ARTICLE IN PRESS Cancer Letters ■■ (2014) ■■–■■
Contents lists available at ScienceDirect
Cancer Letters j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / c a n l e t
Mini-review
Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors Jing Nie a,1, Lin Liu b,1, Xiang Li a, Weidong Han a,* a b
Department of Molecular Biology, Institute of Basic Medical Science, PLA General Hospital, Beijing 100853, China Department of General Surgery, PLA General Hospital, Beijing 100853, China
A R T I C L E
I N F O
Article history: Received 25 June 2014 Received in revised form 5 August 2014 Accepted 6 August 2014 Keywords: Decitabine Demethylating drug Tumor therapy Drug sensitivity
A B S T R A C T
Epigenetic alterations are strongly associated with cancer development and drug resistance. The use of the DNA methylation inhibitor decitabine (Dacogen®) has been approved in the treatment of hematological malignancies, and its clinical effects on solid tumors have gained attention. Here, we present a review of the molecular regulation mechanisms, clinical experiences and biological evaluation for novel decitabinebased therapies in solid tumors. We also discuss the following questions: What is the best administration schedule of decitabine in solid tumors? Is there tumor type specificity for decitabine-based epigenetic therapy? What are the biological function and mechanism of decitabine in suppressing tumor development? Is there a correlation between DNA demethylation and clinical response? Importantly, low-dose decitabine and combined therapy show significant improvement in solid tumor treatment. However, the correlation studies are preliminary, and key biomarkers for prognosis need further investigation. © 2014 Elsevier Ireland Ltd. All rights reserved.
Introduction Surgery, radiotherapy and chemotherapy are the mainstays of tumor therapy. Recently, cancer immunotherapy gains worldwide attention and is regarded as one of the most promising cancer treatments in 21st century. Besides targeting the checkpoint inhibitors, such as CTLA-1 and PD-1, modification of other immune system components would also be prospective. DNA methylation is an epigenetic event that regulates chromatin compaction and repression of gene expression. In cancer cells, a variety of genes are abnormally silenced by DNA methylation, including tumor suppressors, and genes controlling the immune response and drug sensitivity [1,2]. The inhibition of DNA methylation with cytidine analogues such as decitabine (5-aza-2′deoxycytidine, Dacogen®, DAC for short) reactivates the expression of genes silenced by hypermethylation. DAC incorporates into replicating DNA and forms irreversible covalent bonds with the active sites of DNA methyltransferase [3]. Various animal model and cell line studies indicated that DAC induced the expression of genes that control cell apoptosis, cell cycle arrest, and cancer testis antigens (CTA), MHC molecules and costimulation molecules by DNA hypomethylation. As a result, DAC enhanced the anti-tumor immune response and inhibited tumor development [4–6]. Clinically, previous observations indicated that
* Corresponding author. Tel.: +86 10 66937463; fax: +86 10 66937516. E-mail address:
[email protected] (W. Han). 1 These authors contributed equally to this study.
higher doses of DAC induce cytotoxicity, while low-dose, prolonged infusion with DAC correlates with clinical response in hematological disorders [7]. DAC has been used as an approved therapy for the treatment of hematological malignancies such as, myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). The data indicate that approximately 50% of MDS patients demonstrate a clinical response after DAC treatment and that with myelosuppression is the major adverse effect [8]. Over the past two decades, besides treatment of hematological diseases, many clinical trials have focused on utilizing DAC in solid tumors as a lone agent or as part of a combination chemotherapy approach. The clinical effectiveness of the DAC-based therapy in patients with solid tumors has recently been demonstrated. The remarkable response and mild toxicity caused by DAC treatment in certain patients highlighted the excellent prospective use in cancer treatment. This review focuses on the biological mechanism and therapeutic effect of DAC treatment in solid tumors. We summarize the potential marker genes, methylation status, administration regimen and clinical response for this epigenetic therapy. Additionally, we discuss the correlation between gene demethylation and clinical response and provide the potential early targets for DAC-based epigenetic therapy. The molecular regulation mechanisms of DAC in tumor treatment Increase the sensitivity of chemotherapeutic drugs Surgery, radiotherapy, chemotherapy, and targeted therapy are widely used as first-line cancer regimens. However, overall survival is still limited by drug resistance. Numerous DNA methylation
http://dx.doi.org/10.1016/j.canlet.2014.08.010 0304-3835/© 2014 Elsevier Ireland Ltd. All rights reserved.
