Skip to main content
Gates Foundation Author Manuscripts logoLink to Gates Foundation Author Manuscripts
. Author manuscript; available in PMC: 2018 Sep 25.
Published in final edited form as: N Engl J Med. 2018 Sep 25;379(17):1621–1634. doi: 10.1056/NEJMoa1803484

Phase 2b placebo-controlled trial of M72/AS01E candidate vaccine to prevent active tuberculosis in adults

Olivier Van Der Meeren 1,*, Mark Hatherill 2,*, Videlis Nduba 3, Robert J Wilkinson 4, Monde Muyoyeta 5, Elana Van Brakel 6, Helen M Ayles 7, German Henostroza 8,§, Friedrich Thienemann 9, Thomas J Scriba 10, Andreas Diacon 11, Gretta L Blatner 12,#, Marie-Ange Demoitié 13, Michele Tameris 14, Mookho Malahleha 15, James C Innes 16, Elizabeth Hellstrom 17, Neil Martinson 18, Tina Singh 19, Elaine Jacqueline Akite 20, Aisha Khatoon 21, Anne Bollaerts 22, Ann M Ginsberg 23, Thomas G Evans 24,, Paul Gillard 25,**, Dereck R Tait 26,**
PMCID: PMC6151253  PMID: 30280651

Abstract

Background:

A tuberculosis vaccine to interrupt transmission is urgently needed. We assessed the safety and efficacy of the candidate tuberculosis vaccine, M72/AS01E, against progression to bacteriologically-confirmed active pulmonary tuberculosis disease in adults with latent Mycobacterium tuberculosis (Mtb) infection.

Methods:

In a randomized, double-blind, placebo-controlled, phase 2b trial conducted in Kenya, South Africa and Zambia, human immunodeficiency virus (HIV)-negative adults aged 18-50 years with latent Mtb infection (positive by interferon-gamma release assay) were randomized (1:1) to receive two doses of either M72/AS01E or placebo intramuscularly on days 0 and 30. Clinical suspicion of tuberculosis was confirmed from sputum using a polymerase chain reaction test and/or mycobacterial culture.

Results:

This paper reports the primary analysis, conducted after a mean follow-up of 2.3 years. 1786 participants received M72/AS01E and 1787 received placebo. In the vaccine group, 10 cases met the primary case definition (bacteriologically-confirmed active pulmonary tuberculosis confirmed prior to treatment, not associated with HIV infection) versus 22 cases in the placebo group (0.3 vs. 0.6 cases per 100 person-years, respectively): vaccine efficacy 54.0% (90% confidence interval 13.9-75.4; 95%CI 2.9-78.2; p=0.04). Solicited and unsolicited adverse events within 7 days post-injection were more frequent among M72/AS01E recipients (91.2%) than placebo recipients (68.9%), the difference attributed mainly to injection site reactions and flu-like symptoms. Serious adverse events, potential immune-mediated diseases and deaths occurred with similar low frequencies between groups.

Conclusions:

M72/AS01E was associated with a clinically acceptable safety profile and provided 54.0% protection for Mtb-infected adults against active pulmonary tuberculosis disease.

Introduction

One-quarter of the global population is estimated to be infected with Mycobacterium tuberculosis (Mtb), and tuberculosis (TB) is the leading infectious cause of death worldwide.1,2 There were an estimated 10.4 million new TB cases and 1.7 million TB deaths in 2016. An effective TB vaccine for Mtb-infected persons would have a marked impact on TB control, including drug-resistant TB, through interruption of transmission.3,4 Modelling suggests the most effective contribution to TB control would be a vaccine preventing pulmonary TB in adolescents and young adults.4 The only licensed TB vaccine, BCG (bacille Calmette-Guérin), does not offer significant protection against pulmonary TB in Mtb-infected adults.5

The M72/AS01E (GSK) candidate vaccine contains the M72 recombinant fusion protein derived from two immunogenic Mtb antigens (Mtb32A and Mtb39A), combined with the Adjuvant System AS01, which is also a component of the malaria (Mosquirix, GSK) and recombinant zoster (Shingrix, GSK) vaccines. The Mtb39A and Mtb32A components of the recombinant antigen elicited specific lymphoproliferation and/or interferon-gamma (IFN-γ) production in individuals with latent and active TB.6-8 In phase 2 studies, M72/AS01 exhibited a clinically acceptable safety profile, and induced humoral and cell-mediated immune (CMI) responses in healthy and human immunodeficiency virus (HIV)-infected individuals, Mtbinfected adults and adolescents, and in BCG-vaccinated infants (Table S1). 9-16

Despite the caveats associated with the available animal models,17 non-clinical evaluations (antigen-selection approach and in vivo preclinical data), clinical safety and immunogenicity evidence, based on the ability of the candidate vaccine to induce Th-1 type responses, supported a proof-of-concept human trial.6-9,18-22

We conducted a proof-of-concept phase 2b trial to evaluate M72/AS01E in preventing bacteriologically-confirmed pulmonary TB in HIV-negative adults with Mtb infection, defined by a positive interferon-gamma release assay (IGRA) (www.clinicaltrials.gov. NCT01755598). This population was selected based on its higher incidence of pulmonary TB compared to IGRA-negative individuals, which allowed a smaller sample size for proofof- concept.23

Methods

Study design

The study is a multi-center, double-blind, randomized (1:1), placebo-controlled trial conducted in three TB endemic African countries (Kenya, South Africa and Zambia). The randomization was not stratified but was performed using a minimization algorithm accounting for gender and center (see Supplement for details). Eleven study sites were selected based on local TB prevalence and ability to perform the study according to Good Clinical Practice guidelines (GCP). The QuantiFERON TB Gold in-Tube assay (QFT, Qiagen) was used at the manufacturer’s recommended cut-off to identify latent Mtb infection. The study population is being followed up for 3 years after vaccination with M72/AS01E or placebo. A pre-specified primary analysis was performed when all participants had completed at least 2 years of follow-up. Immunogenicity and reactogenicity were assessed in a subgroup of 300 participants. The final analysis after 3 years of follow-up and secondary study objectives including CMI responses will be reported in a subsequent publication.

