Diarrhea Induced by Small Molecule Tyrosine Kinase Inhibitors Compared With Chemotherapy: Potential Role of the Microbiome

Kate R Secombe, Ysabella Z A Van Sebille, Bronwen J Mayo, Janet K Coller, Rachel J Gibson, Joanne M Bowen, Kate R Secombe, Ysabella Z A Van Sebille, Bronwen J Mayo, Janet K Coller, Rachel J Gibson, Joanne M Bowen

Abstract

Small molecule receptor tyrosine kinase inhibitors (SM-TKIs) are among a group of targeted cancer therapies, intended to be more specific to cancer cells compared with treatments, such as chemotherapy, hence reducing adverse events. Unfortunately, many patients report high levels of diarrhea, the pathogenesis of which remains under investigation. In this article, we compare the current state of knowledge of the pathogenesis of chemotherapy-induced diarrhea (CID) in comparison to SM-TKI-induced diarrhea, and investigate how a similar research approach in both areas may be beneficial. To this end, we review evidence that both treatment modalities may interact with the gut microbiome, and as such the microbiome should be investigated for its ability to reduce the risk of diarrhea.

Keywords: chemotherapy; diarrhea; microbiome; supportive care; tyrosine kinase inhibitors.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Figures

Figure 1.
Figure 1.
Potential interactions of the gut microbiome with tyrosine kinase inhibitor (TKI) treatments leading to diarrhea. (A) Chemotherapy causes vast changes to the gut microbiome,, as well as activation of inflammatory pathways via pattern recognition receptors (PRRs), such as Toll-like receptor 4,, that lead to ulceration and eventual diarrhea. TKI treatment also leads to diarrhea, but the mechanism is not well understood. We propose that the gut microbiome may play a key role. (B) Long-term TKI treatment may lead to a dysbiotic microbiome. Additionally, direct inhibition of EGFRs or VEGFRs in the gut can lead to altered gut function (eg, changes in cell proliferation and capillary networks) that can alter microbial composition.- This could lead to similar inflammatory outcomes as in chemotherapy. (C) Alternatively, increased chloride secretion in the gut (causing diarrhea itself) could lead to a significant shift in the microbiome that may lead to additive effect on the diarrhea.,,

References

    1. Gibson RJ, Stringer AM. Chemotherapy-induced diarrhoea. Curr Opin Support Palliat Care. 2009;3:31-35.
    1. Stein A, Voigt W, Jordan K. Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management. Ther Adv Med Oncol. 2010;2:51-63.
    1. Di Fiore F, Van Cutsem E. Acute and long-term gastrointestinal consequences of chemotherapy. Best Pract Res Clin Gastroenterol. 2009;23:113-124.
    1. Carlotto A, Hogsett VL, Maiorini EM, Razulis JG, Sonis ST. The economic burden of toxicities associated with cancer treatment: review of the literature and analysis of nausea and vomiting, diarrhoea, oral mucositis and fatigue. Pharmacoeconomics. 2013;31:753-766.
    1. Vouk K, Benter U, Amonkar MM, et al. Cost and economic burden of adverse events associated with metastatic melanoma treatments in five countries. J Med Econ. 2016;19:900-912.
    1. Elting LS, Shih YCT. The economic burden of supportive care of cancer patients. Support Care Cancer. 2004;12:219-226.
    1. Rugo HS, Di Palma JA, Tripathy D, et al. The characterization, management, and future considerations for ErbB-family TKI-associated diarrhea. Breast Cancer Res Treat. 2019;175:5-15.
    1. Hojjat-Farsangi M. Small-molecule inhibitors of the receptor tyrosine kinases: promising tools for targeted cancer therapies. Int J Mol Sci. 2014;15:13768-13801.
    1. Jiao Q, Bi L, Ren Y, Song S, Wang Q, Wang YS. Advances in studies of tyrosine kinase inhibitors and their acquired resistance. Mol Cancer. 2018;17:36.
