Supporting the gastrointestinal microenvironment during high-dose chemotherapy and stem cell transplantation by inhibiting IL-1 signaling with anakinra

H R Wardill, C E M de Mooij, A R Da Silva Ferreira, H Havinga, H J M Harmsen, W J F M van der Velden, L F J van Groningen, W J E Tissing, N M A Blijlevens, H R Wardill, C E M de Mooij, A R Da Silva Ferreira, H Havinga, H J M Harmsen, W J F M van der Velden, L F J van Groningen, W J E Tissing, N M A Blijlevens

Abstract

High-dose chemotherapy causes intestinal inflammation and subsequent breakdown of the mucosal barrier, permitting translocation of enteric pathogens, clinically manifesting as fever. Antibiotics are mainstay for controlling these complications, however, they are increasingly recognized for their detrimental effects, including antimicrobial resistance and dysbiosis. Here, we show that mucosal barrier injury induced by the mucotoxic chemotherapeutic agent, high-dose melphalan (HDM), is characterized by hyper-active IL-1b/CXCL1/neutrophil signaling. Inhibition of this pathway with IL-1RA, anakinra, minimized the duration and intensity of mucosal barrier injury and accompanying clinical symptoms, including diarrhea, weight loss and fever in rats. 16S analysis of fecal microbiome demonstrated a more stable composition in rats receiving anakinra, with reduced pathogen expansion. In parallel, we report through Phase IIA investigation that anakinra is safe in stem cell transplant patients with multiple myeloma after HDM. Ramping-up anakinra (100-300 mg administered intravenously for 15 days) did not cause any adverse events or dose limiting toxicities, nor did it change time to neutrophil recovery. Our results reinforce that strengthening the mucosal barrier may be an effective supportive care strategy to mitigate local and systemic clinical consequences of HDM. We are now conducting a Phase IIB multicenter, placebo-controlled, double-blinded trial to assess clinical efficacy of anakinra (AFFECT-2).Trial registration: ClinicalTrials.gov identifier: NCT03233776.

Conflict of interest statement

The authors declare no competing interests.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
Melphalan causes hyperactivation of the CXCL1 (GROα) / IL-1β axis in rats. (A) CXCL1 and (B) Il-1β were assessed in repeated blood samples collected after melphalan treatment. Data presented as mean ± SEM. N = 8/group, all analyzed longitudinally using a mixed-effects model with Geisser-Greenhouse correction for multiple comparisons.
Figure 2
Figure 2
Anakinra mitigates melphalan-induced mucosal barrier injury and clinical manifestations of gut toxicity. (A) Mucosal barrier injury defined by plasma citrulline showed a significant decrease in MEL + ANA at D + 2 and D + 4 compared to melphalan only. (BC) Anakinra prevented melphalan-induced weight loss and food intake (DE). Data presented as mean ± SEM. N = 8/group, all analyzed longitudinally using a mixed-effects model with Geisser-Greenhouse correction, with the exception of panel (E) which used a one-way ANOVA with Tukey’s post-hoc.
Figure 3
Figure 3
Anakinra does not impair the myeloablative properties of melphalan but controls fever. (A) Neutrophils, (B) lymphocytes and (C) monocytes assessed in whole blood at termination. Melphalan induced fever (D), which was controlled by anakinra. Data presented as mean ± SEM. N = 8/group with all data analyzed using mixed-effects model with Geisser-Greenhouse correction for multiple comparisons.
Figure 4
Figure 4
Anakinra reduces mucosal barrier injury-associated dysbiosis and controls enteric pathogen expansion. Melphalan caused compositional changes in the fecal microbiota that persisted for the entire experimental period (A). MEL + ANA rats only showed significant PCoA changes at day 4 (B). Taxonomic analysis (C) showed expansion of Enterobacteriales in MEL only rats, with increases in E.Coli (D), Bacteroidetes vulgates (E) and Clostridium sp (F). Data shown as individual samples, with the exclusion of Panel C which shows mean relative abundance. N = 8/group, all analyzed longitudinally via repeated stool collection. All statistical analyses for microbiota data are outlined in supplementary methods.
Figure 5
Figure 5
Anakinra promotes commensal stability and controls pathogen expansion. Melphalan alone depleted key commensal microbes and caused expansion of pathogens at day 4 (A), 7 (B) and 10 (C), each of which were minimized by anakinra. Data show median difference between groups (median ± 95% CI). All statistical analyses for microbiota data are outlined in supplementary methods.
Figure 6
Figure 6
Clinical assessments in Phase IIA AFFECT-1 cohort treated with 100, 200 or 300 mg anakinra. (A) Neutrophils, (B) temperature with data on antbiotic use, (C) CRP and (D) albumin were assessed longitudinally in all participants. No differences were detected in any parameter. Data shown as individual participant trajectories.

