1
|
Chan SPY, Yeo CPX, Hong BH, Tan EMC, Beh CY, Yeo ELL, Poon DJJ, Chu PL, Soo KC, Chua MLK, Chow EKH. Combinatorial functionomics identifies HDAC6-dependent molecular vulnerability of radioresistant head and neck cancer. Exp Hematol Oncol 2025; 14:5. [PMID: 39800760 PMCID: PMC11727331 DOI: 10.1186/s40164-024-00590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Radiotherapy is the primary treatment modality for most head and neck cancers (HNCs). Despite the addition of chemotherapy to radiotherapy to enhance its tumoricidal effects, almost a third of HNC patients suffer from locoregional relapses. Salvage therapy options for such recurrences are limited and often suboptimal, partly owing to divergent tumor and microenvironmental factors underpinning radioresistance. In this study, we utilized a combinatorial functionomics approach, the Quadratic Phenotypic Optimization Platform (QPOP), to rationally design drug pairings that exploit the molecular fingerprint and vulnerability of established in vitro isogenic radioresistant (RR)-HNC models. METHODS A QPOP-specific protocol was applied to RR-HNC models to rank and compare all possible drug combinations from a 12-drug set comprising standard chemotherapy, small molecule inhibitors and targeted therapies specific to HNC. Drug combination efficacy was evaluated by computing combination index scores, and by measuring apoptotic response. Drug targeting was validated by western blot analyses, and the Comet assay was used to quantify DNA damage. Enhanced histone deacetylase inhibitor (HDACi) efficacy in RR models was further examined by in vivo studies, and genetic and chemical inhibition of major Class I/II HDACs. Regulatory roles of HDAC6/SP1 axis were investigated using immunoprecipitation, gel shift and ChIP-qPCR assays. Comparative transcriptomic analyses were employed to determine the prognostic significance of targeting HDAC6. RESULTS We report the therapeutic potential of combining panobinostat (pan-HDAC inhibitor) with AZD7762 (CHK1/2 inhibitor; AstraZeneca) or ionizing radiation (IR) to re-sensitize RR-HNC cells and showed increased DNA damage underlying enhanced synergy. We further refined this RR-specific drug combination and prioritized HDAC6 as a targetable dependency in reversing radioresistance. We provide mechanistic insights into HDAC6-mediated regulation via a crosstalk involving SP1 and oncogenic and repair genes. From two independent patient cohorts, we identified a four-gene signature that may have discriminative ability to predict for radioresistance and amenable to HDAC6 inhibition. CONCLUSION We have uncovered HDAC6 as a promising molecular vulnerability that should be explored to treat RR-HNC.
Collapse
Affiliation(s)
- Sharon Pei Yi Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Celestia Pei Xuan Yeo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Boon Hao Hong
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Evelyn Mui Cheng Tan
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Chaw Yee Beh
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Eugenia Li Ling Yeo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Dennis Jun Jie Poon
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Pek Lim Chu
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Cancer and Stem Cell Biology Programme, Singapore, Singapore
| | - Khee Chee Soo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Melvin Lee Kiang Chua
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, Oncology Academic Programme, Singapore, Singapore.
- Department of Head and Neck and Thoracic Cancers, Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
2
|
Xiao Y, Zhang Y, Hu Y, Zhang X, Tan J, Yao S, Wang X, Qin Y. Advances in the study of posttranslational modifications of histones in head and neck squamous cell carcinoma. Clin Epigenetics 2024; 16:165. [PMID: 39574168 PMCID: PMC11580233 DOI: 10.1186/s13148-024-01785-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024] Open
Abstract
The pathogenesis of head and neck squamous cell carcinoma (HNSCC) is notably complex. Early symptoms are often subtle, and effective early screening methods are currently lacking. The tumors associated with HNSCC develop rapidly, exhibit high aggressiveness, and respond poorly to existing treatments, leading to low survival rates and poor prognosis. Numerous studies have demonstrated that histone posttranslational modifications (HPTMs), including acetylation, methylation, phosphorylation, and ubiquitination, play a critical role in the occurrence and progression of HNSCC. Moreover, targeting histone posttranslationally modified molecules with specific drugs has shown potential in enhancing therapeutic outcomes and improving prognosis, underscoring their significant clinical value. This review aims to summarize the role of histone posttranslational modifications in the pathogenesis and progression of HNSCC and to discuss their clinical significance, thereby providing insights into novel therapeutic approaches and drug development for this malignancy.
Collapse
Affiliation(s)
- Yuyang Xiao
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Yikai Zhang
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Yuyang Hu
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Xupeng Zhang
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Jiaqi Tan
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Shanhu Yao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
- Key Laboratory of Medical Information Research, Central South University, Changsha, 410013, Hunan Province, China
| | - Xingwei Wang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
| | - Yuexiang Qin
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, China.
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
3
|
Jin L, Jiang Q, Huang H, Zhou X. Topical histone deacetylase inhibitor remetinostat improves IMQ-induced psoriatic dermatitis via suppressing dendritic cell maturation and keratinocyte differentiation and inflammation. Eur J Pharmacol 2024; 983:177011. [PMID: 39304110 DOI: 10.1016/j.ejphar.2024.177011] [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: 06/05/2024] [Revised: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by excessive proliferation of keratinocytes and infiltration of immune cells. Although psoriasis has entered the era of biological treatment, there is still a need to explore more effective therapeutic targets and drugs due to the presence of resistance and adverse reactions to biologics. Remetinostat, an HDAC inhibitor, can maintain its potency within the skin with minimal systemic effects, making it a promising topical medication for treating psoriasis. But its effectiveness in treating psoriasis has not been evaluated. In this study, the topical application of remetinostat significantly improved psoriasiform inflammation in an imiquimod-induced mice model by inhibiting CD86 expression of CD11C+I-A/I-E+ dendritic cells (DCs) in the skin. Moreover, remetinostat could dampen the maturation and activation of bone marrow-derived DCs in vitro, as well as the expression of psoriasis-related inflammatory mediators by keratinocytes. In addition, remetinostat could promote keratinocyte differentiation without affecting its proliferation. Our findings demonstrate that remetinostat improves psoriasis by inhibiting the maturation and activation of DCs and the differentiation and inflammation of keratinocytes, which may facilitate the potential application of remetinostat in anti-psoriasis therapy.
Collapse
Affiliation(s)
- Liping Jin
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huining Huang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xingchen Zhou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Sacco JL, Gomez EW. Epithelial-Mesenchymal Plasticity and Epigenetic Heterogeneity in Cancer. Cancers (Basel) 2024; 16:3289. [PMID: 39409910 PMCID: PMC11475326 DOI: 10.3390/cancers16193289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The tumor microenvironment comprises various cell types and experiences dynamic alterations in physical and mechanical properties as cancer progresses. Intratumoral heterogeneity is associated with poor prognosis and poses therapeutic challenges, and recent studies have begun to identify the cellular mechanisms that contribute to phenotypic diversity within tumors. This review will describe epithelial-mesenchymal (E/M) plasticity and its contribution to phenotypic heterogeneity in tumors as well as how epigenetic factors, such as histone modifications, histone modifying enzymes, DNA methylation, and chromatin remodeling, regulate and maintain E/M phenotypes. This review will also report how mechanical properties vary across tumors and regulate epigenetic modifications and E/M plasticity. Finally, it highlights how intratumoral heterogeneity impacts therapeutic efficacy and provides potential therapeutic targets to improve cancer treatments.
Collapse
Affiliation(s)
- Jessica L. Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Esther W. Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
5
|
Liu Y, Zhang N, Wen Y, Wen J. Head and neck cancer: pathogenesis and targeted therapy. MedComm (Beijing) 2024; 5:e702. [PMID: 39170944 PMCID: PMC11338281 DOI: 10.1002/mco2.702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Head and neck cancer (HNC) is a highly aggressive type of tumor characterized by delayed diagnosis, recurrence, metastasis, relapse, and drug resistance. The occurrence of HNC were associated with smoking, alcohol abuse (or both), human papillomavirus infection, and complex genetic and epigenetic predisposition. Currently, surgery and radiotherapy are the standard treatments for most patients with early-stage HNC. For recurrent or metastatic (R/M) HNC, the first-line treatment is platinum-based chemotherapy combined with the antiepidermal growth factor receptor drug cetuximab, when resurgery and radiation therapy are not an option. However, curing HNC remains challenging, especially in cases with metastasis. In this review, we summarize the pathogenesis of HNC, including genetic and epigenetic changes, abnormal signaling pathways, and immune regulation mechanisms, along with all potential therapeutic strategies such as molecular targeted therapy, immunotherapy, gene therapy, epigenetic modifications, and combination therapies. Recent preclinical and clinical studies that may offer therapeutic strategies for future research on HNC are also discussed. Additionally, new targets and treatment methods, including antibody-drug conjugates, photodynamic therapy, radionuclide therapy, and mRNA vaccines, have shown promising results in clinical trials, offering new prospects for the treatment of HNC.