    1. Takeda M, Okamoto I, Nakagawa K. Pooled safety analysis of EGFR-TKI treatment for EGFR mutation-positive non–small cell lung cancer. Lung Cancer. 2015;88:74-79.
    1. McQuade RM, Stojanovska V, Abalo R, Bornstein JC, Nurgali K. Chemotherapy-induced constipation and diarrhea: pathophysiology, current and emerging treatments. Front Pharmacol. 2016;7:414.
    1. Upadhyay A, Bodar V, Malekzadegan M, et al. Loperamide induced life threatening ventricular arrhythmia. Case Rep Cardiol. 2016;2016:5040176.
    1. Loriot Y, Perlemuter G, Malka D, et al. Drug insight: gastrointestinal and hepatic adverse effects of molecular-targeted agents in cancer therapy. Nat Clin Pract Oncol. 2008;5:268-278.
    1. Bowen J, Al-Dasooqi N, Bossi P, et al. The pathogenesis of mucositis: updated perspectives and emerging targets. Support Care Cancer. 2019;27:4023-4033.
    1. Bowen JM. Development of the rat model of lapatinib-induced diarrhoea. Scientifica (Cairo). 2014;2014:194185.
    1. Van Sebille YZ, Gibson RJ, Wardill HR, Bowen JM. ErbB small molecule tyrosine kinase inhibitor (TKI) induced diarrhoea: chloride secretion as a mechanistic hypothesis. Cancer Treat Rev. 2015;41:646-652.
    1. Secombe KR, Ball IA, Shirren J, et al. Targeting neratinib-induced diarrhea with budesonide and colesevelam in a rat model. Cancer Chemother Pharmacol. 2019;83:531-543.
    1. Pal SK, Li SM, Wu X, et al. Stool bacteriomic profiling in patients with metastatic renal cell carcinoma receiving vascular endothelial growth factor-tyrosine kinase inhibitors. Clin Cancer Res. 2015;21:5286-5293.
    1. Liu K, Zhang W, Tan Q, Jiang G, Jia J. Antibiotic use is a negative predictor of the efficacy and toxicity of epidermal growth factor receptor-targeted therapy in advanced non-small cell lung cancer. Oncol Lett. 2019;18:2677-2683.
    1. van Vliet MJ, Tissing WJ, Dun CA, et al. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin Infect Dis. 2009;49:262-270.
    1. Viaud S, Saccheri F, Mignot G, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342:971-976.
    1. National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE). Published 2018. Accessed January 21, 2020
    1. Ribeiro RA, Wanderley CWS, Wong DVT, et al. Irinotecan- and 5-fluorouracil-induced intestinal mucositis: insights into pathogenesis and therapeutic perspectives. Cancer Chemother Pharmacol. 2016;78:881-893.
    1. Falcone A, Ricci S, Brunetti I, et al. Phase III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) Compared with infusional fluorouracil, leucovorin, and irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal cancer: the Gruppo Oncologico Nord Ovest. J Clin Oncol. 2007;25:1670-1676.
    1. Lalla RV, Bowen J, Barasch A, et al. MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer. 2014;120:1453-1461.
    1. Mego M, Chovanec J, Vochyanova-Andrezalova I, et al. Prevention of irinotecan induced diarrhea by probiotics: a randomized double blind, placebo controlled pilot study. Complement Ther Med. 2015;23:356-362.
    1. Michael M, Brittain M, Nagai J, et al. Phase II study of activated charcoal to prevent irinotecan-induced diarrhea. J Clin Oncol. 2004;22:4410-4417.
    1. Que Y, Liang Y, Zhao J, et al. Treatment-related adverse effects with pazopanib, sorafenib and sunitinib in patients with advanced soft tissue sarcoma: a pooled analysis. Cancer Manag Res. 2018;10:2141-2150.
