. RESULTS MEC cells enter a quiescent, non-proliferative yet rapidly reversible drug tolerant state upon EGFR inhibition. Despite robust suppression of MEC cell proliferation, no discernable apoptosis is detected. Combination of EGFR and HDAC inhibitors exhibits synergistic effects, exerting ~5-fold more potent cell cytotoxicity compared to HDAC or EGFR monotherapy. CUDC-101, a single molecule with dual EGFR-HDAC inhibitor moieties, exerts irreversible and potent cytotoxic activity against MEC cells and blunts MEC cancer stem-cell tumorigenicity. CONCLUSION MEC cells are intrinsically tolerant to EGFR inhibition. Combining EGFR and HDAC inhibitors exerts synergistic and potent cytotoxic effects, suggesting that EGFR inhibitors still hold significant promise against MEC. Future studies are needed to assess the applicability and efficacy of dual EGFR-HDAC inhibitors for the clinical management of MEC.
Collapse
Affiliation(s)
- Kshitij Parag-Sharma
- Graduate Curriculum in Cell Biology and Physiology, Biological and Biomedical Sciences Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason Tasoulas
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Oral and Craniofacial Health Sciences, UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adele M Musicant
- Graduate Curriculum in Genetics and Molecular Biology, Biological and Biomedical Sciences, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carlos H Viesi do Nascimento-Filho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Zhichuan Zhu
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Biochemistry and Biophysics, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chloe Twomey
- Carolina Research Scholar, Undergraduate Curriculum in Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Biochemistry and Biophysics, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rogerio M Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Antonio L Amelio
- Division of Oral and Craniofacial Health Sciences, UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Cell Biology and Physiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Cancer Cell Biology Program, Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Biomedical Research Imaging Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
14
|
Pouloudi D, Manou M, Sarantis P, Tsoukalas N, Tsourouflis G, Dana E, Karamouzis MV, Klijanienko J, Theocharis S. Clinical Significance of Histone Deacetylase (HDAC)-1, -2, -4 and -6 Expression in Salivary Gland Tumors. Diagnostics (Basel) 2021; 11:diagnostics11030517. [PMID: 33799478 PMCID: PMC8000873 DOI: 10.3390/diagnostics11030517] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Salivary gland tumors (SGTs) comprise a group of rare neoplasms. Locally aggressive, recurrent and/or metastatic SGTs are notorious for their resistance to systemic therapy, making the need for carefully designed, prospective and randomized trials with useful predictive markers mandatory to define new effective therapeutic protocols. Histone Deacetylases (HDACs), are thought to play a crucial role in carcinogenesis. They affect the DNA structure, being also able to regulate its transcription, repair, and replication. This study aimed to evaluate-to our knowledge for the first time-the HDAC-1, -2, -4 and -6 immunohistochemical expression in SGTs and their potential use as prognostic biomarkers. Medical records and archival histopathological material of 58 (36 benign and 22 malignant) SGT patients were included in this study. The H-score was statistically correlated with the clinicopathological characteristics for all cases and patients' survival rate in malignant SGTs. HDAC-2 positivity was significantly associated with more prolonged overall survival (OS) of patients with malignant SGTs (p = 0.028), while HDAC-2 positivity and no HDAC-6 expression were associated with prolonged OS of patients with HG malignant SGT (p = 0.003 and p = 0.043, respectively). Additionally, a high HDAC-2 H-score was significantly associated with longer OS for HG malignant SGT patients (p = 0.027). In our study, HDAC-2 expression is a marker for good prognosis, whereas HDAC-6 expression indicated poor prognosis; thus, an inhibitor of HDAC-6 may be used to improve patients' survival.
Collapse
Affiliation(s)
- Despoina Pouloudi
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (D.P.); (M.M.); (P.S.); (N.T.); (E.D.)
| | - Maria Manou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (D.P.); (M.M.); (P.S.); (N.T.); (E.D.)
| | - Panagiotis Sarantis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (D.P.); (M.M.); (P.S.); (N.T.); (E.D.)
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Nikolaos Tsoukalas
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (D.P.); (M.M.); (P.S.); (N.T.); (E.D.)
| | - Gerasimos Tsourouflis
- 2nd Department of Propedeutic Surgery, School of Medicine, National and Kapodistrian, University of Athens, 115 27 Athens, Greece;
| | - Eougken Dana
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (D.P.); (M.M.); (P.S.); (N.T.); (E.D.)
| | - Michalis V. Karamouzis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | | | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (D.P.); (M.M.); (P.S.); (N.T.); (E.D.)
