Adjunctive host-directed therapies for pulmonary tuberculosis: a prospective, open-label, phase 2, randomised controlled trial

Robert S Wallis, Sibuse Ginindza, Trevor Beattie, Nishanee Arjun, Morongwe Likoti, Vinodh A Edward, Mohammed Rassool, Khatija Ahmed, Katherine Fielding, Bintou A Ahidjo, Mboyo D T Vangu, Gavin Churchyard, Robert S Wallis, Sibuse Ginindza, Trevor Beattie, Nishanee Arjun, Morongwe Likoti, Vinodh A Edward, Mohammed Rassool, Khatija Ahmed, Katherine Fielding, Bintou A Ahidjo, Mboyo D T Vangu, Gavin Churchyard

Abstract

Background: Current tuberculosis treatments leave patients with clinically significant lung injury and increased all-cause mortality post-cure. Adjunctive host-directed therapies could protect the lungs, improve long-term survival, and shorten treatment duration; however, few have been tested clinically. Therefore, we aimed to assess the safety and preliminary efficacy of four host-directed therapies for tuberculosis.

Methods: In this prospective, open-label, phase 2, randomised controlled trial, patients with pulmonary tuberculosis were recruited at three clinical sites in South Africa. Eligible patients were aged 18-65 years, HIV-1-negative, and had rifampicin-susceptible Mycobacterium tuberculosis, a sputum Xpert cycle threshold of less than 20, and moderately advanced or far advanced disease on chest radiography. By use of numbers generated in blocks of ten and stratification by site, eligible patients were randomly assigned (1:1:1:1:1) to receive one of the four oral host-directed treatments plus standard tuberculosis treatment or standard treatment alone (the control group). Host-directed treatments were: CC-11050 (200 mg twice daily, taken with food; day 1-112); everolimus (0·5 mg/day; day 1-112); auranofin (3 mg/day for seven doses, then 6 mg/day; day 1-112); and ergocalciferol (5 mg on day 1, then 2·5 mg on day 28 and day 56). All study participants received oral rifabutin-substituted standard tuberculosis treatment for 180 days. Patients and clinicians were not masked to treatment assignment. Spirometry and sputum culture with solid and liquid media were done at baseline and up to 180 days at specified intervals throughout treatment. The primary endpoint was safety and tolerability up to day 210. Secondary preliminary efficacy endpoints were treatment effects on sputum microbiology (culture status at day 56 and the hazard ratio for stable culture conversion up to day 180) and lung function (FEV1 and forced vital capacity [FVC]) measured by spirometry at day 56, day 180, and day 540. Safety was analysed in the intention-to-treat population and preliminary efficacy primarily in the per-protocol population. The trial is registered at ClinicalTrials.gov, NCT02968927. Post-treatment follow-up was completed in 2020.

Findings: Between Nov 18, 2016, and Sept 27, 2018, 200 patients were screened and randomly assigned to different treatment groups (n=40 per group, apart from n=39 in the everolimus group after one patient withdrew consent). 11 treatment-emergent serious adverse events occurred either during treatment or within 30 days after treatment discontinuation, of which three were attributable to a host-directed treatment. Life-threatening thrombocytopenia occurred in an auranofin recipient; apparent intra-abdominal sepsis leading to death occurred in another auranofin recipient and was classified as a suspected unexpected serious adverse reaction. Tuberculous spondylitis occurred as an apparent paradoxical reaction in a patient receiving ergocalciferol. Two patients in the control group had life-threatening, treatment-attributable liver injury. No treatment-emergent, treatment-attributable serious adverse events occurred in patients receiving CC-11050 or everolimus. Mean FEV1 in the control group was 61·7% of predicted (95% CI 56·3-67·1) at baseline and 69·1% (62·3-75·8) at day 180. Patients treated with CC-11050 and everolimus had increased recovery of FEV1 at day 180 relative to the control group (mean difference from control group 6·30%, 95% CI 0·06-12·54; p=0·048; and 6·56%, 0·18-12·95; p=0·044, respectively), whereas auranofin and ergocalciferol recipients did not. None of the treatments had an effect on FVC during 180 days of follow-up or on measures of sputum culture status over the course of the study.

Interpretation: CC-11050 and everolimus were safe and reasonably well tolerated as adjunctive therapies for tuberculosis, and analysis of preliminary efficacy suggests they might also enhance the recovery of FEV1, a key measure of lung function and predictor of all-cause mortality. Further studies of these candidates are warranted.

Funding: The Bill & Melinda Gates Foundation and the South African Medical Research Council.

