1,611
Views
43
CrossRef citations to date
0
Altmetric
Editorial

Could valproic acid be an effective anticancer agent? The evidence so far

&

Abstract

Valproic acid is an inhibitor of class I histone deacetylases. Epigenetic therapies in cancer have been focus of a keen interest and histone deacetylase inhibitors, in particular, have been approved for certain types of hematologic malignancies. Valproic acid is an attractive candidate for cancer therapy due to its mechanism of action, its low cost and generally good clinical tolerability. In the following editorial, we will review its role as monotherapy for cancer, its place in combination epigenetic therapy, and its role as chemosensitizer, and cancer preventative agent.

With the discovery of its function as an inhibitor of class I and IIa histone deacetylases (HDACs), there arose a significant interest in assessing the possible role of valproic acid (VPA), a well-established anti-seizure drug, in cancer therapy Citation[1,2]. Epigenetic changes such as aberrant DNA methylation and histone acetylation are common in cancer, providing a strong rationale for the use of epigenetic therapies. Moreover, the completion of The Cancer Genome Atlas (TCGA) project has demonstrated frequent mutations in critical epigenetic regulators, further strengthening a link between genetic and epigenetic events in cancer. Due to its low cost, favorable side-effect profile and its ease in crossing the blood–brain barrier, VPA is an attractive drug candidate for a variety of possible indications. In the following paragraphs, we will summarize the available evidence for VPA’s role in cancer therapy and in cancer prevention.

VPA as monotherapy for cancer

The experience with VPA as monotherapy for cancer is limited. Interesting findings exist in metastatic neuroendocrine carcinomas, where VPA exposure has been shown to increase Notch1 expression Citation[3], considered to act as a tumor-suppressor gene. In a small Phase I study with eight patients, treated at target concentrations between 50 and 100 µg/ml, one patient achieved a partial response, while five patients had stable disease. High-level Notch-1 induction was associated with partial response to VPA. In myeloid malignancies, VPA has been shown in vitro to induce both apoptosis and differentiation in leukemic blasts, leading to trials assessing its role as monotherapy or in combination with all-trans-retinoic acid in either acute myelogenous leukemia (AML) or myelodysplastic syndrome (MDS). Response rates in a Phase II study of 58 patients with AML were 5% Citation[4] compared to 44% in a separate study of 18 patients with MDS Citation[5].

VPA as epigenetic combination therapy for cancer

Given the modest benefit with VPA monotherapy in cancer, it is not surprising that the majority of evidence for a possible role of VPA as an anticancer drug is based on its effects observed in combination with other drugs. For simplicity sake, these can be categorized into combination therapies with other epigenetic modifiers, combinations with cytotoxic chemotherapy agents and combinations with immune modulators. The discovery in 2006 that treatment with the DNA methyltransferase 1 (DNMT1) inhibitor azacytidine was associated with dramatically improved overall survival compared with conventional care (hazard ratio [HR] 0.58; 95% CI: 0.43–0.77) in patients with poor-risk myelodysplasia was proof of concept for a major role of epigenetic therapies in cancer Citation[6]. Since HDAC inhibition was found to be synergistic with azacytidine in vitro, numerous studies have focused on combinations of demethylating agents with HDAC inhibitors including VPA. In a Phase II study of VPA with all-trans-retinoic acid and azacytidine in patients with poor-risk AML and MDS, responses were observed in 23% of patients and median overall survival was 12.4 months. In this study, the responses correlated with the demethylation of several aberrantly methylated promoter regions. These survival rates were inferior to those observed with azacytidine alone in either high-risk MDS or AML (24.5 months) Citation[6,7]. However, this could partly be explained by patient selection and the number of previous treatment regimens. It does raise the important question, however, if HDAC inhibition adds clear benefit to that of demethylating agents alone. A randomized Phase II study of decitabine plus VPA versus decitabine alone in 76 patients with AML and MDS found a marginally improved response rate in the VPA arm (53 vs 42%, p = NS) without improvement in survival. These findings are supported by the recent lack of benefit when the HDAC inhibitor entinostat was added to azacytidine in the E1905 trial for the treatment of AML and MDS Citation[8]. Neurotoxicity was also a major dose-limiting toxicity in a Phase I study of eight patients with advanced non-small cell lung cancer. No responses were seen in this small cohort and somnolence, lethargy and disorientation was observed at low concentrations Citation[9].