The study was undertaken in accordance with GCP and the Declaration of Helsinki. The protocol was approved by ethics committees and regulatory authorities in each participating country. All participants provided informed consent. The study protocol is available at NEJM.org. Unblinded safety data are reviewed by an independent data monitoring committee (IDMC). Anonymized individual participant data and study documents can be requested for further research from www.clinicalstudydatarequest.com.

Population

Adults 18-50 years of age were eligible if they were healthy or had stable chronic medical conditions, were HIV-negative, had no TB symptoms, were QFT-positive, and had a sputum sample negative for Mtb at baseline using polymerase chain reaction (PCR) (GeneXpert MTB/RIF, Cepheid).

Vaccination

Two doses of M72/AS01E or placebo were administered intramuscularly into the deltoid one month apart.

Efficacy endpoints

The primary study objective was to evaluate M72/AS01E efficacy to prevent active pulmonary TB according to the first case definition (primary endpoint; see Table 1 for case definitions). Secondary study objectives were vaccine efficacy (VE) according to additional case definitions, immunogenicity, safety and reactogenicity of the vaccine.

Table 1. Case definitions of tuberculosis (TB).

Case definition Clinical suspicion* Culture results PCR results HIV status Other condition
1st definition (Primary endpoint): Definite pulmonary TB disease not associated with HIV infection X Either or both positive Negative Sputum collected before initiation of TB treatment
Definition used for the sensitivity analysis of the primary endpoint: Definite pulmonary TB disease (any two positive sputum tests) not associated with HIV infection X Any two tests positive** Negative
2nd Definition: Definite PCR-positive pulmonary TB disease not associated with HIV infection X Any Positive Negative
3rd Definition: Definite pulmonary TB, not associated with HIV-infection X Either or both positive Negative Sputum collected up to 4 weeks
4th Definition: Definite pulmonary TB X Either or both positive Any after initiation of TB treatment
5th Definition: Clinical TB (any location) - Any Any Any Clinician has diagnosed TB disease and has decided to treat the patient
5th Modified definition: Clinical TB (any location) not associated with HIV-infection - Any Any Negative

PCR = polymerase chain reaction

*

Presenting with one or more of cough >1-2 weeks, fever >1 week, night sweats, weight loss, pleuritic chest pain, hemoptysis, fatigue, shortness of breath on exertion.

**

either two positive cultures, or two positive by PCR, or one positive by culture and one by PCR.

Possible deaths due to TB have not been included in any of the case definitions unless the case definition criteria as stated were met.

Evaluation of safety and reactogenicity

Serious adverse events (SAEs), potential immune-mediated diseases (pIMDs) and pregnancies were recorded until 6 months after the second vaccination. SAEs deemed related to study product were recorded until study end. Unsolicited AEs were recorded for 30 days after each dose. Local and systemic symptoms were solicited from the subgroup using diary cards for 7 days after each injection. Laboratory testing for clinical chemistry and hematology was performed in the subgroup on Days 0, 7, 30 and 37.

Evaluation of immunogenicity

Blood samples were collected from the subgroup before dose 1, one month post-dose 2, and annually until year 3. Anti-M72 IgG antibodies were measured using enzyme-linked immunosorbent assay (ELISA) as previously described (cut-off 2.8 ELISA units/ml).13

TB surveillance

TB surveillance used both active (visits, phone calls and text messages), and passive (selfreporting) methods. Participants with clinical suspicion of pulmonary TB provided three sputum samples collected over one week for PCR and liquid culture by Mycobacterial Growth Indicator Tube. Samples were preferably to be taken before initiation of TB treatment, but samples collected up to 4 weeks after treatment initiation were accepted (Table 1, case definitions 3 and 4). Diagnostic and treatment decisions were made by treating non-study physicians. HIV retesting and screening for diabetes (HbA1c) were performed in all participants with confirmed TB disease.

Statistical analysis

Using a log rank test with 80% power assuming a true VE of 70% (hazard ratio of 30%) and a 2-sided 10% significance level, 21 cases were required for a fixed sample design assuming proportional hazard rates. To obtain 21 cases, assuming a mean yearly attack rate of 0.55% in the control group, 2 years of follow-up for each subject and an attrition rate of 15% over the 2-year period, 3,506 participants needed to be enrolled. As per protocol, the primary analysis could occur at 21 cases or at completion of 24-month follow-up.

VE was analyzed in the according-to-protocol efficacy cohort, using a Cox proportional hazard regression models (VE=1-hazard ratio) with 90% confidence intervals (CIs) and Wald p-value. Descriptive post-hoc 95% CIs are also provided. The primary endpoint was met if the lower limit of the 2-sided 90% CI for VE against bacteriologically-confirmed pulmonary TB (first case definition) was >0%. If the primary endpoint was met, the first secondary endpoint (VE for the second case definition) was to be analyzed using the same success criterion. A pre-planned exploratory analysis compared the effect of 6 pre-specified covariates (giving 14 subgroups) on VE (interpretation should be performed cautiously as the risk of having at least one false significant result ranges between 51%–77%).

The total vaccinated cohort (all subjects who received at least one vaccination) was used to assess safety. Analysis of immunogenicity was performed on the according-to-protocol immunogenicity cohort for the subgroup.

Statistical analyses were performed with SAS version 9.2 or above on SAS Drug Development. Only the external statisticians and IDMC have been unblinded at the level of individual subject data.

The Supplementary Appendix provides the eligibility criteria and screening procedures, vaccine and placebo composition, safety monitoring and surveillance activities.

Reults

Out of 3,575 randomized participants, 3,573 received at least one dose of M72/AS01E or placebo between August 2014 and November 2015. The mean age of participants was 28.9 (standard deviation [SD] 8.3 years); 43% were women. The study groups were balanced in terms of pre-specified demographic characteristics (Table S2).

