Metabolomic and BH3 profiling of esophageal cancers: novel assessment methods for precision therapy

R Taylor Ripley, Deborah R Surman, Laurence P Diggs, Jane B Trepel, Min-Jung Lee, Jeremy Ryan, Jeremy L Davis, Seth M Steinberg, Jonathan M Hernandez, Choung Hoang, Cara M Kenney, Colleen D Bond, Tricia F Kunst, Anthony Letai, David S Schrump, R Taylor Ripley, Deborah R Surman, Laurence P Diggs, Jane B Trepel, Min-Jung Lee, Jeremy Ryan, Jeremy L Davis, Seth M Steinberg, Jonathan M Hernandez, Choung Hoang, Cara M Kenney, Colleen D Bond, Tricia F Kunst, Anthony Letai, David S Schrump

Abstract

Background: Esophageal cancers accounted for nearly 16,000 deaths in 2016. The number of patients with esophageal cancers increases every year. Neoadjuvant chemoradiotherapy (nCRT) prior to esophagectomy is a standard treatment for esophageal cancers. The patients who have no residual tumor (pathological complete response (pCR)) at surgery are the most likely to experience long term survival. Accurately determining which patients will have a pCR will improve prognostic information for patients and families, confirm lack of response to nCRT, or avoid surgery if no residual tumor is present. Imaging, endoscopy, and liquid biomarkers have all failed to detect pCR without performing an esophagectomy.

Methods: In this study, we are enrolling patients with esophageal adenocarcinoma and squamous cell carcinoma. Patients will undergo standard evaluation including CT scans, laboratory tests, endoscopy with biopsies, and evaluation by a thoracic surgeon. Tissue biopsy is required for enrollment that will be sent for BH3 profiling and metabolomics. Patients will be treated with standard nCRT followed by surgery. Patients with metastatic disease are not eligible. Surgery at the National Cancer Institute will be minimally-invasive robotic surgery. Patients will remain on study indefinitely with regular clinic visits and imaging tests.

Discussion: The mitochondria are critically involved in the intrinsic pathway apoptosis. Bcl-2 homology domain 3 (BH3) profiling is a technique to measure a cell's susceptibility to apoptosis. BH3 profiling measures the relative interactions of proteins that induce or block apoptosis. The collective balance of these proteins determines whether a cell is near the threshold to undergo apoptosis. If the cell is near this threshold, then the tumor may be more likely to die when treated with nCRT. The mitochondria secrete metabolites that may be detectable as biomarkers. Metabolomics is a global assessment of all metabolite changes that has been performed for detection, monitoring, prognosis, and treatment response in cancers. Stratification of patients based on whether pCR occurs or not may elucidate metabolomic signatures that may be associated with response. We are asking whether BH3 profiling or a metabolomic signature will correlate with tumor death after nCRT for esophageal cancer.

Trial registration: NCT03223662 ; Clinicaltrials.gov. July 21, 2017.

Keywords: BH3 profiling; Esophageal adenocarcinoma; Esophageal squamous cell carcinoma; Metabolomics; Pathologic complete response.

Conflict of interest statement

Authors’ information

Principal Investigator:

R. Taylor Ripley, M.D., National Cancer Institute/CCR, NIH.

10 Center Drive, CRC Room 4–3952.

Bethesda, MD 20892.

Phone: 301–496-2127.

Taylor.Ripley@nih.gov

https://ccr.cancer.gov/Thoracic-and-Gastrointestinal-Oncology-Branch/r-taylor-ripley

Research Nurse/Coordinator:

Cara M. Kenney, RN, OCD, CCR, NCI.

10 Center Drive, CRC Room 4–3752.

Bethesda, MD 20892.

Phone: 240–760-6233.

kenneycara@mail.nih.gov

Ethics approval and consent to participate

The ‘Metabolomic and BH3 Profiling of Esophageal Cancers: Identification of Novel Assessment Methods of Treatment Response for Precision Therapy’ trial was approved by the Institutional Review Board (IRB) of the National Cancer Institute, National Institutes of Health, Bethesda, MD.

