1
|
Kijowska J, Grzegorczyk J, Gliwa K, Jędras A, Sitarz M. Epidemiology, Diagnostics, and Therapy of Oral Cancer-Update Review. Cancers (Basel) 2024; 16:3156. [PMID: 39335128 PMCID: PMC11430737 DOI: 10.3390/cancers16183156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Oral cavity and lip cancers are the 16th most common cancer in the world. It is widely known that a lack of public knowledge about precancerous lesions, oral cancer symptoms, and risk factors leads to diagnostic delay and therefore a lower survival rate. Risk factors, which include drinking alcohol, smoking, HPV infection, a pro-inflammatory factor-rich diet, and poor oral hygiene, must be known and avoided by the general population. Regular clinical oral examinations should be enriched in an oral cancer search protocol for the most common symptoms, which are summarized in this review. Moreover, new diagnostic methods, some of which are already available (vital tissue staining, optical imaging, oral cytology, salivary biomarkers, artificial intelligence, colposcopy, and spectroscopy), and newly researched techniques increase the likelihood of stopping the pathological process at a precancerous stage. Well-established oral cancer treatments (surgery, radiotherapy, chemotherapy, and immunotherapy) are continuously being developed using novel technologies, increasing their success rate. Additionally, new techniques are being researched. This review presents a novel glance at oral cancer-its current classification and epidemiology-and will provide new insights into the development of new diagnostic methods and therapies.
Collapse
Affiliation(s)
- Julia Kijowska
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, ul. Chodźki 6, 20-093 Lublin, Poland
| | - Julia Grzegorczyk
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, ul. Chodźki 6, 20-093 Lublin, Poland
| | - Katarzyna Gliwa
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, ul. Chodźki 6, 20-093 Lublin, Poland
| | - Aleksandra Jędras
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, ul. Chodźki 6, 20-093 Lublin, Poland
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, ul. Chodźki 6, 20-093 Lublin, Poland
| |
Collapse
|
2
|
Muniz IDAF, Araujo M, Bouassaly J, Farshadi F, Atique M, Esfahani K, Bonan PRF, Hier M, Mascarella M, Mlynarek A, Alaoui-Jamali M, da Silva SD. Therapeutic Advances and Challenges for the Management of HPV-Associated Oropharyngeal Cancer. Int J Mol Sci 2024; 25:4009. [PMID: 38612819 PMCID: PMC11012756 DOI: 10.3390/ijms25074009] [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: 02/08/2024] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The use of conventional chemotherapy in conjunction with targeted and immunotherapy drugs has emerged as an option to limit the severity of side effects in patients diagnosed with head and neck cancer (HNC), particularly oropharyngeal cancer (OPC). OPC prevalence has increased exponentially in the past 30 years due to the prevalence of human papillomavirus (HPV) infection. This study reports a comprehensive review of clinical trials registered in public databases and reported in the literature (PubMed/Medline, Scopus, and ISI web of science databases). Of the 55 clinical trials identified, the majority (83.3%) were conducted after 2015, of which 77.7% were performed in the United States alone. Eight drugs have been approved by the FDA for HNC, including both generic and commercial forms: bleomycin sulfate, cetuximab (Erbitux), docetaxel (Taxotere), hydroxyurea (Hydrea), pembrolizumab (Keytruda), loqtorzi (Toripalimab-tpzi), methotrexate sodium (Trexall), and nivolumab (Opdivo). The most common drugs to treat HPV-associated OPC under these clinical trials and implemented as well for HPV-negative HNC include cisplatin, nivolumab, cetuximab, paclitaxel, pembrolizumab, 5-fluorouracil, and docetaxel. Few studies have highlighted the necessity for new drugs specifically tailored to patients with HPV-associated OPC, where molecular mechanisms and clinical prognosis are distinct from HPV-negative tumors. In this context, we identified most mutated genes found in HPV-associated OPC that can represent potential targets for drug development. These include TP53, PIK3CA, PTEN, NOTCH1, RB1, FAT1, FBXW7, HRAS, KRAS, and CDKN2A.
