1
|
Xu Q, Huang S, Yang K. Combination immunochemotherapy for recurrent or metastatic head and neck squamous cell carcinoma: a systematic review and meta-analysis. BMJ Open 2023; 13:e069047. [PMID: 37311638 DOI: 10.1136/bmjopen-2022-069047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2023] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of combination immunochemotherapy regimens for the treatment of recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC). DESIGN Meta-analysis and systematic review. DATA SOURCES PubMed, Embase, Web of Science and Cochrane library and the Clinicaltrials.gov clinical trials registry were searched up to 14 March 2022. ELIGIBILITY CRITERIA FOR SELECTING STUDIES We included randomised controlled trials that compared combination immunochemotherapy with conventional chemotherapy for R/M HNSCC. Primary outcomes of interest were overall survival (OS), progression-free survival (PFS), objective response rate (ORR) and adverse effects (AEs). DATA EXTRACTION AND SYNTHESIS Two reviewers independently extracted data and assessed the risk of bias of the included studies. The HR and its 95% CI were used as the effect analysis statistic for survival analysis, while the OR and its 95% CI were used as the effect analysis statistic for dichotomous variables. These statistics were extracted by the reviewers and aggregated using a fixed-effects model to synthesise the data. RESULTS A total of 1214 relevant papers were obtained after the initial search, and five papers that met the inclusion criteria were included; these studies included a total of 1856 patients with R/M HNSCC. Meta-analysis showed that the OS and PFS of patients with R/M HNSCC in the combination immunochemotherapy group were significantly longer than those in the conventional chemotherapy group (HR=0.84; 95% CI 0.76, 0.94; p=0.002; HR=0.67; 95% CI 0.61, 0.75; p<0.0001), and the ORR was significantly higher (OR=1.90; 95% CI 1.54, 2.34; p<0.00001). The analysis of AEs showed that there was no significant difference in the overall incidence rate of AEs between two groups (OR=0.80; 95% CI 0.18, 3.58; p=0.77), but the rate of grade III and IV AEs was significantly higher in patients in the combination immunochemotherapy group (OR=1.39; 95% CI 1.12, 1.73; p=0.003). CONCLUSIONS Combination immunochemotherapy prolonged OS and PFS in patients with R/M HNSCC and improved the ORR; while this approach did not increase the overall incidence of AEs in patients, it increased the rate of grade III and IV AEs. PROSPERO REGISTRATION NUMBER CRD42022344166.
Collapse
Affiliation(s)
- Qiudong Xu
- Department of Oral and Maxillofacial Surgery, Wuxi Stomatology Hospital, Wuxi, China
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuang Huang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Yang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Prieto-Fernández L, Montoro-Jiménez I, de Luxan-Delgado B, Otero-Rosales M, Rodrigo JP, Calvo F, García-Pedrero JM, Álvarez-Teijeiro S. Dissecting the functions of cancer-associated fibroblasts to therapeutically target head and neck cancer microenvironment. Biomed Pharmacother 2023; 161:114502. [PMID: 37002578 DOI: 10.1016/j.biopha.2023.114502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/22/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Head and neck cancers (HNC) are a diverse group of aggressive malignancies with high morbidity and mortality, leading to almost half-million deaths annually worldwide. A better understanding of the molecular processes governing tumor formation and progression is crucial to improve current diagnostic and prognostic tools as well as to develop more personalized treatment strategies. Tumors are highly complex and heterogeneous structures in which growth and dissemination is not only governed by the cancer cells intrinsic mechanisms, but also by the surrounding tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) emerge as predominant TME components and key players in the generation of permissive conditions that ultimately impact in tumor progression and metastatic dissemination. Although CAFs were initially considered a consequence of tumor development, it is now well established that they actively contribute to numerous cancer hallmarks i.e., tumor cell growth, migration and invasion, cancer cell stemness, angiogenesis, metabolic reprograming, inflammation, and immune system modulation. In this scenario, therapeutic strategies targeting CAF functions could potentially have a major impact in cancer therapeutics, providing avenues for new treatment options or for improving efficacy in established approaches. This review is focused on thoroughly dissecting existing evidences supporting the contribution of CAFs in HNC biology with an emphasis on current knowledge of the key molecules and pathways involved in CAF-tumor crosstalk, and their potential as novel biomarkers and/or therapeutic targets to effectively interfere the tumor-stroma crosstalk for HNC patients benefit. involved in CAF-tumor crosstalk, and their potential as novel biomarkers and/or therapeutic targets to effec- tively interfere the tumor-stroma crosstalk for HNC patients benefit.
Collapse
Affiliation(s)
- Llara Prieto-Fernández
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Montoro-Jiménez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz de Luxan-Delgado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - María Otero-Rosales
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Abstract
EGFR is a member of the ERBB family. It plays a significant role in cellular processes such as growth, survival and differentiation via the activation of various signaling pathways. EGFR deregulation is implicated in various human malignancies, and therefore EGFR has emerged as an attractive anticancer target. EGFR inhibition using strategies such as tyrosine kinase inhibitors and monoclonal antibodies hinders cellular proliferation and promotes apoptosis in cancer cells in vitro and in vivo. EGFR inhibition by tyrosine kinase inhibitors has been shown to be a better treatment option than chemotherapy for advanced-stage EGFR-driven non-small-cell lung cancer, yet de novo and acquired resistance limits the clinical benefit of these therapeutic molecules. This review discusses the cellular signaling pathways activated by EGFR. Further, current therapeutic strategies to target aberrant EGFR signaling in cancer and mechanisms of resistance to them are highlighted.
Collapse
|
4
|
Goel B, Tiwari AK, Pandey RK, Singh AP, Kumar S, Sinha A, Jain SK, Khattri A. Therapeutic approaches for the treatment of head and neck squamous cell carcinoma-An update on clinical trials. Transl Oncol 2022; 21:101426. [PMID: 35460943 PMCID: PMC9046875 DOI: 10.1016/j.tranon.2022.101426] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common non-skin cancer with a tobacco consumption and infection with high-risk human papillomavirus (HPV) being major risk factors. Despite advances in numerous therapy modalities, survival rates for HNSCC have not improved considerably; a vast number of clinical outcomes have demonstrated that a combination strategy (the most well-known docetaxel, cisplatin, and 5-fluorouracil) is the most effective treatment choice. Immunotherapy that targets immunological checkpoints is being tested in a number of clinical trials, either alone or in conjunction with chemotherapeutic or targeted therapeutic drugs. Various monoclonal antibodies, such as cetuximab and bevacizumab, which target the EGFR and VEGFR, respectively, as well as other signaling pathway inhibitors, such as temsirolimus and rapamycin, are also being studied for the treatment of HNSCC. We have reviewed the primary targets in active clinical studies in this study, with a particular focus on the medications and drug targets used.
Collapse
Affiliation(s)
- Bharat Goel
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Anoop Kumar Tiwari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Rajeev Kumar Pandey
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Akhand Pratap Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Sujeet Kumar
- Centre for Proteomics and Drug Discovery, Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai - 410206, Maharashtra, India
| | - Abhishek Sinha
- Department of Oral Medicine & Radiology, Sardar Patel Post Graduate Institute of Dental & Medical Sciences, Lucknow - 226025, Uttar Pradesh, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Arun Khattri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India.
| |
Collapse
|
5
|
Gomes INF, da Silva-Oliveira RJ, da Silva LS, Martinho O, Evangelista AF, van Helvoort Lengert A, Leal LF, Silva VAO, dos Santos SP, Nascimento FC, Lopes Carvalho A, Reis RM. Comprehensive Molecular Landscape of Cetuximab Resistance in Head and Neck Cancer Cell Lines. Cells 2022; 11:154. [PMID: 35011716 PMCID: PMC8750399 DOI: 10.3390/cells11010154] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/17/2021] [Accepted: 12/31/2021] [Indexed: 12/15/2022] Open
Abstract
Cetuximab is the sole anti-EGFR monoclonal antibody that is FDA approved to treat head and neck squamous cell carcinoma (HNSCC). However, no predictive biomarkers of cetuximab response are known for HNSCC. Herein, we address the molecular mechanisms underlying cetuximab resistance in an in vitro model. We established a cetuximab resistant model (FaDu), using increased cetuximab concentrations for more than eight months. The resistance and parental cells were evaluated for cell viability and functional assays. Protein expression was analyzed by Western blot and human cell surface panel by lyoplate. The mutational profile and copy number alterations (CNA) were analyzed using whole-exome sequencing (WES) and the NanoString platform. FaDu resistant clones exhibited at least two-fold higher IC50 compared to the parental cell line. WES showed relevant mutations in several cancer-related genes, and the comparative mRNA expression analysis showed 36 differentially expressed genes associated with EGFR tyrosine kinase inhibitors resistance, RAS, MAPK, and mTOR signaling. Importantly, we observed that overexpression of KRAS, RhoA, and CD44 was associated with cetuximab resistance. Protein analysis revealed EGFR phosphorylation inhibition and mTOR increase in resistant cells. Moreover, the resistant cell line demonstrated an aggressive phenotype with a significant increase in adhesion, the number of colonies, and migration rates. Overall, we identified several molecular alterations in the cetuximab resistant cell line that may constitute novel biomarkers of cetuximab response such as mTOR and RhoA overexpression. These findings indicate new strategies to overcome anti-EGFR resistance in HNSCC.
