A proof-of-principle study of epigenetic therapy added to neoadjuvant doxorubicin cyclophosphamide for locally advanced breast cancer

Claudia Arce, Carlos Pérez-Plasencia, Aurora González-Fierro, Erick de la Cruz-Hernández, Alma Revilla-Vázquez, Alma Chávez-Blanco, Catalina Trejo-Becerril, Enrique Pérez-Cárdenas, Lucia Taja-Chayeb, Enrique Bargallo, Patricia Villarreal, Teresa Ramírez, Teresa Vela, Myrna Candelaria, Maria F Camargo, Elizabeth Robles, Alfonso Dueñas-González, Claudia Arce, Carlos Pérez-Plasencia, Aurora González-Fierro, Erick de la Cruz-Hernández, Alma Revilla-Vázquez, Alma Chávez-Blanco, Catalina Trejo-Becerril, Enrique Pérez-Cárdenas, Lucia Taja-Chayeb, Enrique Bargallo, Patricia Villarreal, Teresa Ramírez, Teresa Vela, Myrna Candelaria, Maria F Camargo, Elizabeth Robles, Alfonso Dueñas-González

Abstract

Background: Aberrant DNA methylation and histone deacetylation participate in cancer development and progression; hence, their reversal by inhibitors of DNA methylation and histone deacetylases (HDACs) is at present undergoing clinical testing in cancer therapy. As epigenetic alterations are common to breast cancer, in this proof-of-concept study demethylating hydralazine, plus the HDAC inhibitor magnesium valproate, were added to neoadjuvant doxorubicin and cyclophosphamide in locally advanced breast cancer to assess their safety and biological efficacy.

Methodology: This was a single-arm interventional trial on breast cancer patients (ClinicalTrials.gov Identifier: NCT00395655). After signing informed consent, patients were typed for acetylator phenotype and then treated with hydralazine at 182 mg for rapid-, or 83 mg for slow-acetylators, and magnesium valproate at 30 mg/kg, starting from day -7 until chemotherapy ended, the latter consisting of four cycles of doxorubicin 60 mg/m2 and cyclophosphamide 600 mg/m2 every 21 days. Core-needle biopsies were taken from primary breast tumors at diagnosis and at day 8 of treatment with hydralazine and valproate.

Main findings: 16 patients were included and received treatment as planned. All were evaluated for clinical response and toxicity and 15 for pathological response. Treatment was well-tolerated. The most common toxicity was drowsiness grades 1-2. Five (31%) patients had clinical CR and eight (50%) PR for an ORR of 81%. No patient progressed. One of 15 operated patients (6.6%) had pathological CR and 70% had residual disease <3 cm. There was a statistically significant decrease in global 5mC content and HDAC activity. Hydralazine and magnesium valproate up- and down-regulated at least 3-fold, 1,091 and 89 genes, respectively.

Conclusions: Hydralazine and magnesium valproate produce DNA demethylation, HDAC inhibition, and gene reactivation in primary tumors. Doxorubicin and cyclophosphamide treatment is safe, well-tolerated, and appears to increase the efficacy of chemotherapy. A randomized phase III study is ongoing to support the efficacy of so-called epigenetic or transcriptional cancer therapy.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Flow diagram of the trial.
Figure 1. Flow diagram of the trial.
Figure 2. Global DNA methylation.
Figure 2. Global DNA methylation.
The five patients had a reduction in 5mC content varying from 0.7−2.59% decrease. The mean 5mC in the five pre-treatment samples was 5.71% (standard deviation [SD], 1.43) which decreased to 4.2% (SD, 0.95). This difference was statistically significant (p = 0.043).
Figure 3. Histone deacetylase activity.
Figure 3. Histone deacetylase activity.
The five patients assayed showed a reduction in enzymatic activity. In Y axis are the absolute values in optical density (OD) units. Higher and lower decreases were 0.3845 and 0.0175, for a mean decrease of 0.1624. This difference was statistically significant (p = 0.042).
Figure 4. Hierarchical Cluster Analysis.
Figure 4. Hierarchical Cluster Analysis.
The cluster shown represents 3,117 genes. Each row represents a gene, whereas each column corresponds to a tissue sample. The relative abundance of the gene in the tissue correlates with color intensity (red, induced; green, repressed; black, no change). On the dendogram, post-treated clinical samples clustered together.
Figure 5. Principal Component Analysis.
Figure 5. Principal Component Analysis.
Principal Component Analysis, showing in three-dimensional space relationship between samples.
Figure 6. Gene expression by RT-PCR.
Figure 6. Gene expression by RT-PCR.
RT-PCR of NDUFA13 and DAPPER genes in biopsies from the primary tumor showing that these genes were reactivated after treatment. This corresponds to one of the three patients whose biopsy (only post-treatment) was analyzed by microarray and showed over-expression of these genes.

