1
|
Wahyudianingsih R, Sanjaya A, Jonathan T, Pranggono EH, Achmad D, Hernowo BS. Chemotherapy's effects on autophagy in the treatment of Hodgkin's lymphoma: a scoping review. Discov Oncol 2024; 15:269. [PMID: 38976168 PMCID: PMC11231119 DOI: 10.1007/s12672-024-01142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Classical Hodgkin Lymphomas (HL) are a unique malignant growth with an excellent initial prognosis. However, 10-30% of patients will still relapse after remission. One primary cellular function that has been the focus of tumor progression is autophagy. This process can preserve cellular homeostasis under stressful conditions. Several studies have shown that autophagy may play a role in developing HL. Therefore, this review aimed to explore chemotherapy's effect on autophagy in HL, and the effects of autophagy on HL. METHODS A scoping review in line with the published PRISMA extension for scoping reviews (PRISMA-ScR) was conducted. A literature search was conducted on the MEDLINE database and the Cochrane Central Register of Controlled Trials (CENTRAL). All results were retrieved and screened, and the resulting articles were synthesized narratively. RESULTS The results showed that some cancer chemotherapy also induces autophagic flux. Although the data on HL is limited, since the mechanisms of action of these drugs are similar, we can infer a similar relationship. However, this increased autophagy activity may reflect a mechanism for increasing tumor growth or a cellular compensation to inhibit its growth. Although evidence supports both views, we argued that autophagy allowed cancer cells to resist cell death, mainly due to DNA damage caused by cytotoxic drugs. CONCLUSION Autophagy reflects the cell's adaptation to survive and explains why chemotherapy generally induces autophagy functions. However, further research on autophagy inhibition is needed as it presents a viable treatment strategy, especially against drug-resistant populations that may arise from HL chemotherapy regimens.
Collapse
Affiliation(s)
- Roro Wahyudianingsih
- Postgraduate Program of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
- Department of Anatomical Pathology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Ardo Sanjaya
- Department of Anatomy, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia.
| | - Timothy Jonathan
- Undergraduate Program in Medicine, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia
| | - Emmy Hermiyanti Pranggono
- Department of Internal Medicine, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| | - Dimyati Achmad
- Department of Oncological Surgery, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| | - Bethy Suryawathy Hernowo
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| |
Collapse
|
2
|
Ho T, Coleman C, Shah P, Yazbeck V. Advances in Hodgkin's lymphoma pharmacotherapy: a focus on histone deacetylase inhibitors. Expert Opin Pharmacother 2023; 24:1427-1438. [PMID: 37249399 DOI: 10.1080/14656566.2023.2219392] [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: 03/16/2023] [Accepted: 05/25/2023] [Indexed: 05/31/2023]
Abstract
INTRODUCTION Classical Hodgkin lymphomas (cHL) usually have excellent cure rates. Yet, for patients with refractory or relapsed cHL, prognosis deteriorates as the disease becomes resistant to subsequent lines of therapies: autologous stem cell transplantation, brentuximab vedotin, and checkpoint inhibitors. Immune escape and drug resistance are hallmarks of Hodgkin Reed Sternberg cell survival, prompting the need for additional therapeutic strategies. Histone modification-based combination is an effective clinical strategy. AREAS COVERED In this review, we discuss the different histone deacetylase (HDAC) inhibitor molecules that have been developed and studied in cancer therapy with a focus on cHL. We review their preclinical and clinical activities both as single agents and in combination studies. Literature search was conducted in PubMed, Google Scholar, and ClinicalTrials.gov databases, using search terms 'Hodgkin lymphoma,' 'histone deacetylase inhibitor', and variations on such (e.g. 'HDAC' and individual drug names) in combination using operators 'AND,' 'OR,' and 'NOT' according to Boolean logic. EXPERT OPINION HDAC inhibitors alone will not be sufficient for the treatment of R/RcHL, but given their disease control capacity, synergistic interaction with currently approved drugs, and ability to overcome drug resistance, particularly PD-1 inhibitors, we believe that HDACinhibitors will eventually become incorporated into the treatment armamentarium of cHL.
Collapse
Affiliation(s)
- Thuy Ho
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Cara Coleman
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Palak Shah
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Victor Yazbeck
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
3
|
Yang FF, Hu T, Liu JQ, Yu XQ, Ma LY. Histone deacetylases (HDACs) as the promising immunotherapeutic targets for hematologic cancer treatment. Eur J Med Chem 2023; 245:114920. [PMID: 36399875 DOI: 10.1016/j.ejmech.2022.114920] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/25/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Bone marrow transplantation is regarded as the most effective immunotherapy for hematologic cancer, but it generally faces difficulties in matching. Aberrant expression of histone deacetylases (HDACs) is closely related to the occurrence and development of hematological cancer. Recent studies suggested that HDACs might play a critical role in initiating anti-cancer immune response or enhancing anti-cancer immunotherapy. Besides, combining HDAC inhibition and immunotherapy could prevent immunotherapy resistance in some degree and reach an extended treatment window. This review summarized the relationship between HDACs and immune and described the current understanding of HDACs in immunotherapy for hematologic cancer.
