1
|
Liu J, Ouyang Y, Xia Z, Mai W, Song H, Zhou F, Shen L, Chen K, Li X, Zhuang SM, Liao J. FAP is a prognostic marker, but not a viable therapeutic target for clinical translation in HNSCC. Cell Oncol (Dordr) 2024; 47:623-638. [PMID: 37856075 DOI: 10.1007/s13402-023-00888-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2023] [Indexed: 10/20/2023] Open
Abstract
PURPOSE PD-1 targeted immunotherapy has imparted a survival benefit to advanced head and neck squamous cell carcinoma (HNSCC), but less than 20% patients produce a durable response to this therapy. Here we aimed to investigate the potential biomarkers for predicting the clinical outcome and resistance to PD-1 targeted immunotherapy in HNSCC patients, and to examine the involvement of FAP+ cancer-associated fibroblasts (CAFs). METHODS Bioinformatics methods were applied to analyze multiple datasets and explore the role of PD-1 and FAP in HNSCC. Immunohistochemistry was used to detect the expression of FAP protein. Fap gene knockout mice (Fap-/-) and L929 cells with different levels of Fap overexpression (L929-Fap-Low/High) were established to demonstrate the role of FAP+ CAFs in tumor development and immune checkpoint blockade (ICB) resistance. RESULTS The expression level of PD-1 gene was positively correlated with better overall survival and therapeutic response to PD-1 blockade in HNSCC, but not all tumors with high expression of both PD-1 and PD-L1 were responsive. Moreover, FAP gene was overexpressed in pan-cancer tissues, and could serve as a prognostic biomarker for several cancers, including HNSCC. However, FAP protein was undetectable in mouse MTCQ1 tumors and barely expressed in human HNSCC tumors. Furthermore, FAP+ CAFs did not promote tumor growth or enhance the resistance to PD-1 inhibitor treatment. CONCLUSION Although FAP+ CAFs have attracted increasing attention for their role in cancer, the feasibility and efficacy of FAP-targeting therapies for HNSCC remain doubtful.
Collapse
Affiliation(s)
- Jie Liu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
- Department of General Surgery of Otorhinolaryngology Head and Neck, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangzhou, China
| | - Yeling Ouyang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, China
| | - Zijin Xia
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wenhao Mai
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Hongrui Song
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Fang Zhou
- Central Sterile Supply Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lichun Shen
- Department of General Surgery of Otorhinolaryngology Head and Neck, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kaiting Chen
- Department of General Surgery of Otorhinolaryngology Head and Neck, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaochen Li
- Department of General Surgery of Otorhinolaryngology Head and Neck, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shi-Min Zhuang
- Department of General Surgery of Otorhinolaryngology Head and Neck, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangzhou, China.
| | - Jing Liao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Chen HH, Yu HI, Chang JJS, Li CW, Yang MH, Hung MC, Tarn WY. DDX3 regulates cancer immune surveillance via 3' UTR-mediated cell-surface expression of PD-L1. Cell Rep 2024; 43:113937. [PMID: 38489268 DOI: 10.1016/j.celrep.2024.113937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Programmed death-1 (PD-1)/PD ligand-1 (PD-L1)-mediated immune escape contributes to cancer development and has been targeted as an anti-cancer strategy. Here, we show that inhibition of the RNA helicase DDX3 increased CD8+ T cell infiltration in syngeneic oral squamous cell carcinoma tumors. DDX3 knockdown compromised interferon-γ-induced PD-L1 expression and, in particular, reduced the level of cell-surface PD-L1. DDX3 promoted surface PD-L1 expression by recruiting the adaptor protein 2 (AP2) complex to the 3' UTR of PD-L1 mRNA. DDX3 depletion or 3' UTR truncation increased the binding of the coatomer protein complexes to PD-L1, leading to its intracellular accumulation. Therefore, this 3' UTR-dependent mechanism may counteract cellular negative effects on surface trafficking of PD-L1. Finally, pharmaceutic disruption of DDX3's interaction with AP2 reduced surface PD-L1 expression, supporting that the DDX3-AP2 pathway routes PD-L1 to the cell surface. Targeting DDX3 to modulate surface trafficking of immune checkpoint proteins may provide a potential strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Hung-Hsi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsin-I Yu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Chia-Wei Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chao-Tung University, Taipei, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Woan-Yuh Tarn
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
3
|
Su NW, Dai SH, Hsu K, Chang KM, Ko CC, Kao CW, Chang YF, Chen CG. PD-L1-positive circulating endothelial progenitor cells associated with immune response to PD-1 blockade in patients with head and neck squamous cell carcinoma. Cancer Immunol Immunother 2024; 73:3. [PMID: 38175307 DOI: 10.1007/s00262-023-03595-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024]
Abstract
A number of the inhibitors against programmed death protein 1 (PD-1) have been approved to treat recurrent or metastatic squamous cell carcinoma of head and neck (HNSCC). The interaction between PD-1 and its ligand (PD-L1) serves as an immune checkpoint that governs cytotoxic immune effectors against tumors. Numerous clinical trials of PD-1/PD-L1 inhibitors have so far been discordant about having sufficient PD-L1 expression in the tumor as a prerequisite for a successful anti-PD-1 treatment. On the other hand, vascular endothelial cells modulate immune activities through PD-L1 expression, and thus it is possible that the expressions of circulating endothelial cells (CECs) and circulating endothelial progenitor cells (CPCs) could affect antitumor immunity as well as neoangiogenesis. Here we investigated the potential involvement of PD-L1+ CECs and PD-L1+ CPCs in PD-1 blockade treatments for HNSCC patients. We measured CD8+ T cells, CECs, and CPCs in the peripheral blood of the HNSCC patients treated by anti-PD-1 therapies. We found that their PD-L1+ CPC expression before anti-PD1 therapies was strongly correlated with treatment responses and overall survival. Moreover, if the first infusion of PD-1 inhibitors reduced ≥ 50% PD-L1+ CPCs, a significantly better outcome could be predicted. In these patients as well as in an animal model of oral cancer, Pd-l1+ CPC expression was associated with limited CD8+ T-cell infiltration into the tumors, and anti-PD-1 treatments also targeted Pd-l1+ CPCs and increased CD8+ T-cell infiltration. Our results highlight PD-L1+ CPC as a potential regulator in the anti-PD-1 treatments for HNSCC.
