Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Ex vivo drug sensitivity testing as a means for drug repurposing in esophageal adenocarcinoma

  • Ines Lohse,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Writing – original draft, Writing – review & editing

    Affiliations Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Molecular Therapeutics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America

  • Hassan Al-Ali,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Methodology, Resources, Software, Validation, Writing – original draft, Writing – review & editing

    Affiliations Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America, Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Peggy and Harold Katz Drug Discovery Center, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, United States of America

  • Claude-Henry Volmar,

    Roles Investigation, Methodology, Writing – review & editing

    Affiliations Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Molecular Therapeutics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America

  • Annamil D. Alvarez Trotta,

    Roles Investigation, Writing – review & editing

    Affiliations Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America, Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, United States of America

  • Shaun P. Brothers,

    Roles Conceptualization, Data curation, Funding acquisition, Methodology, Resources, Writing – review & editing

    Affiliations Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Molecular Therapeutics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America

  • Anthony J. Capobianco,

    Roles Conceptualization, Funding acquisition, Project administration, Resources, Writing – review & editing

    Affiliations Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America, Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, United States of America

  • Claes Wahlestedt

    Roles Conceptualization, Funding acquisition, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing

    cwahlestedt@med.miami.edu

    Affiliations Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, Florida, United States of America, Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, Florida, United States of America

Abstract

Background

Esophageal cancer remains one of the hardest cancers to treat with rising incidence rates, low overall survival and high levels of treatment resistance. The lack of clinically available biomarkers hinder diagnosis and treatment stratification. While large scale sequencing approaches have uncovered a number of molecular makers, little has translated in the routine treatment of esophageal cancer patients.

Material and methods

We evaluate the treatment response towards a panel of 215 FDA-approved and 163 epigenetic compounds of 4 established and 2 patient-derived esophageal cancer cell lines. Cell viability was evaluated after 72h of treatment using cell titer glow. The drug sensitivity testing results for gemcitabine and cisplatin were validated using clonogenic assays.

Results

The tested cell lines display different drug sensitivity profiles, although we found compounds that display efficacy in all of the tested established or patient-derived cell lines. Clonogenic assays confirmed the validity of the drug sensitivity testing results. Using the epigenetic library, we observed high sensitivity towards a number of epigenetic modifiers.

Discussion

Ex vivo drug sensitivity testing may present a viable option for the treatment stratification of esophageal cancer patients and holds the potential to greatly improve patient outcome while reducing treatment toxicity.

Introduction

Esophageal cancer is the 8th most common cause of cancer death in Western countries with rising incidence rates of adenocarcinoma of the lower esophagus and esophagogastric junction [1].

Although the prognosis for early stage disease is favorable, overall survival remains low at 19% despite advances in surgical techniques and multimodal therapies [1, 2]. Apart from the ERBB2/HER2 status, no biomarkers that complement or improve the diagnosis, risk stratification and therapy of the disease as part of routine clinical practice [3].

A number of recent high-throughput investigations revealed histological, genetic and epigenetic changes typical for esophageal adenocarcinoma [3]. While, these studies have uncovered a number of mutations and revealed an aberrant methylation of the genome [47], little has translated into clinical practice to date. However, a number of clinical trials currently evaluate whether the utility of genomic and epigenetic markers for early detection, prognosis and prediction of response to treatment [46, 8]. Optimal pre-clinical staging based on the American Joint Committee on Cancer (AJCC) Staging System based on tumor-node-metastasis (TNM) sub-classifications [2, 9, 10] remains crucial in optimizing outcomes for patients with esophageal adenocarcinoma [2].

Precision medicine approaches for the treatment stratification of patients with esophageal adenocarcinoma have recently come into the focus of the field in order to improve patient outcome. Apart from improvements in PET imaging techniques [11, 12], the lack of available material from biopsies, long turn over times of sequencing approaches and failure to detect targetable mutations have prevented the clinical application [13].

We have overcome these issues by adapting a drug sensitivity testing (DST) platform previously established for the stratification of patients with relapsed/refractory AML [14]. Ex vivo DST screening allows the identification of patient-specific treatments from a library of 215 FDA-approved compounds that are available on compassionate care and may present a new stratification approach for esophageal adenocarcinoma patients [14].

