Safety and efficacy assessment of two new leprosy skin test antigens: randomized double blind clinical study

Becky L Rivoire, Nathan A Groathouse, Stephen TerLouw, Kapil Dev Neupane, Chaman Ranjit, Bishwa Raj Sapkota, Saraswoti Khadge, Chhatra B Kunwar, Murdo Macdonald, Rachel Hawksworth, Min B Thapa, Deanna A Hagge, Melinda Tibbals, Carol Smith, Tina Dube, Dewei She, Mark Wolff, Eric Zhou, Mamodikoe Makhene, Robin Mason, Christine Sizemore, Patrick J Brennan, Becky L Rivoire, Nathan A Groathouse, Stephen TerLouw, Kapil Dev Neupane, Chaman Ranjit, Bishwa Raj Sapkota, Saraswoti Khadge, Chhatra B Kunwar, Murdo Macdonald, Rachel Hawksworth, Min B Thapa, Deanna A Hagge, Melinda Tibbals, Carol Smith, Tina Dube, Dewei She, Mark Wolff, Eric Zhou, Mamodikoe Makhene, Robin Mason, Christine Sizemore, Patrick J Brennan

Abstract

Background: New tools are required for the diagnosis of pre-symptomatic leprosy towards further reduction of disease burden and its associated reactions. To address this need, two new skin test antigens were developed to assess safety and efficacy in human trials.

Methods: A Phase I safety trial was first conducted in a non-endemic region for leprosy (U.S.A.). Healthy non-exposed subjects (n = 10) received three titrated doses (2.5 µg, 1.0 µg and 0.1 µg) of MLSA-LAM (n = 5) or MLCwA (n = 5) and control antigens [Rees MLSA (1.0 µg) and saline]. A randomized double blind Phase II safety and efficacy trial followed in an endemic region for leprosy (Nepal), but involved only the 1.0 µg (high dose) and 0.1 µg (low dose) of each antigen; Tuberculin PPD served as a control antigen. This Phase II safety and efficacy trial consisted of three Stages: Stage A and B studies were an expansion of Phase I involving 10 and 90 subjects respectively, and Stage C was then conducted in two parts (high dose and low dose), each enrolling 80 participants: 20 borderline lepromatous/lepromatous (BL/LL) leprosy patients, 20 borderline tuberculoid/tuberculoid (BT/TT) leprosy patients, 20 household contacts of leprosy patients (HC), and 20 tuberculosis (TB) patients. The primary outcome measure for the skin test was delayed type hypersensitivity induration.

Findings: In the small Phase I safety trial, reactions were primarily against the 2.5 µg dose of both antigens and Rees control antigen, which were then excluded from subsequent studies. In the Phase II, Stage A/B ramped-up safety study, 26% of subjects (13 of 50) showed induration against the high dose of each antigen, and 4% (2 of 50) reacted to the low dose of MLSA-LAM. Phase II, Stage C safety and initial efficacy trial showed that both antigens at the low dose exhibited low sensitivity at 20% and 25% in BT/TT leprosy patients, but high specificity at 100% and 95% compared to TB patients. The high dose of both antigens showed lower specificity (70% and 60%) and sensitivity (10% and 15%). BL/LL leprosy patients were anergic to the leprosy antigens.

Interpretation: MLSA-LAM and MLCwA at both high (1.0 µg) and low (0.1 µg) doses were found to be safe for use in humans without known exposure to leprosy and in target populations. At a sensitivity rate of 20-25% these antigens are not suitable as a skin test for the detection of the early stages of leprosy infection; however, the degree of specificity is impressive given the presence of cross-reactive antigens in these complex native M. leprae preparations.

Trial registration: ClinicalTrials.gov NCT01920750 (Phase I), NCT00128193 (Phase II).

Conflict of interest statement

I have read the journal's policy and have the following conflict: The EMMES Corporation served as the Data Coordinating Center for these studies. This does not alter our adherence to all PLOS NTDs policies on sharing data and materials.