Collapse
Affiliation(s)
- Isis de Araújo Ferreira Muniz
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
- Graduate Program in Dentistry, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Megan Araujo
- Division of Experimental Medicine and Oncology, Department of Medicine and Health Sciences, McGill University, Montreal, QC HC3 1E2, Canada; (M.A.); (J.B.)
| | - Jenna Bouassaly
- Division of Experimental Medicine and Oncology, Department of Medicine and Health Sciences, McGill University, Montreal, QC HC3 1E2, Canada; (M.A.); (J.B.)
| | - Fatemeh Farshadi
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
- Division of Experimental Medicine and Oncology, Department of Medicine and Health Sciences, McGill University, Montreal, QC HC3 1E2, Canada; (M.A.); (J.B.)
| | - Mai Atique
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
| | - Khashayar Esfahani
- Department of Oncology, McGill University, Montreal, QC HC3 1E2, Canada;
| | - Paulo Rogerio Ferreti Bonan
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
- Graduate Program in Dentistry, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Michael Hier
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
| | - Marco Mascarella
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
| | - Alex Mlynarek
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
| | - Moulay Alaoui-Jamali
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
| | - Sabrina Daniela da Silva
- Department of Otolaryngology and Head and Neck Surgery, McGill University, Montreal, QC HC3 1E2, Canada; (I.d.A.F.M.); (F.F.); (M.A.); (P.R.F.B.); (M.H.); (M.M.); (A.M.); (M.A.-J.)
- Division of Experimental Medicine and Oncology, Department of Medicine and Health Sciences, McGill University, Montreal, QC HC3 1E2, Canada; (M.A.); (J.B.)
| |
Collapse
|
3
|
Peña-Oyarzún D, Flores T, Torres VA, Quest AFG, Lobos-González L, Kretschmar C, Contreras P, Maturana-Ramírez A, Criollo A, Reyes M. Inhibition of PORCN Blocks Wnt Signaling to Attenuate Progression of Oral Carcinogenesis. Clin Cancer Res 2024; 30:209-223. [PMID: 37812478 DOI: 10.1158/1078-0432.ccr-23-0318] [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: 02/01/2023] [Revised: 05/12/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE Oral squamous cell carcinoma (OSCC) is commonly preceded by potentially malignant lesions, referred to as oral dysplasia. We recently reported that oral dysplasia is associated with aberrant activation of the Wnt/β-catenin pathway, due to overexpression of Wnt ligands in a Porcupine (PORCN)-dependent manner. Pharmacologic inhibition of PORCN precludes Wnt secretion and has been proposed as a potential therapeutic approach to treat established cancers. Nevertheless, there are no studies that explore the effects of PORCN inhibition at the different stages of oral carcinogenesis. EXPERIMENTAL DESIGN We performed a model of tobacco-induced oral cancer in vitro, where dysplastic oral keratinocytes (DOK) were transformed into oral carcinoma cells (DOK-TC), and assessed the effects of inhibiting PORCN with the C59 inhibitor. Similarly, an in vivo model of oral carcinogenesis and ex vivo samples derived from patients diagnosed with oral dysplasia and OSCC were treated with C59. RESULTS Both in vitro and ex vivo oral carcinogenesis approaches revealed decreased levels of nuclear β-catenin and Wnt3a, as observed by immunofluorescence and IHC analyses. Consistently, reduced protein and mRNA levels of survivin were observed after treatment with C59. Functionally, treatment with C59 in vitro resulted in diminished cell migration, viability, and invasion. Finally, by using an in vivo model of oral carcinogenesis, we found that treatment with C59 prevented the development of OSCC by reducing the size and number of oral tumor lesions. CONCLUSIONS The inhibition of Wnt ligand secretion with C59 represents a feasible treatment to prevent the progression of early oral lesions toward OSCC.
