Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jun 1.
Published in final edited form as: Lancet Infect Dis. 2015 May 6;15(6):711–720. doi: 10.1016/S1473-3099(15)00007-9

Towards earlier inclusion of Children in Tuberculosis (TB) drugs trials: Consensus statements from an Expert Panel

Sharon Nachman 1, Amina Ahmed 1, Farhana Amanullah 1, Mercedes C Becerra 1, Radu Botgros 1, Grania Brigden 1, Renee Browning 1, Elizabeth Gardiner 1, Richard Hafner 1, Anneke Hesseling 1, Cleotilde How 1, Patrick Jean-Philippe 1, Erica Lessem 1, Mamodikoe Makhene 1, Nontombi Mbelle 1, Ben Marais 1, Helen McIlleron 1, David F Mc Neeley 1, Carl Mendel 1, Stephen Murray 1, Eileen Navarro 1, Gloria E Oramasionwu 1, Ariel R Porcalla 1,*, Clydette Powell 1, Mair Powell 1, Mona Rigaud 1, Vanessa Rouzier 1, Pearl Samson 1, H Simon Schaaf 1, Seema Shah 1, Jeff Starke 1, Soumya Swaminathan 1, Eric Wobudeya 1, Carol Worrell 1
PMCID: PMC4471052  NIHMSID: NIHMS699644  PMID: 25957923

Abstract

Children represent a significant proportion of the global tuberculosis (TB) burden, and may be disproportionately more affected by its most severe clinical manifestations. Currently available treatments for pediatric drug-susceptible (DS) and drug-resistant (DR) TB, albeit generally effective, are hampered by high pill burden, long duration of treatment, coexistent toxicities, and an overall lack of suitable, child-friendly formulations. The complex and burdensome nature of administering the existing regimens to treat DS TB also contributes to the rise of DR TB strains. Despite the availability and use of these therapies for decades, a dearth of dosing evidence in children underscores the importance of sustained efforts for TB drug development to better meet the treatment needs of children with TB. Several new TB drugs and regimens with promising activity against both DS and DR TB strains have recently entered clinical development and are in various phases of clinical evaluation in adults or have received marketing authorization for adults. However, initiation of clinical trials to evaluate these drugs in children is often deferred, pending the availability of complete safety and efficacy data in adults or after drug approval. This document summarizes consensus statements from an international panel of childhood TB opinion leaders which support the initiation of evaluation of new TB drugs and regimens in children at earlier phases of the TB Drug development cycle.

Keywords: childhood TB, drug resistant TB, pharmacokinetics, ethics, clinical trial, new anti-TB drugs

INTRODUCTION

TB is a major, but often unrecognized, cause of morbidity and mortality among children in the developing world. Current figures probably underestimate the global disease burden, with childhood TB cases accounting for an estimated 6% of reported cases, 1 and at least double this percentage in highly TB-endemic areas. 2 Underdiagnosis (and thus underreporting) is of special concern in younger children who are at greatest risk of disease progression following TB exposure and infection, and in whom microbiologic or other diagnostic confirmation of both tuberculous infection and disease is most problematic. 3 HIV infection increases the risk of TB disease and death, particularly in the absence of antiretroviral treatment. 410 Of further concern is that the number of children with DR-TB is increasing globally. 1116 TB treatment, including treatment for DR-TB, can be effective for children, 17 but is limited by poor service delivery, a lack of child-friendly drug formulations, and a scarcity of data on safety, dosing, and drug-drug interactions. 1722 A critical gap persists for the treatment of DR-TB: new drugs and regimens are needed for children, and more data are needed to strengthen the evidence base and guide the use of existing second-line drugs 18, 23 decrease pediatric TB medicines’ market fragmentation and improve access to these drugs. 24 This document, which builds on previous similar efforts 25, 26, presents consensus statements from an expert panel to promote strategies for the timely collection of evidence on safety and dosing of TB drugs in children to guide clinical management and optimize the care of children with TB.

CONSENSUS STATEMENTS PREPARATION

Search strategy and selection criteria

Before the workshop, relevant literature was surveyed (SN, RB2, PJP) to review evidence and prepare statements for discussion. The databases we searched included PubMed, Medline, Embase with an emphasis on English language papers published during the past 10 years in peer-reviewed journals. Some older papers were also included if they were judged to be important by the authors. Search terms included “TB”, “childhood TB”, “Anti TB treatment”, “MDR-TB treatment”, “MDR-TB outcomes”, “Drug Exposure”, “Pharmacovigilance”, “clinical trials”, “Drug Development”, “HIV-infected”, “Pharmacokinetics”, “Ethics”.

Consensus generation

Expert pediatric TB clinicians, researchers, and opinion leaders were invited to a workshop, “Towards Earlier Involvement of Children and Pregnant Women in Trials of New TB Drugs”, organized by the National Institutes of Health (NIH) in Bethesda, MD, in May 2013. The expert Panel’s consensus on pregnant or lactating women is reported separately. Members from regulatory agencies were invited to attend as non-voting panelists.

Draft statements were circulated to panelists for review and comment, discussed on teleconference calls, revised accordingly and drafts distributed to participants before the workshop. During timed discussions, a group consensus approach was used, that included modifying the statements in real time based on panelists’ suggested modifications. Edited statements underwent panel vote. Voting rules included 2 options: agree or disagree, as indicated by a show of hands. Consensus was declared for a statement if ≥75% of panelists agreed to the final draft statement. All statements were further reviewed in a final plenary workshop session. After the workshop, additional conference calls were held with panelists to finalize consensus.

STATE OF RESEARCH ON NEW TB DRUGS IN CHILDREN

Ongoing and planned trials

This is a particularly exciting time in TB drug research. New drugs, many with novel mechanisms of action, novel drug combinations and strategies to treat TB are being investigated. Clinical trials for DR-TB in adults are currently underway for new nitroimidazoles (delamanid, PA-824), oxazolidinones (sutezolid, linezolid, AZD5847), bedaquiline, and clofazimine. 2730 Some of these drugs have received accelerated approvals for marketing. 3133 Novel combinations that include both new chemical entities and older or repurposed drugs are being tested in adults in studies such as ‘REMox’(Rapid Evaluation of Moxifloxacin in the treatment of sputum smear positive tuberculosis), the Global Alliance for TB Drug Development’s ‘NC002’ (PA-824, moxifloxacin, and pyrazinamide) and ‘NC003’ (clofazimine, bedaquiline, PA-824 and pyrazinamide) trials, and the ‘STREAM’(The Evaluation of a Standardised Treatment Regimen of Anti-Tuberculosis Drugs for Patients with multidrug-resistant [MDR]-TB) trial. 3436 Unfortunately, studies in children lag significantly behind adult studies, and safety and pharmacokinetic studies to support a pediatric indication are currently on-going or planned in children for very few of these novel treatments (delamanid and bedaquiline). This lag in developing studies in children is reflected in research and development investments for pediatric TB accounting for just two percent of the total funding invested in TB research overall 37 but a quarter of the estimated need. 38 Figure 1 illustrates main phases in TB drug development and Table 1 summarizes relevant TB treatment studies in children, known to the authors.

Figure 1. TB Drug Development phases.

Figure 1

Reproduced with permission 74

Table 1.

