Randomized phase II study of valproic acid in combination with bevacizumab and oxaliplatin/fluoropyrimidine regimens in patients with RAS-mutated metastatic colorectal cancer: the REVOLUTION study protocol

Antonio Avallone, Maria Carmela Piccirillo, Elena Di Gennaro, Carmela Romano, Filomena Calabrese, Maria Serena Roca, Fabiana Tatangelo, Vincenza Granata, Antonio Cassata, Ernesta Cavalcanti, Nicola Maurea, Piera Maiolino, Lucrezia Silvestro, Alfonso De Stefano, Francesco Giuliani, Gerardo Rosati, Emiliano Tamburini, Pasquale Aprea, Valeria Vicario, Anna Nappi, Carlo Vitagliano, Rossana Casaretti, Alessandra Leone, Antonella Petrillo, Gerardo Botti, Paolo Delrio, Francesco Izzo, Francesco Perrone, Alfredo Budillon, Antonio Avallone, Maria Carmela Piccirillo, Elena Di Gennaro, Carmela Romano, Filomena Calabrese, Maria Serena Roca, Fabiana Tatangelo, Vincenza Granata, Antonio Cassata, Ernesta Cavalcanti, Nicola Maurea, Piera Maiolino, Lucrezia Silvestro, Alfonso De Stefano, Francesco Giuliani, Gerardo Rosati, Emiliano Tamburini, Pasquale Aprea, Valeria Vicario, Anna Nappi, Carlo Vitagliano, Rossana Casaretti, Alessandra Leone, Antonella Petrillo, Gerardo Botti, Paolo Delrio, Francesco Izzo, Francesco Perrone, Alfredo Budillon

Abstract

Background: Despite effective treatments, metastatic colorectal cancer (mCRC) prognosis is still poor, mostly in RAS-mutated tumors, thus suggesting the need for novel combinatorial therapies. Epigenetic alterations play an important role in initiation and progression of cancers, including CRC. Histone-deacetylase inhibitors (HDACi) have shown activity in combination with chemotherapy in the treatment of solid tumors. Owing to its HDACi activity and its safe use for epileptic disorders, valproic acid (VPA) is a good candidate for anticancer therapy that we have largely explored preclinically translating our findings in currently ongoing clinical studies. We have shown in CRC models that HDACi, including VPA, induces synergistic antitumor effects in combination with fluoropyrimidines. Furthermore, unpublished results from our group demonstrated that VPA induces differentiation and sensitization of CRC stem cells to oxaliplatin. Moreover, preclinical and clinical data suggest that HDACi may prevent/reverse anti-angiogenic resistance.

Methods/design: A randomized, open-label, two-arm, multicenter phase-II study will be performed to explore whether the addition of VPA to first line bevacizumab/oxaliplatin/fluoropyrimidine regimens (mFOLFOX-6/mOXXEL) might improve progression-free survival (PFS) in RAS-mutated mCRC patients. A sample size of 200 patients was calculated under the hypothesis that the addition of VPA to chemotherapy/bevacizumab can improve PFS from 9 to 12 months, with one-sided alpha of 0.20 and a power of 0.80. Secondary endpoints are overall survival, objective response rate, metastases resection rate, toxicity, and quality of life. Moreover, the study will explore several prognostic and predictive biomarkers on blood samples, primary tumors, and on resected metastases.

Discussion: The "Revolution" study aims to improve the treatment efficacy of RAS-mutated mCRC through an attractive strategy evaluating the combination of VPA with standard cancer treatment. Correlative studies could identify novel biomarkers and could add new insight in the mechanism of interaction between VPA, fluoropyrimidine, oxaliplatin, and bevacizumab.

Trial registration: EudraCT: 2018-001414-15; ClinicalTrials.gov identifier: NCT04310176.

Keywords: KRAS mutation; anti-VEGF; colorectal cancer; epigenetics; predictive biomarker.

Conflict of interest statement

Conflict of interest statement: The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

© The Author(s), 2020.

Figures

Figure 1.
Figure 1.
Study design.
Figure 2.
Figure 2.
Study procedures.