Collapse
Affiliation(s)
- Yan Liu
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
- National Facility for Translational Medicine (Sichuan)West China Hospital of Sichuan UniversityChengduChina
| | - Nannan Zhang
- National Center for Birth Defect MonitoringKey Laboratory of Birth Defects and Related Diseases of Women and ChildrenMinistry of EducationWest China Second University HospitalSichuan UniversityChengduChina
| | - Yi Wen
- State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Jiaolin Wen
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
| |
Collapse
|
6
|
Teresa BGD, Ayala-Zambrano C, González-Suárez M, Molina B, Torres L, Rodríguez A, Frías S. Reversion from basal histone H4 hypoacetylation at the replication fork increases DNA damage in FANCA deficient cells. PLoS One 2024; 19:e0298032. [PMID: 38820384 PMCID: PMC11142588 DOI: 10.1371/journal.pone.0298032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 06/02/2024] Open
Abstract
The FA/BRCA pathway safeguards DNA replication by repairing interstrand crosslinks (ICL) and maintaining replication fork stability. Chromatin structure, which is in part regulated by histones posttranslational modifications (PTMs), has a role in maintaining genomic integrity through stabilization of the DNA replication fork and promotion of DNA repair. An appropriate balance of PTMs, especially acetylation of histones H4 in nascent chromatin, is required to preserve a stable DNA replication fork. To evaluate the acetylation status of histone H4 at the replication fork of FANCA deficient cells, we compared histone acetylation status at the DNA replication fork of isogenic FANCA deficient and FANCA proficient cell lines by using accelerated native immunoprecipitation of nascent DNA (aniPOND) and in situ protein interactions in the replication fork (SIRF) assays. We found basal hypoacetylation of multiple residues of histone H4 in FA replication forks, together with increased levels of Histone Deacetylase 1 (HDAC1). Interestingly, high-dose short-term treatment with mitomycin C (MMC) had no effect over H4 acetylation abundance at the replication fork. However, chemical inhibition of histone deacetylases (HDAC) with Suberoylanilide hydroxamic acid (SAHA) induced acetylation of the FANCA deficient DNA replication forks to levels comparable to their isogenic control counterparts. This forced permanence of acetylation impacted FA cells homeostasis by inducing DNA damage and promoting G2 cell cycle arrest. Altogether, this caused reduced RAD51 foci formation and increased markers of replication stress, including phospho-RPA-S33. Hypoacetylation of the FANCA deficient replication fork, is part of the cellular phenotype, the perturbation of this feature by agents that prevent deacetylation, such as SAHA, have a deleterious effect over the delicate equilibrium they have reached to perdure despite a defective FA/BRCA pathway.
Collapse
Affiliation(s)
- Benilde García-de Teresa
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Ciudad de México, Mexico
| | - Cecilia Ayala-Zambrano
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Ciudad de México, Mexico
| | - Mirna González-Suárez
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
| | - Bertha Molina
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
| | - Leda Torres
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
| | - Alfredo Rodríguez
- Laboratorio de Falla Medular y Carcinogénesis, Unidad de Genética de la Nutrición, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Ciudad de México, Mexico
| | - Sara Frías
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Ciudad de México, Mexico
| |
Collapse
|
7
|
Jiang R, Fritz M, Que SKT. Cutaneous Squamous Cell Carcinoma: An Updated Review. Cancers (Basel) 2024; 16:1800. [PMID: 38791879 PMCID: PMC11119634 DOI: 10.3390/cancers16101800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Representing the second most common skin cancer, the incidence and disease burden of cutaneous squamous cell carcinoma (cSCC) continues to increase. Surgical excision of the primary site effectively cures the majority of cSCC cases. However, an aggressive subset of cSCC persists with clinicopathological features that are indicative of higher recurrence, metastasis, and mortality risks. Acceleration of these features is driven by a combination of genetic and environmental factors. The past several years have seen remarkable progress in shaping the treatment landscape for advanced cSCC. Risk stratification and clinical management is a top priority. This review provides an overview of the current perspectives on cSCC with a focus on staging, treatment, and maintenance strategies, along with future research directions.
Collapse
Affiliation(s)
- Rina Jiang
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Mike Fritz
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Syril Keena T. Que
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| |
Collapse
|
8
|
Seane EN, Nair S, Vandevoorde C, Joubert A. Mechanistic Sequence of Histone Deacetylase Inhibitors and Radiation Treatment: An Overview. Pharmaceuticals (Basel) 2024; 17:602. [PMID: 38794172 PMCID: PMC11124271 DOI: 10.3390/ph17050602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Histone deacetylases inhibitors (HDACis) have shown promising therapeutic outcomes in haematological malignancies such as leukaemia, multiple myeloma, and lymphoma, with disappointing results in solid tumours when used as monotherapy. As a result, combination therapies either with radiation or other deoxyribonucleic acid (DNA) damaging agents have been suggested as ideal strategy to improve their efficacy in solid tumours. Numerous in vitro and in vivo studies have demonstrated that HDACis can sensitise malignant cells to both electromagnetic and particle types of radiation by inhibiting DNA damage repair. Although the radiosensitising ability of HDACis has been reported as early as the 1990s, the mechanisms of radiosensitisation are yet to be fully understood. This review brings forth the various protocols used to sequence the administration of radiation and HDACi treatments in the different studies. The possible contribution of these various protocols to the ambiguity that surrounds the mechanisms of radiosensitisation is also highlighted.
Collapse
Affiliation(s)
- Elsie Neo Seane
- Department of Radiography, School of Health Care Sciences, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
- Department of Medical Imaging and Therapeutic Sciences, Faculty of Health and Wellness, Cape Peninsula University of Technology, Cape Town 7530, South Africa
- Radiation Biophysics Division, Separate Sector Cyclotron (SSC) Laboratory, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiation Biophysics Division, Separate Sector Cyclotron (SSC) Laboratory, iThemba LABS, Cape Town 7131, South Africa;
| | - Charlot Vandevoorde
- GSI Helmholtz Centre for Heavy Ion Research, Department of Biophysics, 64291 Darmstadt, Germany;
| | - Anna Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa;
| |
Collapse
|
9
|
Xu M, Hou Y, Li N, Yu W, Chen L. Targeting histone deacetylases in head and neck squamous cell carcinoma: molecular mechanisms and therapeutic targets. J Transl Med 2024; 22:418. [PMID: 38702756 PMCID: PMC11067317 DOI: 10.1186/s12967-024-05169-9] [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: 12/27/2023] [Accepted: 04/05/2024] [Indexed: 05/06/2024] Open
Abstract
The onerous health and economic burden associated with head and neck squamous cell carcinoma (HNSCC) is a global predicament. Despite the advent of novel surgical techniques and therapeutic protocols, there is an incessant need for efficacious diagnostic and therapeutic targets to monitor the invasion, metastasis and recurrence of HNSCC due to its substantial morbidity and mortality. The differential expression patterns of histone deacetylases (HDACs), a group of enzymes responsible for modifying histones and regulating gene expression, have been demonstrated in neoplastic tissues. However, there is limited knowledge regarding the role of HDACs in HNSCC. Consequently, this review aims to summarize the existing research findings and explore the potential association between HDACs and HNSCC, offering fresh perspectives on therapeutic approaches targeting HDACs that could potentially enhance the efficacy of HNSCC treatment. Additionally, the Cancer Genome Atlas (TCGA) dataset, CPTAC, HPA, OmicShare, GeneMANIA and STRING databases are utilized to provide supplementary evidence on the differential expression of HDACs, their prognostic significance and predicting functions in HNSCC patients.