References

    1. Zhu WG, Otterson GA. The interaction of histone deacetylase inhibitors and DNA methyltransferase inhibitors in the treatment of human cancer cells. Curr Med Chem Anti-Cancer Agents. 2003;3:187–199.
    1. Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet. 1999;21:103–107.
    1. Li L, Shi H, Yiannoutsos C, Huang TH, Nephew KP. Epigenetic hypothesis tests for methylation and acetylation in a triple microarray system. J Comput Biol. 2005;12:370–390.
    1. Primeau M, Gagnon J, Momparler RL. Synergistic antineoplastic action of DNA methylation inhibitor 5-AZA-2′-deoxycytidine and histone deacetylase inhibitor depsipeptide on human breast carcinoma cells. Int J Cancer. 2003;103:177–184.
    1. Zhu WG, Lakshmanan RR, Beal MD, Otterson GA. DNA methyltransferase inhibition enhances apoptosis induced by histone deacetylase inhibitors. Cancer Res. 2001;61:1327–1333.
    1. Klisovic MI, Maghraby EA, Parthun MR, Guimond M, Sklenar AR, et al. Depsipeptide (FR 901228) promotes histone acetylation, gene transcription, apoptosis and its activity is enhanced by DNA methyltransferase inhibitors in AML1/ETO-positive leukemic cells. Leukemia. 2003;17:350–358.
    1. Gagnon J, Shaker S, Primeau M, Hurtubise A, Momparler RL. Interaction of 5-aza-2′-deoxycytidine and depsipeptide on antineoplastic activity and activation of 14-3-3sigma, E-cadherin and tissue inhibitor of metalloproteinase 3 expression in human breast carcinoma cells. Anticancer Drugs. 2003;14:193–202.
    1. Bovenzi V, Momparler RL. Antineoplastic action of 5-aza-2′-deoxycytidine and histone deacetylase inhibitor and their effect on the expression of retinoic acid receptor beta and estrogen receptor alpha genes in breast carcinoma cells. Cancer Chemother Pharmacol. 2001;48:71–76.
    1. Gore SD, Baylin S, Sugar E, Carraway H, Miller CB, et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res. 2006;66:6361–6369.
    1. García-Manero G, Kantarjian HM, Sánchez-González B, Yang H, Rosner G, et al. Phase I/II study of the combination of 5-aza-2′ -deoxycytidine with valproic acid in patients with leukemia. Blood. 2006 Aug 1 [Epub ahead of print]
    1. Karagiannis TC, El-Osta A. Modulation of cellular radiation responses by histone deacetylase inhibitors. Oncogene. 2006;25:3885–3893.
    1. Kim MS, Blake M, Baek JH, Kohlhagen G, Pommier Y, et al. Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer Res. 2003;63:7291–7300.
    1. Chávez-Blanco A, Pérez-Plasencia C, Párez-Cárdenas E, Carrasco-Legleu C, Rangel-López E, et al. Antineoplastic effects of the DNA methylation inhibitor hydralazine and the histone deacetylase inhibitor valproic acid in cancer cell lines. Cancer Cell Int. 2006;6:2.
    1. Kim MS, Blake M, Baek JH, Kohlhagen G, Pommier Y, Carrier F. Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer Res. 2003;63:7291–7300.
    1. Sonnemann J, Kumar KS, Heesch S, Muller C, Hartwig C, et al. Histone deacetylase inhibitors induce cell death and enhance the susceptibility to ionizing radiation, etoposide, and TRAIL in medulloblastoma cells. Int J Oncol. 2006;28:755–766.
    1. Pérez-Plasencia C, Dueñas-González A. Can the state of cancer chemotherapy resistance be reverted by epigenetic therapy? Mol Cancer. 2006;5:27.
    1. Huang TH, Perry MR, Laux DE. Methylation profiling of CpG islands in human breast cancer cells. Hum Mol Genet. 1999;8:459–470.
    1. Yang X, Yan L, Davidson NE. DNA methylation in breast cancer. Endocr Relat Cancer. 2001;8:115–127.
