Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Dec 1.
Published in final edited form as: Curr Pharmacol Rep. 2017 Oct 14;3(6):360–373. doi: 10.1007/s40495-017-0110-5

Epigenetic Therapeutics and Their Impact in Immunotherapy of Lung Cancer

Ju Hwan Cho 1, Filiz Oezkan 1,2, Michael Koenig 1, Gregory A Otterson 1, James Gordon Herman 3, Kai He 1
PMCID: PMC5831502  NIHMSID: NIHMS913522  PMID: 29503796

Abstract

Lung cancer is the leading cause of cancer-related death in the United States and worldwide. Novel therapeutic developments are critically necessary to improve outcomes for this disease. Aberrant epigenetic change plays an important role in lung cancer development and progression. Therefore, drugs targeting the epigenome are being investigated in the treatment of lung cancer. Monotherapy of epigenetic therapeutics such as DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi) have so far not shown any apparent benefit while one of the clinical trials with the combinations of DNMTi and HDACi showed a small positive signal for treating lung cancer. Combinations of DNMTi and HDACi with chemotherapies have some efficacy but are often limited by increased toxicities. Preclinical data and clinical trial results suggest that combining epigenetic therapeutics with targeted therapies might potentially improve outcomes in lung cancer patients. Furthermore, several clinical studies suggest that the HDACi vorinostat could be used as a radiosensitizer in lung cancer patients receiving radiation therapy. Immune checkpoint blockade therapies are revolutionizing lung cancer management. However, only a minority of lung cancer patients experience long-lasting benefits from immunotherapy. The role of epigenetic reprogramming in boosting the effects of immunotherapy is an area of active investigation. Preclinical studies and early clinical trial results support this approach which may improve lung cancer treatment, with potentially prolonged survival and tolerable toxicity. In this review, we discuss the current status of epigenetic therapeutics and their combination with other antineoplastic therapies, including novel immunotherapies, in lung cancer management.

1. Introduction

Lung cancer is the leading cause of cancer-related death and a major healthcare challenge globally [1]. Non-small cell lung cancer (NSCLC), accounting for about 85% of all cases, is the major histologic subtype. Small cell lung cancer (SCLC) accounts for 10–12% of all lung cancer cases [2]. At the time of diagnosis more than 40% of patients are already in an advanced tumor stage. Despite the recent development of targeted therapies and immunotherapies, the overall prognosis for patient is still poor, with less than 15–18% of patients surviving at 5 years after diagnosis. The primary treatment for the majority of advanced lung cancer patients continues to be cytotoxic chemotherapy [3]. Novel lung cancer treatment strategies using epigenetic therapeutics alone or in combination with other therapies have been preclinically developed and clinically tested over the last decade, with numerous ongoing clinical trials. Epigenetic therapeutics were first shown to be effective in the treatment of hematological malignancies such as acute myeloid leukemia (AML), myeloid dysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and some types of lymphoma. Some are approved by the US Food and Drug Administration (FDA) as shown detailed in Supplementary Table 1. Epigenetic therapeutics such as DNA methyltransferase inhibitors (DNMTis) and histone deacetylase inhibitors (HDACis) were first tested as monotherapies, and subsequently as combination therapies. In this review, we discuss the current status of their potential application in lung cancer management with perspectives on combination with other novel therapies, including immunotherapy.

2. Epigenetics in lung cancer

Epigenetic alterations such as DNA methylation and histone modifications are known to be involved in tumor development and tumor progression of lung cancer and other cancers [15].

2.1 DNA-methylation

DNA methylation affects the transcription of genes without altering the DNA nucleotide sequence and is found sparsely but globally in human cells. In eukaryotic DNA, cytosine is methylated and then converted into 5-methylcytosine by DNA methyltransferases (DNMTs) [16]. There are three enzymatically active DNMTs in human cells: DNMT1, 3a and 3b [1719]. Global hypomethylation is characteristic in the transformation of benign cells to malignant cells and accelerates as cancer progresses. On the other hand, hypermethylation of specific regions, such as the CpG islands of tumor suppressor genes, plays an important role in carcinogenesis for many types of cancers, including lung cancer [20, 21]. Hypermethylation of these sequences can induce inappropriate silencing of growth regulatory genes and tumor suppressor genes. Inactivation of tumor suppressor genes via promoter hypermethylation is an early event in carcinogenesis and reported to be an early sign of lung cancer development [22].

2.1.1 DNA-methyltransferase

inhibitors In the 1960s, Vesely et al. first described the DNMTis azacitidine and decitabine and showed their cancerostatic effect in preclinical leukemia studies [23, 24]. In 1980 Jones et al. discovered that azanucleotides could induce DNA hypomethylation, especially when lower doses were used [25]. Momparler et al. conducted preclinical and clinical studies proving that azanucleotides were effectively targeting DNA methylation in leukemic cells [26, 27]. After numerous further trials, azacitidine and decitabine were finally approved by the FDA for hematological malignancies (see Supplement Table 1).

Table 1.

Epigenetic therapeutics (mono- and combined) in NSCLC patients

Epigenetic agent Phase Total patient # Patient # with NSCLC Response rate CR/PR SD OS month Side effects Study status NCT number Reference
DNMTi

decitabine 1 EC, LC, pleural meso (35) 20 - - - grade 4 neutropenia (15), grade 3 leukopenia, thrombocytopenia or anemia in (20) of 35, (2) of 35, and (3) of 35 patients completed NCT00019825 [4]

HDACi   

belinostat 1 - - - - - not yet published completed NCT00413075 [5]
entinostat 1 refractory solid tumors and lymphomas (27) 2 better in lower dose 1/2 - frequent hypophosphatemia completed NCT00020579 [6, 7]
romidepsin 2 19 16 no objective responses 9 - grade 3 anemia (3), grade 3/4 neutropenia (4), grade 4 thrombocytopenia (1), grade 3 hypoxia (3), pneumonitis (1), tumor pain (1), thrombosis (1), cellulitis at injection site (2), pulmonary embolism (1) completed NCT00020202 [8]
vorinostat 2 16 16 2 progressed after 1 cycle (2), included in further analyses (14) - 7.1 9 SAE: neutropenia (3), pulmonary embolism (2), hyperglycemia (1), thrombosis (1), dyspnea (1), cerebrovascular accident (1), 16 AE: neutropenia (6), hyperglycemia (5), lymphopenia (3), fatigue (4), pneumonia (1) completed NCT00138203 [9]

DNMTi HDACi

azacitidine entinostat 1/2 45 45 see next columns 1/1 10 therapy cycle 1: grade 3: hematologic toxicities (9), gastrointestinal symptoms (6), electrolyte disturbances (3), general symptoms (5), grade 4: hematologic toxicities (3); therapy cycle ≥2: grade 3: hematologic toxicities (9), gastro intestinal (1), endocrine (1), general symptoms (5); grade 4: hematological toxicities (3) completed NCT00387465 [10]
azacitidine entinostat - - Stage I NSCLC - - - not yet published terminated NCT01207726 [11]
azacitidine entinostat 1 - Stage I NSCLC - - - not yet published terminated NCT01886573 [12]
azacitidine entinostat - - - - - - not yet published terminated NCT01935947 [13]
decitabine valproic acid 1 8 8 - - 1 grade 3 neurotoxicity (2) terminated earlier (initially 25 planned) NCT00084981 [14]

Abbreviations: AE: adverse event; CR: complete response; DNMTi: DNA methyltransferase inhibitor; EC: esophageal cancer; HDACi: histone deacetylase inhibitor; LC: lung cancer; meso: mesothelioma; NCT Number: ClinicalTrials.gov identifier; NSCLC: non-small cell lung cancer; OS: overall survival; PR: partial response; SAE: serious adverse event; SD: stable disease; (−): data not available.

2.1.2 DNMTi-monotherapy in lung cancer

A pilot phase I-II study on decitabine in patients with stage IV NSCLC was conducted by Momparler et al. [28, 29]. One patient was reported to have survived 81 months. This promising finding led to further DNMTi trials in lung cancer patients. Most of these trials combined DNMTis with other agents. To our knowledge, only one monotherapy trial with decitabine was conducted in NSCLC patients; no objective clinical response was observed and severe toxicities occured. Grade 4 neutropenia was observed in 15 patients, and was dose limiting in four patients; grade 3 neutropenia, thrombocytopenia or anemia were frequently reported as well. Two patients with extensive liver metastases experienced grade 3 hepatotoxicity [4] (Table 1). Due to limited efficacy in NSCLC as monotherapy, further trials combined DNMTis with other agents [4, 30].