Collapse
Affiliation(s)
- Fei-Fei Yang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Ting Hu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Jian-Quan Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Xiao-Qian Yu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Li-Ying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China; China Meheco Topfond Pharmaceutical Co., Key Laboratory of Cardio-cerebrovascular Drug, Zhumadian, 463000, PR China.
| |
Collapse
|
4
|
Autophagy in Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14205072. [PMID: 36291856 PMCID: PMC9600546 DOI: 10.3390/cancers14205072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Autophagy is a dynamic and tightly regulated process that seems to have dual effects in cancer. In some contexts, it can induce carcinogenesis and promote cancer cell survival, whereas in others, it acts preventing tumor cell growth and tumor progression. Thus, autophagy functions seem to strictly depend on cancer ontogenesis, progression, and type. Here, we will dive into the current knowledge of autophagy in hematological malignancies and will highlight the main genetic components involved in each cancer type. Abstract Autophagy is a highly conserved metabolic pathway via which unwanted intracellular materials, such as unfolded proteins or damaged organelles, are digested. It is activated in response to conditions of oxidative stress or starvation, and is essential for the maintenance of cellular homeostasis and other vital functions, such as differentiation, cell death, and the cell cycle. Therefore, autophagy plays an important role in the initiation and progression of tumors, including hematological malignancies, where damaged autophagy during hematopoiesis can cause malignant transformation and increase cell proliferation. Over the last decade, the importance of autophagy in response to standard pharmacological treatment of hematological tumors has been observed, revealing completely opposite roles depending on the tumor type and stage. Thus, autophagy can promote tumor survival by attenuating the cellular damage caused by drugs and/or stabilizing oncogenic proteins, but can also have an antitumoral effect due to autophagic cell death. Therefore, autophagy-based strategies must depend on the context to create specific and safe combination therapies that could contribute to improved clinical outcomes. In this review, we describe the process of autophagy and its role on hematopoiesis, and we highlight recent research investigating its role as a potential therapeutic target in hematological malignancies. The findings suggest that genetic variants within autophagy-related genes modulate the risk of developing hemopathies, as well as patient survival.
Collapse
|
5
|
Lu X, Liu M, Yang J, Yi Q, Zhang X. Panobinostat enhances NK cell cytotoxicity in soft tissue sarcoma. Clin Exp Immunol 2022; 209:127-139. [PMID: 35867577 DOI: 10.1093/cei/uxac068] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/15/2022] [Accepted: 07/21/2022] [Indexed: 11/12/2022] Open
Abstract
Sarcoma is a rare and heterogeneous class of mesenchymal malignancies with poor prognosis. Panobinostat (LBH589) as one of histone deacetylase (HDAC) inhibitors, has demonstrated anti-tumor activity in patients with sarcoma, but its mechanisms remains unclear. Here, we found that LBH589 alone inhibited the proliferation and colony formation of soft tissue sarcoma(STS) cell lines. Transcriptome analysis showed that treatment with LBH589 augmented the NK cell mediated cytotoxicity. Quantitative real-time PCR and flow cytometric analysis (FACS) further confirmed that LBH589 increased the expression of NKG2D ligands MICA/MICB. Mechanistically, LBH589 activated the Wnt/β-catenin pathway by upregulating the histone acetylation in β-catenin promoter. In vitro co-culture experiments and in vivo animal experiments showed that LBH589 increased the cytotoxicity of natural killer (NK) cells while Wnt/β-catenin inhibitor decreased the effects. Our findings suggests that LBH589 facilitates the anti-tumor effect of NK cells, highlights LBH589 an effective assistance drug in NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Xiuxia Lu
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Jing Yang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Que Yi
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| |
Collapse
|
6
|
Larragoite ET, Nell RA, Martins LJ, Barrows LR, Planelles V, Spivak AM. Histone deacetylase inhibition reduces deleterious cytokine release induced by ingenol stimulation. Biochem Pharmacol 2022; 195:114844. [PMID: 34801521 PMCID: PMC8712404 DOI: 10.1016/j.bcp.2021.114844] [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] [Received: 09/29/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 01/03/2023]
Abstract
Latency reversing agents (LRAs), such as protein kinase C (PKC) agonists, constitute a promising strategy for exposing and eliminating the HIV-1 latent reservoir. PKC agonists activate NF-κB and induce deleterious pro-inflammatory cytokine production. Adjuvant pharmacological agents, such as ruxolitinib, a JAK inhibitor, have previously been combined with LRAs to reduce deleterious pro-inflammatory cytokine secretion without inhibiting HIV-1 reactivation in vitro. Histone deacetylase inhibitors (HDACi) are known to dampen pro-inflammatory cytokine secretion in the context of other diseases and synergize with LRAs to reactivate latent HIV-1. This study investigates whether a panel of epigenetic modifiers, including HDACi, could dampen PKC-induced pro-inflammatory cytokine secretion during latency reversal. We screened an epigenetic modifier library for compounds that reduced intracellular IL-6 production induced by the PKC agonist Ingenol-3,20-dibenzoate. We further tested the most promising epigenetic inhibitor class, HDACi, for their ability to reduce pro-inflammatory cytokines and reactivate latent HIV-1 ex vivo. We identified nine epigenetic modulators that reduced PKC-induced intracellular IL-6. In cells from aviremic individuals living with HIV-1, the HDAC1-3 inhibitor, suberohydroxamic acid (SBHA), reduced secretion of pro-inflammatory cytokines TNF-α, IL-5, IL-2r, and IL-17 but did not significantly reactivate latent HIV-1 when combined with Ingenol-3,20-dibenzoate. Combining SBHA and Ingenol-3,20-dibenzoate reduces deleterious cytokine production during latency reversal but does not induce significant viral reactivation in aviremic donor PBMCs. The ability of SBHA to reduce PKC-induced pro-inflammatory cytokines when combined with Ingenol-3,20-dibenzoate suggests SBHA can be used to reduced PKC induced pro-inflammatory cytokines but not to achieve latency reversal in the context of HIV-1.
