Diurnal cortisol and survival in epithelial ovarian cancer

Andrew Schrepf, Premal H Thaker, Michael J Goodheart, David Bender, George M Slavich, Laila Dahmoush, Frank Penedo, Koen DeGeest, Luis Mendez, David M Lubaroff, Steven W Cole, Anil K Sood, Susan K Lutgendorf, Andrew Schrepf, Premal H Thaker, Michael J Goodheart, David Bender, George M Slavich, Laila Dahmoush, Frank Penedo, Koen DeGeest, Luis Mendez, David M Lubaroff, Steven W Cole, Anil K Sood, Susan K Lutgendorf

Abstract

Introduction: Hypothalamic-pituitary-adrenal (HPA) deregulation is commonly observed in cancer patients, but its clinical significance is not well understood. We prospectively examined the association between HPA activity, tumor-associated inflammation, and survival in ovarian cancer patients prior to treatment.

Materials and methods: Participants were 113 women with ovarian cancer who provided salivary cortisol for three days prior to treatment for calculation of cortisol slope, variability, and night cortisol. Cox proportional hazard regression analyses were used to examine associations between cortisol and survival in models adjusting for disease stage, tumor grade, cytoreduction and age. On a subsample of 41 patients with advanced disease ascites fluid was assayed for levels of interleukin-6 (IL-6) and correlated with cortisol variables.

Results: Each cortisol measure was associated with decreased survival time, adjusting for covariates (all p<.041). A one standard deviation increase in night cortisol was associated with a 46% greater likelihood of death. Patients in the high night cortisol group survived an estimated average of 3.3 years compared to 7.3 years for those in the low night cortisol group. Elevated ascites IL-6 was associated with each cortisol measure (all r>36, all p<.017).

Discussion: Abnormal cortisol rhythms assessed prior to treatment are associated with decreased survival in ovarian cancer and increased inflammation in the vicinity of the tumor. HPA abnormalities may reflect poor endogenous control of inflammation, dysregulation caused by tumor-associated inflammation, broad circadian disruption, or some combination of these factors. Nocturnal cortisol may have utility as a non-invasive measure of HPA function and/or disease severity.

Keywords: Biological markers; Chronobiology disorders; Hydrocortisone; Inflammation; Ovarian neoplasms.

Copyright © 2015 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Flow chart of patient recruitment.
Figure 2
Figure 2
Kaplan Meier survival curve for patients with high nocturnal cortisol (median 3.3 years, 95% CI=2.6, 3.8 years) vs. patients with low nocturnal cortisol (7.3 years, 95% CI =3.8, 10.8 years). Cox regression adjusted for covariates indicates that patients with lower nocturnal cortisol had longer survival times (p=.021).
Figure 3
Figure 3
Kaplan Meier survival curve for patients with flat diurnal cortisol slope (median 3.2 years, 95% CI=2.1, 4.3 years) vs. patients with steep diurnal cortisol slope (6.8 years, 95% CI =4.5, 9.0 years). Cox regression adjusted for covariates indicates that patients with steeper cortisol slopes had longer survival times (p=.040).
Figure 4
Figure 4
Scatterplot of Interleukin-6 assayed in ascites fluid and diurnal cortisol variability.