On-going and planned studies in children (current as of December 2013)

Study Sponsor Status Endpoint
Pharmacokinetics of first-and second-line agents in children with DS-TB and DR-TB NIH on-going PK and safety
Pharmacokinetics of delamanid in children Otsuka on-going PK, safety
Pharmacokinetics of bedaquiline in children Janssen, IMPAACT1 Planned PK, safety
SHINE –Treatment shortening in children with paucibacillary TB British MRC2/ DFID3/Wellcome Trust Planned Efficacy, safety, PK
Pharmacokinetics of first-line agents in infants TB Alliance Planned PK
Rifapentine+isoniazid in children in LTBI5 TBTC4 Planned PK, safety
Levofloxacin and isoniazid in children exposed to DR-TB IMPAACT Planned Efficacy
1

International Maternal Pediatric Adolescent AIDS Clinical Trials Group

2

Medical Research Council

3

Drugs For Neglected Infectious Diseases

4

Tuberculosis Trials Consortium

5

Latent TB Infection

Ethical considerations

Children are a vulnerable group with limited or developing autonomy and are legally disallowed from providing informed consent. Children, therefore, require special measures to protect them from exploitation and harm. Many international guidance documents and regulations specify acceptable risk/benefit ratios and require that research involving children offers a prospect of benefit, or poses minimal risk. 39 An acceptable risk/benefit balance for the involvement of children in clinical trials depends not only on the risk/benefit ratio of a study for the individual child, but also on the available alternatives and the social value of the research 40, which for children depends on the burden of the disease being studied and the need for the intervention in that population.

A concern is that involvement of children in research at earlier stages of drug development may expose them unnecessarily to investigational drugs with uncertain future and undocumented safety risks. Some ethics guidance documents require that children be enrolled in research only if the research cannot be conducted in adults. 41 Others propose initiating pediatric studies (Phase I or II), particularly in children with serious and life-threatening diseases who could benefit from the study intervention, after obtaining preclinical safety data and evidence of efficacy from adult studies. 42, 43 Both TB in young children, 10 and DR-TB in all children, 20, 44 are serious and life-threatening conditions with limited treatment options, with affected children potentially further harmed by the dearth of data to guide use of existing drugs. Thus, TB drug research can and does, in such instances, offer a prospect of direct benefit to children that outweighs the risks of proceeding to pediatric trials with relatively incomplete (i.e. before Phase III) adult data. For these reasons, the earlier involvement of children in specific TB trials may be ethically justified.

Regulatory environment

Although drug regulatory legislation in both the United States (US) 45 and Europe 46 similarly provides incentives for the inclusion of children as part of any product’s development plan, the requirement for studies in children in an orphan indication differ. In the US, drugs intended to treat TB generally qualify for orphan designation under the orphan drug regulations, 47 and inclusion of children in pre-licensure trials is not required. 45 Similarly, drugs intended to treat TB can enjoy orphan medicinal product status in Europe, however no exemption is given for inclusion of children in trials investigating new TB therapies, and a pediatric investigation plan has to be agreed with the European Union regulators. 48 In South Africa, the Medicines Control Council (MCC) pays special attention to the conduct of research in minors, ensuring that clinical trials in TB are consistent with the National Health Act. 49 To ensure that prospects of direct benefit accrue to the participant, the Act requires that all research be therapeutic; non-therapeutic trials must be specifically authorized and deemed to contribute significantly to generalizable knowledge.

SUMMARY OF EXPERT PANEL CONSENSUS

The following sections summarize consensus statements around four main questions and topics.

1. What types of drugs or regimens should be prioritized for clinical trials in children?

When considering a new TB drug or regimen for study in children, characteristics (preclinical and adult clinical data) that suggest outcomes at least as favorable as established alternatives in the study setting should guide the TB drug/regimen selection and prioritization. These characteristics include the following: (a) similar or improved efficacy/effectiveness compared to an available alternative; (b) improved safety/toxicity and tolerance profile as compared to a standard regimen; (c) prospect for TB treatment shortening or simplification of the administration schedule; (d) prospect for administration of a fully oral regimen; (e) fewer drug-drug interactions, especially with antiretroviral drugs; and (f) availability in an appropriate formulation for dosing the targeted age group(s).

Development of child-friendly formulations for accurate pediatric dosing is important, and planning should be initiated once minimally acceptable adult safety data have been constituted, sufficient pharmacokinetic and pharmacodynamics information is available, and when at least an efficacious dose range in adults has been established (Phase IIA results are available). Care must be taken to investigate tolerability, palatability and formulations (e.g., fixed dose combination, dispersible pills, granules, or sprinkles) for children across the pediatric age spectrum. However, the development of an appropriate formulation to allow accurate pediatric dosing, while preferred, should not delay the initiation of clinical trials in children but can be developed in parallel.

When designing trials for new regimens, in addition to the criteria for drug prioritization, the practicality and clinical effectiveness of any new drug or regimen needs to be considered. Key principles that ensure correct treatment and ease of programmatic use in high-burden settings in particular should be followed, and the feasibility for use in resource-limited settings (e.g. the need for refrigeration and the shelf-life of a drug) considered. 50 Table 2 presents the World Health Organization (WHO) classification of existing TB drugs and Table 3 briefly summarizes existing information on selected priority TB drugs in children including criteria for their prioritization and lists current knowledge gaps.

Table 2.

WHO grouping of drugs used for DR-TB

Group Drugs (abbreviations)
Group 1:
First-line oral agents
pyrazinamide (Z)
ethambutol (E)
rifabutin (Rfb)
Group 2:
Injectable agents
kanamycin (Km)
amikacin (Am)
capreomycin (Cm)
streptomycin (S)
Group 3:
Fluoroquinolones
levofloxacin (Lfx)
moxifloxacin (Mfx)
ofloxacin (Ofx)
Group 4:
Oral bacteriostatic second-line
Agents
para-aminosalicylic acid (PAS)
cycloserine (Cs)
terizidone (Trd)
ethionamide (Eto)
protionamide (Pto)
Group 5:
Agents with unclear role in
treatment of drug resistant-TB
clofazimine (Cfz)
linezolid (Lzd)
amoxicillin/clavulanate (Amx/Clv)
thioacetazone (Thz)
imipenem/cilastatin (Ipm/Cln)v high-dose isoniazid (high-dose H)b
clarithromycin (Clr)

Table 3.

