1,981
Views
3
CrossRef citations to date
0
Altmetric
Review

Opportunities for enteral drug delivery for neonates, infants, and toddlers: a critical exploration

ORCID Icon, ORCID Icon & ORCID Icon
Pages 475-519 | Received 23 Dec 2021, Accepted 06 Apr 2022, Published online: 29 May 2022

ABSTRACT

Introduction

The field of neonatal, infant and toddler pharmaceutical development is constantly improving, however a lag still remains in comparison to older children and adults. Their rapid anatomical, physiological and behavioral developmental rates pose extra challenges in diagnosing, treating, or preventing their disease. In turn, this brings complexity in formulating truly age-appropriate medicinal products that suit this heterogeneous pediatric subset. Progress in the availability of such products has ensued following the introduction of the 2007 European Union Pediatric Regulation, and in recent years, oral multiparticulate and dispersible solid formulations have gained interest alongside liquid formulations. However, the need is still great for dosage forms that do not compromise on pharmaceutical efficacy, safety and global accessibility in those aged under 2.

Areas covered

This article highlights some of the formulation challenges correlated with this age group and critically explores recent solid age-appropriate formulations and their administration devices for enteral drug delivery.

Expert opinion

There are many formulation requirements to consider when formulating drug products for children aged under 2. Efforts are required into understanding acceptability in this age group and of their carers, and whether innovation or optimization is required, to help guide formulators toward optimal approaches without impacting access.

1. Introduction

The 2007 European Union (EU) Pediatric Regulation has improved the research for developing children’s medicines, increased the quality of information concerning their safety, efficacy and dosing but also the availability of pediatric medicines designed with the age and capability of the unwell children in mind. With the regulation came the mandatory Pediatric Investigation Plan (PIP) scrutinized by the European Medicines Agency Pediatric Committee (EMA PDCO); safeguarding a pharmaceutical company’s consideration for the entire pediatric population for a new indication, a new route of administration or a new age-appropriate dosage form [Citation1] at early stages of development. The ICH E11 guideline on clinical investigation of medicinal products [Citation2] sets out the pediatric subsets to consider as shown below, based on developmental biology and pharmacology, for clinical study design:

- Preterm and term new-born infants (up to 27 days after birth)

- Infants and toddlers (28 days to 23 months)

- Children (2 to 11 years)

- Adolescents (12 to 16–18 years)

Article 10 and 11 of the regulation also allows for the application of ‘deferrals’ and ‘waivers’ The deferral provision allows pediatric research to occur when it is ethical and safe to do so by permitting a delay in commencement and/or completion of all or some PIP measures; its purpose is to prevent the delay of the adult product’s marketing authorization. However, it does elongate the time for pediatric formulations to be authorized and available. A waiver can be sought if the disease/condition indication does not occur in part of/the entire pediatric population or if it has likelihood for insignificant therapeutic benefit, effectiveness or safety over standard of care. This waiver could be a reason of the lack of advancement in certain pediatric therapies [Citation3]. However, there is also a provision in the EU regulation for off-label (OL) or off-patent (OP) medicines to be reformulated appropriately for children, with a 10-year Pediatric Use Marketing Authorization (PUMA) in return. Unfortunately, only 6 PUMAs have been approved and completed so far [Citation3], [a detailed table can be found in [Citation4]]. Thus, many older formulations commonly expose to risk of inappropriate dosing especially in under 2s, namely neonates [Citation5]. Balan et al., 2018 reviewed 101 studies [1996–2016] and showed that up to 99.5% of patients in neonatal intensive care units (NICU) were prescribed at least one medicine OL, with administered drug dose and age commonly associated with this OL prescribing [Citation6]. To quote Tomasi et al., 2017, ‘children are not small adults and neonates are not small children’ [Citation1].

‘Key binding elements’ in PIP decisions [Citation7] aid understanding of formulation quality requirements in early stages of its development. They revolve around developing product characteristics and of its delivery that foster patient ‘acceptability’ to ensure the formulation can achieve optimal patient adherence and thus treatment effectiveness. For example, the requirement of a particular pharmaceutical form, formulation, dose strength, administration route, possible excipient issues [e.g. level of exposure, type of excipient], administration device [e.g. syringe, nasogastric tube (NGT)], product manipulation (if any) to be detailed in the Summary of Product Characteristics (SmPC) [e.g. mix with food/drink], and palatability [Citation7,Citation8]. Regulations in the USA contain similarities to EU regulations. A comparison is detailed in [Citation9].

Ten years post-EU regulation implementation, the EMA reported that the research, development, and supply of pediatric medicinal products had increased but also that gaps remained, particularly in developing age-appropriate dosage forms (only 43 approved between 2007 and 2016 in EU [Citation1]), as seen in [Citation3].

Figure 1. Trends from completed PIPs for medicines authorized for the pediatric population between 2008–2016. from EMA 2017 10-year report [Citation3].

Figure 1. Trends from completed PIPs for medicines authorized for the pediatric population between 2008–2016. from EMA 2017 10-year report [Citation3].

Noticeably, neonates (term/preterm), infants, and toddlers fill the largest portion of this gap [Citation2] Hence this article is explicitly targeted toward providing a holistic awareness and update of the current standpoint on their dosing with appropriate formulations, to help inform on advancements made and existing limitations.