Collapse
Affiliation(s)
- Nai-Wen Su
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei, 25245, Taiwan
| | - Shuen-Han Dai
- Department of Pathology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Kate Hsu
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei, 25245, Taiwan
- Department of Medicine, Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, 25245, Taiwan
- The Immunogenetics Laboratory, Department of Medical Research, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
| | - Kuo-Ming Chang
- Department of Pathology and Laboratory Medicine, MacKay Memorial Hospital, Hsinchu, 35071, Taiwan
| | - Chun-Chuan Ko
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, 25160, Taiwan
| | - Chen-Wei Kao
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
| | - Yi-Fang Chang
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
| | - Caleb G Chen
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan.
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei, 25245, Taiwan.
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan.
- Institute of Molecular Medicine, National Tsing-Hua University, Hsin-Chu, Taiwan.
| |
Collapse
|
4
|
Lin Y, Chen CY, Ku YC, Wang LC, Hung CC, Lin ZQ, Chen BH, Hung JT, Sun YC, Hung KF. A modified SELEX approach to identify DNA aptamers with binding specificity to the major histocompatibility complex presenting ovalbumin model antigen. RSC Adv 2023; 13:32681-32693. [PMID: 37936644 PMCID: PMC10626974 DOI: 10.1039/d3ra04686a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/04/2023] [Indexed: 11/09/2023] Open
Abstract
Aptamers have sparked significant interest in cell recognition because of their superior binding specificity and biocompatibility. Cell recognition can be mediated by targeting the major histocompatibility complex (MHC) that presents short peptides derived from intracellular antigens. Although numerous antibodies have demonstrated a specific affinity for the peptide-MHC complex, the number of aptamers that exhibit comparable characteristics is limited. Aptamers are usually selected from large libraries via the Systemic Evolution of Ligands by Exponential Enrichment (SELEX), an iterative process of selection and PCR amplification to enrich a pool of aptamers with high affinity. However, the success rate of aptamer identification is low, possibly due to the presence of complementary sequences or sequences rich in guanine and cytosine that are less accessible for primers. Here, we modified SELEX by employing systemic consecutive selections with minimal PCR amplification. We also modified the analysis by selecting aptamers that were identified in multiple selection rounds rather than those that are highly enriched. Using this approach, we were able to identify two aptamers with binding specificity to cells expressing the ovalbumin alloantigen as a proof of concept. These two aptamers were also discovered among the top 150 abundant candidates, despite not being highly enriched, by performing conventional SELEX. Additionally, we found that highly enriched aptamers tend to contain fractions of the primer sequence and have minimal target affinity. Candidate aptamers are easily missed in the conventional SELEX process. Therefore, our modification for SELEX may facilitate the identification of aptamers for more application in diverse biomedical fields. Significance: we modify the conventional method to improve the efficiency in the identification of the aptamer, a single strand of nucleic acid with binding specificity to the target molecule, showing as a proof of concept that this approach is particularly useful to select aptamers that can selectively bind to cells presenting a particular peptide by the major histocompatibility complex (MHC) on the cell surface. Given that cancer cells may express mutant peptide-MHC complexes that are distinct from those expressed by normal cells, this study sheds light on the potential application of aptamers to cancer cell targeting.