Information on selected priority TB drugs in children1, prioritization criteria and knowledge gaps

Current Drugs of
interest
(Drug Class/
Mechanism of
Action)
Current drug knowledge Criteria for prioritization Priority
conditions
/subgroups
Knowledge gaps
Better
efficacy
Better
toxicity
profile
Treatment
shortening
Fully oral
Regimen
Fewer DDI2 DR TB <5 years HIV3
Delamanid433
(Dinitroimidazole/
Inhibits mycolic
acid synthesis)
-Phase IIB data; adult Phase III studies underway
-Good safety profile
-Lacks interaction with CYP 450
-attractive for ARV co-administration
Concern:
-QTc prolongation
x75, 76 x ? x x x x x77, 78 -Optimal dosing in young
children
-Safety profile in children
-Drug interactions with ARVs
Bedaquiline5
(Diarylquinoline/
Inhibits bacterial
ATP synthase)
-Promising efficacy,
CYP 3A4 substrate, Long half-life,
efficacious dose available
-full efficacy studies underway
Concern:
-higher death rate in treatment group
-QTc prolongation
x - ? x - x x x -No PK/PD information
-Optimal dosing in children
-Drug interactions with ARVs
Rifapentine5
(Rifamycin/
inhibits DNA-
dependent
RNA polymerase)
-Low MIC, long half-life, potent
activity against TB
-Effective in once-weekly treatment of LTBI
Concern:
-hypersensitivity
x x79 x x - - x x -Optimal dosing in very
young children
Levofloxacin &
Moxifloxacin5,6
(Fluoro
Quinolones/
inhibit
topoisomerase
II DNA gyrase)
Levofloxacin:
-Approved in children for anthrax, plague;
-Dosing from CAP2 studies;
-solution formulation
Moxifloxacin:
-potential for shortening TB treatment but
current data conflicting
Concern:
-arthropathy, tendon rupture, nerve damage 80,
QTc prolongation
x x ? x x x x x -Optimal dosing in young
children
-Role in TB treatment
Linezolid6
(Oxazolidinone/
Inhibits protein
synthesis by
binding to 23S
ribosomal RNA)
-Approved for multiple indications other than TB
from birth -17 yrs. Used in DR-TB with some
success
No QTc prolongation
Concerns:
-Visual loss, neuropathy, lactic acidosis,
-Myelosuppression
x8184 - ? x ? x x x -Optimal dosing in
TB/efficacy at reduced doses
-Toxicity at prolonged
exposure
Clofazimine5
(Riminophenazine/
Unclear, possible
production of
reactive oxygen
species of M.tb)
Prolonged half-life
Skin discoloration (reversible),
Concern:
-QTc prolongation
x - x x ? x ? x -Dosing in TB not established
-No data from juvenile animal
studies
-no PK/PD data available
PA-82485
(nitroimidazole/
Inhibits M.tb F420-
dependant
synthesis of protein
and cell wall lipids)
-Adult Phase IIB studies underway
-Good safety profile
-Potential for shortening TB treatment (DS-and
DR-TB)
-Lacks interaction with CYP 450
-Attractive for ARV co-administration
Concern:
-None to date
x x x x x x x x -Optimal dosing in children
-Safety profile in children
-Adult efficacy studies not yet
complete
-Possibility for Rx shortening
-Drug interactions with ARVs

“X”: some evidence to support effect/ proposed use;

“-“: some evidence against effect/proposed use;

“?”: lack of evidence for/against effect/proposed use

1

Drugs are not listed in order of priority and absence from this list does not necessarily indicate low priority

2

DDI: drug-drug interactions; CAP: community acquired pneumonia

3

Or other immune compromised states

4

Delamanid has received conditional market authorization from the EMA

5

Bedaquiline, Rifapentine and Quinolones (except Levofloxacin) are licensed in adults, but not children

6

Levofloxacin, Linezolid and Clofazimine are licensed in both adults and children

2. Which populations of children are among the highest priority for study?

Once sufficient adult safety and efficacy data are available (discussed in the following section), it is recommended to initiate pediatric Phase I and II clinical trials. To this end, certain pediatric populations should be prioritized based on their medical needs. The greatest need for more effective, child-friendly, and less toxic drugs/regimens is in the management of children with DR-TB, 20 children in younger age groups 10, 51, 52 and, for preventative treatment, in those children exposed to or infected by an index case with DRTB. 5356 Studies are particularly essential in children under 2 years of age (with specific inclusion of infants) in whom pharmacokinetics may be substantially different compared to older children and adults. 57 Children with conditions such as HIV infection or malnutrition that: (a) increase their vulnerability to TB, (b) influence the pharmacokinetic profiles of TB drugs, or (c) increase the likelihood of drug interactions, are also important populations to prioritize for studies. 4, 5860

Although evaluation of efficacy is not the main objective of studies of new drugs/regimens in children, optimizing benefit for and limiting unnecessary risks in children continue to be major driving aims of pediatric studies. Therefore, only children with a diagnosis of confirmed or probable TB as per published case definitions for DS or DR-TB should be enrolled in TB treatment trials. 6163 Similarly, only children with documented significant exposure to DR-TB and evidence of infection as per accepted definitions (e.g., positive result from a tuberculin skin test or interferon gamma release assay) should be enrolled in TB prevention trials of new drugs/regimens.

3. When can Phase I or II studies be conducted in children, and what data are needed to facilitate their inclusion?

Risks from initiating trials of new TB drugs at earlier phases of TB drug development can be mitigated when sufficient adult pre-clinical and/or clinical data are available to allow adequate evaluation of the risk/benefit ratio. Enrollment of children in TB drug research is acceptable when the following are available: (a) results are available from a full range of non-clinical studies including repeated dose toxicity studies of appropriate duration in adult animals, (b) a complete package of safety pharmacology and genotoxicity studies and appropriate juvenile animal toxicity studies are available, and when those results do not indicate serious cause for concern; (c) animal and adult human studies (early bactericidal activity [EBA] or other appropriate studies) have confirmed anti-Mycobacterium tuberculosis (M.tb) activity; (d) data on drug pharmacokinetics and pharmacodynamics in adult participants allow for the selection of appropriate pharmacokinetic targets in children or, alternatively, an efficacious and safe adult dose has been established (Phase IIB); and (e) for HIV-infected children, drug interaction information with antiretroviral drug(s) of interest is available from adult studies prior to opening similar studies for the pediatric population(s) of interest. Concurrent evaluations of more than one (unapproved) drug in a TB regimen may be appropriate when such studies have already been completed in adults and have acceptable safety, efficacy, and pharmacokinetic profiles with manageable drug-drug interactions. 64

When the above criteria are met, a smaller safety database or a higher threshold for acceptable risk may be acceptable for initiating studies in pediatric groups with the greatest medical needs. In most situations, safety data from Phase IIB trials in adults should be sufficient to allow for determination of an acceptable risk/ benefit profile for children. However, before undertaking pediatric studies, the following steps should ideally be in place: development of child-friendly formulations, and a feasible pediatric investigation plan. Therefore, TB drug developers should consider preparing for pediatric studies when a drug shows promising efficacy and safety in Phase IIA adult trials.

4. What are the relevant elements of clinical trial design?

Investigational approach

General considerations

Efficient and ethical study designs that produce the highest achievable quality of evidence should be adopted to determine the doses that are safe and achieve pharmacokinetic goals. This will help to limit the number of children exposed to experimental doses of a new drug or treatment regimen. Based on developmental pharmacokinetic principles, particularly rapid pharmacokinetic changes are expected in the first weeks of life, 65 while after 2 years of age, allometric scaling for size will, in the case of many drugs, allow prediction of pharmacokinetic targets based on those in adults. However, differences are expected between pediatric age groups. Therefore, the following age groups are proposed, as a guideline, for pediatric pharmacokinetic evaluations: 0 - < 3 months, 3 - < 24 months, 2 - < 5 years, 5 - 10 years, and > 10 years of age to adulthood. In most instances, novel TB agents should be evaluated in children concurrently receiving appropriate standard of care TB treatment. For children with mild disease, initial single agent therapy may be considered for pharmacokinetic studies, typically for up to 2 weeks.

