Editorial Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Dec 10, 2015; 6(6): 212-215
Published online Dec 10, 2015. doi: 10.5306/wjco.v6.i6.212
Combination therapies improve the anticancer activities of retinoids in neuroblastoma
Belamy B Cheung, Molecular Carcinogenesis Program, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney NSW 2031, Australia
Author contributions: Cheung BB solely contributed to this manuscript.
Conflict-of-interest statement: The author declares no conflicts of interest regarding this manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Belamy B Cheung, PhD, Molecular Carcinogenesis Program, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, PO Box 81, Randwick, Sydney NSW 2031, Australia. bcheung@ccia.unsw.edu.au
Telephone: +61-2-93852450 Fax: +61-2-96626584
Received: May 19, 2015
Peer-review started: May 20, 2015
First decision: August 4, 2015
Revised: September 10, 2015
Accepted: October 12, 2015
Article in press: October 13, 2015
Published online: December 10, 2015
Processing time: 204 Days and 6.7 Hours

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.

Key Words: Retinoids, Histone deacetylase inhibitors, Combination therapies, Neuroblastoma, Fenretinide

Core tip: Neuroblastoma (NB) begins in embryonal neural crest cells, which later give rise to the sympathetic nervous system, and is caused in part by factors which arrest differentiation. In vitro, retinoids force susceptible cancer cells down a pathway of terminal differentiation and, have been part of the routine treatment of advanced NB for number of decades. Synergistic anti-tumour activity between histone deacetylase inhibitors and retinoids has been observed in a variety of preclinical models. This editorial note discusses some of these findings on the combination therapies for improving the anticancer activities of retinoids in NB.



INTRODUCTION

Neuroblastoma (NB) is a tumor of the sympathetic nervous system and the most common extracranial solid tumour in childhood[1]. NB accounts for more than 7% of malignancies in patients younger than 15 years and around 15% of all paediatric oncology deaths[2]. Some infants experience spontaneous regression, whereas older patients have maturation of their tumor into benign ganglioneuromas. However, the outcome for children with a high-risk clinical phenotype has improved only modestly, with long-term survival still less than 40%[3]. The introduction of 13-cis-retinoic acid (13-cis-RA) in the therapy of NB has improved the prognosis of this disease. Currently, the standard treatment for high risk of NB consists of myeloablative therapy followed by autologous hematopoietic stem cell transplantation and maintenance with 13-cis-RA for the treatment of minimal residual disease (MRD), leading to a 3-year disease-free survival rate of about 50%[4]. 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.

Retinoid therapy in paediatric cancer

Retinoids are vital for the growth and differentiation of a variety of normal adult and embryonic tissues, and have potent antiproliferative effects on many malignant cell types[5]. Retinoids mediate their widespread effects on cells by regulating the transcription of target genes through a complex system of ligand-inducible nuclear transcription factors: The retinoic acid receptors and retinoid X receptors[6]. RA exists in several stereoisomeric forms: Predominantly all-trans retinoic acid (ATRA) and 13-cis-RA, but also as less-stable isomers such as 9-cis retinoic acid. In the last few decades they have been widely studied in cancer prevention and therapy because of their ability to induce differentiation of tumor cells[7]. Retinoids are successfully used for the treatment of one pediatric cancer: Acute promyelocytic leukemia. ATRA converts the PML-RAR-α fusion protein into activator of transcription and restores cell differentiation[8]. Retinoids have also been widely investigated in solid tumors, especially in NB. In a long-term study for children with high-risk NB treated on a randomized trial of myeloablative therapy followed by 13-cis-RA, which given after intensive therapy resulted in significant improvement in 5-year overall survival rates, regardless of the type of consolidation[9]. However, as many high-risk patients still ultimately die due to relapse of persistent MRD after initial cytotoxic chemotherapy, novel therapies effective against MDR NB are needed.

