1
|
Makimoto A, Fujisaki H, Matsumoto K, Takahashi Y, Cho Y, Morikawa Y, Yuza Y, Tajiri T, Iehara T. Retinoid Therapy for Neuroblastoma: Historical Overview, Regulatory Challenges, and Prospects. Cancers (Basel) 2024; 16:544. [PMID: 38339295 PMCID: PMC10854948 DOI: 10.3390/cancers16030544] [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: 11/11/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Retinoids are vitamin A derivatives and include trans-retinoic acid, isotretinoin, tamibarotene, and bexarotene, all of which are currently available for clinical use. The clinical development of retinoid therapy for neuroblastoma has a history spanning more than four decades. The most promising agent is isotretinoin, which can contribute to improving event-free survival in patients with high-risk neuroblastoma by approximately 10% when administered over six months as maintenance therapy. Although isotretinoin is regarded as an essential component in the standard clinical management of high-risk neuroblastoma, its use for this purpose in the US and EU is off-label. To promote isotretinoin use in Japan as a treatment for neuroblastoma, our clinical research team is planning to launch an investigator-initiated, registration-directed clinical trial. The present review article discusses the basic science behind retinoid therapy, pre-clinical/clinical evidence on neuroblastoma, the concept of the proposed clinical trial, and prospects for this therapy.
Collapse
Affiliation(s)
- Atsushi Makimoto
- Department of Laboratory Medicine, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan
- Clinical Research Support Center, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
- Department of Hematology/Oncology, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
| | - Hiroyuki Fujisaki
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka 534-0021, Japan;
| | - Kimikazu Matsumoto
- Children’s Cancer Center, National Center for Child Health and Development, Tokyo 157-8535, Japan;
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan;
| | - Yuko Cho
- Department of Pediatrics, Hokkaido University Hospital, Sapporo 060-8648, Japan;
| | - Yoshihiko Morikawa
- Clinical Research Support Center, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
| | - Yuki Yuza
- Department of Hematology/Oncology, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Tomoko Iehara
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| |
Collapse
|
2
|
Bishayee K, Nazim UM, Kumar V, Kang J, Kim J, Huh SO, Sadra A. Reversing the HDAC-inhibitor mediated metabolic escape in MYCN-amplified neuroblastoma. Biomed Pharmacother 2022; 150:113032. [PMID: 35486977 DOI: 10.1016/j.biopha.2022.113032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/08/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022] Open
Abstract
In MYCN-amplified neuroblastoma (NB), we noticed that the single compound treatment with the HDAC inhibitor vorinostat led to a reprogramming of the glycolytic pathway in these cells. This reprogramming was upregulation of fatty acid oxidation (FAO) and oxidative phosphorylation (OXPHOS), allowing the cells to generate ATP, albeit at a reduced rate. This behavior was dependent on reduced levels of MYCN and a corresponding increase in the levels of PPARD transcription factors. By integrating metabolic and functional studies in NB cells and mouse xenografts, we demonstrate a compensatory upregulation of FAO/OXPHOS metabolism that promotes resistance to HDAC inhibitors. From the additional compounds that could reverse this metabolic reprogramming, the mTORC1 inhibitor sirolimus was selected. Besides both a block of glycolysis and OXPHOS, the HDAC/mTORC1 inhibitor combination produced significantly higher levels of reactive oxygen species (ROS) in the treated cells and in xenograft tumor samples, also a consequence of increased glycolytic block. The lead compounds were also tested for changes in the message levels of the glycolytic enzymes and their pathway activity, and HK2 and GPI glycolytic enzymes were most affected at their RNA message level. This combination was seen with no overall toxicity in treated mice in terms of weight loss or liver/kidney function.