References

    1. American Cancer Society: Cancer Facts & Figures. Atlanta, GA: American Cancer Society; 2014. Available from URL: .
    1. Bristow RE, Tomacruz RS, Armstrong DK, Trimble EL, Montz FJ. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J. Clin. Oncol. 2002;20:1248–1259.
    1. Burger RA, Grosen EA, Ioli GR, Van Eden ME, Park M, Berman ML, Manetta A, Disaia PJ, Granger GA, Gatanaga T. Spontaneous release of interleukin-6 by primary cultures of lymphoid and tumor cell populations purified from human ovarian carcinoma. J. Interferon Cytokine Res. 1995;15:255–260.
    1. Buysse DJ, Reynolds CF, 3rd, Monk TH, Berman SR, Kupfer DJ. The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research. Psychiatry Res. 1989;28:193–213.
    1. Cella DF, Tulsky DS, Gray G, Sarafian B, Linn E, Bonomi A, Silberman M, Yellen SB, Winicour P, Brannon J, Eckberg K, Lloyd S, Purl S, Blendowski C, Goodman M, Barnicle M, Stewart I, McHale M, Bonomi P, Kaplan E, Taylor SIV, Thomas CR, Jr, Harris J. The Functional Assessment of Cancer Therapy scale: development and validation of the general measure. J Clin Oncol. 1993;11:570–579.
    1. Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N. Engl. J. Med. 1995;332:1351–1362.
    1. Clevenger L, Schrepf A, Degeest K, Bender D, Goodheart M, Ahmed A, Dahmoush L, Penedo F, Lucci J, 3rd, Thaker PH, Mendez L, Sood AK, Slavich GM, Lutgendorf SK. Sleep disturbance, distress, and quality of life in ovarian cancer patients during the first year after diagnosis. Cancer. 2013;119:3234–3241.
    1. Clow A, Hucklebridge F, Stalder T, Evans P, Thorn L. The cortisol awakening response: more than a measure of HPA axis function. Neurosci. Biobehav. Rev. 2010;35:97–103.
    1. Cohen L, Cole SW, Sood AK, Prinsloo S, Kirschbaum C, Arevalo JM, Jennings NB, Scott S, Vence L, Wei Q, Kentor D, Radvanyi L, Tannir N, Jonasch E, Tamboli P, Pisters L. Depressive symptoms and cortisol rhythmicity predict survival in patients with renal cell carcinoma: role of inflammatory signaling. PLoS One. 2012;7:e42324.
    1. Cole SW, Sood AK. Molecular pathways: beta-adrenergic signaling in cancer. Clin. Cancer. Res. 2012;18:1201–1206.
    1. Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav. Immun. 2007;21:153–160.
    1. Dickinson RE, Fegan KS, Ren X, Hillier SG, Duncan WC. Glucocorticoid regulation of SLIT/ROBO tumour suppressor genes in the ovarian surface epithelium and ovarian cancer cells. PLoS One. 2011;6:e27792.
    1. Edgell T, Martin-Roussety G, Barker G, Autelitano DJ, Allen D, Grant P, Rice GE. Phase II biomarker trial of a multimarker diagnostic for ovarian cancer. J. Cancer Res. Clin. Oncol. 2010;136:1079–1088.
    1. Flint MS, Kim G, Hood BL, Bateman NW, Stewart NA, Conrads TP. Stress hormones mediate drug resistance to paclitaxel in human breast cancer cells through a CDK-1-dependent pathway. Psychoneuroendocrinology. 2009;34:1533–1541.
    1. Innominato PF, Roche VP, Palesh OG, Ulusakarya A, Spiegel D, Lévi FA. The circadian timing system in clinical oncology. Ann. Med. 2014;46:191–207.
    1. Jeannin P, Duluc D, Delneste Y. IL-6 and leukemia-inhibitory factor are involved in the generation of tumor-associated macrophage: regulation by IFN-γ. Immunotherapy. 2011;3:23–26.
