Hypomethylating agents synergize with irinotecan to improve response to chemotherapy in colorectal cancer cells

Anup Sharma, Rajita Vatapalli, Eihab Abdelfatah, K Wyatt McMahon, Zachary Kerner, Angela A Guzzetta, Jasvinder Singh, Cynthia Zahnow, Stephen B Baylin, Sashidhar Yerram, Yue Hu, Nilofer Azad, Nita Ahuja, Anup Sharma, Rajita Vatapalli, Eihab Abdelfatah, K Wyatt McMahon, Zachary Kerner, Angela A Guzzetta, Jasvinder Singh, Cynthia Zahnow, Stephen B Baylin, Sashidhar Yerram, Yue Hu, Nilofer Azad, Nita Ahuja

Abstract

Colorectal cancer (CRC) is the second leading cause of cancer death in the United States. In the metastatic setting, the majority of patients respond to initial therapies but eventually develop resistance and progress. In this study, we test the hypothesis that priming with epigenetic therapy sensitizes CRC cell lines, which were previously resistant to subsequent chemotherapeutic agents. When multiple CRC cell lines are first exposed to 500 nM of the DNA demethylating agent, 5-aza-cytidine (AZA) in-vitro, and the cells then established as in-vivo xenografts in untreated NOD-SCID mice; there is an enhanced response to cytotoxic chemotherapy with agents commonly used in CRC treatment. For irinotecan (IRI), growth diminished by 16-62 fold as assessed, by both proliferation (IC50) and anchorage independent cell growth soft agar assays. Treatment of resistant HCT116 cell line along with in-vivo, for CRC line xenografts, AZA plus IRI again exhibits this synergistic response with significant improvement in survival and tumor regression in the mice. Genome-wide expression correlates changes in pathways for cell adhesion and DNA repair with the above responses. A Phase 1/2 clinical trial testing this concept is already underway testing the clinical efficacy of this concept in IRI resistant, metastatic CRC (NCT01896856).