VPA in combination with cytotoxic chemotherapy for cancer

An interesting and unexpected finding of a Phase I/II trial of azacytidine in combination with the HDACi entinostat in non-small cell lung cancer was the observation that epigenetic therapy seemed to restore sensitivity to previously used chemotherapeutics Citation[10]. The precise mechanism for this observation remains unclear. Based on the hypothesis that HDACi mediated changes in chromatin structure favoring a more euchromatic chromatin pattern, VPA has been studied in combination with cytotoxic chemotherapy, particularly with DNA-damaging agents: in combination with the topoisomerase II inhibitor epirubicin, responses were seen in 22% of 44 patients including those with tumors considered to be refractory to anthracyclines such as melanoma or in patients with previous anthracycline exposure Citation[11]. In a subsequent Phase II extension cohort of 15 patients with metastatic breast cancer, VPA in combination with 5-FU, epirubicin and cyclophosphamide produced objective responses in 64% of patients with acceptable toxicities Citation[12]. Potential synergy between VPA and doxorubicin was also observed in a Phase II study of 16 patients with unresectable and platinum-refractory mesothelioma Citation[13], a clinical scenario for which no accepted treatment options exist. VPA and doxorubicin yielded encouraging response rates of 16% and disease control rates of 36%. In combination with a new topoisomerase I inhibitor karenitecin, VPA use led to disease stabilization in 47% of patients treated in a Phase I/II study for metastatic melanoma Citation[14]. In a small randomized study of 36 patients with advanced cervical cancer, the addition of epigenetic therapy with hydralazine and VPA to cisplatin and topotecan led to a statistically significant improvement in the progression-free survival of 10 versus 6 months (p = 0.034) Citation[15] compared to chemotherapy alone, suggesting for the first time in a randomized fashion the possible superiority of VPA-based epigenetic therapy as chemosensitizer. Similar results were observed in other studies in which VPA-based epigenetic therapies were combined with platinum-based chemotherapies in an attempt to overcome previous platinum resistance Citation[16]. Interestingly, a VPA-induced increase in H3 acetylation has also been shown to prevent the emergence of resistance to mammalian target of rapamycin inhibitors in renal cell cancer Citation[17].

VPA in cancer prevention

It is known that HDAC inhibition can lead to reduced levels of DNMT1 expression Citation[18]. A recent report from our laboratory showed that class I HDAC-mediated stabilization of DNMT1 protein expression is an early event in smoke carcinogen-induced transformation of bronchial epithelial cells Citation[19]. This was associated with the uncoupling of DNMT1 expression from the usually tight limitation to the S-phase of the cell cycle, leading to de-novo methylation and epigenetic silencing of tumor-suppressor genes. Importantly, treatment with VPA partially reversed aberrant DNA methylation, leading to re-expression of previously silenced genes and suppression of anchorage-independent colony formation. We hypothesized based on these data that VPA may play an important role in chemoprevention of smoke-related malignancies such as lung, head and neck and bladder cancer. In a retrospective cohort study of 439,628 US veterans with indications for routine clinical use of VPA (bipolar disorder, seizure d/o, PTSD, migraines), the risk only for squamous cell carcinomas of the head and neck was significantly reduced in the 26,911 patients with long-term VPA use (HR: 0.66; 95% CI: 0.48–0.92) Citation[20]. The risk for lung, bladder, prostate and colon cancers was not statistically different between VPA users and non-users. Risk reduction was only observed in patients with median VPA levels in the therapeutic range (>40 µM) for seizure prevention and HDAC inhibition and only after at least 3 years of use, reducing the likelihood that the conclusions were based on spurious results. The lack of effect on lung, colon and prostate cancer risk is confirmed in a study from Denmark, which did not find a significant correlation between VPA use and cancer risk between VPA users and non-users, but did not specifically investigate head and neck cancer risk Citation[21]. A third study based on the UK General Practice Research database found no impact of VPA on total cancer incidence, but did detect an increase in colorectal cancers (HR: 3.95; 95% CI: 1.97–7.92, p = 0.001) and a trend towards increased prostate cancer risk (HR: 2.15; 95% CI: 0.92–5.02; p = 0.08) and decreased breast cancer risk (HR: 0.40; 95% CI: 0.14–1.30; p = 0.08) Citation[22]. The incidence of head and neck cancer was low, preventing conclusions about VPA’s role in further reducing its risk in this study. The increased risk for colon cancer was neither seen in our study nor in the Danish study, making an uneven distribution of risk factors the most likely explanation for this phenomenon. The risk reduction for head and neck cancer in our study is interesting and seems to be largely limited to patients with non-oropharyngeal head and neck cancer, making an antiviral effect of VPA against human papilloma virus driven cancers less likely as etiology. It will need to be seen in further studies if this effect can be generalized to other smoking-related squamous cell carcinomas of the upper aerodigestive tract such as those of the lung and the esophagus. Despite its large size, our study lacked the sufficient number of cases to conclusively answer this question. Likewise, our study does not answer the question if resistance to VPA emerges in the epithelium of the head and neck after long-term exposure and the mechanism by which it is mediated.

Conclusion

While VPA’s role in cancer therapy as monotherapy or in combination epigenetic therapy may only be moderate both due to toxicity and limited efficacy, its use in combination regimens with cytotoxic chemotherapy and in chemoprevention against upper aerodigestive malignancies may hold promise and deserves further study.