    1. Mielnicki LM, Asch HL, Asch BB. Genes, chromatin, and breast cancer: an epigenetic tale. J Mammary Gland Biol Neoplasia. 2001;6:169–182.
    1. Shi H, Rahmatpanah F, Wei SH, Caldwell CW, Huang TH. Dissecting complex epigenetic alterations in breast cancer using CpG island microarrays. Cancer Res. 2001;61:8375–8380.
    1. Kaufman M, Hortobagyi GN, Goldhrisch A. Recommendations from an International Expert Panel on the use of neoadjuvant (primary) systemic treatment of operable breast cancer: an update. J Clin Oncol. 2006;24:1940–1949.
    1. van der Hage JA, van der Hage JA, van de Velde CJ, Julien JP, Tubiana-Hulin M, et al. Preoperative chemotherapy in primary operable breast cancer: results from the European Organization for Research and Treatment of Cancer Trial 10902. J Clin Oncol. 2001;19:4224–4237.
    1. Fisher B, Fisher B, Brown A, Mamounas E. Effect of preoperative chemotherapy on local regional disease in women with operable breast cancer: findings from National Surgical Adjuvant Breast and Bowel Project B-18. J Clin Oncol. 1997;15:2483–2493.
    1. Scholl SM, Pierga J, Asselain B, et al. Breast tumor response to primary chemotherapy predicts local and distant control as well as survival. Eur J Cancer. 1995;31:1969–1975.
    1. Kuerer HM, Newman LA, Smith TL. Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy. J Clin Oncol. 1999;17:460–469.
    1. Castañeda-Hernández G, Falcón-Neri A, Herrera-Abarca A, Herrera JE, Flores-Murrieta FJ. Determination of three acetylator phenotypes in a Mexican population using sulfamethazine metabolic ratio. Am J Ther. 1995;2:57–60.
    1. Sandoval Guerrero K, Revilla Vázquez A, Segura-Pacheco B, Dueñas-González A. Determination of 5-methyl-cytosine and cytosine in tumor DNA samples of cancer patients. Electrophoresis. 2005;26:1057–1062.
    1. Chávez-Blanco A, Segura-Pacheco B, Pérez-Cárdenas E, Taja-Chayeb L, Cetina L, 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. Reece PA, Cozamanis I, Zacest R. Selective high-performance liquid chromatographic assays for hydralazine and its metabolites in plasma of man. J Chromatogr. 1980;181:427–440.
    1. Sturn A, Quaknebush J, Trajanoski Z. Genesis: cluster analysis of microarray data. Bioinformatics. 2002;18:207–208.
    1. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98:5116–5121.
    1. Mauri D, Pavlidis N, Ioannidis JPA. Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta analysis. J Natl Cancer Inst. 2005;97:188–194.
    1. Scholl SM, Fourquet A, Asselain B. Neoadjuvant versus adjuvant chemotherapy in premenopausal patients with tumours considered too large for breast conserving surgery: preliminary results of a randomised trial: S6. Eur J Cancer. 1994;5:645–652.
    1. Schwartz GF, Birchansky CA, Komarnicky LTl. Induction chemotherapy followed by breast conservation for locally advanced carcinoma of the breast. Cancer. 1994;73:362–369.
    1. Powles TJ, Hickish TF, Makris A. Randomized trial of chemoendocrine therapy started before or after surgery for treatment of primary breast cancer. J Clin Oncol. 1995;13:547–552.
    1. Brain E, Garrino C, Misset JL, Carbonero IG, Itzhaki M, et al. Long-term prognostic and predictive factors in 107 stage II/III breast cancer patients treated with anthracycline-based neoadjuvant chemotherapy. Br J Cancer. 1997;79:1360–1367.
    1. Morrell LE, Lee YJ, Hurley J, et al. A phase II trial of neoadjuvant methotrexate, vinblastine, doxorubicin, and cisplatin in the treatment of patients with locally advanced breast carcinoma. Cancer. 1998;82:503–511.