Collapse
Affiliation(s)
- Izabela N. F. Gomes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
| | - Renato J. da Silva-Oliveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
- Barretos School of Medicine Dr. Paulo Prata—FACISB, Barretos 14785-002, Brazil
| | - Luciane Sussuchi da Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
| | - Olga Martinho
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, 4710-057 Braga, Portugal; (O.M.); (F.C.N.)
| | - Adriane F. Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
| | - André van Helvoort Lengert
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
| | - Letícia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
- Barretos School of Medicine Dr. Paulo Prata—FACISB, Barretos 14785-002, Brazil
| | - Viviane Aline Oliveira Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
| | | | - Flávia Caroline Nascimento
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, 4710-057 Braga, Portugal; (O.M.); (F.C.N.)
| | - André Lopes Carvalho
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (R.J.d.S.-O.); (L.S.d.S.); (A.F.E.); (A.v.H.L.); (L.F.L.); (V.A.O.S.); (A.L.C.)
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, 4710-057 Braga, Portugal; (O.M.); (F.C.N.)
- Laboratory of Molecular Diagnosis, Barretos Cancer Hospital, Barretos 14784-400, Brazil;
- 3ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
6
|
Marcu LG, Marcu DC. Current Omics Trends in Personalised Head and Neck Cancer Chemoradiotherapy. J Pers Med 2021; 11:jpm11111094. [PMID: 34834445 PMCID: PMC8625829 DOI: 10.3390/jpm11111094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Chemoradiotherapy remains the most common management of locally advanced head and neck cancer. While both treatment components have greatly developed over the years, the quality of life and long-term survival of patients undergoing treatment for head and neck malignancies are still poor. Research in head and neck oncology is equally focused on the improvement of tumour response to treatment and on the limitation of normal tissue toxicity. In this regard, personalised therapy through a multi-omics approach targeting patient management from diagnosis to treatment shows promising results. The aim of this paper is to discuss the latest results regarding the personalised approach to chemoradiotherapy of head and neck cancer by gathering the findings of the newest omics, involving radiotherapy (dosiomics), chemotherapy (pharmacomics), and medical imaging for treatment monitoring (radiomics). The incorporation of these omics into head and neck cancer management offers multiple viewpoints to treatment that represent the foundation of personalised therapy.
Collapse
Affiliation(s)
- Loredana G. Marcu
- Faculty of Informatics & Science, University of Oradea, 410087 Oradea, Romania
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- Correspondence:
| | - David C. Marcu
- Faculty of Electrical Engineering & Information Technology, University of Oradea, 410087 Oradea, Romania;
| |
Collapse
|
7
|
Maržić D, Marijić B, Braut T, Janik S, Avirović M, Hadžisejdić I, Tudor F, Radobuljac K, Čoklo M, Erovic BM. IMP3 Protein Overexpression Is Linked to Unfavorable Outcome in Laryngeal Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13174306. [PMID: 34503117 PMCID: PMC8430545 DOI: 10.3390/cancers13174306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 02/02/2023] Open
Abstract
Simple Summary IMP3 expression was analyzed in patients with malignant (laryngeal squamous cell carcinoma), semi-malignant (dysplasia) and benign (nodules, polyps) laryngeal lesions and correlated with clinical characteristics. Higher IMP3 stains were particularly found in malignant laryngeal pathologies, which might be useful for differentiation between premalignant and malignant lesions. In laryngeal cancer patients, higher IMP3 expression was associated with positive neck nodes and worse disease-specific survival. Abstract Background: The aim of this study was to (i) determine IMP3 protein expression in benign and malignant laryngeal lesions, (ii) compare its expression to Ki-67, p53, cyclin D1, and (iii) finally, to examine the prognostic power of IMP3 in squamous cell carcinomas of the larynx (LSSC). Methods: IMP3 protein expression was evaluated in 145 patients, including 62 LSCC, 45 dysplasia (25 with low and 20 with high-grade dysplasia), and 38 benign lesions (vocal cord polyps and nodules). Results: IMP3 was significantly higher expressed in LSCC compared to dysplasia and benign lesions (p < 0.001; p < 0.001, respectively). Similarly, higher expression patterns were observed for Ki-67 and p53, whereas cyclin D1 was equally distributed in all three lesions. IMP3 (p = 0.04) and Ki-67 (p = 0.02) expressions were significantly linked to neck node positivity, and IMP3 overexpression to worse disease-specific survival (p = 0.027). Conclusion: Since IMP3 showed significantly higher expression in laryngeal carcinomas, but not in high- or low-grade dysplasia, it serves as a useful marker to differentiate between invasive and noninvasive lesions. Higher IMP3 expression represented a significantly worse prognosticator for clinical outcomes of patients with squamous cell carcinoma of the larynx.
Collapse
Affiliation(s)
- Diana Maržić
- Department of Audiology and Phoniatrics, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (D.M.); (K.R.)
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (B.M.); (T.B.); (M.A.); (I.H.); (F.T.)
| | - Blažen Marijić
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (B.M.); (T.B.); (M.A.); (I.H.); (F.T.)
- Institute of Head and Neck Diseases, Evangelical Hospital, 1180 Vienna, Austria
- Department of Otorhinolaryngology, Head and Neck Surgery, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Tamara Braut
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (B.M.); (T.B.); (M.A.); (I.H.); (F.T.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Stefan Janik
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University Vienna, 1190 Vienna, Austria;
| | - Manuela Avirović
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (B.M.); (T.B.); (M.A.); (I.H.); (F.T.)
- Clinical Department of Pathology and Cytology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Ita Hadžisejdić
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (B.M.); (T.B.); (M.A.); (I.H.); (F.T.)
- Clinical Department of Pathology and Cytology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Filip Tudor
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (B.M.); (T.B.); (M.A.); (I.H.); (F.T.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Katarina Radobuljac
- Department of Audiology and Phoniatrics, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (D.M.); (K.R.)
| | - Miran Čoklo
- Center for Applied Bioanthropology, Institute for Anthropological Research, 10000 Zagreb, Croatia;
| | - Boban M. Erovic
- Institute of Head and Neck Diseases, Evangelical Hospital, 1180 Vienna, Austria
- Correspondence:
| |
Collapse
|
8
|
Riestra-Ayora J, Sánchez-Rodríguez C, Palao-Suay R, Yanes-Díaz J, Martín-Hita A, Aguilar MR, Sanz-Fernández R. Paclitaxel-loaded polymeric nanoparticles based on α-tocopheryl succinate for the treatment of head and neck squamous cell carcinoma: in vivo murine model. Drug Deliv 2021; 28:1376-1388. [PMID: 34180747 PMCID: PMC8245075 DOI: 10.1080/10717544.2021.1923863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The prognosis of patients with recurrent or metastatic head and neck squamous cell cancer (HNSCC) is generally poor. New treatments are required to supplement the current standard of care. Paclitaxel (PTX), an effective chemotherapeutic for HNSCC, has serious side effects. A polymeric nanocarrier system was developed for the delivery of PTX to improve HNSCC treatment. This study aimed to evaluate the antitumor efficacy of PTX-loaded polymeric nanoparticles based on α-TOS (PTX-NPs) administered by direct intratumoral injection into a Hypopharynx carcinoma squamous cells (FaDu) tumor xenograft mouse model. The nanocarrier system based on block copolymers of polyethylene glycol (PEG) and a methacrylic derivative of α-TOS was synthesized and PTX was loaded into the delivery system. Tumor volume was measured to evaluate the antitumor effect of the PTX-NPs. The relative mechanisms of apoptosis, cell proliferation, growth, angiogenesis, and oxidative and nitrosative stress were detected by Western blotting, fluorescent probes, and immunohistochemical analysis. The antitumor activity results showed that compared to free PTX, PTX-NPs exhibited much higher antitumor efficacy and apoptosis-inducing in a FaDu mouse xenograft model and demonstrated an improved safety profile. Ki-67, EGFR, and angiogenesis markers (Factor VIII, CD31, and CD34) expression were significantly lower in the PTX-NPs group compared with other groups (p < .05). Also, PTX-NPs induced oxidative and nitrosative stress in tumor tissue. Direct administration of PTX-loaded polymeric nanoparticles based on α-Tocopheryl Succinate at the tumor sites, proved to be promising for HNSCC therapy.