Collapse
Affiliation(s)
- Mengchen Xu
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Shandong Provincial Clinical Research Center for Oral Diseases, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yiming Hou
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Shandong Provincial Clinical Research Center for Oral Diseases, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Na Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250022, Shandong, China
- Center of Clinical Laboratory, Shandong Second Provincial General Hospital, Jinan, 250022, Shandong, China
| | - Wenqian Yu
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Lei Chen
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Shandong Provincial Clinical Research Center for Oral Diseases, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
10
|
Barry B, Dolivet G, Clatot F, Huguet F, Abdeddaim C, Baujat B, Blanchard N, Calais G, Carrat X, Chatellier A, Coste F, Cupissol D, Cuvelier P, De Mones Del Pujol E, Deneuve S, Duffas O, Dupret-Bories A, Even C, Evrard C, Evrard D, Faivre S, Fakhry N, Garrel R, Gorphe P, Houliat T, Kaminsky MC, Krebs L, Lapeyre M, Lindas P, Malard O, Mirghani H, Mondina M, Moriniere S, Mouawad F, Pestre-Munier J, Pham Dang N, Picard A, Ramin L, Renard S, Salvan D, Schernberg A, Sire C, Thariat J, Vanbockstael J, Vo Tan D, Wojcik T, Klein I, Block V, Baumann-Bouscaud L, De Raucourt D. [French national standard for the treatment of squamous cell carcinoma of upper aero-digestive tract - General principles of treatment]. Bull Cancer 2024; 111:393-415. [PMID: 38418334 DOI: 10.1016/j.bulcan.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/14/2023] [Accepted: 12/31/2023] [Indexed: 03/01/2024]
Abstract
OBJECTIVES The management of upper aerodigestive tract cancers is a complex specialty. It is essential to provide an update to establish optimal care. At the initiative of the INCa and under the auspices of the SFORL, the scientific committee, led by Professor Béatrix Barry, Dr. Gilles Dolivet, and Dr. Dominique De Raucourt, decided to develop a reference framework aimed at defining, in a scientific and consensus-based manner, the general principles of treatment for upper aerodigestive tract cancers applicable to all sub-locations. METHODOLOGY To develop this framework, a multidisciplinary team of practitioners was formed. A systematic analysis of the literature was conducted to produce recommendations classified by grades, in accordance with the standards of the French National Authority for Health (HAS). RESULTS The grading of recommendations according to HAS standards has allowed the establishment of a reference for patient care based on several criteria. In this framework, patients benefit from differentiated care based on prognostic factors they present (age, comorbidities, TNM status, HPV status, etc.), conditions of implementation, and quality criteria for indicated surgery (operability, resectability, margin quality, mutilation, salvage surgery), as well as quality criteria for radiotherapy (target volume, implementation time, etc.). The role of medical and postoperative treatments was also evaluated based on specific criteria. Finally, supportive care must be organized from the beginning and throughout the patients' care journey. CONCLUSION All collected data have led to the development of a comprehensive framework aimed at harmonizing practices nationally, facilitating decision-making in multidisciplinary consultation meetings, promoting equality in practices, and providing a state-of-the-art and reference practices for assessing the quality of care. This new framework is intended to be updated every 5 years to best reflect the latest advances in the field.
Collapse
Affiliation(s)
- Béatrix Barry
- AP-HP, hôpital Bichat-Claude-Bernard, ORL et CCF, Paris (75), France
| | - Gilles Dolivet
- Institut de cancérologie de Lorraine, ORL et CCF, Nancy (54), France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Olivier Duffas
- Centre hospitalier de Libourne, ORL et CMF, Libourne, France
| | | | | | | | - Diane Evrard
- AP-HP, hôpital Bichat-Claude-Bernard, ORL et CCF, Paris (75), France
| | | | - Nicolas Fakhry
- Assistance publique-Hôpitaux de Marseille, ORL et CCF, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Didier Salvan
- Centre hospitalier Sud Francilien, ORL et CCF, Corbeil-Essonnes, France
| | | | | | | | | | | | | | - Isabelle Klein
- Dispositif Spécifique Régional du Cancer Grand Est - NEON, Nancy (54), France
| | - Véronique Block
- Dispositif Spécifique Régional du Cancer Grand Est - NEON, Nancy (54), France
| | | | | |
Collapse
|
11
|
Viktorsson K, Rieckmann T, Fleischmann M, Diefenhardt M, Hehlgans S, Rödel F. Advances in molecular targeted therapies to increase efficacy of (chemo)radiation therapy. Strahlenther Onkol 2023; 199:1091-1109. [PMID: 37041372 PMCID: PMC10673805 DOI: 10.1007/s00066-023-02064-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/19/2023] [Indexed: 04/13/2023]
Abstract
Recent advances in understanding the tumor's biology in line with a constantly growing number of innovative technologies have prompted characterization of patients' individual malignancies and may display a prerequisite to treat cancer at its patient individual tumor vulnerability. In recent decades, radiation- induced signaling and tumor promoting local events for radiation sensitization were explored in detail, resulting the development of novel molecular targets. A multitude of pharmacological, genetic, and immunological principles, including small molecule- and antibody-based targeted strategies, have been developed that are suitable for combined concepts with radiation (RT) or chemoradiation therapy (CRT). Despite a plethora of promising experimental and preclinical findings, however, so far, only a very limited number of clinical trials have demonstrated a better outcome and/or patient benefit when RT or CRT are combined with targeted agents. The current review aims to summarize recent progress in molecular therapies targeting oncogenic drivers, DNA damage and cell cycle response, apoptosis signaling pathways, cell adhesion molecules, hypoxia, and the tumor microenvironment to impact therapy refractoriness and to boost radiation response. In addition, we will discuss recent advances in nanotechnology, e.g., RNA technologies and protein-degrading proteolysis-targeting chimeras (PROTACs) that may open new and innovative ways to benefit from molecular-targeted therapy approaches with improved efficacy.
Collapse
Affiliation(s)
- Kristina Viktorsson
- Department of Oncology/Pathology, Karolinska Institutet, Visionsgatan 4, 17164, Solna, Sweden
| | - Thorsten Rieckmann
- Department of Radiation Oncology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Otolaryngology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Maximilian Fleischmann
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Markus Diefenhardt
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
- German Cancer Consortium (DKTK) partner site: Frankfurt, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
12
|
Aleksandrova Y, Neganova M. Deciphering the Mysterious Relationship between the Cross-Pathogenetic Mechanisms of Neurodegenerative and Oncological Diseases. Int J Mol Sci 2023; 24:14766. [PMID: 37834214 PMCID: PMC10573395 DOI: 10.3390/ijms241914766] [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: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The relationship between oncological pathologies and neurodegenerative disorders is extremely complex and is a topic of concern among a growing number of researchers around the world. In recent years, convincing scientific evidence has accumulated that indicates the contribution of a number of etiological factors and pathophysiological processes to the pathogenesis of these two fundamentally different diseases, thus demonstrating an intriguing relationship between oncology and neurodegeneration. In this review, we establish the general links between three intersecting aspects of oncological pathologies and neurodegenerative disorders, i.e., oxidative stress, epigenetic dysregulation, and metabolic dysfunction, examining each process in detail to establish an unusual epidemiological relationship. We also focus on reviewing the current trends in the research and the clinical application of the most promising chemical structures and therapeutic platforms that have a modulating effect on the above processes. Thus, our comprehensive analysis of the set of molecular determinants that have obvious cross-functional pathways in the pathogenesis of oncological and neurodegenerative diseases can help in the creation of advanced diagnostic tools and in the development of innovative pharmacological strategies.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 420088 Kazan, Russia
| |
Collapse
|
13
|
Ling R, Wang J, Fang Y, Yu Y, Su Y, Sun W, Li X, Tang X. HDAC-an important target for improving tumor radiotherapy resistance. Front Oncol 2023; 13:1193637. [PMID: 37503317 PMCID: PMC10368992 DOI: 10.3389/fonc.2023.1193637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Radiotherapy is an important means of tumor treatment, but radiotherapy resistance has been a difficult problem in the comprehensive treatment of clinical tumors. The mechanisms of radiotherapy resistance include the repair of sublethal damage and potentially lethal damage of tumor cells, cell repopulation, cell cycle redistribution, and reoxygenation. These processes are closely related to the regulation of epigenetic modifications. Histone deacetylases (HDACs), as important regulators of the epigenetic structure of cancer, are widely involved in the formation of tumor radiotherapy resistance by participating in DNA damage repair, cell cycle regulation, cell apoptosis, and other mechanisms. Although the important role of HDACs and their related inhibitors in tumor therapy has been reviewed, the relationship between HDACs and radiotherapy has not been systematically studied. This article systematically expounds for the first time the specific mechanism by which HDACs promote tumor radiotherapy resistance in vivo and in vitro and the clinical application prospects of HDAC inhibitors, aiming to provide a reference for HDAC-related drug development and guide the future research direction of HDAC inhibitors that improve tumor radiotherapy resistance.
