The chemo-gut study: investigating the long-term effects of chemotherapy on gut microbiota, metabolic, immune, psychological and cognitive parameters in young adult Cancer survivors; study protocol

Julie M Deleemans, Faye Chleilat, Raylene A Reimer, Jan-Willem Henning, Mohamad Baydoun, Katherine-Ann Piedalue, Andrew McLennan, Linda E Carlson, Julie M Deleemans, Faye Chleilat, Raylene A Reimer, Jan-Willem Henning, Mohamad Baydoun, Katherine-Ann Piedalue, Andrew McLennan, Linda E Carlson

Abstract

Background: The gut microbiota is an important modulator of immune, metabolic, psychological and cognitive mechanisms. Chemotherapy adversely affects the gut microbiota, inducing acute dysbiosis, and alters physiological and psychological function. Cancer among young adults has risen 38% in recent decades. Understanding chemotherapy's long-term effects on gut microbiota and psycho-physiological function is critical to improve survivors' physical and mental health, but remains unexamined. Restoration of the gut microbiota via targeted therapies (e.g. probiotics) could potentially prevent or reverse the psycho-physiological deficits often found in young survivors following chemotherapy, ultimately leading to reduced symptom burden and improved health.

Methods: This longitudinal study investigates chemotherapy induced long-term gut dysbiosis, and associations between gut microbiota, and immune, metabolic, cognitive and psychological parameters using data collected at < 2 month (T1), 3-4 months (T2), and 5-6 months (T3) post-chemotherapy. Participants will be 18-39 year old blood or solid tumor cancer survivors (n = 50), and a healthy sibling, partner or friend as a control (n = 50). Gut microbiota composition will be measured from fecal samples using 16 s RNA sequencing. Psychological and cognitive patient reported outcome measures will include depression, anxiety, post-traumatic stress disorder symptoms, pain, fatigue, and social and cognitive function. Dual-energy X-ray Absorptiometry (DXA) will be used to measure fat and lean mass, and bone mineral concentration. Pro-inflammatory cytokines, C-reactive protein (CRP), lipopolysaccharide (LPS), serotonin, and brain derived neurotrophic factor (BDNF) will be measured in serum, and long-term cortisol will be assayed from hair. Regression and linear mixed model (LMM) analyses will examine associations across time points (T1 - T3), between groups, and covariates with gut microbiota, cognitive, psychological, and physiological parameters.

Conclusion: Knowing what bacterial species are depleted after chemotherapy, how long these effects last, and the physiological mechanisms that may drive psychological and cognitive issues among survivors will allow for targeted, integrative interventions to be developed, helping to prevent or reverse some of the late-effects of treatment that many young cancer survivors face. This protocol has been approved by the Health Research Ethics Board of Alberta Cancer Committee (ID: HREBA.CC-19-0018).

Keywords: Cancer; Chemotherapy; Cortisol; Cytokines; Gut microbiota; Psycho-oncology; Young adult.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Chemotherapy Driven Dysbiosis of the Microbiota-Gut-Brain Axis. The Chemotherapy Driven Dysbiosis of the Microbiota-Gut-Brain Axis model posits that chemotherapy given to young cancer patients induces long-term gut dysbiosis, increasing intestinal permeability (i.e. “leaky gut”), which allows bacterial toxins, such as lipopolysaccharide (LPS), to enter the blood stream. This subsequently leads to systemic inflammation via increases in pro-inflammatory cytokines, especially IL-6, IL-1b, and TNF-a, and C-reactive protein, as well as dysregulation of the HPA-axis. This creates a feedback loop in which inflammatory mechanisms trigger the stress response to increase systemic cortisol, feeding back into the immune system to exacerbate levels of inflammation. Gut microbiota dysbiosis and dysregulation of the immune system and HPA-axis attenuate levels of serotonin (5-HT) and brain derived neurotrophic factor (BDNF), which then work in tandem to induce changes in psychological and cognitive function, including increased symptoms of anxiety and depression (i.e. “sickness behaviours”), pain, fatigue, and social and cognitive deficits. Finally, gut dysbiosis, immune, and HPA-axis dysregulation may also augment patients’ vulnerability to increased adipose tissue, and subsequent overweight and obesity, further compromising their health