Copyright © 2021 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile *Withdrew consent before receiving any treatment. †Found to have spinal tuberculosis on baseline PET-CT scan. ‡Found to have elevated blood glucose at screening. §Required an excluded concomitant medication.
Figure 2
Figure 2
Proportion of patients with stable culture conversion with time by treatment group These data are based on combined results of solid and liquid cultures. The table indicates the numbers of patients at risk at key timepoints.
Figure 3
Figure 3
Lung function over time in the per-protocol population (A) Mean FEV1%. (B) Mean FVC. Please note the varying y-axes. HDTs were given until day 112, except for ergocalciferol, for which administration ceased on day 56. The symbols indicate timepoints that differ significantly from control after adjusting for differences at baseline. FEV1=forced expiratory volume in 1 s. FVC=forced vital capacity. HDT=host-directed therapy. *Everolimus at day 180. †CC-11050 at day 180. ‡Auranofin at day 84. §Ergocalciferol at day 14.

References

    1. WHO Global tuberculosis report. 2019.
    1. Willcox PA, Ferguson AD. Chronic obstructive airways disease following treated pulmonary tuberculosis. Respir Med. 1989;83:195–198.
    1. Duong M, Islam S, Rangarajan S. Mortality and cardiovascular and respiratory morbidity in individuals with impaired FEV1 (PURE): an international, community-based cohort study. Lancet Glob Health. 2019;7:e613–e623.
    1. Wallis RS, Hafner R. Advancing host-directed therapy for tuberculosis. Nat Rev Immunol. 2015;15:255–263.
    1. Subbian S, Koo MS, Tsenova L. Pharmacologic inhibition of host phosphodiesterase-4 improves isoniazid-mediated clearance of Mycobacterium tuberculosis. Front Immunol. 2016;7:238.
    1. Subbian S, Tsenova L, Holloway J. Adjunctive phosphodiesterase-4 inhibitor therapy improves antibiotic response to pulmonary tuberculosis in a rabbit model. EBioMedicine. 2016;4:104–114.
    1. Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI, Deretic V. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell. 2004;119:753–766.
    1. Han Q, Lin L, Zhao B, Wang N, Liu X. Inhibition of mTOR ameliorates bleomycin-induced pulmonary fibrosis by regulating epithelial-mesenchymal transition. Biochem Biophys Res Commun. 2018;500:839–845.
    1. Cabahug VLO, Uy HS, Yu-Keh E, Sapno KJD. Outcomes of treatment with sirolimus for non-infectious uveitis: a meta-analysis and systematic review. Clin Ophthalmol. 2019;13:649–669.
    1. Harbut MB, Vilchèze C, Luo X. Auranofin exerts broad-spectrum bactericidal activities by targeting thiol-redox homeostasis. Proc Natl Acad Sci USA. 2015;112:4453–4458.
    1. Martineau AR, Nhamoyebonde S, Oni T. Reciprocal seasonal variation in vitamin D status and tuberculosis notifications in Cape Town, South Africa. Proc Natl Acad Sci USA. 2011;108:19013–19017.
    1. Wallis RS, Zumla A. Vitamin D as adjunctive host-directed therapy in tuberculosis: a systematic review. Open Forum Infect Dis. 2016;3
    1. Coussens AK, Wilkinson RJ, Hanifa Y. Vitamin D accelerates resolution of inflammatory responses during tuberculosis treatment. Proc Natl Acad Sci USA. 2012;109:15449–15454.
    1. Mannick JB, Del Giudice G, Lattanzi M. mTOR inhibition improves immune function in the elderly. Sci Transl Med. 2014;6
    1. Kean WF, Hart L, Buchanan WW. Auranofin. Br J Rheumatol. 1997;36:560–572.
    1. Wallis RS, Peppard T, Hermann D. Month 2 culture status and treatment duration as predictors of recurrence in pulmonary tuberculosis: model validation and update. PLoS One. 2015;10
    1. Wallis RS, Wang C, Meyer D, Thomas N. Month 2 culture status and treatment duration as predictors of tuberculosis relapse risk in a meta-regression model. PLoS One. 2013;8
    1. Graham BL, Steenbruggen I, Miller MR. Standardization of spirometry 2019 update. An official American Thoracic Society and European Respiratory Society Technical statement. Am J Respir Crit Care Med. 2019;200:e70–e88.