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
ARTICLE IN PRESS 2
J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
alterations in the cancer methylome are related to drug resistance. For example, there are genes involved in DNA repair, apoptosis, cell cycle control and drug metabolism [9–11]. Pretreatment with low-dose DAC could function as a potential sensitizer of chemotherapeutic drugs via the epigenetic modulation of aberrant DNA methylation in cancers. Hypermethylation and inactivation of the DNA mismatch repair gene MLH1 were observed in 25–35% of ovarian cancer patients following platinum-based chemotherapy. This result was related to a poor survival rate in ovarian cancer patients [12]. In vitro restoration of MLH1 could confer sensitivity of DNA damaging drugs and DAC treatment resensitized tumor cells to chemotherapeutic agents in vivo [13]. The methylation-mediated silence of pro-apoptotic genes such as caspase-8 occurs in multiple solid tumors and could be a key mechanism of resistance to chemotherapy. A study in neuroblastoma patients showed that caspase-8 promoter demethylation and gene expression were observed in 2/7 bone marrow samples following DAC treatment [14]. Membrane transporters play an important role in the uptake of the chemotherapeutic agents. Therefore, the reduced expression of these genes may be another critical factor causing drug resistance. For example, the ATP-binding cassette (ABC) drug transporters control tumor multidrug resistance [15,16]. Strikingly, administration of DAC dramatically enhances the expression of transporter CTR1, ABCB1 and ABCG2 in solid tumors [17]. Zeller et al. detected 9 genes that acquired methylation in ovarian tumors at relapse after chemotherapy or in chemoresistant cell lines. These genes included the following: ARHGD1B, ARMCX2, COL1A, FLNA, FLNC, MEST, MLH1, NTS and PSMB9 [6]. Overcome the resistance of molecular targeted therapy The anti-epidermal growth factor receptor (EGFR) therapy is a common regimen for patients with wild-type (wt) KRAS metastatic colorectal cancer (mCRC). However, a substantial portion of these patients are also resistant to treatment and most responders will become resistant to anti-EGFR therapy probably by EGFR hypermethylation and silencing. Scartozzi et al. observed a higher response rate to anti-EGFR therapy in the absence of EGFR promoter methylation [18]. DAC induces EGFR expression and restores the sensitivity to EGFR inhibitors. A recent phase I/II clinical trial of DAC in combination with panitumumab in patients with wt KRAS mCRC was performed. Of note, 2 of the 20 patients tested had a partial response, and 10 patients had stable disease (3 patients longer than 16 weeks). Furthermore, the time to progression (TTP) was longer in 7 patients when compared to their previous TTP on ceruximab [19]. It is possible that the combination regimen resensitized patients to the anti-EGFR treatment. However, the roles and mechanism of DAC in overcoming resistance of mAbs against EGFR in CRC cells need further elucidation. Induce cell reprogramming The cancer stem cell theory indicates that although chemotherapy kills most cells in a tumor, the tumor stem cells survive and contribute to drug resistance. This hypothesis is based on the evidence suggesting the tumor initiating cell may be innately resistant to various therapies. As a result, the cancer stem cells were able to survive cytotoxic or targeted therapies and caused tumor development and relapse. Many studies have observed a limited clinical response using only a demethylating drug. However, improved effects were found when followed by other subsequent treatments such as chemotherapy, immunotherapy, and targeted therapy. This result suggests there is a “priming” ability of the epigenetic agent [20]. Transient low doses of DNA demethylating drugs exert durable antitumor effects on
hematological and epithelial tumor cells. The demethylating drugs might inhibit the tumor initiating or cancer stem-like and selfrenewing cells, and provide a memory for the reprogramminglike status [21]. A recent study showed that low-dose 5-azatidine elicited a DNMT3B-dependent activation of p53 target genes and also caused both DNA damage and DNA demethylation. The result was the downregulation of the pluripotency genes including NANOG, SOX2, GDF3 and Myc target genes in NT2/D1 cells [22]. Thus, epigenetic therapy combined with chemotherapy would be a potential new strategy for tumor treatment and could overcome drug resistance by killing both cancer cells and cancer stem cells.
Elicit immune response Demethylating drugs regulate multiple pathways for tumorigenesis such as cell cycle control, DNA replication, mRNA splicing and translation. It was recently demonstrated that there was a significant enrichment for immunomodulatory pathways in tumor cell lines after treatment with 5-azacitidine. The pathways included interferon signaling, antigen processing and presentation, cytokines/ chemokines, and the upregulation of cancer testis antigens [23]. An analysis of gene expression profiles of PBMCs or biopsies during DAC treatment in solid tumor patients confirmed the regulation of immune activity. George et al. detected that genes enriched on day 7 post-decitabine play roles in synaptogenesis, B cell activation, antigen processing and peptide presentation, and inhibition of cell growth. The day 28 samples also included genes with biogenic amine catabolism, regulation of interferon gamma, calcium mediated signaling and regulation of cdc42 GTPase activity [14]. Several large studies have demonstrated that 5-azacytidine activates antitumor immunity in different ways. In addition to regulating cancer testis antigens such as MAGE family [24], the demethylating drug up-regulated other tumor immune stimulating molecules, including MHC-I antigens, beta-2-microglobulin (B2M), CD58, TAP1, and the immune-proteasome subunits PSMB9 and PSMB8 [25–27], and the transcription factors IRF7 and IRF5 [24,28]. Furthermore, deletion of DNMT1 and subsequent DNA methylation influenced the proliferative potential of antigen-specific CD8+ T cells [29], and 5-azacytidine treatment (5 μM or 20 μM) promoted an inhibitory T cell phenotype and impaired immune mediated anti-leukemic activity [30]. Hence, methylation modification and silencing of specific genes may be critical during T cell maturation and commitment. Treatment with 5-azacitidine may sensitize tumors to immune checkpoint therapy by targeting PD-L1/ PD-1 interaction. Thus, the combination of epigenetic therapy and immunotherapy augments the clinical response in tumor patients [27,31]. These findings indicated the possibility that the demethylating drugs might play an immune stimulatory role in cancer therapy by sensitizing the patients to immune responses. The potential molecular mechanisms of DAC function in cancer therapy are summarized in Fig. 1, while the detailed regulation pathways and molecular networks need further investigation (Fig. 1). In addition, identifying the crosstalk between immunotherapy and chemotherapy will be promising.
Clinical experiences of DAC in solid tumors In the past 10 years, clinical trials treating solid tumors with DAC have improved. We collected the recent clinical studies in the NCBI database in May 2014 that administered DAC to patients with solid tumors. There are 13 phase I or II clinical trials (305 patients, 18 tumor types) using DAC agent at different doses examined in this review. The studies are shown in Table 1.
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
ARTICLE IN PRESS J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
Increase chemotherapeutic drugs sensitivity Apoptosis
DNA damage
3
Sensitize anti-EGFR therapy
Drug uptake
ABC transporters, CTR1
EGFR
Caspase-8 Tumor suppressor (p16, p15, BRCA1, RASSF1A)
MLH1
pluripotency genes (Ocr4, Nanog, Myc)
Transcription factor (IRF7, IF5) Proteasome (PSMB9, PSMB8) Cancer testis antigen (MAGE-1, MAGE-3, NY-ESO-1)
Interferon signaling MHC-I co-stimulation molecule Inhibitory molecule
Elicit immune response
Cell reprogramming
Fig. 1. The molecular regulation mechanisms of DAC in anti-tumor treatment. The demethylating drug of DAC shows significant clinical response in solid tumors probably through several mechanisms. DAC treatment could increase the sensitivity of tumor cells to the chemotherapeutic drugs via the upregulation of genes related to cell apoptosis, DNA damage, and drug uptake. DAC is able to sensitize the anti-EGFR therapy through the induction of EGFR. Moreover, DAC treatment stimulates the immune responses, owing to the regulation of cancer testis antigen, MHC-I, co-stimulation and inhibitory molecules, as well as some immune-related transcription factors or other regulators. In addition, DAC treatment may influence cell reprogramming by the modification of the pluripotency genes.