Collapse
Affiliation(s)
- Juan Riestra-Ayora
- Department otolaryngology, Hospital Universitario de Getafe, Getafe (Madrid), Carretera de Toledo, km 12.500, Getafe, Madrid, Spain.,Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - Carolina Sánchez-Rodríguez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - Raquel Palao-Suay
- Department of Polymeric Nanomaterials and Biomaterials Institute of Polymer Science and Technology CSIC, Networking Biomedical Research Centre in Bioengineering Biomaterials, and Nanomedicine CIBER-BBN, C/Juan de la Cierva, 3, Madrid, Spain
| | - Joaquín Yanes-Díaz
- Department otolaryngology, Hospital Universitario de Getafe, Getafe (Madrid), Carretera de Toledo, km 12.500, Getafe, Madrid, Spain.,Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - Ana Martín-Hita
- Department Pathology, Hospital, Universitario de Getafe, Getafe (Madrid), Carretera de Toledo, km 12.500, Getafe, Madrid, Spain
| | - María Rosa Aguilar
- Department of Polymeric Nanomaterials and Biomaterials Institute of Polymer Science and Technology CSIC, Networking Biomedical Research Centre in Bioengineering Biomaterials, and Nanomedicine CIBER-BBN, C/Juan de la Cierva, 3, Madrid, Spain
| | - Ricardo Sanz-Fernández
- Department otolaryngology, Hospital Universitario de Getafe, Getafe (Madrid), Carretera de Toledo, km 12.500, Getafe, Madrid, Spain.,Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| |
Collapse
|
9
|
Wang H, Nan S, Wang Y, Xu C. CDX2 enhances natural killer cell-mediated immunotherapy against head and neck squamous cell carcinoma through up-regulating CXCL14. J Cell Mol Med 2021; 25:4596-4607. [PMID: 33733587 PMCID: PMC8107099 DOI: 10.1111/jcmm.16253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
(NK) cells are at the first line of defence against tumours, but their anti‐tumour mechanisms are not fully understood. We aimed to investigate the mechanism by which NK cells can mediate immunotherapy against head and neck squamous cell carcinoma (HNSCC). We collected fifty‐two pairs of HNSCC tissues and corresponding adjacent normal tissues; analysis by RT‐qPCR showed underexpression of CXCL14 in HNSCC tissues. Primary NK cells were then isolated from the peripheral blood of HNSCC patients and healthy donors. CXCL14 was found to be consistently under‐expressed in the primary NK cells from the HNSCC patients. However, CXCL14 expression was increased in IL2‐activated primary NK cells and NK‐92 cells. We next evaluated NK cell migration, IFN‐γ and TNF‐α expression, cytotoxicity and infiltration in response to CXCL14 overexpression or knockdown using gain‐ and loss‐of‐function approach. The results exhibited that CXCL14 overexpression promoted NK cell migration, cytotoxicity and infiltration. Subsequent in vivo experiments revealed that CXCL14 suppressed the growth of HNSCC cells via activation of NK cells. ChIP was applied to study the enrichment of H3K27ac, p300, H3K4me1 and CDX2 in the enhancer region of CXCL14, which showed that CDX2/p300 activated the enhancer of CXCL14 to up‐regulate its expression. Rescue experiments demonstrated that CDX2 stimulated NK cell migration, cytotoxicity and infiltration through up‐regulating CXCL14. In vivo data further revealed that CDX2 suppressed tumorigenicity of HNSCC cells through enhancement of CXCL14. To conclude, CDX2 promotes CXCL14 expression by activating its enhancer, which promotes NK cell–mediated immunotherapy against HNSCC.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Otolaryngology Head and Neck Surgery, Jilin University Second Hospital, Changchun, China
| | - Shanji Nan
- Department of Neurology, Jilin University Second Hospital, Changchun, China
| | - Ying Wang
- Department of Gastroenterology, Jilin University First Hospital, Changchun, China
| | - Chengbi Xu
- Department of Otolaryngology Head and Neck Surgery, Jilin University Second Hospital, Changchun, China
| |
Collapse
|
10
|
Jureczek J, Feldmann A, Bergmann R, Arndt C, Berndt N, Koristka S, Loureiro LR, Mitwasi N, Hoffmann A, Kegler A, Bartsch T, Bachmann M. Highly Efficient Targeting of EGFR-Expressing Tumor Cells with UniCAR T Cells via Target Modules Based on Cetuximab ®. Onco Targets Ther 2020; 13:5515-5527. [PMID: 32606767 PMCID: PMC7297505 DOI: 10.2147/ott.s245169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/19/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Since epithelial growth factor receptor (EGFR) overexpression is linked to a variety of malignancies, it is an attractive target for immune therapy including chimeric antigen receptor (CAR)-engineered T cells. Unfortunately, CAR T cell therapy harbors the risk of severe, even life-threatening side effects. Adaptor CAR T cell platforms such as the previously described UniCAR system might be able to overcome these problems. In contrast to conventional CARs, UniCAR T cells are per se inert. Their redirection towards target cells occurs only in the presence of a tumor-specific target molecule (TM). TMs are bifunctional molecules being able to recognize a tumor-associated antigen and to cross-link the CAR T cell via a peptide epitope recognized by the UniCAR domain. Materials and Methods Here, we compare αEGFR TMs: a nanobody (nb)-based αEGFR TM derived from the camelid αEGFR antibody 7C12 with a murine and humanized single-chain fragment variable (scFv) based on the clinically used antibody Cetuximab®. Results In principle, both the nb- and scFv-based TM formats are able to redirect UniCAR T cells to eliminate EGFR-expressing tumor cells in an antigen-specific and TM-dependent manner. However, the scFv-based αEGFR TM was significantly superior to the nb-based TM especially with respect to lysis of tumor cells. Discussion Improved efficiency of the scFv-based TM allowed the redirection of UniCAR T cells towards tumor cells expressing high as well as low EGFR levels in comparison to nb-based αEGFR TMs.
Collapse
Affiliation(s)
- Justyna Jureczek
- German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicole Berndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Stefanie Koristka
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Liliana Rodrigues Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Nicola Mitwasi
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Hoffmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Alexandra Kegler
- German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Tabea Bartsch
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Michael Bachmann
- German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.,Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
11
|
Crombet Ramos T, Mestre Fernández B, Mazorra Herrera Z, Iznaga Escobar NE. Nimotuzumab for Patients With Inoperable Cancer of the Head and Neck. Front Oncol 2020; 10:817. [PMID: 32537431 PMCID: PMC7266975 DOI: 10.3389/fonc.2020.00817] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
EGFR activation induces cell proliferation, neoformation of blood vessels, survival, and metastasis of the cancer cells. Nimotuzumab is an engineered, intermediate affinity anti-EGFR antibody, that apart from other drugs in its class, is very safe and does not cause hypomagnesemia or grade 3–4 cutaneous rash. The antibody inhibits cell proliferation and angiogenesis, activates natural killer cells, stimulates dendritic cell maturation, and induces cytotoxic T cells. Nimotuzumab restores MHC-I expression on tumor cells, hindering one of the EGFR immune-escape ways. The antibody has been extensively studied in 7 clinical trials, concurrently with irradiation or irradiation plus chemotherapy in subjects with inoperable head and neck tumors. Nimotuzumab was safe and efficacious in unfit patients receiving irradiation alone and in subjects treated with cisplatin and radiotherapy. In patients with locally advanced squamous cell carcinomas of the head and neck, nimotuzumab in combination with low dose cisplatin and radiotherapy was superior to cisplatin and radiotherapy in progression free survival, disease free survival, and locoregional tumor control.