Figures

Figure 1. CONSORT flow diagram, Phase II,…
Figure 1. CONSORT flow diagram, Phase II, Stage C-1 Trial.
Figure 2. Phase II, Stage A/B –…
Figure 2. Phase II, Stage A/B – DTH induration by subject.
Phase II, Stage A/B graph depicting DTH indurations elicited by leprosy skin test antigens at the high dose (1.0 µg) and low dose (0.1 µg), and PPD at 5 TU: A) MLCwA, and B) MLSA-LAM. The first five subjects on both graphs represent subjects from Stage A, and the remaining 45 subjects on both graphs represent subjects from Stage B.
Figure 3. Dot plot of induration measurements.
Figure 3. Dot plot of induration measurements.
Induration results are provided across five subject groups, including BL/LL leprosy patients (n = 19 in low and n = 20 in high dose group), BT/TT leprosy patients (n = 20), HC (n = 20), TB (n = 20), and ECs (n = 50). Low and high dose groups were combined to show PPD responses: BL/LL leprosy patients (n = 39) and all other groups (n = 40). Mean and standard deviation are shown.
Figure 4. Distribution frequency of induration.
Figure 4. Distribution frequency of induration.
Frequency distribution graphs were used to establish cut off points for each skin test antigen at each dosage tested: A) MLSA-LAM low dose, B) MLCwA low dose, C) MLSA-LAM high dose, D) MLCwA high dose. Frequency of induration reaction (mm) of EC and TB groups were graphed against BT/TT and BL/LL leprosy groups. The anti-mode between the control and leprosy patient group represents the cut off for each antigen and antigen dose.