Vaccine efficacy

There were 3,283 participants included in the according-to-protocol efficacy analysis (Figure 1). Ten cases of active pulmonary TB in the vaccine group and 22 cases in the placebo group met the primary case definition after mean follow-up of 2.3 years (SD 0.4) (Table 2). The incidence of pulmonary TB (first case definition) per 100 person-years was 0.3 in the M72/AS01E and 0.6 in the placebo group, with overall VE 54.0% (90% CI 13.9-75.4; 95% CI 2.9-78.2; p=0.04). Analysis using a Cox regression model adjusted for country, gender, diabetes, age strata, smoking, and BCG history, gave nearly identical results (data not shown). VE for the second case definition (secondary endpoint) was 58.3% (90% CI 12.8- 80.1; 95% CI -0.5-82.7; p=0.05), and ranged from 28%–36% for protocol-defined case definitions 3 to 5 (Table 2). Kaplan-Meier curves are displayed in Figure 2 for the first case definition. Results were comparable in the total vaccinated efficacy cohort analysis (VE 57.0% [90% CI: 19.9-76.9; 95% CI: 9.7-79.5]; Table 2).

Figure 1. Study flow.

Figure 1

ATP = according to protocol, N = number of participants, TB = tuberculosis

*Participants eliminated from the ATP efficacy cohort: administration of vaccine forbidden in the protocol (19), randomization error (2), randomization code broken at the investigator site (1), study vaccine not administered according to protocol (3), did not receive two vaccine doses (236), did not enter the efficacy evaluation period one month post-dose 2 (11), active tuberculosis (any case definition) diagnosed up to one month post-dose 2 (1), administration of medication forbidden by the protocol (2), non-compliance with vaccination schedule (3), did not meet inclusion/exclusion criteria (7).

**Participants eliminated from the ATP immunogenicity cohort: administration of vaccine forbidden in the protocol (4), sputum Mtb positive at baseline (1), did not meet inclusion/exclusion criteria (1), concomitant infection (active TB) related to the vaccine which may influence immune response (1), concomitant infection (became HIV -infected) not related to the vaccine which may influence immune response (7), non- compliance with the vaccination schedule (3), non-compliance with the blood sampling schedule (9), essential serological data missing (all post-vaccination time points month 2 and month 12 missing) (15), did not receive two vaccine doses (15).

Table 2. Vaccine efficacy of M72/AS01E versus placebo against pulmonary tuberculosis (TB) in adults with evidence of TB infection (unadjusted Cox regression model).

N = number of participants included in each group, n = number of participants having pulmonary TB according to the definitions specified in Table 1, person-years = sum of follow-up periods (expressed in years). Follow-up starts 30 days after dose 2 for according-to-protocol analysis and from the day of dose 1 for total vaccinated efficacy cohort analysis, and ends for both analyses at the first occurrence of pulmonary TB for cases, or for non-cases at either the individual end of the follow-up or at the data lock point, whichever comes first.

CI = confidence interval, P-value = 2-sided p-value from Cox regression model

According-to-protocol efficacy cohort M72/AS01E N=1623 Placebo N=1660 Vaccine efficacy
TB case definition n Person-years Rate per 100 person-years (90% CI) n Person-years Rate per 100 person-years (90% CI) % (90% CI) % (95% CI) p-value
1st definition 10 3707.03 0.3 (0.2-0.5) 22 3747.43 0.6 (0.4-0.8) 54.0 (13.9-75.4) 54.0 (2.9-78.2) 0.04
Sensitivity analysis 5 3709.42 0.1 (0.1-0.3) 17 3751.23 0.5 (0.3-0.7) 70.3 (31.3-87.1) 70.3 (19.4-89.0)
2nd definition 7 3709.42 0.2 (0.1-0.4) 17 3751.23 0.5 (0.3-0.7) 58.3 (12.8-80.1) 58.3 (-0.5-82.7) 0.05
3rd definition 16 3707.03 0.4 (0.3-0.7) 25 3747.43 0.7 (0.5-0.9) 35.3 (-9.5-61.8) 35.3 (-21.2-65.5)
4th definition 17 3707.03 0.5 (0.3-0.7) 27 3747.43 0.7 (0.5-1.0) 36.4 (-5.9-61.8) 36.4 (-16.8-65.3)
5th definition 21 3711.87 0.6 (0.4-0.8) 30 3753.43 0.8 (0.6-1.1) 29.2 (-13.1-55.7) 29.2 (-23.7-59.5)
5th modified definition 20 3711.87 0.5 (0.4-0.8) 28 3753.03 0.7 (0.5-1.0) 27.7 (-17.0-55.3) 27.7 (-28.3-59.3)
Total vaccinated efficacy cohort M72/AS01E N=1785 Placebo N=1783 Vaccine efficacy
TB case definition n Person-years Rate per 100 person-years (90% CI) n Person-years Rate per 100 personyears (90% CI) % (90% CI) % (95% CI) p-value
1st definition 10 4301.70 0.2 (0.1-0.4) 23 4253.72 0.5 (0.4-0.8) 57.0 (19.9-76.9) 57.0 (9.7-79.5) 0.03
Sensitivity analysis 5 4304.09 0.1 (0.1-0.2) 17 4258.75 0.4 (0.3-0.6) 70.9 (32.9-87.4) 70.9 (21.2-89.3)
2nd definition 7 4304.09 0.2 (0.1-0.3) 17 4258.75 0.4 (0.3-0.6) 59.3 (14.8-80.5) 59.3 (1.8-83.1) <0.05
3rd definition 17 4301.70 0.4 (0.3-0.6) 26 4253.72 0.6 (0.4-0.8) 35.4 (-7.9-61.3) 35.4 (-19.1-64.9)
4th definition 18 4301.70 0.4 (0.3-0.6) 28 4253.72 0.7 (0.5-0.9) 36.5 (-4.4-61.3) 36.5 (-14.9-64.9)
5th definition 23 4306.54 0.5 (0.4-0.8) 30 4260.96 0.7 (0.5-1.0) 24.1 (-19.7-51.9) 24.1 (-30.6-55.9)
5th modified definition 22 4306.54 0.5 (0.4-0.7) 28 4260.55 0.7 (0.5-0.9) 22.2 (-24.2-51.3) 22.2 (-35.9-55.5)

Figure 2. Kaplan- Meier estimate of definite pulmonary tuberculosis disease not associated with HIV -infection (first case definit ion) (According to protocol efficacy cohort) .