- Department of Pathology, Institut Curie, 75248 Paris, France;
- Correspondence: or ; Tel.: +30-210-7462116; Fax: +30-210-7462157
| |
Collapse
|
15
|
Martins MD, Silveira FM, Martins MAT, Almeida LO, Bagnato VS, Squarize CH, Castilho RM. Photobiomodulation therapy drives massive epigenetic histone modifications, stem cells mobilization and accelerated epithelial healing. JOURNAL OF BIOPHOTONICS 2021; 14:e202000274. [PMID: 33025746 DOI: 10.1002/jbio.202000274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 06/11/2023]
Abstract
Emerging evidence indicates the clinical benefits of photobiomodulation therapy (PBMT) in the management of skin and mucosal wounds. Here, we decided to explore the effects of different regiments of PBMT on epithelial cells and stem cells, and the potential implications over the epigenetic circuitry during healing. Scratch-wound migration, immunofluorescence (anti-acetyl-Histone H3, anti-acetyl-CBP/p300 and anti-BMI1), nuclear morphometry and western blotting (anti-Phospho-S6, anti-methyl-CpG binding domain protein 2 [MBD2]) were performed. Epithelial stem cells were identified by the aldehyde dehydrogenase enzymatic levels and sphere-forming assay. We observed that PBMT-induced accelerated epithelial migration and chromatin relaxation along with increased levels of histones acetylation, the transcription cofactors CBP/p300 and mammalian target of rapamycin. We further observed a reduction of the transcription repression-associated protein MBD2 and a reduced number of epithelial stem cells and spheres. In this study, we showed that PBMT could induce epigenetic modifications of epithelial cells and control stem cell fate, leading to an accelerated healing phenotype.
Collapse
Affiliation(s)
- Manoela D Martins
- Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Felipe Martins Silveira
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Marco A T Martins
- Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Department of Oral Medicine, Hospital de Clínicas de Porto Alegre (HCPA/UFRGS), Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Luciana O Almeida
- Laboratory of Tissue Culture, Department of Basic and Oral Biology, University of Sao Paulo School of Dentistry, Ribeirao Preto, Rio Grande do Sul, Brazil
| | - Vanderlei S Bagnato
- São Carlos Institute of Physics, University of São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Rogerio M Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Survival of salivary gland cancer stem cells requires mTOR signaling. Cell Death Dis 2021; 12:108. [PMID: 33479203 PMCID: PMC7820616 DOI: 10.1038/s41419-021-03391-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 01/02/2023]
Abstract
Advanced salivary gland mucoepidermoid carcinoma (MEC) is a relentless cancer that exhibits resistance to conventional chemotherapy. As such, treatment for patients with advanced MEC is tipically radical surgery and radiotherapy. Facial disfigurement and poor quality of life are frequent treatment challenges, and many patients succumb to loco-regional recurrence and/or metastasis. We know that cancer stem-like cells (CSC) drive MEC tumorigenesis. The current study tests the hypothesis that MEC CSC are sensitive to therapeutic inhibition of mTOR. Here, we report a correlation between the long-term clinical outcomes of 17 MEC patients and the intratumoral expression of p-mTOR (p = 0.00294) and p-S6K1 (p = 0.00357). In vitro, we observed that MEC CSC exhibit constitutive activation of the mTOR signaling pathway (i.e., mTOR, AKT, and S6K1), unveiling a potential strategy for targeted ablation of these cells. Using a panel of inhibitors of the mTOR pathway, i.e., rapamycin and temsirolimus (mTOR inhibitors), buparlisib and LY294002 (AKT inhibitors), and PF4708671 (S6K1 inhibitor), we observed consistently dose-dependent decrease in the fraction of CSC, as well as inhibition of secondary sphere formation and self-renewal in three human MEC cell lines (UM-HMC-1,-3A,-3B). Notably, therapeutic inhibition of mTOR with rapamycin or temsirolimus induced preferential apoptosis of CSC, when compared to bulk tumor cells. In contrast, conventional chemotherapeutic drugs (cisplatin, paclitaxel) induced preferential apoptosis of bulk tumor cells and accumulation of CSC. In vivo, therapeutic inhibition of mTOR with temsirolimus caused ablation of CSC and downregulation of Bmi-1 expression (major inducer of stem cell self-renewal) in MEC xenografts. Transplantation of MEC cells genetically silenced for mTOR into immunodeficient mice corroborated the results obtained with temsirolimus. Collectively, these data demonstrated that mTOR signaling is required for CSC survival, and unveiled the therapeutic potential of targeting the mTOR pathway for elimination of highly tumorigenic cancer stem-like cells in salivary gland mucoepidermoid carcinoma.