Collapse
Affiliation(s)
- Yang Lin
- Department of Medical Research, Taipei Veterans General Hospital 201, Section 2, Shi-Pai Road Taipei 112 Taiwan +886-2-28712121-7382
| | - Cho-Yi Chen
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University Taipei Taiwan
| | - Yu-Chia Ku
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University Taipei Taiwan
| | - Li-Chin Wang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University Taipei Taiwan
| | - Chia-Chien Hung
- School of Computer Science, Georgia Institute of Technology Atlanta GA USA
| | - Zhi-Qian Lin
- Department of Medical Research, Taipei Veterans General Hospital 201, Section 2, Shi-Pai Road Taipei 112 Taiwan +886-2-28712121-7382
| | - Bing-Hong Chen
- Department of Medical Research, Taipei Veterans General Hospital 201, Section 2, Shi-Pai Road Taipei 112 Taiwan +886-2-28712121-7382
| | | | - Yi-Chen Sun
- School of Medicine, Tzu-Chi University Hualien Taiwan
- Department of Ophthalmology, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation New Taipei City Taiwan
| | - Kai-Feng Hung
- Department of Medical Research, Taipei Veterans General Hospital 201, Section 2, Shi-Pai Road Taipei 112 Taiwan +886-2-28712121-7382
- Department of Dentistry, School of Dentistry, National Yang Ming Chiao Tung University Taipei Taiwan
| |
Collapse
|
5
|
Zaryouh H, De Pauw I, Baysal H, Melis J, Van den Bossche V, Hermans C, Lau HW, Lambrechts H, Merlin C, Corbet C, Peeters M, Vermorken JB, De Waele J, Lardon F, Wouters A. Establishment of head and neck squamous cell carcinoma mouse models for cetuximab resistance and sensitivity. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:709-728. [PMID: 38239393 PMCID: PMC10792481 DOI: 10.20517/cdr.2023.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/07/2023] [Accepted: 10/10/2023] [Indexed: 01/22/2024]
Abstract
Aim: Acquired resistance to the targeted agent cetuximab poses a significant challenge in finding effective anti-cancer treatments for head and neck squamous cell carcinoma (HNSCC). To accurately study novel combination treatments, suitable preclinical mouse models for cetuximab resistance are key yet currently limited. This study aimed to optimize an acquired cetuximab-resistant mouse model, with preservation of the innate immunity, ensuring intact antibody-dependent cellular cytotoxicity (ADCC) functionality. Methods: Cetuximab-sensitive and acquired-resistant HNSCC cell lines, generated in vitro, were subcutaneously engrafted in Rag2 knock-out (KO), BALB/c Nude and CB17 Scid mice with/without Matrigel or Geltrex. Once tumor growth was established, mice were intraperitoneally injected twice a week with cetuximab for a maximum of 3 weeks. In addition, immunohistochemistry was used to evaluate the tumor and its microenvironment. Results: Despite several adjustments in cell number, cell lines and the addition of Matrigel, Rag2 KO and BALB/C Nude mice proved to be unsuitable for xenografting our HNSCC cell lines. Durable tumor growth of resistant SC263-R cells could be induced in CB17 Scid mice. However, these cells had lost their resistance phenotype in vivo. Immunohistochemistry revealed a high infiltration of macrophages in cetuximab-treated SC263-R tumors. FaDu-S and FaDu-R cells successfully engrafted into CB17 Scid mice and maintained their sensitivity/resistance to cetuximab. Conclusion: We have established in vivo HNSCC mouse models with intact ADCC functionality for cetuximab resistance and sensitivity using the FaDu-R and FaDu-S cell lines, respectively. These models serve as valuable tools for investigating cetuximab resistance mechanisms and exploring novel drug combination strategies.
Collapse
Affiliation(s)
- Hannah Zaryouh
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Ines De Pauw
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Hasan Baysal
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Jöran Melis
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Valentin Van den Bossche
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels B-1200, Belgium
- Institut Roi Albert II, Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Brussels B-1200, Belgium
| | - Christophe Hermans
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Ho Wa Lau
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Hilde Lambrechts
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Céline Merlin
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels B-1200, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
- Department of Medical Oncology, Antwerp University Hospital, Edegem 2650, Belgium
| | - Jan Baptist Vermorken
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
- Department of Medical Oncology, Antwerp University Hospital, Edegem 2650, Belgium
| | - Jorrit De Waele
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
- The authors contributed equally
| | - An Wouters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Antwerp 2610, Belgium
- The authors contributed equally
| |
Collapse
|
6
|
Yassin-Kassab A, Wang N, Foley J, Stewart TM, Burns MR, Casero RA, Harbison RA, Duvvuri U. Polyamine transport inhibition and cisplatin synergistically enhance tumor control through oxidative stress in murine head and neck cancer models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550524. [PMID: 37546993 PMCID: PMC10402081 DOI: 10.1101/2023.07.25.550524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background Surgery and/or platinum-based chemoradiation remain standard of care for patients with head and neck squamous cell carcinoma (HNSCC). While these therapies are effective in a subset of patients, a substantial proportion experience recurrence or treatment resistance. As cisplatin mediates cytotoxicity through oxidative stress while polyamines play a role in redox regulation, we posited that combining cisplatin with polyamine transport inhibitor, AMXT-1501, would increase oxidative stress and tumor cell death in HNSCC cells. Methods Cell proliferation was measured in syngeneic mouse HNSCC cell lines treated with cisplatin ± AMXT-1501. Synergy was determined by administering cisplatin and AMXT-1501 at a ratio of 1:10 to cancer cells in vitro . Cancer cells were transferred onto mouse flanks to test the efficacy of treatments in vivo . Reactive oxygen species (ROS) were measured. Cellular apoptosis was measured with flow cytometry using Annexin V/PI staining. High-performance liquid chromatography (HPLC) was used to quantify polyamines in cell lines. Cell viability and ROS were measured in the presence of exogenous cationic amino acids. Results The combination of cisplatin and AMXT-1501 synergize in vitro on HNSCC cell lines. In vivo combination treatment resulted in tumor growth inhibition greater than either treatment individually. The combination treatment increased ROS production and induced apoptotic cell death. HPLC revealed the synergistic mechanism was independent of intracellular polyamine levels. Supplementation of cationic amino acids partially rescued cancer cell viability and reduced ROS. Conclusion AMXT-1501 enhances the cytotoxic effects of cisplatin in vitro and in vivo in aggressive HNSCC cell lines through a polyamine-independent mechanism.