Collapse
Affiliation(s)
- Kausik Bishayee
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, South Korea
| | - Uddin Md Nazim
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, South Korea
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, South Korea
| | - Jieun Kang
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, South Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, South Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, South Korea.
| | - Ali Sadra
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, South Korea.
| |
Collapse
|
3
|
Ciaccio R, De Rosa P, Aloisi S, Viggiano M, Cimadom L, Zadran SK, Perini G, Milazzo G. Targeting Oncogenic Transcriptional Networks in Neuroblastoma: From N-Myc to Epigenetic Drugs. Int J Mol Sci 2021; 22:12883. [PMID: 34884690 PMCID: PMC8657550 DOI: 10.3390/ijms222312883] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma (NB) is one of the most frequently occurring neurogenic extracranial solid cancers in childhood and infancy. Over the years, many pieces of evidence suggested that NB development is controlled by gene expression dysregulation. These unleashed programs that outline NB cancer cells make them highly dependent on specific tuning of gene expression, which can act co-operatively to define the differentiation state, cell identity, and specialized functions. The peculiar regulation is mainly caused by genetic and epigenetic alterations, resulting in the dependency on a small set of key master transcriptional regulators as the convergence point of multiple signalling pathways. In this review, we provide a comprehensive blueprint of transcriptional regulation bearing NB initiation and progression, unveiling the complexity of novel oncogenic and tumour suppressive regulatory networks of this pathology. Furthermore, we underline the significance of multi-target therapies against these hallmarks, showing how novel approaches, together with chemotherapy, surgery, or radiotherapy, can have substantial antineoplastic effects, disrupting a wide variety of tumorigenic pathways through combinations of different treatments.
Collapse
|
4
|
Clark RA, Newton M, Qiao J, Lee S, Chung DH. Reactivation of silenced α-N-catenin induces retinoic acid sensitivity in neuroblastoma cells. Surgery 2021; 170:1546-1553. [PMID: 34092372 DOI: 10.1016/j.surg.2021.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/08/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND High-risk neuroblastoma remains the most difficult pediatric solid tumor to treat and is associated with chemotherapy and radiation resistance that may be secondary to epigenetic modifications. We have previously found that α-N-catenin, a cell-adhesion protein encoded by the gene CTNNA2, plays a tumor suppressor role in neuroblastoma by inhibiting the NF-κB signaling pathway. A subset of neuroblastoma tumors that lack α-N-catenin are resistant to all-trans retinoic acid. However, the mechanism of CTNNA2 silencing in neuroblastoma remains unknown. Herein, we sought to determine the mechanism of α-N-catenin silencing in neuroblastoma. METHODS Two human neuroblastoma cell lines, SK-N-AS and BE(2)-C, were stably transfected with a plasmid expressing CTNNA2. Both cell lines were treated with the histone deacetylase inhibitor Trichostatin A alone and in combination with retinoic acid. Cell survival and colony formation were measured. Cellular differentiation and expression of cell survival signaling pathways were analyzed. Immunoblotting and reverse transcription quantitative polymerase chain reaction were used to examine protein and messenger RNA expression. RESULTS Retinoic acid treatment induced cellular differentiation and inhibited cellular proliferation in BE(2)-C cells but did not induce differentiation in SK-N-AS cells. Re-expression of α-N-catenin enhanced the sensitivity to retinoic acid-induced cell growth arrest and downregulated key cell survival pathways in both cell lines. Trichostatin A treatment induced CTNNA2 expression in SK-N-AS cells, and combination treatment with Trichostatin A induced retinoic acid sensitivity in retinoic acid-resistant cells. CONCLUSION Re-expression of α-N-catenin in retinoic acid-resistant cells induced sensitivity to retinoic acid treatment and is controlled epigenetically via histone deacetylase. α-N-catenin is a potential biomarker for retinoic acid sensitivity and combination treatment with Trichostatin A and retinoic acid may improve survival among children with high-risk, retinoic acid-resistant neuroblastoma.