1.1. Neonate, infant, and toddler ontogenesis considerations

The lack of appropriate dosage forms is profoundly linked to their sharp dynamic increase in growth and maturation rate: not only visible height/weight changes but also organ maturation, body composition, developmental and behavioral changes during these two first years of life. This deepens the complexity to treat this subset as disease diagnosis can be blurred due to overlap in symptomatic presentation, and also since it creates a moving target product profile for formulation scientists.

The Infant and Toddler Forum proposes 5 key stages categories [Birth-1 month, 1–3 months, 3–6 months, 6–12 months, and 12 months-2 years] encompassing developmental transformations including cognitive understanding, recognition, (oral)motor functions, language, feeding changes from liquids to semi-solids, teething, and declination of gag reflex. Behavioral changes such as sleeping times, nappy habits, attitudes, taste/texture preferences, and teething discomfort could also have effects on medicine administration [Citation10]. Plausibly, children <2 years cannot proactively partake in their treatment to any extent, unlike older children, with complete dependence on their caregivers. Therefore, consideration weighs even more heavily toward ease of administration of the dosage form by the caregiver [Citation11].

There are still many knowledge gaps on the physiology of the pediatric gastrointestinal system relevant to the absorption of enterally administered medicines; how this system develops from birth, how it differs to adults, and how it changes with disease. Key physiological parameters of the gastrointestinal system relevant to the design of age-appropriate enteral drug products are discussed in [Citation12,Citation13]. Regarding pharmacokinetics, in the first two years of life, drug Absorption, Distribution, Metabolism and Excretion (ADME) profiles are greatly affected [Citation14–16]. Some of these physiological parameters such as, increased gastric pH, gastric emptying time, decreased absorption capacity, microbiome status, intestinal fluid composition, and diet are known to affect drug absorption (more so in preterm/term neonates) [Citation14,Citation15,Citation17]. These factors can show inter-individual variation, especially of the different developmental stages within these first two years; resulting in inter-individual drug absorption and exposure. The European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) recognize this and use physiologically based pharmacokinetic modeling (PBPK) as a method to investigate formulation approaches closer during the drug development stages to reduce this pharmacokinetic variability [Citation17]. Drug distribution can also be affected due to lower plasma protein concentrations, higher body water content, and lower blood volumes. From birth, immaturity in metabolic processes and lower body fat content can result in reduced drug clearance and increased drug half-life. Immaturity in glomerular filtration, tubular secretion, and reabsorption can also cause a reduction in renal drug excretion [Citation14,Citation15]. Unlike in older children, these factors mature and change rapidly at different rates between birth and the second birthday, creating a need for dosing regimens to be adapted. shows the sharp increase in weight [approximately triples] from term birth to two years relative to the rest of the pediatric population. But for preterm neonates, their weight may increase threefold over several weeks when receiving postnatal care [Citation18]. Hence, dose adjustment constantly required brings constant concern/risk of mis-dosing, possibly even more to emotional parents returning home with their child from the hospital. Complex medicine regimes, polypharmacy, and difficulty in administration introduce dosing error risks and cause much anxiety as it is likely to cause significant effects the more vulnerable the patient is e.g. discharged babies form NICU. The PADDINGToN study [Citation19] shows that supporting medicine administration with technology such as a phone application or QR codes on packaging could help provide easy access to information and advice when required by parents.

Figure 2. Body weight as a function of age of children aged between term birth to 14 years. Data obtained from 2009 UK-World Health Organization (WHO) growth charts and 1990 UK standard centile charts as per the British National Formulary [Citation20].

Figure 2. Body weight as a function of age of children aged between term birth to 14 years. Data obtained from 2009 UK-World Health Organization (WHO) growth charts and 1990 UK standard centile charts as per the British National Formulary [Citation20].

Flexible and acceptable (swallowable, palatable, easy to administer) dosage forms are therefore desired to allow for safe dose adjustment and optimal medical treatment, without impairing the quality, performance, and commercial viability of the pharmaceutical product, principally when considering (preterm)neonates [Citation14,Citation16,Citation18].

1.2. Neonate, infant, and toddler excipient safety and administration considerations

Excipients generally recognized as safe for adults and possibly for older children/adolescents can be unsafe when dosed in under 2s due to their immaturity in ADME [Citation11,Citation18]. Those in intensive care are often polymedicated, leading to cumulative effects, drug interactions, and exacerbated adverse reactions, especially for preterm babies, with excipients in their treatment regime potentially exceeding the adult accepted daily intake. Therefore, when formulating suitable dosage forms for this group, the need for, type, and quantity of excipient used that does not deter its quality nor the bioavailability of the active pharmaceutical ingredient (API) should be considered [Citation2,Citation18,Citation21]. Recent papers [Citation22,Citation23] highlight excipient concerns and mention studies such as ▣Safe Excipient Exposure in Neonates and Small Children (SEEN) and ▣European Study on Neonatal Exposure to Excipients (ESNEE) and their generation of an ‘excipients of interest list’ that demonstrate the achievability of progress in this area. The safety, tolerability, and exposure of excipients remain an underlying core consideration but will not be central to this review as it has been extensively detailed elsewhere [Walsh, et al., 2021 give an overview [Citation4]].