Collapse
|
12
|
Montanuy H, Martínez-Barriocanal Á, Antonio Casado J, Rovirosa L, Ramírez MJ, Nieto R, Carrascoso-Rubio C, Riera P, González A, Lerma E, Lasa A, Carreras-Puigvert J, Helleday T, Bueren JA, Arango D, Minguillón J, Surrallés J. Gefitinib and Afatinib Show Potential Efficacy for Fanconi Anemia-Related Head and Neck Cancer. Clin Cancer Res 2020; 26:3044-3057. [PMID: 32005748 DOI: 10.1158/1078-0432.ccr-19-1625] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/29/2019] [Accepted: 01/28/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Fanconi anemia rare disease is characterized by bone marrow failure and a high predisposition to solid tumors, especially head and neck squamous cell carcinoma (HNSCC). Patients with Fanconi anemia with HNSCC are not eligible for conventional therapies due to high toxicity in healthy cells, predominantly hematotoxicity, and the only treatment currently available is surgical resection. In this work, we searched and validated two already approved drugs as new potential therapies for HNSCC in patients with Fanconi anemia. EXPERIMENTAL DESIGN We conducted a high-content screening of 3,802 drugs in a FANCA-deficient tumor cell line to identify nongenotoxic drugs with cytotoxic/cytostatic activity. The best candidates were further studied in vitro and in vivo for efficacy and safety. RESULTS Several FDA/European Medicines Agency (EMA)-approved anticancer drugs showed cancer-specific lethality or cell growth inhibition in Fanconi anemia HNSCC cell lines. The two best candidates, gefitinib and afatinib, EGFR inhibitors approved for non-small cell lung cancer (NSCLC), displayed nontumor/tumor IC50 ratios of approximately 400 and approximately 100 times, respectively. Neither gefitinib nor afatinib activated the Fanconi anemia signaling pathway or induced chromosomal fragility in Fanconi anemia cell lines. Importantly, both drugs inhibited tumor growth in xenograft experiments in immunodeficient mice using two Fanconi anemia patient-derived HNSCCs. Finally, in vivo toxicity studies in Fanca-deficient mice showed that administration of gefitinib or afatinib was well-tolerated, displayed manageable side effects, no toxicity to bone marrow progenitors, and did not alter any hematologic parameters. CONCLUSIONS Our data present a complete preclinical analysis and promising therapeutic line of the first FDA/EMA-approved anticancer drugs exerting cancer-specific toxicity for HNSCC in patients with Fanconi anemia.
Collapse
Affiliation(s)
- Helena Montanuy
- Department of Genetics and Microbiology. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Águeda Martínez-Barriocanal
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Group of Molecular Oncology, IRB Lleida, Lleida, Spain
| | - José Antonio Casado
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM), Madrid, Spain
| | - Llorenç Rovirosa
- Department of Genetics and Microbiology. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria José Ramírez
- Department of Genetics and Microbiology. Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Genetics Department and Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rocío Nieto
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos Carrascoso-Rubio
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM), Madrid, Spain
| | - Pau Riera
- Genetics Department and Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Alan González
- Department of Anatomic Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Enrique Lerma
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Adriana Lasa
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Genetics Department and Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jordi Carreras-Puigvert
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Molecular Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Helleday
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Molecular Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Juan A Bueren
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM), Madrid, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Group of Molecular Oncology, IRB Lleida, Lleida, Spain
| | - Jordi Minguillón
- Department of Genetics and Microbiology. Universitat Autònoma de Barcelona, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM), Madrid, Spain
| | - Jordi Surrallés
- Department of Genetics and Microbiology. Universitat Autònoma de Barcelona, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM), Madrid, Spain
| |
Collapse
|
13
|
Borges GÁ, Elias ST, Araujo TSD, Souza PM, Nascimento-Filho CHV, Castilho RM, Squarize CH, Magalhães PDO, Guerra ENS. Asparaginase induces selective dose- and time-dependent cytotoxicity, apoptosis, and reduction of NFκB expression in oral cancer cells. Clin Exp Pharmacol Physiol 2020; 47:857-866. [PMID: 31943292 DOI: 10.1111/1440-1681.13256] [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] [Received: 05/31/2019] [Revised: 11/19/2019] [Accepted: 01/08/2020] [Indexed: 01/10/2023]
Abstract
Asparaginase is fundamental to the treatment of haematological malignancies. However, little has been studied on the effects that asparaginase could exert on solid tumours. Thus, this study aimed to evaluate the effects of asparaginase on an oral carcinoma cell line. The cytotoxicity of asparaginase in SCC-9 (tongue squamous cell carcinoma) and HaCaT (human keratinocyte) cell lines was evaluated with MTT cell viability assay. The cells were treated with asparaginase at 0.04, 0.16, 0.63, 1.0, 1.5, 2.5, and 5.0 IU/mL. Dose-response curves and IC50 values were obtained and the Tumour Selectivity Index (TSI) was calculated. The effect of asparaginase on procaspase-3 and nuclear factor κB (NFκB) expression was evaluated with western blot because it was reported that the overexpression of NFκB has been shown to contribute to tumour cell survival, proliferation, and migration. Caspase 3/7 staining was performed to identify cell death using flow cytometry. Effective asparaginase concentrations were lower for SCC-9 cells when compared to HaCaT cells. The cytotoxicity results at 48 and 72 hours were significantly different for SCC-9 cells. The TSI indicated that asparaginase was selective for the tumour cells. A decrease in procaspase-3 and NFκB protein levels was observed in SCC-9 cells. Furthermore, asparaginase resulted in significant apoptosis after 48 and 72 hours. Based on these results, asparaginase was cytotoxic in a dose- and time-dependent manner, induces apoptosis, and reduces NFκB expression in oral cancer cells. These results encourage further studies on the effectiveness of this enzyme as a treatment for solid tumours, especially head and neck cancer.
Collapse
Affiliation(s)
- Gabriel Álvares Borges
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasilia, Brazil.,Epithelial Biology Laboratory, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Silvia Taveira Elias
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Tassiana Souza De Araujo
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Paula Monteiro Souza
- Natural Products Laboratory, Faculty of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Carlos Henrique Viesi Nascimento-Filho
- Epithelial Biology Laboratory, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Rogerio M Castilho
- Epithelial Biology Laboratory, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Cristiane H Squarize
- Epithelial Biology Laboratory, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | | | - Eliete Neves Silva Guerra
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasilia, Brazil.,Epithelial Biology Laboratory, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
14
|
Ion Channel Dysregulation in Head and Neck Cancers: Perspectives for Clinical Application. Rev Physiol Biochem Pharmacol 2020; 181:375-427. [PMID: 32789787 DOI: 10.1007/112_2020_38] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Head and neck cancers are a highly complex and heterogeneous group of malignancies that involve very diverse anatomical structures and distinct aetiological factors, treatments and clinical outcomes. Among them, head and neck squamous cell carcinomas (HNSCC) are predominant and the sixth most common cancer worldwide with still low survival rates. Omic technologies have unravelled the intricacies of tumour biology, harbouring a large diversity of genetic and molecular changes to drive the carcinogenesis process. Nonetheless, this remarkable heterogeneity of molecular alterations opens up an immense opportunity to discover novel biomarkers and develop molecular-targeted therapies. Increasing evidence demonstrates that dysregulation of ion channel expression and/or function is frequently and commonly observed in a variety of cancers from different origin. As a consequence, the concept of ion channels as potential membrane therapeutic targets and/or biomarkers for cancer diagnosis and prognosis has attracted growing attention. This chapter intends to comprehensively and critically review the current state-of-art ion channel dysregulation specifically focusing on head and neck cancers and to formulate the major challenges and research needs to translate this knowledge into clinical application. Based on current reported data, various voltage-gated potassium (Kv) channels (i.e. Kv3.4, Kv10.1 and Kv11.1) have been found frequently aberrantly expressed in HNSCC as well as precancerous lesions and are highlighted as clinically and biologically relevant features in both early stages of tumourigenesis and late stages of disease progression. More importantly, they also emerge as promising candidates as cancer risk markers, tumour markers and potential anti-proliferative and anti-metastatic targets for therapeutic interventions; however, the oncogenic properties seem to be independent of their ion-conducting function.