References

    1. Modest DP, Ricard I, Heinemann V, et al. Outcome according to KRAS-, NRAS- and BRAF-mutation as well as KRAS mutation variants: pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO colorectal cancer study group. Ann Oncol 2016; 27: 1746–1753.
    1. Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 2016; 27: 1386–1422.
    1. Avallone A, Piccirillo MC, Aloj L, et al. A randomized phase 3 study on the optimization of the combination of bevacizumab with FOLFOX/OXXEL in the treatment of patients with metastatic colorectal cancer-OBELICS (Optimization of BEvacizumab scheduLIng within Chemotherapy Scheme). BMC Cancer 2016; 16: 69.
    1. Budillon A, Di Gennaro E, Bruzzese F, et al. Histone deacetylase inhibitors: a new wave of molecular targeted anticancer agents. Recent Pat Anticancer Drug Discov 2007; 2: 119–134.
    1. Mann BS, Johnson JR, Cohen MH, et al. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 2007; 12: 1247–1252.
    1. Khot A, Dickinson M, Prince HM. Romidepsin for peripheral T-cell lymphoma. Expert Rev Hematol 2013; 6: 351–359.
    1. Lee HZ, Kwitkowski VE, Del Valle PL, et al. FDA approval: belinostat for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma. Clin Cancer Res 2015; 21: 2666–2670.
    1. Greig SL. Panobinostat: a review in relapsed or refractory multiple myeloma. Target Oncol 2016; 11: 107–114.
    1. Raha P, Thomas S, Munster PN. Epigenetic modulation: a novel therapeutic target for overcoming hormonal therapy resistance. Epigenomics 2011; 3: 451–470.
    1. Duenas-Gonzalez A, Candelaria M, Perez-Plascencia C, et al. Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors. Cancer Treat Rev 2008; 34: 206–222.
    1. Brodie MJ, Dichter MA. Antiepileptic drugs. N Engl J Med 1996; 334: 168–175.
    1. Münster 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: 1979–1985.
    1. Chateauvieux S, Morceau F, Dicato M, et al. Molecular and therapeutic potential and toxicity of valproic acid. J Biomed Biotechnol 2010; 2010: 479364.
    1. Atmaca A, Al-Batran SE, Maurer A, et al. Valproic acid (VPA) in patients with refractory advanced cancer: a dose escalating phase I clinical trial. Br J Cancer 2007; 97: 177–182.
    1. Chavez-Blanco A, Segura-Pacheco B, Perez-Cardenas E, et al. Histone acetylation and histone deacetylase activity of magnesium valproate in tumor and peripheral blood of patients with cervical cancer. A phase I study. Mol Cancer 2005; 4: 22.
    1. Arce C, Pérez-Plasencia C, González-Fierro A, et al. A proof-of-principle study of epigenetic therapy added to neoadjuvant doxorubicin cyclophosphamide for locally advanced breast cancer. PLoS One 2006; 1: e98.
    1. 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: 2488–2496.
    1. Su JM, Li XN, Thompson P, et al. Phase 1 study of valproic acid in pediatric patients with refractory solid or CNS tumors: a children’s oncology group report. Clin Cancer Res 2011; 17: 589–597.
    1. Sandor V, Bakke S, Robey RW, et al. Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res 2002; 8: 718–728.
    1. Byrd JC, Marcucci G, Parthun MR, et al. A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia. Blood 2005; 105: 959–967.
    1. Shah MH, Binkley P, Chan K, et al. Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clin Cancer Res 2006; 12: 3997–4003.
    1. Di Gennaro E, Bruzzese F, Pepe S, et al. Modulation of thymidilate synthase and p53 expression by HDAC inhibitor vorinostat resulted in synergistic antitumor effect in combination with 5FU or raltitrexed. Cancer Biol Ther 2009; 8: 782–791.
    1. Bruzzese F, Rocco M, Castelli S, et al. Synergistic antitumor effect between vorinostat and topotecan in small cell lung cancer cells is mediated by generation of reactive oxygen species and DNA damage-induced apoptosis. Mol Cancer Ther 2009; 8: 3075–3087.
    1. Di Gennaro E, Piro G, Chianese MI, et al. Vorinostat synergises with capecitabine through upregulation of thymidine phosphorylase. Br J Cancer 2010; 103: 1680–1691.
    1. Bruzzese F, Leone A, Rocco M, et al. HDAC inhibitor vorinostat enhances the antitumor effect of gefitinib in squamous cell carcinoma of head and neck by modulating ErbB receptor expression and reverting EMT. J Cell Physiol 2011; 226: 2378–2390.
    1. Terranova-Barberio M, Roca MS, Zotti AI, et al. Valproic acid potentiates the anticancer activity of capecitabine in vitro and in vivo in breast cancer models via induction of thymidine phosphorylase expression. Oncotarget 2016; 7: 7715–7731.