Collapse
Affiliation(s)
- Rui Ling
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jingzhi Wang
- Department of Radiotherapy Oncology, Affiliated Yancheng First Hospital of Nanjing University Medical School, First People’s Hospital of Yancheng, Yancheng, China
| | - Yuan Fang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yunpeng Yu
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuting Su
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wen Sun
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Tang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
14
|
Winge MCG, Kellman LN, Guo K, Tang JY, Swetter SM, Aasi SZ, Sarin KY, Chang ALS, Khavari PA. Advances in cutaneous squamous cell carcinoma. Nat Rev Cancer 2023:10.1038/s41568-023-00583-5. [PMID: 37286893 DOI: 10.1038/s41568-023-00583-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2023] [Indexed: 06/09/2023]
Abstract
Human malignancies arise predominantly in tissues of epithelial origin, where the stepwise transformation from healthy epithelium to premalignant dysplasia to invasive neoplasia involves sequential dysregulation of biological networks that govern essential functions of epithelial homeostasis. Cutaneous squamous cell carcinoma (cSCC) is a prototype epithelial malignancy, often with a high tumour mutational burden. A plethora of risk genes, dominated by UV-induced sun damage, drive disease progression in conjunction with stromal interactions and local immunomodulation, enabling continuous tumour growth. Recent studies have identified subpopulations of SCC cells that specifically interact with the tumour microenvironment. These advances, along with increased knowledge of the impact of germline genetics and somatic mutations on cSCC development, have led to a greater appreciation of the complexity of skin cancer pathogenesis and have enabled progress in neoadjuvant immunotherapy, which has improved pathological complete response rates. Although measures for the prevention and therapeutic management of cSCC are associated with clinical benefit, the prognosis remains poor for advanced disease. Elucidating how the genetic mechanisms that drive cSCC interact with the tumour microenvironment is a current focus in efforts to understand, prevent and treat cSCC.
Collapse
Affiliation(s)
- Mårten C G Winge
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
- Department of Dermatology, Stanford University, Redwood City, CA, USA
| | - Laura N Kellman
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Stanford Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Konnie Guo
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Jean Y Tang
- Department of Dermatology, Stanford University, Redwood City, CA, USA
| | - Susan M Swetter
- Department of Dermatology, Stanford University, Redwood City, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Sumaira Z Aasi
- Department of Dermatology, Stanford University, Redwood City, CA, USA
| | - Kavita Y Sarin
- Department of Dermatology, Stanford University, Redwood City, CA, USA
| | - Anne Lynn S Chang
- Department of Dermatology, Stanford University, Redwood City, CA, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA.
- Department of Dermatology, Stanford University, Redwood City, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Stanford Program in Cancer Biology, Stanford University, Stanford, CA, USA.
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA.
| |
Collapse
|
15
|
Yuan F, Yong J, Liu X, Wang Y. Selinexor assists vorinostat in inhibiting HDAC activity via promoting the accumulation of maspin in the nucleus of oral tongue squamous cell carcinoma cells. Cytotechnology 2023; 75:1-16. [PMID: 36713062 PMCID: PMC9880106 DOI: 10.1007/s10616-022-00555-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/07/2022] [Indexed: 02/01/2023] Open
Abstract
Oral tongue squamous cell carcinoma (OTSCC) is the most common oral cancer with a low overall survival rate, necessitating effective treatments. This study reports the anti-OTSCC effect of vorinostat and selinexor. OTSCC cell lines SCC-4 and SCC-25 were cultured to determine the effects of vorinostat and/or selinexor on cell survival, invasion, migration, and apoptosis. The transplanted tumor model of SCC-25 in nude mice was established to observe the therapeutic effects of vorinostat and/or selinexor. Western blotting was used to determine protein expressions in tumor cells. The results showed that histone deacetylase 1 (HDAC1) and exportin 1 (XPO1) were highly expressed, while nuclear maspin was expressed at a low rate in SCC-4 and SCC-25 compared to the normal tongue tissue. In vitro, both vorinostat and selinexor effectively inhibited cell viability, invasion, and migration, promoted cell apoptosis, down-regulated HDAC1, Matrix Metalloproteinase 2 (MMP2), and B cell leukemia/lymphoma 2 (Bcl-2), and up-regulated nuclear maspin and cleaved caspase 3. In vivo, both vorinostat and selinexor inhibited the growth of SCC-25-bearing tumors, down-regulated the expression of Ki67, HDAC1, MMP2, and Bcl-2, and promoted the expression of nuclear maspin and cleaved caspase 3. The combination of these two drugs exhibited synergistic effects both in vivo and in vitro. Our evidence shows that vorinostat combined with selinexor is an effective treatment for OTSCC. The mechanism may be that selinexor promotes the accumulation of maspin in the nucleus, an endogenous HDAC1 inhibitory protein to inhibit the HDAC1 activity of vorinostat and exert a synergistic anti-OTSCC effect.
Collapse
Affiliation(s)
- Fenqian Yuan
- Department of Head and Neck Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, 519 East Beijing Road, Nanchang, 330029 Jiangxi China
| | - Jingkang Yong
- Department of Head and Neck Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, 519 East Beijing Road, Nanchang, 330029 Jiangxi China
| | - Xueming Liu
- Department of Head and Neck Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, 519 East Beijing Road, Nanchang, 330029 Jiangxi China
| | - Yifeng Wang
- Department of Head and Neck Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, 519 East Beijing Road, Nanchang, 330029 Jiangxi China
| |
Collapse
|
16
|
Sullivan JK, Fahey PP, Agho KE, Hurley SP, Feng Z, Day RO, Lim D. Valproic acid as a radio-sensitizer in glioma: A systematic review and meta-analysis. Neurooncol Pract 2023; 10:13-23. [PMID: 36659976 PMCID: PMC9837785 DOI: 10.1093/nop/npac078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Histone deacetylase inhibitors (HDACi) including valproic acid (VPA) have the potential to improve radiotherapy (RT) efficacy and reduce treatment adverse events (AE) via epigenetic modification and radio-sensitization of neoplastic cells. This systematic review and meta-analysis aimed to assess the efficacy and AE associated with HDACi used as radio-sensitizers in adult solid organ malignancy patients. Methods A systematic review utilized electronic searches of MEDLINE(Ovid), Embase(Ovid), The Cochrane Library, and the International Clinical Trials Registry Platform to identify studies examining the efficacy and AEs associated with HDACi treatment in solid organ malignancy patients undergoing RT. Meta-analysis was performed with overall survival (OS) reported as hazard ratios (HR) as the primary outcome measure. OS reported as median survival difference, and AEs were secondary outcome measures. Results Ten studies reporting on the efficacy and/or AEs of HDACi in RT-treated solid organ malignancy patients met inclusion criteria. All included studies focused on HDACi valproic acid (VPA) in high-grade glioma patients, of which 9 studies (n = 6138) evaluated OS and 5 studies (n = 1055) examined AEs. The addition of VPA to RT treatment protocols resulted in improved OS (HR = 0.80, 95% CI 0.67-0.96). No studies focusing on non-glioma solid organ malignancy patients, or non-VPA HDACi met the inclusion criteria for this review. Conclusions This review suggests that glioma patients undergoing RT may experience prolonged survival due to HDACi VPA administration. Further randomized controlled trials are required to validate these findings. Additionally, more research into the use of HDACi radio-adjuvant treatment in non-glioma solid organ malignancies is warranted.