Collapse
|
7
|
Chung CH, Lin CY, Chen CY, Hsueh CW, Chang YW, Wang CC, Chu PY, Tai SK, Yang MH. Ferroptosis Signature Shapes the Immune Profiles to Enhance the Response to Immune Checkpoint Inhibitors in Head and Neck Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204514. [PMID: 37026630 DOI: 10.1002/advs.202204514] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 01/23/2023] [Indexed: 05/27/2023]
Abstract
As a type of immunogenic cell death, ferroptosis participates in the creation of immunoactive tumor microenvironments. However, knowledge of spatial location of tumor cells with ferroptosis signature in tumor environments and the role of ferroptotic stress in inducing the expression of immune-related molecules in cancer cells is limited. Here the spatial association of the transcriptomic signatures is demonstrated for ferroptosis and inflammation/immune activation located in the invasive front of head and neck squamous cell carcinoma (HNSCC). The association between ferroptosis signature and inflammation/immune activation is more prominent in HPV-negative HNSCC compared to HPV-positive ones. Ferroptotic stress induces PD-L1 expression through reactive oxygen species (ROS)-elicited NF-κB signaling pathway and calcium influx. Priming murine HNSCC with the ferroptosis inducer sensitizes tumors to anti-PD-L1 antibody treatment. A positive correlation between the ferroptosis signature and the active immune cell profile is shown in the HNSCC samples. This study reveals a subgroup of ferroptotic HNSCC with immune-active signatures and indicates the potential of priming HNSCC with ferroptosis inducers to increase the antitumor efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Chih-Hung Chung
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 115201, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Chun-Yu Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
- Institute of Data Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
- School of Dentistry, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Chih-Yu Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Chun-Wei Hsueh
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Yao-Wen Chang
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Chen-Chi Wang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Otolaryngology Head & Neck Surgery, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Pen-Yuan Chu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Otolaryngology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
| | - Shyh-Kuan Tai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Otolaryngology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
| | - Muh-Hwa Yang
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 115201, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
| |
Collapse
|
8
|
Chiu WC, Ou DL, Tan CT. Mouse Models for Immune Checkpoint Blockade Therapeutic Research in Oral Cancer. Int J Mol Sci 2022; 23:ijms23169195. [PMID: 36012461 PMCID: PMC9409124 DOI: 10.3390/ijms23169195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The most prevalent oral cancer globally is oral squamous cell carcinoma (OSCC). The invasion of adjacent bones and the metastasis to regional lymph nodes often lead to poor prognoses and shortened survival times in patients with OSCC. Encouraging immunotherapeutic responses have been seen with immune checkpoint inhibitors (ICIs); however, these positive responses to monotherapy have been limited to a small subset of patients. Therefore, it is urgent that further investigations into optimizing immunotherapies are conducted. Areas of research include identifying novel immune checkpoints and targets and tailoring treatment programs to meet the needs of individual patients. Furthermore, the advancement of combination therapies against OSCC is also critical. Thus, additional studies are needed to ensure clinical trials are successful. Mice models are advantageous in immunotherapy research with several advantages, such as relatively low costs and high tumor growth success rate. This review paper divided methods for establishing OSCC mouse models into four categories: syngeneic tumor models, chemical carcinogen induction, genetically engineered mouse, and humanized mouse. Each method has advantages and disadvantages that influence its application in OSCC research. This review comprehensively surveys the literature and summarizes the current mouse models used in immunotherapy, their advantages and disadvantages, and details relating to the cell lines for oral cancer growth. This review aims to present evidence and considerations for choosing a suitable model establishment method to investigate the early diagnosis, clinical treatment, and related pathogenesis of OSCC.
Collapse
Affiliation(s)
- Wei-Chiao Chiu
- Department of Medical Research, Fu-Jen Catholic University Hospital, Fu-Jen Catholic University, New Taipei City 24352, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei City 100225, Taiwan
| | - Da-Liang Ou
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei City 10051, Taiwan
- YongLin Institute of Health, National Taiwan University, Taipei City 10672, Taiwan
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei City 100225, Taiwan
- Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei City 10051, Taiwan
- Department of Otolaryngology, College of Medicine, National Taiwan University, Taipei City 100233, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302058, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 88649)
| |
Collapse
|
9
|
Shi Y, Xie T, Wang B, Wang R, Cai Y, Yuan B, Gleber-Netto FO, Tian X, Rodriguez-Rosario AE, Osman AA, Wang J, Pickering CR, Ren X, Sikora AG, Myers JN, Rangel R. Mutant p53 drives an immune cold tumor immune microenvironment in oral squamous cell carcinoma. Commun Biol 2022; 5:757. [PMID: 35902768 PMCID: PMC9334280 DOI: 10.1038/s42003-022-03675-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 07/06/2022] [Indexed: 02/03/2023] Open
Abstract
The critical role of the tumor immune microenvironment (TIME) in determining response to immune checkpoint inhibitor (ICI) therapy underscores the importance of understanding cancer cell-intrinsic mechanisms driving immune-excluded ("cold") TIMEs. One such cold tumor is oral cavity squamous cell carcinoma (OSCC), a tobacco-associated cancer with mutations in the TP53 gene which responds poorly to ICI therapy. Because altered TP53 function promotes tumor progression and plays a potential role in TIME modulation, here we developed a syngeneic OSCC models with defined Trp53 (p53) mutations and characterized their TIMEs and degree of ICI responsiveness. We observed that a carcinogen-induced p53 mutation promoted a cold TIME enriched with immunosuppressive M2 macrophages highly resistant to ICI therapy. p53-mutated cold tumors failed to respond to combination ICI treatment; however, the combination of a programmed cell death protein 1 (PD-1) inhibitor and stimulator of interferon genes (STING) agonist restored responsiveness. These syngeneic OSCC models can be used to gain insights into tumor cell-intrinsic drivers of immune resistance and to develop effective immunotherapeutic approaches for OSCC and other ICI-resistant solid tumors.