Collapse
Affiliation(s)
- Rachael A Clark
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Micah Newton
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jingbo Qiao
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Sora Lee
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Dai H Chung
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
5
|
Cheung BB, Kleynhans A, Mittra R, Kim PY, Holien JK, Nagy Z, Ciampa OC, Seneviratne JA, Mayoh C, Raipuria M, Gadde S, Massudi H, Wong IPL, Tan O, Gong A, Suryano A, Diakiw SM, Liu B, Arndt GM, Liu T, Kumar N, Sangfelt O, Zhu S, Norris MD, Haber M, Carter DR, Parker MW, Marshall GM. A novel combination therapy targeting ubiquitin-specific protease 5 in MYCN-driven neuroblastoma. Oncogene 2021; 40:2367-2381. [PMID: 33658627 PMCID: PMC8016666 DOI: 10.1038/s41388-021-01712-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 01/31/2023]
Abstract
Histone deacetylase (HDAC) inhibitors are effective in MYCN-driven cancers, because of a unique need for HDAC recruitment by the MYCN oncogenic signal. However, HDAC inhibitors are much more effective in combination with other anti-cancer agents. To identify novel compounds which act synergistically with HDAC inhibitor, such as suberanoyl hydroxamic acid (SAHA), we performed a cell-based, high-throughput drug screen of 10,560 small molecule compounds from a drug-like diversity library and identified a small molecule compound (SE486-11) which synergistically enhanced the cytotoxic effects of SAHA. Effects of drug combinations on cell viability, proliferation, apoptosis and colony forming were assessed in a panel of neuroblastoma cell lines. Treatment with SAHA and SE486-11 increased MYCN ubiquitination and degradation, and markedly inhibited tumorigenesis in neuroblastoma xenografts, and, MYCN transgenic zebrafish and mice. The combination reduced ubiquitin-specific protease 5 (USP5) levels and increased unanchored polyubiquitin chains. Overexpression of USP5 rescued neuroblastoma cells from the cytopathic effects of the combination and reduced unanchored polyubiquitin, suggesting USP5 is a therapeutic target of the combination. SAHA and SE486-11 directly bound to USP5 and the drug combination exhibited a 100-fold higher binding to USP5 than individual drugs alone in microscale thermophoresis assays. MYCN bound to the USP5 promoter and induced USP5 gene expression suggesting that USP5 and MYCN expression created a forward positive feedback loop in neuroblastoma cells. Thus, USP5 acts as an oncogenic cofactor with MYCN in neuroblastoma and the novel combination of HDAC inhibitor with SE486-11 represents a novel therapeutic approach for the treatment of MYCN-driven neuroblastoma.
Collapse
Affiliation(s)
- Belamy B Cheung
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.
- School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia.
| | - Ane Kleynhans
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Rituparna Mittra
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Patrick Y Kim
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Jessica K Holien
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, VIC, Australia
| | - Zsuzsanna Nagy
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Olivia C Ciampa
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Janith A Seneviratne
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Mukesh Raipuria
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Satyanarayana Gadde
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Chemistry, UNSW Sydney, Sydney, NSW, Australia
| | - Hassina Massudi
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Iris Poh Ling Wong
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Owen Tan
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew Gong
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Aldwin Suryano
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Sonya M Diakiw
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Bing Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Greg M Arndt
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Tao Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Naresh Kumar
- School of Chemistry, UNSW Sydney, Sydney, NSW, Australia
| | - Olle Sangfelt
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Cancer Center and Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Murray D Norris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- University of New South Wales Centre for Childhood Cancer Research, Sydney, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel R Carter
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Michael W Parker
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, VIC, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Glenn M Marshall
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia.