Collapse
|
15
|
Thomas R, Weihua Z. Rethink of EGFR in Cancer With Its Kinase Independent Function on Board. Front Oncol 2019; 9:800. [PMID: 31508364 PMCID: PMC6716122 DOI: 10.3389/fonc.2019.00800] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/06/2019] [Indexed: 12/23/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is one of most potent oncogenes that are commonly altered in cancers. As a receptor tyrosine kinase, EGFR's kinase activity has been serving as the primary target for developing cancer therapeutics, namely the EGFR inhibitors including small molecules targeting its ATP binding pocket and monoclonal antibodies targeting its ligand binding domains. EGFR inhibitors have produced impressive therapeutic benefits to responsive types of cancers. However, acquired and innate resistances have precluded current anti-EGFR agents from offering sustainable benefits to initially responsive cancers and benefits to EGFR-positive cancers that are innately resistant. Recent years have witnessed a realization that EGFR possesses kinase-independent (KID) pro-survival functions in cancer cells. This new knowledge has offered a different angle of understanding of EGFR in cancer and opened a new avenue of targeting EGFR for cancer therapy. There are already many excellent reviews on the role of EGFR with a focus on its kinase-dependent functions and mechanisms of resistance to EGFR targeted therapies. The present opinion aims to initiate a fresh discussion about the function of EGFR in cancer cells by laying out some unanswered questions pertaining to EGFR in cancer cells, by rethinking the unmet therapeutic challenges from a view of EGFR's KID function, and by proposing novel approaches to target the KID functions of EGFR for cancer treatment.
Collapse
Affiliation(s)
- Rintu Thomas
- Department of Biology and Biochemistry, College of Natural Science and Mathematics, University of Houston, Houston, TX, United States
| | - Zhang Weihua
- Department of Biology and Biochemistry, College of Natural Science and Mathematics, University of Houston, Houston, TX, United States
| |
Collapse
|
16
|
Targeted Biological Drugs and Immune Check Point Inhibitors for Locally Advanced or Metastatic Cancers of the Conjunctiva, Eyelid, and Orbit. Int Ophthalmol Clin 2019; 59:13-26. [PMID: 30908276 DOI: 10.1097/iio.0000000000000271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
17
|
Wang Q, Song X, Zhao Y, He Q, Shi M, Xu P, Ni S, Chen Y, Lin J, Zhang L. Preoperative high c-reactive protein/albumin ratio is a poor prognostic factor of oral squamous cell carcinoma. Future Oncol 2019; 15:2277-2286. [PMID: 31237166 DOI: 10.2217/fon-2019-0063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: To explore whether c-reactive protein/albumin (CRP/Alb) ratio is a poor prognostic factor for patients with oral squamous cell carcinoma (OSCC). Patients & methods: Receiver-operating characteristic analysis was performed to evaluate the optimal cut-off value of CRP/Alb ratio in 240 patients with OSCC. The Kaplan-Meier method was used to plot the overall survival and disease-free survival curves. Cox proportional hazards model was used to implement univariate and multivariate analyses. Results: Preoperative high CRP/Alb ratio was associated with age, advanced stage, lymphatic metastasis, platelet-to-lymphocyte ratio and neutrophil-to-lymphocyte ratio (all p < 0.05). Elevated CRP/Alb ratio independently predicts worse overall survival and disease-free survival of patients with OSCC. Conclusion: Preoperative high CRP/Alb ratio was a poor independent prognostic marker of OSCC.
Collapse
Affiliation(s)
- Qiuju Wang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Xiaoyu Song
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Yanzhen Zhao
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Qiao He
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Min Shi
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Pingyao Xu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Sujiao Ni
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Yibo Chen
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Jingying Lin
- West China Second University Hospital, Sichuan University, & The Key Laboratory of Birth Defects & Related Diseases of Women & Children (Sichuan University), Ministry of Education, Chengdu, PR China
| | - Li Zhang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| |
Collapse
|
18
|
Rahman S, Kraljević Pavelić S, Markova-Car E. Circadian (De)regulation in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2019; 20:ijms20112662. [PMID: 31151182 PMCID: PMC6600143 DOI: 10.3390/ijms20112662] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/26/2019] [Accepted: 05/28/2019] [Indexed: 12/15/2022] Open
Abstract
Head and neck cancer encompass different malignancies that develop in and around the throat, larynx, nose, sinuses and mouth. Most head and neck cancers are squamous cell carcinomas (HNSCC) that arise in the flat squamous cells that makeup the thin layer of tissue on the surface of anatomical structures in the head and neck. Each year, HNSCC is diagnosed in more than 600,000 people worldwide, with about 50,000 new cases. HNSCC is considered extremely curable if detected early. But the problem remains in treatment of inoperable cases, residues or late stages. Circadian rhythm regulation has a big role in developing various carcinomas, and head and neck tumors are no exception. A number of studies have reported that alteration in clock gene expression is associated with several cancers, including HNSCC. Analyses on circadian clock genes and their association with HNSCC have shown that expression of PER1, PER2, PER3, CRY1, CRY2,CKIε, TIM, and BMAL1 are deregulated in HNSCC tissues. This review paper comprehensively presents data on deregulation of circadian genes in HNSCC and critically evaluates their potential diagnostics and prognostics role in this type of pathology.
Collapse
Affiliation(s)
- Sadia Rahman
- University of Rijeka, Department of Biotechnology, Centre for High-Throughput Technologies, 51000 Rijeka, Croatia.
| | - Sandra Kraljević Pavelić
- University of Rijeka, Department of Biotechnology, Centre for High-Throughput Technologies, 51000 Rijeka, Croatia.
| | - Elitza Markova-Car
- University of Rijeka, Department of Biotechnology, Centre for High-Throughput Technologies, 51000 Rijeka, Croatia.
| |
Collapse
|
19
|
Peitzsch C, Nathansen J, Schniewind SI, Schwarz F, Dubrovska A. Cancer Stem Cells in Head and Neck Squamous Cell Carcinoma: Identification, Characterization and Clinical Implications. Cancers (Basel) 2019; 11:cancers11050616. [PMID: 31052565 PMCID: PMC6562868 DOI: 10.3390/cancers11050616] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/21/2019] [Accepted: 04/26/2019] [Indexed: 12/19/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most commonly diagnosed cancer worldwide. Despite advances in the treatment management, locally advanced disease has a poor prognosis, with a 5-year survival rate of approximately 50%. The growth of HNSCC is maintained by a population of cancer stem cells (CSCs) which possess unlimited self-renewal potential and induce tumor regrowth if not completely eliminated by therapy. The population of CSCs is not only a promising target for tumor treatment, but also an important biomarker to identify the patients at risk for therapeutic failure and disease progression. This review aims to provide an overview of the recent pre-clinical and clinical studies on the biology and potential therapeutic implications of HNSCC stem cells.
Collapse
Affiliation(s)
- Claudia Peitzsch
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner site Dresden, 01307 Dresden, Germany.
| | - Jacqueline Nathansen
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Sebastian I Schniewind
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Franziska Schwarz
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner site Dresden, 01307 Dresden, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01307 Dresden, Germany.
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner site Dresden, 01307 Dresden, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01307 Dresden, Germany.
| |
Collapse
|
20
|
Jiang X, Ye J, Dong Z, Hu S, Xiao M. Novel genetic alterations and their impact on target therapy response in head and neck squamous cell carcinoma. Cancer Manag Res 2019; 11:1321-1336. [PMID: 30799957 PMCID: PMC6371928 DOI: 10.2147/cmar.s187780] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is highly variable by tumor site, histologic type, molecular characteristics, and clinical outcome. During recent years, emerging targeted therapies have been focused on driver genes. HNSCC involves several genetic alterations, such as co-occurrence, multiple feedback loops, and cross-talk communications. These different kinds of genetic alterations interact with each other and mediate targeted therapy response. In the current review, it is emphasized that future treatment strategy in HNSCC will not solely be based on "synthetic lethality" approaches directed against overactivated genes. More importantly, biologic, genetic, and epigenetic alterations of HNSCC will be taken into consideration to guide the therapy. The emerging genetic alterations in HNSCC and its effect on targeted therapy response are discussed in detail. Hopefully, novel combination regimens for the treatment of HNSCC can be developed.