References

    1. van der Velden WJ, et al. Intestinal damage determines the inflammatory response and early complications in patients receiving conditioning for a stem cell transplantation. PLoS ONE. 2010;5:e15156. doi: 10.1371/journal.pone.0015156.
    1. Goldberg J, Jacobsohn DA, Zahurak ML, Vogelsang GB. Gastrointestinal toxicity from the preparative regimen is associated with an increased risk of graft-versus-host disease. Biol. Blood Marrow Transpl. 2005;11:101–107. doi: 10.1016/j.bbmt.2004.10.007.
    1. Hill GR, Ferrara JL. The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: Rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 2000;95:2754–2759. doi: 10.1182/blood.V95.9.2754.009k25_2754_2759.
    1. Dandoy CE, et al. Incidence, risk factors, and outcomes of patients who develop mucosal barrier injury-laboratory confirmed bloodstream infections in the first 100 days after allogeneic hematopoietic stem cell transplant. JAMA Netw. Open. 2020;3:e1918668. doi: 10.1001/jamanetworkopen.2019.18668.
    1. Ustun C, et al. Bacterial blood stream infections (BSIs), particularly post-engraftment BSIs, are associated with increased mortality after allogeneic hematopoietic cell transplantation. Bone Marrow Transpl. 2019;54:1254–1265. doi: 10.1038/s41409-018-0401-4.
    1. Dandoy CE, et al. Healthcare burden, risk factors, and outcomes of mucosal barrier injury laboratory-confirmed bloodstream infections after stem cell transplantation. Biol. Blood Marrow Transpl. 2016;22:1671–1677. doi: 10.1016/j.bbmt.2016.06.002.
    1. Dandoy CE, Ardura MI, Papanicolaou GA, Auletta JJ. Bacterial bloodstream infections in the allogeneic hematopoietic cell transplant patient: New considerations for a persistent nemesis. Bone Marrow Transpl. 2017;52:1091–1106. doi: 10.1038/bmt.2017.14.
    1. Holler E, et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: Loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol. Blood Marrow Transpl. 2014;20:640–645. doi: 10.1016/j.bbmt.2014.01.030.
    1. Peled JU, et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 2020;382:822–834. doi: 10.1056/NEJMoa1900623.
    1. van Vliet MJ, Harmsen HJ, de Bont ES, Tissing WJ. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog. 2010;6:e1000879. doi: 10.1371/journal.ppat.1000879.
    1. Shono Y, et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci. Transl. Med. 2016 doi: 10.1126/scitranslmed.aaf2311.
    1. Shono Y, van den Brink MRM. Gut microbiota injury in allogeneic haematopoietic stem cell transplantation. Nat. Rev. Cancer. 2018;18:283–295. doi: 10.1038/nrc.2018.10.
    1. Taur Y, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124:1174–1182. doi: 10.1182/blood-2014-02-554725.
    1. Rashidi A, et al. Specific gut microbiota changes heralding bloodstream infection and neutropenic fever during intensive chemotherapy. Leukemia. 2020;34:312–316. doi: 10.1038/s41375-019-0547-0.
    1. Verlinden A, et al. Current antimicrobial practice in febrile neutropenia across Europe and Asia: the EBMT Infectious Disease Working Party survey. Bone Marrow Transpl. 2020;55:1588–1594. doi: 10.1038/s41409-020-0811-y.
    1. Blennow O, Ljungman P. The challenge of antibiotic resistance in haematology patients. Br. J. Haematol. 2016;172:497–511. doi: 10.1111/bjh.13816.
    1. Palumbo A, et al. Autologous transplantation and maintenance therapy in multiple myeloma. N. Engl. J. Med. 2014;371:895–905. doi: 10.1056/NEJMoa1402888.