Figure 2

The decreased number at risk after 24 months reflects the participants for whom follow-up after this time point had not occurred at the data lock point.

HR = hazard ratio

CI = confidence interval

TB = tuberculosis

A planned sensitivity analysis of the first case definition was retricted to participants positive for Mtb by at least two diagnostic tests (culture and/or PCR) performed on the sputa collected (Table S3). This analysis included 5 cases in the M72/AS01E group and 17 cases in the placebo group; VE was 70.3% (90% CI 31.3-87.1%; p=0.02) (Table 2). Piece-wise analysis of cases (first case definition) occurring before versus after the median follow-up time (1.12 years) showed VE of 39.0% (90% CI -42.5-73.9) in the first period and 66.5% (90% CI 13.3-87.0) in the second period.

Pre-specified subgroup analyses using case definition 1 showed VE in men of 75.2% (p=0.03) and in women of 27.4% (p=0.52), and VE in participants aged <=25 years of 84.4% (p=0.01) and VE for those aged >25-50 years of 10.2% (p=0.82) (Table 3). A post-hoc hierarchical test was performed to assess the interaction between group and gender (p=0.31) and between group and age (p=0.07) in the complete model containing all main effects as well as the two interaction terms (Table S4).

Table 3. Vaccine efficacy of definite pulmonary tuberculosis disease not associated with HIV-infection (case definition 1) for each covariate and overall (unadjusted Cox regression model, according-to-protocol efficacy cohort).

Covariates Group N n Person-years Rate per 100 person-years (90% CI) Vaccine efficacy
% (90% CI) % (95% CI)
Overall M72/AS01E 1623 10 3707.03 0.3 (0.2-0.5) 54.0 (13.9-75.4) 54.0 (2.9-78.2)
Placebo 1660 22 3747.43 0.6 (0.4-0.8)
Diabetes (No) M72/AS01E 1615 10 3688.14 0.3 (0.2-0.5) 53.9 (13.8-75.4) 53.9 (2.8-78.2)
Placebo 1655 22 3735.22 0.6 (0.4-0.8)
Diabetes (Yes) M72/AS01E 7 0 16.29 0.0 0.0 0.0
Placebo 5 0 12.21 0.0
Female M72/AS01E 679 7 1572.39 0.4 (0.2-0.8) 27.4 (-63.4-67.7) 27.4 (-90.8-72.4)
Placebo 708 10 1627.29 0.6 (0.4-1.0)
Male M72/AS01E 944 3 2134.63 0.1 (0.1-0.4) 75.2 (28.3-91.4) 75.2 (12.2-93.0)
Placebo 952 12 2120.13 0.6 (0.4-0.9)
Kenya M72/AS01E 242 2 549.09 0.4 (0.1-1.2) -101.6 (-1411.7-73.1) -101.6 (-2123.7-81.7)
Placebo 246 1 550.84 0.2 (0.0-0.9)
South Africa M72/AS01E 1307 8 3008.71 0.3 (0.1-0.5) 59.3 (19.0-79.6) 59.3 (7.6-82.1)
Placebo 1344 20 3058.97 0.7 (0.5-0.9)
Zambia M72/AS01E 74 0 - -
Placebo 70 1 - -
Current smoker M72/AS01E 831 7 1891.62 0.4 (0.2-0.7) 53.3 (0.8-78.0) 53.3 (-14.6- 80.9)
Placebo 842 15 1891.36 0.8 (0.5-1.2)
Not a current smoker M72/AS01E 791 3 1812.82 0.2 (0.1-0.4) 56.0 (-36.9-85.9) 56.0 (-70.1- 88.6)
Placebo 818 7 1856.06 0.4 (0.2-0.7)
Age ≤25 years M72/AS01E 705 2 1599.77 0.1 (0.0-0.4) 84.4 (45.7-95.5) 84.4 (31.0-96.5)
Placebo 724 13 1616.66 0.8 (0.5-1.3)
Age >25 years M72/AS01E 918 8 2107.25 0.4 (0.2-0.7) 10.2 (-99.6-59.6) 10.2 (-132.7-65.4)
Placebo 936 9 2130.77 0.4 (0.2-0.7)
No BCG history or scar M72/AS01E 136 *1* - -
Placebo 149 *1* - -
BCG history and/or scar M72/AS01E 1243 8 2823.92 0.3 (0.2-0.5) 55.8 (11.0-78.0) 55.8 (-1.8-80.8)
Placebo 1247 18 2808.34 0.6 (0.4-0.9)
Unknown BCG status M72/AS01E 243 1 555.68 0.2 (0.0-0.9) 73.1 (-69.1-95.7) 73.1 (-140.5-97.0)
Placebo 246 4 591.74 0.7 (0.3-1.5)

N = number of participants included in each group, n = number of participants meeting case definition 1, BCG = bacille Calmette-Guérin, person-years = sum of follow-up period (up to the first occurrence of pulmonary TB, or to either the individual end of the follow-up or to the data lock point, which ever occurred first) expressed in years, CI = confidence interval, p-value = Two-sided p-value from Cox regression model, *1* = one case that remains blinded

Reactogenicity and safety

The percentage of participants who experienced at least one SAE within 6 months postvaccination was similar between the groups (1.6% in the M72/AS01E group and 1.8% in the placebo group) (Table 4). One SAE in each group was considered causally related to vaccination (pyrexia and hypertensive encephalopathy, currently blinded to group). pIMDs were reported by two participants in the M72/AS01E group and 5 in the placebo group. There were 24 deaths (14 trauma-related) during the study, 7 in the M72/AS01E group (5 trauma-related) and 17 in the placebo group (9 trauma-related) (Table 4). No death was assessed as related to study vaccination. One participant died of pneumonia for whom there was also a suspicion of intestinal TB, but this latter diagnosis was not confirmed. Vaccination did not significantly affect hematology and biochemistry parameters (Figure S1). A post-hoc analysis showed 33 pregnancies, of which 28 resulted in delivery of a healthy infant. There were three ectopic pregnancies, one spontaneous abortion, and one pregnant woman was lost-to-follow-up. No birth defects were noted. Regular IDMC review of unblinded safety data resulted in recommendations to continue the study unchanged.