    1. Jehn CF, Kuehnhardt D, Bartholomae A, Pfeiffer S, Krebs M, Regierer AC, Schmid P, Possinger K, Flath BC. Biomarkers of depression in cancer patients. Cancer. 2006;107:2723–2729.
    1. Kirschbaum C, Hellhammer DH. Salivary cortisol in psychoneuroendocrine research: recent developments and applications. Psychoneuroendocrinology. 1994;19:313–333.
    1. Kraemer HC, Giese-Davis J, Yutsis M, O'Hara R, Neri E, Gallagher-Thompson D, Taylor CB, Spiegel D. Design decisions to optimize reliability of daytime cortisol slopes in an older population. Am. J. Geriatr. Psychiatry. 2006;14:325–33.
    1. Kulbe H, Chakravarty P, Leinster DA, Charles KA, Kwong J, Thompson RG, Coward JI, Schioppa T, Robinson SC, Gallagher WM, Galletta L, Australian Ovarian Cancer Study Group. Salako MA, Smyth JF, Hagemann T, Brennan DJ, Bowtell DD, Balkwill FR. A dynamic inflammatory cytokine network in the human ovarian cancer microenvironment. Cancer Res. 2012;72:66–75.
    1. Kurman RJ. Origin and molecular pathogenesis of ovarian high-grade serous carcinoma. Ann. Oncol. 2013;(Suppl 10):x16–x21.
    1. Lamkin DM, Lutgendorf SK, Lubaroff D, Sood AK, Beltz TG, Johnson AK. Cancer induces inflammation and depressive-like behavior in the mouse: modulation by social housing. Brain Behav. Immun. 2011;25:555–564.
    1. Lane D, Matte I, Rancourt C, Piché A. Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients. BMC Cancer. 2011;11:210.
    1. Lutgendorf SK, Weinrib AZ, Penedo F, Russell D, DeGeest K, Costanzo ES, Henderson PJ, Sephton SE, Rohleder N, Lucci JA, 3rd, Cole SW, Sood AK, Lubaroff DM. Interleukin-6, cortisol, and depressive symptoms in ovarian cancer patients. J. Clin. Oncol. 2008;26:4820–4827.
    1. Mavroudis PD, Scheff JD, Calvano SE, Lowry SF, Androulakis IP. Entrainment of peripheral clock genes by cortisol. Physiol. Genomics. 2012;44:607–621.
    1. Obata NH, Tamakoshi K, Shibata K, Kikkawa F, Tomoda Y. Effects of interleukin-6 on in vitro cell attachment, migration and invasion of human ovarian carcinoma. Anticancer Res. 1997;17:337–342.
    1. Offner FA, Obrist P, Stadlmann S, Feichtingera H, Klinglerb P, Heroldc M, Zwierzinac H, Hittmaira A, Mikuza G, Abendsteind B, Zeimete A, Marth C. IL-6 secretion by human peritoneal mesothelial and ovarian cancer cells. Cytokine. 1995;7:542–547.
    1. Pace TW, Miller AH. Cytokines and glucocorticoid receptor signaling. Relevance to major depression. Ann. N. Y. Acad. Sci. 2009;1179:86–105.
    1. Radloff LS. The CES-D scale: a self-report depression scale for research in the general population. Appl. Psychol. Meas. 1977;1:385–401.
    1. Reinartz S, Schumann T, Finkernagel F, Wortmann A, Jansen JM, Meissner W, Krause M, Schwörer AM, Wagner U, Müller-Brüsselbach S, Müller R. Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma: correlation of CD163 expression, cytokine levels and early relapse. Int. J. Cancer. 2014;134:32–42.
    1. Rhen T, Cidlowski JA. Antiinflammatory action of glucocorticoids--new mechanisms for old drugs. N. Engl. J. Med. 2005;353:1711–1723.
    1. Rich T, Innominato PF, Boerner J, Mormont MC, Iacobelli S, Baron B, Jasmin C, Lévi F. Elevated serum cytokines correlated with altered behavior, serum cortisol rhythm, and dampened 24-hour rest-activity patterns in patients with metastatic colorectal cancer. Clin. Cancer Res. 2005;11:1757–1764.