Collapse
|
17
|
TrkB-Targeted Therapy for Mucoepidermoid Carcinoma. Biomedicines 2020; 8:biomedicines8120531. [PMID: 33255325 PMCID: PMC7759804 DOI: 10.3390/biomedicines8120531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/10/2020] [Accepted: 11/22/2020] [Indexed: 12/21/2022] Open
Abstract
The brain-derived neurotrophic factor (BDNF)/tyrosine receptor kinase B (TrkB) pathway was previously associated with key oncogenic outcomes in a number of adenocarcinomas. The aim of our study was to determine the role of this pathway in mucoepidermoid carcinoma (MEC). Three MEC cell lines (UM-HMC-2, H253 and H292) were exposed to Cisplatin, the TrkB inhibitor, ANA-12 and a combination of these drugs. Ultrastructural changes were assessed through transmission electron microscopy; scratch and Transwell assays were used to assess migration and invasion; and a clonogenic assay and spheroid-forming assay allowed assessment of survival and percentage of cancer stem cells (CSC). Changes in cell ultrastructure demonstrated Cisplatin cytotoxicity, while the effects of ANA-12 were less pronounced. Both drugs, used individually and in combination, delayed MEC cell migration, invasion and survival. ANA-12 significantly reduced the number of CSC, but the Cisplatin effect was greater, almost eliminating this cell population in all MEC cell lines. Interestingly, the spheroid forming capacity recovered, following the combination therapy, as compared to Cisplatin alone. Our studies allowed us to conclude that the TrkB inhibition, efficiently impaired MEC cell migration, invasion and survival in vitro, however, the decrease in CSC number, following the combined treatment of ANA-12 and Cisplatin, was less than that seen with Cisplatin alone; this represents a limiting factor.
Collapse
|
18
|
Theocharis S, Tasoulas J, Masaoutis C, Kokkali S, Klijanienko J. Salivary gland cancer in the era of immunotherapy: can we exploit tumor microenvironment? Expert Opin Ther Targets 2020; 24:1047-1059. [PMID: 32744127 DOI: 10.1080/14728222.2020.1804863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Salivary gland cancers (SGCs) consist of a rare family of neoplasms with varying histology and biological behavior. Therapeutic regimens have been relatively unchanged for decades. The recent successes of immunotherapy have raised hopes for the development of more effective strategies in SGC, thus emphasizing the role of tumor microenvironment (TME) in the design for more effective therapies. AREAS COVERED This review presents an overview of the current knowledge on the pathobiology of SGC TME and discusses the potential of immunotherapeutic targeting. EXPERT OPINION Most data on the role of TME in SGC carcinogenesis are derived from preclinical studies. Signaling cascades of immunotherapeutic interest, PD-1/PD-L1 and PD-1/PD-L2, are active in many SGCs and might be associated with biological behavior and prognosis. Immunotherapeutic attempts are very limited, but recent findings in other tumors on the role of exosomes and PD-L2 signaling suggest that TME of SGCs warrants further research, emphasizing larger cohorts, histology-based stratification, and standardized evaluation of immunomodulatory molecules, to explore the potential of targeting tumor stroma and its signaling cascades. Furthermore, combination of immunotherapies or immunotherapies with the antineoplastic agents targeting AR, HER2, and tyrosine kinases, recently introduced in SGC treatment, constitutes a promising approach for the future.