Collapse
Affiliation(s)
- Daniel Peña-Oyarzún
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Interdisciplinary Center for Research in Territorial Health of the Aconcagua Valley (CIISTe Aconcagua), School of Medicine, Faculty of Medicine, San Felipe Campus, Universidad de Valparaiso, Chile
| | - Tania Flores
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Research Centre in Dental Science (CICO), Faculty of Dentistry, Universidad de La Frontera, Temuco, Chile
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular Communication, Center for studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Catalina Kretschmar
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Pamela Contreras
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular Communication, Center for studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrea Maturana-Ramírez
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Montserrat Reyes
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
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
|
de Kort WWB, de Ruiter EJ, Haakma WE, Driehuis E, Devriese LA, van Es RJJ, Willems SM. p-mTOR, p-ERK and PTEN Expression in Tumor Biopsies and Organoids as Predictive Biomarkers for Patients with HPV Negative Head and Neck Cancer. Head Neck Pathol 2023; 17:697-707. [PMID: 37486536 PMCID: PMC10514008 DOI: 10.1007/s12105-023-01576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Survival rates of head and neck squamous cell carcinoma (HNSCC) have only marginally improved in the last decades. Hence there is a need for predictive biomarkers for long-time survival that can help to guide treatment decisions and might lead to the development of new therapies. The phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR signaling pathway is the most frequently altered pathway in HNSCC, genes are often mutated, amplificated and overexpressed causing aberrant signaling affecting cell growth and differentiation. Numerous genetic alterations of upstream and downstream factors have currently been clarified. However, their predictive value has yet to be established. Therefore we assess the predictive value of p-mTOR, p-ERK and PTEN expression. METHODS Tissue microarrays (TMA's) of HPV-negative patients with oropharyngeal (n = 48), hypopharyngeal (n = 16) or laryngeal (n = 13) SCC, treated with primary chemoradiation (cisplatin/carboplatin/cetuximab and radiotherapy), were histologically stained for p-mTOR, PTEN and p-ERK. Expression was correlated to overall survival (OS), disease free survival (DFS) and locoregional control (LRC). Also p-mTOR was histologically stained in a separate cohort of HNSCC organoids (n = 8) and correlated to mTOR-inhibitor everolimus response. RESULTS High p-mTOR expression correlated significantly with worse OS in multivariate analysis in the whole patient cohort [Hazar Ratio (HR) 1.06, 95%CI 1.01-1.11, p = 0.03] and in the cisplatin/carboplatin group with both worse OS (HR 1.09, 95%CI 1.02-1.16, p = 0.02) and DFS (HR 1.06, 95%CI 1.00-1.12, p = 0,04). p-ERK expression correlated significantly with DFS in univariate analysis in the whole patient cohort (HR 1.03, 95%CI 1.00-1.05, p = 0.04) and cisplatin/carboplatin group (HR 1.03, 95%CI 1.00-1.07, p = 0.04). PTEN-expression did not correlate with OS/DFS/LRC. Better organoid response to everolimus correlated significantly to higher p-mTOR expression (Rs = - 0.731, p = 0.04). CONCLUSIONS High p-mTOR expression predicts and high p-ERK expression tends to predict worse treatment outcome in HPV negative HNSCC patients treated with chemoradiation, providing additional evidence that these markers are candidate prognostic biomarkers for survival in this patient population. Also this study shows that the use of HNSCC organoids for biomarker research has potential. The role of PTEN expression as prognostic biomarker remains unclear, as consistent evidence on its prognostic and predictive value is lacking.