References

    1. Petra AI, Panagiotidou S, Hatziagelaki E, Stewart JM, Conti P, Theoharides TC. Gut-microbiota-brain Axis and its effect on neuropsychiatric disorders with suspected immune Dysregulation. Clin Ther. 2015;37(5):984–995. doi: 10.1016/j.clinthera.2015.04.002.
    1. Foster JA, Neufeld KM. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013;36(5):305–312. doi: 10.1016/j.tins.2013.01.005.
    1. Maes M, Kubera M, Leunis JC, Berk M, Geffard M, Bosmans E. In depression, bacterial translocation may drive inflammatory responses, oxidative and nitrosative stress (O&NS), and autoimmune responses directed against O&NS-damaged neoepitopes. Acta Psychiatr Scand. 2013;127(5):344–354. doi: 10.1111/j.1600-0447.2012.01908.x.
    1. Dinan TG, Cryan JF. The microbiome-gut-brain Axis in health and disease. Gastroenterol Clin N Am. 2017;46(1):77–89. doi: 10.1016/j.gtc.2016.09.007.
    1. Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for microbiome-brain-gut axis communication. Adv Exp Med Biol. 2014;817:115–133. doi: 10.1007/978-1-4939-0897-4_5.
    1. Bai J, Behera M, Bruner DW. The gut microbiome, symptoms, and targeted interventions in children with cancer: a systematic review. Support Care Cancer. 2018;26(2):427–439. doi: 10.1007/s00520-017-3982-3.
    1. Jordan KR, Loman BR, Bailey MT, Pyter LM. Gut microbiota-immune-brain interactions in chemotherapy-associated behavioral comorbidities. Cancer. 2018;124(20):3990–3999. doi: 10.1002/cncr.31584.
    1. Jacola LM, Edelstein K, Liu W, Pui CH, Hayashi R, Kadan-Lottick NS, Srivastava D, Henderson T, Leisenring W, Robison LL, et al. Cognitive, behaviour, and academic functioning in adolescent and young adult survivors of childhood acute lymphoblastic leukaemia: a report from the childhood Cancer survivor study. Lancet Psychiatry. 2016;3(10):965–972. doi: 10.1016/S2215-0366(16)30283-8.
    1. Schulte F, Brinkman TM, Li C, Fay-McClymont T, Srivastava DK, Ness KK, Howell RM, Mueller S, Wells E, Strother D, et al. Social adjustment in adolescent survivors of pediatric central nervous system tumors: a report from the childhood Cancer survivor study. Cancer. 2018;124(17):3596–3608. doi: 10.1002/cncr.31593.
    1. Burkhamer J, Kriebel D, Clapp R. The increasing toll of adolescent cancer incidence in the US. PLoS One. 2017;12(2):e0172986. doi: 10.1371/journal.pone.0172986.
    1. Borre YE, O'Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014;20(9):509–518. doi: 10.1016/j.molmed.2014.05.002.
    1. Cancer CPA. Adolescents and young adults with Cancer: system performance report. In. Toronto, Canada. 2017.
    1. Barr RD, Ferrari A, Ries L, Whelan J, Bleyer WA. Cancer in adolescents and young adults: a narrative review of the current status and a view of the future. JAMA Pediatr. 2016;170(5):495–501. doi: 10.1001/jamapediatrics.2015.4689.
    1. Asher A. Cognitive dysfunction among cancer survivors. Am J Phys Med Rehabil. 2011;90(5 Suppl 1):S16–S26. doi: 10.1097/PHM.0b013e31820be463.
    1. Seitz DC, Besier T, Debatin KM, Grabow D, Dieluweit U, Hinz A, Kaatsch P, Goldbeck L. Posttraumatic stress, depression and anxiety among adult long-term survivors of cancer in adolescence. Eur J Cancer. 2010;46(9):1596–1606. doi: 10.1016/j.ejca.2010.03.001.
    1. Grenham S, Clarke G, Cryan JF, Dinan TG. Brain-gut-microbe communication in health and disease. Front Physiol. 2011;2:94. doi: 10.3389/fphys.2011.00094.
    1. Huang Y, Yang W, Liu H, Duan J, Zhang Y, Liu M, Li H, Hou Z, Wu KK. Effect of high-dose methotrexate chemotherapy on intestinal Bifidobacteria, Lactobacillus and Escherichia coli in children with acute lymphoblastic leukemia. Exp Biol Med (Maywood) 2012;237(3):305–311. doi: 10.1258/ebm.2011.011297.
    1. Montassier E, Gastinne T, Vangay P, Al-Ghalith GA, Bruley des Varannes S, Massart S, Moreau P, Potel G, de La Cochetiere MF, Batard E, et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment Pharmacol Ther. 2015;42(5):515–528. doi: 10.1111/apt.13302.