Collapse
Affiliation(s)
- Erin T. Larragoite
- Department of Pathology, University of Utah, Salt Lake City, United States
| | - Racheal A. Nell
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, United States
| | - Laura J. Martins
- Department of Pathology, University of Utah, Salt Lake City, United States
| | - Louis R. Barrows
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, United States
| | - Vicente Planelles
- Department of Pathology, University of Utah, Salt Lake City, United States
| | - Adam M. Spivak
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, United States, Corresponding Author: Adam M. Spivak, 50 North Medical Drive, Division of Infectious Diseases, Room 4B319, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, Phone: 801-587-1964, Fax: 801-585-3377,
| |
Collapse
|
7
|
Afolabi LO, Bi J, Li X, Adeshakin AO, Adeshakin FO, Wu H, Yan D, Chen L, Wan X. Synergistic Tumor Cytolysis by NK Cells in Combination With a Pan-HDAC Inhibitor, Panobinostat. Front Immunol 2021; 12:701671. [PMID: 34531855 PMCID: PMC8438531 DOI: 10.3389/fimmu.2021.701671] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/17/2021] [Indexed: 01/18/2023] Open
Abstract
Histone deacetylases (HDAC) are frequently overexpressed in tumors, and their inhibition has shown promising anti-tumor effects. However, the synergistic effects of HDAC inhibition with immune cell therapy have not been fully explored. Natural killer (NK) cells are cytotoxic lymphocytes for anti-tumor immune surveillance, with immunotherapy potential. We showed that a pan-HDAC inhibitor, panobinostat, alone demonstrated anti-tumor and anti-proliferative activities on all tested tumors in vitro. Additionally, panobinostat co-treatment or pretreatment synergized with NK cells to mediate tumor cell cytolysis. Mechanistically, panobinostat treatment increased the expression of cell adhesion and tight junction-related genes, promoted conjugation formation between NK and tumor cells, and modulates NK cell-activating receptors and ligands on tumor cells, contributing to the increased tumor cytolysis. Finally, panobinostat therapy led to better tumor control and synergized with anti-PD-L1 therapy. Our data highlights the anti-tumor potential of HDAC inhibition through tumor-intrinsic toxicity and enhancement of NK -based immunotherapy.
Collapse
Affiliation(s)
- Lukman O. Afolabi
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiacheng Bi
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xuguang Li
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, China
| | - Adeleye O. Adeshakin
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Funmilayo O. Adeshakin
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Haisi Wu
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dehong Yan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liang Chen
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaochun Wan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
8
|
Study of the antilymphoma activity of pracinostat reveals different sensitivities of DLBCL cells to HDAC inhibitors. Blood Adv 2021; 5:2467-2480. [PMID: 33999145 DOI: 10.1182/bloodadvances.2020003566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
Histone deacetylase inhibitors (HDACis) are antitumor agents with distinct efficacy in hematologic tumors. Pracinostat is a pan-HDACi with promising early clinical activity. However, similar to other HDACis, its activity as a single agent is limited. Diffuse large B-cell lymphoma (DLBCL) includes distinct molecular subsets or metabolically defined subtypes that rely in different ways on the B-cell receptor signaling pathway, oxidative phosphorylation, and glycolysis for their survival. The antitumor activity of pracinostat has not been determined in lymphomas. We performed preclinical in vitro activity screening of 60 lymphoma cell lines that included 25 DLBCLs. DLBCL cells belonging to distinct metabolic subtypes were treated with HDACis for 6 hours or 14 days followed by transcriptional profiling. DLBCL xenograft models enabled assessment of the in vivo antilymphoma activity of pracinostat. Combination treatments with pracinostat plus 10 other antilymphoma agents were performed. Western blot was used to assess acetylation levels of histone and nonhistone proteins after HDACi treatment. Robust antiproliferative activity was observed across all lymphoma histotypes represented. Focusing on DLBCL, we identified a low-sensitivity subset that almost exclusively consists of the oxidative phosphorylation (OxPhos)-DLBCL metabolic subtype. OxPhos-DLBCL cells also showed poorer sensitivity to other HDACis, including vorinostat. Transcriptomic analysis revealed fewer modulated transcripts but an enrichment of antioxidant pathway genes after HDACi treatment of OxPhos-DLBCLs compared with high-sensitivity B-cell receptor (BCR)-DLBCLs. Pharmacologic inhibition of antioxidant production rescued sensitivity of OxPhos-DLBCLs to pracinostat whereas BCR-DLBCLs were unaffected. Our study provides novel insights into the antilymphoma activity of pracinostat and identifies a differential response of DLBCL metabolic subtypes to HDACis.
Collapse
|
9
|
Lin HC, Chang Y, Chen RY, Hung LY, Chen PCH, Chen YP, Medeiros LJ, Chiang PM, Chang KC. Epstein-Barr virus latent membrane protein-1 upregulates autophagy and promotes viability in Hodgkin lymphoma: Implications for targeted therapy. Cancer Sci 2021; 112:1589-1602. [PMID: 33525055 PMCID: PMC8019199 DOI: 10.1111/cas.14833] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/17/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Hodgkin lymphoma (HL) is composed of neoplastic Hodgkin and Reed‐Sternberg cells in an inflammatory background. The neoplastic cells are derived from germinal center B cells that, in most cases, are infected by Epstein‐Barr virus (EBV), which may play a role in tumorigenesis. Given that EBV‐latent membrane protein 1 (LMP1) regulates autophagy in B cells, we explored the role of autophagy mediated by EBV or LMP1 in HL. We found that EBV‐LMP1 transfection in HL cells induced a modest increase in autophagy signals, attenuated starvation‐induced autophagic stress, and alleviated autophagy inhibition‐ or doxorubicin‐induced cell death. LMP1 knockdown leads to decreased autophagy LC3 signals. A xenograft mouse model further showed that EBV infection significantly increased expression of the autophagy marker LC3 in HL cells. Clinically, LC3 was expressed in 15% (19/127) of HL samples, but was absent in all cases of nodular lymphocyte‐predominant and lymphocyte‐rich classic HL cases. Although expression of LC3 was not correlated with EBV status or clinical outcome, autophagic blockade effectively eradicated LMP1‐positive HL xenografts with better efficacy than LMP1‐negative HL xenografts. Collectively, these results suggest that EBV‐LMP1 enhances autophagy and promotes the viability of HL cells. Autophagic inhibition may be a potential therapeutic strategy for treating patients with HL, especially EBV‐positive cases.