Placebo-controlled studies are not generally necessary or helpful in children if the novel TB agent has proven efficacy in adult studies and sufficient adult data exists to suggest initial safe pediatric dosing. Use of a placebo should only be considered when (a) there is an extraordinary scientific need to evaluate complex toxicity and tolerance issues in children; (b) when placebo use does not pose a risk of serious harm or risk to trial feasibility; and (c) if the research addresses a question that is relevant to health priorities in the countries where it is undertaken. 43 Situations where placebo use may be appropriate include when there is need to evaluate safety signals for novel therapies or in situations where a high background of adverse events from the disease or from co-administered medications is anticipated.

Study outcomes and extrapolation of adult efficacy data

With the scarcity of data for drugs and regimens in children, evaluation of a new drug or regimen should preferably include the following outcomes: (a) equivalent serum concentrations to those achieved in adults at optimal dose including formulations bioequivalence studies, (b) safety and tolerability of child-friendly formulations, and when feasible or appropriate (c) time to culture conversion, mortality and morbidity data. Extrapolation of adult efficacy data to pediatric populations limits the number and size of pediatric trials while allowing efficient use of resources. As a result, children can have earlier access to safe, efficacious, and evidence-based therapies. Extrapolation is possible when the following three assumptions apply: a) the progression of disease is sufficiently similar in adults and the pediatric populations; b) response to intervention is similar in adults and the pediatric populations and, c) adult and pediatric populations have a similar exposure-response relationship. (See Figure 2) 6668 Thus, efficacy studies in children for new drugs for intrathoracic TB, may not be necessary to allow for pediatric labeling. Similar response to treatment and exposure-response relationships in adults and children can be assumed for intrathoracic TB. However, efficacy studies may be needed for extrathoracic forms of TB and prevention studies in children.

Figure 2. Pediatric studies decision tree.

Figure 2

Reproduced from: http://www.fda.gov.ezproxy.nihlibrary.nih.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM072109.pdf. Abbreviations: PK/PD: pharmacokinetics/pharmacodynamics; ER: exposure-response

Enrollment strategies

Although sometimes cited as an important safety protection, 69 enrollment strategies using sequential age de-escalation are not currently required by any regulatory body and delay drug evaluation in the youngest age groups. 70 If the TB drug to be evaluated in children does not exhibit any significant safety signals in pre-clinical and adult clinical studies, pediatric studies should be allowed to proceed directly to concurrent evaluation across all pediatric age groups, to the extent that appropriate formulations are available. Particular emphasis should be placed on inclusion of the youngest children. Enrollment by sequential age de-escalation should be used only rarely, such as when specific safety or pharmacokinetic concerns that warrant testing older children before proceeding to younger children are identified. Sequential enrollment of age cohorts may actually raise ethical concerns by delaying collection of critical pharmacokinetic and safety data in the age groups that are most likely to benefit from a new agent or regimen.

Regardless of the approach used, sufficient evaluable subjects within each age cohort must be included to strengthen the quality of evidence generated. Furthermore, and to the extent possible, Phase IIB and later phase studies in adults should be designed to enroll children aged > 10 years, who are expected to have TB disease presentations similar to adults and are able to routinely provide sputum specimens due to adult type intrathoracic TB disease. Weight (and body surface area) differences within this group should be taken into account when establishing a- correct dose. Experts in studies of adolescents should be available to the investigators, and safeguards for protection of pediatric participants should be in place. Alternatively, if no expectation exists that the drug may interfere with progression through puberty or have a different safety profile in adolescents, the drug should be licensed for use in that age group without waiting for specific adolescent studies.

Dosing approach

Pharmacokinetic evaluation of single-dose administration of new drugs should be considered as a first step to inform multiple-dose pharmacokinetic studies; this approach has the potential to minimize risks of unwanted drug exposure. Alternatively, multiple dosing in a mini-cohort (i.e., using an initial sample size of no more than 3–6 children) can be used initially to provide preliminary safety and pharmacokinetic data while exposing fewer children. Subsequently, a final recommended dose may be established using a larger cohort. Modeling and simulation should be used to predict initial dosing in children for each age category. Selection of the initial dose in children can be informed by semi-mechanistic models adjusted for weight and other age-related changes such as volume of distribution, metabolizing enzyme maturation, and rate of drug excretion. As they become more accurate, physiologic-based pharmacokinetic models may increasingly contribute to initial dose selection. 71 Both safety and pharmacokinetic data from children should be incorporated into these models as soon as they become available, and should be used to improve subsequent dose prediction in successive cohorts of children. Real-time drug concentration analysis in individual study participants and multiple interim analyses of drug exposures in small cohorts within studies allow reduction of risk through rapid dose adjustment in individuals and cohorts.

Pharmacokinetic sampling strategies

Approaches that include methods to minimize pharmacokinetic sampling and sample volumes, rapid analysis of pharmacokinetic results to inform more accurate dosing in adaptively designed studies, and stratification by age, weight, and drug formulation schemas should be used when appropriate. Semi-mechanistic pharmacokinetic modeling using a population approach can enable opportunistic sampling and facilitate the use of variable sampling times and relatively sparse sampling schedules. The efficiency of this approach is enhanced further by the use of optimal sampling designs based on knowledge of the drug’s pharmacokinetics, limiting the number of blood samples needed from each child. Special attention must be paid to the volume of blood sampled and the timing of samples in very young children.

Additional design considerations

In order to guide dose adjustments, important drug-drug interactions should be specifically studied in young children receiving TB treatment. The magnitude of drug-drug interactions in this age group may not be predicted by either adult studies or other pediatric age cohorts. As with adults, children should be recruited from diverse racial and ethnic backgrounds to explore relevant pharmacogenomic differences. 72 Safety and adverse event data should be disaggregated and analyzed by age group. Data pooling can be used to generate models from diverse sources. Mechanisms using similar study designs (as appropriate) and standardized data collection forms and procedures should be supported to facilitate both collaborative data sharing, combined analyses across studies and incorporation into models. Children aged 0–3 months usually benefit from specific pharmacokinetic and safety evaluations. Studies should plan to extend duration of drug treatment in children who tolerate the drug (and experience no safety issues), if it is expected to add benefit to the standard of care; however the duration should not exceed length of treatment from adult studies. These methods may increase the prospect of direct benefit from the intervention and allow for collection of extended safety data and limited treatment response data with more prolonged exposure.

Safety monitoring and long term follow up

Safety monitoring principles

Due to major biological differences between children and adults, adverse event (AE) profiles and drug interactions that occur in pediatric patients may not be exactly as predicted by adult studies. Depending on the drug(s) being investigated or anticipated AEs, initial dosing in an in-patient setting or other intensely monitored study setting may be warranted. Special monitoring (e.g. EKG monitoring or other specific laboratory measurements) may/may not be needed if data from adults do not suggest any specific associated toxicity. Passive reporting should be used only when safety parameters of the agent have been well defined in children. Caregivers should be carefully instructed and encouraged to promptly report observed signs and symptoms to investigators. Establishment of independent safety monitoring committees (SMC), which include experts in pediatric pharmacology, pediatric TB experts, researchers with special experience in pediatric trials, or other specialists as needed, can provide additional protection.