Solid tumor pooled studies Several trials were performed with different schedules of DACbased therapy to determine the tolerable administration dose and time in solid tumor patients. A phase I study of 19 solid tumor patients used DAC 20–40 mg/m 2 /day for a 72-hour continuous intravenous infusion on days 1–3 of a 28-day cycle. The authors reported that the tolerated dose of DAC in these patients was 30 mg/ m2/day during the 72-hour continuous infusion [32]. In another clinical trial, Samlowski et al. applied lower doses of DAC (2 mg/ m2/d) via long-term 7-day continuous infusion to patients with refractory solid tumors. They found that the level of toxicity was suitable for further DNA methylation evaluation. However, patients with extensive radiotherapy to marrow-containing bones or extensive nucleoside analog pre-treatment should be excluded, due to the possible adverse effects [33]. DAC showed a clinical effect in hematological diseases, but the combination with other therapies would be more effective in solid tumors. Appleton reported that combination administration of DAC (90 mg/m2/day) as a 6-hour infusion on day 1 followed by the chemotherapeutic agent carboplatin on day 8 in a 28-day cycle showed a favorable response rate [34]. Recently, our group proposed the regimen of extreme low-dose DAC (7 mg/m2/day) as a half-hour infusion per day for 5 consecutive days. This therapy could also be combined with chemotherapy or adoptive immunotherapy (for example, cytokine-induced killer cells (CIKs)) in solid tumor patients. Strikingly, clinical benefits were found in 60% of patients. Moreover, the median progression free survival (PFS) was prolonged compared with the PFS of previous treatments. Furthermore, we observed a significant correlation between the PFS of previous treatment and clinical response [35]. A phase I trial used DAC in combination with the HDAC inhibitor vorinostat in patients with advanced solid tumors. Stathis et al. compared the sequential schedule (DAC 10 mg/m2/day for 5-day infusion, combined with vorinostat 200 mg 3 times a day on days 6–12) and a concurrent schedule (vorinostat 200 mg 2 times a day on days 3–9). The authors found
that the sequential schedule were easier to deliver and was more tolerable [36]. Stewart observed no significant correlation between the clinical responses and DAC doses or tumor characteristics when using different doses of DAC in distinct patient cohorts [17]. In pediatric patients with neuroblastoma and other solid tumors, the maximumtolerated dose (MTD) of DAC was 5 mg/m2/day for 7 days when administered in combination with doxorubicin and cyclophosphamide. The patients showed significant myelosuppression as the primary toxicity. There were only limited clinical responses observed in this study because the doses of DAC needed to produce clinically relevant biologic effects were not well tolerated with this combination in children. Ovarian cancer The first-line chemotherapy regimen for ovarian cancer patients generally consists of a platinum compound (cisplatin or carboplatin) and a taxane, while many patients will relapse and acquire resistance to the current treatment and resistance is a key limiting factor in patient survival [37]. Numerous preclinical studies have suggested that the development of epithelial ovarian cancer is correlated with the accumulation of aberrant DNA methylation. Thus, hypomethylating agents can reverse platinum resistance in ovarian cancer cells [38]. Several phase I/II clinical trials used lowdose DAC combined with carboplatin in platinum-resistant ovarian cancer patients. The Nephew group has observed DAC tolerability at a dose of 10 mg/m2 daily for 5 days followed by carboplatin administration on day 8 in ovarian cancer patients [39]. Using this regimen, 35% of 17 patients showed clinical response with progression-free survival (PFS) at 10.2 months, and 9 patients (53%) had PFS at 6 months [40]. Colorectal cancer A clinical trial was conducted in metastatic colorectal cancer (mCRC) patients with wild type (wt)-KRAS. The patients were treated
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
4
Regimen
Total doses (mg)
Phase
Patients number
Results
Response plus stable disease rate (%)
Year
References
Advanced cervical cancer
DAC 50 mg/m2/day followed by cisplatin 30 g/m2 for 3-day infusion (21-day cycle) DAC 20–40 mg/m2/day 3-day continuous infusion (28-day cycle) DAC 2 mg/m2/day 7-day continuous infusion (28-day cycle) DAC 20–30 mg/m2/day 3-day continuous infusion (35-day cycle) DAC 90 mg/m2 on day 1 6-hour infusion followed by carboplatin on day 8 (28-day cycle) DAC 2.5–20 mg/m2/day 1-hour infusion for 5 or 10 days DAC (5–10 mg/m2) 1-hour infusion for 7 days, followed by doxorubicin (45 mg/m2) and cyclophosphamide (1 g/m2) on day 7 DAC (10 or 20 mg/m2) for 5 days and carboplatin on day 8 (28-day cycle) DAC 10 mg/m2/day on days 1–5 and vorinostat 200 mg twice a day on days 6–12 (28-day cycle) DAC 10 mg/m2 for 5 days and carboplatin on day 8 (28-day cycle) DAC 45 mg/m2 on days 1 and 15 and anti-EGFR 6 mg/kg on days 8 and 22 (28-day cycle) Escalating doses of DAC (5–15 mg/m2) for 10 days in combination with VPA (10–20 mg/kg/day) on days 5–21 (28day cycle) DAC 7 mg/m2 for 5 days or combined with chemotherapy or CIKs
150
II
25
PR:8/21 SD:5/21 PD:8/21
62
2002
[48]
60–120
I
19
PD:14/14
0
2003
[32]
14
I
10
SD:2/9 PD:7/9
22
2005
[33]
60–90
I
35
SD:13/28 PD:15/28
46
2006
[42]
90
I
35
PR:1/10 SD:3/10 PD:6/10
40
2007
[34]
25–100
I
31
PR:1/28 SD:17/28
64
2009
[17]
35–70
I
21
SD:7/21 PD:14/21
33
2010
[14]
50–100
I
10
CR:1/10 SD:6/10 PD:3/10
70
2010
50
II
43
SD:11/38 PD:27/38
29
2011
[36]
50
II
17
CR:1/17 PR:5/17 SD:6/17 PD:5/17
70
2012
[40]
90
I/II
20
PR:2/20 SD:10/20 PD:8/20
60
2013
[19]
50–100
I
SD:1/8 PD:7/8
13
2013
[27]
35
I/II
PR:3/31 SD:14/31 PD:14/31
55
2014
[35]
Metastatic solid tumors Solid tumors (ovarian, renal, breast, colon) Solid tumors (lung, esophagus and pleura) Solid tumors (colon, breast, ovary, melanoma, sarcoma, gall bladder and pleural mesothelioma) Solid tumors and lymphomas Children refractory solid tumors (neuroblastoma, rhabdomyosarcoma, osteosarcoma) Platinum-resistant ovarian cancer Advanced solid tumors and nonHodgkin’s lymphomas Platinum-resistant ovarian cancer wt KRAS metastatic colorectal cancer
Non-small cell lung cancer
Solid tumors (gastric, colorectal, liver, esophageal, lung, cervical, ovarian and pancreatic)
8
31
ARTICLE IN PRESS
Tumor types
J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
Table 1 Clinical trials of DAC-based therapy on solid tumors.