    1. Mikhael J, et al. Treatment of multiple myeloma: ASCO and CCO joint clinical practice guideline. J. Clin. Oncol. 2019;37:1228–1263. doi: 10.1200/jco.18.02096.
    1. Herbers AH, Blijlevens NM, Donnelly JP, de Witte TJ. Bacteraemia coincides with low citrulline concentrations after high-dose melphalan in autologous HSCT recipients. Bone Marrow Transpl. 2008;42:345–349. doi: 10.1038/bmt.2008.170.
    1. van der Velden WJ, Blijlevens NM, Feuth T, Donnelly JP. Febrile mucositis in haematopoietic SCT recipients. Bone Marrow Transpl. 2009;43:55–60. doi: 10.1038/bmt.2008.270.
    1. Wardill HR, et al. Translational model of melphalan-induced gut toxicity reveals drug-host-microbe interactions that drive tissue injury and fever. Cancer Chemother. Pharmacol. 2021;88:173–188. doi: 10.1007/s00280-021-04273-7.
    1. Bowen JM, et al. Systematic review of agents for the management of cancer treatment-related gastrointestinal mucositis and clinical practice guidelines. Support Care Cancer. 2019;27:4011–4022. doi: 10.1007/s00520-019-04892-0.
    1. Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood. 2011;117:3720–3732. doi: 10.1182/blood-2010-07-273417.
    1. de Mooij CEM, Netea MG, van der Velden WJFM, Blijlevens NMA. Targeting the interleukin-1 pathway in patients with hematological disorders. Blood. 2017 doi: 10.1182/blood-2016-12-754994.
    1. Wang X, et al. Exogenous IL-1Ra attenuates intestinal mucositis induced by oxaliplatin and 5-fluorouracil through suppression of p53-dependent apoptosis. Anticancer Drugs. 2015;26:35–45. doi: 10.1097/CAD.0000000000000142.
    1. Wu Z, et al. Interleukin 1 receptor antagonist reduces lethality and intestinal toxicity of 5-Fluorouracil in a mouse mucositis model. Biomed. Pharmacother. 2011;65:339–344. doi: 10.1016/j.biopha.2011.04.013.
    1. Xiang D, et al. Interleukin-1 receptor antagonist attenuates cyclophosphamide-induced mucositis in a murine model. Cancer Chemother. Pharmacol. 2011;67:1445–1453. doi: 10.1007/s00280-010-1439-1.
    1. Rossi-Semerano L, et al. Tolerance and efficacy of off-label anti-interleukin-1 treatments in France: A nationwide survey. Orphanet J. Rare Dis. 2015;10:19. doi: 10.1186/s13023-015-0228-7.
    1. Fleischmann RM, et al. Safety of extended treatment with anakinra in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2006;65:1006–1012. doi: 10.1136/ard.2005.048371.
    1. van der Velden WJ, Herbers AH, Netea MG, Blijlevens NM. Mucosal barrier injury, fever and infection in neutropenic patients with cancer: Introducing the paradigm febrile mucositis. Br. J. Haematol. 2014;167:441–452. doi: 10.1111/bjh.13113.
    1. Trivedi PJ, Adams DH. Chemokines and chemokine receptors as therapeutic targets in inflammatory bowel disease; pitfalls and promise. J. Crohns Colitis. 2018;12:S641–S652. doi: 10.1093/ecco-jcc/jjx145.
    1. Wardill HR, et al. Irinotecan-induced gastrointestinal dysfunction and pain are mediated by common TLR4-dependent mechanisms. Mol. Cancer Ther. 2016;15:1376–1386. doi: 10.1158/1535-7163.MCT-15-0990.
    1. Bowen J, et al. The pathogenesis of mucositis: Updated perspectives and emerging targets. Support Care Cancer. 2019;27:4023–4033. doi: 10.1007/s00520-019-04893-z.
    1. Apetoh L, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007;13:1050–1059. doi: 10.1038/nm1622.