The consent (‘The Consent to Participate in a Clinical Research Study)’ was approved by the NCI IRB and is giving to all patients prior to enrollment. The consent reference number is based on the intramural trial number, 17-C-0135, and is available as Additional file 1.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30. doi: 10.3322/caac.21332.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29. doi: 10.3322/caac.21254.
    1. van Hagen P, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366(22):2074–2084. doi: 10.1056/NEJMoa1112088.
    1. Davies AR, et al. Tumor stage after neoadjuvant chemotherapy determines survival after surgery for adenocarcinoma of the esophagus and esophagogastric junction. J Clin Oncol. 2014;32(27):2983–2990. doi: 10.1200/JCO.2014.55.9070.
    1. Holscher AH, et al. Prognostic classification of histopathologic response to neoadjuvant therapy in esophageal adenocarcinoma. Ann Surg. 2014;260(5):779–784. doi: 10.1097/SLA.0000000000000964.
    1. Berger AC, et al. Complete response to neoadjuvant chemoradiotherapy in esophageal carcinoma is associated with significantly improved survival. J Clin Oncol. 2005;23(19):4330–4337. doi: 10.1200/JCO.2005.05.017.
    1. Chirieac LR, et al. Posttherapy pathologic stage predicts survival in patients with esophageal carcinoma receiving preoperative chemoradiation. Cancer. 2005;103(7):1347–1355. doi: 10.1002/cncr.20916.
    1. Donohoe CL, Ryan AM, Reynolds JV. Cancer cachexia: mechanisms and clinical implications. Gastroenterol Res Pract. 2011;2011:601434. doi: 10.1155/2011/601434.
    1. Shaikh T, et al. Increased time from neoadjuvant chemoradiation to surgery is associated with higher pathologic complete response rates in esophageal cancer. Ann Thorac Surg. 2015;99(1):270–276. doi: 10.1016/j.athoracsur.2014.08.033.
    1. van Rossum PS, et al. The incremental value of subjective and quantitative assessment of 18F-FDG PET for the prediction of pathologic complete response to preoperative Chemoradiotherapy in esophageal Cancer. J Nucl Med. 2016;57(5):691–700. doi: 10.2967/jnumed.115.163766.
    1. Piessen G, et al. Is there a role for surgery for patients with a complete clinical response after chemoradiation for esophageal cancer? An intention-to-treat case-control study. Ann Surg. 2013;258(5):793–799. doi: 10.1097/SLA.0000000000000228.
    1. Lee JL, et al. A single institutional phase III trial of preoperative chemotherapy with hyperfractionation radiotherapy plus surgery versus surgery alone for resectable esophageal squamous cell carcinoma. Ann Oncol. 2004;15(6):947–954. doi: 10.1093/annonc/mdh219.
    1. Yuan H, et al. PET/CT in the evaluation of treatment response to neoadjuvant chemoradiotherapy and prognostication in patients with locally advanced esophageal squamous cell carcinoma. Nucl Med Commun. 2016;37(9):947–955. doi: 10.1097/MNM.0000000000000527.
    1. Hellmann MD, et al. Pathological response after neoadjuvant chemotherapy in resectable non-small-cell lung cancers: proposal for the use of major pathological response as a surrogate endpoint. Lancet Oncol. 2014;15(1):e42–e50. doi: 10.1016/S1470-2045(13)70334-6.
    1. Bruzzi JF, et al. Detection of interval distant metastases: clinical utility of integrated CT-PET imaging in patients with esophageal carcinoma after neoadjuvant therapy. Cancer. 2007;109(1):125–134. doi: 10.1002/cncr.22397.
    1. Elliott JA, et al. Value of CT-PET after neoadjuvant chemoradiation in the prediction of histological tumour regression, nodal status and survival in oesophageal adenocarcinoma. Br J Surg. 2014;101(13):1702–1711. doi: 10.1002/bjs.9670.
    1. Cheedella NK, et al. Association between clinical complete response and pathological complete response after preoperative chemoradiation in patients with gastroesophageal cancer: analysis in a large cohort. Ann Oncol. 2013;24(5):1262–1266. doi: 10.1093/annonc/mds617.
    1. Nagrath D, et al. Metabolomics for mitochondrial and cancer studies. Biochim Biophys Acta. 2011;1807(6):650–663. doi: 10.1016/j.bbabio.2011.03.006.
    1. Cao DL, et al. Efforts to resolve the contradictions in early diagnosis of prostate cancer: a comparison of different algorithms of sarcosine in urine. Prostate Cancer Prostatic Dis. 2011;14(2):166–172. doi: 10.1038/pcan.2011.2.
    1. Sreekumar A, et al. Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression. Nature. 2009;457(7231):910–914. doi: 10.1038/nature07762.
    1. Yan H, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360(8):765–773. doi: 10.1056/NEJMoa0808710.
    1. Spratlin JL, Serkova NJ, Eckhardt SG. Clinical applications of metabolomics in oncology: a review. Clin Cancer Res. 2009;15(2):431–440. doi: 10.1158/1078-0432.CCR-08-1059.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Certo M, et al. Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell. 2006;9(5):351–365. doi: 10.1016/j.ccr.2006.03.027.
    1. Potter DS, Letai A. To prime, or not to prime: that is the question. Cold Spring Harb Symp Quant Biol. 2016;81:131–140. doi: 10.1101/sqb.2016.81.030841.
    1. Ryan JA, Brunelle JK, Letai A. Heightened mitochondrial priming is the basis for apoptotic hypersensitivity of CD4+ CD8+ thymocytes. Proc Natl Acad Sci U S A. 2010;107(29):12895–12900. doi: 10.1073/pnas.0914878107.
    1. Ni Chonghaile T, et al. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science. 2011;334(6059):1129–1133. doi: 10.1126/science.1206727.
    1. Montero J, et al. Drug-induced death signaling strategy rapidly predicts cancer response to chemotherapy. Cell. 2015;160(5):977–989. doi: 10.1016/j.cell.2015.01.042.
    1. Stachler MD, et al. Paired exome analysis of Barrett's esophagus and adenocarcinoma. Nat Genet. 2015;47(9):1047–1055. doi: 10.1038/ng.3343.
    1. Muller PA, Vousden KH. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell. 2014;25(3):304–317. doi: 10.1016/j.ccr.2014.01.021.
    1. Galluzzi L, et al. Targeting p53 to mitochondria for cancer therapy. Cell Cycle. 2008;7(13):1949–1955. doi: 10.4161/cc.7.13.6222.
    1. Madani K, et al. Prognostic value of p53 mutations in oesophageal adenocarcinoma: final results of a 15-year prospective study. Eur J Cardiothorac Surg. 2010;37(6):1427–1432. doi: 10.1016/j.ejcts.2009.12.018.
    1. Kandioler D, et al. The biomarker TP53 divides patients with neoadjuvantly treated esophageal cancer into 2 subgroups with markedly different outcomes. A p53 research group study. J Thorac Cardiovasc Surg. 2014;148(5):2280–2286. doi: 10.1016/j.jtcvs.2014.06.079.
    1. Bruzzi JF, et al. Detection of Richter's transformation of chronic lymphocytic leukemia by PET/CT. J Nucl Med. 2006;47(8):1267–1273.
    1. Lawrence MS, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499(7457):214–218. doi: 10.1038/nature12213.

Source: PubMed

3
Subscribe