Collapse
Affiliation(s)
- Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens , Athens, Greece.,Department of Pathology, Institut Curie , Paris, France
| | - Jason Tasoulas
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens , Athens, Greece
| | - Christos Masaoutis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens , Athens, Greece
| | - Stefania Kokkali
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens , Athens, Greece.,First Medical Oncology Clinic, Saint-Savvas Anticancer Hospital , Athens, Greece
| | | |
Collapse
|
19
|
Magno Guimarães D, deLucas da Silva Almeida F, Moraes Castilho R, Eduardo Nor J, Daumas Nunes F. DNA methyltransferase expression is associated with cell proliferation in salivary mucoepidermoid carcinoma. J Oral Pathol Med 2020; 49:1053-1060. [PMID: 32740989 DOI: 10.1111/jop.13092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/11/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The present study investigated the correlation between the expression of DNA methyltransferase (DNMT)1, DNMT3A, and DNMT3B and the proliferation of mucoepidermoid carcinomas (MECs) using the molecular markers Ki-67 and cyclin D1. This study also demonstrates the effects of 5-aza-2-deoxycytidine (5AC) on the MEC tumor cell lines in relation to DNMT1 and DNMT3A expression, and cell-cycle arrest. MATERIALS AND METHODS The immunohistochemistry of DNMT1, DNMT3A, DNMT3B, Ki-67, and cyclin D1 was analyzed in 40 samples of MEC and 15 samples of healthy minor salivary glands. The effects of 5AC on DNMT1 and DNMT3B expression in MEC cell lines were analyzed by Western blot, and the effects of 5AC on the cell cycle were analyzed using flow cytometry. RESULTS The expression of DNMT1 and DNMT3B was more intense in MECs than in healthy salivary glands. A strong correlation was found between the expression of the DNMTs and the proliferation markers. This correlation was validated In Vitro, where treatment with 5AC reduced the expression of the DNMTs and the percentage of cells at the G2/M phase of the cell cycle. CONCLUSION The expression of DNMT1, DNMT3A, DNMT3B is correlated significantly with the expression of Ki-67 and cyclin D1. The treatment with 5AC reduces DNMT expression and decreases the percentage of cells at the G2/M phase of the cell cycle, while increasing the cells at the G0/G1 phase.
Collapse
Affiliation(s)
- Douglas Magno Guimarães
- Dental School, University Center of Pará, Belém, Pará, Brazil.,Department of Surgery and Oral Pathology, João de Barros Barreto University Hospital, Belém, Pará, Brazil
| | | | - Rogerio Moraes Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Jaques Eduardo Nor
- Department of Restorative Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Fabio Daumas Nunes
- Department of Oral Pathology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Dos Santos ES, Ramos JC, Normando AGC, Mariano FV, Paes Leme AF. Epigenetic alterations in salivary gland tumors. Oral Dis 2020; 26:1610-1618. [PMID: 31829479 DOI: 10.1111/odi.13253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/15/2019] [Accepted: 11/26/2019] [Indexed: 12/22/2022]
Abstract
Salivary gland tumors (SGTs) comprise a heterogeneous group of benign and malignant neoplasms that exhibit significant variability in their microscopic appearance, clinical presentation, and biological behavior. The etiologic factors are unknown; however, chromosomic translocation, secondary radiation, and chemotherapy can be associated with the development of SGT. It has been indicated that epigenetic alterations can be responsible for the development and progress of these neoplasms. The epigenetic mechanisms are defined as a set of DNA changes that do not alter the sequence of nucleotide bases but alter the expression of the proteins. These alterations have been studied in the SGT, and they were associated with the development and progress of these neoplasms and may influence on SGT prognosis. Hence, we critically review the currently available data on the participation of epigenetic events on salivary gland tumors.
Collapse
Affiliation(s)
- Erison S Dos Santos
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Joab C Ramos
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Ana Gabriela C Normando
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Fernanda V Mariano
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Adriana F Paes Leme
- Brazilian Bioscience National Laboratory, Center for Research in Energy and Materials, Campinas, Brazil
| |
Collapse
|
21
|
Cancer Stem Cells: Powerful Targets to Improve Current Anticancer Therapeutics. Stem Cells Int 2019; 2019:9618065. [PMID: 31781251 PMCID: PMC6874936 DOI: 10.1155/2019/9618065] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/25/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023] Open
Abstract
A frequent observation in several malignancies is the development of resistance to therapy that results in frequent tumor relapse and metastasis. Much of the tumor resistance phenotype comes from its heterogeneity that halts the ability of therapeutic agents to eliminate all cancer cells effectively. Tumor heterogeneity is, in part, controlled by cancer stem cells (CSC). CSC may be considered the reservoir of cancer cells as they exhibit properties of self-renewal and plasticity and the capability of reestablishing a heterogeneous tumor cell population. The endowed resistance mechanisms of CSC are mainly attributed to several factors including cellular quiescence, accumulation of ABC transporters, disruption of apoptosis, epigenetic reprogramming, and metabolism. There is a current need to develop new therapeutic drugs capable of targeting CSC to overcome tumor resistance. Emerging in vitro and in vivo studies strongly support the potential benefits of combination therapies capable of targeting cancer stem cell-targeting agents. Clinical trials are still underway to address the pharmacokinetics, safety, and efficacy of combination treatment. This review will address the main characteristics, therapeutic implications, and perspectives of targeting CSC to improve current anticancer therapeutics.