2.2 Histone modifications

In eukaryotes, 147 base pairs (bp) of DNA are wrapped around an octamer of histones consisting of two copies each of H2A, H2B, H3 and H4 [31]. The resulting nucleosomes are further compacted to form higher-order chromatin structures. There are several types of histone modifications, including acetylation, methylation and ubiquitination. These modifications regulate gene expression by altering the interactions of histones with chromatin-associated proteins, marking regions of transcriptionally active euchromatin and inactive heterochromatin [32]. Histone post-translational modification is not dependent on the cell cycle and is potentially reversible [33, 34]. Histones can be post-translationally modified by histone acetyltransferases (HATs) and histone deacetylases (HDACs) [35]. HDACs are responsible for removing the acetyl-group from lysine residues in histones, inducing a condensed state of inactivated-chromatin (heterochromatin) and transcriptional repression; HATs perform the opposite function by adding acetyl-groups to lysine residues and inducing a euchromatin state and transcriptional activation [36]. There are four classes of HDAC enzymes based on their structures and functions: class I (HDAC 1–3 and 8), II (HDAC 4–7, 9 and 10), III (Sir-2 related - SIRT1-7) and IV (HDAC 11) [37] HDAC expression can be altered in various cancers. Overexpression of HDACs was observed in several solid tumors including lung cancer [3840]. A synergistic interaction between HDAC-mediated histone deacetylation and DNMT-mediated DNA methylation can collaboratively cause gene silencing [15, 41, 42]. These mechanisms are known to be involved in cancer development [36].

2.2.1 Histone deacetylase inhibitors

HDACis were developed to reverse the gene silencing effect of HDACs and are classified into the following four major classes: 1) hydroxamic acids, 2) amino-benzamides, 3) cyclic peptides and 4) short-chain fatty acids [31]. The most commonly used HDACi in clinical trials with solid tumors and hematological malignancies belong to the first two groups. Three HDACi have been FDA-approved for the treatment of T-cell lymphomas: vorinostat, romidepsin, and belinostat. The HDACi panobinostat has been FDA-approved for the treatment of multiple myeloma since 2015 (Supplementary Table 1).

2.2.2 HDACi-monotherapy in lung cancer

HDACi monotherapies were investigated in NSCLC and SCLC clinical trials. Romidepsin was tested in three single-arm monotherapy trials. Among them, a phase I trial in patients with neuroendocrine tumors was terminated early due to an increased number of severe cardiac toxicities [43]. Two later trials, one in NSCLC and one in SCLC, did not identify severe cardiac toxicities despite the fact that the dosage was increased in the NSCLC trial [44, 45] (Table 1 and Supplementary Table 3). Romidepsin was ultimately found to be clinically ineffective in a monotherapy setting. Safety and efficacy of entinostat, vorinostat, belinostat and panabinostat were investigated in monotherapy settings in NSCLC patients [6, 7, 9]. Entinostat showed only minimal efficacy, but was reported to be safe and tolerable in NSCLC [6, 7]. Vorinostat did not show any objective antitumor response in NSCLC patients, and severe toxicities were reported [9]. Panobinostat, a pan-deacetylase inhibitor, is the only HDACi that induced tumor-shrinkage as a monotherapy in SCLC. However, the trial was terminated earlier than planned, as only a small percentage of patients responded [46]. Thus, HDACi monotherapy has not proven to be effective in lung cancer.

2.3 Epigenetic therapeutic combinations for the treatment of lung cancer

As the antitumor efficacy of epigenetic monotherapies is low, more recent trials have combined epigenetic therapeutics in an effort to improve outcome. The observation both in vitro and in vivo that HDAC-mediated histone deacetylation and DNMT mediated DNA methylation collaboratively cause gene silencing supported clinical trials to test the efficacy of combining HDAC inhibition and DNMT inhibition in cancer treatment [15, 19, 41, 42]. Several such trials were terminated earlier than planned. Chu et al. published a clinical trial combining HDAC inhibition with valproic acid and DNMT inhibition with decitabine in NSCLC patients. Unacceptable neurotoxic adverse events were reported and there was no survival benefit [14]. Juergens et al. conducted a phase I/II trial of combined azacitidine and entinostat in NSCLC patients. Median overall survival (OS) and median progression free survival (PFS) were encouraging, 8.6 months and 7.4 weeks respectively, after completion of at least one cycle of epigenetic treatment, although the objective response rate was low [10]. The vast majority of patients (87%) discontinued the therapy due to disease progression [10] (Table 1). Another interesting finding from this study was an increased objective response (21%) of those patients continuing with other chemotherapies. Subsequently, further trials combining chemotherapy with epigenetic agents were conducted.

3. Epigenetic therapeutics combined with non-immune therapies in lung cancer

To improve therapeutic efficacy in lung cancer, clinical trials with combinations of epigenetic therapeutics with chemotherapeutics, radiotherapy, targeted therapy and more recently immunotherapy have been conducted.

3.1. Epigenetic therapeutics combined with chemotherapy

In preclinical studies, taxanes and platinum-based agents led to an increased antitumor effect when combined with HDACi [47, 48]. This was investigated in clinical trials combining HDACi with chemotherapeutics. Ramalingam et al. published a trial combining carboplatin and paclitaxel with vorinostat or placebo in 94 NSCLC patients. Among them, twenty completed the vorinostat arm and, showed a prolonged PFS (6 vs 4.1 months), a prolonged OS (13 vs. 9.7 months) and an improved response rate (RR) of 34% compared to 12% in the placebo arm. The 1-year OS was 51% in the vorinostat group and 33% in the placebo group [45]. Toxicities were substantial with 3 deaths occurring in the vorinostat arm [49, 50], Table 2a. Jones et al. reported partial response (PR) in 1 out of 5 lung cancer patients treated with panobinostat, paclitaxel, and carboplatin [50]. A recent phase I study combining belinostat with carboplatin and paclitaxel, presented at the 2016 World Conference on Lung Cancer by Waqar et al., demonstrated encouraging antitumor efficacy. In 13 out of 23 patients RR was available. PR was seen in 35%, stable disease (SD) in 17% and only one patient had a progressive disease (PD) [51]. Compared to carboplatin/paclitaxel alone, the combination with an HDACi appears promising [49].

Table 2.

a Epigenetic therapies combined with non-immune therapies in NSCLC patients

Epigenetic agent Phase Total
patient
#
Patient
#
with
NSCLC
Response
rate
CR/PR PFS
month
SD OS
month
Side effects Study
status
NCT
number
Reference
DNMTi Chemotherapy

azacitidine cisplatin 1 - - - - - - - not yet published - NCT00901537 [54]
azacitidine carboplatin or paclitaxel 1 - - - - - - - not yet published completed NCT01478685 [55]

HDACi Chemotherapy

belinostat paclitaxel + carboplatin 1 23 23 35% 8 - 4 - fatigue (91%), nausea (78%), constipation (74%), anemia (65%), diarrhea (65%), alopecia, arthralgia, decreased appetite, insomnia, neutropenia (61%), dizziness, vomiting (57%), headache (52%) completed NCT01310244 [51]
belinosta standard of care chemotherapy 1 aimed 7 aimed 7 - - - - not yet published terminated NCT01090830 [56]
panobinostat pemetrexed 1 - - - - - - - not yet published terminated NCT00907179 [57]
panobinostat carboplatin + etoposid 1 6 4 - - - - dose-limiting toxicity (2), grade 4 thrombocytopenia and grade 4 febrile neutropenia (1) terminated because of inacceptable toxicities NCT00958022 [58]
panobinostat cisplatin+ pemetrexed 1 - - - - - - - not yet published active, not recruiting NCT01336842 [59]
panobinostat paclitaxel, carboplatin, bevacizumab 1 22 4 - 3 11 - neutropenia (90%, 67% grade 4), thrombocytopenia (90%), anemia (76%), fatigue (71%), diarrhea (52%), vomiting (48%) completed NCT00556088 [50]
vorinostat bevacizumab + carboplatin + paclitaxel 1/2 25 25 - - - - - not yet published terminated NCT00702572 [60]
vorinostat topotecan 1/2 - - - - - - - not yet published terminated NCT00697476 [61]
vorinostat gemcitabine + platinum-based agent 1 61 (10 completed) 61 - - - - - 39 SAE, 61 AE (most frequent anemia and asthenia) completed NCT00423449 [62]
vorinostat docetaxel 1 12 3 - - - - - not separately documented for LC NCT00565227 [63]
vorinostat vs placebo carboplatin + paclitaxel 2 62 (20 completed in vorinostat-arm) 32 (12 completed in placebo-arm) 94 34% (vorinostat-arm) vs 12% - 6 vs 4.1 13 vs 9.7 29 SAE (vorinostat-arm):febrile neutropenia (2), anemia (2), cardiac disorders (3), gastrointestinal disorders (10), death (3), other general disorders (9), 100% other AE in vorinostat-arm: anemia (43), constipation (23), diarrhea (20), nausea (37), vomiting (25), PD (20), fatigue (52), leukocyte amount decreased (28), thrombocyte count decreased (36), anorexia (35), hyperglycemia (29), peripheral neuropathy (29), and others completed NCT00481078 [49]
vorinostat vs placebo (phase 2) paclitaxel (phase 1), carboplatin (phase 2) 1/2 12 (vorino-stat-arm 4) 12 (vorino-stat-arm 4) - - - - - SAE 3/4: nausea (1), sepsis (1), platelet count decreased (1), hypotension (1), dehydratation (1), neutrophil count decreased (1), platelet count decreased (1), WBC decreased (1) AE 4/4: anemia (2), blurred vision (1), diarrhea (2), and others terminated NCT01413750 [64]
vorinostat vs placebo carboplatin + paclitaxel 2/3 126 (vorino-stat-arm), completed 43 126 (vorino-stat-arm) - - - - - SAE 63 (vorinostat-arm) vs 45, AE 114 vs 117 - NCT00473889 [65]
romidepsin flavopiridol 1 aimed 23 - - - - - - not yet published terminated NCT00094978 [66]
vorinostat bortezomib 1 21 neo-adjuvant (21) 6 patients > 60% necrosis - - - - most common toxicities included: grade 1 fatigue (14/20, 70%), grade I nausea (8/20, 40%), grade 1 neuropathy (4/20, 20%), and grade 1 diarrhea (4/20, 20%). DLT (2) completed NCT00731952 [52]
vorinostat bortezomib 2 18 18 - - 1.5 5 4.7 grade 3 toxicities: thrombocytopenia (7), lymphopenia (3), fatigue (4), hyponatremia (3), dizziness (2), vomiting (2), syncope (2), neuropathy (2), grade 4 toxicities: thrombocytopenia (1), fatigue (1) completed NCT00798720 [67]