Collapse
Affiliation(s)
- Yewen Shi
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA ,grid.452672.00000 0004 1757 5804Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Tongxin Xie
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Bingbing Wang
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Rong Wang
- grid.49470.3e0000 0001 2331 6153Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu Cai
- grid.49470.3e0000 0001 2331 6153Department of Oral & Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bo Yuan
- grid.240145.60000 0001 2291 4776Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Frederico O. Gleber-Netto
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Xiangjun Tian
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Alanis E. Rodriguez-Rosario
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA ,grid.449853.70000 0001 2051 0540Department of Biology, University of Puerto Rico, Bayamon, Puerto Rico USA
| | - Abdullah A. Osman
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Jing Wang
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Curtis R. Pickering
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Xiaoyong Ren
- grid.452672.00000 0004 1757 5804Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Andrew G. Sikora
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Jeffrey N. Myers
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Roberto Rangel
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| |
Collapse
|
10
|
Chen HH, Yu HI, Rudy R, Lim SL, Chen YF, Wu SH, Lin SC, Yang MH, Tarn WY. DDX3 modulates the tumor microenvironment via its role in endoplasmic reticulum-associated translation. iScience 2021; 24:103086. [PMID: 34568799 PMCID: PMC8449240 DOI: 10.1016/j.isci.2021.103086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/15/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Using antibody arrays, we found that the RNA helicase DDX3 modulates the expression of secreted signaling factors in oral squamous cell carcinoma (OSCC) cells. Ribo-seq analysis confirmed amphiregulin (AREG) as a translational target of DDX3. AREG exerts important biological functions in cancer, including promoting cell migration and paracrine effects of OSCC cells and reprogramming the tumor microenvironment (TME) of OSCC in mice. DDX3-mediated translational control of AREG involves its 3′-untranslated region. Proteomics identified the signal recognition particle (SRP) as an unprecedented interacting partner of DDX3. DDX3 and SRP54 were located near the endoplasmic reticulum, regulated the expression of a common set of secreted factors, and were essential for targeting AREG mRNA to membrane-bound polyribosomes. Finally, OSCC-associated mutant DDX3 increased the expression of AREG, emphasizing the role of DDX3 in tumor progression via SRP-dependent, endoplasmic reticulum-associated translation. Therefore, pharmacological targeting of DDX3 may inhibit the tumor-promoting functions of the TME. DDX3-AREG axis promotes cancer progression through microenvironment remodeling DDX3 activates AREG translation via binding to its 3′ UTR DDX3 interacts with the signal recognition particle (SRP) DDX3-SRP-mediated mRNA recruitment assists ER-associated translation
Collapse
Affiliation(s)
- Hung-Hsi Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Academy Road Section 2, Nankang, Taipei 11529, Taiwan
| | - Hsin-I Yu
- Institute of Biomedical Sciences, Academia Sinica, 128 Academy Road Section 2, Nankang, Taipei 11529, Taiwan
| | - Rudy Rudy
- Institute of Biomedical Sciences, Academia Sinica, 128 Academy Road Section 2, Nankang, Taipei 11529, Taiwan
| | - Sim-Lin Lim
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Fen Chen
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Shu-Hsing Wu
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Shu-Chun Lin
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Woan-Yuh Tarn
- Institute of Biomedical Sciences, Academia Sinica, 128 Academy Road Section 2, Nankang, Taipei 11529, Taiwan
| |
Collapse
|
11
|
Spenlé C, Loustau T, Burckel H, Riegel G, Abou Faycal C, Li C, Yilmaz A, Petti L, Steinbach F, Ahowesso C, Jost C, Paul N, Carapito R, Noël G, Anjuère F, Salomé N, Orend G. Impact of Tenascin-C on Radiotherapy in a Novel Syngeneic Oral Squamous Cell Carcinoma Model With Spontaneous Dissemination to the Lymph Nodes. Front Immunol 2021; 12:636108. [PMID: 34290694 PMCID: PMC8287883 DOI: 10.3389/fimmu.2021.636108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/11/2021] [Indexed: 12/05/2022] Open
Abstract
Radiotherapy, the most frequent treatment of oral squamous cell carcinomas (OSCC) besides surgery is employed to kill tumor cells but, radiotherapy may also promote tumor relapse where the immune-suppressive tumor microenvironment (TME) could be instrumental. We established a novel syngeneic grafting model from a carcinogen-induced tongue tumor, OSCC13, to address the impact of radiotherapy on OSCC. This model revealed similarities with human OSCC, recapitulating carcinogen-induced mutations found in smoking associated human tongue tumors, abundant tumor infiltrating leukocytes (TIL) and, spontaneous tumor cell dissemination to the local lymph nodes. Cultured OSCC13 cells and OSCC13-derived tongue tumors were sensitive to irradiation. At the chosen dose of 2 Gy mimicking treatment of human OSCC patients not all tumor cells were killed allowing to investigate effects on the TME. By investigating expression of the extracellular matrix molecule tenascin-C (TNC), an indicator of an immune suppressive TME, we observed high local TNC expression and TIL infiltration in the irradiated tumors. In a TNC knockout host the TME appeared less immune suppressive with a tendency towards more tumor regression than in WT conditions. Altogether, our novel syngeneic tongue OSCC grafting model, sharing important features with the human OSCC disease could be relevant for future anti-cancer targeting of OSCC by radiotherapy and other therapeutic approaches.