Collapse
|
22
|
Interference with the bromodomain epigenome readers drives p21 expression and tumor senescence. Cancer Lett 2019; 461:10-20. [PMID: 31265875 DOI: 10.1016/j.canlet.2019.06.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 01/14/2023]
Abstract
Head and neck cancer (HNSCC) are one of the most common solid malignancies of the world, being responsible for over 350,000 deaths every year. Much of the complications in managing and treating HNSCC advent from the complex genetic and epigenetic landscape of the disease. Emerging information has shown promising results in targeting BRD4, an epigenetic regulator bromodomain that functions as a scaffold for transcription factors at promoters and super-enhancers. Here we show that by disrupting the interaction between BRD4 and histones using the bromodomain inhibitor JQ1, HNSCC cells undergo cell growth arrest followed by cellular senescence. Mechanistically, JQ1 negatively impacted the phosphorylation levels of SIRT1 along with the acetylation levels of mutant p53 (active). In vivo administration of JQ1 resulted in disruption of HNSCC growth along with the activation of cellular senescence, observed by the accumulation of DNA double-strand breaks, p16ink4, accumulation of senescence-associated beta-galactosidase, and loss of phosphorylated Sirt1ser47. Furthermore, we also demonstrate that JQ1 was efficient in reducing the population of cancer stem cells from HNSCC xenografts.
Collapse
|
23
|
Hai L, Szwarc MM, Lonard DM, Rajapakshe K, Perera D, Coarfa C, Ittmann M, Fernandez-Valdivia R, Lydon JP. Short-term RANKL exposure initiates a neoplastic transcriptional program in the basal epithelium of the murine salivary gland. Cytokine 2019; 123:154745. [PMID: 31226438 DOI: 10.1016/j.cyto.2019.154745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022]
Abstract
Although salivary gland cancers comprise only ∼3-6% of head and neck cancers, treatment options for patients with advanced-stage disease are limited. Because of their rarity, salivary gland malignancies are understudied compared to other exocrine tissue cancers. The comparative lack of progress in this cancer field is particularly evident when it comes to our incomplete understanding of the key molecular signals that are causal for the development and/or progression of salivary gland cancers. Using a novel conditional transgenic mouse (K5:RANKL), we demonstrate that Receptor Activator of NFkB Ligand (RANKL) targeted to cytokeratin 5-positive basal epithelial cells of the salivary gland causes aggressive tumorigenesis within a short period of RANKL exposure. Genome-wide transcriptomic analysis reveals that RANKL markedly increases the expression levels of numerous gene families involved in cellular proliferation, migration, and intra- and extra-tumoral communication. Importantly, cross-species comparison of the K5:RANKL transcriptomic dataset with The Cancer Genome Atlas cancer signatures reveals the strongest molecular similarity with cancer subtypes of the human head and neck squamous cell carcinoma. These studies not only provide a much needed transcriptomic resource to mine for novel molecular targets for therapy and/or diagnosis but validates the K5:RANKL transgenic as a preclinical model to further investigate the in vivo oncogenic role of RANKL signaling in salivary gland tumorigenesis.