    1. Bonadonna G, Valagussa P, Brambilla C, et al. Primary chemotherapy in operable breast cancer: eight-year experience at the Milan Cancer Institute. J Clin Oncol. 1998;16:93–100.
    1. Fernández-Sánchez M, Gamboa-Domínguez A, Uribe N, García-Ulloa AC, Flores-Estrada D, et al. Clinical and pathological predictors of the response to neoadjuvant anthracycline chemotherapy in locally advanced breast cancer. Med Oncol. 1998;23:171–183.
    1. Rouzier R, Extra JM, Klijanienko J. Incidence and prognostic significance of complete axillary downstaging after primary chemotherapy in breast cancer patients with T1 to T3 tumors and cytologically proven axillary metastatic lymph nodes. J Clin Oncol. 2002;20:1304–1310.
    1. Rouzier R, Pusztai L, Delaloge S, González-Angulo AM, Andre F, et al. Nomograms to predict pathologic complete response and metastasis-free survival alter preoperative chemotherapy for breast cancer. J Clin Oncol. 2005;23:8331–8339.
    1. Acharya S, Bussel JB. Hematologic toxicity of sodium valproate. J Pediatr Hematol Oncol. 2000;22:62–65.
    1. Bourg V, Lebrun C, Chichmanian RM, Thomas P, Frenay M. Nitroso-urea-cisplatin-based chemotherapy associated with valproate: increase of haematologic toxicity. Ann Oncol. 2001;12:217–219.
    1. Bug G, Gul H, Schwarz K, Pfeifer H, Kampfmann M, Zheng X, et al. Valproic acid stimulates proliferation and self-renewal of hematopoietic stem cells. Cancer Res. 2005;65:2537–2541.
    1. Lesser JM, Israili ZH, Davis DC, Dayton PG. Metabolism and disposition of hydralazine-14C in man and dog. Drug Metab Dispos. 1974;2:351–360.
    1. Goffin J, Eisenhauer E. DNA methyltransferase inhibitors- state of the art. Ann Oncol. 2002;13:1699–716.
    1. Claus R, Almstedt M, Lubbert M. Epigenetic treatment of hematopoietic malignancies: in vivo targets of demethylating agents. Semin Oncol. 2005;32:511–520.
    1. Mai A, Massa S, Rotili D, Cerbara I, Valente S, et al. Histone deacetylation in epigenetics: an attractive target for anticancer therapy. Med Res Rev. 2005;25:261–309.
    1. Segura-Pacheco B, Trejo-Becerril C, Pérez-Cárdenas E, Taja-Chayeb L, Mariscal I, et al. Reactivation of tumor suppressor genes by the cardiovascular drugs hydralazine and procainamide and their potential use in cancer therapy. Clin Cancer Res. 2003;9:1596–1603.
    1. Angeles E, Vázquez-Valadez VH, Vázquez-Valadez O, Velázquez-Sánchez AM, Ramírez A, et al. Computational studies of 1-hydrazinophtalazine (hydralazine) as antineoplastic agent. Docking studies on methyltransferase. Lett Drug Des Discov. 2005;2:282–286.
    1. Zambrano P, Segura-Pacheco B, Pérez-Cárdenas E, Cetina L, Revilla-Vázquez A, et al. A phase I study of hydralazine to demethylate and reactivate the expression of tumor suppressor genes. BMC Cancer. 2005;5:44.
    1. Samlowski WE, Leachman SA, Wade M, Cassidy P, Porter-Gill P, et al. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J Clin Oncol. 2005;23:3897–3905.
    1. Tang B, Jiang J. Study of the CpG methylation status of ER alpha gene in estrogen receptor alpha-negative breast cancer cell lines and the role of hydralazine demethylation. Zhonghua Bing Li Xue Za Zhi. 2005;34:283–287.
    1. Gurvich N, Tsygankova OM, Meinkoth JL, Klein PS. Histone deacetylase is a target of valproic acid-mediated cellular differentiation. Cancer Res. 2004;64:1079–1086.