    1. Carrato A, Benavides M, Massutí B, et al. First-line single-agent regorafenib in frail patients with metastatic colorectal cancer: a pilot phase II study of the Spanish Cooperative Group for the Treatment of Digestive Tumours (TTD). BMC Cancer. 2019;19:533.
    1. Yin X, Yin Y, Shen C, et al. Adverse events risk associated with regorafenib in the treatment of advanced solid tumors: meta-analysis of randomized controlled trials. Onco Targets Ther. 2018;11:6405-6414.
    1. Santoni M, Conti A, De Giorgi U, et al. Risk of gastrointestinal events with sorafenib, sunitinib and pazopanib in patients with solid tumors: a systematic review and meta-analysis of clinical trials. Int J Cancer. 2014;135:763-773.
    1. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28:1061-1068.
    1. Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet. 2011;378:1931-1939.
    1. Miller MJ, Foy KC, Kaumaya PTP. Cancer immunotherapy: present status, future perspective, and a new paradigm of peptide immunotherapeutics. Discov Med. 2013;15:166-176.
    1. Huo Z, Yu S, Hong S, et al. A systematic review and meta-analysis of the risk of diarrhea associated with vandetanib treatment in carcinoma patients. Onco Targets Ther. 2016;9:3621-3631.
    1. Pozo K, Zahler S, Ishimatsu K, et al. Preclinical characterization of tyrosine kinase inhibitor-based targeted therapies for neuroendocrine thyroid cancer. Oncotarget. 2018;9:37662-37675.
    1. Yin Y, Qiu XY, Zhang YH, Zhang B. A rare cutaneous phototoxic rash after vandetanib therapy in a patient with thyroid cancer: a case report. Medicine (Baltimore). 2019;98:e16392.
    1. Berdelou A, Borget I, Godbert Y, et al. Lenvatinib for the treatment of radioiodine-refractory thyroid cancer in real-life practice. Thyroid. 2018;28:72-78. doi:10.1089/thy.2017.0205
    1. Haddad RI, Schlumberger M, Wirth LJ, et al. Incidence and timing of common adverse events in Lenvatinib-treated patients from the SELECT trial and their association with survival outcomes. Endocrine. 2017;56:121-128.
    1. Schmidinger M, Danesi R. Management of adverse events associated with cabozantinib therapy in renal cell carcinoma. Oncologist. 2018;23:306-315.
    1. Fazio N, Cella CA, Del Re M, et al. Pharmacodynamics, clinical findings and approval status of current and emerging tyrosine-kinase inhibitors for pancreatic neuroendocrine tumors. Expert Opin Drug Metab Toxicol. 2019;15:993-1004.
    1. Becker A, van Wijk A, Smit EF, Postmus PE. Side-effects of long-term administration of erlotinib in patients with non-small cell lung cancer. J Thorac Oncol. 2010;5:1477-1480.
    1. Hirsh V, Blais N, Burkes R, Verma S, Croitoru K. Management of diarrhea induced by epidermal growth factor receptor tyrosine kinase inhibitors. Curr Oncol. 2014;21:329-336.
    1. Azim HA, Jr, Agbor-Tarh D, Bradbury I, et al. Pattern of rash, diarrhea, and hepatic toxicities secondary to lapatinib and their association with age and response to neoadjuvant therapy: analysis from the NeoALTTO trial. J Clin Oncol. 2013;31:4504-4511.
    1. Tagliamento M, Genova C, Rijavec E, et al. Afatinib and erlotinib in the treatment of squamous-cell lung cancer. Expert Opin Pharmacother. 2018;19:2055-2062.
    1. Chan A, Delaloge S, Holmes FA, et al. Neratinib after trastuzumab-based adjuvant therapy in patients with HER2-positive breast cancer (ExteNET): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2016;17:367-377.
    1. Van Sebille YZA, Gibson RJ, Wardill HR, Bowen JM. Gastrointestinal toxicities of first and second-generation small molecule human epidermal growth factor receptor tyrosine kinase inhibitors in advanced nonsmall cell lung cancer. Curr Opin Support Palliat Care. 2016;10:152-156.