Collapse
Affiliation(s)
- Caroline Spenlé
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Thomas Loustau
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| | - Hélène Burckel
- Institut de Cancérologie de Strasbourg Europe (ICANS), UNICANCER, Paul Strauss Comprehensive Cancer Center, Radiobiology Laboratory, Université de Strasbourg, Strasbourg, France
| | - Gilles Riegel
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| | - Chérine Abou Faycal
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| | - Chengbei Li
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| | - Alev Yilmaz
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| | - Luciana Petti
- Université Côte d’Azur, CNRS, IPMC, Valbonne-Sophia Antipolis, France
| | - Fanny Steinbach
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| | - Constance Ahowesso
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Camille Jost
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Nicodème Paul
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Platform GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, LabEx TRANSPLANTEX, Strasbourg, France
| | - Raphael Carapito
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Platform GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, LabEx TRANSPLANTEX, Strasbourg, France
| | - Georges Noël
- Institut de Cancérologie de Strasbourg Europe (ICANS), UNICANCER, Paul Strauss Comprehensive Cancer Center, Radiobiology Laboratory, Université de Strasbourg, Strasbourg, France
- Institut de Cancérologie Strasbourg Europe (ICANS), UNICANCER, Department of Radiation Oncology, Strasbourg, France
| | - Fabienne Anjuère
- Université Côte d’Azur, CNRS, IPMC, Valbonne-Sophia Antipolis, France
| | - Nathalie Salomé
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| | - Gertraud Orend
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, Strasbourg, France
| |
Collapse
|
12
|
Tsang O, Wong JWH. Proteogenomic interrogation of cancer cell lines: an overview of the field. Expert Rev Proteomics 2021; 18:221-232. [PMID: 33877947 DOI: 10.1080/14789450.2021.1914594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Cancer cell lines (CCLs) have been a major resource for cancer research. Over the past couple of decades, they have been instrumental in omic profiling method development and as model systems to generate new knowledge in cell and cancer biology. More recently, with the increasing amount of genomic, transcriptomic and proteomic data being generated in hundreds of CCLs, there is growing potential for integrative proteogenomic data analyses to be performed.Areas covered: In this review, we first describe the most commonly used proteome profiling methods in CCLs. We then discuss how these proteomics data can be integrated with genomics data for proteogenomics analyses. Finally, we highlight some of the recent biological discoveries that have arisen from proteogenomics analyses of CCLs.Expert opinion: Protegeonomics analyses of CCLs have so far enabled the discovery of novel proteins and proteoforms. It has also improved our understanding of biological processes including post-transcriptional regulation of protein abundance and the presentation of antigens by major histocompatibility complex alleles. With proteomics data to be generated in hundreds to thousands of CCLs in coming years, there will be further potential for large-scale proteogenomics analyses and data integration with the phenotypically well-characterized CCLs.
Collapse
Affiliation(s)
- Olson Tsang
- Centre for PanorOmic Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Jason W H Wong
- Centre for PanorOmic Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR.,School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR
| |
Collapse
|
13
|
Sagheer SH, Whitaker-Menezes D, Han JYS, Curry JM, Martinez-Outschoorn U, Philp NJ. 4NQO induced carcinogenesis: A mouse model for oral squamous cell carcinoma. Methods Cell Biol 2021; 163:93-111. [PMID: 33785171 DOI: 10.1016/bs.mcb.2021.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common subsite of head and neck cancer, with a 5-year survival rate of only 50%. There is a pressing need for animal models that recapitulate the human disease to understand the factors driving OSCC carcinogenesis. Many laboratories have used the chemical carcinogen 4-nitroquinoline-1-oxide (4NQO) to investigate OSCC formation. The importance of the 4NQO mouse model is that it mimics the stepwise progression observed in OSCC patients. The 4NQO carcinogen model has the advantage that it can be used with transgenic mice with genetic modification in specific tissue types to investigate their role in driving cancer progression. Herein, we describe the basic approach for administering 4NQO to mice to induce OSCC and methods for assessing the tissue and disease progression.