    1. Coller JK, et al. Potential safety concerns of TLR4 antagonism with irinotecan: A preclinical observational report. Cancer Chemother. Pharmacol. 2017;79:431–434. doi: 10.1007/s00280-016-3223-3.
    1. Fucikova J, et al. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020;11:1013. doi: 10.1038/s41419-020-03221-2.
    1. Kim CW, Kim KD, Lee HK. The role of dendritic cells in tumor microenvironments and their uses as therapeutic targets. BMB Rep. 2021;54:31–43. doi: 10.5483/BMBRep.2021.54.1.224.
    1. Diaz R, et al. Distinct diarrhea profiles during outpatient chemotherapy. Support Care Cancer. 2020 doi: 10.1007/s00520-020-05753-x.
    1. Hueso T, et al. Impact and consequences of intensive chemotherapy on intestinal barrier and microbiota in acute myeloid leukemia: The role of mucosal strengthening. Gut. Microbes. 2020;12:1800897. doi: 10.1080/19490976.2020.1800897.
    1. Wang H, Jatmiko YD, Bastian SE, Mashtoub S, Howarth GS. Effects of supernatants from escherichia coli nissle 1917 and faecalibacterium prausnitzii on Intestinal epithelial cells and a rat model of 5-fluorouracil-induced mucositis. Nutr. Cancer. 2017;69:307–318. doi: 10.1080/01635581.2017.1263747.
    1. Roychowdhury S, et al. Faecalibacterium prausnitzii and a prebiotic protect intestinal health in a mouse model of antibiotic and clostridium difficile exposure. JPEN J. Parenter. Enteral Nutr. 2018;42:1156–1167. doi: 10.1002/jpen.1053.
    1. Kyriazopoulou E, et al. An open label trial of anakinra to prevent respiratory failure in COVID-19. Elife. 2021 doi: 10.7554/eLife.66125.
    1. de Mooij CEM, et al. Anakinra: efficacy in the management of fever during neutropenia and mucositis in autologous stem cell transplantation (AFFECT-2)-study protocol for a multicenter randomized double-blind placebo-controlled trial. Trials. 2020;21:948. doi: 10.1186/s13063-020-04847-5.
    1. Vanlancker E, Vanhoecke B, Stringer A, Van de Wiele T. 5-Fluorouracil and irinotecan (SN-38) have limited impact on colon microbial functionality and composition in vitro. PeerJ. 2017;5:e4017. doi: 10.7717/peerj.4017.
    1. Wardill HR, et al. Antibiotic-induced disruption of the microbiome exacerbates chemotherapy-induced diarrhoea and can be mitigated with autologous faecal microbiota transplantation. Eur. J. Cancer. 2021;153:27–39. doi: 10.1016/j.ejca.2021.05.015.
    1. Wardill HR, et al. Translational model of melphalan-induced gut toxicity reveals drug-host-microbe interactions that drive tissue injury and fever. Cancer Chemother. Pharmacol. 2021 doi: 10.1007/s00280-021-04273-7.
    1. Blijlevens NM, Lutgens LC, Schattenberg AV, Donnelly JP. Citrulline: A potentially simple quantitative marker of intestinal epithelial damage following myeloablative therapy. Bone Marrow Transplant. 2004;34:193–196. doi: 10.1038/sj.bmt.1704563.
    1. Crenn P, et al. Plasma citrulline: A marker of enterocyte mass in villous atrophy-associated small bowel disease. Gastroenterology. 2003;124:1210–1219. doi: 10.1016/s0016-5085(03)00170-7.
    1. Fijlstra M, et al. Lactose maldigestion during methotrexate-induced gastrointestinal mucositis in a rat model. Am. J. Physiol. Gastrointest. Liver Physiol. 2011;300:G283–291. doi: 10.1152/ajpgi.00462.2010.
    1. National Cancer Institute. Cancer therapy evaluation program. Common terminology criteria for adverse events v5.0 (CTCAE). Bethesda, MD: National Cancer Institute. (2017).

Source: PubMed

3
订阅