DNMTi Targeted therapy

azacitidine erlotinib 1 30 2 * 1 >4 in NSCLC, n/a for SCLC 2 - not separately documented for LC, conjunctivitis, infusion reaction (2/5) completed NCT00996515 [68]
azacitidine erlotinib + mTOR inhibitor CC-223 1b - - - - - - - not yet published completed, Celgene NCT01545947 [69]

HDACi Targeted therapy

belinostat erlotinib 1/2 - 5 (preliminary) - - - - grade 3 diarrhea (3), grade 2 diarrhea(1), grade 2 rash (1), grade 1 diarrhea (1), grade 1 nausea (1), terminated after 5 patients due to severe toxicities NCT01188707 [70]
entinostat erlotinib 2 132 132 prolonged OS with high expression of E-cadherin - - - 9.4 (patients with high levels of e-cadherin) vs5.9 Fatigue (32), rash (35), diarrhea (30), dermatitis acneiformis (12), dyspnea (11), anemia (7), asthenia (7), hypokalemia (7), abdominal pain (7), hypoxia (4), pleural effusion (4), pneumonia (3), hypophosphatemia (3), syncope (1), completed NCT00750698 [71]
panobinostat erlotinib 1 42 35 - 3 2.5 14 7.4 most common AEs were fatigue and nausea (grades 1–3) and rash and anorexia (grades 1–2), not specified for LC completed NCT00738751 [72]
panobinostat sorafenib 1 - - - - - - - not yet published completed NCT01005797 [73]
romidepsin (phase 2) erlotinib (phase 1) 1/2 17 17 - 3.3, prolonged PFS >6 7 - nausea, vomiting, and fatigue (each 82%), diarrhea (65%), anorexia (53%), and rash (41%) completed NCT01302808 [74]
vorinostat erlotinib 1/2 33 EGFR mutant NSCLC 33 - - - 7 no significant difference anemia (20), diarrhea (19), rash (12), fatigue (16), nausea (11), anorexia (12), vomiting (9), xerosis cutis (8), xerostomia (6), conjuncitivis (5), epigastralgia (4), leukopenia (3), neutropenia (3), mucositis (4), pneumonitis (1). completed NCT00503971 [75]
vorinostat (phase 1) erlotinib (phase 2) 1/2 16 (1 completed)/ 9 discontinued due to PD 16 - - - - - only 1 patient completed the study terminated NCT00251589 [76]
vorinostat gefitinib 1 EGFR mutant NSCLC aimed 18 - - - - - not yet published recruiting NCT02151721 [77]
vorinostat gefitinib 1/2 52/43 52/43 see next columns 16 3.2 6 19 toxicities (grade 1–3) in phase I (15) in phase 2 (43) completed NCT01027676 [78]
vorinostat sorafenib 1 35 15 1 2.2 5 - fatigue (8/15), rash (8/15), nausea (8/15), anorexia (7/15), diarrhea (6/15), hand-foot syndrome (5/15), others completed NCT00635791 [79]

DNMTi Other therapy

decitabine genistein 1/2 20 - - - - - - not yet published completed NCT01628471 [80]
decitabine romidepsin + celecoxib 1 34 - - - - - - not yet published completed NCT00037817 [81]
b Epigenetic therapies combined with immune therapies in NSCLC patients

Epigene
tic
agents
Pha
se
Patients
#
Patient
s
# with NSCL
C
Respo
nse
rate
PR SD Side
Effects
Estimat
ed
completi
on
Study
Status
NCT
Number
Refere
nce
DNMTi Immunotherapy

azacitidine pembrolizumab 2 12 (in 2016), aimed 90 12 - - - not yet published - recruiting NCT02546986 [124]

HDACi Immunotherapy

ACY 241 nivolumab + ipilimumab 1b not yet completed, aimed 41 aimed 41 - - - not yet completed 2017 recruiting NCT02635061 [125]
entinostat pembrolizumab 1b/2 NSCLC and melanoma, 22 not yet completed 22 - 1 (PD-1 naive) 3 (PD-1 naive), 2 (prior PD-1 treatment) not yet completed, first 22 patients 5 study related AE; 2017: treatment-related AEs occurred in 5 patients (most common: nausea and fatigue (2); grade 3/4 fatigue and rash (1). 2019 recruiting NCT02437136 [122]
entinostat pembrolizumab 1 NSCLC aimed 30, not yet completed - - - not yet published 2018 recruiting NCT02909452 [126]
entinostat nivolumab + ipilimumab solid tumors mainly BC not yet completed - - - not yet completed 2018 recruiting NCT02453620 [127]
vorinostat pembrolizumab 1/2 aimed 100, not completed aimed 100, not completed - - - not yet completed 2017 recruiting NCT02638090 [128]

DNMTi HDACi Immunotherapy

azacitidine entinostat nivolumab 1 not completed - - - - not yet published Aug-18 recruiting NCT01928576 [129]

DNMTi others Immunotherapy

azacitidine epacadostat pembrolizumab 1/2 solid tumors and NSCLC not completed - - - not yet published 2021 recruiting NCT02959437 [130]
decitabine tetra-hydrouridine nivolumab 2 not completed NSCLC - - - not yet published 2019 not yet recruiting NCT02664181 [131]
azacitidine paclitaxel 1 (1+3, 3 mono, or 3+ durvalumab) 2 - advanced NSCLC - - - not yet published 2018 recruiting NCT02250326 [132]

Abbreviations: AE: adverse event; CR: complete response; DLT: dose limiting toxicities; DNMTi: DNA methyltransferase inhibitor; HDACi: histone deacetylase inhibitor; LC: lung cancer; n/a: not available; NCT Number: ClinicalTrials.gov identifier; NSCLC: non-small cell lung cancer; OS: overall survival; PD: progressive disease; PR: partial response; PFS: Progression free survival; SAE: serious adverse event; SCLC: small-cell lung cancer; SD: stable disease; WBC: white blood cells. (−): data not available.

*

not separately documented for lung cancer.

Abbreviations: AE: adverse event; BC: breast cancer; CR: complete response; CRS: cytokine release syndrome; EC: esophageal cancer; HL: Hodgkin's lymphoma; LC: lung cancer; NCT Number: ClinicalTrials.gov identifier; NHL: Non-Hodgkin's lymphoma; NSCLC: non-small cell lung cancer; PD: progressive disease; PR: partial response; SD: stable disease. (−): data not available.

Unfortunately, the results of most clinical trials combining HDACi with chemotherapy in lung cancer have been negative. Some of these trials were terminated early due to toxicities. Trials with published outcome data are limited, and several trials are still ongoing (Table 2a). While combined treatment of lung cancer patients with either belinostat or vorinostat with carboplatin and paclitaxel shows preliminary efficacy, larger trials must be performed and ongoing trials need to be completed to confirm efficacy. In a neoadjuvant phase-I trial combining the proteasome inhibitor bortezomib with vorinostat, necrosis was detected in 6 out of 20 patients who completed the treatment. Additionally Jones et al. reported reduced SUV-uptake in PET-CT scans performed after treatment completion but before surgery [52]. Erasmus et al. reported that a reduction of SUV-uptake could predict operability and survival [53]. It remains unclear if the necrosis was related to the treatment or caused by the tumor itself. Furthermore the tumor size was not reduced by this neoadjuvant treatment. Toxicities were dose-limiting in two patients (Table 2a).