    1. Alling DW, Bosworth EB. The after-history of pulmonary tuberculosis. VI. The first fifteen years following diagnosis. Am Rev Respir Dis. 1960;81:839–849.
    1. Imperial MZ, Nahid P, Phillips PPJ. A patient-level pooled analysis of treatment-shortening regimens for drug-susceptible pulmonary tuberculosis. Nat Med. 2018;24:1708–1715.
    1. Angel JH, Chu LS, Lyons HA. Corticotropin in the treatment of tuberculosis. A controlled study. Arch Intern Med. 1961;108:353–369.
    1. Marcus H, Yoo OH, Akyol T, Williams MH., Jr A randomized study of the effects of corticosteroid therapy on healing of pulmonary tuberculosis as judged by clinical, roentgenographic, and physiologic measurements. Am Rev Respir Dis. 1963;88:55–64.
    1. Malik SK, Martin CJ. Tuberculosis, corticosteroid therapy, and pulmonary function. Am Rev Respir Dis. 1969;100:13–18.
    1. Jones PW, Beeh KM, Chapman KR, Decramer M, Mahler DA, Wedzicha JA. Minimal clinically important differences in pharmacological trials. Am J Respir Crit Care Med. 2014;189:250–255.
    1. Donaldson GC, Seemungal TA, Patel IS. Airway and systemic inflammation and decline in lung function in patients with COPD. Chest. 2005;128:1995–2004.
    1. Brasier AR. Therapeutic targets for inflammation-mediated airway remodeling in chronic lung disease. Expert Rev Respir Med. 2018;12:931–939.
    1. Romanowski K, Baumann B, Basham CA, Ahmad Khan F, Fox GJ, Johnston JC. Long-term all-cause mortality in people treated for tuberculosis: a systematic review and meta-analysis. Lancet Infect Dis. 2019;19:1129–1137.
    1. Shuldiner J, Leventhal A, Chemtob D, Mor Z. Mortality after anti-tuberculosis treatment completion: results of long-term follow-up. Int J Tuberc Lung Dis. 2016;20:43–48.
    1. Wallis RS, Johnson JL, Okwera A. Pentoxifylline in human immunodeficiency virus- positive tuberculosis: safety at 4 years. J Infect Dis. 1998;178
    1. Wallis RS, Nsubuga P, Whalen C. Pentoxifylline therapy in human immunodeficiency virus-seropositive persons with tuberculosis: a randomized, controlled trial. J Infect Dis. 1996;174:727–733.
    1. Rothan HA, Stone S, Natekar J, Kumari P, Arora K, Kumar M. The FDA-approved gold drug auranofin inhibits novel coronavirus (SARS-COV-2) replication and attenuates inflammation in human cells. Virology. 2020;547:7–11.
    1. Lewis MG, DaFonseca S, Chomont N. Gold drug auranofin restricts the viral reservoir in the monkey AIDS model and induces containment of viral load following ART suspension. AIDS. 2011;25:1347–1356.
    1. Furst DE. Mechanism of action, pharmacology, clinical efficacy and side effects of auranofin. An orally administered organic gold compound for the treatment of rheumatoid arthritis. Pharmacotherapy. 1983;3:284–298.
    1. Keers RY. The gold rush 1925–35. Thorax. 1980;35:884–889.
    1. Amberson JB, McMahon BT, Pinner M. A clinical trial of sanocrysin in pulmonary tuberculosis. Am Rev Tuberc. 1931;24:401–435.
    1. Eaves R, Hansky J, Wallis P. Gold induced enterocolitis: case report and a review of the literature. Aust N Z J Med. 1982;12:617–620.
    1. Langer HE, Hartmann G, Heinemann G, Richter K. Gold colitis induced by auranofin treatment of rheumatoid arthritis: case report and review of the literature. Ann Rheum Dis. 1987;46:787–792.
    1. Jarner D, Nielsen AM. Auranofin (SK + F 39162) induced enterocolitis in rheumatoid arthritis. A case report. Scand J Rheumatol. 1983;12:254–256.
    1. Liu PT, Stenger S, Li H. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311:1770–1773.
    1. Martineau AR, Timms PM, Bothamley GH. High-dose vitamin D3 during intensive-phase antimicrobial treatment of pulmonary tuberculosis: a double-blind randomised controlled trial. Lancet. 2011;377:242–250.
    1. Salahuddin N, Ali F, Hasan Z, Rao N, Aqeel M, Mahmood F. Vitamin D accelerates clinical recovery from tuberculosis: results of the SUCCINCT Study [Supplementary Cholecalciferol in recovery from tuberculosis]. A randomized, placebo-controlled, clinical trial of vitamin D supplementation in patients with pulmonary tuberculosis'. BMC Infect Dis. 2013;13:22.

Source: PubMed

3
Abonner