| |
Collapse
|
6
|
Abstract
Neuroblastoma (NB) is a pediatric cancer of the sympathetic nervous system and one of the most common solid tumors in infancy. Amplification of MYCN, copy number alterations, numerical and segmental chromosomal aberrations, mutations, and rearrangements on a handful of genes, such as ALK, ATRX, TP53, RAS/MAPK pathway genes, and TERT, are attributed as underlying causes that give rise to NB. However, the heterogeneous nature of the disease-along with the relative paucity of recurrent somatic mutations-reinforces the need to understand the interplay of genetic factors and epigenetic alterations in the context of NB. Epigenetic mechanisms tightly control gene expression, embryogenesis, imprinting, chromosomal stability, and tumorigenesis, thereby playing a pivotal role in physio- and pathological settings. The main epigenetic alterations include aberrant DNA methylation, disrupted patterns of posttranslational histone modifications, alterations in chromatin composition and/or architecture, and aberrant expression of non-coding RNAs. DNA methylation and demethylation are mediated by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins, respectively, while histone modifications are coordinated by histone acetyltransferases and deacetylases (HATs, HDACs), and histone methyltransferases and demethylases (HMTs, HDMs). This article focuses predominately on the crosstalk between the epigenome and NB, and the implications it has on disease diagnosis and treatment.
Collapse
Affiliation(s)
- Irfete S Fetahu
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| |
Collapse
|
7
|
Vorinostat-loaded titanium oxide nanoparticles (anatase) induce G2/M cell cycle arrest in breast cancer cells via PALB2 upregulation. 3 Biotech 2020; 10:407. [PMID: 32904337 DOI: 10.1007/s13205-020-02391-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is a group of diseases in which cells divide out of controlled, typically resulting in a mass. Erlotinib is targeted cancer drug which functions as an inhibitor of the epidermal growth factor receptor (EGFR) tyrosine kinase. It is used mainly to treat of non-small cell lung cancer patients and has an action against pancreatic cancer. Vorinostat (aka suberanilohydroxamic acid) is an inhibitor of histone deacetylases (HDAC), which has an epigenetic modulation activity. It is used to treat cutaneous T cell lymphoma. In the present study, the erlotinib (ERL) and vorinostat (SAHA) loaded TiO2 nanoparticles (NPs) were used for the treatment of the breast cancer cells (MDA-MB-231 and MCF-7) and human cancerous amniotic cells (WISH). Cell count and viability were negatively affected in all treatments compared to normal cells and bare TiO2 NPs. Apoptosis results indicated a significant increase in the total apoptosis in all treatments compared with control cells. ERL- and SAHA-loaded TiO2 NPs treatments arrested breast cancer cells at G2/M phase, which indicate the cytotoxic effect of these treatment. Partner and localizer of BRCA2 (PALB2) gene expression was assessed using qPCR. The results indicate that PLAB2 was upregulated in ERL- and SAHA-loaded TiO2 NPs compared with control cells and can be used as nanocarrier for chemotherapy drugs. However, this conclusion necessitates further confirmative investigation.
Collapse
|
8
|
Makowiecka A, Malek N, Mazurkiewicz E, Mrówczyńska E, Nowak D, Mazur AJ. Thymosin β4 Regulates Focal Adhesion Formation in Human Melanoma Cells and Affects Their Migration and Invasion. Front Cell Dev Biol 2019; 7:304. [PMID: 31921836 PMCID: PMC6935720 DOI: 10.3389/fcell.2019.00304] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
Thymosin β4 (Tβ4), a multifunctional 44-amino acid polypeptide and a member of actin-binding proteins (ABPs), plays an important role in developmental processes and wound healing. In recent years an increasing number of data has been published suggesting Tβ4's involvement in tumorigenesis. However, Tβ4's role in melanoma tumor development still remains to be elucidated. In our study we demonstrate that Tβ4 is crucial for melanoma adhesion and invasion. For the purpose of our research we tested melanoma cell lines differing in invasive potential. Moreover, we applied shRNAs to silence TMSB4X (gene encoding Tβ4) expression in a cell line with high TMSB4X expression. We found out that Tβ4 is not only a component of focal adhesions (FAs) and interacts with several FAs components but also regulates FAs formation. We demonstrate that Tβ4 level has an impact on FAs' number and morphology. Moreover, manipulation with TMSB4X expression resulted in changes in cells' motility on non-coated and MatrigelTM (resembling basement membrane composition)-coated surfaces and drastically decreased invasion abilities of the cells. Additionally, a correlation between Tβ4 expression level and exhibition of mesenchymal-like [epithelial-mesenchymal transition (EMT)] features was discovered. Cells with lowered TMSB4X expression were less EMT-progressed than control cells. Summarizing, obtained results show that Tβ4 by regulating melanoma cells' adhesion has an impact on motility features and EMT. Our study not only contributes to a better understanding of the processes underlying melanoma cells' capacity to create metastases but also highlights Tβ4 as a potential target for melanoma management therapy.