    1. Scully C, Epstein J, Sonis S. Oral mucositis: a challenging complication of radiotherapy, chemotherapy, and radiochemotherapy: part 1, pathogenesis and prophylaxis of mucositis. Head Neck. 2003;25(12):1057–1070. doi: 10.1002/hed.10318.
    1. Dash S, Clarke G, Berk M, Jacka FN. The gut microbiome and diet in psychiatry: focus on depression. Curr Opin Psychiatry. 2015;28(1):1–6. doi: 10.1097/YCO.0000000000000117.
    1. Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav Immun. 2007;21(2):153–160. doi: 10.1016/j.bbi.2006.09.006.
    1. Berk M, Williams LJ, Jacka FN, O'Neil A, Pasco JA, Moylan S, Allen NB, Stuart AL, Hayley AC, Byrne ML, et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013;11:200. doi: 10.1186/1741-7015-11-200.
    1. Capuron L, Gumnick JF, Musselman DL, Lawson DH, Reemsnyder A, Nemeroff CB, Miller AH. Neurobehavioral effects of interferon-alpha in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology. 2002;26(5):643–652. doi: 10.1016/S0893-133X(01)00407-9.
    1. O'Mahony SM, Hyland NP, Dinan TG, Cryan JF. Maternal separation as a model of brain-gut axis dysfunction. Psychopharmacology. 2011;214(1):71–88. doi: 10.1007/s00213-010-2010-9.
    1. Beishuizen A, Thijs LG. Endotoxin and the hypothalamo-pituitary-adrenal (HPA) axis. J Endotoxin Res. 2003;9(1):3–24.
    1. Sonis ST. The pathobiology of mucositis. Nat Rev Cancer. 2004;4(4):277–284. doi: 10.1038/nrc1318.
    1. Schurman JV, Singh M, Singh V, Neilan N, Friesen CA. Symptoms and subtypes in pediatric functional dyspepsia: relation to mucosal inflammation and psychological functioning. J Pediatr Gastroenterol Nutr. 2010;51(3):298–303.
    1. Hilimire MR, DeVylder JE, Forestell CA. Fermented foods, neuroticism, and social anxiety: an interaction model. Psychiatry Res. 2015;228(2):203–208. doi: 10.1016/j.psychres.2015.04.023.
    1. Steenbergen L, Sellaro R, van Hemert S, Bosch JA, Colzato LS. A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav Immun. 2015;48:258–264. doi: 10.1016/j.bbi.2015.04.003.
    1. Clapp M, Aurora N, Herrera L, Bhatia M, Wilen E, Wakefield S. Gut microbiota's effect on mental health: the gut-brain axis. Clin Pract. 2017;7(4):987. doi: 10.4081/cp.2017.987.
    1. Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience. 2010;170(4):1179–1188. doi: 10.1016/j.neuroscience.2010.08.005.
    1. Anderson SC, Cryan JF, Dinan T. The Psychobiotic revolution: mood, food, and the new science of the gut-brain connection. Washington, DC: National Geographic Partners, LLC; 2017.
    1. Janssen DG, Caniato RN, Verster JC, Baune BT. A psychoneuroimmunological review on cytokines involved in antidepressant treatment response. Hum Psychopharmacol. 2010;25(3):201–215. doi: 10.1002/hup.1103.
    1. Sarkar A, Harty S, Lehto SM, Moeller AH, Dinan TG, Dunbar RIM, Cryan JF, Burnet PWJ. The microbiome in psychology and cognitive neuroscience. Trends Cogn Sci. 2018;22(7):611–636. doi: 10.1016/j.tics.2018.04.006.
    1. Vazquez E, Barranco A, Ramirez M, Gruart A, Delgado-Garcia JM, Martinez-Lara E, Blanco S, Martin MJ, Castanys E, Buck R, et al. Effects of a human milk oligosaccharide, 2′-fucosyllactose, on hippocampal long-term potentiation and learning capabilities in rodents. J Nutr Biochem. 2015;26(5):455–465. doi: 10.1016/j.jnutbio.2014.11.016.
    1. Prehn-Kristensen A, Zimmermann A, Tittmann L, Lieb W, Schreiber S, Baving L, Fischer A. Reduced microbiome alpha diversity in young patients with ADHD. PLoS One. 2018;13(7):e0200728. doi: 10.1371/journal.pone.0200728.
    1. Naseribafrouei A, Hestad K, Avershina E, Sekelja M, Linlokken A, Wilson R, Rudi K. Correlation between the human fecal microbiota and depression. Neurogastroenterol Motil. 2014;26(8):1155–1162. doi: 10.1111/nmo.12378.
    1. Prasad PK, Hardy KK, Zhang N, Edelstein K, Srivastava D, Zeltzer L, Stovall M, Seibel NL, Leisenring W, Armstrong GT, et al. Psychosocial and neurocognitive outcomes in adult survivors of adolescent and early young adult Cancer: a report from the childhood Cancer survivor study. J Clin Oncol. 2015;33(23):2545–2552. doi: 10.1200/JCO.2014.57.7528.