    1. Shi H, Wei SH, Leu YW, Rahmatpanah F, Liu JC, et al. Triple analysis of the cancer epigenome: an integrated microarray system for assessing gene expression, DNA methylation, and histone acetylation. Cancer Res. 2003;63:2164–2171.
    1. Suzuki H, Gabrielson E, Chen W, Anbazhagan R, van Engeland M, Weijenberg MP, Herman JG, Baylin SB. A genomic screen for genes upregulated by demethylation and histone deacetylase inhibition in human colorectal cancer. Nat Genet. 2002;31:141–149.
    1. Modlich O, Prisack HB, Munnes M, Audretsch W, Bojar H. Immediate gene expression changes after the first course of neoadjuvant chemotherapy in patients with primary breast cancer disease. Clin Cancer Res. 2004;10:6418–6431.
    1. Buchholz TA, Stivers DN, Stec J, Ayers M, Clark E, et al. Global gene expression changes during neoadjuvant chemotherapy for human breast cancer. Cancer J. 2002;8:461–468.
    1. Blagosklonny MV, Trostel S, Kayastha G, Demidenko ZN, Vassilev LT, et al. Depletion of mutant p53 and cytotoxicity of histone deacetylase inhibitors. Cancer Res. 2005;65:7386–7392.
    1. Kitazono M, Bates S, Fok P, Fojo T, Blagosklonny MV. The histone deacetylase inhibitor FR901228 (desipeptide) restores expression and function of pseudo-null p53. Cancer Biol Ther. 2002;2:665–668.
    1. Selivanova G, Kawasaki T, Ryabchenko L, Wiman KG. Reactivation of mutant p53: a new strategy for cancer therapy. Semin Cancer Biol. 1998;8:369–378.
    1. Cristofanilli M, Krishnamurthy S, Guerra L, Broglio K, Arun B, et al. A nonreplicating adenoviral vector that contains the wild-type p53 transgene combined with chemotherapy for primary breast cancer: safety, efficacy, and biologic activity of a novel gene-therapy approach. Cancer. 2006;107:935–944.
    1. Karpf AR, Peterson PW, Rawlins JT, Dalley BK, Yang Q, et al. Inhibition of DNA methyltransferase stimulates the expression of signal transducer and activator of transcription 1, 2, and 3 genes in colon tumor cells. Proc Natl Acad Sc U S A. 1999;96:14007–14012.
    1. Tomasi TB, Magner WJ, Khan AN. Epigenetic regulation of immune escape genes in cancer. Cancer Immunol Immunother. 2006;55:1159–1184.
    1. Gollob JA, Sciambi CJ, Peterson BL, Richmond T, Thoreson M, et al. Phase I trial of sequential low-dose 5-aza-2′-deoxycytidine plus high-dose intravenous bolus interleukin-2 in patients with melanoma or renal cell carcinoma. Clin Cancer Res. 2006;12:4619–4627.
    1. Hartsough MT, Clare SE, Mair M, Elkahloun AG, Sgroi D, et al. Elevation of breast carcinoma Nm23-H1 metastasis suppressor gene expression and reduced motility by DNA methylation inhibition. Cancer Res. 2001;61:2320–2327.
    1. Smeitink J, van den Heuvel L, DiMauro S. The genetics and pathology of oxidative phosphorylation. Nat Rev Genet. 2001;2:342–352.
    1. Mamelak AJ, Kowalski J, Murphy K, Yadava N, Zahurak M, et al. Downregulation of NDUFA1 and other oxidative phosphorylation-related genes is a consistent feature of basal cell carcinoma. Exp Dermatol. 2005;14:336–348.
    1. Huang G, Lu H, Hao A, Ng DC, Ponniah S, et al. GRIM-19, a cell death regulatory protein, is essential for assembly and function of mitochondrial complex I. Mol Cell Biol. 2004;24:8447–8456.
    1. Wong TW, Yu HY, Kong SK, Fung KP, Kwok TT. The decrease of mitochondrial NADH dehydrogenease and drug induced apoptosis in doxorubicin resistant A431 cells. Life Sci. 2000;67:1111–1118.
    1. Frank NY, Margaryan A, Huang Y, Schatton T, Waaga-Gasser AM, et al. ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. Cancer Res. 2005;65:4320–4333.

Source: PubMed

3
Abonneren