References

    1. WHO Expert Committee on Leprosy. World Health Organ Tech Rep Ser 1–61, 61 p following 61.
    1. Snider DE Jr (1982) The tuberculin skin test. Am Rev Respir Dis 125: 108–118.
    1. Huebner RE, Schein MF, Bass JB Jr (1993) The tuberculin skin test. Clin Infect Dis 17: 968–975.
    1. Convit J, Sampson C, Zuniga M, Smith PG, Plata J, et al. (1992) Immunoprophylactic trial with combined Mycobacterium leprae/BCG vaccine against leprosy: preliminary results. Lancet 339: 446–450.
    1. WHO IMMLEP (1982) Vaccination trials against leprosy: a meeting of the epidemiology subgroup of Scientific Working Group on the Immunology of Leprosy, Geneva, 11–13 February, 1985. WHO Document TDR/IMMLEP/EDP 85: 7–8.
    1. Meyers WM, Kvernes S, Binford CH (1975) Comparison of reactions to human and armadillo lepromins in leprosy. Int J Lepr Other Mycobact Dis 43: 218–225.
    1. Millar JW, Gannon C, Chan CS (1975) Comparison in leprosy patients of Fernandex and Mitsuda reactions using human and armadillo antigens. A double-blind study. Int J Lepr Other Mycobact Dis 43: 226–233.
    1. FDA (2011) Current Good Manufacturing Practice in Manufacturing, Processing, Packing, or Holding of Drugs; General. 21 CFR 210
    1. FDA (2011) Current Good Manufacturing Practice for Finished Pharmaceuticals. 21 CFR 211.
    1. Rivoire BL, TerLouw S, Groathouse N, Brennan PJ (2014) The Challenge of Producing Skin Test Antigens with Minimal Resources Suitable for Human Application against a Neglected Tropical Disease; Leprosy. PLoS Negl Trop Dis e2791.
    1. Smelt AH, Rees RJ, Liew FY (1981) Induction of delayed-type hypersensitivity to Mycobacterium leprae in healthy individuals. Clin Exp Immunol 44: 501–506.
    1. Arnadottir T, Rieder HL, Trebucq A, Waaler HT (1996) Guidelines for conducting tuberculin skin test surveys in high prevalence countries. Tuber Lung Dis 77 (Suppl 1) 1–19.
    1. Sanofi Pasteur Limited Toronto Ontario Canada (2009) Tubersol, Tuberculin Purified Protein Derivative (Mantoux) Package Insert R19-0905 Canada.
    1. ICH (2012) MeDRA Term Selection. Technical Requirements for Registration of Pharmaceuticals for Human Use. Points to Consider 4.3
    1. FDA (2011) Investigational New Drug Application; Responsibilities of Sponsors and Investigators. 21 CFR 312 Subpart D.
    1. Britton WJ, Lockwood DN (2004) Leprosy. Lancet 363: 1209–1219.
    1. Global strategy for further reducing the leprosy burden and sustaining leprosy control activities 2006–2010. Operational guidelines. Lepr Rev 77: IX, X, 1–50.
    1. Holland SM (2001) Nontuberculous mycobacteria. Am J Med Sci 321: 49–55.
    1. Parashar D, Das R, Chauhan DS, Sharma VD, Lavania M, et al. (2009) Identification of environmental mycobacteria isolated from Agra, north India by conventional & molecular approaches. Indian J Med Res 129: 424–431.
    1. Fine PE, Floyd S, Stanford JL, Nkhosa P, Kasunga A, et al. (2001) Environmental mycobacteria in northern Malawi: implications for the epidemiology of tuberculosis and leprosy. Epidemiol Infect 126: 379–387.
    1. IMMLEP W (1998) Expert Committee on Leprosy. 7th Report 1–43.
    1. Organization WH (2001) International Union Against Tuberculosis and Lung Disease, Royal Netherlands Tuberculosis Association. Revised international definitions in tuberculosis control. Int J Tuberc Lung Dis 5: 213–215.
    1. W.H K, W.A W (1952) Use of ranks in one-criterion variance analysis. Journal of the American Statistical Association 47: 583–621.
    1. Zhou, Obuchowski, McClish (2002) Statistical Methods in Diagnostic Medicine. Wiley.
    1. Organization WH (2012) Expert Committee on Leprosy, 8th Report.
    1. Nery JA, Bernardes Filho F, Quintanilha J, Machado AM, Oliveira Sde S, et al. (2013) Understanding the type 1 reactional state for early diagnosis and treatment: a way to avoid disability in leprosy. An Bras Dermatol 88: 787–792.
    1. Voorend CG, Post EB (2013) A systematic review on the epidemiological data of erythema nodosum leprosum, a type 2 leprosy reaction. PLoS Negl Trop Dis 7: e2440.
    1. FDA (2011) Investigational New Drug Application. 21 CFR 312
    1. Gupte MD, Anantharaman DS, Nagaraju B, Kannan S, Vallishayee RS (1990) Experiences with Mycobacterium leprae soluble antigens in a leprosy endemic population. Lepr Rev 61: 132–144.
    1. Collins FM, Morrison NE, Watson SR (1983) Fernandez and Mitsuda reactivity in guinea pigs sensitized with heat-killed Mycobacterium leprae: persistence and specificity of skin reactivity to soluble and particulate antigens. Int J Lepr Other Mycobact Dis 51: 481–489.
    1. Gupte MD, Anantharaman DS (1988) Use of soluble antigens in leprosy epidemiology. Lepr Rev 59: 329–335.