3.2 Epigenetic therapeutics combined with targeted therapies

There are several ongoing and completed trials combining epigenetic drugs with targeted therapies (Table 2a). A trial combining the HDACi belinostat with the erlotinib was terminated due to intolerable toxicities. The full publication of this study is still pending. Witta et al. published a phase II, two-arm trial combining entinostat or placebo with erlotinib in 132 advanced NSCLC who previously experienced chemotherapy treatment failure. The trial population was not preselected by actionable EGFR mutation. The combined therapy of erlotinib and entinostat did not result in improved clinical outcome in this unselected patient population. Han et al. published a phase I/II trial combining gefitinib and vorinostat in patients with both EGFR-mutant and EGFR-wildtype advanced NSCLC. Subgroup analysis found that vorinostat potentially improves the efficacy of gefitinib in EGFR-mutant NSCLC [78, 82]. However, only 13 patients in this study were EGFR-mutant. Larger trials will be needed to validate the finding. A preclinical study demonstrated that the combined use of vorinostat and osimertinib could reverse BIM deletion polymorphism–mediated osimertinib resistance in EGFR-mutant NSCLC cells. Therefore, the authors suggest the future development of selective HDAC inhibitors to overcome osimertinib resistance [83]. To our knowledge, combined epigenetic therapy and ALK-inhibitor therapy trials have not yet been conducted. A preclinical study by Fukuda et al. demonstrated that HDAC inhibition with quisinostat could overcome crizotinib resistance by mesenchymal-epithelial transition. This preclinical finding might support related clinical studies in NSCLC with ALK rearrangement [84].

3.3 Epigenetic therapeutics combined with radiation therapy

Preclinical studies combining HDACi and radiation therapy in lung cancer, colon cancer, breast cancer, and other cancers demonstrated increased anti-tumor efficacy [8587]. The combination of HDACi with radiotherapy was investigated in three recent lung cancer trials [8890]. Vorinostat was used as a radiosensitzer in a phase-I study, enrolling twelve NSCLC patients with brain metastases. The combination of vorinostat with radiation therapy was reported to be safe and the median OS was 36 weeks. A recently published study by Choi et al. enrolled 17 NSCLC patients with up to 4 brain metastases and used vorinostat as a radiosensitizer before sterotactic radiotherapy of the brain metastases. Dose-limiting toxicities did not occur (Supplementary Table 3) [89]. Further studies are ongoing.

4. Epigenetic therapeutics combined with immunotherapy

4.1 Lung cancer immunity and immunotherapy

NSCLC has been historically considered to be non-immunogenic. In recent years the role of the immune system in cancer development and progression, and in lung cancer in particular, has been better understood [91, 92]. Both the innate and the adaptive immune systems are involved in destroying cancer cells and inhibiting cancer cell growth [93]. Immature dendritic cells (DC), existing in most human cancers, capture cancer cell antigens [94, 95]. Once activated, DCs present cancer antigens within the major histocompatibility complex (MHC) to naïve T-cells in tumor-draining lymph nodes and induce a T-cell response. Cytotoxic CD8+ T-cells are then enabled to spot and destroy cancer cells [3]. Dysfunction of the immune system is well-known to be involved in cancer development and progression through different mechanisms [93, 96, 97]. Recent publications have demonstrated that various immunological mechanisms play an important role in NSCLC. Impairment of T-cell proliferation and an immunosuppressive microenvironment contribute to lung cancer growth [98100]. One of the major mechanisms of T-cell suppression is the so-called immune checkpoint. Several immune checkpoints have been discovered including CTLA-4/B7, PD-L1/PD-1, LAG-3, TIM-3 [101].

Programmed death receptor-1 (PD-1) is expressed by cytotoxic T-cells infiltrating NSCLCs. The increased expression and activation of PD-1 has a wide immunosuppressive effect [102]. The upregulation of programmed death receptor ligand-1 (PD-L1) on NSCLC cells correlates with the suppression of activating tumor-infiltrating DCs and T-cells [103105]. These recent findings in cancer immunity brought forth the development of novel immunotherapies in lung cancer and other malignancies. Some of the most promising drugs target immune checkpoints such as PD-1/PD-L1 and CTLA-4. An increased OS in NSCLC patients treated with anti-PD-1 or anti-PD-L1 in second-line therapy and also in selected patients in first-line therapy was demonstrated [91, 106108]. Nivolumab (anti-PD1), pembrolizumab (anti-PD1), and atezolizumab (anti-PDL1) have been approved as a 2nd line NSCLC therapy. Pembrolizumab has been approved as 1st line treatment in metastatic NSCLC. In May 2017, pembrolizumab in combination with carboplatin and pemetrexed was granted accelerated approval for metastastic nonsquamous NSCLC as 1st line treatment (Supplementary Table 4).

4.2 Preclinical studies of combined epigenetic therapeutics and immunotherapy

Only a minority of patients treated with immunotherapy shows a long-term benefit [18, 107, 109]. To enhance clinical efficacy, combinations of epigenetic therapies with immunotherapies were studied in lung cancer and other cancers [110]. Several preclinical studies suggest that epigenetic reprogramming enhances immune recognition and response against cancer cells and reverse immune evasion [111, 112]. HDACi and DNMTis significantly augment the effector T-cell tumor-infiltration by removing or inhibiting myeloid-deprived suppressor cells (MDSC) and other immune suppression components [113115].

Preclinical studies demonstrate that combining epigenetic drugs with immunotherapy could lead to alteration of multiple pathways, changing the phenotype of cancer cells and facilitating long-lasting adaptive- and innate- immune responses [111, 113, 116]. The HDACis vorinostat and romidepsin enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma. In vivo experiments with this combined treatment result in nearly complete lung cancer eradication [113]. A mouse-model with colon carcinomas and mammary carcinomas treated by combining azacitidine, entinostat and anti-PD-1 or anti-CTLA-4 therapy revealed a remarkable improvement in treatment outcomes and cure of more than 80% of tumor-bearing mice [116]. Furthermore, epigenetic therapies have been reported to increase tumor antigen expression. Weiser et al. reported that treatment with the DNMTi decitabine alone as well as the sequential treatment with decitabine and the HDACi depsipeptide increase the expression of cancer testis antigen NY-ESO-1 and facilitate the recognition of thoracic cancer cells by CD8+ T-cells specific for NY-ESO-1 [117].

4.3 Clinical combinations of epigenetic therapeutics with immunotherapy

Wrangle et al followed up 6 patients who previously received epigenetic therapy with azacitidine and entinostat within a trial mentioned above [10] and subsequently treated with anti-PD-1 or anti-PD-L1. Of these 6 patients, 3 partial responses and two stable diseases were observed after immune checkpoint blockade [112, 118120]. These recent preclinical and clinical discoveries support the rationale of several clinical combination therapy study designs. Combinations of epigenetic drugs and immunotherapies are currently under investigation in multiple lung cancer trials (Table 2b). To date, only limited outcome data is available from such trials.

Preliminary results of an ongoing trial combining pembrolizumab and entinostat in NSCLC patients and melanoma patients (ENCORE 601) were recently presented as posters at the Society for the Immunotherapy of Cancer Annual Meeting in 2016 and Annual ASCO meeting in 2017 respectively [121, 122]. Out of 22 enrolled NSCLC patients, 17 were evaluable. Of eleven anti-PD-1/PD-L1 naïve patients, one PR, one SD and nine PD were reported. The prior preclinical finding of reduced immunosuppressive myeloid driver suppressor cells and regulatory T-cells could be verified in blood samples of the study patients [116, 121]. Of the remaining six patients who had received prior anti-PD-1/PD-L1 therapy and were now receiving combination therapy, three had SD and the other three had PD. Grade 3/4 treatment-related adverse events included hypophosphatemia (9%), neutropenia (5%), anemia (5%), acute respiratory failure (5%), elevated alkaline phosphatase (5%), and immune-mediated hepatitis (5%) [121]. Syndax Pharmaceuticals recently announced the interim analysis of this trial. The pre-specified objective response threshold to advance into the second stage of the Phase 2 trial was met [123]. At least 2 out of 20 NSCLC patients, previously progressive on anti-PD-1 or anti-PD-L1 therapy or 3 out of 13 NSCLC patients previously naïve to anti-PD-1 or anti-PD-L1 therapy responded objectively, defined as either a PR or complete response (CR) to entinostat/pembrolizumab treatment [123]. Encouraging signals have emerged from preliminary interim analyses, although most clinical trials in this field are still ongoing. The completion of ENCORE 601 and other ongoing trials will provide data to answer weather clinical efficacy could be confirmed for combining epigenetic and immunotherapies (Table 2b).