Collapse
Affiliation(s)
| | - Paul P Fahey
- School of Health Sciences, Western Sydney University, New South Wales, Australia
| | - Kinglsey E Agho
- School of Health Sciences, Western Sydney University, New South Wales, Australia
| | - Simon P Hurley
- School of Medicine, Flinders University, South Australia, Australia
| | - Zhihui Feng
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Richard O Day
- St Vincent’s Clinical Campus, University of New South Wales, New South Wales, Australia
| | - David Lim
- School of Medicine, Flinders University, South Australia, Australia
- School of Health Sciences, Western Sydney University, New South Wales, Australia
- Centre for Remote Health: A JBI Affiliated Centre, Alice Springs, Australia
| |
Collapse
|
17
|
Mireștean CC, Iancu RI, Iancu DPT. Capecitabine-A "Permanent Mission" in Head and Neck Cancers "War Council"? J Clin Med 2022; 11:5582. [PMID: 36233450 PMCID: PMC9573684 DOI: 10.3390/jcm11195582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Capecitabine, an oral pro-drug that is metabolized to 5-FU, has been used in clinical practice for more than 20 years, being part of the therapeutic standard for digestive and breast cancers. The use of capecitabine has been evaluated in many trials including cases diagnosed in recurrent or metastatic settings. Induction regimens or a combination with radiation therapy were evaluated in head and neck cancers, but 5-FU still remained the fluoropyrimidine used as a part of the current therapeutic standard. Quantifications of levels or ratios for enzymes are involved in the capecitabine metabolism to 5-FU but are also involved in its conversion and elimination that may lead to discontinuation, dose reduction or escalation of treatment in order to obtain the best therapeutic ratio. These strategies based on biomarkers may be relevant in the context of the implementation of precision oncology. In particular for head and neck cancers, the identification of biomarkers to select possible cases of severe toxicity requiring discontinuation of treatment, including "multi-omics" approaches, evaluate not only serological biomarkers, but also miRNAs, imaging and radiomics which will ensure capecitabine a role in both induction and concomitant or even adjuvant and palliative settings. An approach including routine testing of dihydropyrimidine dehydrogenase (DPD) or even the thymidine phosphorylase (TP)/DPD ratio and the inclusion of miRNAs, imaging and radiomics parameters in multi-omics models will help implement "precision chemotherapy" in HNC, a concept supported by the importance of avoiding interruptions or treatment delays in this type of cancer. The chemosensitivity and prognostic features of HPV-OPC cancers open new horizons for the use of capecitabine in heavily pretreated metastatic cases. Vorinostat and lapatinib are agents that can be associated with capecitabine in future clinical trials to increase the therapeutic ratio.
Collapse
Affiliation(s)
- Camil Ciprian Mireștean
- Department of Medical Oncology and Radiotherapy, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
- Department of Surgery, Railways Clinical Hospital, 700506 Iasi, Romania
| | - Roxana Irina Iancu
- Oral Pathology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Clinical Laboratory, St. Spiridon Emergency Hospital, 700111 Iasi, Romania
| | - Dragoș Petru Teodor Iancu
- Department of Medical Oncology and Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Radiation Oncology, Regional Institute of Oncology, 700483 Iasi, Romania
| |
Collapse
|
18
|
Monge-Cadet J, Moyal E, Supiot S, Guimas V. DNA repair inhibitors and radiotherapy. Cancer Radiother 2022; 26:947-954. [PMID: 35987813 DOI: 10.1016/j.canrad.2022.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022]
Abstract
Radiotherapy (RT) is one of the main cancer treatments and grows in importance due to improved techniques. DNA damage caused by ionizing radiation creates DNA strand breaks that trigger an intervention of DNA repair pathways involving numerous proteins and enzymes. In recent years, we have identified DNA repair inhibitors as targets for inhibiting cellular repair systems and thus causing cell death. Combining RT with these DNA repair inhibitors appears to be a new approach for cancer treatment, but safety and real efficiency of this combination in practice is unclear. Numerous trials are underway in various diseases and initial results are promising overall, yet remain controversial.
Collapse
Affiliation(s)
- J Monge-Cadet
- Radiothérapie, institut universitaire du cancer de Toulouse, 1, avenue Irène-Joliot-Curie, 31100 Toulouse, France.
| | - E Moyal
- Radiothérapie, institut universitaire du cancer de Toulouse, 1, avenue Irène-Joliot-Curie, 31100 Toulouse, France
| | - S Supiot
- Radiothérapie, institut de cancérologie de l'Ouest, boulevard Professeur Jacques-Monod, 44800 Saint-Herblain, France
| | - V Guimas
- Radiothérapie, institut de cancérologie de l'Ouest, boulevard Professeur Jacques-Monod, 44800 Saint-Herblain, France
| |
Collapse
|
19
|
Suchanti S, Stephen BJ, Awasthi S, Awasthi SK, Singh G, Singh A, Mishra R. Harnessing the role of epigenetic histone modification in targeting head and neck squamous cell carcinoma. Epigenomics 2022; 14:279-293. [PMID: 35184601 DOI: 10.2217/epi-2020-0348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most prevalent form of cancer worldwide. Despite advancements made in treatment strategies, the fatality rate of HNSCC is very high. An accumulating body of evidence suggests that epigenetic modification of histones plays an influential role in the development and progression of the disease. In this review we discuss the role of epigenetic modifications in HNSCC and the inter-relationships of human papillomavirus oncoproteins and histone-modifying agents. Further, we explore the possibility of identifying these modifications as biomarkers for their use as drugs in treatment strategies.
Collapse
Affiliation(s)
- Surabhi Suchanti
- Department of Biosciences, Manipal University Jaipur, Rajasthan, 303007, India
| | - Bjorn J Stephen
- Department of Biosciences, Manipal University Jaipur, Rajasthan, 303007, India
| | - Sonali Awasthi
- Department of Life Sciences, CSJM University, Kanpur, Uttar Pradesh, 208024, India
| | - Sudhir K Awasthi
- Department of Life Sciences, CSJM University, Kanpur, Uttar Pradesh, 208024, India
| | - Gyanendra Singh
- Toxicology Division, ICMR-National Institute of Occupational Health, Ahmedabad, Gujarat, 380016, India
| | - Abhijeet Singh
- Department of Biosciences, Manipal University Jaipur, Rajasthan, 303007, India
| | - Rajeev Mishra
- Department of Life Sciences, CSJM University, Kanpur, Uttar Pradesh, 208024, India
| |
Collapse
|
20
|
Yuan B, Zhao X, Wang X, Liu E, Liu C, Zong Y, Jiang Y, Hou M, Chen Y, Chen L, Zhang Y, Wang H, Fu J. Patient-derived organoids for personalized gallbladder cancer modelling and drug screening. Clin Transl Med 2022; 12:e678. [PMID: 35075805 PMCID: PMC8786696 DOI: 10.1002/ctm2.678] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/15/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gallbladder carcinoma (GBC) is a relatively rare but highly aggressive cancer with late clinical detection and a poor prognosis. However, the lack of models with features consistent with human gallbladder tumours has hindered progress in pathogenic mechanisms and therapies. METHODS We established organoid lines derived from human GBC as well as normal gallbladder and benign gallbladder adenoma (GBA) tissues. The histopathology signatures of organoid cultures were identified by H&E staining, immunohistochemistry and immunofluorescence. The genetic and transcriptional features of organoids were analysed by whole-exome sequencing and RNA sequencing. A set of compounds targeting the most active signalling pathways in GBCs were screened for their ability to suppress GBC organoids. The antitumour effects of candidate compounds, CUDC-101 and CUDC-907, were evaluated in vitro and in vivo. RESULTS The established organoids were cultured stably for more than 6 months and closely recapitulated the histopathology, genetic and transcriptional features, and intratumour heterogeneity of the primary tissues at the single-cell level. Notably, expression profiling analysis of the organoids revealed a set of genes that varied across the three subtypes and thus may participate in the malignant progression of gallbladder diseases. More importantly, we found that the dual PI3K/HDAC inhibitor CUDC-907 significantly restrained the growth of various GBC organoids with minimal toxicity to normal gallbladder organoids. CONCLUSIONS Patient-derived organoids are potentially a useful platform to explore molecular pathogenesis of gallbladder tumours and discover personalized drugs.