1.3. Enteral feeding tubes

It is a norm for patients <2 years in ICU, or even preterm neonates returning home to have nutrition and treatment administered through enteral feeding tubes. Currently, many medicines given require unlicensed (UL) manipulation for this administration. Not only is there risk of incompatibility with enteral tubes and affecting the pharmaceutical quality and efficacy of the drug product but also these unstandardized manipulations introduce a true mis-dosing risk with possible significant side effects. This is the case especially for potent drugs given in small volumes. Liquids are more easily dosed through enteral tubes, however where solid formulations are concerned detailed guidance on suitable manipulation methods in SmPCs should be provided. Whether solid or liquid, future age-appropriate formulations should consider safe and efficacious administration (to the product’s best ability) through enteral tubes by undertaking feasibility studies [Citation24] if relevant to the condition treated.

1.4. Preferences toward route of drug delivery

Currently, treating children under 2 is skewed toward parenteral drug delivery, responding to a clinical need for fast and acute treatment of the seriously ill. For perspective, the European Union Clinical Trials Register (EudraCT) records 760 clinical trials conducted in children <2 since it began in 2004, of which ~72% were parenteral products [Citation25]. However, oral drug delivery remains highly desirable, as suggested by O’Brien et al, 2019 [Citation11], as it is better suited to long-term treatments, can be administered (or via enteral tubes) at home, and off-sets parenteral route limitations: punctuation and pain, risk of phlebitis, infection, electrolyte imbalance, fluid overload, incorrect dose volume measurement, and limited/difficult intravenous access [Citation2,Citation18,Citation26]. There is prospect for the rectal route of administration as it bypasses these limitations and those associated with oral delivery for fast treatment, but there is an attached stigma to this route.

Prior to the regulation enforcement, the 2006 EMA reflection paper was first to propose a matrix to evaluate ‘applicability’ (i.e. age suitability) and ‘preferability’ of oral dosage forms for children but only based on general opinion of a few healthcare workers and carers. outlines the variability in responses for preterm, term neonates, infants, and toddlers.

Figure 3. Healthcare worker and carer (N = 40) opinion on the ‘applicability’ of various oral dosage forms to preterm and term neonates, infants and toddlers. Adapted from EMA 2006 reflection paper [Citation27].

Figure 3. Healthcare worker and carer (N = 40) opinion on the ‘applicability’ of various oral dosage forms to preterm and term neonates, infants and toddlers. Adapted from EMA 2006 reflection paper [Citation27].

Tablets, capsules, and chewable tablets were equally considered ‘not applicable’ for all age groups but, with all other formulations becoming more ‘applicable’ and ‘preferable’ with age. Interestingly, multiparticulate and monolithic formulations (powders, orodispersible) likewise were ‘not deemed applicable’ at all for preterm neonates and just slightly more ‘applicable’ yet still problematic for term neonates, infants, and toddlers. Liquids were deemed to have the greatest ‘applicability’ and ‘preferability’ across all the age groups, the assumption being a lower aspiration/choking risk [Citation28] and that they can accommodate different age/weight-based doses via volumetric measurement (syringe, cup, and spoon). This highlights the deeper drug delivery gap for neonates. Although this study shows useful comparison between dosage forms, it is slightly outdated and shows how there is a lack of available and recent evidence that can be reflected upon to gain a more current perspective, which may include other types of dosage forms for alternative administration routes, e.g. the rectal route.

Liquid oral drug delivery has been the gold standard for pediatric dosing, but is it the golden standard? As part of this, the 2007 WHO ‘Make medicines child size’ initiative has raised awareness and shifted the paradigm from liquids toward flexible solid dosage forms: orodispersible tablets (ODT), dispersible/soluble tablets (DT) used to form a liquid preparation upon administration or smaller solids (i.e. multiparticulate) that may be mixed into a vehicle such as food, breastmilk to aid dosing particularly for <6 years [Citation29]. Recent publications such as Mfoafo et al., 2021 [Citation30], consider them as a future trend to oral drug delivery. Hence, this review will not explore liquid, tablet, and chewable formulations but will present the advancement, trends, age-suitability, and an evidence-based analysis of these novel dosage forms for neonates, infants, and toddlers in order to assess if they could offer new innovation-fits-all approaches. The rectal route of administration and its associated barriers will also be explored.

2. Criteria to assess the age-appropriateness of dosage forms and evidence available

Acceptability of dosage forms is a concept that has emerged in the past decade. The 2013 EMA document ‘Guideline on pharmaceutical development of medicines for paediatric use’ formally defines ‘acceptability’ as ‘The overall ability and willingness of the patient to use and its caregiver to administer the medicine as intended’ [Citation31]. It indirectly encompasses factors such as illness status, treatment duration, mood, cultural influence, and general habits [Citation27].

shows the framework that will be used for the purposes of this review, to consider the added values and limitations of commercially available enteral formulations for children under 2.