Table 4. Vaccine safety summary (Total vaccinated cohort).

M72/AS01E Placebo
N=1786 N=1787
n % (95% CI) n % (95% CI) RR (95% CI)
30 days post-vaccination
At least one unsolicited symptom 1203 67.4 (65.1-69.5) 812 45.4 (43.1-47.8) 1.48 (1.35-1.62)
At least one causally-related unsolicited symptom 992 55.5 (53.2-57.9) 371 20.8 (18.9-22.7 2.68 (2.37-3.02)
At least one grade 3 symptom 234 13.1 (11.6-14.8) 124 6.9 (5.8-8.2) 1.89 (1.51-2.37)
At least one causally-related grade 3 symptom 177 9.9 (8.6-11.4) 27 1.5 (1.0-2.2) 6.56 (4.36-10.23)
At least one SAE 10 0.6 (0.3-1.0) 17 1.0 (0.6-1.5)
At least one causally-related SAE 1 0.1 (0.0-0.3) 1 0.1 (0.0-0.3)
Within 6 months post-vaccination
At least one SAE 29 1.6 (1.1-2.3) 33 1.8 (1.3-2.6)
At least one causally-related SAE 1 0.1 (0.0-0.3) 1 0.1 (0.0-0.3)
pIMD 2 0.1 (0.0-0.4) 5 0.3 (0.1-0.7)
   Immune thrombocytopenic purpura 1 case that remains blinded
   Basedow’s (Graves) disease 1 case that remains blinded
   Gout 1 case that remains blinded
   Optic neuritis 2 cases that remain blinded
   Erythema multiforme 1 case that remains blinded
   Rash morbilliform 1 case that remains blinded
Whole study period
Fatal SAE, all 7 0.4 (0.2-0.8) 17 1.0 (0.6-1.5)
   Injury (gunshot, stab wound, road traffic accident, burn) 5 - 8 -
   Cardiac disorder 1 case that remains blinded
   Unknown cause/ Sudden death 3 cases that remain blinded
   Hepatic cirrhosis and hepatic encephalopathy 1 case that remains blinded
   Acute HIV infection 1 case that remains blinded
   Pneumonia and gastrointestinal tuberculosis suspicion 1 case that remains blinded
   Cerebrovascular accident 1 case that remains blinded
   Completed suicide 1 case that remains blinded
   Dyspnoea (drug overdose) 1 case that remains blinded
   Not coded (stab wound) 1 case that remains blinded
Fatal SAE, causally-related 0 - 0 -

pIMD = potential immune-mediated disease, SAE = serious adverse event, CI = confidence interval, N = number of participants in the indicated cohort, n = number of participants reporting the symptom; RR = relative risk.

More participants reported unsolicited AE in the M72/AS01E group (67.4%) than in the placebo group (45.4%). The excess was driven by injection-site reactions and influenza-like symptoms (Table S5). Swelling reactions larger than 100mm diameter were reported by 53 participants (3.0%) in the M72/AS01E group and by one participant in the placebo group. The median duration of these large swelling reactions was 4 days.

In the subgroup, local and systemic solicited symptoms were reported more frequently by M72/AS01E recipients than placebo recipients (Table S6). Among local solicited symptoms, pain was the most frequently reported (81.8% of M72/AS01E recipients and 34.4% of placebo recipients, with 24.3% and 3.3%, respectively, reporting grade 3 pain). Redness and swelling were uncommon in both groups. Fatigue, headache, malaise, or myalgia was each reported by 58.1%-68.9% of M72/AS01E recipients and 26.5%-47.0% of placebo recipients. Fever >38.0°C was reported by 18.9% and 6.6%, respectively. Fever >39.5°C was uncommon (4.1% versus 1.3%, respectively).

The immunogenicity results indicate 100% of participants in the M72/AS01E group seroconverted at Month 2 and 99% were still seropositive at Month 12 (Figure S2). Figure S3 elaborates on the clinical relevance of this study that could be shared with patients by healthcare professionals.

Discussion

There is no TB vaccine recommended for use in Mtb-infected adults, who represent a large reservoir of future cases of active TB. Here, we demonstrate that protection against TB disease may be achieved by vaccination of Mtb-infected adults with an adjuvanted subunit vaccine containing two Mtb proteins. The finding of efficacy for the primary endpoint was supported by the more stringent sensitivity analysis, and by the analysis of the second case definition. Less stringent case definitions 3-5 showed similar, but non-significant, trends for efficacy. This is the first efficacy trial with M72/AS01E, and the results confirm the clinically acceptable safety and reactogenicity profile observed previously. Antibody responses were in the same range as previously observed in M72/AS01E-vaccinated adults living in TB endemic regions.9,10

Since the trial included only Mtb-infected individuals, it is not possible to determine the extent to which Mtb infection influences VE. In previous TB efficacy trials, the viral-vectored candidate vaccine MVA85A showed no additional protection beyond that provided by BCG in Mtb-uninfected infants;24 multiple doses of inactivated M. vaccae (obuense) administered to HIV-infected adults reduced the hazard of definite TB, which was a secondary endpoint, by 39%, with no effect modification by baseline Mtb infection status.25 A comprehensive global TB vaccination strategy should be targeted at both Mtb-uninfected and -infected adolescents and adults.4 Our study in Mtb-infected adults complements the findings of a recent trial that demonstrated 45% efficacy of BCG revaccination for protection of Mtb non-infected adolescents against sustained QFT seroconversion (In press).26 These results suggest a potential role for M72/AS01E among vaccination strategies against TB.