    1. American Thoracic Society. Diagnostic standards and classification of tuberculosis. Am Rev Respir Dis 142: 725–735.
    1. Scollard DM, Adams LB, Gillis TP, Krahenbuhl JL, Truman RW, et al. (2006) The continuing challenges of leprosy. Clin Microbiol Rev 19: 338–381.
    1. Leeflang MM, Deeks JJ, Gatsonis C, Bossuyt PM (2008) Systematic reviews of diagnostic test accuracy. Ann Intern Med 149: 889–897.
    1. Zweig MH, Campbell G (1993) Receiver-operating characteristic (ROC) plots: a fundamental evaluation tool in clinical medicine. Clin Chem 39: 561–577.
    1. Sterne JA, Fine PE, Ponnighaus JM, Rees RJ, Chavula D (1998) Delayed-type hypersensitivity to Mycobacterium leprae soluble antigens as a test for infection with the leprosy bacillus. Int J Epidemiol 27: 713–721.
    1. Organization. WH (1982) Report of the Eleventh and Twelfth Meetings of the Steering Committee of the Scientific Working Group on the Chemotherapy of Leprosy. Geneva. March 30–31 and Oct 10. TDR/THELEP-SC (11–12) 82: 1–6.
    1. Geluk A (2013) Challenges in immunodiagnostic tests for leprosy. Expert Opin Med Diagn 7: 265–274.
    1. Douglas JT, Cellona RV, Fajardo TT Jr, Abalos RM, Balagon MV, et al. (2004) Prospective study of serological conversion as a risk factor for development of leprosy among household contacts. Clin Diagn Lab Immunol 11: 897–900.
    1. Manandhar R, LeMaster JW, Butlin CR, Brennan PJ, Roche PW (2000) Interferon-gamma responses to candidate leprosy skin-test reagents detect exposure to leprosy in an endemic population. Int J Lepr Other Mycobact Dis 68: 40–48.
    1. Sengupta U, Sinha S, Ramu G, Lamb J, Ivanyi J (1987) Suppression of delayed hypersensitivity skin reactions to tuberculin by M. leprae antigens in patients with lepromatous and tuberculoid leprosy. Clin Exp Immunol 68: 58–64.
    1. Ottenhoff TH (2012) New pathways of protective and pathological host defense to mycobacteria. Trends Microbiol 20: 419–428.
    1. Modlin RL (2010) The innate immune response in leprosy. Curr Opin Immunol 22: 48–54.
    1. Bhat RM, Prakash C (2012) Leprosy: an overview of pathophysiology. Interdiscip Perspect Infect Dis 2012: 181089.
    1. Kumar S, Naqvi RA, Khanna N, Rao DN (2011) Disruption of HLA-DR raft, deregulations of Lck-ZAP-70-Cbl-b cross-talk and miR181a towards T cell hyporesponsiveness in leprosy. Mol Immunol 48: 1178–1190.
    1. Chatterjee D, Khoo KH (1998) Mycobacterial lipoarabinomannan: an extraordinary lipoheteroglycan with profound physiological effects. Glycobiology 8: 113–120.
    1. Torrelles JB, Sieling PA, Zhang N, Keen MA, McNeil MR, et al. (2012) Isolation of a distinct Mycobacterium tuberculosis mannose-capped lipoarabinomannan isoform responsible for recognition by CD1b-restricted T cells. Glycobiology 22: 1118–1127.
    1. Barnes PF, Chatterjee D, Abrams JS, Lu S, Wang E, et al. (1992) Cytokine production induced by Mycobacterium tuberculosis lipoarabinomannan. Relationship to chemical structure. J Immunol 149: 541–547.
    1. Marques MA, Neves-Ferreira AG, da Silveira EK, Valente RH, Chapeaurouge A, et al. (2008) Deciphering the proteomic profile of Mycobacterium leprae cell envelope. Proteomics 8: 2477–2491.
    1. Geluk A, Bobosha K, van der Ploeg-van Schip JJ, Spencer JS, Banu S, et al. (2012) New biomarkers with relevance to leprosy diagnosis applicable in areas hyperendemic for leprosy. J Immunol 188: 4782–4791.
    1. Aggerbeck H, Giemza R, Joshi P, Tingskov PN, Hoff ST, et al. (2013) Randomised clinical trial investigating the specificity of a novel skin test (C-Tb) for diagnosis of M. tuberculosis infection. PLoS One 8: e64215.
    1. Spencer JS, Brennan PJ (2011) The role of Mycobacterium leprae phenolic glycolipid I (PGL-I) in serodiagnosis and in the pathogenesis of leprosy. Lepr Rev 82: 344–357.
    1. Sampaio LH, Stefani MM, Oliveira RM, Sousa AL, Ireton GC, et al. (2011) Immunologically reactive M. leprae antigens with relevance to diagnosis and vaccine development. BMC Infect Dis 11: 26.
    1. Moet FJ, Oskam L, Faber R, Pahan D, Richardus JH (2004) A study on transmission and a trial of chemoprophylaxis in contacts of leprosy patients: design, methodology and recruitment findings of COLEP. Lepr Rev 75: 376–388.
    1. Moet FJ, Pahan D, Oskam L, Richardus JH (2008) Effectiveness of single dose rifampicin in preventing leprosy in close contacts of patients with newly diagnosed leprosy: cluster randomised controlled trial. BMJ 336: 761–764.
    1. Feenstra SG, Pahan D, Moet FJ, Oskam L, Richardus JH (2012) Patient-related factors predicting the effectiveness of rifampicin chemoprophylaxis in contacts: 6 year follow up of the COLEP cohort in Bangladesh. Lepr Rev 83: 292–304.

Source: PubMed

3
Subscribe