Conflict of interest statement

Competing Interests: There is a competing interest: Dr. Ahuja receives research grant funding from Astex Inc. and the Van Andel Research Institute. She has licensed methylation biomarkers to Cepheid. Dr. Ahuja also has served as consultant to Johnson and Johnson. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Low dose HMA causes global…
Fig 1. Low dose HMA causes global demethylation in CRC cell lines.
(A) Colon cancer cell lines (HCT116, SW480, Caco-2, RKO, SW620, Colo205, Colo320) showed very little or no cytotoxicity. Percent viability after 72 hrs treatment with 500 nM AZA, relative to mock control. Bars represent the mean of 3 replicates ±SD; (B & C) Colon Cancer cell lines saw basal or low expression of DNMT1 protein. Upper and the lower panel is a representative blot for DNMT1 and β-Actin as a housekeeper. (D) Protein expression of DNMT1 in colon cancer cells (Caco-2-, HCT116 and SW480) changes after treatment with a various concentration of AZA. Cells were treated with AZA at the indicated concentrations for 72hr and then subjected to immunoblot analysis using anti-DNMT1 antibody. Actin served as a loading control. B, C, and D are representative blots of at least three independent experiments. (E) Quantitation of DNA methylation using bisulfite LINE-1 PCR and Pyrosequencing in Caco-2, SW480 and HCT116 (3-day treatment followed by 3 rest period). Representative LINE-1 quantitation presented for the cell lines. The pyrogram quantitates C for methylated, and T for unmethylated DNA was plotted as a line graph.
Fig 2. In vivo epigenetic therapy sensitize…
Fig 2. In vivo epigenetic therapy sensitize CRC cell line xenografts to decrease tumor burden.
Colorectal cancer cells were treated for 72 hours with AZA at 500nM and saline, or PBS inject Mock, were allowed to recover from the short-term cytotoxic effects of AZA for seven days before injection into NOD-SCID mice (n = 10 in each case). NOD-SCID mice were xenografted with various CRC cell line and monitored until mice showed 2000 mm3 tumor development. (A-G) Represents xenografted mice responded to therapy and (H & I) are the mice which did not respond to the treatment (S3 Fig). Mean tumor volume (±SEM) over time are plotted. Statistical significance determined by two-tailed paired t-test.
Fig 3. Epigenetic therapy sensitizes chemotherapy in…
Fig 3. Epigenetic therapy sensitizes chemotherapy in CRC cell lines.
(A-C) Log dose response curves for CRC cell lines treated (in triplicate) with IRI for 72 hours three days’ post epigenetic therapy. Individual curves represent the percentage of viable cells (±SD) for each epigenetic pretreatment condition normalized to its untreated control cells, such that the highest values for each pretreatment state represent 100%, and 0%. Data shown from representative experiments. Visualization of drug interaction is illustrated by CI-Fa plot. The CI-Fa plot represents the combination index plotted versus Fa, the fraction of affected enzyme or biological function. CI values were calculated from each Fa (i.e. various drug concentration) for CRC cell lines Caco-2, SW480, and HCT116. Average synergism (CI<1) at Fa>0.5 for all three CRC lines. The AZA and IRI chemotherapeutic doses may be significantly reduced for combinations that are synergistic at Fa>0.5 for all three cell lines. (D-F) Caco-2, SW480, and HCT116 cells were seeded on a solidified Matrigel layer six days after epigenetic therapy. Beginning the following day, cells were treated with chemotherapy for 72 hrs. The drug was then removed, and colonies were permitted to grow 2–4 additional days. Representative Caco-2, SW480 and HCT116 colonies following treatment with 80 nM IRI. Three independent experiments (total nine replicates. (G) HCT116 colony forming assay examined for prolonged exposure to AZA as a pretreatment to IRI on HCT116 cells. HCT116 cells were pretreated with AZA for 72 hours, followed by five days’ rest and the schedule was repeated three more times before IRI treatment in soft agar assay. This modification increased colony growth inhibition in HCT116 significantly more than IRI alone, which were previously resistant.
Fig 4. Epigenetic therapy in vivo sensitize…
Fig 4. Epigenetic therapy in vivo sensitize SW480 xenografts to subsequent chemotherapy.
NOD-SCID mice bearing SW480 and HCT116 xenografts were treated with 0.5 mg/kg AZA (sc, qd × 5) and 10 mg/kg IRI (i.p., day 5), or vehicle, until the end of experiment cycles. (A&D) Mean tumor volume (±SEM) over time. Statistical significance determined by two-tailed paired t-test. (B&E) Survival analysis showed that the median survival for SW480 and HCT116 tumor-bearing mice was significantly reduced in AZA and IRI treated compared to mice which were untreated tumors. (C&F) Indicates weight changes over the course of treatment. Mice were either treated with AZA alone, AZA + IRI or IRI alone or saline vehicle. Mice were treated until the end of the experiment. N = 10 mice in each.
Fig 5. Relative expression of Caco-2 and…
Fig 5. Relative expression of Caco-2 and SW480 colon cancer cell lines.
Heatmap showing relative expression (Mock is shown as reference) of the Sensitization Genes in the IRI and AZA+IRI-treated samples.