Material and methods

Esophageal cell lines

OE33, OE19 and SKGT2 human esophageal adenocarcinoma cell lines were obtained from the European Collection of Cell Culture (ATCC). Flo1, EAC42 and EAC47 cells were established in the Capobianco Laboratory. EAC42 and EAC47 human esophageal adenocarcinoma cell lines are derived from patients undergoing surgery at the Miller School of Medicine, University of Miami [15]. The Capobianco laboratory obtained written informed consent from all patients and approval from the Institutional Research Ethics Committee.

The normal esophageal epithelium cell line EACN42 was established in the Capobianco laboratory. The cell lines was derived from human normal epithelium and immortalized using HPV E6-E7.

FDA library drug sensitivity testing

Drug sensitivity testing (DST) was performed as described previously [14]. Briefly, a range of 215 FDA/EMA (Food and Drug Administration/European Medicines Agency)-approved anti-cancer drugs were represented in the compound library, covering a variety of targets and pathways relevant to cancer in general and esophageal cancer specifically (Table 1). Individual drugs were dissolved in 100% DMSO and tested in duplicates starting at a maximal test concentration of 10μM then over a 20,000-fold concentration range to generate dose response curves. Wells with assay buffer containing DMSO served as negative controls. 1000 exponentially growing cells were seeded per well in 384-well micro-titer plates and incubated in the presence of compounds in a humidified environment at 37°C and 5% CO2. After 72 hours of treatment, cell viability was assessed by measuring ATP levels via bioluminescence (CellTiter-Glo, Promega) according to manufactures recommendations. Interpretation of curve parameters was performed according to the modified drug sensitivity scoring (DSSmod) function developed by Swords et al [14]. As a final step, the selective DSSmod (sDSSmod) for each drug in each patient screen is calculated according to the formula sDSSmod = DSSmod (patient cells)—DSSmod (normal cells). Given in this way, the sDSSmod incorporated information on each drug’s potency, efficacy, effect range and therapeutic index, making it possible to prioritize compounds over multiple parameters using a single numerical metric. In addition, this methodology allows us to rank compounds by cancer-selective efficacy for each individual patient. For example, a large positive sDSSmod means that a compound is highly selective for esophageal cancer cells over normal cells in a given sample (favorable scenario), while a large negative score means that the effect was preferential for normal cells (unfavorable scenario). All calculations and scoring routines are implemented in MatLab and additional curve fitting and statistical analyses are performed in GraphPad prism.

thumbnail
Table 1. Compound library for ex vivo drug sensitivity screening.

All listed agents are FDA-approved and classified according to mechanism of action where available.

https://doi.org/10.1371/journal.pone.0203173.t001

Clonogenic assay

Clonogenic assays were performed as described previously [16]. Briefly, the patient-derived cell lines EAC42 and EAC47 were treated with 100nM gemcitabine and 50μM or 10 μM cisplatin for 2h and 24h respectively. Colony formation was evaluated 14 days after seeding. All experiments were performed in triplicates and two biological replicates. Statistical analyses were performed in GraphPad prism.

Epigenetic compound library screen

We tested a 163 compound epigenetic library that consists of inhibitors and activators of epigenetic modifying enzymes (writers, erasers and readers). All compounds were dissolved in 100% DMSO and tested in duplicate at a nominal test concentration of 6μM. Wells with assay buffer containing DMSO served as negative controls. 1000 exponentially growing cells were seeded per well in 384-well micro-titer plates and incubated in the presence of compounds in a humidified environment at 37°C and 5% CO2. After 72 hours of treatment, cell viability was assessed by measuring ATP levels via bioluminescence (CellTiter-Glo, Promega). Positive hits were defined as any compound that showed cell killing higher that 3 standard deviations of the negative control. Curve fitting and statistical analyses are performed in GraphPad prism.