Collapse
Affiliation(s)
- Hui-Chen Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yao Chang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Ruo-Yu Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Liang-Yi Hung
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan.,PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | | | - Ya-Ping Chen
- Division of Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Po-Min Chiang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kung-Chao Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, Kaohsiung Medical University Hospital, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
10
|
He Y, Xu L, Feng J, Wu K, Zhao Y, Huang H. HDAC Inhibitor LBH589 Suppresses the Proliferation but Enhances the Antileukemic Effect of Human γδT Cells. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:623-630. [PMID: 33005729 PMCID: PMC7515977 DOI: 10.1016/j.omto.2020.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 08/06/2020] [Indexed: 12/17/2022]
Abstract
γδT cells have potent effects on hematological malignancies, and their functions can be regulated by anti-tumor agents. Histone deacetylase inhibitors (HDACis) not only have antileukemic activity on leukemia but also affect immune cells during therapeutic application. In this in vitro study, we showed that LBH589, a pan-HDACi, impaired the proliferation of human γδT cells, as well as their proportions in peripheral blood mononuclear cells (PBMCs). At the specific concentration, LBH589 induced significant antileukemic activity of γδT cells against the HL-60 cells and Kasumi cells in a dose-dependent manner. However, the expression levels of activating receptor and molecules, as well as interferon-γ (IFN-γ) expression on γδT cells, were not affected by LBH589. After treatment with LBH589 for indicated times, extracellular-regulated protein kinase (ERK), Akt, and c-Jun N-terminal kinase (JNK) signaling pathways in γδT cells were not activated. In contrast, a stronger expression of Notch was observed and sustained for 72 h. Inhibition of Notch signaling by FLI-06, the γ-secretase inhibitor, significantly reversed the enhanced antileukemic ability of γδT cells induced by LBH589. For the first time, our investigations demonstrate that LBH589 can inhibit proliferation of γδT cells but facilitate their antileukemic effects via activation of Notch signaling.
Collapse
Affiliation(s)
- Ying He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
- Zhejiang Provincial People’s Hospital, 158 Shangtang Road, Hangzhou, Zhejiang 310014, China
- Institute of Hematology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Lin Xu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jingjing Feng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Kangni Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yanmin Zhao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
- Corresponding author: Yanmin Zhao, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
- Corresponding author: He Huang, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.
| |
Collapse
|
11
|
Wang X, Waschke BC, Woolaver RA, Chen SMY, Chen Z, Wang JH. HDAC inhibitors overcome immunotherapy resistance in B-cell lymphoma. Protein Cell 2020; 11:472-482. [PMID: 32162275 PMCID: PMC7305292 DOI: 10.1007/s13238-020-00694-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has been applied successfully to treat B-cell lymphomas in preclinical models or clinical settings. However, immunotherapy resistance is a major challenge for B-cell lymphoma treatment. To overcome this issue, combinatorial therapeutic strategies have been pursued to achieve a better efficacy for treating B-cell lymphomas. One of such strategies is to combine immunotherapy with histone deacetylase (HDAC) inhibitors. HDAC inhibitors can potentially increase tumor immunogenicity, promote anti-tumor immune responses, or reverse immunosuppressive tumor environments. Thus, the combination of HDAC inhibitors and immunotherapy has drawn much attention in current cancer treatment. However, not all HDAC inhibitors are created equal and their net effects are highly dependent on the specific inhibitors used and the HDACs they target. Hence, we suggest that optimal treatment efficacy requires personalized design and rational combination based on prognostic biomarkers and unique profiles of HDAC inhibitors. Here, we discuss the possible mechanisms by which B-cell lymphomas acquire immunotherapy resistance and the effects of HDAC inhibitors on tumor cells and immune cells that could help overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Xiaoguang Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Brittany C Waschke
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Rachel A Woolaver
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Samantha M Y Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Zhangguo Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Jing H Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA.
| |
Collapse
|
12
|
Abstract
Introduction: T-cell lymphomas represent a broad group of malignant T-cell neoplasms with marked molecular, clinical, and biologic heterogeneity. Survival rates after conventional chemotherapy regimens are poor for most subtypes and new therapies are needed. Rapidly expanding knowledge in the field of epigenomics and the development of an increasing number of epigenetic-modifying agents have created new opportunities for epigenetic therapies for patients with this complex group of diseases.Areas covered: The present review summarizes current knowledge on epigenetic alterations in T-cell lymphomas, availability, and mechanisms of action of epigenetic-modifying agents, results of clinical trials of epigenetic therapies in T-cell lymphomas, status of FDA approval, and biomarker approaches to guide therapy. Promising future directions are discussed.Expert opinion: Mutations in epigenetic-modifying genes are among the most common genetic alterations in T-cell lymphomas, highlighting the potential for epigenetic therapies to improve management of this group of diseases. Single-agent efficacy is well documented, leading to FDA approval for several indications, but overall response rates and durability of responses remain modest. Critical next steps for the field include optimizing combination therapies that incorporate epigenetic-modifying agents and developing predictive biomarkers that help guide patient and drug selection.