Recent research suggests that progression from latent Mtb infection to active TB is not a single definitive event, but rather a transition through a spectrum of inflammatory and infected states reflecting the activity of individual granulomas.27 Clinically, this spectrum results in heterogeneous disease states within and between individuals. In this study, participants with clinical suspicion of TB underwent diagnostic investigation. Approximately one-third of confirmed pulmonary TB cases were only confirmed by a single test out of the six performed (either culture or PCR). ‘Single positive’ cases were evenly distributed between the vaccine and placebo arms and became positive by culture (7 cases) after an unusually long period or by PCR (3 cases) after an unusually high number of amplification cycles. We hypothesize that active surveillance of study participants detected pulmonary TB with low bacterial load, consistent with early stages of disease or reinfection. Interestingly, three (out of 10) ‘single positive’ participants (blinded as to group) did not receive TB treatment and remain well, suggesting successful immune control and lack of disease progression. The sensitivity analysis suggested higher VE in participants with at least two positive tests, consistent with higher bacterial load. Piece-wise and time-to-event analyses did not demonstrate significant VE during year 1. We hypothesize this may be because at least some individuals who developed active TB during this time already had incipient TB at baseline, against which the vaccine could not be expected to have impact, or that the study did not have power to demonstrate a difference in the first year, or that this was a chance finding. Whilst we made reasonable efforts to exclude participants with active TB at screening (single PCR test on one sputum specimen), a limitation of the study was that we could not exclude that early active cases were missed, given the frequently low bacillary load and sporadic nature of bacillary shedding in early stages of TB disease.28

Unexpectedly, we observed a higher point estimate for VE in men than women (attack rate in the placebo group 0.6 per 100 person-years for men and women), and in those aged <=25 years versus 26-50 years (attack rate 0.8 versus 0.4 per 100 person-years, respectively). A post-hoc demographic analysis showed an imbalance in gender in the group aged <=25 years (66% men and 34% women), while the older age group was well-balanced, suggesting that the apparent difference observed by gender was confounded by the effect of age and is artefactual. Additionally, in a post-hoc interaction test, VE did not seem to be different by gender (p=0.3), while efficacy tended to be heterogeneous across age groups (p=0.07 in a hierarchical model containing both interactions). Interpretation of all post-hoc and exploratory subgroup analyses should be performed cautiously because the study was not powered to detect differences between subgroups and multiplicity was not accounted for.

Age could potentially affect VE through a differential vaccine effect according to time since primary Mtb infection or BCG priming.29 We hypothesize that those further from primary infection are more likely to have infection under immune system control, with little additional benefit conveyed by vaccination. Increasing age is associated with increased probability of more remote infection based on several studies 30-34and screening data from the current study, in which 55.1%–66.6% of the screened individuals were already Mtb-infected.31 Alternatively, the circumstances that lead to reactivation may be less amenable to immunologic control by booster vaccination further from primary BCG vaccination or initial Mtb infection, and therefore the benefits of vaccination may be more limited. Given the age of the study population, immune senescence is unlikely to impact VE.

PCR had sensitivity of 80% compared to culture (Table S7), consistent with more events meeting the first case definition than the second. Future VE trials should therefore utilize automated liquid culture in addition to PCR to maximize case detection. TB treatment of adult drug-sensitive pulmonary TB leads to negative sputum culture and PCR in 8 weeks in some participants;35 therefore case definitions 3 and 4 likely underestimate TB incidence.

Strengths of the study were the inclusion of a large, well-defined cohort, exclusion of active TB disease at baseline, statistical power to address the primary endpoint, and the use of alternative case definitions for the efficacy endpoint that reflect applicability in the real world. Finally, 99% of participants consented to biobanking of pre- and post-vaccination blood samples. These samples offer the opportunity to discover potential immune correlates of vaccine-mediated protection against TB, which if confirmed, will be critical to reduce the size of future efficacy trials (www.clinicaltrials.gov. NCT02097095).

In conclusion, M72/AS01E had an acceptable safety and reactogenicity profile, and significantly reduced the incidence of pulmonary TB in healthy Mtb-infected HIV-negative adults. These promising results provide a unique opportunity to better understand the mechanisms by which this vaccine confers protection against TB and could lead to future improvements in global tuberculosis control.

Supplementary Appendix

Supplementary Material

Acknowledgments

Trademark statement

Mosquirix and Shingrix are trademarks of the GSK group of companies.

Funding

GlaxoSmithKline Biologicals SA and Aeras; Aeras funders are The Bill & Melinda Gates Foundation, DFID UK, DGIS and AusAID.

Contributors

Conceived and designed the study: AB, AD, AG, DT, GB, GH, HA, MAD, MH, PG, TE, TS, and VN.

Collected the data: AD, AK, DT, EA, EH, EV, FT, JI, TS, GB, GH, HA, MH, MMa, MMu, MT, NM, OV, PG, RJW, TJS, and VN.

Analyzed and interpreted the data: AG, AK, DT, EA, EV, FT, GB, MAD, MH, MMu, NM, OV, PG, RJW, TE, TS, and VN.

Were part of the core writing team: AG, DT, EV, MH, MMu, OV, PG, RJW, VN.

All authors reviewed and approved the final submitted version of the paper.

Conflict of interest

OV, MAD, EA, AB, PG, TS and AK are employed by the GSK group of companies and OV, MAD, PG and TS hold restrictive shares of the GSK group of companies as part of their employee remuneration. MAD has a pending patent for novel methods capable to induce an immune response relating to certain uses of M72/AS01E. GB and TE are former Aeras employees. VN, MMu, EV, GH, FT, AD, MT, MMa, JI, EH, have nothing to disclose. MH and TJS received institutional grants from the GSK group of companies for the conduct of the study. MH and RJW have participated as GSK Advisory Board Members, Therapeutic Vaccines, in London, in 2014. RJW has received grants from Aeras and from Wellcome Trust during the conduct of the study. HA reports grants from the GSK group of companies. NM has received grants from Aeras, from the GSK group of companies and from two Tuberculosis diagnostic devices companies during the conduct of the study. AG and DT are employed by Aeras. AG and DT report grants from Bill and Melinda Gates Foundation and from DFID, DGIS and AusAID during the conduct of the study.