Table 1. Consideration criteria involved in assessing the age-appropriateness of a dosage form. Adapted from [Citation28,Citation29,Citation31]

As mentioned, there has been a shift away from oral liquids for children <2 years. Although advantageous in many regards, especially swallowability, oral liquids carry a higher risk of unsafe excipients and dosing error that oral solids can overcome. Illustrated by , in the past decade, there has been a surge of placebo clinical studies designed to generate evidence around acceptability of multiparticulates, DTs, ODTs, and orodispersible films (ODFs). More studies were found for multiparticulates, less so for ODFs, and none for (O)DTs, but it should be noted that this does not sway particular favorability toward multiparticulates.

Table 2. Recent placebo studies showing the shift and increase in acceptability toward solid formulations in children, participants including <2 years

These studies of children physically ingesting the dosage forms revealed that against general consensus, flexible solid dosage forms were acceptable, carrying less (if any) administration safety issues than expected, i.e. no significant cases of aspiration/choking. However, longer term usage, multiple (daily) administration and API inclusion were not explored, meaning palatability was not considered despite being a key limiting factor.

The driver for the switch to these flexible solid dosage forms is based on increased chemical and physical stability, fewer problematic excipients in comparison to liquids [Citation28], potential for more efficient taste masking, smaller administration volumes and no swallowability issues. Section 3.0 of this article details currently licensed solid oral multiparticulate and monolithic formulations for children <2 years. No systematic search strategy was used; products were found in the literature as a starting point and then refined using electronic sources such as the Electronic Medicines Compendium.

3. Oral solid dosage forms

3.1. Multiparticulate dosage forms

There is potential for flexible dosing of multiparticulates, providing a suitable device or unit-dose packaging is integrated into product development and/or available; a field that has developed quickly in parallel [Citation41].

lists multiparticulate formulations (including two of the six PUMAs approved) for use in the under 2s grouped by their therapeutic indications.

Table 3. Commercially available solid multiparticulate formulations for neonates, infants, and toddlers

It is to be noted that may not be exhaustive as there is no official, easily searchable database that lists all currently licensed pediatric medicines globally. It should be noted that although more commercially available multiparticulate formulations have been sourced, this does not show favorability bias toward this dosage from type compared to the others. Moreover, most limitations listed are from the authors’ interpretation and opinion as there is little public reports available.

3.1.1 – Minitablet Formulations

They are generally regarded as tablets measuring less than 4 mm in diameter [Citation42,Citation43], with similar low production costs but with enhanced swallowability. They can offer dose flexibility, but where smaller doses/counts are needed for younger/lighter children this can result in higher or significant risk of dosing error if counted inaccurately, leading to dosing devices or packaging considerations being central to this application. Additionally, the number of minitablets for higher doses should not reach uncomfortable levels for the child to swallow [Citation34,Citation42].

Whilst there are more formulations of the granule/sprinkle type compared to minitablets approved for use in the under 2s, there are more placebo acceptability studies present for minitablets. The primary study conducted by Thomson et al. in 2009 [Citation44] with participants 2–6 years, paved the way for further studies in younger children. These showed excellent acceptability and swallowability of at least 1 minitablet administered with <3 mouthfuls of drink in children as young as 2 days (including preterm neonates), with as many as 100 minitablets administered with a drink in infants as young as 6 months. When compared to the acceptability and swallowability of 15% glucose (0.5 ml in neonates; 3–5 ml in infants and toddlers), minitablets were always superior with parents being in their favor. Acceptability/swallowability of coated and uncoated minitablets were not significantly different. As these studies showed promising swallowability and ease of administration in neonates as young as 2 days, including preterm neonates, more licensed minitablets for under 2s might emerge in the coming years or as evidenced in some recent studies mentioned below.

To demystify concerns about swallowability safety in young infants, the concept of orodispersible minitablets has been pondered, e.g. Stoltenberg et al. 2011 [Citation45]. Before the age of 5 months, infants only swallow liquids safely due to an extrusion reflex. From 6 months they should be able to swallow soft foods. Thus, combining the benefits of a minitablet with properties of disintegration in a small amount of saliva seems a way forward to dose the youngest of patients.

Other minitablets are marketed for slightly older children: Slenyto (melatonin) ~3 mm diameter minitablet [>2 years for insomnia treatment [Citation46]] and PancreaseTM HL Capsules containing ~2 mm enteric coated minitablets [>15 years for cystic fibrosis exocrine insufficiency [Citation47]].

Merck & Co. addressed in a recent study [Citation43] unpleasant minitablet palatability (taste, texture) containing amorphous solid dispersion APIs (undisclosed), when administered with semi-solids i.e. applesauce (type unspecified). Their aim was to investigate a coating that prevented early API release to provide a pragmatic timeframe for administration whilst avoiding palatability issues that may deter the child from future dosing/feeding sessions.