Fenretinide is an effective retinoid therapy

Retinoids are vitamin A analogues required for normal morphogenesis and maintenance of diverse embryologic and adult tissues, which act on cells by binding nuclear receptors[10]. Fenretinide or N-4-hydroxyphenyl retinamide (4-HPR) is a synthetic derivative of retinoic acid which works on cancer cells through nuclear receptor-dependent and -independent signalling mechanisms[11]. 4-HPR has a broad spectrum of cytotoxic activity against primary tumor cells, cell lines, and/or xenografts of various cancers, including NB [11-13] and has been tested in early phase clinical trials in recurrent NB [14,15]. 4-HPR was anti-angiogenic in multiple tumour types and cytopathic in some cancer cells which were resistant to other retinoids or chemotherapeutics[13]. Clinical trials have revealed that 4-HPR is a highly active therapeutic and chemo-preventive agent with minimal side-effects in NB[14]. A phase I/II trial of oral 4-HPR in children with high-risk, relapsed solid tumours demonstrated minimal 4-HPR toxicity, but only stable disease as the best clinical response[16].

Combination therapy improves the anticancer activity of histone deacetylase inhibitors and retinoids

Increased histone deacetylase activity is a common causal factor in human cancer that causes transcriptional silencing of tumour suppressor genes[17]. Histone deacetylase inhibitors prevent deacetylases removing acetyl groups from histone tails, thereby promoting gene transcription[18]. Several histone deacetylase inhibitors have entered early phase trials, and, suberoylanilide hydroxamic acid (SAHA) has been approved for use in adult cutaneous T cell lymphoma[19]. The histone deacetylase inhibitor side-effect profile is low when compared with cytotoxic chemotherapy[20]. Moreover, two unbiased preclinical screens identified retinoid signal activation as the most effective method of augmenting the histone deacetylase inhibitor anti-cancer signal[21,22]. Retinoic acid receptor α (RARα), and, preferentially expressed antigen of melanoma, both repressor proteins for the retinoid signal, was shown to mediate resistance to histone deacetylase inhibitors[21]. Furthermore, RARα-deficient cells showed enhanced sensitivity to histone deacetylase inhibitors in vitro and in vivo[22]. These studies suggest that retinoid signal activation is necessary for histone deacetylase inhibitor activity. Hahn et al[23] used an HDAC inhibitor (valproicacid) as an enhancer to screen a small-molecule library for compounds inducing NB maturation, the top hit identified in the screen was all-trans-retinoic acid. These studies demonstrated that investigation of HDAC inhibitors and retinoids in combination are warranted to improve the anticancer activities in cancer.

Combination therapies improve the anticancer activities of retinoids in NB

Synergistic anti-tumour activity between histone deacetylase inhibitors and retinoids has been observed in a variety of preclinical models[24,25]. A study suggested that the HDAC inhibitor LAQ824 has a greater antitumor activity in combination with 13-cis-retinoic acid in melanoma tumors[24]. Another study showed that the intracranial tumors in ND2:SmoA1 mice treated with retinoid acid + SAHA + cisplatin showed a 4-fold increase in apoptosis over controls, and a 2-fold increase over animals receiving only SAHA or retinoid acid + SAHA[25]. We and others have shown that retinoids combined with histone deacetylase inhibitors are synergistic[26,27]. However, SAHA combined with 13-cis-retinoic acid, was well-tolerated in a phase I/II paediatric trial, but the best response for relapsed solid tumour patients was stable disease[28]. Recently, our study showed that 4-HPR+SAHA as a more effective therapy for NB than 13-cis-RA alone or with SAHA[29]. 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 RARα. Importantly, the up-regulation of Tβ4 and down-regulation of RARα were both necessary for the 4-HPR + SAHA cytotoxic effect on NB cells. Moreover, Tβ4 knockdown in NB cells increased cell migration and blocked the effect of 4-HPR + SAHA on cell migration and focal adhesion formation[29]. This study demonstrates that Tβ4 is a novel therapeutic target in NB, and that 4-HPR and SAHA is a potential combination therapy for the disease.