Collapse
Affiliation(s)
- S Hamad Sagheer
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Diana Whitaker-Menezes
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - John Y S Han
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Joseph M Curry
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, PA, United States.
| | | | - Nancy J Philp
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
14
|
Chang KW, Lin CE, Tu HF, Chung HY, Chen YF, Lin SC. Establishment of a p53 Null Murine Oral Carcinoma Cell Line and the Identification of Genetic Alterations Associated with This Carcinoma. Int J Mol Sci 2020; 21:ijms21249354. [PMID: 33302499 PMCID: PMC7764333 DOI: 10.3390/ijms21249354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/27/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), including oral squamous cell carcinoma (OSCC), ranks sixth in cancer incidence worldwide. To generate OSCC cells lines from human or murine tumors, greatly facilitates investigations into OSCC. This study describes the establishing of a mouse palatal carcinoma cell line (designated MPC-1) from a spontaneous tumor present in a heterozygous p53 gene loss C57BL/6 mouse. A MPC-1-GFP cell subclone was then generated by lentivirus infection resulting in stable expression of green fluorescent protein. Assays indicated that MPC-1 was a p53 null polygonal cell that was positive for keratinocyte markers; it also expressed vimentin and showed a loss of E-cadherin expression. Despite that MPC-1 having strong proliferation and colony formation capabilities, the potential for anchorage independent growth and tumorigenesis was almost absent. Like other murine MOC-L and MTCQ cell line series we have previously established, MPC-1 also expresses a range of stemness markers, various oncogenic proteins, and a number of immune checkpoint proteins at high levels. However, the synergistic effects of the CDK4/6 inhibitor palbociclib on other therapeutic drugs were not observed with MPC-1. Whole exon sequencing revealed that there were high rates of non-synonymous mutations in MPC-1 affecting various genes, including Akap9, Arap2, Cdh11, Hjurp, Mroh2a, Muc4, Muc6, Sp110, and Sp140, which are similar to that the mutations present in a panel of chemical carcinogenesis-related murine tongue carcinoma cell lines. Analysis has highlighted the dis-regulation of Akap9, Cdh11, Muc4, Sp110, and Sp140 in human HNSCC as indicated by the TCGA and GEO OSCC databases. Sp140 expression has also been associated with patient survival. This study describes the establishment and characterization of the MPC-1 cell line and this new cell model should help to advance genetic research into oral cancer.
Collapse
Affiliation(s)
- Kuo-Wei Chang
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan; (K.-W.C.); (C.-E.L.); (H.-Y.C.); (Y.-F.C.)
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Stomatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chia-En Lin
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan; (K.-W.C.); (C.-E.L.); (H.-Y.C.); (Y.-F.C.)
| | - Hsi-Feng Tu
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Hsin-Yao Chung
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan; (K.-W.C.); (C.-E.L.); (H.-Y.C.); (Y.-F.C.)
| | - Yi-Fen Chen
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan; (K.-W.C.); (C.-E.L.); (H.-Y.C.); (Y.-F.C.)
| | - Shu-Chun Lin
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan; (K.-W.C.); (C.-E.L.); (H.-Y.C.); (Y.-F.C.)
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Stomatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Correspondence:
| |
Collapse
|
15
|
Marcazzan S, Dadbin A, Brachi G, Blanco E, Varoni EM, Lodi G, Ferrari M. Development of lung metastases in mouse models of tongue squamous cell carcinoma. Oral Dis 2020; 27:494-505. [PMID: 32767730 DOI: 10.1111/odi.13592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) represents 3%-4% of all cancers. Despite the increasing incidence of OSCC distant metastasis and poor prognosis, few animal models of OSCC distant metastasis have been reported. In this study, we established mouse models of OSCC lung metastasis by orthotopic and tail vein injection of new OSCC cell lines. METHODS For the tail vein model, we used a novel cell line isolated from lung metastases reproduced in vivo after intravenous injection of HSC-3 GFP/luciferase cells and sorted for GFP expression (HSC-3 M1 GFP/luciferase). Lung metastases were assessed by imaging techniques and further confirmed by histology. For the orthotopic model, HSC-3 GFP/luciferase cells were injected into the tongue of athymic nude mice. The primary tumor and metastases were assessed by in vivo imaging, histology, and immunohistochemistry. RESULTS The orthotopic model presented spontaneous lung metastases in 50% of the animals and lymph node metastases were present in 83% of cases. In the tail vein model, a lung metastasis rate of 60% was observed. CONCLUSIONS Lung metastases were successfully reproduced by orthotopic and tail vein injection. Since lymph node metastases were present, the orthotopic model with HSC-3 GFP/luciferase cells may be suitable to investigate metastatic dissemination in OSCC.