3.1.2 – Granule, pellet, powder, sprinkle formulations

Granules are nonuniform in shape with a broad size distribution. Two placebo studies [Citation35,Citation37] (), showed good scores and suitability in children 1–4 years when administering granules with/without food or drink.

lists 21 formulations of which the majority are granules and only 1 pellet and 1 powder formulation. 12 are packaged in unit-dose sachets, 9 in unit-dose capsules and only one product (Creon® Micro Pancreatin 60.12 mg Gastro-resistant Granules, 2004) is in a multi-dose bottle with a dosing scoop. As mentioned earlier, there is a larger gap in drug development for neonates. This is evidenced in with only 7 licensed products for neonatal use: Lopimune® (lopinavir/ritonavir) and Co formulated lopinavir+ritonavir, Episenta® (sodium valproate) and Epilim Chronosphere® (sodium valproate/valproic acid), Alkindi® (hydrocortisone), Tamiflu® (oseltamivir phosphate) and Xuriden® (uridine triacetate). The other 14 are licensed for use in infants onwards.

Analyzing the SmPC’s shows some disparity/clarity of information provided for carers which may affect administration ease, for example, unspecifying the type or volume of dosing vehicle required. Too little may result in unpalatable taste/texture, with too much meaning incomplete dose ingestion. This may also become an issue when extra food/liquid needs to be added to mixing containers to disperse remaining product. Vague terms such as to mix with ‘food’ ‘liquid’ or ‘drink’ are used. This could pose incompatibility risks of reduced drug efficacy/taste masking if administered with certain foods/drinks, especially if a coating is disrupted.

Some additional information can make administration guidance clearer such as: recommendations to not administer via a feeding bottle as blockage may occur hindering accurate dosing, explicitly saying bottles may be used, stating no mixing with liquids due to effects on taste masking as well as ability to administer a full dose, recommending certain vehicle types e.g. acidic (fruit juice/sauce), fat-containing media or even avoidance of a food type (e.g. grapefruit). Epilim® Chronosphere (sodium valproate/valproic acid) seems to be the only example from making a clear statement in the SmPC that its drug PK is not altered when mixed with food. It should be noted that dosing vehicles to aid swallowing or taste can help to increase acceptability and adherence to the medicine (particularly for chronic use) and so, where warnings are provided in SmPCs such as those for Alkindi® (hydrocortisone) (i.e. no mixing with liquids), unless there is clear clinical evidence of significant negative effects, stating so should be carefully considered [Citation37].

14 of the 21 formulations have a coating. To note on palatability, 6 of the 21 formulations contain flavors including: strawberry, orange-vanilla, apricot, and natural orange juice and 7 of the 21 formulations contain sweeteners and/or polyols: sucrose, mannitol, glucose, aspartame, sucralose, and acesulfame potassium.

The CHAPAS-2 study for Lopimune® (lopinavir/ritonavir) illustrates the importance of palatability for children under 2. Uncoated granules dissolved in a small quantity of breast milk were administered to unweaned infants with a spoon or placed directly on the tongue before breastfeeding. For weaned infants/toddlers, unspecified amounts were mixed into porridge, with recommendations to add honey/sugar to enhance palatability. Although swallowability of granules was superior compared to the syrup equivalent in children aged between 3 months and 4 years, parents reported poor taste resulted in the child struggling to eat or perhaps later refusing feedings. There were concerns on how much honey/sugar may be required to make Lopimune® (lopinavir/ritonavir) more acceptable; especially for higher doses, with added concerns around extra expense, and that perhaps the syrup equivalent would ensure more dose ingestion [Citation48]. The LOLIPOP trial for Quadrimune® (abacavir/lamivudine/lopinavir/ritonavir) is underway with granules coated with strawberry flavor hopefully offering better taste. It should be noted that other palatability characteristics (texture: hardness, roughness, cohesiveness and fracturability/shape) as well as dose volume are concomitantly important for multiparticulate acceptability [Citation49,Citation50].

Although NGTs may be used as a method of drug administration, it cannot be expected that all formulations may be suited to this form of drug delivery. Only 4 products [Desitrend® (levetiracetam), Nexium® (esomeprazole), Vistogard® (uridine triacetate), Prevacid® (lansoprazole)] in mention that an NGT may be used. All four give instructions on its use but are not necessarily clear on details such as flush volume, composition/frequency of flushing, tube lumen diameter, compatibility data of the prepared formulation with the tube material or blockage issues. Dose/flush volumes listed may also seem unsuitably high for the target age especially if fluid restricted [Desitrend® (levetiracetam) and Vistogard® (uridine triacetate)].

A recent Q&A on the quality of medicines by the EMA [Citation24] outlines what data is required to show a formulation’s feasibility to be administered optimally via NGT as well as what should be included in the SmPC and patient information leaflet (PIL). For the former, feasibility studies should encompass how modifying the drug product may affect bioavailability or risk of tube blockage: fine dispersal/complete dissolving of the solid dosage form is required and particle size, dispersion medium/volume/viscosity, tube lumen diameter/material and data on dose recovery (>90% is acceptable), and minimum flush volumes (a consideration for fluid restricted patients).