Collapse
Affiliation(s)
- Aleksandra Makowiecka
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Natalia Malek
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Ewa Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Ewa Mrówczyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| |
Collapse
|
9
|
Makena MR, Cho HE, Nguyen TH, Koneru B, Verlekar DU, Hindle A, Kang MH, Reynolds CP. Cytotoxic activity of difluoromethylornithine compared with fenretinide in neuroblastoma cell lines. Pediatr Blood Cancer 2018; 65:e27447. [PMID: 30251395 PMCID: PMC9621602 DOI: 10.1002/pbc.27447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/31/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Maintenance therapy with 13-cis-retinoic acid and immunotherapy (given after completion of intensive cytotoxic therapy) improves outcome for high-risk neuroblastoma patients. The synthetic retinoid fenretinide (4-HPR) achieved multiple complete responses in relapse/refractory neuroblastoma in early-phase clinical trials, has low systemic toxicity, and has been considered for maintenance therapy clinical trials. Difluoromethylornithine (DFMO, an irreversible inhibitor of ornithine decarboxylase with minimal single-agent clinical response data) is being used for maintenance therapy of neuroblastoma. We evaluated the cytotoxic activity of DFMO and fenretinide in neuroblastoma cell lines. PROCEDURE We tested 16 neuroblastoma cell lines in bone marrow-level hypoxia (5% O2 ) using the DIMSCAN cytotoxicity assay. Polyamines were measured by HPLC-mass spectrometry and apoptosis by transferase dUTP nick end labeling (TUNEL) using flow cytometry. RESULTS At clinically achievable levels (100 μM), DFMO significantly decreased (P < 0.05) polyamine putrescine and achieved modest cytotoxicity (<1 log (90% cytotoxicity). Prolonged exposures (7 days) or culture in 2% and 20% O2 did not enhance DFMO cytotoxicity. However, fenretinide (10 μM) even at a concentration lower than clinically achievable in neuroblastoma patients (20 μM) induced ≥ 1 log cell kill in 14 cell lines. The average IC90 and IC99 of fenretinide was 4.7 ± 1 μM and 9.9 ± 1.8 μM, respectively. DFMO did not induce a significant increase (P > 0.05) in apoptosis (TUNEL assay). Apoptosis by fenretinide was significantly higher (P < 0.001) compared with DFMO or controls. CONCLUSIONS DFMO as a single agent has minimal cytotoxic activity for neuroblastoma cell lines.