ARTICLE IN PRESS J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
with DAC at 45 mg/m2 on days 1 and 15, and panitumumab (mAb against EGFR) 6 mg/kg on days 8 and 22 every 28 days. Excitingly, 10% patients had a partial response and 50% patients had stable disease. This result suggested that the combination of DAC and targeted therapy was well tolerated and showed clinical response in mCRC patients [19].
Lung cancer Epigenetic modifications have an important role in lung cancer tumorigenesis. Several recent clinical trials have combined DAC and chemotherapeutic drugs in patients with non-small cell lung cancer (NSCLC). The patients were treated with escalating doses of DAC (5– 15 mg/m2) for 10 days in combination with valproic acid (VPA) (10– 20 mg/kg/day) on days 5–21 of a 28-day cycle. However, the clinical response was disappointing due to the unacceptable neurological toxicity caused by VPA. There was stable disease only in one patient. Thus, an alternative combination or schedule with less toxicity should be investigated [41].
The biological evaluation in DAC-based therapy The demethylating activity of the DAC regimen was examined in vivo to evaluate its biological effects. DNA from peripheralblood mononuclear cells (PBMCs), plasma and tumor biopsies was extracted and assessed for global DNA methylation and site-specific DNA methylation, and gene expression profile.
5
Biomarkers for DAC-based therapy DAC-based therapy has exhibited favorable effects in some types of solid tumor patients. Therefore, further investigations of prognostic biomarkers for hypomethylating strategies in solid tumors are required. Many studies have focused on the detection of MAGE-1 methylation status in peripheral blood as a potential biomarker for DAC treatment. The percentage of patients who displayed significant demethylation of MAGE-1 in PBMCs during the first course of treatment ranged from 60% to 100% [2,14]. The increased expression of HbF mRNA was reported to be 88% of blood samples and 100% of bone marrow samples in neuroblastoma patients following DAC treatment [14]. Consistent with these data, HbF protein levels were enhanced by Western blotting analysis post-decitabine [34]. Following treatment with a combination of DAC and VPA, the HbF levels increased in 100% of PBMC samples from NSCLC patients [41]. Analyses of DAC-altered genes in ovarian cancer patients postdecitabine treatment revealed that the epigenetic silencing of tumor suppressor genes, such as DLEC1 [43], OPCML [44], and RASSF1A [45] occurred frequently. A phase I/II clinical trial reported that RASSF1A, AKT1S1, and MLH1 genes were hypomethylated in all patients after DAC therapy. The demethylation of the developmentassociated transcription factors HOXA10 and HOXA11 was specifically related to ovarian cancer responders with PFS more than 6 months and associated with chemotherapy sensitivity [40].
Discussion The optimal regimen for DAC treatment
DAC schedule and DNA methylation/gene re-expression Due to limitations in acquiring tumor biopsies, analyses of plasma DNA or PBMCs were performed. In ovarian cancer patients, the methylation specific polymerase analysis indicated the HOXA11 and BRCA1 DNA promoters were hypomethylated in plasma DNA on day 8 and 15 post-decitabine treatment [39]. Appleton et al. observed that the global and MAGE-1 promoter DNA demethylation levels were maximal during days 8–12 in PBMCs and buccal cells. The demethylation levels then returned to near baseline levels by day 22 [34]. Similarly, Samlowski et al. reported that the levels of global DNA and MAGE-1 promoter methylation were apparent by day 7 (immediately after termination of continuous infusion), but declined 14 days after the start of the first cycle [33]. The immunohistochemistry analysis of available tumor biopsies from non-small cell lung cancer (NSCLC) patients suggested a marked induction of NY-ESO-1, MAGE-3, and p16, after DAC treatment [42]. Nevertheless, significant global demethylation was reduced or not found in tumor biopsies compared to PBMCs. This result may be due to the reduced uptake of DAC and lower proliferation rate in tumor cells compared to PBMCs [40,42]. In addition to the gene alterations in tumor cells, the levels of fetal hemoglobin (HbF) in lymphocytes increased 8–12 days after DAC treatment and then returned to the initial levels by day 15 in cycle 1. However, HbF levels accumulated in the subsequent cycles of treatment. These results suggest that repeated treatment with DAC is necessary to reverse gene silencing [34]. George et al. reported that DAC had a long-term activity within a 28-day cycle, and they observed that the MAGE-1 promoter was demethylated in the PBMC of 18/21 patient samples on day 28 following DAC treatment. Additionally, 14/16 patients exhibited increased HbF mRNA expression on day 28. Of these patients, 10/ 14 patients had increased HbF expression on day 7 and the expression increased on day 28 [14]. Thus, different DAC schedules may cause differential DNA methylation and gene expression profiles. The molecular mechanism of DAC action in solid tumor therapies is still unclear.