Collapse
Affiliation(s)
- Xiaohua Jiang
- Department of Otolaryngology Head and Neck Surgery, Sir Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,
| | - Jing Ye
- Department of Otolaryngology Head and Neck Surgery, Sir Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,
| | - Zhihuai Dong
- Department of Otolaryngology Head and Neck Surgery, Sir Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,
| | - Sunhong Hu
- Department of Otolaryngology Head and Neck Surgery, Sir Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,
| | - Mang Xiao
- Department of Otolaryngology Head and Neck Surgery, Sir Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,
| |
Collapse
|
21
|
Pidugu VK, Wu MM, Yen AH, Pidugu HB, Chang KW, Liu CJ, Lee TC. IFIT1 and IFIT3 promote oral squamous cell carcinoma metastasis and contribute to the anti-tumor effect of gefitinib via enhancing p-EGFR recycling. Oncogene 2019; 38:3232-3247. [PMID: 30626937 DOI: 10.1038/s41388-018-0662-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/12/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023]
Abstract
IFIT1 and IFIT3 are abundant products of interferon-stimulating genes. While the importance of IFIT1 and IFIT3 in the prognosis of cancer has been reported, the molecular basis of IFIT1 and IFIT3 in cancer progression remains unexplored. In the present study, we investigated the modes of action and the clinical significance of IFIT1 and IFIT3 in oral squamous cell carcinoma (OSCC). Ectopic expression of IFIT1 or IFIT3 induced OSCC cell invasion by promoting the epithelial-mesenchymal transition, whereas IFIT1 or IFIT3 knockdown exhibited opposite effects. Overexpression of IFIT1 or IFIT3 promoted tumor growth, regional and distant metastasis in xenograft and orthotopic nude mice models. Most importantly, IFIT1 or IFIT3 overexpression increased the levels of p-EGFRY1068 and p-AKTS473 in OSCC cells and also enhanced tumor inhibitory effect of gefitinib. By immunoprecipitation and LC-MS/MS analysis, we found that IFIT1 and IFIT3 interacted with ANXA2 that enhanced p-EGFRY1068 endosomal recycling. Depletion of ANXA2 using siRNA therefore abolished p-EGFRY1068 and p-AKTS473 expression in IFIT1- or IFIT3-overexpressed cells. Furthermore, a significant positive association of increased IFIT1 and IFIT3 expression with advanced T-stage, lymph node metastasis, perineural invasion, lymphovascular invasion, extranodal extension, and poor overall survival rate was confirmed in OSCC patients. We also found a statistically positive correlation of p-EGFRY1068 expression with IFIT1 and IFIT3 in OSCC tumors and poor clinical outcome in patients. Collectively, we demonstrated a novel role of IFIT1 and IFIT3 in driving OSCC progression and metastasis by interacting with ANXA2 and hence enhancing p-EGFR recycling and its downstream signaling.
Collapse
Affiliation(s)
- Vijaya Kumar Pidugu
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University, and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Meei-Maan Wu
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ai-Hsin Yen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Hima Bindu Pidugu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Kuo-Wei Chang
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Chung-Ji Liu
- Department of Oral and Maxillofacial Surgery, Mackay Memorial Hospital, Taipei, 10449, Taiwan.
| | - Te-Chang Lee
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University, and Academia Sinica, Taipei, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan. .,Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.
| |
Collapse
|
22
|
Buch S, Chatra L. Immunotherapy and its advances in the management of head-and-neck cancer. CHRISMED JOURNAL OF HEALTH AND RESEARCH 2019. [DOI: 10.4103/cjhr.cjhr_155_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
23
|
Chopra M. Annual Congress of the European Society for Medical Oncology (ESMO): Munich, Germany, 19-23 October 2018. Target Oncol 2018; 13:673-677. [PMID: 30426327 DOI: 10.1007/s11523-018-0608-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Martin Chopra
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
24
|
Abstract
OPINION STATEMENT In head and neck cancer, we continue to work towards a more personalized approach to treatment of patients, where analysis of a patient's tumor guides targeting of molecular or immunologic pathways. Critically important to this pursuit is a better understanding of the direct biologic effect of a drug or combination on the tumor microenvironment in humans, as well as biomarker discovery. These goals are consistent with the primary purpose of a "window of opportunity" trial and while conduct of these trials requires a careful balance of benefits and potential risks, to date these trials have been both feasible and safe in HNSCC in the curative intent setting. In the era of immunotherapy, with countless possible combinations and ongoing clinical trials, window trials are even more important for informing clinical trial design and appropriate combination therapy, and ultimately a more personalized approach to our patients that leads to improvement in outcomes.
Collapse
|
25
|
Tanaka N, Osman AA, Takahashi Y, Lindemann A, Patel AA, Zhao M, Takahashi H, Myers JN. Head and neck cancer organoids established by modification of the CTOS method can be used to predict in vivo drug sensitivity. Oral Oncol 2018; 87:49-57. [PMID: 30527243 DOI: 10.1016/j.oraloncology.2018.10.018] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Currently there are no standard biomarkers of head and neck squamous cell carcinoma (HNSCC) response to therapy. This is, due to a lack of adequate predictive tumor models. To this end, we established cancer organoid lines from individual patient's tumors, and characterized their growth characteristics and response to different drug treatments with the objective of using these models for prediction of treatment response. MATERIALS AND METHODS Forty-three patients' samples were processed to establish organoids. To analyze the character of these organoids, immunohistochemistry, Western blotting, drug sensitivity assays, clonogenic survival assays, and animal experiments were performed. The HPV status and TP53 mutational status were also confirmed in these lines. RESULTS HNSCC organoids were successfully established with success rate of 30.2%. Corresponding two-dimensional cell lines were established from HNSCC organoids at higher success rate (53.8%). These organoids showed similar histological features and stem cell, epithelial and mesenchymal marker expression to the original tumors, thus recapitulating many of the characteristics of the original tumor cells. The cisplatin and docetaxel IC50 were determined for HNSCC organoids and the corresponding 2D cell lines using drug sensitivity and clonogenic survival assays. Responses to drug treatment in vivo were found to be similar to the IC50 calculated from organoids by drug sensitivity assays in vitro. CONCLUSION We established novel in vitro HNSCC cancer organoid lines retaining many properties of the original tumors from they were derived. These organoids can predict in vivo drug sensitivity and may represent useful tools to develop precision treatments for HNSCC.
Collapse
Affiliation(s)
- Noriaki Tanaka
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abdullah A Osman
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yoko Takahashi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Antje Lindemann
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ameeta A Patel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mei Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hideaki Takahashi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
26
|
Lin XL, Xiao XY. Progress in neoadjuvant drug therapy of rectal cancer. Shijie Huaren Xiaohua Zazhi 2018; 26:1340-1347. [DOI: 10.11569/wcjd.v26.i22.1340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rectal cancer is one of the most common malignant tumors of the digestive system. A portion of patients have had locally advanced disease at the time of diagnosis and have lost the chance of radical surgery. How to increase the R0 resection rate has always been a major difficulty in clinical practice. Some studies have shown that for some patients, neoadjuvant drug therapy can achieve reduction of clinical stage, increase the R0 resection rate and anus-preserving rate, reduce the local recurrence and micro-metastasis, and prolong the survival time of patients. This article mainly reviews the progress in neoadjuvant drug therapy of rectal cancer.
Collapse
Affiliation(s)
- Xiao-Lin Lin
- Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiu-Ying Xiao
- Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
27
|
Kartha VK, Alamoud KA, Sadykov K, Nguyen BC, Laroche F, Feng H, Lee J, Pai SI, Varelas X, Egloff AM, Snyder-Cappione JE, Belkina AC, Bais MV, Monti S, Kukuruzinska MA. Functional and genomic analyses reveal therapeutic potential of targeting β-catenin/CBP activity in head and neck cancer. Genome Med 2018; 10:54. [PMID: 30029671 PMCID: PMC6053793 DOI: 10.1186/s13073-018-0569-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/11/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy characterized by tumor heterogeneity, locoregional metastases, and resistance to existing treatments. Although a number of genomic and molecular alterations associated with HNSCC have been identified, they have had limited impact on the clinical management of this disease. To date, few targeted therapies are available for HNSCC, and only a small fraction of patients have benefited from these treatments. A frequent feature of HNSCC is the inappropriate activation of β-catenin that has been implicated in cell survival and in the maintenance and expansion of stem cell-like populations, thought to be the underlying cause of tumor recurrence and resistance to treatment. However, the therapeutic value of targeting β-catenin activity in HNSCC has not been explored. METHODS We utilized a combination of computational and experimental profiling approaches to examine the effects of blocking the interaction between β-catenin and cAMP-responsive element binding (CREB)-binding protein (CBP) using the small molecule inhibitor ICG-001. We generated and annotated in vitro treatment gene expression signatures of HNSCC cells, derived from human oral squamous cell carcinomas (OSCCs), using microarrays. We validated the anti-tumorigenic activity of ICG-001 in vivo using SCC-derived tumor xenografts in murine models, as well as embryonic zebrafish-based screens of sorted stem cell-like subpopulations. Additionally, ICG-001-inhibition signatures were overlaid with RNA-sequencing data from The Cancer Genome Atlas (TCGA) for human OSCCs to evaluate its association with tumor progression and prognosis. RESULTS ICG-001 inhibited HNSCC cell proliferation and tumor growth in cellular and murine models, respectively, while promoting intercellular adhesion and loss of invasive phenotypes. Furthermore, ICG-001 preferentially targeted the ability of subpopulations of stem-like cells to establish metastatic tumors in zebrafish. Significantly, interrogation of the ICG-001 inhibition-associated gene expression signature in the TCGA OSCC human cohort indicated that the targeted β-catenin/CBP transcriptional activity tracked with tumor status, advanced tumor grade, and poor overall patient survival. CONCLUSIONS Collectively, our results identify β-catenin/CBP interaction as a novel target for anti-HNSCC therapy and provide evidence that derivatives of ICG-001 with enhanced inhibitory activity may serve as an effective strategy to interfere with aggressive features of HNSCC.