Results

Drug sensitivity testing of normal esophageal cells

The normal esophageal epithelium cell line EACN42 was exposed to the 215 compound FDA-approved library cell viability was evaluated after 72 of treatment. The esophageal epithelial cells display toxicity to 71 compounds (Fig 1A). Significant responses (DSS ≥5), however, were only observed in response to treatment with 34 compounds. The highest toxicity was observed in cells treated with topoisomerase 1/2 inhibitors (Camptothecin (DSS 52.69), Idarubicin (DSS 26.1), Topotecan (DSS 43.21)) and antimitotics (Docetaxel (DSS 40.14), Paclitaxel (DSS 34.94), Vincristine (DSS 26.09), Cephalomannine (DSS 23.35)) and antimetabolites (Flurada (DSS 46.34), Fludarabine (DSS 42.19)). Immunomodulators (Aspirin, Maraviroc) the other hand display low or no activity (S1 Table). Similarly, alkylating agents such as Cisplatin, Carboplatin and Oxaliplatin only displayed minimal treatment effects.

thumbnail
Fig 1. Drug sensitivity profile of the normal control cell line.

(A) Bar graphs of treatment responses of the normal esophageal epithelium cell line EACN42. (B) Comparison of response towards the FDA-approved compound library of patient-derived white blood cells and normal esophageal epithelium.

https://doi.org/10.1371/journal.pone.0203173.g001

Because matched normal tissue is not available at sufficient quantities from patients with esophageal adenocarcinoma from either biopsies or surgical intervention, we compared the response of the esophageal epithelium to the treatment response of white blood cells derived from a healthy donor (Fig 1B). While we do observe a correlation in treatment response in a small number of compounds including rapamycin, gemcitabine, capcecitabine and vinblastine, the majority of compounds display vastly different treatment responses.

Drug sensitivity testing of esophageal cancer cell lines

Drug sensitivity of 4 established and 2 low passage patient-derived esophageal cancer cell lines towards a panel of 215 FDA-approved compounds was established using the DST platform.

Similar to what we have previously observed in AML [14], the 4 tested established esophageal cancer cell lines (OE19, OE33, Flo-1, SK-GT2) display vastly different drug sensitivity profiles (Fig 2, S1 Fig). Nevertheless, we identified a small subset of compounds that displayed activity in all of the cell lines (Belinostat, Carfilzomib, Panbinostat, Bleomycin, Bortezomib, Dasatinib, Rapamycin (Sirolimus) and Temsirolimus) although the magnitude of response was different between cell lines (Fig 2A, S2 Table). The HDAC inhibitor Panbinostat and the proteasome inhibitors Bortezomib and Carfilzomib display the highest treatment efficacy in all cell lines.

thumbnail
Fig 2. Ex vivo drug sensitivity testing.

(A) The heatmap of sDSSmod profiles reveals large variability in both direction and magnitude of drug responses in the esophageal adenocarcinoma cell lines tested. The sDSSmod profile for each cell line is depicted with all drugs that had a score of more than +5 or less than -5 in at least one cell line (drugs that had no effect in any cell linewere excluded). Cell lines and drugs were clustered using hierarchical clustering with a tanimoto distance metric. Red color indicates a positive sDSSmod score while green color indicates a negative sDSSmod score. Bar graphs of clinically actionable drug responses for (B) OE19 and (C) OE33.

https://doi.org/10.1371/journal.pone.0203173.g002

In addition to the established esophageal cancer cell lines, we evaluated EAC42 and EAC47, two patient-derived esophageal cell lines at low passages. Although both lines differ in the magnitude of response, the drug sensitivity profiles are very similar (Figs 2A and 3). The antimethabolites Floxuridine and Methotrexate (Abitrexate) display the highest levels of cancer selective efficacy (S2 Table).

thumbnail
Fig 3. Individual screening results.

Bar graphs of clinically actionable drug responses for (A) EAC42 and (B) EAC47.

https://doi.org/10.1371/journal.pone.0203173.g003

Gemcitabine (Gemzar) was the 3rd and 6th treatment suggestion in EAC42 and EAC47, respectively (Fig 3). Cisplatin on the other hand did not result in a reduction of cell survival in either patient-derived line at a concentration of 10μM. In order to verify the results of the DST screen, we performed clonogenic survival assays using EAC42 and EAC47. The calculated EC50 for gemcitabine of 6.6nM for EAC42 and 10.7nM for EAC47. No EC50 was determined for cisplatin treatment. Treatment with 10nM of gemcitabine for 2h reduced survival to 59.26% in EAC42 and 63.28% in EAC47 (Fig 4A). Treatment with 10μM and 50μM of cisplatin for 24h reduced survival to 89.19% and 50.1%, respectively in EAC42. Treatment of EAC47 reduced survival to 95.75% in response to treatment with 10μM cisplatin and 28.64% in response to treatment with 50μM cisplatin (Fig 4B).

thumbnail
Fig 4. Clonogenic survival assay.