References

    1. Flis S, Gnyszka A, Flis K. DNA methyltransferase inhibitors improve the effect of chemotherapeutic agents in SW48 and HT-29 colorectal cancer cells. PloS one. 2014;9(3):e92305 Epub 2014/03/29. 10.1371/journal.pone.0092305
    1. Labianca R, Beretta GD, Kildani B, Milesi L, Merlin F, Mosconi S, et al. Colon cancer. Critical reviews in oncology/hematology. 2010;74(2):106–33. Epub 2010/02/09. 10.1016/j.critrevonc.2010.01.010
    1. Sundermeyer ML, Meropol NJ, Rogatko A, Wang H, Cohen SJ. Changing patterns of bone and brain metastases in patients with colorectal cancer. Clinical colorectal cancer. 2005;5(2):108–13. Epub 2005/08/16.
    1. Grothey A, Van Cutsem E, Sobrero A, Siena S, Falcone A, Ychou M, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet (London, England). 2013;381(9863):303–12. Epub 2012/11/28.
    1. Lenz HJ, Stintzing S, Loupakis F. TAS-102, a novel antitumor agent: a review of the mechanism of action. Cancer treatment reviews. 2015;41(9):777–83. Epub 2015/10/03. 10.1016/j.ctrv.2015.06.001
    1. Ahuja N, Sharma AR, Baylin SB. Epigenetic Therapeutics: A New Weapon in the War Against Cancer. Annual review of medicine. 2016;67:73–89. Epub 2016/01/16. 10.1146/annurev-med-111314-035900
    1. Jones PA, Issa JP, Baylin S. Targeting the cancer epigenome for therapy. Nature reviews Genetics. 2016;17(10):630–41. Epub 2016/09/16. 10.1038/nrg.2016.93
    1. Baylin SB, Ohm JE. Epigenetic gene silencing in cancer—a mechanism for early oncogenic pathway addiction? Nature reviews Cancer. 2006;6(2):107–16. Epub 2006/02/24. 10.1038/nrc1799
    1. Crea F, Giovannetti E, Cortesi F, Mey V, Nannizzi S, Gallegos Ruiz MI, et al. Epigenetic mechanisms of irinotecan sensitivity in colorectal cancer cell lines. Mol Cancer Ther. 2009;8:1964–73. 10.1158/1535-7163.MCT-09-0027
    1. Abdelfatah E, Kerner Z, Nanda N, Ahuja N. Epigenetic therapy in gastrointestinal cancer: the right combination. Therapeutic advances in gastroenterology. 2016;9(4):560–79. Epub 2016/07/02. 10.1177/1756283X16644247
    1. Qin L, Dong Z, Zhang JT. Reversible epigenetic regulation of 14-3-3sigma expression in acquired gemcitabine resistance by uhrf1 and DNA methyltransferase 1. Molecular pharmacology. 2014;86(5):561–9. Epub 2014/09/06. 10.1124/mol.114.092544
    1. Kaminskas E, Farrell AT, Wang YC, Sridhara R, Pazdur R. FDA drug approval summary: azacitidine (5-azacytidine, Vidaza) for injectable suspension. The oncologist. 2005;10(3):176–82. Epub 2005/03/29. 10.1634/theoncologist.10-3-176
    1. Silverman LR, Demakos EP, Peterson BL, Kornblith AB, Holland JC, Odchimar-Reissig R, et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2002;20(10):2429–40. Epub 2002/05/16.
    1. Tsai HC, Li H, Van Neste L, Cai Y, Robert C, Rassool FV, et al. Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer cell. 2012;21(3):430–46. Epub 2012/03/24. 10.1016/j.ccr.2011.12.029
    1. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141:69–80. 10.1016/j.cell.2010.02.027
    1. Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. International journal of cancer. 2008;123(1):8–13. Epub 2008/04/22. 10.1002/ijc.23607
    1. Ahuja N, Easwaran H, Baylin SB. Harnessing the potential of epigenetic therapy to target solid tumors. The Journal of clinical investigation. 2014;124(1):56–63. Epub 2014/01/03. 10.1172/JCI69736
    1. Valdespino V, Valdespino PM. Potential of epigenetic therapies in the management of solid tumors. Cancer management and research. 2015;7:241–51. Epub 2015/09/09. 10.2147/CMAR.S70358
    1. Azad Nilofer Saba E-K AB, Mahoney Michelle R., Adkins Douglas, Flynn Patrick J., Bahary Nathan, Kim George P., Pitot Henry Clement, Erlichman Charles, Donehower Ross C., Herman James Gordon, Baylin Stephen, Ahuja Nita. A phase II study of combination epigenetic therapy in metastatic colorectal cancer (mCRC): A phase II consortium (P2C)/Stand Up 2 Cancer (SU2C) study. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2013;(21).
    1. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA Approval Summary: Vorinostat for Treatment of Advanced Primary Cutaneous T-Cell Lymphoma. The oncologist. 2007;12(10):1247–52. 10.1634/theoncologist.12-10-1247
    1. Kantarjian H, Issa JP, Rosenfeld CS, Bennett JM, Albitar M, DiPersio J, et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer. 2006;106(8):1794–803. Epub 2006/03/15. 10.1002/cncr.21792