3.2 – Monolithic dosage forms

Although monolithic formulations [DT and orodispersible dosage forms (ODT, ODF)] may be administered flexibly to a young child (e.g. ODT/ODF can be predispersed or swallowed whole), they are not necessarily as dose flexible as multiparticulate formulations since they may require manipulation e.g. splitting. They would require multiple dose strengths to be fully flexible [Citation11,Citation87]. Compared to conventional tablets however, they do not require crushing associated with potential dosing error and altered biopharmaceutical properties [Citation49]. Their stability is also regarded as being more challenging than conventional tablets due to their reactivity with moisture, yet are still more stable than liquid formulations [Citation28]. Overall, they offer another option to dose the <2 years. summarizes relevant key information on monolithic DT, ODT and ODF licensed currently for ages <2 years. In total, 11 products were retrieved; much less than for multiparticulates (), most being licensed after 2007. 1 of the 6 PUMAs approved to date is a DT [Kigabeq® (Vigabatrin)]. Only 4 of the 11 products, are licensed for use in neonates, with the other 7 for use from infancy. Like for , may not be a comprehensive inventory of all licensed DT, ODT, and ODF for under 2s, and should be noted that there is no favorability bias toward any one dosage from type based upon how many commercially available products have been sourced for each.

Table 4. Commercially available solid monolithic formulations: dispersible/orodispersible tablet/orodispersible film available for neonates, infants, and toddlers

3.2.1. – Dispersible tablets

The majority (8 out of the 13) of products are in are DT’s, with detailed methods of administration. However, instructions are not always clear, e.g. perhaps not specifying the volume of water required for dispersal which could affect acceptability.

5 of the 10 DT are flavored (strawberry, tutti frutti and orange). 7 of the 10 DT contain polyols and/or intense sweetener excipients (isomalt, sorbitol, sucralose, aspartame, sodium saccharin and acesulfame potassium).

3 formulations can also be administered via a device: i.e. syringe/NGT but a lack of details in the SmPC and PIL on administration through NGT is noticeable. The issue also presents that if tablet splitting is required, it is not clear if a lesser dispersion volume of water is required. Consider Ucedane® and Carbaglu® (carglumic acid), they are licensed for use from birth and are given across 2–4 daily doses using a volume of water in each administration appropriate for a whole tablet. This may be considered too high cumulatively, especially in fluid restricted patients. However, the 2013 EMA guideline [Citation31] advises against taking a portion of liquid prepared from a tablet that has been dispersed, suspended, or dissolved to form a flexible age-appropriate medicine for a pediatric patient; as risk of dosing error is increased with such multi-step procedures, unless it can be verified that the preparation is easily prepared, has homogeneity and the correct volume can be measured [Citation31].

With no placebo studies or insufficient acceptability studies, it is difficult to inform with evidence common limitations that may be present for DTs intended for under 2s, but others may include: product being left behind in the drinking glass/syringe during preparation, which can lead to significant dosing error if small dispersal volumes (5–10 ml) are used or drugs have a narrow therapeutic index (NTI), NGT blockages and poor palatability which may interfere with subsequent feedings.

Developments for more DT for the under 2s is underway. Eurartesim® DT for treatment of Malaria, is not currently licensed, with studies ongoing. It is a fixed-dose combination of Dihydroartemisinin-piperaquine phosphate. The tablet dispersed in 10 ml of water has the advantage of not requiring food to increase absorption, easy administration, better palatability compared to the crushed film-coated tablet (flavors and sweeteners included) and increased compliance [Citation88,Citation89].

3.2.2. – Orodispersible tablets/films

ODTs and ODFs are formulated to disintegrate in situ in the saliva to be swallowed [Citation87]. They can be useful for the delivery of larger drug doses in comparison to solutions and suspensions formed from DTs that can be higher in volume. Their rapid disintegration within the oral cavity and lack of requirement for solid/fluid vehicles also makes it easier (no extra steps) upon administration [Citation90]. However, incorporating high drug loads into these dosage forms can be problematic from a manufacturing perspective and may compromise on dosage form size. Therefore inclusion of more potent drugs are favored [Citation87]. Low drug loads per unit may also result in multiple administrations [Citation91]. There is still a large absence on studies examining acceptable sizes of ODT/ODF for young children, but generally they are larger than regular tablets [Citation50].

No placebo studies for ODT were found but two recent placebo studies looked at ODFs in children as young as 2 days showing good acceptability, with participant palatability and swallowability assessment favoring ODFs when compared to glucose syrup. Despite parents being unfamiliar with the dosage form, it was not a barrier in its administration, with children less likely to spit it out. The majority found 6cm2 film size manageable but stated that flavors (e.g. strawberry commonly suggested) and colors should be included, although that was deliberately avoided in the placebo study to avoid preference bias [Citation39]. Klingmann et al. 2020 [Citation40] did not intend to investigate administration of ODF without a dosing vehicle (milk, water, fruit juice, tea, and 15% maltodextrin solution); 80% of caregivers preferring milk. This reiterates milk, a substantial part of neonatal, infant and toddler diets as an obvious and practical vehicle. Further studies would be required to discern palatability issues as well as ODF size and appearance, which govern ingestion ease () in the acceptability of medicines. The importance of explicitly detailing in the SmPC precise instructions on the ODF’s correct administration seems paramount: the ODF in this study [Citation40] was intended to be orodispersible but not mucoadhesive and hence there were cases of the film not sufficient sticking to the cheek pouch, particularly in children <6 months, where part of it was retained on the feeding bottle/mother’s breast. Therefore, ODF adhesive property analysis is important during development, although there are no standardized approaches [Citation91].