Clonogenic survival in response to treatment with (A) 10nM gemcitabine and (B) 10μM or 50μM of cisplatin. Error bars represent SD.

https://doi.org/10.1371/journal.pone.0203173.g004

Epigenetic compound library screen

OE33, Flo-1 and EAC47 cells were exposed to a 163 compound library containing epigenetic modifiers at a concentration of 6μM. Positive hits were defined as any compound that showed cell killing higher that 3 standard deviations of the negative control.

17 compounds displayed a significant reduction in cell viability in OE33, the majority of which were HDAC inhibitors (Fig 5A). Flo-1 and EAC47 cell viability was reduced by 20 compounds (Fig 5B) and 18 compounds (Fig 5C), respectively. In addition to HDAC inhibitors, Flo-1 and EAC47 display sensitivity towards the BET-inhibitors JQ1 and CPI203 (S3 Table).

thumbnail
Fig 5. Epigenetics compound library screen.

Percent cell killing in response to treatment with the epigenetics compound library in (A) OE33, (B) Flo-1 and (C) EAC47. (D) 10 compounds displayed significant reductions of cell viability in all of the tested cell lines.

https://doi.org/10.1371/journal.pone.0203173.g005

10 compounds displayed a significant reduction of cell survival in all of the tested cell lines (Fig 5D). With the exception of the PLK inhibitor Bl-2536, all common hits are HDAC inhibitors.

Discussion

Treatment stratification of patients with esophageal adenocarcinoma is problematic due to the lack of molecular markers predicting outcome and treatment response, and the optimal patient management remains controversial. While the collection of large sequencing data sets revealed genetic changes, including alterations in DNA methylation patterns [4, 68], these datasets can only supplement the histological classification without providing patient-specific treatment suggestions. Additionally, the aggressive chemotherapy regimen commonly used in esophageal cancer patients come at the price of high normal tissue toxicity. This is especially critical in the light of the high rate of unsuccessful treatment attempts observed in this patient population. Ex vivo DST screening may present a new stratification approach for patients with esophageal adenocarcinoma that allows the identification of patient-specific treatments while reducing normal tissue toxicity [14].

The esophageal adenocarcinoma cell lines display a wide range of treatment responses and vastly different individual sensitivity profiles. In contrast to our previous observations in AML, we uncovered a small subset of compounds that displayed treatment efficacy in all of the tested samples. Specifically, Bortezomib displayed high activity in all lines. The DST screen revealed a number of treatment options not part of standard therapy in esophageal adenocarcinoma, while compounds commonly used in patients such as platinum drugs (cisplatin, carboplatin, oxaliplatin), taxanes (docetaxel, placlitaxel), 5-FU, irinotecan, capecitabine and epirubicin display treatment efficacy in few or none of the lines. However, similar to previous observations, a treatment response can be observed in response to treatment with cisplatin when doses are escalated beyond the maximum dose of the DST screen. Due to high levels of normal tissue toxicity and numerous difficult to manage side effects, a similar dose escalation is not feasible in a clinical setting. The dose range used on the DST screen was chosen to represent a clinically relevant treatment dose. Therefore, a negative result on the DST screen does not imply a complete lack of treatment response. Nevertheless, as seen in the case of cisplatin treatment, the dose needed for a clinically relevant response lies outside of the dose range that is clinically feasible. The data derived from the FDA screen suggests that phenotypic-based treatment stratification may present a way to overcome the current issues with patient stratification and classification.

The impact of epigenetic modifications has recently come into the focus of the esophageal adenocarcinoma community and studies have shown numerous changes in epigenetic marks in patient samples. A small number of studies have provided encouraging results for the use of HDAC inhibitors for the treatment of esophageal cancer [1720]. We observed high levels of treatment responses towards a panel of HDAC inhibitors using an epigenetic library screen containing 163 compounds targeting epigenetic modifiers. This suggests that compounds targeting epigenetic modifiers in esophageal adenocarcinoma should be added to the screening library and may present a promising therapeutic option of sensitive patients.