Collapse
Affiliation(s)
- Monish R. Makena
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Departments of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| | - Hwang Eui Cho
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Departments of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| | - Thinh H. Nguyen
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Pharmacology and Neuroscience, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| | - Balakrishna Koneru
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Departments of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| | - Dattesh U. Verlekar
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Departments of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| | - Ashly Hindle
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Departments of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| | - Min H. Kang
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Departments of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Pharmacology and Neuroscience, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| | - C. Patrick Reynolds
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Departments of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Pharmacology and Neuroscience, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Pediatrics, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX.,Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX
| |
Collapse
|
10
|
Jubierre L, Jiménez C, Rovira E, Soriano A, Sábado C, Gros L, Llort A, Hladun R, Roma J, Toledo JSD, Gallego S, Segura MF. Targeting of epigenetic regulators in neuroblastoma. Exp Mol Med 2018; 50:1-12. [PMID: 29700278 PMCID: PMC5938021 DOI: 10.1038/s12276-018-0077-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 01/13/2018] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
Approximately 15,000 new cases of pediatric cancer are diagnosed yearly in Europe, with 8–10% corresponding to neuroblastoma, a rare disease with an incidence of 8–9 cases per million children <15 years of age. Although the survival rate for low-risk and intermediate-risk patients is excellent, half of children with high-risk, refractory, or relapsed tumors will be cured, and two-thirds of the other half will suffer major side effects and life-long disabilities. Epigenetic therapies aimed at reversing the oncogenic alterations in chromatin structure and function are an emerging alternative against aggressive tumors that are or will become resistant to conventional treatments. This approach proposes targeting epigenetic regulators, which are proteins that are involved in the creation, detection, and interpretation of epigenetic signals, such as methylation or histone post-translational modifications. In this review, we focused on the most promising epigenetic regulators for targeting and current drugs that have already reached clinical trials. Treatments that target chromatin, the combination of DNA and proteins, are emerging as alternative ways to treat aggressive neuroblastomas, cancers of neural tissue. Altering the structure and function of chromatin is a form of “epigenetic therapy”, treatment that affects inheritable molecular signals controlling the activity of genes, rather than targeting the genes directly. Researchers in Spain led by Miguel Segura at the Vall d’Hebron Research Institute in Barcelona review progress in developing epigenetic therapies for neuroblastomas. A growing body of fundamental research and evidence from clinical trials suggest this approach could open promising new avenues to treating aggressive and drug-resistant cancers. The authors recommend an increased effort to identify and explore the activities of small molecules that could form the basis of effective epigenetic therapies for various cancers.
Collapse
Affiliation(s)
- Luz Jubierre
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Carlos Jiménez
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Eric Rovira
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Aroa Soriano
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Constantino Sábado
- Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Luis Gros
- Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Anna Llort
- Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Raquel Hladun
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.,Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Josep Roma
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Josep Sánchez de Toledo
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.,Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Soledad Gallego
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.,Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Miguel F Segura
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.
| |
Collapse
|
11
|
Chlapek P, Slavikova V, Mazanek P, Sterba J, Veselska R. Why Differentiation Therapy Sometimes Fails: Molecular Mechanisms of Resistance to Retinoids. Int J Mol Sci 2018; 19:ijms19010132. [PMID: 29301374 PMCID: PMC5796081 DOI: 10.3390/ijms19010132] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022] Open
Abstract
Retinoids represent a popular group of differentiation inducers that are successfully used in oncology for treatment of acute promyelocytic leukemia in adults and of neuroblastoma in children. The therapeutic potential of retinoids is based on their key role in the regulation of cell differentiation, growth, and apoptosis, which provides a basis for their use both in cancer therapy and chemoprevention. Nevertheless, patients treated with retinoids often exhibit or develop resistance to this therapy. Although resistance to retinoids is commonly categorized as either acquired or intrinsic, resistance as a single phenotypic feature is usually based on the same mechanisms that are closely related or combined in both of these types. In this review, we summarize the most common changes in retinoid metabolism and action that may affect the sensitivity of a tumor cell to treatment with retinoids. The availability of retinoids can be regulated by alterations in retinol metabolism or in retinoid intracellular transport, by degradation of retinoids or by their efflux from the cell. Retinoid effects on gene expression can be regulated via retinoid receptors or via other molecules in the transcriptional complex. Finally, the role of small-molecular-weight inhibitors of altered cell signaling pathways in overcoming the resistance to retinoids is also suggested.