Collapse
Affiliation(s)
- Sabrina Marcazzan
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy.,Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Ali Dadbin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Giulia Brachi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.,Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Elena Maria Varoni
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | - Giovanni Lodi
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
16
|
Development and Radiation Response Assessment in A Novel Syngeneic Mouse Model of Tongue Cancer: 2D Culture, 3D Organoids and Orthotopic Allografts. Cancers (Basel) 2020; 12:cancers12030579. [PMID: 32131500 PMCID: PMC7139805 DOI: 10.3390/cancers12030579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/31/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) are aggressive cancers that contribute to significant morbidity and mortality in humans. Although numerous human xenograft models of OSCC have been developed, only a few syngeneic models of OSCC exist. Here, we report on a novel murine model of OSCC, RP-MOC1, derived from a tongue tumor in a C57Bl/6 mouse exposed to the carcinogen 4-nitroquinoline-1-oxide. Phenotypic characterization and credentialing (STR profiling, exome sequencing) of RP-MOC1 cells was performed in vitro. Radiosensitivity was evaluated in 2D culture, 3D organoids, and in vivo using orthotopic allografts. RP-MOC1 cells exhibited a stable epithelial phenotype with proliferative, migratory and invasive properties. Exome sequencing identified several mutations commonly found in OSCC patients. The LD50 for RP-MOC1 cells in 2D culture and 3D organoids was found to be 2.4 Gy and 12.6 Gy, respectively. Orthotopic RP-MOC1 tumors were pan-cytokeratin+ and Ki-67+. Magnetic resonance imaging of orthotopic RP-MOC1 tumors established in immunocompetent mice revealed marked growth inhibition following 10 Gy and 15 Gy fractionated radiation regimens. This radiation response was completely abolished in tumors established in immunodeficient mice. This novel syngeneic model of OSCC can serve as a valuable platform for the evaluation of combination strategies to enhance radiation response against this deadly disease.
Collapse
|
17
|
Li Q, Dong H, Yang G, Song Y, Mou Y, Ni Y. Mouse Tumor-Bearing Models as Preclinical Study Platforms for Oral Squamous Cell Carcinoma. Front Oncol 2020; 10:212. [PMID: 32158692 PMCID: PMC7052016 DOI: 10.3389/fonc.2020.00212] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
Preclinical animal models of oral squamous cell carcinoma (OSCC) have been extensively studied in recent years. Investigating the pathogenesis and potential therapeutic strategies of OSCC is required to further progress in this field, and a suitable research animal model that reflects the intricacies of cancer biology is crucial. Of the animal models established for the study of cancers, mouse tumor-bearing models are among the most popular and widely deployed for their high fertility, low cost, and molecular and physiological similarity to humans, as well as the ease of rearing experimental mice. Currently, the different methods of establishing OSCC mouse models can be divided into three categories: chemical carcinogen-induced, transplanted and genetically engineered mouse models. Each of these methods has unique advantages and limitations, and the appropriate application of these techniques in OSCC research deserves our attention. Therefore, this review comprehensively investigates and summarizes the tumorigenesis mechanisms, characteristics, establishment methods, and current applications of OSCC mouse models in published papers. The objective of this review is to provide foundations and considerations for choosing suitable model establishment methods to study the relevant pathogenesis, early diagnosis, and clinical treatment of OSCC.
Collapse
Affiliation(s)
- Qiang Li
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Heng Dong
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guangwen Yang
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuxian Song
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yongbin Mou
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Yongbin Mou
| | - Yanhong Ni
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Yanhong Ni
| |
Collapse
|
18
|
ERK Activation Modulates Cancer Stemness and Motility of a Novel Mouse Oral Squamous Cell Carcinoma Cell Line. Cancers (Basel) 2019; 12:cancers12010061. [PMID: 31878324 PMCID: PMC7016611 DOI: 10.3390/cancers12010061] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
We established the NHRI-HN1 cell line from a mouse tongue tumor induced by 4-nitroquinoline 1-oxide (4-NQO)/arecoline, with further selection for cell stemness via in vitro sphere culture, to evaluate potential immunotherapies for oral squamous cell carcinoma (OSCC) in East and Southeast Asia. In vivo and in vitro phenotypic characterization, including tumor growth, immune modulator administration, gene expression, morphology, migration, invasion, and sphere formation assays, were conducted. NHRI-HN1 cells are capable of generating orthotopic tumors in syngeneic mice. Interestingly, immune stimulation via CpG oligodeoxynucleotide (CpG-ODN) dramatically reduced the tumor growth in NHRI-HN1 cell-injected syngeneic mice. The pathways enriched in genes that were differentially expressed in NHRI-HN1 cells when compared to non-tumorigenic cells were similar to those that were identified when comparing human OSCC and non-tumorous tissues. NHRI-HN1 cells have characteristics of epithelial-mesenchymal transition (EMT), including enhanced migration and invasion. NHRI-HN1 cells showed aggressive cell growth and sphere formation. The blockage of extracellular signal-regulated kinase (ERK) activation suppressed cell migration and reduced stemness characteristics in NHRI-HN1 cells, similar to human OSCC cell lines. Our data suggest that NHRI-HN1 cells, showing tumorigenic characteristics of EMT, cancer stemness, and ERK activation, are sufficient in modeling human OSCC and also competent for use in investigating oral cancer immunotherapies.
Collapse
|
19
|
Chen YF, Chang KW, Yang IT, Tu HF, Lin SC. Establishment of syngeneic murine model for oral cancer therapy. Oral Oncol 2019; 95:194-201. [DOI: 10.1016/j.oraloncology.2019.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/03/2019] [Accepted: 06/24/2019] [Indexed: 01/09/2023]
|