Collapse
Affiliation(s)
- Nada Ahmed
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.,Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
13
|
Epigenetic Control of Autophagy in Cancer Cells: A Key Process for Cancer-Related Phenotypes. Cells 2019; 8:cells8121656. [PMID: 31861179 PMCID: PMC6952790 DOI: 10.3390/cells8121656] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/19/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Although autophagy is a well-known and extensively described cell pathway, numerous studies have been recently interested in studying the importance of its regulation at different molecular levels, including the translational and post-translational levels. Therefore, this review focuses on the links between autophagy and epigenetics in cancer and summarizes the. following: (i) how ATG genes are regulated by epigenetics, including DNA methylation and post-translational histone modifications; (ii) how epidrugs are able to modulate autophagy in cancer and to alter cancer-related phenotypes (proliferation, migration, invasion, tumorigenesis, etc.) and; (iii) how epigenetic enzymes can also regulate autophagy at the protein level. One noteable observation was that researchers most often reported conclusions about the regulation of the autophagy flux, following the use of epidrugs, based only on the analysis of LC3B-II form in treated cells. However, it is now widely accepted that an increase in LC3B-II form could be the consequence of an induction of the autophagy flux, as well as a block in the autophagosome-lysosome fusion. Therefore, in our review, all the published results describing a link between epidrugs and autophagy were systematically reanalyzed to determine whether autophagy flux was indeed increased, or inhibited, following the use of these potentially new interesting treatments targeting the autophagy process. Altogether, these recent data strongly support the idea that the determination of autophagy status could be crucial for future anticancer therapies. Indeed, the use of a combination of epidrugs and autophagy inhibitors could be beneficial for some cancer patients, whereas, in other cases, an increase of autophagy, which is frequently observed following the use of epidrugs, could lead to increased autophagy cell death.
Collapse
|
14
|
Sermer D, Pasqualucci L, Wendel HG, Melnick A, Younes A. Emerging epigenetic-modulating therapies in lymphoma. Nat Rev Clin Oncol 2019; 16:494-507. [PMID: 30837715 DOI: 10.1038/s41571-019-0190-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite considerable advances in the treatment of lymphoma, the prognosis of patients with relapsed and/or refractory disease continues to be poor; thus, a continued need exists for the development of novel approaches and therapies. Epigenetic dysregulation might drive and/or promote tumorigenesis in various types of malignancies and is prevalent in both B cell and T cell lymphomas. Over the past decade, a large number of epigenetic-modifying agents have been developed and introduced into the clinical management of patients with haematological malignancies. In this Review, we provide a concise overview of the most promising epigenetic therapies for the treatment of lymphomas, including inhibitors of histone deacetylases (HDACs), DNA methyltransferases (DNMTs), enhancer of zeste homologue 2 (EZH2), bromodomain and extra-terminal domain proteins (BETs), protein arginine N-methyltransferases (PRMTs) and isocitrate dehydrogenases (IDHs), and highlight the most promising future directions of research in this area.
Collapse
Affiliation(s)
- David Sermer
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Hans-Guido Wendel
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ari Melnick
- Weill-Cornell Medical College, New York, NY, USA
| | - Anas Younes
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
15
|
Zhao LM, Zhang JH. Histone Deacetylase Inhibitors in Tumor Immunotherapy. Curr Med Chem 2019; 26:2990-3008. [PMID: 28762309 DOI: 10.2174/0929867324666170801102124] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 04/26/2017] [Accepted: 06/27/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND With an increasing understanding of the antitumor immune response, considerable progress has been made in the field of tumor immunotherapy in the last decade. Inhibition of histone deacetylases represents a new strategy in tumor therapy and histone deacetylase inhibitors have been recently developed and validated as potential antitumor drugs. In addition to the direct antitumor effects, histone deacetylase inhibitors have been found to have the ability to improve tumor recognition by immune cells that may contribute to their antitumor activity. These immunomodolutory effects are desirable, and their in-depth comprehension will facilitate the design of novel regimens with improved clinical efficacy. OBJECTIVE Our goal here is to review recent developments in the application of histone deacetylase inhibitors as immune modulators in cancer treatment. METHODS Systemic compilation of the relevant literature in this field. RESULTS & CONCLUSION In this review, we summarize recent advances in the understanding of how histone deacetylase inhibitors alter immune process and discuss their effects on various cytokines. We also discuss the challenges to optimize the use of these inhibitors as immune modulators in cancer treatment. Information gained from this review will be valuable to this field and may be helpful for designing tumor immunotherapy trials involving histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Li-Ming Zhao
- School of Chemistry and Chemical Engineering, and Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.,State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Jie-Huan Zhang
- School of Chemistry and Chemical Engineering, and Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| |
Collapse
|
16
|
Ribeiro ML, Reyes-Garau D, Armengol M, Fernández-Serrano M, Roué G. Recent Advances in the Targeting of Epigenetic Regulators in B-Cell Non-Hodgkin Lymphoma. Front Genet 2019; 10:986. [PMID: 31681423 PMCID: PMC6807552 DOI: 10.3389/fgene.2019.00986] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
In the last 10 years, major advances have been made in the diagnosis and development of selective therapies for several blood cancers, including B-cell non-Hodgkin lymphoma (B-NHL), a heterogeneous group of malignancies arising from the mature B lymphocyte compartment. However, most of these entities remain incurable and current treatments are associated with variable efficacy, several adverse events, and frequent relapses. Thus, new diagnostic paradigms and novel therapeutic options are required to improve the prognosis of patients with B-NHL. With the recent deciphering of the mutational landscapes of B-cell disorders by high-throughput sequencing, it came out that different epigenetic deregulations might drive and/or promote B lymphomagenesis. Consistently, over the last decade, numerous epigenetic drugs (or epidrugs) have emerged in the clinical management of B-NHL patients. In this review, we will present an overview of the most relevant epidrugs tested and/or used so far for the treatment of different subtypes of B-NHL, from first-generation epigenetic therapies like histone acetyl transferases (HDACs) or DNA-methyl transferases (DNMTs) inhibitors to new agents showing selectivity for proteins that are mutated, translocated, and/or overexpressed in these diseases, including EZH2, BET, and PRMT. We will dissect the mechanisms of action of these epigenetic inhibitors, as well as the molecular processes underlying their lack of efficacy in refractory patients. This review will also provide a summary of the latest strategies being employed in preclinical and clinical settings, and will point out the most promising lines of investigation in the field.
