Plasma TNF-α and Soluble TNF Receptor Levels after Doxorubicin with or without Co-Administration of Mesna-A Randomized, Cross-Over Clinical Study

John Hayslip, Emily V Dressler, Heidi Weiss, Tammy J Taylor, Mara Chambers, Teresa Noel, Sumitra Miriyala, Jeriel T R Keeney, Xiaojia Ren, Rukhsana Sultana, Mary Vore, D Allan Butterfield, Daret St Clair, Jeffrey A Moscow, John Hayslip, Emily V Dressler, Heidi Weiss, Tammy J Taylor, Mara Chambers, Teresa Noel, Sumitra Miriyala, Jeriel T R Keeney, Xiaojia Ren, Rukhsana Sultana, Mary Vore, D Allan Butterfield, Daret St Clair, Jeffrey A Moscow

Abstract

Purpose: Chemotherapy-induced cognitive impairment (CICI) is a common sequelae of cancer therapy. Recent preclinical observations have suggested that CICI can be mediated by chemotherapy-induced plasma protein oxidation, which triggers TNF-α mediated CNS damage. This study evaluated sodium-2-mercaptoethane sulfonate (Mesna) co-administration with doxorubicin to reduce doxorubicin-induced plasma protein oxidation and resultant cascade of TNF-α, soluble TNF receptor levels and related cytokines.

Methods: Thirty-two evaluable patients were randomized using a crossover design to receive mesna or saline in either the first or second cycle of doxorubicin in the context of a standard chemotherapy regimen for either non-Hodgkin lymphoma or breast cancer. Mesna (360 mg/m2) or saline administration occurred 15 minutes prior and three hours post doxorubicin. Pre-treatment and post-treatment measurements of oxidative stress, TNF-α and related cytokines were evaluated during the two experimental cycles of chemotherapy.

Results: Co-administration of mesna with chemotherapy reduced post-treatment levels of TNF-related cytokines and TNF-receptor 1 (TNFR1) and TNF-receptor 2 (TNFR2) (p = 0.05 and p = 0.002, respectively). Patients with the highest pre-treatment levels of each cytokine and its receptors were the most likely to benefit from mesna co-administration.

Conclusions: The extracellular anti-oxidant mesna, when co-administered during a single cycle of doxorubicin, reduced levels of TNF-α and its receptors after that cycle of therapy, demonstrating for the first time a clinical interaction between mesna and doxorubicin, drugs often coincidentally co-administered in multi-agent regimens. These findings support further investigation to determine whether rationally-timed mesna co-administration with redox active chemotherapy may prevent or reduce the cascade of events that lead to CICI.

Trial registration: clinicaltrials.gov NCT01205503.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Flow of patient enrollment.
Fig 1. Flow of patient enrollment.
Fig 2. Study design.
Fig 2. Study design.
Fig 3. Mean estimates of TNF-α levels…
Fig 3. Mean estimates of TNF-α levels for each time point by mesna and saline groups calculated from linear mixed model adjusting for baseline level, time, group (mesna vs saline), chemotherapy regimen and baseline by group interaction.
Fig 4. Mean estimates of TNF receptor…
Fig 4. Mean estimates of TNF receptor 1 levels for each time point by mesna and saline groups calculated from linear mixed model adjusting for baseline level, time, group (mesna vs saline), chemotherapy regimen and baseline by group interaction.
Fig 5. Mean estimates of TNF receptor…
Fig 5. Mean estimates of TNF receptor 2 levels for each time point by mesna and saline groups calculated from linear mixed model adjusting for baseline level, time, group (mesna vs saline), chemotherapy regimen and baseline by group interaction.