Previous solid tumor trials commonly used higher doses of DAC as a single day administration for one cycle. This approach exhibited high toxicity and had limited clinical effects. Momparler treated lung cancer patients with DAC at doses ranging from 200 to 660 mg/ m2 every 5–7 weeks. This treatment prolonged the survival of one in nine lung cancer patients post-DAC therapy [46]. In 2002, Aparicio and Weber reviewed eight clinical trials containing a total of 198 patients. There were no patients with a complete response, 3 had partial responses, and 17 had stable disease [47]. The effects of the demethylating drugs are not instant because they require a cell division and proliferation period for DAC to be incorporated into DNA. Thus, the repeated DAC treatments are crucial. Moreover, the appropriate dose of DAC is another key factor in obtaining demethylation activity while minimizing toxicity. Various treatment schedules were performed (Table 1). Stewart et al. showed that low-dose (2–10 mg/m2) DAC administered 1-hour daily using a 5 or 10 day infusion regimen decreased DNA methylation at the lowest doses in a trial with 31 solid tumor patients, and the changes at lower doses of DAC were comparable with those reported at higher doses or prolonged administration schedules. This low-dose DAC regimen had favorable responses when combined with other chemotherapeutic agents [17,48]. The use of the lower doses with high intensity, multi-day, and multi-cycle administration may be a rational epigenetic strategy in solid tumors. Specifically, the administration of DAC as low as 7 mg/ m2 for 5 days exerted significant clinical response and had reduced toxicity and side effects. Therefore, this approach may be an optimal scheme in solid tumor patients. Methylation and demethylation are interchangeable, and the DNA methylation status would revert to baseline levels by the start of the next cycle if treatment was delayed by more than 35 days. Thus, the 28-day treatment cycle is recommended. However, there are no definitive data on the number of cycles required for this epigenetic treatment. It is also unclear whether more cycles lead to better outcomes.
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
ARTICLE IN PRESS J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
Clinical response rate (%)
6
70 60 50 40
Response rate (%)
30 20
Response plus stable disease rate (%)
10 0
Fig. 2. The clinical response rates of DAC-based therapy in solid tumors. Both the response rates and response plus stable disease rates of DAC treatment in different solid tumor patients are summarized, based on the 13 phase I or II clinical trials in Table 1.
Monotherapy versus combined therapy Although DAC showed clinical activity as single agent, it was more effective in combination therapies with cytotoxic drugs, molecular targeted agents and other epigenetic agents or immunotherapy. Nevertheless, the drug combinations might generate undesired hematological toxicity and relatively low doses of DAC are suggested. Moreover, the sequential administration regimen was superior to concurrent administration because it permitted cancer cells to complete several division cycles and reactivated chemotherapy related genes [39]. The fittest population to DAC-based epigenetic therapy DAC-based therapy has exhibited favorable effects in some types of solid tumor patients based on our observations and these studies. As seen in Fig. 2, the response plus stable disease rates for ovarian, colorectal and lung cancers are 60%, 64%, and 47%, respectively (Fig. 2). Thus, for some tumor types such as ovarian cancer there may be a good clinical response when used in combination with conventional chemotherapeutic agents. The distinct regimens for each tumor type are listed in Table 2. Interestingly, immune pathway reactivation was highest in ovarian cancers, and the upregulation of cancer testis antigens was more pronounced for colorectal and ovarian cancers [23]. This result was consistent with our observations that ovarian and colorectal cancer patients benefit most from DAC epigenetic therapy. Approximately 15–20% of CRC patients possess a “CpG island methylator” phenotype (CIMP) that is resistant to the anti-EGFR therapy [18,49]. Thus, the combination of the epigenetic agent and anti-EGFR therapy may be beneficial for these CIMP-high patients [19]. DNA methylation and clinical response DNA methylation plays a central role in controlling tumor gene expression and multiple clinical trials confirmed that DAC reduced DNA methylation in solid tumors. Genes including MAGE-1, MAGE3, HOXA11, HOXA10, p16, NY-ESO-1 and p15 have reduced methylation following DAC treatment. MAGE-1, p16, and HOXA11 are the most widely examined genes responding to the biologic effects of DAC (Fig. 3). A phase II trial of DAC and carboplatin in ovarian cancer suggested that the number of demethylated genes was greater in tumor biopsies from patients with PFS of more than 6 months versus less than 6 months (311 vs. 244 genes). This result was also found in PBMCs (630 genes vs. 474 genes, PFS > 6 vs. < 6
months) [40]. Furthermore, they analyzed the enriched functional clusters for genes that were hypermethylated in biopsies from DACtherapy responders (PFS > 6 months) versus non-responders (PFS < 6 months). The most notable pathways enriched at baseline in DAC responsive patients were cytokine-cytokine receptor interactions, drug transporters, and mitogen-activated protein kinase (MAPK), toll-like receptor and Jak-STAT signaling pathways [40]. Large clinical trials have ascertained the demethylating activity of DAC (Fig. 3). However, the more critical issue is whether the DNA demethylation status of some specific genes could reflect the clinical effects. MAGE-1, HbF, or some other potential biomarkers were reported to be up-regulated in numerous solid tumor patients post-DAC treatment. Unfortunately, the demethylation levels of these known target genes could not definitely predict the prognosis of DAC-based treatment according to the current literature. However, the correlation between the demethylation rates of key genes and the clinical response rates should exist. It is important to explore the comprehensive regulation mechanism of DAC in vivo in suppressing tumor development. It is also critical to discover the crucial genes most correlated with clinical responses. Additionally, it will be necessary to investigate biomarkers for the early prediction of outcome for this novel epigenetic therapy. Conclusion The epigenetic treatment of patients with hematological diseases provided an opportunity for DAC-based therapy in solid tumors. Numerous clinical trials indicated that lower doses but high intensity, multi-day, and multi-cycle DAC administration was suggested for solid tumor treatment, especially patients with ovarian cancer, colorectal cancer, or cervical cancer. Moreover, DAC combined with other therapies showed better outcome than DAC alone. This result may be due to the finding that DAC enhances the sensitivity of the current available agents to tumor cells, inhibits the development of cancer cells and activates the immune responses. Several candidate genes were detected to verify the biological activity of DAC in vivo including MAGE1/3, p16, HOXA11. However, the correlation between DNA demethylation rate and the clinical response rate was unclear. The use of this novel epigenetic therapy has just begun in solid tumors. It will be valuable to understand the function and mechanism of DAC. The identification of critical biomarkers for prognosis requires further investigation. Conflicts of interest statement Nothing to report.