Publisher's Disclaimer: This is an Author Final Manuscript, which is the version after external peer review and before publication in the Journal. The publisher’s version of record, which includes all New England Journal of Medicine editing and enhancements, is available at 10.1056/NEJMoa1803484..

Contributor Information

Olivier Van Der Meeren, GSK, Wavre and Rixensart, Belgium.

Mark Hatherill, South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, South Africa.

Videlis Nduba, Kenya Medical Research Institute (KEMRI/CRDR), Nairobi, Kenya.

Robert J Wilkinson, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa, and Francis Crick Institute London, and Department of Medicine, Imperial College, London, United Kingdom.

Monde Muyoyeta, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambiaa.

Elana Van Brakel, TASK Applied Science, Cape Town, South Africa.

Helen M Ayles, Zambart, University of Zambia, Lusaka, Zambia, and London School of Hygiene and Tropical Medicine, London, United Kingdom.

German Henostroza, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia.

Friedrich Thienemann, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa, and the Department of Internal Medicine, University Hospital of Zurich, Switzerland.

Thomas J. Scriba, South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, South Africa.

Andreas Diacon, TASK Applied Science, Cape Town, South Africa; Stellenbosch University, Cape Town, South Africa.

Gretta L. Blatner, AERAS, Rockville, United States of America.

Marie-Ange Demoitié, GSK, Wavre and Rixensart, Belgium.

Michele Tameris, South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, South Africa.

Mookho Malahleha, Setshaba Research Centre, Pretoria, Gauteng South Africa.

James C. Innes, The Aurum Institute, Klerksdorp and Tembisa Research Centres, South Africa.

Elizabeth Hellstrom, Be PART Yoluntu Centre, Paarl, South Africa.

Neil Martinson, Perinatal HIV Research Unit (PHRU), Chris Hani Baragwanath Hospital, South African MRC Collaborating Centre for HIV/AIDS and TB, and NRF Centre of Excellence in Biomedical TB Research, University of the Witwatersrand Johannesburg, South Africa, and Johns Hopkins University Center for TB Research, Baltimore MD, United States of America.

Tina Singh, GSK, Wavre and Rixensart, Belgium.

Elaine Jacqueline Akite, GSK, Wavre and Rixensart, Belgium.

Aisha Khatoon, GSK, Wavre and Rixensart, Belgium.

Anne Bollaerts, GSK, Wavre and Rixensart, Belgium.

Ann M. Ginsberg, AERAS, Rockville, United States of America.

Thomas G. Evans, AERAS, Rockville, United States of America.

Paul Gillard, GSK, Wavre and Rixensart, Belgium.

Dereck R. Tait, AERAS Global TB Vaccine Foundation, Cape Town, South Africa.