Collapse
Affiliation(s)
- Vinay K Kartha
- Bioinformatics Program, Boston University, Boston, MA, USA
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Khalid A Alamoud
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Khikmet Sadykov
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Bach-Cuc Nguyen
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Fabrice Laroche
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Hui Feng
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jina Lee
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara I Pai
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Ann Marie Egloff
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jennifer E Snyder-Cappione
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Anna C Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | - Manish V Bais
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Stefano Monti
- Bioinformatics Program, Boston University, Boston, MA, USA
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Maria A Kukuruzinska
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA.
| |
Collapse
|
28
|
Blas K, Wilson TG, Tonlaar N, Galoforo S, Hana A, Marples B, Wilson GD. Dual blockade of PI3K and MEK in combination with radiation in head and neck cancer. Clin Transl Radiat Oncol 2018; 11:1-10. [PMID: 30014041 PMCID: PMC6019866 DOI: 10.1016/j.ctro.2018.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023] Open
Abstract
Background and purpose In this study we have combined fractionated radiation treatment (RT) with two molecular targeted agents active against key deregulated signaling pathways in head and neck cancer. Materials and methods We used two molecularly characterized, low passage HNSCC cell lines of differing biological characteristics to study the effects of binimetinib and buparlisib in combination with radiation in vitro and in vivo. Results Buparlisib was active against both cell lines in vitro whereas binimetinib was more toxic to UT-SCC-14. Neither agent modified radiation sensitivity in vitro. Buparlisib significantly inhibited growth of UT-SSC-15 alone or in combination with RT but was ineffective in UT-SCC-14. Binimetinib did cause a significant delay with RT in UT-SCC-14 and it significantly reduced growth of the UT-SCC-15 tumors both alone and with RT. The tri-modality treatment was not as effective as RT with a single effective agent. Conclusions No significant benefit was gained by the combined use of the two agents with RT even though each was efficacious when used alone.
Collapse
Affiliation(s)
- Kevin Blas
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Thomas G Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Nathan Tonlaar
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Sandra Galoforo
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Alaa Hana
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Brian Marples
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - George D Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States.,Beaumont BioBank, William Beaumont Hospital, Royal Oak, MI, United States
| |
Collapse
|
29
|
Polverini PJ, D'Silva NJ, Lei YL. Precision Therapy of Head and Neck Squamous Cell Carcinoma. J Dent Res 2018; 97:614-621. [PMID: 29649374 DOI: 10.1177/0022034518769645] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Precision medicine is an approach to disease prevention and treatment that takes into account genetic variability and environmental and lifestyle influences that are unique to each patient. It facilitates stratification of patient populations that vary in their susceptibility to disease and response to therapy. Shared databases and the implementation of new technology systems designed to advance the integration of this information will enable health care providers to more accurately predict and customize prevention and treatment strategies for patients. Although precision medicine has had a limited impact in most areas of medicine, it has been shown to be an increasingly successful approach to cancer therapy. Despite early promising results targeting aberrant signaling pathways or inhibitors designed to block tumor-driven processes such as angiogenesis, limited success emphasizes the need to discover new biomarkers and treatment targets that are more reliable in predicting response to therapy and result in better health outcomes. Recent successes in the use of immunity-inducing antibodies have stimulated increased interest in the use of precision immunotherapy of head and neck squamous cell carcinoma. Using next-generation sequencing, the precise profiling of tumor-infiltrating lymphocytes has great promise to identify hypoimmunogenic cancer that would benefit from a rationally designed combinatorial approach. Continued interrogation of tumors will reveal new actionable targets with increasing therapeutic efficacy and fulfill the promise of precision therapy of head and neck cancer.
Collapse
Affiliation(s)
- P J Polverini
- 1 Department of Periodontics and Oral Medicine, Division of Oral Medicine, Pathology, and Radiology, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,2 Department of Pathology, University of Michigan Health System, Ann Arbor, MI, USA.,3 Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - N J D'Silva
- 1 Department of Periodontics and Oral Medicine, Division of Oral Medicine, Pathology, and Radiology, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,2 Department of Pathology, University of Michigan Health System, Ann Arbor, MI, USA.,3 Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Y L Lei
- 1 Department of Periodontics and Oral Medicine, Division of Oral Medicine, Pathology, and Radiology, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,3 Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.,4 Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| |
Collapse
|
30
|
van Rosmalen M, Ni Y, Vervoort DFM, Arts R, Ludwig SKJ, Merkx M. Dual-Color Bioluminescent Sensor Proteins for Therapeutic Drug Monitoring of Antitumor Antibodies. Anal Chem 2018; 90:3592-3599. [PMID: 29443503 PMCID: PMC5843950 DOI: 10.1021/acs.analchem.8b00041] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Monitoring the levels of therapeutic antibodies in individual patients would allow patient-specific dose optimization, with the potential for major therapeutic and financial benefits. Our group recently developed a new platform of bioluminescent sensor proteins (LUMABS; LUMinescent AntiBody Sensor) that allow antibody detection directly in blood plasma. In this study, we targeted four clinically important therapeutic antibodies, the Her2-receptor targeting trastuzumab, the anti-CD20 antibodies rituximab and obinutuzumab, and the EGFR-blocking cetuximab. A strong correlation was found between the affinity of the antibody binding peptide and sensor performance. LUMABS sensors with physiologically relevant affinities and decent sensor responses were obtained for trastuzumab and cetuximab using mimotope and meditope peptides, respectively, with affinities in the 10-7 M range. The lower affinity of the CD20-derived cyclic peptide employed in the anti-CD20 LUMABS sensor ( Kd = 10-5 M), translated in a LUMABS sensor with a strongly attenuated sensor response. The trastuzumab and cetuximab sensors were further characterized with respect to binding kinetics and their performance in undiluted blood plasma. For both antibodies, LUMABS-based detection directly in plasma compared well to the analytical performance of commercial ELISA kits. Besides identifying important design parameters for the development of new LUMABS sensors, this work demonstrates the potential of the LUMABS platform for point-of-care detection of therapeutic antibodies.
Collapse
Affiliation(s)
- Martijn van Rosmalen
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Yan Ni
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Daan F M Vervoort
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Remco Arts
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Susann K J Ludwig
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Maarten Merkx
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| |
Collapse
|
31
|
Brands RC, Scheurer MJJ, Hartmann S, Seher A, Kübler AC, Müller-Richter UDA. Apoptosis-sensitizing activity of birinapant in head and neck squamous cell carcinoma cell lines. Oncol Lett 2018; 15:4010-4016. [PMID: 29467909 DOI: 10.3892/ol.2018.7783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/20/2017] [Indexed: 02/07/2023] Open
Abstract
Inhibitor of apoptosis proteins, which are overexpressed in head and neck squamous cell carcinoma (HNSCC), may cause therapeutic resistance. Using SMAC mimetic compounds, including birinapant, to degrade and/or inhibit these proteins and sensitize apoptosis may enhance therapies in HNSCC. Fas expression was analyzed in nine HNSCC cell lines and one keratinocyte cell line via flow cytometry. These cell lines were treated with Fas ligand-Fc (FasL) and birinapant, a bivalent SMAC mimetic, in mono and combination therapies. Cytotoxicity was measured using a crystal violet assay. Annexin V assay was performed for detection of apoptosis. The treatment efficacy of mono and combination therapies was statistically analyzed. Nonlinear regression analysis was performed to determine the inhibitory concentration (IC10) of birinapant. Fas expression was detected in each cell line tested. Mono treatment with FasL revealed minor to no apoptotic effects in the majority of the cell lines. Crystal violet and Annexin V staining revealed increased apoptosis rates for all cell lines following incubation with birinapant in mono treatment. Combination treatment with FasL and birinapant (IC10) revealed additional and synergistic effects in eight out of the ten cell lines. To the best of our knowledge, the present study provided the first evidence of the apoptosis-sensitizing activity of combination treatment with FasL and birinapant in HNSCC cell lines.