    1. Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, Coleman B, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer discovery. 2011;1(7):598–607. Epub 2012/05/16. 10.1158/-11-0214
    1. Matei D, Fang F, Shen C, Schilder J, Arnold A, Zeng Y, et al. Epigenetic resensitization to platinum in ovarian cancer. Cancer research. 2012;72(9):2197–205. Epub 2012/05/03. 10.1158/0008-5472.CAN-11-3909
    1. Azad N, Zahnow CA, Rudin CM, Baylin SB. The future of epigenetic therapy in solid tumours—lessons from the past. Nat Rev Clin Oncol. 2013;10:256–66. 10.1038/nrclinonc.2013.42
    1. Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer research. 2010;70(2):440–6. Epub 2010/01/14. 10.1158/0008-5472.CAN-09-1947
    1. Issa J-PJ, Roboz G, Rizzieri D, Jabbour E, Stock W, O'Connell C, et al. Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicentre, randomised, dose-escalation phase 1 study. The Lancet Oncology. 2015;16(9):1099–110. 10.1016/S1470-2045(15)00038-8.
    1. Li H, Chiappinelli KB, Guzzetta AA, Easwaran H, Yen RW, Vatapalli R, et al. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget. 2014;5(3):587–98. Epub 2014/03/04. 10.18632/oncotarget.1782
    1. 2016.
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic acids research. 2015;43(7):e47 Epub 2015/01/22. 10.1093/nar/gkv007
    1. Ritchie ME, Silver J, Oshlack A, Holmes M, Diyagama D, Holloway A, et al. A comparison of background correction methods for two-colour microarrays. Bioinformatics (Oxford, England). 2007;23(20):2700–7. Epub 2007/08/28.
    1. Smyth GK, Speed T. Normalization of cDNA microarray data. Methods. 2003;31(4):265–73. 10.1016/S1046-2023(03)00155-5.
    1. Smyth GK. limma: Linear Models for Microarray Data In: Gentleman R, Carey VJ, Huber W, Irizarry RA, Dudoit S, editors. Bioinformatics and Computational Biology Solutions Using R and Bioconductor. New York, NY: Springer New York; 2005. p. 397–420.
    1. Smyth GK, Altman NS. Separate-channel analysis of two-channel microarrays: recovering inter-spot information. BMC bioinformatics. 2013;14:165 Epub 2013/05/28. 10.1186/1471-2105-14-165
    1. Smyth GK, Michaud J, Scott HS. Use of within-array replicate spots for assessing differential expression in microarray experiments. Bioinformatics (Oxford, England). 2005;21(9):2067–75. Epub 2005/01/20.
    1. Issa JP. DNA methylation as a therapeutic target in cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2007;13(6):1634–7. Epub 2007/03/17.
    1. Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, Coleman B, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 2011;1:598–607. 10.1158/-11-0214
    1. Hatziapostolou M, Iliopoulos D. Epigenetic aberrations during oncogenesis. Cellular and molecular life sciences: CMLS. 2011;68(10):1681–702. Epub 2011/01/21. 10.1007/s00018-010-0624-z
    1. Hong X, Michalski CW, Kong B, Zhang W, Raggi MC, Sauliunaite D, et al. ALCAM is associated with chemoresistance and tumor cell adhesion in pancreatic cancer. Journal of surgical oncology. 2010;101(7):564–9. Epub 2010/05/13. 10.1002/jso.21538
    1. Liu T, Li R, Zhao H, Deng J, Long Y, Shuai MT, et al. eIF4E promotes tumorigenesis and modulates chemosensitivity to cisplatin in esophageal squamous cell carcinoma. Oncotarget. 2016. Epub 2016/09/03.
    1. Vishnubalaji R, Yue S, Alfayez M, Kassem M, Liu FF, Aldahmash A, et al. Bone morphogenetic protein 2 (BMP2) induces growth suppression and enhances chemosensitivity of human colon cancer cells. Cancer cell international. 2016;16:77 Epub 2016/10/07. 10.1186/s12935-016-0355-9
    1. Xu Y, Villalona-Calero MA. Irinotecan: mechanisms of tumor resistance and novel strategies for modulating its activity. Annals of oncology: official journal of the European Society for Medical Oncology. 2002;13(12):1841–51. Epub 2002/11/28.
    1. Valerie Lee JW, Anthony El Khoueiry, Henk Verheul, Elske Gootjes, Anup Sharma, Zachary Kerner, Peter Jones, Stephen Baylin, Ellen Lilly, Nita Ahuja, Thomas Brown, Nilofer Azad. A phase I study of guadecitabine (GUA) combined with irinotecan (IRI) in previously treated metastatic colorectal cancer (mCRC) patients In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research;. 2016 Aprl:16–20; New Orleans, LA. Philadelphia (PA): AACR; 2016; Abstract nr CT017.

Source: PubMed

3
구독하다