    1. Linger RM, Keating AK, Earp HS, Graham DK. TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv Cancer Res. 2008;100:35-83.
    1. Gaumann AK, Kiefer F, Alfer J, Lang SA, Geissler EK, Breier G. Receptor tyrosine kinase inhibitors: are they real tumor killers? Int J Cancer. 2016;138:540-554.
    1. Bowen JM. Mechanisms of TKI-induced diarrhea in cancer patients. Curr Opin Support Palliat Care. 2013;7:162-167.
    1. Liu J, Nicum S, Reichardt P, et al. Assessment and management of diarrhea following VEGF receptor TKI treatment in patients with ovarian cancer. Gynecol Oncol. 2018;150:173-179.
    1. Ledermann JA, Embleton AC, Raja F, et al. Cediranib in patients with relapsed platinum-sensitive ovarian cancer (ICON6): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2016;387:1066-1074.
    1. Sonis ST. The pathobiology of mucositis. Nat Rev Cancer. 2004;4:277-284.
    1. Sonis ST. New thoughts on the initiation of mucositis. Oral Dis. 2010;16:597-600.
    1. Sonis ST. A biological approach to mucositis. J Support Oncol. 2004;2:21-32.
    1. O’Brien DP, Nelson LA, Williams JL, Kemp CJ, Erwin CR, Warner BW. Selective inhibition of the epidermal growth factor receptor impairs intestinal adaptation after small bowel resection. J Surg Res. 2002;105:25-30.
    1. Echavarria I, Lopez-Tarruella S, Marquez-Rodas I, Jerez Y, Martin M. Neratinib for the treatment of HER2-positive early stage breast cancer. Expert Rev Anticancer Ther. 2017;17:669-679.
    1. Fan L, Iseki S. Immunohistochemical localization of vascular endothelial growth factor in the endocrine glands of the rat. Arch Histol Cytol. 1998;61:17-28.
    1. Kamba T, Tam BYY, Hashizume H, et al. VEGF-dependent plasticity of fenestrated capillaries in the normal adult microvasculature. Am J Physiol Heart Circ Physiol. 2006;290:H560-H576.
    1. Kitagawa D, Yokota K, Gouda M, et al. Activity-based kinase profiling of approved tyrosine kinase inhibitors. Genes Cells. 2013;18:110-122.
    1. Gao X, Le X, Costa DB. The safety and efficacy of osimertinib for the treatment of EGFR T790M mutation positive non–small-cell lung cancer. Expert Rev Anticancer Ther. 2016;16:383-390.
    1. Tan CS, Kumarakulasinghe NB, Huang YQ, et al. Third generation EGFR TKIs: current data and future directions. Mol Cancer. 2018;17:29.
    1. Jänne PA, Yang JCH, Kim DW, et al. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med. 2015;372:1689-1699.
    1. Schmidinger M. Understanding and managing toxicities of vascular endothelial growth factor (VEGF) inhibitors. EJC Suppl. 2013;11:172-191.
    1. Bowen JM, Mayo BJ, Plews E, et al. Determining the mechanisms of lapatinib-induced diarrhoea using a rat model. Cancer Chemother Pharmacol. 2014;74:617-627.
    1. Van Sebille YZA, Gibson RJ, Wardill HR, et al. Dacomitinib-induced diarrhoea is associated with altered gastrointestinal permeability and disruption in ileal histology in rats. Int J Cancer. 2017;140:2820-2829.
    1. Thiagarajah JR, Donowitz M, Verkman AS. Secretory diarrhoea: mechanisms and emerging therapies. Nat Rev Gastroenterol Hepatol. 2015;12:446-457.
    1. Duan T, Cil O, Thiagarajah JR, Verkman AS. Intestinal epithelial potassium channels and CFTR chloride channels activated in ErbB tyrosine kinase inhibitor diarrhea. JCI Insight. 2019;4:126444.