Collapse
Affiliation(s)
- Bo Yuan
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Xiaofang Zhao
- Research Center for OrganoidsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiang Wang
- Second Department of Biliary SurgeryEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Erdong Liu
- School of Life SciencesFudan UniversityShanghaiChina
| | - Chunliang Liu
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Yali Zong
- School of Life SciencesFudan UniversityShanghaiChina
| | - Youhai Jiang
- Division of Life Sciences and MedicineCancer Research CenterThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Minghui Hou
- Research Center for OrganoidsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yao Chen
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Lei Chen
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Yongjie Zhang
- Second Department of Biliary SurgeryEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Hongyang Wang
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Jing Fu
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| |
Collapse
|
21
|
Kilgour JM, Shah A, Eichstadt S, Bailey I, Aasi SZ, Sarin KY. Treatment of Cutaneous Squamous Cell Carcinoma With the Topical Histone Deacetylase Inhibitor Remetinostat. JAMA Dermatol 2021; 158:105-107. [PMID: 34787644 DOI: 10.1001/jamadermatol.2021.4549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- James M Kilgour
- Department of Dermatology, Stanford University, Stanford, California
| | - Aatman Shah
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Irene Bailey
- Department of Dermatology, Stanford University, Stanford, California
| | - Sumaira Z Aasi
- Department of Dermatology, Stanford University, Stanford, California
| | - Kavita Y Sarin
- Department of Dermatology, Stanford University, Stanford, California
| |
Collapse
|
22
|
DuBois SG, Granger MM, Groshen S, Tsao-Wei D, Ji L, Shamirian A, Czarnecki S, Goodarzian F, Berkovich R, Shimada H, Villablanca JG, Vo KT, Pinto N, Mosse YP, Maris JM, Shusterman S, Cohn SL, Goldsmith KC, Weiss B, Yanik GA, Twist CJ, Irwin MS, Haas-Kogan DA, Park JR, Marachelian A, Matthay KK. Randomized Phase II Trial of MIBG Versus MIBG, Vincristine, and Irinotecan Versus MIBG and Vorinostat for Patients With Relapsed or Refractory Neuroblastoma: A Report From NANT Consortium. J Clin Oncol 2021; 39:3506-3514. [PMID: 34270348 PMCID: PMC8547934 DOI: 10.1200/jco.21.00703] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/02/2021] [Accepted: 06/23/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE 131I-metaiodobenzylguanidine (MIBG) is an active radiotherapeutic for neuroblastoma. The primary aim of this trial was to identify which of three MIBG regimens was likely associated with the highest true response rate. PATIENTS AND METHODS Patients 1-30 years were eligible if they had relapsed or refractory neuroblastoma, at least one MIBG-avid site, and adequate autologous stem cells. Patients received MIBG 18 mCi/kg on day 1 and autologous stem cell on day 15. Patients randomly assigned to arm A received only MIBG; patients randomly assigned to arm B received intravenous vincristine on day 0 and irinotecan daily on days 0-4; patients randomly assigned to arm C received vorinostat (180 mg/m2/dose) orally once daily on days 1 to 12. The primary end point was response after one course by New Approaches to Neuroblastoma Therapy criteria. The trial was designed with 105 patients to ensure an 80% chance that the arm with highest response rate was selected. RESULTS One hundred fourteen patients were enrolled, with three ineligible and six unevaluable, leaving 105 eligible and evaluable patients (36 in arm A, 35 in arm B, and 34 in arm C; 55 boys; and median age 6.5 years). After one course, the response rates (partial response or better) on arms A, B, and C were 14% (95% CI, 5 to 30), 14% (5 to 31), and 32% (18 to 51). An additional five, five, and four patients met New Approaches to Neuroblastoma Therapy Minor Response criteria on arms A, B, and C, respectively. On arms A, B, and C, rates of any grade 3+ nonhematologic toxicity after first course were 19%, 49%, and 35%. CONCLUSION Vorinostat and MIBG is likely the arm with the highest true response rate, with manageable toxicity. Vincristine and irinotecan do not appear to improve the response rate to MIBG and are associated with increased toxicity.
Collapse
Affiliation(s)
- Steven G. DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | | | - Susan Groshen
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Denice Tsao-Wei
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Lingyun Ji
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Anasheh Shamirian
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Scarlett Czarnecki
- Department of Pediatrics, Loma Linda University Medical Center, Loma Linda, CA
| | - Fariba Goodarzian
- Department of Radiology, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Rachel Berkovich
- Department of Radiology, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Hiroyuki Shimada
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA
| | - Judith G. Villablanca
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Kieuhoa T. Vo
- Department of Pediatrics, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA
| | - Navin Pinto
- Department of Pediatrics, Seattle Children's Hospital and University of Washington School of Medicine, Seattle, WA
| | - Yael P. Mosse
- Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - John M. Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Suzanne Shusterman
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Susan L. Cohn
- Department of Pediatrics, Comer Children's Hospital and University of Chicago Pritzker School of Medicine, Chicago, IL
| | - Kelly C. Goldsmith
- Department of Pediatrics, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA
| | - Brian Weiss
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Gregory A. Yanik
- Department of Pediatrics, CS Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, MI
| | - Clare J. Twist
- Department of Pediatrics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Meredith S. Irwin
- Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daphne A. Haas-Kogan
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Julie R. Park
- Department of Pediatrics, Seattle Children's Hospital and University of Washington School of Medicine, Seattle, WA
| | - Araz Marachelian
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Katherine K. Matthay
- Department of Pediatrics, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA
| |
Collapse
|
23
|
Burkitt K, Saloura V. Epigenetic Modifiers as Novel Therapeutic Targets and a Systematic Review of Clinical Studies Investigating Epigenetic Inhibitors in Head and Neck Cancer. Cancers (Basel) 2021; 13:cancers13205241. [PMID: 34680389 PMCID: PMC8534083 DOI: 10.3390/cancers13205241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Head and neck cancer is the sixth most common malignancy worldwide and it affects approximately 50,000 patients annually in the United States. Current treatments are suboptimal and induce significant long-term toxicities that permanently affect quality of life. Novel therapeutic approaches are thus urgently needed to increase the survival and quality of life of these patients. Epigenetic modifications have been recognized as potential therapeutic targets in various cancer types, including head and neck cancer. The objective of this review is to provide a brief overview of the function of important epigenetic modifiers in head and neck cancer, and to discuss the results of past and ongoing clinical trials evaluating epigenetic interventions targeting these epigenetic modifiers in head and neck cancer patients. The field of epigenetic therapy in head and neck cancer is still nascent; however, it holds significant promise. Although more specific epigenetic drugs are being developed, we envision the rational design of clinical trials that will target a select group of head and neck cancer patients with epigenetic vulnerabilities that can be targeted in combination with immunotherapy, chemotherapy and/or radiotherapy, rendering higher and durable responses while minimizing chronic complications for patients with head and neck cancer. Abstract The survival rate of head and neck squamous cell carcinoma patients with the current standard of care therapy is suboptimal and is associated with long-term side effects. Novel therapeutics that will improve survival rates while minimizing treatment-related side effects are the focus of active investigation. Epigenetic modifications have been recognized as potential therapeutic targets in various cancer types, including head and neck cancer. This review summarizes the current knowledge on the function of important epigenetic modifiers in head and neck cancer, their clinical implications and discusses results of clinical trials evaluating epigenetic interventions in past and ongoing clinical trials as monotherapy or combination therapy with either chemotherapy, radiotherapy or immunotherapy. Understanding the function of epigenetic modifiers in both preclinical and clinical settings will provide insight into a more rational design of clinical trials using epigenetic interventions and the patient subgroups that may benefit from such interventions.
Collapse
Affiliation(s)
- Kyunghee Burkitt
- Head and Neck Medical Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Correspondence: (K.B.); (V.S.)
| | - Vassiliki Saloura
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
- Correspondence: (K.B.); (V.S.)
| |
Collapse
|
24
|
Macedo-Silva C, Benedetti R, Ciardiello F, Cappabianca S, Jerónimo C, Altucci L. Epigenetic mechanisms underlying prostate cancer radioresistance. Clin Epigenetics 2021; 13:125. [PMID: 34103085 PMCID: PMC8186094 DOI: 10.1186/s13148-021-01111-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/02/2021] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy (RT) is one of the mainstay treatments for prostate cancer (PCa), a highly prevalent neoplasm among males worldwide. About 30% of newly diagnosed PCa patients receive RT with a curative intent. However, biochemical relapse occurs in 20–40% of advanced PCa treated with RT either alone or in combination with adjuvant-hormonal therapy. Epigenetic alterations, frequently associated with molecular variations in PCa, contribute to the acquisition of a radioresistant phenotype. Increased DNA damage repair and cell cycle deregulation decreases radio-response in PCa patients. Moreover, the interplay between epigenome and cell growth pathways is extensively described in published literature. Importantly, as the clinical pattern of PCa ranges from an indolent tumor to an aggressive disease, discovering specific targetable epigenetic molecules able to overcome and predict PCa radioresistance is urgently needed. Currently, histone-deacetylase and DNA-methyltransferase inhibitors are the most studied classes of chromatin-modifying drugs (so-called ‘epidrugs’) within cancer radiosensitization context. Nonetheless, the lack of reliable validation trials is a foremost drawback. This review summarizes the major epigenetically induced changes in radioresistant-like PCa cells and describes recently reported targeted epigenetic therapies in pre-clinical and clinical settings. ![]()
Collapse
Affiliation(s)
- Catarina Macedo-Silva
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy.,Cancer Biology and Epigenetics Group, Research Center at Portuguese Oncology Institute of Porto, F Bdg, 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center at Portuguese Oncology Institute of Porto, F Bdg, 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal. .,Department of Pathology and Molecular Immunology at School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal.