    1. Deleemans JM, Reynolds K, Schulte F. Associations among health behaviours and psychosocial outcomes in adolescent and young adult (AYA) cancer survivors. Toronto, Ontario, Canada: Canadian Association of Psychosocial Oncology; 2018.
    1. Tai E, Buchanan N, Townsend J, Fairley T, Moore A, Richardson LC. Health status of adolescent and young adult cancer survivors. Cancer. 2012;118(19):4884–4891. doi: 10.1002/cncr.27445.
    1. Bervoets L, Van Hoorenbeeck K, Kortleven I, Van Noten C, Hens N, Vael C, Goossens H, Desager KN, Vankerckhoven V. Differences in gut microbiota composition between obese and lean children: a cross-sectional study. Gut Pathog. 2013;5(1):10. doi: 10.1186/1757-4749-5-10.
    1. Bomhof MR, Paul HA, Geuking MB, Eller LK, Reimer RA. Improvement in adiposity with oligofructose is modified by antibiotics in obese rats. FASEB J. 2016;30(8):2720–2732. doi: 10.1096/fj.201600151R.
    1. Dudek-Wicher RK, Junka A, Bartoszewicz M. The influence of antibiotics and dietary components on gut microbiota. Prz Gastroenterol. 2018;13(2):85–92.
    1. Mahana D, Trent CM, Kurtz ZD, Bokulich NA, Battaglia T, Chung J, Muller CL, Li H, Bonneau RA, Blaser MJ. Antibiotic perturbation of the murine gut microbiome enhances the adiposity, insulin resistance, and liver disease associated with high-fat diet. Genome Med. 2016;8(1):48. doi: 10.1186/s13073-016-0297-9.
    1. Turnbaugh PJ. Microbes and diet-induced obesity: fast, cheap, and out of control. Cell Host Microbe. 2017;21(3):278–281. doi: 10.1016/j.chom.2017.02.021.
    1. Effect Size (Cohen's d) Calculator for a Student t-Test []. Accessed Feb 2019.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, Huttenhower C. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60. doi: 10.1186/gb-2011-12-6-r60.
    1. Nicolucci AC, Hume MP, Martinez I, Mayengbam S, Walter J, Reimer RA. Prebiotics reduce body fat and Alter intestinal microbiota in children who are overweight or with obesity. Gastroenterology. 2017;153(3):711–722. doi: 10.1053/j.gastro.2017.05.055.
    1. Berruti A, Dogliotti L, Terrone C, Cerutti S, Isaia G, Tarabuzzi R, Reimondo G, Mari M, Ardissone P, De Luca S, et al. Changes in bone mineral density, lean body mass and fat content as measured by dual energy x-ray absorptiometry in patients with prostate cancer without apparent bone metastases given androgen deprivation therapy. J Urol. 2002;167(6):2361–2367. doi: 10.1016/S0022-5347(05)64985-3.
    1. Sandberg E. Meso scale discovery training report. In: The University of Calgary. 2018.
    1. Serotonin ELISA kit []. Accessed Feb 2019.
    1. Steudte S, Kirschbaum C, Gao W, Alexander N, Schonfeld S, Hoyer J, Stalder T. Hair cortisol as a biomarker of traumatization in healthy individuals and posttraumatic stress disorder patients. Biol Psychiatry. 2013;74(9):639–646. doi: 10.1016/j.biopsych.2013.03.011.
    1. Steudte-Schmiedgen S, Kirschbaum C, Alexander N, Stalder T. An integrative model linking traumatization, cortisol dysregulation and posttraumatic stress disorder: insight from recent hair cortisol findings. Neurosci Biobehav Rev. 2016;69:124–135. doi: 10.1016/j.neubiorev.2016.07.015.
    1. Cessna JM, Jim HS, Sutton SK, Asvat Y, Small BJ, Salsman JM, Zachariah B, Fishman M, Field T, Fernandez H, et al. Evaluation of the psychometric properties of the PROMIS Cancer fatigue short form with cancer patients. J Psychosom Res. 2016;81:9–13. doi: 10.1016/j.jpsychores.2015.12.002.
    1. Allan Cheyne J, Solman GJ, Carriere JS, Smilek D. Anatomy of an error: a bidirectional state model of task engagement/disengagement and attention-related errors. Cognition. 2009;111(1):98–113. doi: 10.1016/j.cognition.2008.12.009.
    1. Chen W, Liu F, Ling Z, Tong X, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS One. 2012;7(6):e39743. doi: 10.1371/journal.pone.0039743.
    1. Sears CL, Garrett WS. Microbes, microbiota, and colon cancer. Cell Host Microbe. 2014;15(3):317–328. doi: 10.1016/j.chom.2014.02.007.

Source: PubMed

3
Tilaa