    1. Terranova-Barberio M, Pecori B, Roca MS, et al. Synergistic antitumor interaction between valproic acid, capecitabine and radiotherapy in colorectal cancer: critical role of p53. J Exp Clin Cancer Res 2017; 36: 177.
    1. Miller KM, Tjeertes JV, Coates J, et al. Human HDAC1 and HDAC2 function in the DNA-damage response to promote DNA nonhomologous end-joining. Nat Struct Mol Biol 2010; 17: 1144–1151.
    1. Avallone A, Piccirillo MC, Delrio P, et al. Phase 1/2 study of valproic acid and short-course radiotherapy plus capecitabine as preoperative treatment in low-moderate risk rectal cancer-V-shoRT-R3 (valproic acid-short radiotherapy-rectum 3rd trial). BMC Cancer 2014; 14: 875.
    1. Zhao J. Cancer stem cells and chemoresistance: the smartest survives the raid. Pharmacol Ther 2016; 160: 145–158.
    1. Basseville AS, Sourbier C, Robey RW, et al. Metabolic reprogramming in KRAS mutant cancer cells may cause sensitivity to the histone deacetylase (HDAC) inhibitor romidepsin. Cancer Res 2015; 75(Suppl. 15): Abstract 1771.
    1. Ellis L, Hammers H, Pili R. Targeting tumor angiogenesis with histone deacetylase inhibitors. Cancer Lett 2009; 280: 145–153.
    1. Aggarwal R, Thomas S, Pawlowska N, et al. Inhibiting histone deacetylase as a means to reverse resistance to angiogenesis inhibitors: phase I study of abexinostat plus pazopanib in advanced solid tumor malignancies. J Clin Oncol 2017; 35: 1231–1239.
    1. Friday BB, Anderson SK, Buckner J, et al. Phase II trial of vorinostat in combination with bortezomib in recurrent glioblastoma: a North central cancer treatment group study. Neuro Oncol 2012; 14: 215–221.
    1. Chinnaiyan P, Chowdhary S, Potthast L, et al. Phase I trial of vorinostat combined with bevacizumab and CPT-11 in recurrent glioblastoma. Neuro Oncol 2012; 14: 93–100.
    1. Ramaswamy B, Fiskus W, Cohen B, et al. Phase I-II study of vorinostat plus paclitaxel and bevacizumab in metastatic breast cancer: evidence for vorinostat-induced tubulin acetylation and Hsp90 inhibition in vivo. Breast Cancer Res Treat 2012; 132: 1063–1072.
    1. Pili R, Liu G, Chintala S, et al. Combination of the histone deacetylase inhibitor vorinostat with bevacizumab in patients with clear-cell renal cell carcinoma: a multicentre, single-arm phase I/II clinical trial. Br J Cancer 2017; 116: 874–883.
    1. Peters KB, Lipp ES, Miller E, et al. Phase I/II trial of vorinostat, bevacizumab, and daily temozolomide for recurrent malignant gliomas. J Neurooncol 2018; 137: 349–356.
    1. Ghiaseddin A, Reardon D, Massey W, et al. Phase II study of bevacizumab and vorinostat for patients with recurrent World Health Organization grade 4 malignant glioma. Oncologist 2018; 23: 157–e21.
    1. Lee EQ, Reardon DA, Schiff D, et al. Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma. Neuro Oncol 2015; 17: 862–867.
    1. Strickler JH, Starodub AN, Jia J, et al. Phase I study of bevacizumab, everolimus, and panobinostat (LBH-589) in advanced solid tumors. Cancer Chemother Pharmacol 2012; 70: 251–258.
    1. Drappatz J, Lee EQ, Hammond S, et al. Phase I study of panobinostat in combination with bevacizumab for recurrent high-grade glioma. J Neurooncol 2012; 107: 133–138.
    1. Wheler JJ, Janku F, Falchook GS, et al. Phase I study of anti-VEGF monoclonal antibody bevacizumab and histone deacetylase inhibitor valproic acid in patients with advanced cancers. Cancer Chemother Pharmacol 2014; 73: 495–501.
    1. Michaelis M, Michaelis UR, Fleming I, et al. Valproic acid inhibits angiogenesis in vitro and in vivo. Mol Pharmacol 2004; 65: 520–527.
    1. Michaelis M, Suhan T, Michaelis UR, et al. Valproic acid induces extracellular signal-regulated kinase 1/2 activation and inhibits apoptosis in endothelial cells. Cell Death Differ 2006; 13: 446–453.
    1. Gao D, Xia Q, Lv J, et al. Chronic administration of valproic acid inhibits PC3 cell growth by suppressing tumor angiogenesis in vivo. Int J Urol 2007; 14: 838–845.
    1. Isenberg JS, Jia Y, Field L, et al. Modulation of angiogenesis by dithiolethione-modified NSAIDs and valproic acid. Br J Pharmacol 2007; 151: 63–72.
    1. Shabbeer S, Kortenhorst MS, Kachhap S, et al. Multiple molecular pathways explain the anti-proliferative effect of valproic acid on prostate cancer cells in vitro and in vivo. Prostate 2007; 67: 1099–1110.