Collapse
Affiliation(s)
- Lan Hai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou, Henan Province, PR China
| | - Maria M Szwarc
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Dimuthu Perera
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Michael Ittmann
- Department of Pathology, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | | | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
24
|
SERPINB2 Is a Novel Indicator of Cancer Stem Cell Tumorigenicity in Multiple Cancer Types. Cancers (Basel) 2019; 11:cancers11040499. [PMID: 30965654 PMCID: PMC6520756 DOI: 10.3390/cancers11040499] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/12/2022] Open
Abstract
Drug resistance is one of the major characteristics of cancer stem cells (CSCs) and a mechanism of tumor recurrence. Therefore, selectively targeting CSCs may be an effective therapeutic strategy to overcome cancer recurrence. In the present study, we found that exposure to tumorigenic compounds significantly increased the growth potential and stem-cell-like properties of various CSCs. Early-response genes involved in tumorigenesis can be used as specific markers to predict potential tumorigenicity. Importantly, for the first time we identified, a labile tumorigenic response gene—SERPINB2—and showed that tumorigenic compound exposure more profoundly affected its expression in CSCs than in non-stem cancer cells, although both cells exhibit basal expression of SERPINB2 in multiple cancer types. Our data also revealed a strong relationship between the significantly enhanced expression of SERPINB2 and metastatic progression in multiple cancer types. To the best of our knowledge, this is the first study to focus on the functions of SERPINB2 in the tumorigenicity of various CSCs and these findings will facilitate the development of promising tumorigenicity test platforms.
Collapse
|
25
|
Interfering with bromodomain epigenome readers as therapeutic option in mucoepidermoid carcinoma. Cell Oncol (Dordr) 2018; 42:143-155. [PMID: 30539410 DOI: 10.1007/s13402-018-0416-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2018] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Emerging evidence indicates that bromodomains comprise a conserved class of epigenome readers involved in cancer development and inflammation. Bromodomains are associated with epigenetic modifications of gene transcription through interactions with lysine residues of histone tails. Particularly, the bromodomain and extra-terminal domain (BET) family member BRD4 has been found to be involved in the control over oncogenes, including c-MYC, and in the maintenance of downstream inflammatory processes. The objective of this study was to evaluate the effect of pharmacologically displacing BRD4 in mucoepidermoid carcinoma (MEC) cells. METHODS We assessed the presence of BRD4 levels in a panel of human MEC tissue samples in conjunction with histological grading and clinical information. In vitro studies were carried out using human MEC-derived cell lines. The BET inhibitor iBET762 was administered to MEC cells to assess the impact of disrupted BRD4 signaling on colony forming capacities and cell cycle status. The activation of cellular senescence induced by iBET762 was determined by immunohistochemical staining for p16ink4. Flow cytometry was used to identify populations of cancer stem cells in MEC-derived cell lines. RESULTS We found that primary human MECs and MEC-derived cell lines are endowed with high BRD4 expression levels compared to those in normal salivary glands. We also found that, by displacing BRD4 from chromatin using the BET inhibitor iBET762, MEC cells lose their colony forming capacities and undergo G1 cell cycle arrest and senescence. Finally, we found that targeted displacement of BRD4 from chromatin results in depletion of cancer stem cells from the overall MEC cell populations. CONCLUSIONS Our findings indicate that bromodomain-mediated gene regulation constitutes an epigenetic mechanism that is deregulated in MEC cells and that the use of BET inhibitors may serve as a feasible therapeutic strategy to manage MECs.
Collapse
|
26
|
Wagner VP, Martins MD, Martins MAT, Almeida LO, Warner KA, Nör JE, Squarize CH, Castilho RM. Targeting histone deacetylase and NFκB signaling as a novel therapy for Mucoepidermoid Carcinomas. Sci Rep 2018; 8:2065. [PMID: 29391537 PMCID: PMC5794736 DOI: 10.1038/s41598-018-20345-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/15/2018] [Indexed: 02/06/2023] Open
Abstract
Malignancies from the salivary glands are rare and represent 11% of all cancers from the oropharyngeal anatomical area. Mucoepidermoid Carcinomas (MEC) is the most common malignancy from the salivary glands. Low survival rates of high-grade Mucoepidermoid Carcinomas (MEC) are particularly associated with the presence of positive lymph nodes, extracapsular lymph node spread, and perineural invasion. Most recently, the presence of cancer stem cells (CSC), and the activation of the NFκB signaling pathway have been suggested as cues for an acquired resistance phenotype. We have previously shown that NFκB signaling is very active in MEC tumors. Herein, we explore the efficacy of NFκB inhibition in combination with class I and II HDAC inhibitor to deplete the population of CSC and to destroy MEC tumor cells. Our finding suggests that disruption of NFκB signaling along with the administration of HDAC inhibitors constitute an effective strategy to manage MEC tumors.