A pilot trial evaluating a set of patients with esophageal adenocarcinoma will be necessary to examine the clinical feasibility of this precision medicine approach. Clinically the treatment of esophageal adenocarcinoma consist of a multimodal approach. Although our platform evaluates the response towards single treatments only, our experience using the DST platform for the treatment stratification of patients with relapsed/refractory AML [14] suggests that rational combinations can be successfully added to the DST suggestion.

Although all patients receive at least one biopsy, the material obtained during the procedure is essential for diagnostic purposes and the material superfluous to pathological staging is rarely of sufficient quantity and quality to support and ex vivo screen of this magnitude. This can be overcome by the use of surgical specimen which yield sufficient cellular material to support the DST screen. Although a matched normal tissue sample represents the ideal toxicity control for the DST screen, normal esophageal epithelium is not obtainable from either biopsies or surgical samples. White blood cells, while a matched control, display a vastly different sensitivity profile than the esophageal epithelium and cannot be used to evaluate epithelial toxicity in the esophagus. This can be overcome by using a catchment area matched panel of normal esophageal epithelium samples as a normal reference for future patient screens.

This precision medicine approach has the potential to improve treatment stratification and outcome for patients with esophageal adenocarcinoma. Additionally, DST screens on patient populations may allow us further insight into the biology of the disease through the accumulation of sensitivity information on different classes of therapeutic compounds.

Supporting information

S1 Fig. Individual screening results.

Bar graphs of clinically actionable drug responses for (A) Flo1 and (B) SK-GT2.

https://doi.org/10.1371/journal.pone.0203173.s001

(TIF)

S1 Table. DSS and EC50 values for normal esophageal cells.

https://doi.org/10.1371/journal.pone.0203173.s002

(DOCX)

S3 Table. Results of the epigenetic modifier screen.

https://doi.org/10.1371/journal.pone.0203173.s004

(DOCX)

Acknowledgments

SB received support from the National Institutes of Health (R01NS092671 and R01MH110441). CW’s laboratory is currently publically funded by NIH grants DA035592, DA035055 and AA023781, and Florida Department of Health grants 6AZ08 and 7AZ26.