CONCLUSION

A therapeutic role for retinoids and HDAC inhibitors in several human cancer types, including NB, is well established. However, retinoids and HDAC inhibitors are not completely effective anti-cancer agents when used alone; thus, a better understanding of their mechanism of actions will lead to more evidence-based retinoid combination therapies. Because differentiation is aberrant in NB, compounds that modulate transcription and induce differentiation, such as HDAC inhibitors and retinoids, are of particular interest. Further studies to understand the mechanism of drug actions and the clinical trials with large cohort of patients to determine the efficacy of HDAC inhibitors and retinoids for patients with high-risk NB are warranted.

Footnotes

P- Reviewer: Hohenegger M, Munoz M

S- Editor: Ji FF L- Editor: A E- Editor: Li D

References
1.  Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003;3:203-216.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Maris JM, Hogarty MD, Bagatell R, Cohn SL. Neuroblastoma. Lancet. 2007;369:2106-2120.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Maris JM. Recent advances in neuroblastoma. N Engl J Med. 2010;362:2202-2211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1434]  [Cited by in F6Publishing: 1372]  [Article Influence: 98.0]  [Reference Citation Analysis (0)]
4.  Masetti R, Biagi C, Zama D, Vendemini F, Martoni A, Morello W, Gasperini P, Pession A. Retinoids in pediatric onco-hematology: the model of acute promyelocytic leukemia and neuroblastoma. Adv Ther. 2012;29:747-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
5.  Smith MA, Adamson PC, Balis FM, Feusner J, Aronson L, Murphy RF, Horowitz ME, Reaman G, Hammond GD, Fenton RM. Phase I and pharmacokinetic evaluation of all-trans-retinoic acid in pediatric patients with cancer. J Clin Oncol. 1992;10:1666-1673.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Lotan R. Retinoids in cancer chemoprevention. FASEB J. 1996;10:1031-1039.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Sun SY, Lotan R. Retinoids and their receptors in cancer development and chemoprevention. Crit Rev Oncol Hematol. 2002;41:41-55.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Fang J, Chen SJ, Tong JH, Wang ZG, Chen GQ, Chen Z. Treatment of acute promyelocytic leukemia with ATRA and As2O3: a model of molecular target-based cancer therapy. Cancer Biol Ther. 2002;1:614-620.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Matthay KK, Reynolds CP, Seeger RC, Shimada H, Adkins ES, Haas-Kogan D, Gerbing RB, London WB, Villablanca JG. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children’s oncology group study. J Clin Oncol. 2009;27:1007-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 645]  [Cited by in F6Publishing: 629]  [Article Influence: 41.9]  [Reference Citation Analysis (0)]
10.  Germain P, Iyer J, Zechel C, Gronemeyer H. Co-regulator recruitment and the mechanism of retinoic acid receptor synergy. Nature. 2002;415:187-192.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Sogno I, Venè R, Sapienza C, Ferrari N, Tosetti F, Albini A. Anti-angiogenic properties of chemopreventive drugs: fenretinide as a prototype. Recent Results Cancer Res. 2009;181:71-76.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Maurer BJ, Metelitsa LS, Seeger RC, Cabot MC, Reynolds CP. Increase of ceramide and induction of mixed apoptosis/necrosis by N-(4-hydroxyphenyl)- retinamide in neuroblastoma cell lines. J Natl Cancer Inst. 1999;91:1138-1146.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Wu JM, DiPietrantonio AM, Hsieh TC. Mechanism of fenretinide (4-HPR)-induced cell death. Apoptosis. 2001;6:377-388.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Formelli F, Cavadini E, Luksch R, Garaventa A, Villani MG, Appierto V, Persiani S. Pharmacokinetics of oral fenretinide in neuroblastoma patients: indications for optimal dose and dosing schedule also with respect to the active metabolite 4-oxo-fenretinide. Cancer Chemother Pharmacol. 2008;62:655-665.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Villablanca JG, London WB, Naranjo A, McGrady P, Ames MM, Reid JM, McGovern RM, Buhrow SA, Jackson H, Stranzinger E. Phase II study of oral capsular 4-hydroxyphenylretinamide (4-HPR/fenretinide) in pediatric patients with refractory or recurrent neuroblastoma: a report from the Children’s Oncology Group. Clin Cancer Res. 2011;17:6858-6866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