Collapse
Affiliation(s)
- Vivian P Wagner
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-1078, USA.,Experimental Pathology Unit, Clinics Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Manoela D Martins
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-1078, USA.,Experimental Pathology Unit, Clinics Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Department of Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Marco A T Martins
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-1078, USA.,Experimental Pathology Unit, Clinics Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Luciana O Almeida
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-1078, USA
| | - Kristy A Warner
- Department of Restorative Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Jacques E Nör
- Department of Restorative Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-1078, USA.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rogerio M Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-1078, USA. .,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
27
|
Epigenetic Modifications and Head and Neck Cancer: Implications for Tumor Progression and Resistance to Therapy. Int J Mol Sci 2017; 18:ijms18071506. [PMID: 28704968 PMCID: PMC5535996 DOI: 10.3390/ijms18071506] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 02/06/2023] Open
Abstract
Head and neck squamous carcinoma (HNSCC) is the sixth most prevalent cancer and one of the most aggressive malignancies worldwide. Despite continuous efforts to identify molecular markers for early detection, and to develop efficient treatments, the overall survival and prognosis of HNSCC patients remain poor. Accumulated scientific evidences suggest that epigenetic alterations, including DNA methylation, histone covalent modifications, chromatin remodeling and non-coding RNAs, are frequently involved in oral carcinogenesis, tumor progression, and resistance to therapy. Epigenetic alterations occur in an unsystematic manner or as part of the aberrant transcriptional machinery, which promotes selective advantage to the tumor cells. Epigenetic modifications also contribute to cellular plasticity during tumor progression and to the formation of cancer stem cells (CSCs), a small subset of tumor cells with self-renewal ability. CSCs are involved in the development of intrinsic or acquired therapy resistance, and tumor recurrences or relapse. Therefore, the understanding and characterization of epigenetic modifications associated with head and neck carcinogenesis, and the prospective identification of epigenetic markers associated with CSCs, hold the promise for novel therapeutic strategies to fight tumors. In this review, we focus on the current knowledge on epigenetic modifications observed in HNSCC and emerging Epi-drugs capable of sensitizing HNSCC to therapy.
Collapse
|
28
|
Wagner VP, Martins MD, Guimaraes DM, Vasconcelos AC, Meurer L, Vargas PA, Fonseca FP, Squarize CH, Castilho RM. Reduced chromatin acetylation of malignant salivary gland tumors correlates with enhanced proliferation. J Oral Pathol Med 2017; 46:792-797. [DOI: 10.1111/jop.12557] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Vivian Petersen Wagner
- Department of Oral Pathology; School of Dentistry; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
- Department of Experimental Pathology; Hospital de Clínicas de Porto Alegre; Porto Alegre Rio Grande do Sul Brazil
- Laboratory of Epithelial Biology; Department of Periodontics and Oral Medicine; University of Michigan School of Dentistry; Ann Arbor MI USA
| | - Manoela Domingues Martins
- Department of Oral Pathology; School of Dentistry; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
- Department of Experimental Pathology; Hospital de Clínicas de Porto Alegre; Porto Alegre Rio Grande do Sul Brazil
- Laboratory of Epithelial Biology; Department of Periodontics and Oral Medicine; University of Michigan School of Dentistry; Ann Arbor MI USA
| | - Douglas Magno Guimaraes
- Laboratory of Epithelial Biology; Department of Periodontics and Oral Medicine; University of Michigan School of Dentistry; Ann Arbor MI USA
| | - Artur Cunha Vasconcelos
- Department of Oral Pathology; School of Dentistry; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
- Department of Experimental Pathology; Hospital de Clínicas de Porto Alegre; Porto Alegre Rio Grande do Sul Brazil
| | - Luise Meurer
- Department of Experimental Pathology; Hospital de Clínicas de Porto Alegre; Porto Alegre Rio Grande do Sul Brazil
- Department of Oral Medicine; Hospital de Clínicas de Porto Alegre (HCPA/UFRGS); Porto Alegre Rio Grande do Sul RS Brazil