Collapse
Affiliation(s)
- Petr Chlapek
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
| | - Viera Slavikova
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
| | - Pavel Mazanek
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Jaroslav Sterba
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| |
Collapse
|
12
|
Functional imaging in combination with mutation status aids prediction of response to inhibiting B-cell receptor signaling in lymphoma. Oncotarget 2017; 8:78917-78929. [PMID: 29108275 PMCID: PMC5668008 DOI: 10.18632/oncotarget.20551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022] Open
Abstract
Aberrant B-cell receptor (BCR) signaling is known to contribute to malignant transformation. Two small molecule inhibitors targeting BCR pathway signaling include ibrutinib, a Bruton’s tyrosine kinase (BTK) inhibitor, and idelalisib, a specific Phosphatidylinositol-4,5-bisphosphate 3-kinase delta (PI3Kδ) inhibitor, both of which have been approved for use in haematological malignancies. Despite the identification of various diffuse large B-cell lymphoma (DLBCL) subtypes, mutation status alone is not sufficient to predict patient response and therapeutic resistance can arise. Herein we apply early molecular imaging across alternative activated B-cell (ABC) and germinal center B-cell (GCB) DLBCL subtypes to investigate the effects of BCR pathway inhibition. Treatment with both inhibitors adversely affected cell growth and viability. These effects were partially predictable based upon mutation status. Accordingly, very early 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography (18F-FDG-PET) and 3’-deoxy-3’[18F]-fluorothymidine positron emission tomography (18F-FLT-PET) reported tumour regression and reductions in tumour metabolism and proliferation upon treatment. Furthermore, matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI IMS) identified alterations in the proteome of a model of ABC DLBCL upon treatment with ibrutinib or idelalisib. In conclusion we demonstrate that very early molecular imaging adds predictive value in addition to mutational status of DLBCL that may be useful in directing patient therapy.
Collapse
|
13
|
Vittorio O, Cojoc M, Curcio M, Spizzirri UG, Hampel S, Nicoletta FP, Iemma F, Dubrovska A, Kavallaris M, Cirillo G. Polyphenol Conjugates by Immobilized Laccase: The Green Synthesis of Dextran-Catechin. MACROMOL CHEM PHYS 2016. [DOI: 10.1002/macp.201600046] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Orazio Vittorio
- Children's Cancer Institute Australia; Randwick 2031 NSW Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for NanoMedicine; UNSW Australia; Sydney 2052 NSW Australia
| | - Monica Cojoc
- OncoRay-National Center for Radiation Research in Oncology; Medical Faculty and University Hospital Carl Gustav Carus; Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf; Fetscherstr. 74 01307 Dresden Germany
| | - Manuela Curcio
- Department of Pharmacy, Health and Nutritional Science; University of Calabria; Rende 87036 Italy
| | | | - Silke Hampel
- Leibniz Institute of Solid State and Material Research Dresden; 01171 Dresden Germany
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy, Health and Nutritional Science; University of Calabria; Rende 87036 Italy
| | - Francesca Iemma
- Department of Pharmacy, Health and Nutritional Science; University of Calabria; Rende 87036 Italy
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology; Medical Faculty and University Hospital Carl Gustav Carus; Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf; Fetscherstr. 74 01307 Dresden Germany
- German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ); 69120 Heidelberg Germany
| | - Maria Kavallaris
- Children's Cancer Institute Australia; Randwick 2031 NSW Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for NanoMedicine; UNSW Australia; Sydney 2052 NSW Australia
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Science; University of Calabria; Rende 87036 Italy
| |
Collapse
|
14
|
Cheung BB. Combination therapies improve the anticancer activities of retinoids in neuroblastoma. World J Clin Oncol 2015; 6:212-215. [PMID: 26677433 PMCID: PMC4675905 DOI: 10.5306/wjco.v6.i6.212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/25/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023] Open
Abstract
Most therapeutic protocols for child cancers use cytotoxic agents which have a narrow therapeutic index, and resulting in severe acute and chronic toxicities to normal tissues. Despite the fact that most child cancer patients achieve complete remission after chemotherapy, death still occurs due to relapse of persistent minimal residual disease (MRD) which remaining after initial cytotoxic chemotherapy. Advanced neuroblastoma (NB) is a leading cause of cancer deaths in young children. Retinoids are an important component of advanced NB therapy at the stage of MRD, yet half of all patients treated with 13-cis-retinoic acid still relapse and die. More effective combination therapies, with a lower side-effect profile, are required to improve outcomes for NB. Fenretinide or N-4-hydroxyphenyl retinamide is a synthetic derivative of retinoic acid which works on cancer cells through nuclear receptor-dependent and -independent signalling mechanisms. Moreover, several histone deacetylase inhibitors have entered early phase trials, and, suberoylanilide hydroxamic acid has been approved for use in adult cutaneous T cell lymphoma. A number of studies suggest that retinoid signal activation is necessary for histone deacetylase inhibitor activity. A better understanding of their mechanism of actions will lead to more evidence-based retinoid combination therapies.