Collapse
Affiliation(s)
- W. W. B. de Kort
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - E. J. de Ruiter
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - W. E. Haakma
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - E. Driehuis
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - L. A. Devriese
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R. J. J. van Es
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Head and Neck Surgical Oncology, Utrecht Cancer Center, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - S. M. Willems
- Department of Pathology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
6
|
Kleszcz R. Advantages of the Combinatorial Molecular Targeted Therapy of Head and Neck Cancer-A Step before Anakoinosis-Based Personalized Treatment. Cancers (Basel) 2023; 15:4247. [PMID: 37686523 PMCID: PMC10486994 DOI: 10.3390/cancers15174247] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The molecular initiators of Head and Heck Squamous Cell Carcinoma (HNSCC) are complex. Human Papillomavirus (HPV) infection is linked to an increasing number of HNSCC cases, but HPV-positive tumors generally have a good prognosis. External factors that promote the development of HPV-negative HNSCC include tobacco use, excessive alcohol consumption, and proinflammatory poor oral hygiene. On a molecular level, several events, including the well-known overexpression of epidermal growth factor receptors (EGFR) and related downstream signaling pathways, contribute to the development of HNSCC. Conventional chemotherapy is insufficient for many patients. Thus, molecular-based therapy for HNSCC offers patients a better chance at a cure. The first molecular target for therapy of HNSCC was EGFR, inhibited by monoclonal antibody cetuximab, but its use in monotherapy is insufficient and induces resistance. This article describes attempts at combinatorial molecular targeted therapy of HNSCC based on several molecular targets and exemplary drugs/drug candidates. The new concept of anakoinosis-based therapy, which means treatment that targets the intercellular and intracellular communication of cancer cells, is thought to be the way to improve the clinical outcome for HNSCC patients. The identification of a link between molecular targeted therapy and anakoinosis raises the potential for further progress in HPV-negative HNSCC therapy.
Collapse
Affiliation(s)
- Robert Kleszcz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, 4, Święcickiego Str., 60-781 Poznan, Poland
| |
Collapse
|
7
|
Wang J, Cui B, Chen Z, Ding X. The regulation of skin homeostasis, repair and the pathogenesis of skin diseases by spatiotemporal activation of epidermal mTOR signaling. Front Cell Dev Biol 2022; 10:950973. [PMID: 35938153 PMCID: PMC9355246 DOI: 10.3389/fcell.2022.950973] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
The epidermis, the outmost layer of the skin, is a stratified squamous epithelium that protects the body from the external world. The epidermis and its appendages need constantly renew themselves and replace the damaged tissues caused by environmental assaults. The mechanistic target of rapamycin (mTOR) signaling is a central controller of cell growth and metabolism that plays a critical role in development, homeostasis and diseases. Recent findings suggest that mTOR signaling is activated in a spatiotemporal and context-dependent manner in the epidermis, coordinating diverse skin homeostatic processes. Dysregulation of mTOR signaling underlies the pathogenesis of skin diseases, including psoriasis and skin cancer. In this review, we discuss the role of epidermal mTOR signaling activity and function in skin, with a focus on skin barrier formation, hair regeneration, wound repair, as well as skin pathological disorders. We propose that fine-tuned control of mTOR signaling is essential for epidermal structural and functional integrity.
Collapse
Affiliation(s)
- Juan Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Baiping Cui
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Zhongjian Chen
- School of Medicine, Shanghai University, Shanghai, China
- Shanghai Engineering Research Center for External Chinese Medicine, Shanghai, China
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaolei Ding
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
- *Correspondence: Xiaolei Ding,
| |
Collapse
|
8
|
p-S6 as a Prognostic Biomarker in Canine Oral Squamous Cell Carcinoma. Biomolecules 2022; 12:biom12070935. [PMID: 35883491 PMCID: PMC9313205 DOI: 10.3390/biom12070935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 12/03/2022] Open
Abstract
Scarce information exists on the role of mTOR pathway proteins and their association to aggressiveness and prognosis of patients with canine oral cancers. We aimed to investigate the activated form of mTOR and its downstream S6 protein in canine oral squamous cell carcinoma (OSCC), and to evaluate potential associations between protein expression and clinic-pathologic variables and survival. For that we analysed p-mTOR and p-S6 protein expression by immunohistochemistry in 61 canine OSCCs. Multivariate analysis was conducted to examine their role in patients’ cancer-specific survival (CSS). p-mTOR and p-S6 expression were present in almost all cases. High-expression of p-mTOR was observed in 44 (72.1%) cases using extent score and 52 (85.2%) cases using intensity score. For p-S6, high expression was observed in 53 (86.9%) cases using extent score and in 54 (88.5%) cases using intensity score. An independent prognostic value for p-S6 extension (p = 0.027), tumour stage (p = 0.013) and treatment (p = 0.0009) was found in patients’ CSS analysis. Our data suggest that p-mTOR and p-S6 proteins are commonly expressed in canine OSCC and p-S6 expression is correlated with poor CSS in dogs with OSCC. More studies should be performed to identify possible therapeutic targets related with mTOR pathway for these patients.