5. Discussion

This review focuses on epigenetic therapeutics and their impact on novel therapies including immunotherapy of lung cancer. FDA-approval of these drugs for MDS, AML, ALL and lymphomas treatment encouraged the exploration of the efficacy of epigenetic therapy studies in lung cancer patients. The efficacy of monotherapies in lung cancer was very limited; and when higher doses were applied severe toxicities were observed. The discovery of a possible synergistic effect of different groups of epigenetic therapeutics led to multiple lung cancer trials combining DNMTis and HDACis [10, 14] (Table 1). Again, substantial toxicities occurred and led to the early termination of numerous trials. Most of the dual-agent epigenetic therapy trials completed in lung cancer did not result in a survival benefit. Prolonged survival and PFS were achieved in some lung cancer patients in a clinical trial combining azacitidine and entinostat [10].

An additive effect of epigenetic therapies and chemotherapy was found in 21% of patients who subsequently continued with chemotherapies [10]. Several clinical trials were initiated to verify this improved effect. Ramalingam et al. described a trend toward improvement in median PFS and OS in the vorinostat-group [49]. Unfortunately, the synergistic effect was accompanied by added toxicities, which led to death in several patients [49]. Several other trials with the same approach are to be completed in the near future and might shed light on an optimal regimen when epigenetic therapeutics are combined with chemotherapy in lung cancer patients (Table 2a).

Some combined targeted therapy and epigenetic therapy trials were designed before the necessity of EGFR mutation testing for effective targeted therapy was known. Therefore, the clinical impact of these combinations remains unclear. The clinical efficacy of such combinations should be investigated in a preselected cohort of EGFR-mutated NSCLC patients. At least one such trial currently enrolls EGFR-mutant NSCLC patients (Table 2a). Preclinical data suggest that HDACi could reverse the acquired resistance to 3rd generation EGFR inhibitors and ALK inhibitors in NSCLC patients with actionable EGFR or ALK mutations. These findings need to be verified in clinical trials designed to test this strategy. Another promising approach is the combination of epigenetic therapies with radiotherapy. Only a few trials investigated this approach, but the available data suggests a survival benefit and tolerable toxicities.

Immune checkpoint blockade and other emerging immunotherapy are changing the landscape of lung cancer therapeutics. Positive signals from preclinical and clinical NSCLC studies suggested the efficacy of epigenetic therapeutics in combination with immunotherapy. Several clinical NSCLC studies combining HDACis, DNMTis, or both with anti-PD-L1 therapy and anti-PD-1 therapy with or without anti-CTLA-4 are recruiting patients (Table 2b). Preliminary results of some of those studies including ENCORE 601 support that this approach is clinically meaningful. To date, there are still gaps in the understanding of how epigenetic therapeutics can improve the efficacy of immunotherapies. Further understanding of epigenetic modulation not only in cancer cells, but also in the tumor microenvironment and immune system will help to optimize the clinical trial design and lung cancer management.

Epigenetic therapies have the potential for improving outcomes for lung cancer patients. These therapies can impact varieties of genes and pathways in cancer cells as well as other cells. Currently there is no reliable predictive biomarker for epigenetic therapies. Bringing these therapies to treat lung cancer and other cancers will require further studies confirming efficacy, minimizing side effects, and optimizing management. The most encouraging developments come from combination therapy, particularly with immunotherapy. Advancement of our understanding of tumor epigenetics and immunology, insight from previous and ongoing studies, and continuing the search for new ways to optimize treatment regimens will help us integrate epigenetic treatment into real world management of lung cancer and change the outcome of this disease.

On behalf of all authors, the corresponding author states that there is no conflict of interest.

Supplementary Material

40495_2017_110_MOESM1_ESM

Supplementary Table 1 FDA-approved epigenetic drugs

Supplementary Table 2 Epigenetic therapeutics (mono- and combined) in SCLC patients

Supplementary Table 3 Epigenetic therapies combined with radiation therapy in NSCLC patients

Supplementary Table 4 FDA-approved immune checkpoint inhibitors in lung cancer

Footnotes

Disclosure

The authors have no conflicts of interest to declare.