16.  Villablanca JG, Krailo MD, Ames MM, Reid JM, Reaman GH, Reynolds CP. Phase I trial of oral fenretinide in children with high-risk solid tumors: a report from the Children’s Oncology Group (CCG 09709). J Clin Oncol. 2006;24:3423-3430.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Steele N, Finn P, Brown R, Plumb JA. Combined inhibition of DNA methylation and histone acetylation enhances gene re-expression and drug sensitivity in vivo. Br J Cancer. 2009;100:758-763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 111]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
18.  Johnstone RW. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov. 2002;1:287-299.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, Chiao JH, Reilly JF, Ricker JL, Richon VM. Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood. 2007;109:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Hainsworth JD, Infante JR, Spigel DR, Arrowsmith ER, Boccia RV, Burris HA. A phase II trial of panobinostat, a histone deacetylase inhibitor, in the treatment of patients with refractory metastatic renal cell carcinoma. Cancer Invest. 2011;29:451-455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 77]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
21.  Epping MT, Wang L, Plumb JA, Lieb M, Gronemeyer H, Brown R, Bernards R. A functional genetic screen identifies retinoic acid signaling as a target of histone deacetylase inhibitors. Proc Natl Acad Sci USA. 2007;104:17777-17782.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Epping MT, Meijer LA, Bos JL, Bernards R. UNC45A confers resistance to histone deacetylase inhibitors and retinoic acid. Mol Cancer Res. 2009;7:1861-1870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
23.  Hahn CK, Ross KN, Warrington IM, Mazitschek R, Kanegai CM, Wright RD, Kung AL, Golub TR, Stegmaier K. Expression-based screening identifies the combination of histone deacetylase inhibitors and retinoids for neuroblastoma differentiation. Proc Natl Acad Sci USA. 2008;105:9751-9756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
24.  Kato Y, Salumbides BC, Wang XF, Qian DZ, Williams S, Wei Y, Sanni TB, Atadja P, Pili R. Antitumor effect of the histone deacetylase inhibitor LAQ824 in combination with 13-cis-retinoic acid in human malignant melanoma. Mol Cancer Ther. 2007;6:70-81.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Spiller SE, Ditzler SH, Pullar BJ, Olson JM. Response of preclinical medulloblastoma models to combination therapy with 13-cis retinoic acid and suberoylanilide hydroxamic acid (SAHA). J Neurooncol. 2008;87:133-141.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Raif A, Marshall GM, Bell JL, Koach J, Tan O, D’andreti C, Thomas W, Sekyere E, Norris M, Haber M. The estrogen-responsive B box protein (EBBP) restores retinoid sensitivity in retinoid-resistant cancer cells via effects on histone acetylation. Cancer Lett. 2009;277:82-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
27.  De los Santos M, Zambrano A, Aranda A. Combined effects of retinoic acid and histone deacetylase inhibitors on human neuroblastoma SH-SY5Y cells. Mol Cancer Ther. 2007;6:1425-1432.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Fouladi M, Park JR, Stewart CF, Gilbertson RJ, Schaiquevich P, Sun J, Reid JM, Ames MM, Speights R, Ingle AM. Pediatric phase I trial and pharmacokinetic study of vorinostat: a Children’s Oncology Group phase I consortium report. J Clin Oncol. 2010;28:3623-3629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 151]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
29.  Cheung BB, Tan O, Koach J, Liu B, Shum MS, Carter DR, Sutton S, Po’uha ST, Chesler L, Haber M. Thymosin-β4 is a determinant of drug sensitivity for Fenretinide and Vorinostat combination therapy in neuroblastoma. Mol Oncol. 2015;9:1484-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]