Collapse
|
9
|
Gilbert M, Liang E, Li P, Salgia R, Abouljoud M, Siddiqui F. Outcomes of Primary Mucosal Head and Neck Squamous Cell Carcinoma in Solid Organ Transplant Recipients. Cureus 2022; 14:e24305. [PMID: 35607526 PMCID: PMC9123412 DOI: 10.7759/cureus.24305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Patients who undergo solid organ transplants have a higher risk of developing malignancies and subsequent recurrences. Clinical outcomes in transplant recipients with primary mucosal head and neck squamous cell carcinoma (HNSCC) are not well described in the published literature. Therefore, we retrospectively studied the outcomes in this group of patients. Methods This Institutional Review Board (IRB)-approved analysis included patients who had previously undergone solid organ transplants and subsequently were diagnosed with primary mucosal HNSCC between 2006 and 2021. Our institutional database of solid organ transplant recipients was cross-referenced with our head and neck cancer database to identify the patients included in this cohort. In addition, Kaplan-Meier analyses were performed to calculate overall and disease-free survival. Results Of 1,221 patients, 20 met the inclusion criteria. The median time from organ transplant to HNSCC diagnosis was 5.9 years (range: 0.5-18.5 years). A total of 11 (55.0%) and 9 (45.0%) patients presented with localized and locally advanced disease, respectively. Two-year overall and disease-free survivals were 59.1% and 73.5%, respectively. After initial treatment, six (30.0%) patients experienced a recurrence. All patients who developed a recurrence died within the follow-up period. The median time of death after recurrence for all six patients was 11.5 months (range: 2-22 months). Conclusion This series highlights a high mortality rate following recurrence among patients with primary mucosal HNSCC and a solid organ transplant history. A better understanding of how solid organ transplant history adversely impacts the course of HNSCC could help properly guide treatment, follow-up, and survivorship decisions.
Collapse
Affiliation(s)
| | - Evan Liang
- Radiation Oncology, Henry Ford Health, Detroit, USA
| | - Pin Li
- Public Health Sciences, Henry Ford Health, Detroit, USA
| | - Reena Salgia
- Gastroenterology and Hepatology, Henry Ford Health, Detroit, USA
| | - Marwan Abouljoud
- Transplant and Hepatobiliary Surgery, Henry Ford Health, Detroit, USA
| | | |
Collapse
|
10
|
Sunavala-Dossabhoy G. Disorder at the Start: The Contribution of Dysregulated Translation Initiation to Cancer Therapy Resistance. FRONTIERS IN ORAL HEALTH 2022; 2:765931. [PMID: 35048066 PMCID: PMC8757695 DOI: 10.3389/froh.2021.765931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/02/2021] [Indexed: 11/26/2022] Open
Abstract
Translation of cellular RNA to protein is an energy-intensive process through which synthesized proteins dictate cellular processes and function. Translation is regulated in response to extracellular effectors and availability of amino acids intracellularly. Most eukaryotic mRNA rely on the methyl 7-guanosine (m7G) nucleotide cap to recruit the translation machinery, and the uncoupling of translational control that occurs in tumorigenesis plays a significant role in cancer treatment response. This article provides an overview of the mammalian translation initiation process and the primary mechanisms by which it is regulated. An outline of how deregulation of initiation supports tumorigenesis and how initiation at a downstream open reading frame (ORF) of Tousled-like kinase 1 (TLK1) leads to treatment resistance is discussed.