Collapse
Affiliation(s)
- Marcelo L. Ribeiro
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista, São Paulo, Brazil
| | - Diana Reyes-Garau
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Marc Armengol
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Miranda Fernández-Serrano
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Gaël Roué
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Walewski J, Paszkiewicz-Kozik E, Borsaru G, Hellmann A, Janikova A, Warszewska A, Mais A, Ammendola A, Herz T, Krauss B, Henning SW. Resminostat in patients with relapsed or refractory Hodgkin lymphoma: results of the phase II SAPHIRE study. Leuk Lymphoma 2018; 60:675-684. [DOI: 10.1080/10428194.2018.1492122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Jan Walewski
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | - Ewa Paszkiewicz-Kozik
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | | | - Andrzej Hellmann
- Department for Hematology and Transplantology, University Clinical Centre, Medical University of Gdansk, Gdansk, Poland
| | - Andrea Janikova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Agnieszka Warszewska
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | - Anna Mais
- 4SC AG, Martinsried, Planegg, Germany
| | | | | | | | | |
Collapse
|
18
|
Hargarten JC, Williamson PR. Epigenetic Regulation of Autophagy: A Path to the Control of Autoimmunity. Front Immunol 2018; 9:1864. [PMID: 30154791 PMCID: PMC6102341 DOI: 10.3389/fimmu.2018.01864] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
Autoimmune diseases are a significant cause of debilitation and mortality globally and are in need of cost-effective therapeutics. Autophagy is a cellular pathway that facilitates immune modulation involved in both pathogen control and autoimmunity. Regulation is multifactorial and includes a number of epigenetic pathways which can involve modification of DNA-binding histones to induce autophagy-related mRNA synthesis or microRNA and decapping-associated mRNA degradation which results in autophagy suppression. Appreciation of epigenetic-based pathways involved in autophagy and autoimmunity may facilitate application of a burgeoning group of epigenetic pharmaceuticals to these important diseases.
Collapse
Affiliation(s)
- Jessica C Hargarten
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
19
|
Targeting autophagy in lymphomas: a double-edged sword? Int J Hematol 2018; 107:502-512. [DOI: 10.1007/s12185-018-2414-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 12/19/2022]
|
20
|
Lopez G, Braggio D, Zewdu A, Casadei L, Batte K, Bid HK, Koller D, Yu P, Iwenofu OH, Strohecker A, Choy E, Lev D, Pollock R. Mocetinostat combined with gemcitabine for the treatment of leiomyosarcoma: Preclinical correlates. PLoS One 2017; 12:e0188859. [PMID: 29186204 PMCID: PMC5706733 DOI: 10.1371/journal.pone.0188859] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023] Open
Abstract
Leiomyosarcoma (LMS) is a malignant soft tissue sarcoma (STS) with a dismal prognosis following metastatic disease. Chemotherapeutic intervention has demonstrated to have modest clinical efficacy with no curative potential in LMS patients. Previously, we demonstrated pan-HDAC inhibition to have a superior effect in various complex karyotypic sarcomas. In this study, our goal is to evaluate the therapeutic efficacy of mocetinostat alone and in combination with gemcitabine in LMS. Human leiomyosarcoma (LMS) cell lines were used for in vitro and in vivo studies. Compounds tested included the class I HDAC inhibitor, mocetinostat, and nucleoside analog, gemcitabine. MTS and clonogenic assays were used to evaluate the effect of mocetinostat on LMS cell growth. Cleaved caspase 3/7 analysis was used to determine the effects of mocetinostat on apoptosis. Compusyn software was used to determine in vitro synergy studies for the combination of mocetinostat plus gemcitabine. A LMS xenograft model in SCID mice was used to test the impact of mocetinostat alone, gemcitabine alone and the combination of mocetinostat plus gemcitabine. Mocetinostat abrogated LMS cell growth and clonogenic potential, and enhanced apoptosis in LMS cell lines. The combination of mocetinostat plus gemcitabine exhibited a synergistic effect in LMS cells in vitro. Similarly, mocetinostat combined with gemcitabine resulted in superior anti-LMS effects in vivo. Mocetinostat reduced the expression of gemcitabine-resistance markers RRM1, RRM2, and increased the expression of gemcitabine-sensitivity marker, hENT1, in LMS cells. LMS are aggressive, metastatic tumors with poor prognosis where effective therapeutic interventions are wanting. Our studies demonstrate the potential utility of mocetinostat combined with gemcitabine for the treatment of LMS.
Collapse
Affiliation(s)
- Gonzalo Lopez
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Danielle Braggio
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Abeba Zewdu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Lucia Casadei
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Kara Batte
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Hemant Kumar Bid
- Life Science Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - David Koller
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Peter Yu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Obiajulu Hans Iwenofu
- Department of Pathology, The Ohio State University, Columbus, OH, United States of America
| | - Anne Strohecker
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Edwin Choy
- Division of Hematology Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Dina Lev
- Surgery B, Sheba Medical Center, Tel Aviv, Israel
| | - Raphael Pollock
- Department of Surgery, The Ohio State University, Columbus, OH, United States of America
- * E-mail:
| |
Collapse
|
21
|
Cifaldi L, Locatelli F, Marasco E, Moretta L, Pistoia V. Boosting Natural Killer Cell-Based Immunotherapy with Anticancer Drugs: a Perspective. Trends Mol Med 2017; 23:1156-1175. [PMID: 29133133 DOI: 10.1016/j.molmed.2017.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 12/27/2022]
Abstract
Natural killer (NK) cells efficiently recognize and kill tumor cells through several mechanisms including the expression of ligands for NK cell-activating receptors on target cells. Different clinical trials indicate that NK cell-based immunotherapy represents a promising antitumor treatment. However, tumors develop immune-evasion strategies, including downregulation of ligands for NK cell-activating receptors, that can negatively affect antitumor activity of NK cells, which either reside endogenously, or are adoptively transferred. Thus, restoration of the expression of NK cell-activating ligands on tumor cells represents a strategic therapeutic goal. As discussed here, various anticancer drugs can fulfill this task via different mechanisms. We envision that the combination of selected chemotherapeutic agents with NK cell adoptive transfer may represent a novel strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Loredana Cifaldi
- Department of Pediatric Haematology/Oncology, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy.