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy.
| |
Collapse
|
25
|
HDAC1 regulates the chemosensitivity of laryngeal carcinoma cells via modulation of interleukin-8 expression. Eur J Pharmacol 2021; 896:173923. [PMID: 33539818 DOI: 10.1016/j.ejphar.2021.173923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 01/28/2023]
Abstract
Chemotherapies such as 5-fluorouracil (5-FU) and cisplatin (CDDP) have been widely used to treat laryngeal squamous cell carcinoma (LSCC), the second most common head and neck squamous cell carcinoma. However, chemoresistance seriously impairs chemotherapeutic efficacy. Our present study reveals that 5-FU and CDDP treatment increase the expression of histone deacetylase 1 (HDAC1) in LSCC cells. Consistently, increased levels of HDAC1 are observed in chemoresistant cells. Knockdown of HDAC1 significantly restores the sensitivity of LSCC cells, as HDAC1 increases the expression of interleukin-8 (IL-8), which is essential for LSCC chemoresistance. Mechanistically, HDAC1 directly initiates the transcription of IL-8 though binding to its promoter. Simultaneously, si-HDAC1 increases the levels of miR-93, which binds to the 3'UTR of IL-8 mRNA to trigger its degradation. In summary, the HDAC1/IL-8 axis can confer chemotherapeutic resistance to LSCC cells.
Collapse
|
26
|
Mak MP, Pasini FS, Diao L, Garcia FOT, Takahashi TK, Nakazato D, Martins RE, Almeida CM, Kulcsar MAV, Lamounier VA, Nunes EM, de Souza IC, Garcia MRT, Amadio AV, Siqueira SAC, Snitcovsky IML, Sichero L, Wang J, de Castro G. Valproic acid combined with cisplatin-based chemoradiation in locally advanced head and neck squamous cell carcinoma patients and associated biomarkers. Ecancermedicalscience 2020; 14:1155. [PMID: 33574900 PMCID: PMC7864693 DOI: 10.3332/ecancer.2020.1155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
Background Cisplatin-based chemoradiation (CCRT) offers locally advanced head and neck squamous cell carcinoma (LAHNSCC) patients high local control rate, however, relapses are frequent. Our goal was to evaluate if association of valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, with CCRT improved response rate (RR) and associated biomarkers. Methods This phase II trial included patients with unresectable locally advanced (LA) oropharynx (OP) squamous cell carcinoma. CCRT began after 2 weeks of VPA (P1). Primary goal was RR at 8 weeks after chemoradiation (CRT)+VPA (P2). Biomarkers included microRNA (miR) polymerase chain reaction (PCR)-array profiling in plasma compared to healthy controls by two-sample t-test. Distribution of p-values was analysed by beta-uniform mixture. Findings were validated by real-time PCR quantitative polymerase chain reaction (qPCR) for selected miRs in plasma and saliva. p16, HDAC2 and RAD23 Homolog B, Nucleotide Excision Repair Protein (HR23B) tumour immunohistochemistry were evaluated. Results Given significant toxicities, accrual was interrupted after inclusion of ten LA p16 negative OP patients. All were male, smokers/ex-smokers, aged 41–65 and with previous moderate/high alcohol intake. Nine evaluable patients yielded a RR of 88%. At false discovery rate of 5%, 169 miRs were differentially expressed between patients and controls, including lower expression of tumour suppressors (TSs) such as miR-31, -222, -let-7a/b/e and -145. miR-let-7a/e expression was validated by qPCR using saliva. A HDAC2 H-score above 170 was 90% accurate in predicting 6-month disease-free survival. Conclusions VPA and CRT offered high RR; however, with prohibitive toxicities, which led to early trial termination. Patients and controls had a distinct pattern of miR expression, mainly with low levels of TS miRs targeting Tumor protein P53 (TP53). miR-let-7a/e levels were lower in patients compared to controls, which reinforces the aggressive nature of such tumours (NCT01695122).
Collapse
Affiliation(s)
- Milena Perez Mak
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Fatima Solange Pasini
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas, MD Anderson Cancer Center, 1400 Pressler St. Floor 4, FCT4.6000, Houston, Texas, USA
| | - Fabyane O Teixeira Garcia
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Tiago Kenji Takahashi
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Denyei Nakazato
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Renata Eiras Martins
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Cristiane Maria Almeida
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Marco Aurelio Vamondes Kulcsar
- Head and Neck Surgery Department, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Valdelania Aparecida Lamounier
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Emily Montosa Nunes
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Isabela Cristina de Souza
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Marcio Ricardo Taveira Garcia
- Department of Radiology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Alex Vieira Amadio
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Sheila Aparecida C Siqueira
- Department of Pathology, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Eneas de Carvalho Aguiar, 255, CEP 05403-000, Sao Paulo, SP, Brazil
| | - Igor Moysés Longo Snitcovsky
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Laura Sichero
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Jing Wang
- Head and Neck Surgery Department, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Gilberto de Castro
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| |
Collapse
|
27
|
Hontecillas-Prieto L, Flores-Campos R, Silver A, de Álava E, Hajji N, García-Domínguez DJ. Synergistic Enhancement of Cancer Therapy Using HDAC Inhibitors: Opportunity for Clinical Trials. Front Genet 2020; 11:578011. [PMID: 33024443 PMCID: PMC7516260 DOI: 10.3389/fgene.2020.578011] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/26/2020] [Indexed: 12/25/2022] Open
Abstract
Chemotherapy is one of the most established and effective treatments for almost all types of cancer. However, the elevated toxicity due to the non-tumor-associated effects, development of secondary malignancies, infertility, radiation-induced fibrosis and resistance to treatment limit the effectiveness and safety of treatment. In addition, these multiple factors significantly impact quality of life. Over the last decades, our increased understanding of cancer epigenetics has led to new therapeutic approaches and the promise of improved patient outcomes. Epigenetic alterations are commonly found in cancer, especially the increased expression and activity of histone deacetylases (HDACs). Dysregulation of HDACs are critical to the development and progression of the majority of tumors. Hence, HDACs inhibitors (HDACis) were developed and now represent a very promising treatment strategy. The use of HDACis as monotherapy has shown very positive pre-clinical results, but clinical trials have had only limited success. However, combinatorial regimens with other cancer drugs have shown synergistic effects both in pre-clinical and clinical studies. At the same time, these combinations have enhanced the efficacy, reduced the toxicity and tumor resistance to therapy. In this review, we will examine examples of HDACis used in combination with other cancer drugs and highlight the synergistic effects observed in recent preclinical and clinical studies.
Collapse
Affiliation(s)
- Lourdes Hontecillas-Prieto
- Institute of Biomedicine of Seville, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla/CIBERONC, Seville, Spain
| | - Rocío Flores-Campos
- Institute of Biomedicine of Seville, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla/CIBERONC, Seville, Spain
| | - Andrew Silver
- Centre for Genomics and Child Health, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Enrique de Álava
- Institute of Biomedicine of Seville, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla/CIBERONC, Seville, Spain.,Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, Seville, Spain
| | - Nabil Hajji
- Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Daniel J García-Domínguez
- Institute of Biomedicine of Seville, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla/CIBERONC, Seville, Spain
| |
Collapse
|
28
|
Iannelli F, Zotti AI, Roca MS, Grumetti L, Lombardi R, Moccia T, Vitagliano C, Milone MR, Ciardiello C, Bruzzese F, Leone A, Cavalcanti E, De Cecio R, Iachetta G, Valiante S, Ionna F, Caponigro F, Di Gennaro E, Budillon A. Valproic Acid Synergizes With Cisplatin and Cetuximab in vitro and in vivo in Head and Neck Cancer by Targeting the Mechanisms of Resistance. Front Cell Dev Biol 2020; 8:732. [PMID: 33015030 PMCID: PMC7461984 DOI: 10.3389/fcell.2020.00732] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
Recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC) is a devastating malignancy with a poor prognosis. The combination of cisplatin (CDDP) plus cetuximab (CX) is one of the standard first-line treatments in this disease. However, this therapeutic regimen is often associated with high toxicity and resistance, suggesting that new combinatorial strategies are needed to improve its therapeutic index. In our study, we evaluated the antitumor effects of valproic acid (VPA), a well-known antiepileptic agent with histone deacetylase inhibitory activity, in combination with CDDP/CX doublet in head and neck squamous cell carcinoma (HNSCC) models. We demonstrated, in HNSCC cell lines, but not in normal human fibroblasts, that simultaneous exposure to equitoxic doses of VPA plus CDDP/CX resulted in a clear synergistic antiproliferative and pro-apoptotic effects. The synergistic antitumor effect was confirmed in four different 3D-self-assembled spheroid models, suggesting the ability of the combined approach to affect also the cancer stem cells compartment. Mechanistically, VPA enhanced DNA damage in combination treatment by reducing the mRNA expression of ERCC Excision Repair 1, a critical player in DNA repair, and by increasing CDDP intracellular concentration via upregulation at transcriptional level of CDDP influx channel copper transporter 1 and downregulation of the ATPAse ATP7B involved in CDDP-export. Valproic acid also induced a dose-dependent downregulation of epidermal growth factor receptor (EGFR) expression and of MAPK and AKT downstream signaling pathways and prevent CDDP- and/or CX-induced EGFR nuclear translocation, a well-known mechanism of resistance to chemotherapy. Indeed, VPA impaired the transcription of genes induced by non-canonical activity of nuclear EGFR, such as cyclin D1 and thymidylate synthase. Finally, we confirmed the synergistic antitumor effect also in vivo in both heterotopic and orthotopic models, demonstrating that the combined treatment completely blocked HNSCC xenograft tumors growth in nude mice. Overall, the introduction of a safe and generic drug such as VPA into the conventional treatment for R/M HNSCC represents an innovative and feasible antitumor strategy that warrants further clinical evaluation. A phase II clinical trial exploring the combination of VPA and CDDP/CX in R/M HNSCC patients is currently ongoing in our institute.