Collapse
Affiliation(s)
- Gulshan Sunavala-Dossabhoy
- Department of Biochemistry and Molecular Biology, Louisiana State University Health and Feist Weiller Cancer Center, Shreveport, LA, United States
| |
Collapse
|
11
|
Khalil BD, Sanchez R, Rahman T, Rodriguez-Tirado C, Moritsch S, Martinez AR, Miles B, Farias E, Mezei M, Nobre AR, Singh D, Kale N, Sproll KC, Sosa MS, Aguirre-Ghiso JA. An NR2F1-specific agonist suppresses metastasis by inducing cancer cell dormancy. J Exp Med 2022; 219:e20210836. [PMID: 34812843 PMCID: PMC8614154 DOI: 10.1084/jem.20210836] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 09/20/2021] [Accepted: 10/26/2021] [Indexed: 01/02/2023] Open
Abstract
We describe the discovery of an agonist of the nuclear receptor NR2F1 that specifically activates dormancy programs in malignant cells. The agonist led to a self-regulated increase in NR2F1 mRNA and protein and downstream transcription of a novel dormancy program. This program led to growth arrest of an HNSCC PDX line, human cell lines, and patient-derived organoids in 3D cultures and in vivo. This effect was lost when NR2F1 was knocked out by CRISPR-Cas9. RNA sequencing revealed that agonist treatment induces transcriptional changes associated with inhibition of cell cycle progression and mTOR signaling, metastasis suppression, and induction of a neural crest lineage program. In mice, agonist treatment resulted in inhibition of lung HNSCC metastasis, even after cessation of the treatment, where disseminated tumor cells displayed an NR2F1hi/p27hi/Ki-67lo/p-S6lo phenotype and remained in a dormant single-cell state. Our work provides proof of principle supporting the use of NR2F1 agonists to induce dormancy as a therapeutic strategy to prevent metastasis.
Collapse
Affiliation(s)
- Bassem D. Khalil
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Western Atlantic University School of Medicine, Plantation, FL
| | - Roberto Sanchez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tasrina Rahman
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Stefan Moritsch
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alba Rodriguez Martinez
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Brett Miles
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eduardo Farias
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mihaly Mezei
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ana Rita Nobre
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Deepak Singh
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nupura Kale
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Karl Christoph Sproll
- Department of Oral, Maxillofacial and Plastic Facial Surgery, Medical Faculty, University Hospital of the Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Maria Soledad Sosa
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Julio A. Aguirre-Ghiso
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
12
|
Liu Z, Liu H, Dong Q, Li H, Zhang B, Liu Y, Zhong L, Tang H. Prognostic role of DFNA5 in head and neck squamous cell carcinoma revealed by systematic expression analysis. BMC Cancer 2021; 21:951. [PMID: 34433433 PMCID: PMC8390204 DOI: 10.1186/s12885-021-08692-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023] Open
Abstract
Background The gasdermin E gene (GSDME, also known as DFNA5) is mutated in familial aging-related hearing loss. Recent studies have also revealed that the expression of DFNA5 is suppressed in many cancer types; however, little is known about the function of DFNA5 in head and neck squamous cell carcinoma (HNSCC). Accordingly, the aim of the present study was to evaluate the expression of DFNA5 and explore its prognostic value in HNSCC. Result We used a set of bioinformatics tools, including Oncomine, TIMER, TISIDB, cBioPortal, and GEPIA, to analyze the expression of DFNA5 in patients with HNSCC from public databases. Kaplan-Meier plotter was used to evaluate the potential prognostic significance of DFNA5. DFNA5 mRNA levels were significantly higher in HNSCC tissues than in normal tissues, and high DFNA5 expression was correlated with worse survival. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses showed that DFNA5 expression has a strong positive correlation with cell adhesion and the integrin signaling pathway, whereas its expression was negatively correlated with the levels of infiltrating B cells (cor = − 0.223, P = 8.57e-07) and CD8 T cells (cor = − 0.223, P = 2.99e-07). Conclusion This study demonstrates that DFNA5 expression has prognostic value for HNSCC patients. Moreover, these results suggest that regulation of lymphocyte infiltration is the mechanism underlying the function of DFNA5 in HNSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08692-w.