    1. Abbas R, Hug BA, Leister C, Sonnichsen D. A double-blind, randomized, multiple-dose, parallel-group study to characterize the occurrence of diarrhea following two different dosing regimens of neratinib, an irreversible pan-ErbB receptor tyrosine kinase inhibitor. Cancer Chemother Pharmacol. 2012;70:191-199.
    1. Pohlmann PR. Diarrhea prevention and prophylaxis with crofelemer in HER2 positive breast cancer patients (HALT-D). identifier: NCT02910219. Published September 21, 2016. Accessed February 12, 2020
    1. Gao JJ, Tan M, Pohlmann PR, Swain SM. HALT-D: a phase II evaluation of crofelemer for the prevention and prophylaxis of diarrhea in patients with breast cancer on pertuzumab-based regimens. Clin Breast Cancer. 2017;17:76-78.
    1. Secombe KR, Coller JK, Gibson RJ, Wardill HR, Bowen JM. The bidirectional interaction of the gut microbiome and the innate immune system: implications for chemotherapy-induced gastrointestinal toxicity. Int J Cancer. 2019;144:2365-2376.
    1. Lima-Junior RC, Freitas HC, Wong DV, et al. Targeted inhibition of IL-18 attenuates irinotecan-induced intestinal mucositis in mice. Br J Pharmacol. 2014;171:2335-2350.
    1. Arifa RDN, Madeira MFM, de Paula TP, et al. Inflammasome activation is reactive oxygen species dependent and mediates irinotecan-induced mucositis through IL-1β and IL-18 in mice. Am J Pathol. 2014;184:2023-2034.
    1. Sakai H, Sagara A, Matsumoto K, et al. 5-fluorouracil induces diarrhea with changes in the expression of inflammatory cytokines and aquaporins in mouse intestines. PLoS One. 2013;8:e54788.
    1. Wardill HR, Gibson RJ, Van Sebille YZ, et al. Irinotecan-induced gastrointestinal dysfunction and pain are mediated by common TLR4-dependent mechanisms. Mol Cancer Ther. 2016;15:1376-1386.
    1. Liu S, Kurzrock R. Toxicity of targeted therapy: implications for response and impact of genetic polymorphisms. Cancer Treat Rev. 2014;40:883-891.
    1. Lacouture ME, Mitchell EP, Piperdi B, et al. Skin toxicity evaluation protocol with panitumumab (STEPP), a phase II, open-label, randomized trial evaluating the impact of a pre-emptive skin treatment regimen on skin toxicities and quality of life in patients with metastatic colorectal cancer. J Clin Oncol. 2010;28:1351-1357.
    1. Fakih M, Vincent M. Adverse events associated with anti-EGFR therapies for the treatment of metastatic colorectal cancer. Curr Oncol. 2010;17(suppl 1):S18-S30.
    1. Stringer AM, Al-Dasooqi N, Bowen JM, et al. Biomarkers of chemotherapy-induced diarrhoea: a clinical study of intestinal microbiome alterations, inflammation and circulating matrix metalloproteinases. Support Care Cancer. 2013;21:1843-1852.
    1. Stringer AM, Gibson RJ, Bowen JM, et al. Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile. Int J Exp Pathol. 2009;90:489-499.
    1. Meeker SM, Mears KS, Sangwan N, et al. CFTR dysregulation drives active selection of the gut microbiome. PLoS Pathog. 2020;16:e1008251.
    1. Vernocchi P, Del Chierico F, Russo A, et al. Gut microbiota signatures in cystic fibrosis: Loss of host CFTR function drives the microbiota enterophenotype. PLoS One. 2018;13:e0208171.
    1. Knox NC, Forbes JD, Van Domselaar G, Bernstein CN. The gut microbiome as a target for IBD treatment: are we there yet? Curr Treat Options Gastroenterol. 2019;17:115-126.