References

  • 1.Houben RM, Dodd PJ. The Global Burden of Latent Tuberculosis Infection: A Reestimation Using Mathematical Modelling. PLoS medicine 2016;13:e1002152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.World Health Organization. Global Tuberculosis Report 2017. (http://www.who.int/tb/publications/global_report/en/) [Google Scholar]
  • 3.Dheda K, Gumbo T, Maartens G, et al. The epidemiology, pathogenesis, transmission, diagnosis, and management of multidrug-resistant, extensively drug-resistant, and incurable tuberculosis. Lancet Respir Med 2017;Mar 5. [DOI] [PubMed] [Google Scholar]
  • 4.Harris RC, Sumner T, Knight GM, White RG. Systematic review of mathematical models exploring the epidemiological impact of future TB vaccines. Human vaccines & immunotherapeutics 2016;12:2813-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mangtani P, Abubakar I, Ariti C, et al. Protection by BCG vaccine against tuberculosis: a systematic review of randomized controlled trials. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America 2014;58:470-80. [DOI] [PubMed] [Google Scholar]
  • 6.Al-Attiyah R, Mustafa AS, Abal AT, El-Shamy AS, Dalemans W, Skeiky YA. In vitro cellular immune responses to complex and newly defined recombinant antigens of Mycobacterium tuberculosis. Clin Exp Immunol 2004;138:139-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Skeiky YA, Lodes MJ, Guderian JA, et al. Cloning, expression, and immunological evaluation of two putative secreted serine protease antigens of Mycobacterium tuberculosis. Infect Immun 1999;67:3998-4007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dillon DC, Alderson MR, Day CH, et al. Molecular characterization and human Tcell responses to a member of a novel Mycobacterium tuberculosis mtb39 gene family. Infect Immun 1999;67:2941-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Day CL, Tameris M, Mansoor N, et al. Induction and regulation of T-cell immunity by the novel tuberculosis vaccine M72/AS01 in South African adults. Am J Respir Crit Care Med 2013;188:492-502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gillard P, Yang PC, Danilovits M, et al. Safety and immunogenicity of the M72/AS01E candidate tuberculosis vaccine in adults with tuberculosis: A phase II randomised study. Tuberculosis (Edinb) 2016;100:118-27. [DOI] [PubMed] [Google Scholar]
  • 11.Idoko OT, Owolabi OA, Owiafe PK, et al. Safety and immunogenicity of the M72/AS01 candidate tuberculosis vaccine when given as a booster to BCG in Gambian infants: an open-label randomized controlled trial. Tuberculosis (Edinb) 2014;94:564-78. [DOI] [PubMed] [Google Scholar]
  • 12.Kumarasamy N, Poongulali S, Bollaerts A, et al. A Randomized, Controlled Safety, and Immunogenicity Trial of the M72/AS01 Candidate Tuberculosis Vaccine in HIV-Positive Indian Adults. Medicine (Baltimore) 2016;95:e2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Leroux-Roels I, Forgus S, De Boever F, et al. Improved CD4⁺ T cell responses to Mycobacterium tuberculosis in PPD-negative adults by M72/AS01 as compared to the M72/AS02 and Mtb72F/AS02 tuberculosis candidate vaccine formulations: a randomized trial. Vaccine 2013;31:2196-206. [DOI] [PubMed] [Google Scholar]
  • 14.Montoya J, Solon JA, Cunanan SR, et al. A randomized, controlled dose-finding Phase II study of the M72/AS01 candidate tuberculosis vaccine in healthy PPD-positive adults. J Clin Immunol 2013;33:1360-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Penn-Nicholson A, Geldenhuys H, Burny W, et al. Safety and immunogenicity of candidate vaccine M72/AS01E in adolescents in a TB endemic setting. Vaccine 2015;33:4025-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Thacher EG, Cavassini M, Audran R, et al. Safety and immunogenicity of the M72/AS01 candidate tuberculosis vaccine in HIV-infected adults on combination antiretroviral therapy: a phase I/II, randomized trial. AIDS 2014;28:1769-81. [DOI] [PubMed] [Google Scholar]
  • 17.Williams A, Orme IM. Animal Models of Tuberculosis: An Overview. Microbiology spectrum 2016;4. [DOI] [PubMed] [Google Scholar]
  • 18.Skeiky YA, Alderson MR, Ovendale PJ, et al. Differential immune responses and protective efficacy induced by components of a tuberculosis polyprotein vaccine, Mtb72F, delivered as naked DNA or recombinant protein. J Immunol 2004;172:7618-28. [DOI] [PubMed] [Google Scholar]
  • 19.Brandt L, Skeiky YA, Alderson MR, et al. The protective effect of the Mycobacterium bovis BCG vaccine is increased by coadministration with the Mycobacterium tuberculosis 72-kilodalton fusion polyprotein Mtb72F in M. tuberculosis-infected guinea pigs. Infect Immun 2004;72:6622-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tsenova L, Harbacheuski R, Moreira AL, et al. Evaluation of the Mtb72F polyprotein vaccine in a rabbit model of tuberculous meningitis. Infect Immun 2006;74:2392-401 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Irwin SM, Izzo AA, Dow SW, et al. Tracking antigen-specific CD8 T lymphocytes in the lungs of mice vaccinated with the Mtb72F polyprotein. Infect Immun 2005;73:5809-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lewinsohn DA, Lines RA, Lewinsohn DM. Human dendritic cells presenting adenovirally expressed antigen elicit Mycobacterium tuberculosis-specific CD8+ T cells. Am J Respir Crit Care Med 2002;166:843-8. [DOI] [PubMed] [Google Scholar]
  • 23.Mahomed H, Hawkridge T, Verver S, et al. The tuberculin skin test versus QuantiFERON TB Gold(R) in predicting tuberculosis disease in an adolescent cohort study in South Africa. PloS one 2011;6:e17984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Tameris MD, Hatherill M, Landry BS, et al. Safety and efficacy of MVA85A, a new tuberculosis vaccine, in infants previously vaccinated with BCG: a randomised, placebocontrolled phase 2b trial. Lancet 2013;381:1021-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.von Reyn CF, Mtei L, Arbeit RD, et al. Prevention of tuberculosis in Bacille Calmette-Guerin-primed, HIV-infected adults boosted with an inactivated whole-cell mycobacterial vaccine. AIDS 2010;24:675-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Results from Innovative Phase 2 Tuberculosis Vaccine Trial Offer Potential for New BCG Revaccination Strategies, Hope For Subunit Vaccines. AERAS. Available at http://www.aeras.org/pressreleases/results-from-innovative-phase-2-tuberculosis-vaccinetrial- offer-potential#.Wtg4Q3-ZGbo Accessed 19 April 2018. [Google Scholar]
  • 27.Cadena AM, Fortune SM, Flynn JL. Heterogeneity in tuberculosis. Nature reviews Immunology 2017;17:691-702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Behr MA, Warren SA, Salamon H, et al. Transmission of Mycobacterium tuberculosis from patients smear-negative for acid-fast bacilli. Lancet 1999;353:444-9. [DOI] [PubMed] [Google Scholar]
  • 29.Barreto ML, Pereira SM, Pilger D, et al. Evidence of an effect of BCG revaccination on incidence of tuberculosis in school-aged children in Brazil: second report of the BCGREVAC cluster-randomised trial. Vaccine 2011;29:4875-7. [DOI] [PubMed] [Google Scholar]
  • 30.Andrews JR, Nemes E, Tameris M, et al. Serial QuantiFERON testing and tuberculosis disease risk among young children: an observational cohort study. The Lancet Respiratory Medicine 2017;5:282-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lau Eea. Prevalence of latent TB infection among adults in endemic regions screened for a phase IIb, double-blind, randomized, placebo-controlled study to evaluate GSK candidate vaccine M72/AS01E. . International AIDS Society - TB2016; 2016. [Google Scholar]
  • 32.Mahomed H, Hawkridge T, Verver S, et al. Predictive factors for latent tuberculosis infection among adolescents in a high-burden area in South Africa. The International Journal of Tuberculosis and Lung Disease 2011;15:331-6. [PubMed] [Google Scholar]
  • 33.Mahomed H, Hughes EJ, Hawkridge T, et al. Comparison of Mantoux skin test with three generations of a whole blood IFN-γ assay for tuberculosis infection. The International Journal of Tuberculosis and Lung Disease 2006;10:310-6. [PubMed] [Google Scholar]
  • 34.Wood R, Liang H, Wu H, et al. Changing prevalence of tuberculosis infection with increasing age in high-burden townships in South Africa. The International Journal of Tuberculosis and Lung Disease 2010;14:406-12. [PMC free article] [PubMed] [Google Scholar]
  • 35.Benator D, Bhattacharya M, Bozeman L, et al. Rifapentine and isoniazid once a week versus rifampicin and isoniazid twice a week for treatment of drug-susceptible pulmonary tuberculosis in HIV-negative patients: a randomised clinical trial. Lancet 2002;360:528-34. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Material

RESOURCES