Participant long term follow-up

The need for long-term follow-up for specific populations and for particular study agents should be given special consideration and should be adapted to settings where the study is conducted and where the drug will be used after registration. In addition, while the safety profile from adult trials has relevance for children it may be less helpful when predicting late adverse effects on growth, development and maturation. Hearing loss for example, a known potential complication of aminoglycoside use was reported in 24% of children with DR-TB treated with an aminoglycoside, much higher than that seen in adults, with several patients continuing to have progression of hearing loss months after discontinuation of medication. 19

Long-term follow up and/or drug registry and surveillance data, may be needed to determine possible late effects on skeletal, behavioral, cognitive, sexual, and immune developmental maturation. The duration of follow-up can be drug-specific, based on any signal or concern uncovered during pre-clinical studies or in earlier phase studies in adults. In studies for DR-TB in children, at least 24 months follow-up after treatment completion should be considered routine, since toxicity for some agents is duration-dependent, and the risk of disease relapse is greatest within the first year after treatment completion. Lastly, post marketing surveillance and patient registries may provide additional safety information that could not be detected from the limited pediatric exposures from clinical trials. In particular, post marketing pharmacovigilance activities, because of the greater cumulative drug exposures in the post marketing safety database, could detect rare, serious and/or patient-specific AEs.

FURTHER ISSUES TO BE ADDRESSED

Improving the understanding of TB treatment in children requires participation of all stakeholders involved in TB drug research design and implementation. Regulators should evaluate the existing options to harmonize requirements and streamline processes for pediatric drug development. For new TB drugs, regulators should require and agree upon a formal, time-bound pediatric development plan that includes the development of child-friendly formulations earlier in the drug development cycle. Investigators are encouraged to include children as soon as possible in studies, with appropriate safeguards, and should prioritize research questions most in need of answers, as described earlier. Drug companies/sponsors should initiate pediatric studies at the time points suggested previously, even if not a specific regulatory requirement. Sponsors should make all relevant information, not just safety and dosing information, available to facilitate further evaluation by research consortia and other non-commercial research bodies, particularly when multiple new compounds may be utilized in combination. Sponsors, in conjunction with investigators and community groups, should encourage the inclusion of children > 10 years in initial treatment trials in adults. Advocates should call for clear, harmonized guidance from regulators including requests for: early development of child-friendly drug formulations, the inclusion of children in drug safety, dosing, and efficacy trials, and better understanding of global and local pediatric TB disease burdens. They will also need to advocate for increased funding of pediatric TB research. 37, 73 New mechanisms of collaboration should be developed among all these stakeholders, such as the formation of a standing group to facilitate the earliest possible sharing of data and information on new TB drugs (to help determine when they should enter trials), make coordinated decisions and plans, enable harmonized approaches and address priorities on a consistent, ongoing basis, while minimizing resource duplication.

CONCLUSION

The scarcity of research of TB treatment in children represents a critical gap in global efforts to lessen the burden of TB infection and disease and to control the spread of drug resistance. Children, despite their increased vulnerability to TB, are subject to TB underreporting, and in some settings are at highest risk for exposure, infection and serious TB disease. Extrapolating from the adult treatment experience and adult clinical data may be inadequate for post licensure use of TB drugs in children, even if adult information suggests an acceptable risk/benefit ratio for children. Inclusion of children into studies should occur at the early phases of TB drug development and be an integral part of the clinical development plan, rather than as a post-approval activity. The consensus presented in this paper addresses ethical, regulatory, and methodological considerations that take into account the interests of and features unique to children, and promotes bold concepts that should accelerate the involvement of children in safe, ethical trials of new TB drugs and at earlier stages of the drug development cycle.

Acknowledgement

The authors are grateful to the following individuals for assistance for the planning and conduct of the workshop and/or drafting of the manuscript: Sheryl Zwerski, Sarah Read, Larry Fox, Devasena Gnanashanmugam, Judi Miller, Ellen O’Gara, Paul Sato, Peter Kim, Tyseaia Squirewell McFarlane, Rahel Abebe, Brenda Collins, Andrea Williams. This work was supported with funds from NIAID/NIH. This project has also been funded in part with federal funds from the National Institute of Allergies and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under Contract No. HHSN272200800014C.

Potential conflicts of interest (COI).

The following authors have listed these COI: EL: EL’s employer, the Treatment Action Group (TAG)'s TB/HIV project receives non-commercial support from the Bill & Melinda Gates Foundation and the TB Alliance. These institutions fund research for the development for TB of bedaquiline, clofazimine, a quinolone (moxifloxacin) and PA-824, a drug in the same class as delamanid. The TB Alliance funding that TAG receives is explicitly for the sole purposes of advocating for increased research funding for pediatric TB generally; it is not for a specific drug or regimen. TAG receives funding from the Bill & Melinda Gates Foundation to conduct advocacy to increase research into new tools to fight TB, and to improve access to existing, evidence-based tools and programs-- funding is not linked to advocacy deliverables around any specific drug or regimen; received per diem and travel funding from the World Health Organization to provide community perspective in the Expert Group Meeting on bedaquiline. TAG’s TB/HIV project also receives funding from the U.S. Department of Veteran Affairs to coordinate community engagement for the U.S. Centers for Disease Control Tuberculosis Trials Consortium, which includes rifapentine in many of its trials; my salary is not funded by this source; DFMcN: is employed by Novartis who produces the drug Clofazimine; JS is a member of the Data and Safety Monitoring Board for pediatric trials of delamanid. EG, CM and SM are employed by the TB Alliance who develops new TB drugs/combinations that include bedaquiline, PA-824 clofazimine and moxifloxacin.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Authors’ contribution. All authors have provided significant intellectual contribution to the project by contributing to draft statements preparation for consensus, contributing to final consensus statements as panel members, drafting the article of sections thereof or revising it critically for intellectual content; or providing final approval of the version to be published

Disclaimers. The views expressed in written conference materials or publications and by speakers and moderators at HHS-sponsored conferences, do not necessarily reflect the official policies of the Department of Health and Human Services (DHHS) or individual DHHS or other US government agencies including the National Institute of Health, the Center for Disease Control and Prevention, the Food and Drug Administration or the US Agency for International Development; nor does mention of trade names, commercial practices, or organizations imply endorsement by the U.S. Government. The views expressed in this article are the personal views of the authors and may not be understood or quoted as being made on behalf of or reflecting the position of the European Medicines Agency or one of its committees or working parties

All other authors have indicated no potential COI.