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
DAC dose (mg/m2) × time
Combination with other therapy
Best response
Ovarian cancer
2 × 7 day (10 or 20) × 5 day 10 × 5 day 7 × 5 day 2 × 7 day 45 × 2 day 7 × 5 day 7 × 5 day (20–40) × 3 day 20 × 3 day 25 × 3 day 30 × 3 day 7 × 5 day (5–15) × 10 day 50 × 3 day 7 × 5 day 20 × 3 day 25 × 3 day 30 × 3 day 7 × 5 day 2 × 7 day (20–40) × 3 day 7 × 5 day 7 × 5 day 7 × 5 day 20 × 3 day 25 × 3 day 7 × 5 day 7 × 5 day 7 × 5 day 7 × 5 day 5 × 7 day 5 × 7 day 5 × 7 day 5 × 7 day 7 × 5 day (20–40) × 3 day (20–40) × 3 day
— Carboplatin Carboplatin Carboplatin — Anti-EGFR — Chemotherapy — — — — Chemotherapy — Cisplatin Chemotherapy — — — — — — CIK — CIK — — — Chemotherapy — Chemotherapy — — — — — — —
SD:1/2 PD:1/2 CR:1/10 SD:3/10 PD:6/10 CR:1/17 PR:5/17 SD:6/17 PD:5/17 PR:1/1 PD:1/1 PR:2/20 SD:11/20 PD:7/20 SD:1/1 SD:1/2 PD:1/2 PD:1/1 SD:5/8 PD:3/8 SD:3/6 PD:3/6 SD:2/4 PD:2/4 PR:1/4 SD:2/4 PD:1/4 SD:1/8 PD:7/8 PR:8/21 SD:5/21 PD:8/21 PD:1/1 SD:1/3 PD:2/3 PD:2/2 PD:1/1 PR:1/1 SD:1/5 PD:4/5 PD:1/1 SD:1/1 PD:2/2 SD:3/4 PD:1/4 PD:1/1 SD:1/2 PD:1/2 SD:1/1 SD:1/2 PD:1/2 PD:2/2 SD:1/1 SD:5/14 PD:9/14 SD:2/2 PD:4/4 PD:1/1 PD:1/1 PD:12/12 PD:4/4
Colorectal cancer
Lung cancer
Cervical carcinoma Pleural tumors
Renal cancer Pancreatic cancer Liver cancer Esophageal cancer
Gastric cancer Neuroblastoma Rhabdomyosarcoma Osteosarcoma Synovial sarcoma Alveolar carcinoma Melanoma Breast cancer
Response plus stable disease rate (%)
Response rate (%)
Demethylation genes (post-DAC)
60 (n = 30)
10 (n = 30)
MAGE-1 LINE-1, HOXA11, BRCA1 RASSF1A, AKT1S1, MLH1, HOXA10, HOXA11 MAGEA-3, MAGEA-1, p16, p15
64 (n = 25)
8 (n = 25) MAGE
47 (n = 30)
3 (n = 30)
59 (n = 22)
36 (n = 22)
29 (n = 7)
14 (n = 7)
17 (n = 6)
0 (n = 6)
100 (n = 1) 50 (n = 6)
0 (n = 1) 0 (n = 6)
50 (n = 6)
0 (n = 6)
33 (n = 3)
0 (n = 3)
36 (n = 14) 100 (n = 2) 0 (n = 4) 0 (n = 1) 0 (n = 1) 0 (n = 12) 0 (n = 4)
0 (n = 14) 0 (n = 2) 0 (n = 4) 0 (n = 1) 0 (n = 1) 0 (n = 12) 0 (n = 4)
NY-ESO-1, MAGE-3, p16
MAGE-1
MAGE-1, caspase-8 MAGE-1 MAGE-1 MAGE-1
ARTICLE IN PRESS
Diagnosis
J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
Table 2 Different schedule, responses and biological activity for distinct solid tumors.
7
ARTICLE IN PRESS J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
Relative demethylation frequency
8
6 5 4 3 2
1 0
Fig. 3. Potential biomarker genes for DAC-based therapy. The relative frequencies of the potential biomarkers used in the 13 clinical trials are listed in order.