References

  • 1.Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA: a cancer journal for clinicians. 2005;55(2):74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 2.Yang P, Allen MS, Aubry MC, Wampfler JA, Marks RS, Edell ES, et al. Clinical features of 5,628 primary lung cancer patients: experience at Mayo Clinic from 1997 to 2003. Chest. 2005;128(1):452–62. doi: 10.1378/chest.128.1.452. [DOI] [PubMed] [Google Scholar]
  • 3.Carbone DP, Gandara DR, Antonia SJ, Zielinski C, Paz-Ares L. Non-Small-Cell Lung Cancer: Role of the Immune System and Potential for Immunotherapy. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2015;10(7):974–84. doi: 10.1097/JTO.0000000000000551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, et al. Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12(19):5777–85. doi: 10.1158/1078-0432.CCR-06-0669. [DOI] [PubMed] [Google Scholar]
  • 5.https://clinicaltrials.gov/ct2/show/NCT00413075
  • 6.Kummar S, Gutierrez M, Gardner ER, Donovan E, Hwang K, Chung EJ, et al. Phase I trial of MS-275, a histone deacetylase inhibitor, administered weekly in refractory solid tumors and lymphoid malignancies. Clinical cancer research : an official journal of the American Association for Cancer Research. 2007;13(18 Pt 1):5411–7. doi: 10.1158/1078-0432.CCR-07-0791. [DOI] [PubMed] [Google Scholar]
  • 7.Gore L, Rothenberg ML, O'Bryant CL, Schultz MK, Sandler AB, Coffin D, et al. A phase I and pharmacokinetic study of the oral histone deacetylase inhibitor, MS-275, in patients with refractory solid tumors and lymphomas. Clinical cancer research : an official journal of the American Association for Cancer Research. 2008;14(14):4517–25. doi: 10.1158/1078-0432.CCR-07-1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.https://clinicaltrials.gov/ct2/show/NCT00020202?term=NCT00020202&rank=1
  • 9.Traynor AM, Dubey S, Eickhoff JC, Kolesar JM, Schell K, Huie MS, et al. Vorinostat (NSC# 701852) in patients with relapsed non-small cell lung cancer: a Wisconsin Oncology Network phase II study. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2009;4(4):522–6. doi: 10.1097/jto.0b013e3181952478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.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. doi: 10.1158/2159-8290.CD-11-0214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.https://clinicaltrials.gov/ct2/show/NCT01207726?term=NCT01207726&rank=1
  • 12.https://clinicaltrials.gov/ct2/show/NCT01886573?term=NCT01886573&rank=1
  • 13.https://clinicaltrials.gov/ct2/show/NCT01935947?term=NCT01935947&rank=1
  • 14.Chu BF, Karpenko MJ, Liu Z, Aimiuwu J, Villalona-Calero MA, Chan KK, et al. Phase I study of 5-aza-2'-deoxycytidine in combination with valproic acid in non-small-cell lung cancer. Cancer chemotherapy and pharmacology. 2013;71(1):115–21. doi: 10.1007/s00280-012-1986-8. [DOI] [PubMed] [Google Scholar]
  • 15.Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nature reviews Genetics. 2002;3(6):415–28. doi: 10.1038/nrg816. [DOI] [PubMed] [Google Scholar]
  • 16.Cheng X, Blumenthal RM. Mammalian DNA methyltransferases: a structural perspective. Structure. 2008;16(3):341–50. doi: 10.1016/j.str.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Baylin SB, Jones PA. A decade of exploring the cancer epigenome - biological and translational implications. Nature reviews Cancer. 2011;11(10):726–34. doi: 10.1038/nrc3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. The New England journal of medicine. 2003;349(21):2042–54. doi: 10.1056/NEJMra023075. [DOI] [PubMed] [Google Scholar]
  • 19.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128(4):683–92. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.McGrath J, Trojer P. Targeting histone lysine methylation in cancer. Pharmacology & therapeutics. 2015;150:1–22. doi: 10.1016/j.pharmthera.2015.01.002. [DOI] [PubMed] [Google Scholar]
  • 21.Shames DS, Girard L, Gao B, Sato M, Lewis CM, Shivapurkar N, et al. A genome-wide screen for promoter methylation in lung cancer identifies novel methylation markers for multiple malignancies. PLoS medicine. 2006;3(12):e486. doi: 10.1371/journal.pmed.0030486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Belinsky SA, Klinge DM, Dekker JD, Smith MW, Bocklage TJ, Gilliland FD, et al. Gene promoter methylation in plasma and sputum increases with lung cancer risk. Clinical cancer research : an official journal of the American Association for Cancer Research. 2005;11(18):6505–11. doi: 10.1158/1078-0432.CCR-05-0625. [DOI] [PubMed] [Google Scholar]
  • 23.Sorm F, Vesely J. Effect of 5-aza-2'-deoxycytidine against leukemic and hemopoietic tissues in AKR mice. Neoplasma. 1968;15(4):339–43. [PubMed] [Google Scholar]
  • 24.Sorm F, Piskala A, Cihak A, Vesely J. 5-Azacytidine, a new, highly effective cancerostatic. Experientia. 1964;20(4):202–3. doi: 10.1007/BF02135399. [DOI] [PubMed] [Google Scholar]
  • 25.Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and DNA methylation. Cell. 1980;20(1):85–93. doi: 10.1016/0092-8674(80)90237-8. [DOI] [PubMed] [Google Scholar]
  • 26.Wilson VL, Jones PA, Momparler RL. Inhibition of DNA methylation in L1210 leukemic cells by 5-aza-2'-deoxycytidine as a possible mechanism of chemotherapeutic action. Cancer research. 1983;43(8):3493–6. [PubMed] [Google Scholar]
  • 27.Momparler RL, Bouchard J, Onetto N, Rivard GE. 5-aza-2'-deoxycytidine therapy in patients with acute leukemia inhibits DNA methylation. Leukemia research. 1984;8(2):181–5. doi: 10.1016/0145-2126(84)90141-3. [DOI] [PubMed] [Google Scholar]
  • 28.Momparler RL, Bouffard DY, Momparler LF, Dionne J, Belanger K, Ayoub J. Pilot phase I–II study on 5-aza-2'-deoxycytidine (Decitabine) in patients with metastatic lung cancer. Anti-cancer drugs. 1997;8(4):358–68. doi: 10.1097/00001813-199704000-00008. [DOI] [PubMed] [Google Scholar]
  • 29.Momparler RL, Ayoub J. Potential of 5-aza-2'-deoxycytidine (Decitabine) a potent inhibitor of DNA methylation for therapy of advanced non-small cell lung cancer. Lung Cancer. 2001;34(Suppl 4):S111–5. doi: 10.1016/s0169-5002(01)00397-x. [DOI] [PubMed] [Google Scholar]
  • 30.Aparicio A, Eads CA, Leong LA, Laird PW, Newman EM, Synold TW, et al. Phase I trial of continuous infusion 5-aza-2'-deoxycytidine. Cancer chemotherapy and pharmacology. 2003;51(3):231–9. doi: 10.1007/s00280-002-0563-y. [DOI] [PubMed] [Google Scholar]
  • 31.Khan N, Jeffers M, Kumar S, Hackett C, Boldog F, Khramtsov N, et al. Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. The Biochemical journal. 2008;409(2):581–9. doi: 10.1042/BJ20070779. [DOI] [PubMed] [Google Scholar]
  • 32.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293(5532):1074–80. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
  • 33.Taddei A, Maison C, Roche D, Almouzni G. Reversible disruption of pericentric heterochromatin and centromere function by inhibiting deacetylases. Nature cell biology. 2001;3(2):114–20. doi: 10.1038/35055010. [DOI] [PubMed] [Google Scholar]
  • 34.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 35.Watson JD, Baker TA, Gann A, Levine M, Losick R. Molecular Biology of the Gene. Seventh. [Google Scholar]
  • 36.Vendetti FP, Rudin CM. Epigenetic therapy in non-small-cell lung cancer: targeting DNA methyltransferases and histone deacetylases. Expert opinion on biological therapy. 2013;13(9):1273–85. doi: 10.1517/14712598.2013.819337. [DOI] [PubMed] [Google Scholar]
  • 37.Gigek CO, Chen ES, Calcagno DQ, Wisnieski F, Burbano RR, Smith MA. Epigenetic mechanisms in gastric cancer. Epigenomics. 2012;4(3):279–94. doi: 10.2217/epi.12.22. [DOI] [PubMed] [Google Scholar]
  • 38.Osada H, Tatematsu Y, Saito H, Yatabe Y, Mitsudomi T, Takahashi T. Reduced expression of class II histone deacetylase genes is associated with poor prognosis in lung cancer patients. International journal of cancer. 2004;112(1):26–32. doi: 10.1002/ijc.20395. [DOI] [PubMed] [Google Scholar]
  • 39.Minamiya Y, Ono T, Saito H, Takahashi N, Ito M, Mitsui M, et al. Expression of histone deacetylase 1 correlates with a poor prognosis in patients with adenocarcinoma of the lung. Lung Cancer. 2011;74(2):300–4. doi: 10.1016/j.lungcan.2011.02.019. [DOI] [PubMed] [Google Scholar]
  • 40.Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nature reviews Genetics. 2007;8(4):286–98. doi: 10.1038/nrg2005. [DOI] [PubMed] [Google Scholar]
  • 41.Baylin SB, Jones PA. Epigenetic Determinants of Cancer. Cold Spring Harbor perspectives in biology. 2016;8(9) doi: 10.1101/cshperspect.a019505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Cai Y, Geutjes EJ, de Lint K, Roepman P, Bruurs L, Yu LR, et al. The NuRD complex cooperates with DNMTs to maintain silencing of key colorectal tumor suppressor genes. Oncogene. 2014;33(17):2157–68. doi: 10.1038/onc.2013.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shah MH, Binkley P, Chan K, Xiao J, Arbogast D, Collamore M, et al. Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12(13):3997–4003. doi: 10.1158/1078-0432.CCR-05-2689. [DOI] [PubMed] [Google Scholar]
  • 44.Otterson GA, Hodgson L, Pang H, Vokes EE. Phase II study of the histone deacetylase inhibitor Romidepsin in relapsed small cell lung cancer (Cancer and Leukemia Group B 30304) Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2010;5(10):1644–8. doi: 10.1097/JTO.0b013e3181ec1713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, et al. Clinical and molecular responses in lung cancer patients receiving Romidepsin. Clinical cancer research : an official journal of the American Association for Cancer Research. 2008;14(1):188–98. doi: 10.1158/1078-0432.CCR-07-0135. [DOI] [PubMed] [Google Scholar]