References

  • 1.World Health Organization. [accessed 11/20/2013];Global tuberculosis report 2013. 2013 http://www.who.int/tb/publications/global_report/en/index.html.
  • 2.Marais BJ, Graham SM, Maeurer M, Zumla A. Progress and challenges in childhood tuberculosis. The Lancet infectious diseases. 2013;13(4):287–289. doi: 10.1016/S1473-3099(13)70031-8. [DOI] [PubMed] [Google Scholar]
  • 3.Perez-Velez CM, Marais BJ. Tuberculosis in children. The New England journal of medicine. 2012;367(4):348–361. doi: 10.1056/NEJMra1008049. [DOI] [PubMed] [Google Scholar]
  • 4.Marais BJ, Graham SM, Cotton MF, Beyers N. Diagnostic and management challenges for childhood tuberculosis in the era of HIV. The Journal of infectious diseases. 2007;196(Suppl 1):S76–S85. doi: 10.1086/518659. [DOI] [PubMed] [Google Scholar]
  • 5.Marais BJ, Rabie H, Cotton MF. TB and HIV in children - advances in prevention and management. Paediatric respiratory reviews. 2011;12(1):39–45. doi: 10.1016/j.prrv.2010.09.002. [DOI] [PubMed] [Google Scholar]
  • 6.Hesseling AC, Cotton MF, Jennings T, et al. High incidence of tuberculosis among HIV-infected infants: evidence from a South African population-based study highlights the need for improved tuberculosis control strategies. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2009;48(1):108–114. doi: 10.1086/595012. [DOI] [PubMed] [Google Scholar]
  • 7.Dangor Z, Izu A, Hillier K, et al. Impact of the Antiretroviral Treatment Program on the Burden of Hospitalization for Culture-confirmed Tuberculosis in South African Children: A Time-series Analysis. The Pediatric infectious disease journal. 2013;32(9):972–977. doi: 10.1097/INF.0b013e31828d9aa4. [DOI] [PubMed] [Google Scholar]
  • 8.Martinson NA, Moultrie H, van Niekerk R, et al. HAART and risk of tuberculosis in HIV-infected South African children: a multi-site retrospective cohort. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2009;13(7):862–867. [PMC free article] [PubMed] [Google Scholar]
  • 9.Pensi T, Hemal A, Banerjee T. Simultaneous HAART improves survival in children coinfected with HIV and TB. Tropical medicine & international health : TM & IH. 2013;17(1):52–58. doi: 10.1111/j.1365-3156.2011.02884.x. [DOI] [PubMed] [Google Scholar]
  • 10.Russell GK, Merle CS, Cooke GS, Casas EC, Silveira da Fonseca M, du Cros P. Towards the WHO target of zero childhood tuberculosis deaths: an analysis of mortality in 13 locations in Africa and Asia. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2013;17(12):1518–1523. doi: 10.5588/ijtld.13.0238. [DOI] [PubMed] [Google Scholar]
  • 11.Dramowski A, Morsheimer MM, Jordaan AM, Victor TC, Donald PR, Schaaf HS. Rifampicinmono-resistant Mycobacterium tuberculosis disease among children in Cape Town, South Africa. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2012;16(1):76–81. doi: 10.5588/ijtld.11.0360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.The Sentinel Project for Pediatric Drug-Resistant Tuberculosis and Treatment Action Group. “We Can Heal”: Prevention, diagnosis, treatment, care, and support: Addressing drug-resistant tuberculosis in children. 2013 http://www.treatmentactiongroup.org/sites/tagone.drupalgardens.com/files/201303/tag-we-can-healfinalsm_0.pdf.
  • 13.Mukinda FK, Theron D, van der Spuy GD, et al. Rise in rifampicin-monoresistant tuberculosis in Western Cape, South Africa. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2012;16(2):196–202. doi: 10.5588/ijtld.11.0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Schaaf HS, Moll AP, Dheda K. Multidrug- and extensively drug-resistant tuberculosis in Africa and South America: epidemiology, diagnosis and management in adults and children. Clinics in chest medicine. 2009;30(4):667–683. vii–viii. doi: 10.1016/j.ccm.2009.08.019. [DOI] [PubMed] [Google Scholar]
  • 15.Schaaf HS, Marais BJ, Hesseling AC, Brittle W, Donald PR. Surveillance of antituberculosis drug resistance among children from the Western Cape Province of South Africa--an upward trend. American journal of public health. 2009;99(8):1486–1490. doi: 10.2105/AJPH.2008.143271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zignol M, Sismanidis C, Falzon D, Glaziou P, Dara M, Floyd K. Multidrug-resistant tuberculosis in children: evidence from global surveillance. The European respiratory journal. 2012 doi: 10.1183/09031936.00175812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gegia M, Jenkins HE, Kalandadze I, Furin J. Outcomes of children treated for tuberculosis with second-line medications in Georgia, 2009–2011. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2013;17(5):624–629. doi: 10.5588/ijtld.12.0792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Seddon JA, Hesseling AC, Marais BJ, et al. Paediatric use of second-line anti-tuberculosis agents: a review. Tuberculosis (Edinburgh, Scotland) 2012;92(1):9–17. doi: 10.1016/j.tube.2011.11.001. [DOI] [PubMed] [Google Scholar]
  • 19.Seddon JA, Thee S, Jacobs K, Ebrahim A, Hesseling AC, Schaaf HS. Hearing loss in children treated for multidrug-resistant tuberculosis. The Journal of infection. 2013;66(4):320–329. doi: 10.1016/j.jinf.2012.09.002. [DOI] [PubMed] [Google Scholar]
  • 20.Dooley KE, Mitnick CD, Ann DeGroote M, et al. Old drugs, new purpose: retooling existing drugs for optimized treatment of resistant tuberculosis. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2012;55(4):572–581. doi: 10.1093/cid/cis487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Singh V, Kaur S. Multi-drug resistant childhood tuberculosis. Indian journal of pediatrics. 2011;78(4):456–463. doi: 10.1007/s12098-010-0344-2. [DOI] [PubMed] [Google Scholar]
  • 22.Thee S, Zollner EW, Willemse M, Hesseling AC, Magdorf K, Schaaf HS. Abnormal thyroid function tests in children on ethionamide treatment. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2011;15(9):1191–1193. i. doi: 10.5588/ijtld.10.0707. [DOI] [PubMed] [Google Scholar]
  • 23.Seddon JA, Furin JJ, Gale M, et al. Caring for children with drug-resistant tuberculosis: practice-based recommendations. American journal of respiratory and critical care medicine. 2012;186(10):953–964. doi: 10.1164/rccm.201206-1001CI. [DOI] [PubMed] [Google Scholar]
  • 24.UNITAID. [accessed 11/29/2013];Tuberculosis medicines technology and market landscape--1st edition. UNITAID. 2013 Sep; http://www.unitaid.org/images/marketdynamics/publications/UNITAIDTB_Medicines_Landscape-1st_edition.pdf.
  • 25.Donald PR, Ahmed A, Burman WJ, et al. Requirements for the clinical evaluation of new antituberculosis agents in children. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2013;17(6):794–799. doi: 10.5588/ijtld.12.0567. [DOI] [PubMed] [Google Scholar]
  • 26.Burman WJ, Cotton MF, Gibb DM, Walker AS, Vernon AA, Donald PR. Ensuring the involvement of children in the evaluation of new tuberculosis treatment regimens. PLoS medicine. 2008;5(8):e176. doi: 10.1371/journal.pmed.0050176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zumla A, Nahid P, Cole ST. Advances in the development of new tuberculosis drugs and treatment regimens. Nature reviews Drug discovery. 2013;12(5):388–404. doi: 10.1038/nrd4001. [DOI] [PubMed] [Google Scholar]