Collapse
Affiliation(s)
- Roman C Brands
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, D-97080 Würzburg, Germany
| | - Mario J J Scheurer
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany
| | - Stefan Hartmann
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany.,Interdisciplinary Center for Clinical Research, University Hospital Würzburg, D-97070 Würzburg, Germany
| | - Axel Seher
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany
| | - Alexander C Kübler
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany
| | - Urs D A Müller-Richter
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany
| |
Collapse
|
32
|
Koshizuka K, Kikkawa N, Hanazawa T, Yamada Y, Okato A, Arai T, Katada K, Okamoto Y, Seki N. Inhibition of integrin β1-mediated oncogenic signalling by the antitumor microRNA-29 family in head and neck squamous cell carcinoma. Oncotarget 2017; 9:3663-3676. [PMID: 29423074 PMCID: PMC5790491 DOI: 10.18632/oncotarget.23194] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/01/2017] [Indexed: 12/28/2022] Open
Abstract
Due to their aggressive behavior, local recurrence and distant metastasis, survival rate of advanced stage of the patients with head and neck squamous cell carcinoma (HNSCC) is very poor. Currently available epidermal growth factor receptor (EGFR)-targeted therapies are not considered curative for HNSCC. Therefore, novel approaches for identification of therapeutic targets in HNSCC are needed. All members of the miRNA-29 family (miR-29a, miR-29b, and miR-29c) were downregulated in HNSCC tissues by analysis of RNA-sequencing based microRNA (miRNA) expression signature. Ectopic expression of mature miRNAs demonstrated that the miR-29 family inhibited cancer cell migration and invasion by HNSCC cell lines. Comprehensive gene expression studies and in silico database analyses were revealed that integrin β1 (ITGB1) was regulated by the miR-29 family in HNSCC cells. Overexpression of ITGB1 was confirmed in HNSCC specimens, and high expression of ITGB1 significantly predicted poor survival in patients with HNSCC (p = 0.00463). Knockdown of ITGB1 significantly inhibited cancer cell migration and invasion through regulating downstream of ITGB1-mediated oncogenic signalling. In conclusion, regulation of the antitumor miR-29 family affected integrin-mediated oncogenic signalling to modulate HNSCC pathogenesis; these molecules may be novel therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Keiichi Koshizuka
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan.,Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naoko Kikkawa
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Toyoyuki Hanazawa
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yasutaka Yamada
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | - Atsushi Okato
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | - Takayuki Arai
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | - Koji Katada
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naohiko Seki
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| |
Collapse
|
33
|
Boduc M, Roessler M, Mandic R, Netzer C, Güldner C, Walliczek-Dworschak U, Stuck BA, Mandapathil M. Foxp3 expression in lymph node metastases in patients with head and neck cancer. Acta Otolaryngol 2017; 137:1215-1219. [PMID: 28741409 DOI: 10.1080/00016489.2017.1353705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The prevalence and activity of regulatory T cells in patients with cancer correlates with poor prognosis. These cells are characterized by their expression of Forkhead box protein-3 (Foxp3). Squamous cell carcinoma is the most prevalent type of cancer in the head and neck region with overall poor survival rates, also due to early spread of metastatic cells. MATERIAL AND METHODS Primary tumor specimens as well as lymph node specimens harvested during neck dissection of 65 patients with a diagnosis of HNSCC were subjected to immunohistochemical and H-score analysis of Foxp3 expression. Demographics, diagnoses, histopathology and subsequent outcome were analyzed. RESULTS The primary cancer was squamous cell carcinoma in all patients (male/female 55:10) with the following tumor locations: oral cavity n = 16, oropharynx n = 28, hypopharynx n = 11 and larynx n = 10 (Stage III n = 18; Stage IVA n = 45; Stage IVB n = 2). The H-score for Foxp3 expression in the primary lesion as well as metastatic lymph nodes was significantly higher in advanced stages compared to early stages with differences among tumor locations, which were not significant. High Foxp3 expression was associated with inferior overall survival rates at a mean follow-up of 83.4 months (6-204 months) Conclusions: Foxp3 expression in HNSCC varied from the anatomical site and correlated positively with tumor stage and was associated with poor prognosis. Therefore, Foxp3 expressions in primary lesions as well as lymphogenic metastases appear to predict high-risk HSNCC patients. Novel therapeutic approaches targeting Foxp3+ cells might seem promising for this patient population.
Collapse
Affiliation(s)
- Mehtap Boduc
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-University Marburg, Marburg, Germany
| | - Marion Roessler
- Department of Pathology, Philipps-University Marburg, Marburg, Germany
| | - Robert Mandic
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-University Marburg, Marburg, Germany
| | - Christoph Netzer
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-University Marburg, Marburg, Germany
| | - Christian Güldner
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-University Marburg, Marburg, Germany
| | - Ute Walliczek-Dworschak
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-University Marburg, Marburg, Germany
| | - Boris A. Stuck
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-University Marburg, Marburg, Germany
| | - Magis Mandapathil
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-University Marburg, Marburg, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Asklepios Clinic St. Georg, Hamburg, Germany
| |
Collapse
|
34
|
Zheng Y, Su C, Zhao L, Shi Y. mAb MDR1-modified chitosan nanoparticles overcome acquired EGFR-TKI resistance through two potential therapeutic targets modulation of MDR1 and autophagy. J Nanobiotechnology 2017; 15:66. [PMID: 28978341 PMCID: PMC5628454 DOI: 10.1186/s12951-017-0302-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/23/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) that act against the epithelial growth factor receptor (EGFR) were once widely used in chemotherapy for many human cancers. However, acquired chemoresistance occurred in almost all patients, limiting the clinical application of EGFR-TKI. Thus far, no effective methods existing can resolve this problem. Designing a therapeutic treatment with a specific multi-target profile has been regarded as a possible strategy to overcome acquired EGFR-TKI resistance. METHODS MDR1 antibody-modified chitosan nanoparticles loading gefitinib and autophagy inhibitor chloroquine were prepared by ionic crosslinking and electrostatic attracting method. MTT assay, flow cytometry analysis and western blot assay were all performed to confirm the effect of different formulations of gefitinib on the proliferation of SMMC-7721/gefitinib cells. The preparations demonstrated their multi-target potential to achieve both tumor-targeting selectivity and the desired antitumor effects by blocking cell-surface MDR1 and inhibiting autophagy. RESULTS mAb MDR1-modified CS NPs, when combined with the co-delivery of gefitinib and chloroquine, showed targeting and therapeutic potential on enhancing the delivery of anticancer drugs and inducing significant cell apoptosis against acquired EGFR-TKI resistance through the modulation of autophagy and while blocking the activity of the MDR1 receptor. CONCLUSIONS A new approach to design an excellent nanoparticle drug-delivery system can overcome acquired EGFR-TKI resistance against various multiple antitumor targets.
Collapse
Affiliation(s)
- Yan Zheng
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Chang Su
- School of Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, People's Republic of China.
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, People's Republic of China.
| |
Collapse
|
35
|
Cheung RS, Taniguchi T. Recent insights into the molecular basis of Fanconi anemia: genes, modifiers, and drivers. Int J Hematol 2017; 106:335-344. [PMID: 28631178 PMCID: PMC5904331 DOI: 10.1007/s12185-017-2283-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 06/14/2017] [Indexed: 02/06/2023]
Abstract
Fanconi anemia (FA), the most common form of inherited bone marrow failure, predisposes to leukemia and solid tumors. FA is caused by the genetic disruption of a cellular pathway that repairs DNA interstrand crosslinks. The impaired function of this pathway, and the genetic instability that results, is considered the main pathogenic mechanism behind this disease. The identification of breast cancer susceptibility genes (for example, BRCA1/FANCS and BRCA2/FANCD1) as being major players in the FA pathway has led to a surge in molecular studies, resulting in the concept of the FA-BRCA pathway. In this review, we discuss recent advances in the molecular pathogenesis of FA from three viewpoints: (a) new FA genes, (b) modifier pathways that influence the cellular and clinical phenotypes of FA and (c) non-canonical functions of FA genes that may drive disease progression independently of deficient DNA repair. Potential therapeutic approaches for FA that are relevant to each will also be proposed.
Collapse
Affiliation(s)
- Ronald S Cheung
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., C1-015, Seattle, WA, 98109-1024, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., C1-015, Seattle, WA, 98109-1024, USA
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., C1-015, Seattle, WA, 98109-1024, USA
| | - Toshiyasu Taniguchi
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., C1-015, Seattle, WA, 98109-1024, USA.
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., C1-015, Seattle, WA, 98109-1024, USA.
- Department of Molecular Life Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
| |
Collapse
|