Collapse
Affiliation(s)
- Zhiguo Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055, China.,Guangdong Province Key Laboratory of Stomatology, Guangzhou, China
| | - Hongyan Liu
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, China.,Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Sun Yat-Sen University, 56 Lingyuanxi Road, Guangzhou, 510055, China
| | - Qian Dong
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055, China.,Guangdong Province Key Laboratory of Stomatology, Guangzhou, China
| | - Hongyu Li
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055, China.,Guangdong Province Key Laboratory of Stomatology, Guangzhou, China
| | - Bin Zhang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055, China.,Guangdong Province Key Laboratory of Stomatology, Guangzhou, China
| | - Yufeng Liu
- Guangzhou First People's Hospital, Guangzhou, 510000, China
| | - Limei Zhong
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou, 510317, China.
| | - Haikuo Tang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055, China. .,Guangdong Province Key Laboratory of Stomatology, Guangzhou, China.
| |
Collapse
|
13
|
Dankó T, Petővári G, Sztankovics D, Moldvai D, Raffay R, Lőrincz P, Visnovitz T, Zsiros V, Barna G, Márk Á, Krencz I, Sebestyén A. Rapamycin Plus Doxycycline Combination Affects Growth Arrest and Selective Autophagy-Dependent Cell Death in Breast Cancer Cells. Int J Mol Sci 2021; 22:ijms22158019. [PMID: 34360785 PMCID: PMC8347279 DOI: 10.3390/ijms22158019] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/22/2022] Open
Abstract
Metabolic alteration is characteristic during tumour growth and therapy; however, targeting metabolic rewiring could overcome therapy resistance. mTOR hyperactivity, autophagy and other metabolic processes, including mitochondrial functions, could be targeted in breast cancer progression. We investigated the growth inhibitory mechanism of rapamycin + doxycycline treatment in human breast cancer model systems. Cell cycle and cell viability, including apoptotic and necrotic cell death, were analysed using flow cytometry, caspase activity measurements and caspase-3 immunostainings. mTOR-, autophagy-, necroptosis-related proteins and treatment-induced morphological alterations were analysed by WesTM, Western blot, immunostainings and transmission electron microscopy. The rapamycin + doxycycline combination decreased tumour proliferation in about 2/3rd of the investigated cell lines. The continuous treatment reduced tumour growth significantly both in vivo and in vitro. The effect after short-term treatment was reversible; however, autophagic vacuoles and degrading mitochondria were detected simultaneously, and the presence of mitophagy was also observed after the long-term rapamycin + doxycycline combination treatment. The rapamycin + doxycycline combination did not cause apoptosis or necrosis/necroptosis, but the alterations in autophagy- and mitochondria-related protein levels (LC3-B-II/I, p62, MitoTracker, TOM20 and certain co-stainings) were correlated to autophagy induction and mitophagy, without mitochondria repopulation. Based on these results, we suggest considering inducing metabolic stress and targeting mTOR hyperactivity and mitochondrial functions in combined anti-cancer treatments.
Collapse
Affiliation(s)
- Titanilla Dankó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Gábor Petővári
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Dániel Sztankovics
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Dorottya Moldvai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Regina Raffay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Péter Lőrincz
- Department of Anatomy, Cell and Developmental Biology, Eotvos Lorand University, Pázmány Péter sétány 1/c, H-1117 Budapest, Hungary;
| | - Tamás Visnovitz
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Viktória Zsiros
- Department of Anatomy, Histology and Embryology, Semmelweis University, Tűzoltó utca 58, H-1094 Budapest, Hungary;
| | - Gábor Barna
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Ágnes Márk
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Ildikó Krencz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (T.D.); (G.P.); (D.S.); (D.M.); (R.R.); (G.B.); (Á.M.); (I.K.)