Only two ODTs and one ODF were found to be licensed for children under 2 (). Only Prevacid® allows for alternative administration methods: syringe/NGT use and there are details in its SmPC on tube size and flush volume. These type formulations usually contain intense sweeteners, with both listed ODTs containing strawberry flavor; imperative taste masking aspects to cover poor API taste upon dispersal. It is to be noted that disintegration time would also carry an important acceptability aspect for the youngest of infants and children due to concerns of palatability and possible choking/aspiration hazards. Although sizes for Zofran melt® and Prevacid® (lansoprazole) could not be identified in the SmPC, it is intuitive that for under 2s, a suitable size for their oral cavity that can be retained safely and long enough for disintegration is required. Such aspects require further investigation. Concern of size is why ODminitablets (see 3.1) have been established to include advantages of both dosage forms [Citation49]. A 2019 paper illustrates ongoing Phase II/III clinical trials for a novel 2 mm ODminitablet of enalapril, currently used OL in children. Acceptability and palatability will be assessed in children with congenital heart disease (0 < 6 years) and dilated cardiomyopathy (1 month<12 years). These results can inform other product developments of this dosage form [Citation92].

Palatability (taste/mouthfeel) of ODT/ODF has been scarcely studied in adults let alone in the under 2’s [Citation50]. A 2015 study by Kimura et al [Citation93] investigated this and found that gritty mouthfeel was improved by incorporating core granules (<244 um) into an ODT rather than ingesting them alone, along with adjusting particle size, incorporating a suitable coating and necessary additives. Albeit an adult study, such considerations would be more important in under 2s who are likely to be more sensitized to aversive sensations.

4. Technologies to aid administration

As evident from previous discussion, although the various mentioned examples of commercialized multiparticulate and dispersible monolithic formulations bring innovation to drug delivery for the under 2’s, complexities still exist. There have been recent technological advances and approaches to minimize this impact and aid more compliant drug delivery in a greater number of patients <2 years.

4.1. Dosing vehicles as a swallowing aid and palatability enhancer

Administering medicines with a dosing vehicle effectively aids swallowing [Citation37] but can often help with palatability also (taste, smell, mouthfeel). In Japan, swallowing aid Jellies are available for children that have been weaned onto semi-solids (Ryukakusan Co. Ltd [Citation109]). The product, available in a variety of flavors and multi-dose/unit-dose sachets, has means to taste mask whilst primarily easing swallowing of minitablets, pellets and granular formulations. With no sugar, artificial preservatives or colorings and the simplicity and convenience of carers being able to pour the jelly directly onto a spoon, swallowing aids provide a more standardized alternative to semi-solid foods [Citation28,Citation109]. Similar products are available outside Japan (Gloup® [Citation110]) but seems more common for elderly patients. However, it relies on the ability and willingness of the carer to purchase them in adjunction to the main treatment which may reduce the viability of the formulation. Ideally an all in one product would be better, minimizing the steps required for administration. This is what the proprietary platform ‘ParvuletTM’ offers [Citation111].

Breast milk or infant formula are the sole source of nourishment for a neonate/infant in their first 6 months of life and still forms a large part of their diet once solid foods are introduced. After the age of 1, whole cows’ milk can be given instead. Hence, it could be viable for milk to be seen as a practical dosing vehicle, universally.

Interestingly, recent papers [Citation112,Citation113] depicted the possibility of utilizing infant formula based lipid formulations to enhance drug solubility, with the applicable idea of lipid-fortified infant formula powders as a way to modify the degree of drug solubilization desired; (other milk faces quality/regulatory and component variability issues as an excipient). They also investigated how the products of milk/formula digestion may enhance the solubility and therefore bioavailability of co-administered drugs. Bennett et al, 2012 [Citation114] explore the use of fat within milk (milk as a dosing vehicle) as means to reduce the poor palatability elicited by APIs. Since milk has such potentials, it therefore seems important to explore and clarify within SmPCs which, if at all, milk type may be of greater benefit for medicine administration.

Innovative technologies exist utilizing milk feeding to aid with medicine administration. The MedibottleTM [Citation115] appears as a regular milk bottle with attached nipple and a syringe dispenser on the other end so during feeding, the syringe plunger can be pressed to dispense a little medicine near the bottle nipple at a time. There have been several studies concerning the nipple shield device. A recent study Maier et al., 2019 [Citation116] proposes the superiority of delivering API through a liquid-core sodium alginate hydrogel dosage form in comparison to previously investigated rapidly disintegrating tablets and non-woven fiber dosage forms.

4.2. Devices to aid dose measurement and technologies to aid administration

The 2013 EMA ‘List of criteria for screening PIPs with regard to paediatric specific quality issues’ outlines a key binding element that ‘appropriate dispensing devices’ be developed for film-coated minitablets, pellets, granules [Citation117].

Since minitablets offer high dose flexibility due to their low drug loading per minitablet, they need simply be counted (if stored in multi-dose containers) to adjust to the desired dose and hence for high doses a measuring device can accurately do so. Currently, the few available devices provide concerns of high cost, breakage, misplacement, or are unhygienic to use [Citation49]. The smart mini tablet dispenser (sMTS) by Balda and dispenser commercialized by Philips-Medsize in 2020 [Citation41,Citation49] are both reusable and can fit to standard pharmaceutical bottles. It enables up to 10 and 20 minitablets respectively to be counted and lined up precisely before being dispensed together, minimizing miscounting dosing errors.