  • 28.Working Group on New TB Drugs. [accessed 11/20/2013];The Global TB Drug Pipeline. 2013 http://www.newtbdrugs.org/pipeline.php.
  • 29.Drugs WGoNT. The Global TB Drug Pipeline. 2013. [Google Scholar]
  • 30.Villemagne B, Crauste C, Flipo M, Baulard AR, Deprez B, Willand N. Tuberculosis: the drug development pipeline at a glance. European journal of medicinal chemistry. 2012;51:1–16. doi: 10.1016/j.ejmech.2012.02.033. [DOI] [PubMed] [Google Scholar]
  • 31.Provisional CDC Guidelines for the Use and Safety Monitoring of Bedaquiline Fumarate (Sirturo) for the Treatment of Multidrug-Resistant Tuberculosis. MMWR Recommendations and reports : Morbidity and mortality weekly report Recommendations and reports / Centers for Disease Control. 2013;62(RR-09):1–12. [PubMed] [Google Scholar]
  • 32.The Use of Bedaquiline in the Treatment of Multidrug-Resistant Tuberculosis: Interim Policy Guidance. Geneva: World Health Organization; 2013. 2013. [PubMed] [Google Scholar]
  • 33. [accessed 11/24/2013];European Medicines Agency recommends two new treatment options for tuberculosis. 2013 http://www.ema.europa.eu/docs/en_GB/document_library/Press_release/2013/11/WC500155472.pdf.
  • 34.Development GAfTD. TB Drug Pipeline. 2013 http://www.tballiance.org/downloads/Pipeline/TBA-Pipeline-Q3-2013.pdf.
  • 35.Global Alliance for TB Drug Development. [accessed 11/20/2013];TB Drug Pipeline. 2013 http://www.tballiance.org/downloads/Pipeline/TBA-Pipeline-Q3-2013.pdf.
  • 36. <SQ109.pdf>. [Google Scholar]
  • 37.Lessem E, Jiménez-Levi E Funding for Pediatric TB Research. [accessed 12/05/2013];Supplement to the 2013 Report on Tuberculosis Research Funding Trends, 2005–2012. 2012 http://www.treatmentactiongroup.org/sites/g/files/g450272/f/201310/TB_2013_TB_PEDS.pdf.
  • 38. [accessed 12/05/2013];Roadmap for childhood tuberculosis, towards zero deaths. 2013 http://apps.who.int/iris/bitstream/10665/89506/1/9789241506137_eng.pdf.
  • 39.Evans Chan T. The Search for Minimal Risk in International Paediatric Clinical Trials. Santa Clara Journal of International Law. 2006;5(1) [Google Scholar]
  • 40. [accessed 11/20/2013];World Medical Association Declaration of Helsinki - Ethical Principles for Medical Research Involving Human Subjects. 2008 doi: 10.1001/jama.2013.281053. http://www.wma.net/en/30publications/10policies/b3/index.html.pdf?print-media-type&footerright=[page]/[toPage] [DOI] [PubMed]
  • 41.Macrae DJ. The Council for International Organizations and Medical Sciences (CIOMS) guidelines on ethics of clinical trials. Proceedings of the American Thoracic Society. 2007;4(2):176–178. doi: 10.1513/pats.200701-011GC. discussion 8-9. [DOI] [PubMed] [Google Scholar]
  • 42.Gill D. Ethical principles and operational guidelines for good clinical practice in paediatric research. Recommendations of the Ethics Working Group of the Confederation of European Specialists in Paediatrics (CESP) European journal of pediatrics. 2004;163(2):53–57. doi: 10.1007/s00431-003-1378-5. [DOI] [PubMed] [Google Scholar]
  • 43.Council for International Organizations and Medical Sciences; World Health Organization. [accessed 11/20/2013];International ethical guidelines for biomedical research involving human subjects. 2002 http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResources/UCM049870.pdf.
  • 44.Seddon JA, Visser DH, Bartens M, et al. Impact of drug resistance on clinical outcome in children with tuberculous meningitis. The Pediatric infectious disease journal. 2012;31(7):711–716. doi: 10.1097/INF.0b013e318253acf8. [DOI] [PubMed] [Google Scholar]
  • 45. [accessed March 13-2014];TITLE IV—PEDIATRIC RESEARCH EQUITY ACT OF 2007. http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResources/UCM049870.pdf.
  • 46.Regulation (EC) No 1901/2006 of the European Parliament and of the Council of 12 December 2006 on medicinal products for paediatric use. 2006 [Google Scholar]
  • 47. [accessed 11/20/2013];Orphan Drug Regulations Final Rule June 12, 2013. 2013 http://www.gpo.gov/fdsys/pkg/FR-2013-06-12/pdf/2013-13930.pdf.
  • 48. [accessed 11/20/2013];Regulation (EC) No 141/2000 of the European Parliament and of the Council of 16 December 1999 on orphan medicinal products. 1999 http://ec.europa.eu/health/files/eudralex/vol-1/reg_2000_141/reg_2000_141_en.pdf.
  • 49. [accessed 11/20/2013];National Health Act 2004, Republic of South Africa. 2004 http://www.info.gov.za/view/DownloadFileAction?id=68039.
  • 50.Brigden G, Nyang’wa B-T, Cros Pd, et al. Principles for designing future regimens for multi drug-resistant tuberculosis. [accessed 11/24/2013];Bulletin of the World Health Organization. 2013 doi: 10.2471/BLT.13.122028. http://www.who.int/bulletin/online_first/13-122028.pdf. [DOI] [PMC free article] [PubMed]
  • 51.Drobac PC, Shin SS, Huamani P, et al. Risk factors for in-hospital mortality among children with tuberculosis: the 25-year experience in Peru. Pediatrics. 2012;130(2):e373–e379. doi: 10.1542/peds.2011-3048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vanden Driessche K, Persson A, Marais BJ, Fink PJ, Urdahl KB. Immune vulnerability of infants to tuberculosis. Clinical & developmental immunology. 2013;2013:781320. doi: 10.1155/2013/781320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Seddon JA, Hesseling AC, Finlayson H, et al. Preventive therapy for child contacts of multidrug-resistant tuberculosis: a prospective cohort study. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2013 doi: 10.1093/cid/cit655. [DOI] [PubMed] [Google Scholar]
  • 54.Seddon JA, Hesseling AC, Godfrey-Faussett P, Fielding K, Schaaf HS. Risk factors for infection and disease in child contacts of multidrug-resistant tuberculosis: a cross-sectional study. BMC infectious diseases. 2013;13(1):392. doi: 10.1186/1471-2334-13-392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chiappini E, Sollai S, Bonsignori F, Galli L, de Martino M. Controversies in preventive therapy for children contacts of multidrug-resistant tuberculosis. Journal of chemotherapy (Florence, Italy) 2013 doi: 10.1179/1973947813Y.0000000105. [DOI] [PubMed] [Google Scholar]
  • 56.Seddon JA, Hesseling AC, Finlayson H, et al. Preventive therapy for child contacts of multidrug-resistant tuberculosis: a prospective cohort study. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2013;57(12):1676–1684. doi: 10.1093/cid/cit655. [DOI] [PubMed] [Google Scholar]
  • 57.Thee S, Seddon JA, Donald PR, et al. Pharmacokinetics of isoniazid, rifampin, and pyrazinamide in children younger than two years of age with tuberculosis: evidence for implementation of revised World Health Organization recommendations. Antimicrobial agents and chemotherapy. 2011;55(12):5560–5567. doi: 10.1128/AAC.05429-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chintu C. Tuberculosis and human immunodeficiency virus co-infection in children: management challenges. Paediatric respiratory reviews. 2007;8(2):142–147. doi: 10.1016/j.prrv.2007.05.003. [DOI] [PubMed] [Google Scholar]