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis; Piracicaba Dental School; University of Campinas; Piracicaba São Paulo Brazil
| | - Felipe Paiva Fonseca
- Department of Clinics, Pathology and Surgery; Dental School; Federal University of Minas Gerais; Belo Horizonte Brazil
| | - Cristiane Helena Squarize
- Laboratory of Epithelial Biology; Department of Periodontics and Oral Medicine; University of Michigan School of Dentistry; Ann Arbor MI USA
| | - Rogerio Moraes Castilho
- Laboratory of Epithelial Biology; Department of Periodontics and Oral Medicine; University of Michigan School of Dentistry; Ann Arbor MI USA
| |
Collapse
|
29
|
Unlocking the chromatin of adenoid cystic carcinomas using HDAC inhibitors sensitize cancer stem cells to cisplatin and induces tumor senescence. Stem Cell Res 2017; 21:94-105. [PMID: 28426972 PMCID: PMC7071815 DOI: 10.1016/j.scr.2017.04.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/14/2017] [Accepted: 04/03/2017] [Indexed: 02/06/2023] Open
Abstract
Adenoid cystic carcinoma (ACC) is an uncommon malignancy of the salivary glands that is characterized by local recurrence and distant metastasis due to its resistance to conventional therapy. Platinum-based therapies have been extensively explored as a treatment for ACC, but they show little effectiveness. Studies have shown that a specific group of tumor cells, harboring characteristics of cancer stem cells (CSCs), are involved in chemoresistance of myeloid leukemias, breast, colorectal and pancreatic carcinomas. Therapeutic strategies that target CSCs improve the survival of patients by decreasing the rates of tumor relapse, and epigenetic drugs, such as histone deacetylase inhibitors (HDACi), have shown promising results in targeting CSCs. In this study, we investigated the effect of the HDACi Suberoylanilide hydroxamic acid (Vorinostat), and cisplatin, alone or in combination, on CSCs and non-CSCs from ACC. We used CSCs as a biological marker for tumor resistance to therapy in patient-derived xenograft (PDX) samples and ACC primary cells. We found that cisplatin reduced tumor viability, but enriched the population of CSCs. Systemic administration of Vorinostat reduced the number of detectable CSCs in vivo and in vitro, and a low dose of Vorinostat decreased tumor cell viability. However, the combination of Vorinostat and cisplatin was extremely effective in depleting CSCs and reducing tumor viability in all ACC primary cells by activating cellular senescence. These observations suggest that HDACi and intercalating agents act more efficiently in combination to destroy tumor cells and their stem cells.
Collapse
|
30
|
Ahn MY, Yoon JH. Histone deacetylase 7 silencing induces apoptosis and autophagy in salivary mucoepidermoid carcinoma cells. J Oral Pathol Med 2017; 46:276-283. [PMID: 28178760 DOI: 10.1111/jop.12560] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The overexpression of histone deacetylases (HDACs) has been observed in many cancers, and inhibition of specific HDACs has emerged as a new target for cancer therapy. We found that HDAC7 expression was selectively reduced by HDAC inhibitor apicidin in salivary mucoepidermoid carcinoma (MEC) cells. Here, we show that HDAC7 suppression has a potent antitumor effect in MEC cells. METHODS Histone deacetylases7 was knocked down using HDAC7 siRNAs, and cell proliferation was quantified. Cell cycle progression, apoptosis, and autophagy were measured by flow cytometry and immunoblotting. RESULTS Histone deacetylases 7 siRNAs inhibited cell proliferation and c-Myc expression, increased p27 expression, and caused G2/M phase cell cycle arrest in both YD-15 and Mc3 cells. HDAC7 silencing increased the sub-G1 population, Annexin V positive apoptotic cells and cleaved caspase3 levels. HDAC7 silencing induced an increase in autophagic markers, number of acidic vesicular organelles, and LC3B II levels, and decrease in p62 levels. HDAC7 siRNAs reduced the activation of ERK. HDAC7 knockdown resulted in growth inhibition through G2/M phase cell cycle arrest and induced both apoptosis and autophagy in MEC cells. CONCLUSIONS This study indicates that inhibition of HDAC7 might become a novel and effective therapeutic approach for treating to MEC.
Collapse
Affiliation(s)
- Mee-Young Ahn
- Division of Bio-industry, Major in Pharmaceutical Engineering, College of Medical and Life Sciences, Silla University, Busan, Korea
| | - Jung-Hoon Yoon
- Department of Oral & Maxillofacial Pathology, Wonkwang Bone Regeneration Research Institute, College of Dentistry, Daejeon Dental Hospital, Wonkwang University, Daejeon, Korea
| |
Collapse
|