References

  1. 1. Dubecz A, Gall I, Solymosi N, Schweigert M, Peters JH, Feith M, et al. Temporal trends in long-term survival and cure rates in esophageal cancer: a SEER database analysis. J Thorac Oncol. 2012;7(2):443–7. Epub 2011/12/17. pmid:22173700.
  2. 2. Berry MF. Esophageal cancer: staging system and guidelines for staging and treatment. J Thorac Dis. 2014;6 Suppl 3:S289–97. Epub 2014/05/31. pmid:24876933; PubMed Central PMCID: PMCPMC4037413.
  3. 3. Konda VJA, Souza RF. Biomarkers of Barrett's Esophagus: From the Laboratory to Clinical Practice. Dig Dis Sci. 2018. Epub 2018/05/02. pmid:29713984.
  4. 4. Maley CC, Galipeau PC, Finley JC, Wongsurawat VJ, Li X, Sanchez CA, et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat Genet. 2006;38(4):468–73. Epub 2006/03/28. pmid:16565718.
  5. 5. Clement G, Braunschweig R, Pasquier N, Bosman FT, Benhattar J. Methylation of APC, TIMP3, and TERT: a new predictive marker to distinguish Barrett's oesophagus patients at risk for malignant transformation. J Pathol. 2006;208(1):100–7. Epub 2005/11/10. pmid:16278815.
  6. 6. Jin Z, Cheng Y, Gu W, Zheng Y, Sato F, Mori Y, et al. A multicenter, double-blinded validation study of methylation biomarkers for progression prediction in Barrett's esophagus. Cancer Res. 2009;69(10):4112–5. Epub 2009/05/14. pmid:19435894; PubMed Central PMCID: PMCPMC2752375.
  7. 7. Zhang XM, Guo MZ. The value of epigenetic markers in esophageal cancer. Front Med China. 2010;4(4):378–84. Epub 2010/11/26. pmid:21107750.
  8. 8. Hamilton JP, Sato F, Greenwald BD, Suntharalingam M, Krasna MJ, Edelman MJ, et al. Promoter methylation and response to chemotherapy and radiation in esophageal cancer. Clin Gastroenterol Hepatol. 2006;4(6):701–8. Epub 2006/05/09. pmid:16678495.
  9. 9. Rice TW, Blackstone EH, Rusch VW. 7th edition of the AJCC Cancer Staging Manual: esophagus and esophagogastric junction. Ann Surg Oncol. 2010;17(7):1721–4. Epub 2010/04/07. pmid:20369299.
  10. 10. Rice TW, Mason DP, Murthy SC, Zuccaro G Jr., Adelstein DJ, Rybicki LA, et al. T2N0M0 esophageal cancer. J Thorac Cardiovasc Surg. 2007;133(2):317–24. Epub 2007/01/30. pmid:17258554.
  11. 11. Choi J, Kim SG, Kim JS, Jung HC, Song IS. Comparison of endoscopic ultrasonography (EUS), positron emission tomography (PET), and computed tomography (CT) in the preoperative locoregional staging of resectable esophageal cancer. Surg Endosc. 2010;24(6):1380–6. Epub 2009/12/25. pmid:20033712.
  12. 12. Sandha GS, Severin D, Postema E, McEwan A, Stewart K. Is positron emission tomography useful in locoregional staging of esophageal cancer? Results of a multidisciplinary initiative comparing CT, positron emission tomography, and EUS. Gastrointest Endosc. 2008;67(3):402–9. Epub 2008/01/08. pmid:18178202.
  13. 13. Talukdar FR, di Pietro M, Secrier M, Moehler M, Goepfert K, Lima SSC, et al. Molecular landscape of esophageal cancer: implications for early detection and personalized therapy. Ann N Y Acad Sci. 2018. Epub 2018/06/20. pmid:29917250.
  14. 14. Swords RT, Azzam D, Al-Ali H, Lohse I, Volmar CH, Watts JM, et al. Ex-vivo sensitivity profiling to guide clinical decision making in acute myeloid leukemia: A pilot study. Leuk Res. 2018;64:34–41. Epub 2017/11/28. pmid:29175379; PubMed Central PMCID: PMCPMC5756519.
  15. 15. Wang Z, Da Silva TG, Jin K, Han X, Ranganathan P, Zhu X, et al. Notch signaling drives stemness and tumorigenicity of esophageal adenocarcinoma. Cancer Res. 2014;74(21):6364–74. Epub 2014/08/29. pmid:25164006; PubMed Central PMCID: PMCPMC4527315.
  16. 16. Rafehi H, Orlowski C, Georgiadis GT, Ververis K, El-Osta A, Karagiannis TC. Clonogenic assay: adherent cells. J Vis Exp. 2011;(49). Epub 2011/03/30. pmid:21445039; PubMed Central PMCID: PMCPMC3197314.
  17. 17. Ahrens TD, Timme S, Hoeppner J, Ostendorp J, Hembach S, Follo M, et al. Selective inhibition of esophageal cancer cells by combination of HDAC inhibitors and Azacytidine. Epigenetics. 2015;10(5):431–45. Epub 2015/04/30. pmid:25923331; PubMed Central PMCID: PMCPMC4623041.
  18. 18. Ma J, Guo X, Zhang S, Liu H, Lu J, Dong Z, et al. Trichostatin A, a histone deacetylase inhibitor, suppresses proliferation and promotes apoptosis of esophageal squamous cell lines. Mol Med Rep. 2015;11(6):4525–31. Epub 2015/01/31. pmid:25634603.
  19. 19. Furutani A, Sowa Y, Fujiwara H, Otsuji E, Sakai T. The novel HDAC inhibitor OBP-801/YM753 enhances the effects of 5-fluorouracil with radiation on esophageal squamous carcinoma cells. Oncol Res. 2014;21(5):281–6. Epub 2014/05/24. pmid:24854104.
  20. 20. Zhang B, Wang Y, Pang X. Enhanced radiosensitivity of EC109 cells by inhibition of HDAC1 expression. Med Oncol. 2012;29(1):340–8. Epub 2010/05/14. pmid:20464640.