    1. Byler TK, Leocadio D, Shapiro O, et al. Valproic acid decreases urothelial cancer cell proliferation and induces thrombospondin-1 expression. BMC Urol 2012; 12: 21.
    1. Kvestad H, Evensen L, Lorens JB, et al. In vitro characterization of valproic acid, ATRA, and cytarabine used for disease-stabilization in human acute myeloid leukemia: antiproliferative effects of drugs on endothelial and osteoblastic cells and altered release of angioregulatory mediators by endothelial cells. Leuk Res Treatment 2014; 2014: 143479.
    1. Zhao Y, You W, Zheng J, et al. Valproic acid inhibits the angiogenic potential of cervical cancer cells via HIF-1α/VEGF signals. Clin Transl Oncol 2016; 18: 1123–1130.
    1. Chelluri R, Caza T, Woodford MR, et al. Valproic acid alters angiogenic and trophic gene expression in human prostate cancer models. Anticancer Res 2016; 36: 5079–5086.
    1. Wishart DS, Mandal R, Stanislaus A, et al. Cancer metabolomics and the human metabolome database. Metabolites 2016; 6: 10.
    1. Siravegna G, Mussolin B, Buscarino M, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med 2015; 21: 795–801.
    1. Lanuti P, Rotta G, Almici C, et al. Endothelial progenitor cells, defined by the simultaneous surface expression of VEGFR2 and CD133, are not detectable in healthy peripheral and cord blood. Cytometry A 2016; 89: 259–270.
    1. Avallone A, Di Gennaro E, Delrio P, et al. Neoadjuvant multidisciplinary phase II study (BRANCH) of an early bevacizumab schedule plus chemo-radiation therapy in rectal cancer: efficacy, safety, and biomarkers. In: AACR 103rd Annual Meeting, Mar 31–April 4, 2012, Chicago, IL. Abstract LB-219.
    1. Russo L, Grilli B, Gelzo M, et al. Rapid and accurate determination of 5-fluorouracil in human plasma by LC-UV method. Pharm Chem J 2017; 4: 1–8.
    1. Mlecnik B, Bindea G, Angell HK, et al. Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity 2016; 44: 698–711.
    1. Hughes PE, Caenepeel S, Wu LC. Targeted therapy and checkpoint immunotherapy combinations for the treatment of cancer. Trends Immunol 2016; 37: 462–476.
    1. Terranova-Barberio M, Thomas S, Munster PN. Epigenetic modifiers in immunotherapy: a focus on checkpoint inhibitors. Immunotherapy 2016; 8: 705–719.
    1. Morganroth J, Shah RR, Scott JW. Evaluation and management of cardiac safety using the electrocardiogram in oncology clinical trials: focus on cardiac repolarization (QTc interval). Clin Pharmacol Ther 2010; 87: 166–174.
    1. Ederhy S, Izzedine H, Massard C, et al. Cardiac side effects of molecular targeted therapies: towards a better dialogue between oncologists and cardiologists. Crit Rev Oncol Hematol 2011; 80: 369–379.
    1. Malapelle U, Pisapia P, Sgariglia R, et al. Less frequently mutated genes in colorectal cancer: evidences from next-generation sequencing of 653 routine cases. J Clin Pathol 2016; 69: 767–771.
    1. Malapelle U, De Stefano A, Carlomagno C, et al. Next-generation sequencing in the genomic profiling of synchronous colonic carcinomas: comment on Li et al (2015). J Clin Pathol 2015; 68: 946–947.
    1. Chung EJ, Lee S, Sausville EA, et al. Histone deacetylase inhibitor pharmacodynamic analysis by multiparameter flow cytometry. Ann Clin Lab Sci 2005; 35: 397–406.
    1. Ronzoni S, Faretta M, Ballarini M, et al. New method to detect histone acetylation levels by flow cytometry. Cytometry A 2005; 66: 52–61.
    1. Capone F, Guerriero E, Colonna G, et al. The cytokinome profile in patients with hepatocellular carcinoma and type 2 diabetes. PLoS One 2015; 10: e0134594.
    1. Sorice A, Siano F, Capone F, et al. Potential anticancer effects of polyphenols from chestnut shell extracts: modulation of cell growth, and cytokinomic and metabolomic profiles. Molecules 2016; 21: 1411.
    1. Aaronson NK, Ahmedzai S, Bergman B, et al. The European organization for research and treatment of cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst 1993; 85: 365–376.
    1. Mandard AM, Dalibard F, Mandard JC, et al. Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer 1994; 73: 2680–2686.
    1. Budillon A, Delrio P, Pecori B, et al. Phase I/II study of valproic acid (VPA) and short-course radiotherapy (SCRT) plus capecitabine (CAP) as preoperative treatment in low-moderate risk rectal cancer (V-shoRT-R3). Ann Oncol 2018; 29(Suppl. 8): viii167-viii168.

Source: PubMed

3
Abonnere