| | - Franco Locatelli
- Department of Pediatric Haematology/Oncology, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy; Department of Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Emiliano Marasco
- Department of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Immunology Research Area, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Vito Pistoia
- Immunology Research Area, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
22
|
Combination of a novel HDAC6 inhibitor ACY-241 and anti-PD-L1 antibody enhances anti-tumor immunity and cytotoxicity in multiple myeloma. Leukemia 2017; 32:843-846. [PMID: 29104288 DOI: 10.1038/leu.2017.322] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
23
|
Jiang T, Wang F, Hu L, Cheng X, Zheng Y, Liu T, Jia Y. Chidamide and decitabine can synergistically induce apoptosis of Hodgkin lymphoma cells by up-regulating the expression of PU.1 and KLF4. Oncotarget 2017; 8:77586-77594. [PMID: 29100410 PMCID: PMC5652801 DOI: 10.18632/oncotarget.20659] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 07/25/2017] [Indexed: 02/05/2023] Open
Abstract
Epigenetic abnormalities play important roles in the pathogenesis of Hodgkin lymphoma (HL). Highly expressed class I histone acetyltransferase (HDAC) and hyper-methylation of the promoter region of tumor suppressor genes have been demonstrated in Hodgkin lymphoma. In this paper, we investigated the synergistic effects of combination treatment of chidamide, a selective HDAC inhibitor, and decitabine, a demethylation agent, for HL cell lines and explored a new strategy for treating HL. The apoptosis rates, cell cycle, mitochondrial transmembrane potentials, epigenetic changes and gene expression of HL cell lines treated with chidamide as a single agent and in combination with decitabine were tested. We found that chidamide inhibited the proliferation of HL cells through increased apoptosis. Interestingly, after combined with decitabine, the inhibition rate and apoptotic death in HL cells were significantly increased. Further studies demonstrated that when combined with decitabine the expression of acetylated histone H3 and H3K9 induced by chidamide in HL cells increased, and also the expression of tumor suppressor genes PU.1 and KLF4, which exert inhibition through apoptosis pathway. Therefore, we could come to a conclusion that chidamide and decitabine can synergistically induce apoptosis of Hodgkin lymphoma cells by up-regulating the expression of PU.1 and KLF4. These results provide a new sight in combining two different epigenetic regulatory agents for treating HL.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Department of Hematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan, China
| | - Fujue Wang
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Laboratory of Hematology, West China Hospital of Sichuan University, Tianfu Life Science Park, Chengdu 610041, Sichuan, China
| | - Lianjie Hu
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Laboratory of Hematology, West China Hospital of Sichuan University, Tianfu Life Science Park, Chengdu 610041, Sichuan, China
| | - Xiaomin Cheng
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Department of Hematology, Chengdu Military General Hospital, Chengdu 610083, Sichuan, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Laboratory of Hematology, West China Hospital of Sichuan University, Tianfu Life Science Park, Chengdu 610041, Sichuan, China
| | - Ting Liu
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Laboratory of Hematology, West China Hospital of Sichuan University, Tianfu Life Science Park, Chengdu 610041, Sichuan, China
| | - Yongqian Jia
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Laboratory of Hematology, West China Hospital of Sichuan University, Tianfu Life Science Park, Chengdu 610041, Sichuan, China
| |
Collapse
|
24
|
Desimio MG, Giuliani E, Doria M. The histone deacetylase inhibitor SAHA simultaneously reactivates HIV-1 from latency and up-regulates NKG2D ligands sensitizing for natural killer cell cytotoxicity. Virology 2017; 510:9-21. [PMID: 28689087 DOI: 10.1016/j.virol.2017.06.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/14/2017] [Accepted: 06/29/2017] [Indexed: 01/01/2023]
Abstract
In pilot HIV-1 eradication studies, patients' immune responses were ineffective at killing viral reservoirs reactivated through latency reversing agents (LRAs) like suberoylanilide hydroxamic acid (SAHA). We hypothesized that T cells harboring reactivated HIV-1 express MIC and ULBP ligands for the activating NKG2D receptor of natural killer (NK) cells. Here, we demonstrated that MICA/B and ULBP2 are induced by SAHA on primary T cells harboring reactivated virus. Using latently HIV-1-infected J-Lat 6.3/8.4/9.2 and J1.1 cell lines, we showed that SAHA reverts latency and, simultaneously, up-regulates MICA/B and ULBP2 acting at the transcriptional level and through ATR activation, thus sensitizing T cells with reactivated virus to NKG2D-mediated killing by NK cells. Moreover, IL-2 and IL-15 potently boosted NKG2D expression and cytotoxicity of NK cells against SAHA-reactivated p24+ target cells. Therefore, immunotherapy with cytokines enhancing NKG2D-mediated NK-cell cytotoxicity combined with administration of LRAs up-modulating NKG2D ligands, represents a promising approach towards HIV-1 eradication.