Acknowledgements This project is supported by grants from the National Basic Research Programs (2012CB518103, 2012CB910304, 2011CB910602) and the National Natural Science Foundations of China (31100554, 31270820, 81230061, 31125010, 81302166, 81472838). Reference [1] M. Esteller, P.G. Corn, S.B. Baylin, J.G. Herman, A gene hypermethylation profile of human cancer, Cancer Res. 61 (2001) 3225–3229. [2] Y. Oki, E. Aoki, J.P. Issa, Decitabine–bedside to bench, Crit. Rev. Oncol. Hematol 61 (2007) 140–152. [3] J.P. Issa, Decitabine, Curr. Opin. Oncol. 15 (2003) 446–451. [4] S.J. Adair, K.T. Hogan, Treatment of ovarian cancer cell lines with 5-aza-2′deoxycytidine upregulates the expression of cancer-testis antigens and class I major histocompatibility complex-encoded molecules, Cancer Immunol. Immunother. 58 (2009) 589–601. [5] S. Vijayaraghavalu, J.K. Dermawan, V. Cheriyath, V. Labhasetwar, Highly synergistic effect of sequential treatment with epigenetic and anticancer drugs to overcome drug resistance in breast cancer cells is mediated via activation of p21 gene expression leading to G2/M cycle arrest, Mol. Pharm. 10 (2013) 337–352. [6] C. Zeller, W. Dai, N.L. Steele, A. Siddiq, A.J. Walley, C.S. Wilhelm-Benartzi, et al., Candidate DNA methylation drivers of acquired cisplatin resistance in ovarian cancer identified by methylome and expression profiling, Oncogene 31 (2012) 4567–4576. [7] J.P. Issa, G. Garcia-Manero, F.J. Giles, R. Mannari, D. Thomas, S. Faderl, et al., Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies, Blood 103 (2004) 1635–1640. [8] P. Wijermans, M. Lubbert, G. Verhoef, A. Bosly, C. Ravoet, M. Andre, et al., Low-dose 5-aza-2′-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients, J. Clin. Oncol. 18 (2000) 956–962. [9] X. Chang, C.L. Monitto, S. Demokan, M.S. Kim, S.S. Chang, X. Zhong, et al., Identification of hypermethylated genes associated with cisplatin resistance in human cancers, Cancer Res. 70 (2010) 2870–2879. [10] R.W. Johnstone, A.A. Ruefli, S.W. Lowe, Apoptosis: a link between cancer genetics and chemotherapy, Cell 108 (2002) 153–164. [11] Y.A. Luqmani, Mechanisms of drug resistance in cancer chemotherapy, Med. Princ. Pract. 14 (Suppl. 1) (2005) 35–48. [12] G. Gifford, J. Paul, P.A. Vasey, S.B. Kaye, R. Brown, The acquisition of hMLH1 methylation in plasma DNA after chemotherapy predicts poor survival for ovarian cancer patients, Clin. Cancer Res. 10 (2004) 4420–4426. [13] J.A. Plumb, G. Strathdee, J. Sludden, S.B. Kaye, R. Brown, Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter, Cancer Res. 60 (2000) 6039–6044. [14] R.E. George, J.M. Lahti, P.C. Adamson, K. Zhu, D. Finkelstein, A.M. Ingle, et al., Phase I study of decitabine with doxorubicin and cyclophosphamide in children with neuroblastoma and other solid tumors: a Children’s Oncology Group study, Pediatr. Blood Cancer 55 (2010) 629–638. [15] M. Dean, ABC transporters, drug resistance, and cancer stem cells, J. Mammary Gland Biol. Neoplasia 14 (2009) 3–9. [16] M. Dean, T. Fojo, S. Bates, Tumour stem cells and drug resistance, Nat. Rev. Cancer 5 (2005) 275–284. [17] D.J. Stewart, J.P. Issa, R. Kurzrock, M.I. Nunez, J. Jelinek, D. Hong, et al., Wistuba, II, decitabine effect on tumor global DNA methylation and other parameters in a phase I trial in refractory solid tumors and lymphomas, Clin. Cancer Res. 15 (2009) 3881–3888.
[18] M. Scartozzi, I. Bearzi, A. Mandolesi, R. Giampieri, L. Faloppi, E. Galizia, et al., Epidermal growth factor receptor (EGFR) gene promoter methylation and cetuximab treatment in colorectal cancer patients, Br. J. Cancer 104 (2011) 1786–1790. [19] I. Garrido-Laguna, K.A. McGregor, M. Wade, J. Weis, W. Gilcrease, L. Burr, et al., A phase I/II study of decitabine in combination with panitumumab in patients with wild-type (wt) KRAS metastatic colorectal cancer, Invest. New Drugs 31 (2013) 1257–1264. [20] R.A. Juergens, J. Wrangle, F.P. Vendetti, S.C. Murphy, M. Zhao, B. Coleman, et al., Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer, Cancer Discov. 1 (2011) 598–607. [21] H.C. Tsai, H. Li, L. Van Neste, Y. Cai, C. Robert, F.V. Rassool, et al., Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells, Cancer Cell 21 (2012) 430–446. [22] B.K. Biswal, M.J. Beyrouthy, M.P. Hever-Jardine, D. Armstrong, C.R. Tomlinson, B.C. Christensen, et al., Acute hypersensitivity of pluripotent testicular cancerderived embryonal carcinoma to low-dose 5-aza deoxycytidine is associated with global DNA Damage-associated p53 activation, anti-pluripotency and DNA demethylation, PLoS ONE 7 (2012) e53003. [23] M.X. Liu, M.K. Siu, S.S. Liu, J.W. Yam, H.Y. Ngan, D.W. Chan, Epigenetic silencing of microRNA-199b-5p is associated with acquired chemoresistance via activation of JAG1-Notch1 signaling in ovarian cancer, Oncotarget 5 (2014) 944–958. [24] E. Fonsatti, H.J. Nicolay, L. Sigalotti, L. Calabro, L. Pezzani, F. Colizzi, et al., Functional up-regulation of human leukocyte antigen class I antigens expression by 5-aza-2′-deoxycytidine in cutaneous melanoma: immunotherapeutic implications, Clin. Cancer Res. 13 (2007) 3333–3338. [25] N. Cancer Genome Atlas Research, Comprehensive genomic characterization of