  • 46.de Marinis F, Atmaca A, Tiseo M, Giuffreda L, Rossi A, Gebbia V, et al. A phase II study of the histone deacetylase inhibitor panobinostat (LBH589) in pretreated patients with small-cell lung cancer. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2013;8(8):1091–4. doi: 10.1097/JTO.0b013e318293d88c. [DOI] [PubMed] [Google Scholar]
  • 47.Owonikoko TK, Ramalingam SS, Kanterewicz B, Balius TE, Belani CP, Hershberger PA. Vorinostat increases carboplatin and paclitaxel activity in non-small-cell lung cancer cells. International journal of cancer. 2010;126(3):743–55. doi: 10.1002/ijc.24759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kanzaki M, Kakinuma H, Kumazawa T, Inoue T, Saito M, Narita S, et al. Low concentrations of the histone deacetylase inhibitor, depsipeptide, enhance the effects of gemcitabine and docetaxel in hormone refractory prostate cancer cells. Oncology reports. 2007;17(4):761–7. [PubMed] [Google Scholar]
  • 49.Ramalingam SS, Maitland ML, Frankel P, Argiris AE, Koczywas M, Gitlitz B, et al. Carboplatin and Paclitaxel in combination with either vorinostat or placebo for first-line therapy of advanced non-small-cell lung cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28(1):56–62. doi: 10.1200/JCO.2009.24.9094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Jones SF, Infante JR, Thompson DS, Mohyuddin A, Bendell JC, Yardley DA, et al. A phase I trial of oral administration of panobinostat in combination with paclitaxel and carboplatin in patients with solid tumors. Cancer chemotherapy and pharmacology. 2012;70(3):471–5. doi: 10.1007/s00280-012-1931-x. [DOI] [PubMed] [Google Scholar]
  • 51.Waqar SN. Belinostat in Combination with Carboplatin and Paclitaxel in Patients with Chemotherapy-Naive Metastatic Lung Cancer. WCLC 2016. 2016 abstract no 5996. [Google Scholar]
  • 52.Jones DR, Moskaluk CA, Gillenwater HH, Petroni GR, Burks SG, Philips J, et al. Phase I trial of induction histone deacetylase and proteasome inhibition followed by surgery in non-small-cell lung cancer. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2012;7(11):1683–90. doi: 10.1097/JTO.0b013e318267928d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Erasmus JJ, Rohren E, Swisher SG. Prognosis and reevaluation of lung cancer by positron emission tomography imaging. Proceedings of the American Thoracic Society. 2009;6(2):171–9. doi: 10.1513/pats.200806-059LC. [DOI] [PubMed] [Google Scholar]
  • 54.https://clinicaltrials.gov/ct2/show/NCT00901537?term=NCT00901537&rank=1
  • 55.https://clinicaltrials.gov/ct2/show/NCT01478685?term=NCT01478685&rank=1
  • 56.https://clinicaltrials.gov/ct2/show/NCT01090830?term=NCT01090830&rank=1
  • 57.https://clinicaltrials.gov/ct2/show/NCT00907179?term=NCT00907179&rank=1
  • 58.Tarhini AA, Zahoor H, McLaughlin B, Gooding WE, Schmitz JC, Siegfried JM, et al. Phase I trial of carboplatin and etoposide in combination with panobinostat in patients with lung cancer. Anticancer research. 2013;33(10):4475–81. [PMC free article] [PubMed] [Google Scholar]
  • 59.https://clinicaltrials.gov/ct2/show/NCT01336842?term=NCT01336842&rank=1
  • 60.https://clinicaltrials.gov/ct2/show/NCT00702572?term=NCT00702572&rank=1
  • 61.https://clinicaltrials.gov/ct2/show/NCT00697476?term=NCT00697476&rank=1
  • 62.https://clinicaltrials.gov/ct2/show/NCT00423449?term=NCT00423449&rank=1
  • 63.https://clinicaltrials.gov/ct2/show/NCT00565227?term=NCT00565227&rank=1
  • 64.https://clinicaltrials.gov/ct2/show/NCT01413750?term=NCT01413750&rank=1
  • 65.https://clinicaltrials.gov/ct2/show/NCT00473889?term=NCT00473889&rank=1
  • 66.https://clinicaltrials.gov/ct2/show/NCT00094978?term=NCT00094978&rank=1
  • 67.Hoang T, Campbell TC, Zhang C, Kim K, Kolesar JM, Oettel KR, et al. Vorinostat and bortezomib as third-line therapy in patients with advanced non-small cell lung cancer: a Wisconsin Oncology Network Phase II study. Investigational new drugs. 2014;32(1):195–9. doi: 10.1007/s10637-013-9980-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.https://clinicaltrials.gov/ct2/show/NCT00996515?term=NCT00996515&rank=1
  • 69.https://clinicaltrials.gov/ct2/show/NCT01545947?term=NCT01545947&rank=1
  • 70.Anderson JL. trial of the HDAC inhibitor belinostat in combination with erlotinib in patients with non-small cell lung cancer. WCLC 2013. 2013 Poster Session 3(P 3.11 (abstract no. 2369)) [Google Scholar]
  • 71.Witta SE, Jotte RM, Konduri K, Neubauer MA, Spira AI, Ruxer RL, et al. Randomized phase II trial of erlotinib with and without entinostat in patients with advanced non-small-cell lung cancer who progressed on prior chemotherapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30(18):2248–55. doi: 10.1200/JCO.2011.38.9411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.https://clinicaltrials.gov/ct2/show/NCT00738751?term=NCT00738751&rank=1
  • 73.https://clinicaltrials.gov/ct2/show/NCT01005797?term=NCT01005797&rank=1
  • 74.Gerber DE, Boothman DA, Fattah FJ, Dong Y, Zhu H, Skelton RA, et al. Phase 1 study of romidepsin plus erlotinib in advanced non-small cell lung cancer. Lung Cancer. 2015;90(3):534–41. doi: 10.1016/j.lungcan.2015.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.https://clinicaltrials.gov/ct2/show/NCT00503971?term=NCT00503971&rank=1
  • 76.https://clinicaltrials.gov/ct2/show/NCT00251589?term=NCT00251589&rank=1
  • 77.https://clinicaltrials.gov/ct2/show/NCT02151721?term=NCT02151721&rank=1
  • 78.Han JY, Lee SH, Lee GK, Yun T, Lee YJ, Hwang KH, et al. Phase I/II study of gefitinib (Iressa((R))) and vorinostat (IVORI) in previously treated patients with advanced non-small cell lung cancer. Cancer chemotherapy and pharmacology. 2015;75(3):475–83. doi: 10.1007/s00280-014-2664-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.https://clinicaltrials.gov/ct2/show/NCT00635791?term=NCT00635791&rank=1
  • 80.https://clinicaltrials.gov/ct2/show/NCT01628471?term=NCT01628471&rank=1
  • 81.https://clinicaltrials.gov/ct2/show/NCT00037817?term=NCT00037817&rank=1
  • 82.Song H, Li CW, Labaff AM, Lim SO, Li LY, Kan SF, et al. Acetylation of EGF receptor contributes to tumor cell resistance to histone deacetylase inhibitors. Biochemical and biophysical research communications. 2011;404(1):68–73. doi: 10.1016/j.bbrc.2010.11.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tanimoto A, Takeuchi S, Arai S, Fukuda K, Yamada T, Roca X, et al. Histone Deacetylase 3 Inhibition Overcomes BIM Deletion Polymorphism-Mediated Osimertinib Resistance in EGFR-Mutant Lung Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2016 doi: 10.1158/1078-0432.CCR-16-2271. [DOI] [PubMed] [Google Scholar]
  • 84.Fukuda K, Takeuchi S, Katayama R, Nanjo S, Yamada T, Suzuki T, et al. HDAC Inhibition Overcomes Crizotinib-Resistance by Mesenchymal-Epithelial Transition (MET) in EML4-ALK LungCancer Cells. WCLC 2016. 2016 abstract MA07.10. [Google Scholar]
  • 85.Nicholson J, Jevons SJ, Groselj B, Ellermann S, Konietzny R, Kerr M, et al. E3 Ligase cIAP2 Mediates Downregulation of MRE11 and Radiosensitization in Response to HDAC Inhibition in Bladder Cancer. Cancer research. 2017;77(11):3027–39. doi: 10.1158/0008-5472.CAN-16-3232. [DOI] [PubMed] [Google Scholar]
  • 86.Kim JG, Bae JH, Kim JA, Heo K, Yang K, Yi JM. Combination effect of epigenetic regulation and ionizing radiation in colorectal cancer cells. PloS one. 2014;9(8):e105405. doi: 10.1371/journal.pone.0105405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Artacho-Cordon F, Rios-Arrabal S, Olivares-Urbano MA, Storch K, Dickreuter E, Munoz-Gamez JA, et al. Valproic acid modulates radiation-enhanced matrix metalloproteinase activity and invasion of breast cancer cells. International journal of radiation biology. 2015;91(12):946–56. doi: 10.3109/09553002.2015.1087067. [DOI] [PubMed] [Google Scholar]
  • 88.Decker RH, Gettinger SN, Glazer PM, Wilson LD. Vorinostat, a Histone Deacetylase Inhibitor, in Combination with Thoracic Radiotherapy in Advanced Non-small Cell Lung Cancer: A Dose Escalation Study. International Journal of Radiation Oncology. 2011;81(2):2. [Google Scholar]
  • 89.Choi CYH, Wakelee HA, Neal JW, Pinder-Schenck MC, Michael YH-H, Chang SD, et al. Vorinostat And Concurrent Stereotactic Radiosurgery For Non-Small Cell Lung Cancer Brain Metastases: A Phase I Dose Escalation Trial. International Journal of Radiation Oncology. 2017 doi: 10.1016/j.ijrobp.2017.04.041. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 90.Shi W, Lawrence YR, Choy H, Werner-Wasik M, Andrews DW, Evans JJ, et al. Vorinostat as a radiosensitizer for brain metastasis: a phase I clinical trial. Journal of neuro-oncology. 2014;118(2):313–9. doi: 10.1007/s11060-014-1433-2. [DOI] [PubMed] [Google Scholar]
  • 91.Rizvi NA, Mazieres J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. The Lancet Oncology. 2015;16(3):257–65. doi: 10.1016/S1470-2045(15)70054-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. The New England journal of medicine. 2015;373(17):1627–39. doi: 10.1056/NEJMoa1507643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive immunity to cancer. Annual review of immunology. 2011;29:235–71. doi: 10.1146/annurev-immunol-031210-101324. [DOI] [PubMed] [Google Scholar]