    1. Gevers D, Kugathasan S, Denson LA, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382-392.
    1. Olszak T, An D, Zeissig S, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science. 2012;336:489-493.
    1. Zhou L, Zhang M, Wang Y, et al. Faecalibacterium prausnitzii produces butyrate to maintain Th17/Treg balance and to ameliorate colorectal colitis by inhibiting histone deacetylase 1. Inflamm Bowel Dis. 2018;24:1926-1940.
    1. Musch MW, Wang Y, Claud EC, Chang EB. Lubiprostone decreases mouse colonic inner mucus layer thickness and alters intestinal microbiota. Dig Dis Sci. 2013;58:668-677.
    1. Keely S, Kelly CJ, Weissmueller T, et al. Activated fluid transport regulates bacterial-epithelial interactions and significantly shifts the murine colonic microbiome. Gut Microbes. 2012;3:250-260.
    1. Hecht G, Marrero JA, Danilkovich A, et al. Pathogenic Escherichia coli increase Cl secretion from intestinal epithelia by upregulating galanin-1 receptor expression. J Clin Invest. 1999;104:253-262.
    1. Heuvelin E, Lebreton C, Bichara M, Cerf-Bensussan N, Heyman M. A Bifidobacterium probiotic strain and its soluble factors alleviate chloride secretion by human intestinal epithelial cells. J Nutr. 2010;140:7-11.
    1. Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Keefe DM. Faecal microflora and beta-glucuronidase expression are altered in an irinotecan-induced diarrhea model in rats. Cancer Biol Ther. 2008;7:1919-1925.
    1. Lin XB, Dieleman LA, Ketabi A, et al. Irinotecan (CPT-11) chemotherapy alters intestinal microbiota in tumour bearing rats. PLoS One. 2012;7:e39764.
    1. Von Bultzingslowen I, Adlerberth I, Wold AE, Dahlen G, Jontell M. Oral and intestinal microflora in 5-fluorouracil treated rats, translocation to cervical and mesenteric lymph nodes and effects of probiotic bacteria. Oral Microbiol Immunol. 2003;18:278-284.
    1. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:499-511.
    1. Riehl T, Cohn S, Tessner T, Schloemann S, Stenson WF. Lipopolysaccharide is radioprotective in the mouse intestine through a prostaglandin-mediated mechanism. Gastroenterology. 2000;118:1106-1116.
    1. Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14:356-365.
    1. Mayo BJ, Secombe KR, Wignall AD, et al. The GLP-2 analogue elsiglutide reduces diarrhoea caused by the tyrosine kinase inhibitor lapatinib in rats. Cancer Chemother Pharmacol. 2020;85:793-803. doi:10.1007/s00280-00020-04040-00280
    1. Blijlevens N, Sonis S. Palifermin (recombinant keratinocyte growth factor-1): a pleiotropic growth factor with multiple biological activities in preventing chemotherapy- and radiotherapy-induced mucositis. Ann Oncol. 2007;18:817-826.
    1. Wardill HR, Van Sebille YZA, Ciorba MA, Bowen JM. Prophylactic probiotics for cancer therapy-induced diarrhoea: a meta-analysis. Curr Opin Support Palliat Care. 2018;12:187-197.
    1. Rossi E, Ianiro G, Maiorano BA, et al. Fecal microbiota transplantation for TKI-induced diarrhea in patients with metastatic renal cell carcinoma. J Clin Oncol. 2019;37(7 suppl):615.
    1. Lacouture ME, Keefe DM, Sonis S, et al. A phase II study (ARCHER 1042) to evaluate prophylactic treatment of dacomitinib-induced dermatologic and gastrointestinal adverse events in advanced non–small-cell lung cancer. Ann Oncol. 2016;27:1712-1718.
    1. Ceccarelli G, Schietroma I, Scheri GC, d’Ettorre G, Vullo V. Possible determinants of VSL#3 probiotic failure in preventing gastrointestinal adverse events associated with dacomitinib in patients with advanced non-small-cell lung cancer enrolled in ARCHER-1042 trial. Ann Oncol. 2016;27:2137-2138.