squamous cell lung cancers, Nature 489 (2012) 519–525. [26] M. Challa-Malladi, Y.K. Lieu, O. Califano, A.B. Holmes, G. Bhagat, V.V. Murty, et al., Combined genetic inactivation of beta2-microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma, Cancer Cell 20 (2011) 728–740. [27] J. Wrangle, W. Wang, A. Koch, H. Easwaran, H.P. Mohammad, F. Vendetti, et al., Alterations of immune response of non-small cell lung cancer with azacytidine, Oncotarget 4 (2013) 2067–2079. [28] Q. Li, M.A. Tainsky, Epigenetic silencing of IRF7 and/or IRF5 in lung cancer cells leads to increased sensitivity to oncolytic viruses, PLoS ONE 6 (2011) e28683. [29] C. Chappell, C. Beard, J. Altman, R. Jaenisch, J. Jacob, DNA methylation by DNA methyltransferase 1 is critical for effector CD8 T cell expansion, J. Immunol. 176 (2006) 4562–4572. [30] T. Stubig, A. Badbaran, T. Luetkens, Y. Hildebrandt, D. Atanackovic, T.M. Binder, et al., 5-Azacytidine promotes an inhibitory T-cell phenotype and impairs immune mediated antileukemic activity, Mediators Inflamm. 2014 (2014) 418292. [31] S.L. Topalian, F.S. Hodi, J.R. Brahmer, S.N. Gettinger, D.C. Smith, D.F. McDermott, et al., Safety, activity, and immune correlates of anti-PD-1 antibody in cancer, N. Engl. J. Med. 366 (2012) 2443–2454. [32] A. Aparicio, C.A. Eads, L.A. Leong, P.W. Laird, E.M. Newman, T.W. Synold, et al., Phase I trial of continuous infusion 5-aza-2′-deoxycytidine, Cancer Chemother. Pharmacol. 51 (2003) 231–239. [33] W.E. Samlowski, S.A. Leachman, M. Wade, P. Cassidy, P. Porter-Gill, L. Busby, et al., Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation, J. Clin. Oncol. 23 (2005) 3897–3905. [34] K. Appleton, H.J. Mackay, I. Judson, J.A. Plumb, C. McCormick, G. Strathdee, et al., Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors, J. Clin. Oncol. 25 (2007) 4603–4609. [35] H. Fan, X. Lu, X. Wang, Y. Liu, B. Guo, Y. Zhang, et al., Low-dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: a phase I/II report, J. Immunol. Res. 2014 (2014) 371087. [36] A. Stathis, S.J. Hotte, E.X. Chen, H.W. Hirte, A.M. Oza, P. Moretto, et al., Phase I study of decitabine in combination with vorinostat in patients with advanced solid tumors and non-Hodgkin’s lymphomas, Clin. Cancer Res. 17 (2011) 1582–1590. [37] S.A. Cannistra, Cancer of the ovary, N. Engl. J. Med. 351 (2004) 2519–2529. [38] F. Fang, C. Balch, J. Schilder, T. Breen, S. Zhang, C. Shen, et al., A phase 1 and pharmacodynamic study of decitabine in combination with carboplatin in patients with recurrent, platinum-resistant, epithelial ovarian cancer, Cancer 116 (2010) 4043–4053. [39] D.E. Matei, K.P. Nephew, Epigenetic therapies for chemoresensitization of epithelial ovarian cancer, Gynecol. Oncol. 116 (2010) 195–201. [40] D. Matei, F. Fang, C. Shen, J. Schilder, A. Arnold, Y. Zeng, et al., Epigenetic resensitization to platinum in ovarian cancer, Cancer Res. 72 (2012) 2197–2205. [41] B.F. Chu, M.J. Karpenko, Z. Liu, J. Aimiuwu, M.A. Villalona-Calero, K.K. Chan, et al., Phase I study of 5-aza-2′-deoxycytidine in combination with valproic acid in non-small-cell lung cancer, Cancer Chemother. Pharmacol. 71 (2013) 115–121. [42] D.S. Schrump, M.R. Fischette, D.M. Nguyen, M. Zhao, X. Li, T.F. Kunst, et al., Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura, Clin. Cancer Res. 12 (2006) 5777–5785. [43] J. Kwong, J.Y. Lee, K.K. Wong, X. Zhou, D.T. Wong, K.W. Lo, et al., Candidate tumor-suppressor gene DLEC1 is frequently downregulated by promoter hypermethylation and histone hypoacetylation in human epithelial ovarian cancer, Neoplasia 8 (2006) 268–278. [44] G.C. Sellar, K.P. Watt, G.J. Rabiasz, E.A. Stronach, L. Li, E.P. Miller, et al., OPCML at 11q25 is epigenetically inactivated and has tumor-suppressor function in epithelial ovarian cancer, Nat. Genet. 34 (2003) 337–343.
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010
ARTICLE IN PRESS J. Nie et al./Cancer Letters ■■ (2014) ■■–■■
[45] I. Ibanez de Caceres, C. Battagli, M. Esteller, J.G. Herman, E. Dulaimi, M.I. Edelson, et al., Tumor cell-specific BRCA1 and RASSF1A hypermethylation in serum, plasma, and peritoneal fluid from ovarian cancer patients, Cancer Res. 64 (2004) 6476–6481. [46] R.L. Momparler, D.Y. Bouffard, L.F. Momparler, J. Dionne, K. Belanger, J. Ayoub, Pilot phase I-II study on 5-aza-2′-deoxycytidine (decitabine) in patients with metastatic lung cancer, Anticancer Drugs 8 (1997) 358–368. [47] A. Aparicio, J.S. Weber, Review of the clinical experience with 5-azacytidine and 5-aza-2′-deoxycytidine in solid tumors, Curr. Opin. Investig. Drugs 3 (2002) 627–633.
9
[48] P. Pohlmann, L.P. DiLeone, A.I. Cancella, A.P. Caldas, L. Dal Lago, O. Campos Jr., et al., Phase II trial of cisplatin plus decitabine, a new DNA hypomethylating agent, in patients with advanced squamous cell carcinoma of the cervix, Am. J. Clin. Oncol. 25 (2002) 496–501. [49] N. Cancer Genome Atlas, Comprehensive molecular characterization of human colon and rectal cancer, Nature 487 (2012) 330–337.
Please cite this article in press as: Jing Nie, Lin Liu, Xiang Li, Weidong Han, Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors, Cancer Letters (2014), doi: 10.1016/j.canlet.2014.08.010