  • 59.De Maayer T, Saloojee H. Clinical outcomes of severe malnutrition in a high tuberculosis and HIV setting. Archives of disease in childhood. 2011;96(6):560–564. doi: 10.1136/adc.2010.205039. [DOI] [PubMed] [Google Scholar]
  • 60.Jaganath D, Mupere E. Childhood tuberculosis and malnutrition. The Journal of infectious diseases. 2012;206(12):1809–1815. doi: 10.1093/infdis/jis608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Graham SM, Ahmed T, Amanullah F, et al. Evaluation of tuberculosis diagnostics in children: 1. Proposed clinical case definitions for classification of intrathoracic tuberculosis disease. Consensus from an expert panel. The Journal of infectious diseases. 2012;205(Suppl 2):S199–S208. doi: 10.1093/infdis/jis008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Marais S, Thwaites G, Schoeman JF, et al. Tuberculous meningitis: a uniform case definition for use in clinical research. The Lancet infectious diseases. 2010;10(11):803–812. doi: 10.1016/S1473-3099(10)70138-9. [DOI] [PubMed] [Google Scholar]
  • 63.Seddon JA, Perez-Velez CM, Schaaf HS, et al. Consensus Statement on Research Definitions for Drug-Resistant Tuberculosis in Children. Journal of the Pediatric Infectious Diseases Society. 2013;2(2):100–109. doi: 10.1093/jpids/pit012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.McNeeley DRA, Lin J, Marais B. The Evaluation of New Antituberculosis Drugs in Children. Progress in Respiratory Research: Antituberculosis Chemotherapy. 2011:235–242. [Google Scholar]
  • 65.Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE. Developmental pharmacology--drug disposition, action, and therapy in infants and children. The New England journal of medicine. 2003;349(12):1157–1167. doi: 10.1056/NEJMra035092. [DOI] [PubMed] [Google Scholar]
  • 66.Food and Drug Administration. [accessed 11/24/2013];Guidance for Industry: Exposure-Response Relationships: Study Design, Data Analysis, and Regulatory Applications. 2003 http://www.fda.gov.ezproxy.nihlibrary.nih.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM072109.pdf.
  • 67.Dunne J, Rodriguez WJ, Murphy MD, et al. Extrapolation of adult data and other data in pediatric drug-development programs. Pediatrics. 2011;128(5):e1242–e1249. doi: 10.1542/peds.2010-3487. [DOI] [PubMed] [Google Scholar]
  • 68.European Medicine Agency. [accessed 11/24/2013];Concept paper on extrapolation of efficacy and safety in medicine development. 2013 http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2013/04/WC500142358.pdf.
  • 69. [accessed 12/03/2013];Presidential Commission for the Study of Bioethical Issues. Safeguarding Children. Pediatric Medical Countermeasure Research. 2013 http://bioethics.gov/sites/default/files/PCSBI_Pediatric-MCM_2.pdf.
  • 70.Halpern SD, Randolph AG, Angus DC. No child left behind: Enrolling children and adults simultaneously in critical care randomized trials. Critical care medicine. 2009;37(9):2638–2641. doi: 10.1097/CCM.0b013e3181a59357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Caldwell JC, Evans MV, Krishnan K. Cutting Edge PBPK Models and Analyses: Providing the Basis for Future Modeling Efforts and Bridges to Emerging Toxicology Paradigms. Journal of toxicology. 2012;2012:852384. doi: 10.1155/2012/852384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Leeder JS, Kearns GL, Spielberg SP, van den Anker J. Understanding the relative roles of pharmacogenetics and ontogeny in pediatric drug development and regulatory science. Journal of clinical pharmacology. 2010;50(12):1377–1387. doi: 10.1177/0091270009360533. [DOI] [PubMed] [Google Scholar]
  • 73.E J-L. Tuberculosis Research and Development: 2012 Report on tuberculosis research funding trends, 2005–2011. 2012 < http://www.treatmentactiongroup.org/tbrd2012>.
  • 74.Juan D. Guzman XM-Rn, Wellman Ribón Research and Development of New Drugs Against Tuberculosis. In: Bassam H, Mahboub MGV, editors. Tuberculosis - Current Issues in Diagnosis and Management. 2013. [Google Scholar]
  • 75.Gler MT, Skripconoka V, Sanchez-Garavito E, et al. Delamanid for multidrug-resistant pulmonary tuberculosis. The New England journal of medicine. 2012;366(23):2151–2160. doi: 10.1056/NEJMoa1112433. [DOI] [PubMed] [Google Scholar]
  • 76.Skripconoka V, Danilovits M, Pehme L, et al. Delamanid improves outcomes and reduces mortality in multidrug-resistant tuberculosis. The European respiratory journal. 2013;41(6):1393–1400. doi: 10.1183/09031936.00125812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Paccaly A, Petersen C, Patil S. AIDS. Wshington, DC: 2012. Absence of clinically relevant drug interaction between delamanid, a new drug for multidrug-resistant tuberculosis (MDR-TB) and tenofovir or lopinavir/ritonavir in health subjects. [Google Scholar]
  • 78.Peterson C, Paccaly A, Kim J. ICAAC. San Franscisco, CA: 2012. Delamanid, a New Drug for Multi-Drug Resistant Tuberculosis (MDR-TB), and Efavirenz Do Not Show Clinically Relevant Drug Interactions in Healthy Subjects. [Google Scholar]
  • 79.Sterling TR, Villarino ME, Borisov AS, et al. Three months of rifapentine and isoniazid for latent tuberculosis infection. The New England journal of medicine. 2011;365(23):2155–2166. doi: 10.1056/NEJMoa1104875. [DOI] [PubMed] [Google Scholar]
  • 80.Food and Drug Administration Drug Safety Communication. [accessed 11/24/2013];Fluoroquinolone Antibacterial Drugs: Drug Safety Communication- Risk for possibly permanent nerve damage. 2013 http://www.fda.gov/downloads/Drugs/DrugSafety/UCM365078.pdf.
  • 81.Kjollerstrom P, Brito MJ, Gouveia C, Ferreira G, Varandas L. Linezolid in the treatment of multidrug-resistant/extensively drug-resistant tuberculosis in paediatric patients: experience of a paediatric infectious diseases unit. Scandinavian journal of infectious diseases. 2011;43(6–7):556–559. doi: 10.3109/00365548.2011.564649. [DOI] [PubMed] [Google Scholar]
  • 82.Cox H, Ford N. Linezolid for the treatment of complicated drug-resistant tuberculosis: a systematic review and meta-analysis. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2012;16(4):447–454. doi: 10.5588/ijtld.11.0451. [DOI] [PubMed] [Google Scholar]
  • 83.Rose PC, Hallbauer UM, Seddon JA, Hesseling AC, Schaaf HS. Linezolid-containing regimens for the treatment of drug-resistant tuberculosis in South African children. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. 2012;16(12):1588–1593. doi: 10.5588/ijtld.12.0322. [DOI] [PubMed] [Google Scholar]
  • 84.Lee M, Lee J, Carroll MW, et al. Linezolid for treatment of chronic extensively drug-resistant tuberculosis. The New England journal of medicine. 2012;367(16):1508–1518. doi: 10.1056/NEJMoa1201964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Dawson R, Diacon A. PA-824, moxifloxacin and pyrazinamide combination therapy for tuberculosis. Expert opinion on investigational drugs. 2013;22(7):927–932. doi: 10.1517/13543784.2013.801958. [DOI] [PubMed] [Google Scholar]

RESOURCES