  • 94.Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011;480(7378):480–9. doi: 10.1038/nature10673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nature reviews Cancer. 2012;12(4):265–77. doi: 10.1038/nrc3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ostrand-Rosenberg S. Immune surveillance: a balance between protumor and antitumor immunity. Current opinion in genetics & development. 2008;18(1):11–8. doi: 10.1016/j.gde.2007.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science. 2011;331(6024):1565–70. doi: 10.1126/science.1203486. [DOI] [PubMed] [Google Scholar]
  • 98.Shepherd FA, Douillard JY, Blumenschein GR., Jr Immunotherapy for non-small cell lung cancer: novel approaches to improve patient outcome. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2011;6(10):1763–73. doi: 10.1097/JTO.0b013e31822e28fc. [DOI] [PubMed] [Google Scholar]
  • 99.Forde PM, Reiss KA, Zeidan AM, Brahmer JR. What lies within: novel strategies in immunotherapy for non-small cell lung cancer. The oncologist. 2013;18(11):1203–13. doi: 10.1634/theoncologist.2013-0171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Woo EY, Yeh H, Chu CS, Schlienger K, Carroll RG, Riley JL, et al. Cutting edge: Regulatory T cells from lung cancer patients directly inhibit autologous T cell proliferation. J Immunol. 2002;168(9):4272–6. doi: 10.4049/jimmunol.168.9.4272. [DOI] [PubMed] [Google Scholar]
  • 101.Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10. doi: 10.1016/j.immuni.2013.07.012. [DOI] [PubMed] [Google Scholar]
  • 102.Zhang Y, Huang S, Gong D, Qin Y, Shen Q. Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8+ T lymphocytes in human non-small cell lung cancer. Cellular & molecular immunology. 2010;7(5):389–95. doi: 10.1038/cmi.2010.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Chen YB, Mu CY, Huang JA. Clinical significance of programmed death-1 ligand-1 expression in patients with non-small cell lung cancer: a 5-year-follow-up study. Tumori. 2012;98(6):751–5. doi: 10.1700/1217.13499. [DOI] [PubMed] [Google Scholar]
  • 104.Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol. 2011;28(3):682–8. doi: 10.1007/s12032-010-9515-2. [DOI] [PubMed] [Google Scholar]
  • 105.Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clinical cancer research : an official journal of the American Association for Cancer Research. 2004;10(15):5094–100. doi: 10.1158/1078-0432.CCR-04-0428. [DOI] [PubMed] [Google Scholar]
  • 106.Rajan A, Kim C, Heery CR, Guha U, Gulley JL. Nivolumab, anti-programmed death-1 (PD-1) monoclonal antibody immunotherapy: Role in advanced cancers. Human vaccines & immunotherapeutics. 2016;12(9):2219–31. doi: 10.1080/21645515.2016.1175694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. The New England journal of medicine. 2015;373(2):123–35. doi: 10.1056/NEJMoa1504627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255–65. doi: 10.1016/S0140-6736(16)32517-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. The New England journal of medicine. 2015;372(21):2018–28. doi: 10.1056/NEJMoa1501824. [DOI] [PubMed] [Google Scholar]
  • 110.Heninger E, Krueger TE, Lang JM. Augmenting antitumor immune responses with epigenetic modifying agents. Frontiers in immunology. 2015;6:29. doi: 10.3389/fimmu.2015.00029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sigalotti L, Fratta E, Coral S, Maio M. Epigenetic drugs as immunomodulators for combination therapies in solid tumors. Pharmacology & therapeutics. 2014;142(3):339–50. doi: 10.1016/j.pharmthera.2013.12.015. [DOI] [PubMed] [Google Scholar]
  • 112.Wrangle J, Wang W, Koch A, Easwaran H, Mohammad HP, Vendetti F, et al. Alterations of immune response of Non-Small Cell Lung Cancer with Azacytidine. Oncotarget. 2013;4(11):2067–79. doi: 10.18632/oncotarget.1542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zheng H, Zhao W, Yan C, Watson CC, Massengill M, Xie M, et al. HDAC Inhibitors Enhance T-Cell Chemokine Expression and Augment Response to PD-1 Immunotherapy in Lung Adenocarcinoma. Clinical cancer research : an official journal of the American Association for Cancer Research. 2016;22(16):4119–32. doi: 10.1158/1078-0432.CCR-15-2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ma T, Galimberti F, Erkmen CP, Memoli V, Chinyengetere F, Sempere L, et al. Comparing histone deacetylase inhibitor responses in genetically engineered mouse lung cancer models and a window of opportunity trial in patients with lung cancer. Molecular cancer therapeutics. 2013;12(8):1545–55. doi: 10.1158/1535-7163.MCT-12-0933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Peng D, Kryczek I, Nagarsheth N, Zhao L, Wei S, Wang W, et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature. 2015;527(7577):249–53. doi: 10.1038/nature15520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(32):11774–9. doi: 10.1073/pnas.1410626111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Weiser TS, Guo ZS, Ohnmacht GA, Parkhurst ML, Tong-On P, Marincola FM, et al. Sequential 5-Aza-2 deoxycytidine-depsipeptide FR901228 treatment induces apoptosis preferentially in cancer cells and facilitates their recognition by cytolytic T lymphocytes specific for NY-ESO-1. J Immunother. 2001;24(2):151–61. doi: 10.1097/00002371-200103000-00010. [DOI] [PubMed] [Google Scholar]
  • 118.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. The New England journal of medicine. 2012;366(26):2455–65. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. The New England journal of medicine. 2012;366(26):2443–54. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Brahmer JR, Horn L, Antonia SJ, Spigel DR, Gandhi L, Sequist LV, et al. Survival and long-term follow-up of the phase I trial of nivolumab (Anti-PD-1; BMS-936558; ONO-4538) in patients (pts) with previously treated advanced non-small cell lung cancer (NSCLC) ASCO Meeting Abstracts 2013. 2013;31(15_suppl):8030. [Google Scholar]
  • 121.Johnson ML, Adjei AA, Opyrchal M, Ramalingam S, Janne PA, Dominguez G, et al. Dose escalation/confirmation results of ENCORE 601, a phase Ib/II, open-label study of entinostat (ENT) in combination with pembrolizumab (PEMBRO) in patients with non-small cell lung cancer (NSCLC) 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016) 2016 part two: National Harbor(abstract no. 215) [Google Scholar]
  • 122.Johnson ML, Gonzalez R, Opyrchal M, Gabrilovich D, Ordentlich P, Brouwer S, et al. ENCORE 601: A phase II study of entinostat (ENT) in combination with pembrolizumab (PEMBRO) in patients with melanoma. ASCO Annual Meeting. 2017;35 (suppl; abstract no 9529) [Google Scholar]
  • 123.http://www.syndax.com/wp-content/uploads/2017/05/SNDX-General-Releases-5.16.17.pdf
  • 124.Levy BP, Giaccone G, Besse B, Begic D, Wu X, Fandi A, et al. A phase II multicenter, randomized, placebo-controlled, double-blind study of CC-486 plus pembrolizumab (pembro) vs pembro plus placebo (PBO) in previously treated patients (pts) with locally advanced/metastatic non-small cell lung cancer (NSCLC) ASCO Meeting abstract no TPS9107. 2016 [Google Scholar]
  • 125.https://clinicaltrials.gov/ct2/show/NCT02635061?term=NCT02635061&rank=1
  • 126.https://clinicaltrials.gov/ct2/show/NCT02909452?term=NCT02909452&rank=1
  • 127.https://clinicaltrials.gov/ct2/show/NCT02453620?term=NCT02453620&rank=1
  • 128.https://clinicaltrials.gov/ct2/show/NCT02638090?term=NCT02638090&rank=1
  • 129.https://clinicaltrials.gov/ct2/show/NCT01928576?term=NCT01928576&rank=1
  • 130.https://clinicaltrials.gov/ct2/show/NCT02959437?term=NCT02959437&rank=1
  • 131.Kang K, Schrump D, Thomas A, Schalper K, Saunthararajah Y, Velcheti V. Tetrahydrouridine-decitabine for non-cytotoxic epigenetic therapy of NSCLC to enhance immunotherapeutic effect of anti-PD1 in vivo. ASCO Meeting abstract no 11552. 2017 [Google Scholar]
  • 132.https://clinicaltrials.gov/ct2/show/NCT02250326?term=NCT02250326&rank=1
  • 133.https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/50-794_vidaza.cfm
  • 134.https://www.accessdata.fda.gov/drugsatfda_docs/nda/2006/021790s000_dacogentoc.cfm
  • 135.https://www.accessdata.fda.gov/drugsatfda_docs/nda/2006/021991s000_zolinzatoc.cfm
  • 136.https://www.accessdata.fda.gov/drugsatfda_docs/nda/2008/022152_stavzor_toc.cfm
  • 137.https://www.accessdata.fda.gov/drugsatfda_docs/nda/2009/022393s000toc.cfm
  • 138.https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&applno=206256
  • 139.https://clinicaltrials.gov/ct2/show/NCT00702962?term=NCT00702962&rank=1
  • 140.https://clinicaltrials.gov/ct2/show/NCT00662311?term=NCT00662311&rank=1
  • 141.Sul J, Blumenthal GM, Jiang X, He K, Keegan P, Pazdur R. FDA Approval Summary: Pembrolizumab for the Treatment of Patients With Metastatic Non-Small Cell Lung Cancer Whose Tumors Express Programmed Death-Ligand 1. The oncologist. 2016;21(5):643–50. doi: 10.1634/theoncologist.2015-0498. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

40495_2017_110_MOESM1_ESM

Supplementary Table 1 FDA-approved epigenetic drugs

Supplementary Table 2 Epigenetic therapeutics (mono- and combined) in SCLC patients

Supplementary Table 3 Epigenetic therapies combined with radiation therapy in NSCLC patients

Supplementary Table 4 FDA-approved immune checkpoint inhibitors in lung cancer

RESOURCES