Collapse
Affiliation(s)
- Federica Iannelli
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Andrea Ilaria Zotti
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Laura Grumetti
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Rita Lombardi
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Tania Moccia
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Maria Rita Milone
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Chiara Ciardiello
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Francesca Bruzzese
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Ernesta Cavalcanti
- Laboratory Medicine Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Rossella De Cecio
- Pathology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | | | | | - Franco Ionna
- Maxillo-facial & ENT Surgery Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Francesco Caponigro
- Head and Neck Medical Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| |
Collapse
|
29
|
Epithelial to Mesenchymal Transition: A Mechanism that Fuels Cancer Radio/Chemoresistance. Cells 2020; 9:cells9020428. [PMID: 32059478 PMCID: PMC7072371 DOI: 10.3390/cells9020428] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/27/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) contributes to tumor progression, cancer cell invasion, and therapy resistance. EMT is regulated by transcription factors such as the protein products of the SNAI gene family, which inhibits the expression of epithelial genes. Several signaling pathways, such as TGF-beta1, IL-6, Akt, and Erk1/2, trigger EMT responses. Besides regulatory transcription factors, RNA molecules without protein translation, micro RNAs, and long non-coding RNAs also assist in the initialization of the EMT gene cluster. A challenging novel aspect of EMT research is the investigation of the interplay between tumor microenvironments and EMT. Several microenvironmental factors, including fibroblasts and myofibroblasts, as well as inflammatory, immune, and endothelial cells, induce EMT in tumor cells. EMT tumor cells change their adverse microenvironment into a tumor friendly neighborhood, loaded with stromal regulatory T cells, exhausted CD8+ T cells, and M2 (protumor) macrophages. Several EMT inhibitory mechanisms are instrumental in reversing EMT or targeting EMT cells. Currently, these mechanisms are also significant for clinical use.
Collapse
|
30
|
Kim Y, Wang SE, Jiang YH. Epigenetic therapy of Prader-Willi syndrome. Transl Res 2019; 208:105-118. [PMID: 30904443 PMCID: PMC6527448 DOI: 10.1016/j.trsl.2019.02.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 01/05/2023]
Abstract
Prader-Willi syndrome (PWS) is a complex and multisystem neurobehavioral disorder. The molecular mechanism of PWS is deficiency of paternally expressed gene gene or genes from the chromosome 15q11-q13. Due to imprinted gene regulation, the same genes in the maternal chromosome 15q11-q13 are structurally intact but transcriptionally repressed by an epigenetic mechanism. The unique molecular defect underlying PWS renders an exciting opportunity to explore epigenetic-based therapy to reactivate the expression of repressed PWS genes from the maternal chromosome. Inactivation of H3K9m3 methyltransferase SETDB1 and zinc finger protein ZNF274 results in reactivation of SNRPN and SNORD116 cluster from the maternal chromosomes in PWS patient iPSCs and iPSC-derived neurons, respectively. High content screening of small molecule libraries using cells derived from transgenic mice carrying the SNRPN-EGFP fusion protein has discovered that inhibitors of EHMT2/G9a, a histone 3 lysine 9 methyltransferase, are capable of reactivating expression of paternally expressed SNRPN and SNORD116 from the maternal chromosome, both in cultured PWS patient-derived fibroblasts and in a PWS mouse model. Treatment with an EMHT2/G9a inhibitor also rescues perinatal lethality and failure to thrive phenotypes in a PWS mouse model. These findings present the first evidence to support a proof-of-principle for epigenetic-based therapy for the PWS in humans.
Collapse
Affiliation(s)
- Yuna Kim
- Department of Pediatrics, Duke University of School of Medicine, Durham, North Carolina
| | - Sung Eun Wang
- Department of Pediatrics, Duke University of School of Medicine, Durham, North Carolina
| | - Yong-Hui Jiang
- Department of Pediatrics, Duke University of School of Medicine, Durham, North Carolina; Department of Neurobiology, Duke University of School of Medicine, Durham, North Carolina; Department of Program in Genetics and Genomics, Duke University of School of Medicine, Durham, North Carolina; Department of Program in Cellular and Molecular Biology, Duke University of School of Medicine, Durham, North Carolina.
| |
Collapse
|
31
|
Hamdan FH, Johnsen SA. Perturbing Enhancer Activity in Cancer Therapy. Cancers (Basel) 2019; 11:cancers11050634. [PMID: 31067678 PMCID: PMC6563029 DOI: 10.3390/cancers11050634] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023] Open
Abstract
Tight regulation of gene transcription is essential for normal development, tissue homeostasis, and disease-free survival. Enhancers are distal regulatory elements in the genome that provide specificity to gene expression programs and are frequently misregulated in cancer. Recent studies examined various enhancer-driven malignant dependencies and identified different approaches to specifically target these programs. In this review, we describe numerous features that make enhancers good transcriptional targets in cancer therapy and discuss different approaches to overcome enhancer perturbation. Interestingly, a number of approved therapeutic agents, such as cyclosporine, steroid hormones, and thiazolidinediones, actually function by affecting enhancer landscapes by directly targeting very specific transcription factor programs. More recently, a broader approach to targeting deregulated enhancer programs has been achieved via Bromodomain and Extraterminal (BET) inhibition or perturbation of transcription-related cyclin-dependent kinases (CDK). One challenge to enhancer-targeted therapy is proper patient stratification. We suggest that monitoring of enhancer RNA (eRNA) expression may serve as a unique biomarker of enhancer activity that can help to predict and monitor responsiveness to enhancer-targeted therapies. A more thorough investigation of cancer-specific enhancers and the underlying mechanisms of deregulation will pave the road for an effective utilization of enhancer modulators in a precision oncology approach to cancer treatment.
Collapse
Affiliation(s)
- Feda H Hamdan
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Steven A Johnsen
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
32
|
Gatti V, Fierro C, Annicchiarico-Petruzzelli M, Melino G, Peschiaroli A. ΔNp63 in squamous cell carcinoma: defining the oncogenic routes affecting epigenetic landscape and tumour microenvironment. Mol Oncol 2019; 13:981-1001. [PMID: 30845357 PMCID: PMC6487733 DOI: 10.1002/1878-0261.12473] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 12/20/2022] Open
Abstract
Squamous cell carcinoma (SCC) is a treatment‐refractory tumour which arises from the epithelium of diverse anatomical sites such as oesophagus, head and neck, lung and skin. Accumulating evidence has revealed a number of genomic, clinical and molecular features commonly observed in SCC of distinct origins. Some of these genetic events culminate in fostering the activity of ΔNp63, a potent oncogene which exerts its pro‐tumourigenic effects by regulating specific transcriptional programmes to sustain malignant cell proliferation and survival. In this review, we will describe the genetic and epigenetic determinants underlying ΔNp63 oncogenic activities in SCC, and discuss some relevant transcriptional effectors of ΔNp63, emphasizing their impact in modulating the crosstalk between tumour cells and tumour microenvironment (TME).
Collapse
Affiliation(s)
- Veronica Gatti
- Department of Experimental Medicine, TOR, University of Rome, Tor Vergata, Italy
| | - Claudia Fierro
- Department of Experimental Medicine, TOR, University of Rome, Tor Vergata, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome, Tor Vergata, Italy.,Medical Research Council, Toxicology Unit, University of Cambridge, UK
| | - Angelo Peschiaroli
- National Research Council of Italy, Institute of Translational Pharmacology, Rome, Italy
| |
Collapse
|