Collapse
|
15
|
Cheung BB, Tan O, Koach J, Liu B, Shum MSY, Carter DR, Sutton S, Po'uha ST, Chesler L, Haber M, Norris MD, Kavallaris M, Liu T, O'Neill GM, Marshall GM. Thymosin-β4 is a determinant of drug sensitivity for Fenretinide and Vorinostat combination therapy in neuroblastoma. Mol Oncol 2015; 9:1484-500. [PMID: 25963741 PMCID: PMC5528804 DOI: 10.1016/j.molonc.2015.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 10/23/2022] Open
Abstract
Retinoids are an important component of neuroblastoma therapy at the stage of minimal residual disease, yet 40-50% of patients treated with 13-cis-retinoic acid (13-cis-RA) still relapse, indicating the need for more effective retinoid therapy. Vorinostat, or Suberoylanilide hydroxamic acid (SAHA), is a potent inhibitor of histone deacetylase (HDAC) classes I & II and has antitumor activity in vitro and in vivo. Fenretinide (4-HPR) is a synthetic retinoid which acts on cancer cells through both nuclear retinoid receptor and non-receptor mechanisms. In this study, we found that the combination of 4-HPR + SAHA exhibited potent cytotoxic effects on neuroblastoma cells, much more effective than 13-cis-RA + SAHA. The 4-HPR + SAHA combination induced caspase-dependent apoptosis through activation of caspase 3, reduced colony formation and cell migration in vitro, and tumorigenicity in vivo. The 4-HPR and SAHA combination significantly increased mRNA expression of thymosin-beta-4 (Tβ4) and decreased mRNA expression of retinoic acid receptor α (RARα). Importantly, the up-regulation of Tβ4 and down-regulation of RARα were both necessary for the 4-HPR + SAHA cytotoxic effect on neuroblastoma cells. Moreover, Tβ4 knockdown in neuroblastoma cells increased cell migration and blocked the effect of 4-HPR + SAHA on cell migration and focal adhesion formation. In primary human neuroblastoma tumor tissues, low expression of Tβ4 was associated with metastatic disease and predicted poor patient prognosis. Our findings demonstrate that Tβ4 is a novel therapeutic target in neuroblastoma, and that 4-HPR + SAHA is a potential therapy for the disease.
Collapse
Affiliation(s)
- Belamy B Cheung
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia.
| | - Owen Tan
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Jessica Koach
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Bing Liu
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Michael S Y Shum
- Kids Research Institute, Children's Hospital at Westmead, Sydney, Australia
| | - Daniel R Carter
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Selina Sutton
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Sela T Po'uha
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Louis Chesler
- Division of Clinical Studies, Institute of Cancer Research, Sutton, Surrey, UK
| | - Michelle Haber
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Maria Kavallaris
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Tao Liu
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia
| | - Geraldine M O'Neill
- Kids Research Institute, Children's Hospital at Westmead, Sydney, Australia; Discipline of Paediatrics and Child Health, University of Sydney, Australia
| | - Glenn M Marshall
- Children's Cancer Institute Australia, University of New South Wales, Sydney, Australia; Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia.
| |
Collapse
|