Financial & competing interests disclosure

Grant support for J Brandes: Veterans’ Health Administration Career Development Award 7-IK2BX001283-03; NCI- 5 P50 CA128613-02 Career Development Project to JCB. The contents of this publication do not necessarily reflect the views of the Department of Veterans’ Affairs or the United States Government. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Kramer OH, Zhu P, Ostendorff HP, et al. The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2. EMBO J 2003;22:3411-20
  • Gurvich N, Tsygankova OM, Meinkoth JL, Klein PS. Histone deacetylase is a target of valproic acid-mediated cellular differentiation. Cancer Res 2004;64:1079-86
  • Mohammed TA, Holen KD, Jaskula-Sztul R, et al. A pilot phase II study of valproic acid for treatment of low-grade neuroendocrine carcinoma. Oncologist 2011;16:835-43
  • Pilatrino C, Cilloni D, Messa E, et al. Increase in platelet count in older, poor-risk patients with acute myeloid leukemia or myelodysplastic syndrome treated with valproic acid and all-trans retinoic acid. Cancer 2005;104:101-9
  • Kuendgen A, Knipp S, Fox F, et al. Results of a phase 2 study of valproic acid alone or in combination with all-trans retinoic acid in 75 patients with myelodysplastic syndrome and relapsed or refractory acute myeloid leukemia. Ann Hematol 2005;84(Suppl 1):61-6
  • Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 2009;10:223-32
  • Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol 2010;28:562-9
  • Prebet T, Sun Z, Figueroa ME, et al. Prolonged administration of azacitidine with or without entinostat for myelodysplastic syndrome and acute myeloid leukemia with myelodysplasia-related changes: results of the US Leukemia Intergroup Trial E1905. J Clin Oncol 2014;32:1242-8
  • Chu BF, Karpenko MJ, Liu Z, et al. Phase I study of 5-aza-2’-deoxycytidine in combination with valproic acid in non-small-cell lung cancer. Cancer Chemother Pharmacol 2013;71:115-21
  • Juergens RA, Wrangle J, Vendetti FP, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov 2011;1:598-607
  • Munster P, Marchion D, Bicaku E, et al. Phase I trial of histone deacetylase inhibition by valproic acid followed by the topoisomerase II inhibitor epirubicin in advanced solid tumors: a clinical and translational study. J Clin Oncol 2007;25:1979-85
  • Munster P, Marchion D, Bicaku E, et al. Clinical and biological effects of valproic acid as a histone deacetylase inhibitor on tumor and surrogate tissues: phase I/II trial of valproic acid and epirubicin/FEC. Clin Cancer Res 2009;15:2488-96
  • Scherpereel A, Berghmans T, Lafitte JJ, et al. Valproate-doxorubicin: promising therapy for progressing mesothelioma. A phase II study. Eur Respir J 2011;37:129-35
  • Daud AI, Dawson J, DeConti RC, et al. Potentiation of a topoisomerase I inhibitor, karenitecin, by the histone deacetylase inhibitor valproic acid in melanoma: translational and phase I/II clinical trial. Clin Cancer Res 2009;15:2479-87
  • Coronel J, Cetina L, Pacheco I, et al. A double-blind, placebo-controlled, randomized phase III trial of chemotherapy plus epigenetic therapy with hydralazine valproate for advanced cervical cancer. Preliminary results. Med Oncol 2011;28(Suppl 1):S540-6
  • Falchook GS, Fu S, Naing A, et al. Methylation and histone deacetylase inhibition in combination with platinum treatment in patients with advanced malignancies. Invest New Drugs 2013;31:1192-200
  • Juengel E, Dauselt A, Makarevic J, et al. Acetylation of histone H3 prevents resistance development caused by chronic mTOR inhibition in renal cell carcinoma cells. Cancer Lett 2012;324:83-90
  • Du Z, Song J, Wang Y, et al. DNMT1 stability is regulated by proteins coordinating deubiquitination and acetylation-driven ubiquitination. Sci Signal 2010;3:ra80
  • Brodie SA, Li G, El-Kommos A, et al. Class I HDACs are mediators of smoke carcinogen-induced stabilization of DNMT1 and serve as promising targets for chemoprevention of lung cancer. Cancer Prev Res (Phila) 2014;7:351-61
  • Kang H, Gillespie TW, Goodman M, et al. Long-term use of valproic acid in US veterans is associated with a reduced risk of smoking-related cases of head and neck cancer. Cancer 2014;120:1394-400
  • Hallas J, Friis S, Bjerrum L, et al. Cancer risk in long-term users of valproate: a population-based case-control study. Cancer Epidemiol Biomarkers Prev 2009;18:1714-19
  • Singh G, Bell GS, Driever PH, Sander JW. Cancer risk in people with epilepsy using valproate sodium. Acta Neurol Scand 2012;125:234-40

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.