Collapse
Affiliation(s)
| | - Erica Giuliani
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Margherita Doria
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
25
|
Karantanos T, Politikos I, Boussiotis VA. Advances in the pathophysiology and treatment of relapsed/refractory Hodgkin's lymphoma with an emphasis on targeted therapies and transplantation strategies. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2017; 7:37-52. [PMID: 28701859 PMCID: PMC5502320 DOI: 10.2147/blctt.s105458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Hodgkin’s lymphoma (HL) is highly curable with first-line therapy. However, a minority of patients present with refractory disease or experience relapse after completion of frontline treatment. These patients are treated with salvage chemotherapy followed by autologous stem cell transplantation (ASCT), which remains the standard of care with curative potential for refractory or relapsed HL. Nevertheless, a significant percentage of such patients will progress after ASCT, and allogeneic hematopoietic stem cell transplantation remains the only curative approach in that setting. Recent advances in the pathophysiology of refractory or relapsed HL have provided the rationale for the development of novel targeted therapies with potent anti-HL activity and favorable toxicity profile, in contrast to cytotoxic chemotherapy. Brentuximab vedotin and programmed cell death-1-based immunotherapy have proven efficacy in the management of refractory or relapsed HL, whereas several other agents have shown promise in early clinical trials. Several of these agents are being incorporated with transplantation strategies in order to improve the outcomes of refractory or relapsed HL. In this review we summarize the current knowledge regarding the mechanisms responsible for the development of refractory/relapsed HL and the outcomes with current treatment strategies, with an emphasis on targeted therapies and hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Theodoros Karantanos
- General Internal Medicine Section, Boston Medical Center, Boston University School of Medicine, Boston, MA
| | - Ioannis Politikos
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
CBP/p300 acetyltransferases regulate the expression of NKG2D ligands on tumor cells. Oncogene 2016; 36:933-941. [PMID: 27477692 PMCID: PMC5318661 DOI: 10.1038/onc.2016.259] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/12/2016] [Accepted: 06/13/2016] [Indexed: 01/12/2023]
Abstract
Tumor surveillance of natural killer (NK) cells is mediated by the cytotoxicity receptor natural-killer group 2 member D (NKG2D). Ligands for NKG2D are generally not expressed on healthy cells, but induced on the surface of malignant cells. To date, NKG2D ligand (NKG2D-L) induction was mainly described to depend on the activation of the DNA damage response, although the molecular mechanisms that regulate NKG2D-L expression remain largely unknown. Here, we show that the acetyltransferases CBP (CREB-binding protein) and p300 play a crucial role in the regulation of NKG2D-L on tumor cells. Loss of CBP/p300 decreased the basal cell surface expression of human ligands and reduced the upregulation of MICA/B and ULBP2 in response to histone deacetylase inhibitors or DNA damage. Furthermore, CBP/P300 deficiency abrogated the sensitivity of stressed cells to NK cell-mediated killing. CBP/p300 were also identified as major regulators of mouse NKG2D ligand RAE-1 in vitro and in vivo using the Eμ-Myc lymphoma model. Mechanistically, we observed an enhanced activation of the CBP/p300 binding transcription factor CREB (cAMP response element-binding protein) correlating to the NKG2D-L upregulation. Moreover, increased binding of CREB and CBP/p300 to NKG2D-L promoters and elevated histone acetylation were detectable. This study provides strong evidence for a major role of CBP and p300 in orchestrating NKG2D-L induction and consequently immunosurveillance of tumors in mice and humans. These findings might help to develop novel immunotherapeutic approaches against cancer.
Collapse
|
27
|
Oki Y, Buglio D, Zhang J, Ying Y, Zhou S, Sureda A, Ben-Yehuda D, Zinzani PL, Prince HM, Harrison SJ, Kirschbaum M, Johnston PB, Shen A, von Tresckow B, Younes A. Immune regulatory effects of panobinostat in patients with Hodgkin lymphoma through modulation of serum cytokine levels and T-cell PD1 expression. Blood Cancer J 2014; 4:e236. [PMID: 25105535 PMCID: PMC4219471 DOI: 10.1038/bcj.2014.58] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Y Oki
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D Buglio
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Zhang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Y Ying
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S Zhou
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Sureda
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - D Ben-Yehuda
- Hematology Division, Hadassah Medical Organization, Jerusalem, Israel
| | - P L Zinzani
- Institute of Hematology and Medical Oncology, University of Bologna, Bologna, Italy
| | - H M Prince
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, Victoria, Australia
| | - S J Harrison
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, Victoria, Australia
| | - M Kirschbaum
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - P B Johnston
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - A Shen
- Novartis Pharmaceuticals, Florham Park, NJ, USA
| | | | - A Younes
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
28
|
Romano A, Vetro C, Caocci G, Greco M, Parrinello NL, Di Raimondo F, La Nasa G. Immunological deregulation in classic hodgkin lymphoma. Mediterr J Hematol Infect Dis 2014; 6:e2014039. [PMID: 24959336 PMCID: PMC4063611 DOI: 10.4084/mjhid.2014.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/06/2014] [Indexed: 01/19/2023] Open
Abstract
Classic Hodgkin Lymphoma (cHL) has a unique histology since only a few neoplastic cells are surrounded by inflammatory accessory cells that in the last years have emerged as crucial players in sustaining the course of disease. In addition, recent studies suggest that the abnormal activity of these inflammatory cells (such as deregulation in regulatory T cells signaling, expansion of myeloid derived suppressor cells, HLA-G signaling and natural killer cells dysfunction) may have prognostic significance. This review is focused on summarizing recent advanced in immunological defects in cHL with translational implications.
Collapse
Affiliation(s)
- Alessandra Romano
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Calogero Vetro
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Giovanni Caocci
- Hematology Unit, Department of Medical Sciences “Mario Aresu,” University of Cagliari, Italy
| | - Marianna Greco
- Hematology Unit, Department of Medical Sciences “Mario Aresu,” University of Cagliari, Italy
| | - Nunziatina Laura Parrinello
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Francesco Di Raimondo
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Giorgio La Nasa
- Hematology Unit, Department of Medical Sciences “Mario Aresu,” University of Cagliari, Italy
| |
Collapse
|