References

    1. Bower JE. Behavioral symptoms in patients with breast cancer and survivors. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2008;26(5):768–77. Epub 2008/02/09. 10.1200/JCO.2007.14.3248
    1. Kannarkat G, Lasher EE, Schiff D. Neurologic complications of chemotherapy agents. Current opinion in neurology. 2007;20(6):719–25. 10.1097/WCO.0b013e3282f1a06e .
    1. Aluise CD, Miriyala S, Noel T, Sultana R, Jungsuwadee P, Taylor TJ, et al. 2-Mercaptoethane sulfonate prevents doxorubicin-induced plasma protein oxidation and TNF-alpha release: implications for the reactive oxygen species-mediated mechanisms of chemobrain. Free radical biology & medicine. 2011;50(11):1630–8. 10.1016/j.freeradbiomed.2011.03.009 .
    1. Joshi G, Sultana R, Tangpong J, Cole MP, St Clair DK, Vore M, et al. Free radical mediated oxidative stress and toxic side effects in brain induced by the anti cancer drug adriamycin: insight into chemobrain. Free radical research. 2005;39(11):1147–54. 10.1080/10715760500143478 .
    1. Tangpong J, Cole MP, Sultana R, Joshi G, Estus S, Vore M, et al. Adriamycin-induced, TNF-alpha-mediated central nervous system toxicity. Neurobiology of disease. 2006;23(1):127–39. 10.1016/j.nbd.2006.02.013 .
    1. Tangpong J, Cole MP, Sultana R, Estus S, Vore M, St Clair W, et al. Adriamycin-mediated nitration of manganese superoxide dismutase in the central nervous system: insight into the mechanism of chemobrain. Journal of neurochemistry. 2007;100(1):191–201. 10.1111/j.1471-4159.2006.04179.x .
    1. Tangpong J, Sompol P, Vore M, St Clair W, Butterfield DA, St Clair DK. Tumor necrosis factor alpha-mediated nitric oxide production enhances manganese superoxide dismutase nitration and mitochondrial dysfunction in primary neurons: an insight into the role of glial cells. Neuroscience. 2008;151(2):622–9. 10.1016/j.neuroscience.2007.10.046
    1. Brock N, Hilgard P, Pohl J, Ormstad K, Orrenius S. Pharmacokinetics and mechanism of action of detoxifying low-molecular-weight thiols. Journal of cancer research and clinical oncology. 1984;108(1):87–97. .
    1. Aluise CD, St Clair D, Vore M, Butterfield DA. In vivo amelioration of adriamycin induced oxidative stress in plasma by gamma-glutamylcysteine ethyl ester (GCEE). Cancer letters. 2009;282(1):25–9. 10.1016/j.canlet.2009.02.047 .
    1. James CA, Mant TG, Rogers HJ. Pharmacokinetics of intravenous and oral sodium 2-mercaptoethane sulphonate (mesna) in normal subjects. British journal of clinical pharmacology. 1987;23(5):561–8.
    1. Benjamin RS, Riggs CE Jr, Bachur NR. Plasma pharmacokinetics of adriamycin and its metabolites in humans with normal hepatic and renal function. Cancer research. 1977;37(5):1416–20. .
    1. Piaggio G, Elbourne D, Schulz KF, Villar J, Pinol AP, Gulmezoglu AM. The reporting of methods for reducing and detecting bias: an example from the WHO Misoprostol Third Stage of Labour equivalence randomised controlled trial. BMC medical research methodology. 2003;3:19 Epub 2003/10/04. 10.1186/1471-2288-3-19
    1. Olivier J, Johnson WD, Marshall GD. The logarithmic transformation and the geometric mean in reporting experimental IgE results: what are they and when and why to use them? Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2008;100(4):333–7. 10.1016/S1081-1206(10)60595-9 .
    1. Senn SS. Cross-over Trials in Clinical Research. 2nd ed: Wiley; 2002.
    1. Richardson RP, Rhyne CD, Fong Y, Hesse DG, Tracey KJ, Marano MA, et al. Peripheral blood leukocyte kinetics following in vivo lipopolysaccharide (LPS) administration to normal human subjects. Influence of elicited hormones and cytokines. Annals of surgery. 1989;210(2):239–45.
    1. el-Yazigi A, Ernst P, al-Rawithi S, Legayada E, Raines DA. Pharmacokinetics of mesna and dimesna after simultaneous intravenous bolus and infusion administration in patients undergoing bone marrow transplantation. Journal of clinical pharmacology. 1997;37(7):618–24. .
    1. Brock N, Pohl J, Stekar J, Scheef W. Studies on the urotoxicity of oxazaphosphorine cytostatics and its prevention—III. Profile of action of sodium 2-mercaptoethane sulfonate (mesna). European journal of cancer & clinical oncology. 1982;18(12):1377–87. .
    1. Hyka N, Dayer JM, Modoux C, Kohno T, Edwards CK 3rd, Roux-Lombard P, et al. Apolipoprotein A-I inhibits the production of interleukin-1beta and tumor necrosis factor-alpha by blocking contact-mediated activation of monocytes by T lymphocytes. Blood. 2001;97(8):2381–9. .
    1. Ganz PA, Bower JE, Kwan L, Castellon SA, Silverman DH, Geist C, et al. Does tumor necrosis factor-alpha (TNF-alpha) play a role in post-chemotherapy cerebral dysfunction? Brain, behavior, and immunity. 2013;30 Suppl:S99–108. 10.1016/j.bbi.2012.07.015

Source: PubMed

3
S'abonner