    1. Lacouture ME; Authors of the publication “A phase II study (ARCHER 1042) to evaluate prophylactic treatment of dacomitinib-induced dermatologic and gastrointestinal adverse events in advanced non–small-cell lung cancer.” Reply to the letter to the editor “Possible determinants of VSL#3 probiotic failure in preventing gastrointestinal adverse events associated with dacomitinib in patients with advanced non–small-cell lung cancer enrolled in ARCHER-1042 trial” by Ceccarelli et al. Ann Oncol. 2016;27:2138-2139.
    1. Pal SK. Probiotic yogurt supplement in reducing diarrhea in patients with metastatic kidney cancer being treated with vascular endothelial growth factor-tyrosine kinase inhibitor. Identifier: NCT02944617. Published October 26, 2016. Accessed February 10, 2020
    1. Wardill HR, Secombe KR, Bryant RV, Hazenberg MD, Costello SP. Adjunctive fecal microbiota transplantation in supportive oncology: emerging indications and considerations in immunocompromised patients. EBioMedicine. 2019;44:730-740.
    1. Le Bastard Q, Ward T, Sidiropoulos D, et al. Fecal microbiota transplantation reverses antibiotic and chemotherapy-induced gut dysbiosis in mice. Sci Rep. 2018;8:6219.
    1. Wardill HR, Tissing WJE. Determining risk of severe gastrointestinal toxicity based on pretreatment gut microbial community in patients receiving cancer treatment: a new predictive strategy in the quest for personalized cancer medicine. Curr Opin Support Palliat Care. 2017;11:125-132.
    1. Liu HB, Wu Y, Lv TF, et al. Skin rash could predict the response to EGFR tyrosine kinase inhibitor and the prognosis for patients with non–small cell lung cancer: a systematic review and meta-analysis. PLoS One. 2013;8:e55128.
    1. Widakowich C, de Castro G, Jr de, Azambuja E, Dinh P, Awada A. Review: side effects of approved molecular targeted therapies in solid cancers. Oncologist. 2007;12:1443-1455.
    1. Pinato DJ, Gramenitskaya D, Altmann DM, et al. Antibiotic therapy and outcome from immune-checkpoint inhibitors. J Immunother Cancer. 2019;7:287.
    1. Koschny R, Gotthardt D, Koehler C, Jaeger D, Stremmel W, Ganten TM. Diarrhea is a positive outcome predictor for sorafenib treatment of advanced hepatocellular carcinoma. Oncology. 2013;84:6-13.
    1. Strumberg D, Awada A, Hirte H, et al. Pooled safety analysis of BAY 43-9006 (sorafenib) monotherapy in patients with advanced solid tumours: is rash associated with treatment outcome? Eur J Cancer. 2006;42:548-556.
    1. Thomas SK, Fossella FV, Liu D, et al. Asian ethnicity as a predictor of response in patients with non-small-cell lung cancer treated with gefitinib on an expanded access program. Clin Lung Cancer. 2006;7:326-331.
    1. Hahn AW, Froerer C, VanAlstine S, et al. Targeting bacteroides in stool microbiome and response to treatment with first-line VEGF tyrosine kinase inhibitors in metastatic renal-cell carcinoma. Clin Genitourin Cancer. 2018;16:365-368.
    1. Gong J, Dizman N, Poroyko V, et al. Gut microbiome composition and response to sunitinib in metastatic renal cell carcinoma (mRCC). J Clin Oncol. 2018;36(6 suppl):657.
    1. Boland K, Bedrani L, Turpin W, et al. Persistent diarrhea in patients with Crohn’s disease after mucosal healing is associated with lower diversity of the intestinal microbiome and increased dysbiosis. Preprint. Posted online March 24, 2020. Clin Gastroenterol Hepatol. doi:10.1016/j.cgh.2020.03.044

Source: PubMed

3
Abonnieren