    1. Schauer IG, Zhang J, Xing Z, Guo X, Mercado-Uribe I, Sood AK, Huang P, Liu J. Interleukin-1β promotes ovarian tumorigenesis through a p53/NF-κB-mediated inflammatory response in stromal fibroblasts. Neoplasia. 2013;15:409–420.
    1. Schrepf A, Clevenger L, Christensen D, DeGeest K, Bender D, Ahmed A, Goodheart MJ, Dahmoush L, Penedo F, Lucci JA, 3rd, Ganjei-Azar P, Mendez L, Markon K, Lubaroff DM, Thaker PH, Slavich GM, Sood AK, Lutgendorf SK. Cortisol and inflammatory processes in ovarian cancer patients following primary treatment: relationships with depression, fatigue, and disability. Brain Behav. Immun. 2013;(30 Suppl):S126–S134.
    1. Sephton SE, Sapolsky RM, Kraemer HC, Spiegel D. Diurnal cortisol rhythm as a predictor of breast cancer survival. J. Natl. Cancer Inst. 2000;92:994–1000.
    1. Sephton SE, Lush E, Dedert EA, Floyd AR, Rebholz WN, Dhabhar FS, Spiegel D, Salmon P. Diurnal cortisol rhythm as a predictor of lung cancer survival. Brain Behav. Immun. 2013;(30 Suppl):S163–S70.
    1. Silverman MN, Pearce BD, Biron CA, Miller AH. Immune modulation of the hypothalamic-pituitary-adrenal (HPA) axis during viral infection. Viral Immunol. 2005;18:41–78.
    1. Silverman MN, Sternberg EM. Glucocorticoid regulation of inflammation and its functional correlates: from HPA axis to glucocorticoid receptor dysfunction. Ann. N.Y. Acad. Sci. 2012;1261:55–63.
    1. Singer JD, Willett JB. Applied Longitudinal Data Analysis: Modeling Change and Event Occurrence. New York: Oxford University Press; 2003.
    1. Sood AK, Buller RE. Drug resistance in ovarian cancer: from the laboratory to the clinic. Obstet Gynecol. 1998;92:312–319.
    1. Stone AA, Schwartz JE, Smyth J, Kirschbaum C, Cohen S, Hellhammer D, Grossman S. Individual differences in the diurnal cycle of salivary free cortisol: a replication of flattened cycles for some individuals. Psychoneuroendocrinology. 2001;26:295–306.
    1. Tokunaga H, Takebayashi Y, Utsunomiya H, Akahira J, Higashimoto M, Mashiko M, Ito K, Niikura H, Takenoshita S, Yaegashi N. Clinicopathological significance of circadian rhythm-related gene expression levels in patients with epithelial ovarian cancer. Acta. Obstet. Gynecol. Scand. 2008;87:1060–1070.
    1. Tsigos C, Chrousos GP. Hypothalamic-pituitary-adrenal axis, neuroendocrine factors and stress. J. Psychosom. Res. 2002;53:865–871.
    1. Volden PA, Conzen SD. The influence of glucocorticoid signaling on tumor progression. Brain Behav. Immun. 2013;(30 Suppl):S26–S31.
    1. Weinrib AZ, Sephton SE, Degeest K, Penedo F, Bender D, Zimmerman B, Kirschbaum C, Sood AK, Lubaroff DM, Lutgendorf SK. Diurnal cortisol dysregulation, functional disability, and depression in women with ovarian cancer. Cancer. 2010;116:4410–4419.
    1. Yang X, Wood PA, Oh EY, Du-Quiton J, Ansell CM, Hrushesky WJ. Down regulation of circadian clock gene Period 2 accelerates breast cancer growth by altering its daily growth rhythm. Breast Cancer Res. Treat. 2009;117:423–431.
    1. Yang M, Kim J, Kim JS, Kim SH, Kim JC, Kang MJ, Jung U, Shin T, Wang H, Moon C. Hippocampal dysfunctions in tumor-bearing mice. Brain Behav. Immun. 2014;36:147–55.
    1. Zhang C, Marmé A, Wenger T, Gutwein P, Edler L, Rittgen W, Debatin KM, Altevogt P, Mattern J, Herr I. Glucocorticoid-mediated inhibition of chemotherapy in ovarian carcinomas. Int J Oncol. 2006;28:551–558.

Source: PubMed

3
Sottoscrivi