- Correspondence:
| |
Collapse
|
14
|
Hou F, Liu Y, Cheng Y, Zhang N, Yan W, Zhang F. Exploring the Mechanism of Scutellaria baicalensis Georgi Efficacy against Oral Squamous Cell Carcinoma Based on Network Pharmacology and Molecular Docking Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5597586. [PMID: 34335829 PMCID: PMC8292061 DOI: 10.1155/2021/5597586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Scutellaria baicalensis Georgi (SBG) has been widely shown to induce apoptosis and inhibit invasion and migration of various cancer cells. Increased evidence shows that SBG may be useful to treat oral squamous cell carcinoma (OSCC). However, the biological activity and possible mechanisms of SBG in the treatment of OSCC have not been fully elucidated. This study aimed to clarify the bioactive component and multitarget mechanisms of SBG against OSCC using network pharmacology and molecular docking. METHODS Traditional Chinese Medicine Systems Pharmacology (TCMSP) database was used to predict the active components in SBG, and putative molecular targets of SBG were identified using the Swiss Target Prediction database. OSCC-related targets were screened by GeneCards, Online Mendelian Inheritance in Man (OMIM), and Therapeutic Target Database (TTD). Then, we established protein-protein interaction (PPI), compound-target-disease (C-T-D), and compound-target-pathway (C-T-P) networks by Cytoscape to identify the main components, core targets, and pharmacological pathways of SBG against OSCC via applying data mining techniques and topological parameters. Metascape database was utilized for Gene Ontology (GO) and pathway enrichment analysis. The potential interaction of the main components with core targets was revealed by molecular docking simulation, and for the correlation between core targets and OSCC prognosis analysis, the Kaplan-Meier Plotter online database was used. RESULTS There were 25 active compounds in SBG and 86 genes targeted by OSCC. A total of 141 signaling pathways were identified, and it was found that the PI3K-Akt signaling pathway may occupy core status in the anti-OSCC system. GO analysis revealed that the primary biological processes were related to apoptosis, proliferation, and migration. Molecular docking results confirmed that core targets of OSCC had a high affinity with the main compounds of SBG. CONCLUSION Our study demonstrated multicomponent, multitarget, and multipathway characteristics of SBG in the treatment of OSCC and provided a foundation for further drug development research.
Collapse
Affiliation(s)
- Fanfan Hou
- Stomatology Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Yang Liu
- Stomatology Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - YaHsin Cheng
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Ni Zhang
- Stomatology Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Wenpeng Yan
- Stomatology Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Fang Zhang
- Stomatology Hospital, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
15
|
Usman S, Jamal A, Teh MT, Waseem A. Major Molecular Signaling Pathways in Oral Cancer Associated With Therapeutic Resistance. FRONTIERS IN ORAL HEALTH 2021; 1:603160. [PMID: 35047986 PMCID: PMC8757854 DOI: 10.3389/froh.2020.603160] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Oral cancer is a sub-category of head and neck cancers that primarily initiates in the oral cavity. The primary treatment option for oral cancer remains surgery but it is associated with massive disfigurement, inability to carry out normal oral functions, psycho-social stress and exhaustive rehabilitation. Other treatment options such as chemotherapy and radiotherapy have their own limitations in terms of toxicity, intolerance and therapeutic resistance. Immunological treatments to enhance the body's ability to recognize cancer tissue as a foreign entity are also being used but they are new and underdeveloped. Although substantial progress has been made in the treatment of oral cancer, its complex heterogeneous nature still needs to be explored, to elucidate the molecular basis for developing resistance to therapeutic agents and how to overcome it, with the aim of improving the chances of patients' survival and their quality of life. This review provides an overview of up-to-date information on the complex role of the major molecules and associated signaling, epigenetic changes, DNA damage repair systems, cancer stem cells and micro RNAs in the development of therapeutic resistance and treatment failure in oral cancer. We have also summarized the current strategies being developed to overcome these therapeutic challenges. This review will help not only researchers but also oral oncologists in the management of the disease and in developing new therapeutic modalities.
Collapse
Affiliation(s)
| | | | | | - Ahmad Waseem
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|