SympfinyTM by hs-design, yet to be commercialized, furthers this with its syringe multiparticulate dispensing system that offers precise and controlled dosing of dry powders and microsphere formulations directly into the oral cavity. The system which promises one handed usage, easy cleaning, clog proof and chew resistant shell with spill proof interlocking design to guarantee full dose administration [Citation118] consists of a bulk container that houses the multiparticulates, connecting to a bottle adaptor of which interlocks with the tip of a re-usable syringe (with variable dose settings). Sympfiny™ may also help address syringe administration issues for formulation such as Reyataz® (atazanavir sulfate), Kalydeco® (Ivacaftor), Aciphex® (Rabeprazole) and Tamiflu® (oseltamivir phosphate) (), but high cost may hinder its use in LMICs.

The X-straw® from DS Technology [Citation119] works by containing a unit dose of pellets or granules within a straw structure whereby a filter pushes the medicine up the straw toward the mouth in response to the sipping drinker. Beneficially, the straw may be dipped into a beverage of choice; however, the application of this device may only be appropriate for use in older toddlers since it requires capability/understanding to drink from a straw. Moreover, unlike SympfinyTM, the X-straw® does not offer full dose flexibility as several different strengths would be required.

ParvuletTM technology is an unconventional technology for dispersible tablets. It allows for a DT or even a powder to be quickly dispersed (~30 s) with very minimal water (few mL) upon a spoon to form a semi-solid gel consistency, which can be administered directly [Citation49]. This very simplistic method of preparation/combined administration has possibilities to reduce dosing error which is of particular importance for NTI drugs. The good texture (similar to applesauce) allows for easy swallowing, and thus increases treatment adherence without the need for a food-based vehicle [Citation111]. Combination with taste masking and controlled release elements, with allowances for high drug loading is also conceivable [Citation111].

Although these technologies improve DT administration, their applications to neonates would still be in question. It has been found that children can develop oral motor skills for feeding from around 2 weeks-9 months allowing them to open their mouth for a spoon, and at around 2 months be able to move food from a spoon to the back of the oral cavity. Neonatal diets are composed solely of milk/formula until weaned at the infant age of 6 months. Although the introduction of semi-solid food is not until this age, the gels from these technologies do not involve food and so could be pondered as a means for neonatal dosing. However, the presence of the gag reflex from birth protects against unfamiliar substances and textures (e.g. lumpy food) from being ingested and it may be that such semi-solid gels may initiate this reflex, despite being smooth [Citation10].

4.3. 3D printing opportunities

3D printing has great potential in offering a more patient-centric approach to medicine delivery for a variety of dosage forms and may enable custom on-demand production within healthcare facilities for children, such as precise doses, dosage form size/shape and may enable custom on-demand production within healthcare facilities. Many papers [Citation120–123] speculate upon its logistics and implementation into hospitals and pharmacies but currently it is still an emerging field, with concerns of quality regulation at the core [Citation123]. Most 3D printlets are to be swallowed whole and the smallest size achievable are around 1 mm.

Currently, only one 3D printed medicine is marketed [Spritam® (levetiracetam)] [Citation49]. This ZipDose™ technology addresses some key ODT manufacturing concerns, such as low drug loading (particularly for water soluble drugs), size, slow disintegration times, poor mechanical strength [Citation87], difficulty incorporating taste masking and enabling extended-release features [Citation124]. ZipDose™ gives a smooth mouthfeel with only small amounts of liquid/saliva; due to its layer by layer printing technique of powder to give a porous structure [Citation124].

Despite these technological advances that may negate some of the difficulties still pertaining with the aforementioned formulation types, issues related to cost further arise. The increased price of a medication due to the added cost of using a patented/patenting a technology or device manufacture may oppose their aim of greater medication acceptability and thus compliance, particularly in LMICs or where the market is small. For such drug delivery systems to be maximally utilized and with access unhindered, development and manufacture must allow for profit/minimal loss to be made.

5. Rectal drug delivery

Alternatively, medicines may also be dosed via the rectal route. Much like the oral route, there is no requirement for administration by healthcare professionals (HCP) and can be useful for local and systemic treatment. Rectal formulations include solids (tablets, capsules, powders for reconstitution, suppositories), liquids (solutions, suspensions, emulsions) and semi-solids (foams, creams, gels, ointments). Opposing the limitations of oral delivery, rectal formulations can be administered to the unconscious/vomiting patient or those with swallowing issues; without incorporating concerns associated with taste masking, NGT administration, or to avoid parenteral delivery [Citation49]. Issues with irregularity in upper GI tract are also avoided as well as minimizing first pass metabolism [Citation49,Citation125]. They may also be manufactured at low cost [Citation126].

lists some examples of commercially available rectal formulations licensed for systemic administration in under 2s. The table excludes paracetamol and ibuprofen suppositories indicated for pain/fever for 3 months onwards as many generic formulations of various strengths are available and excludes products for local treatment.

Table 5. Examples of commercially available rectal formulations for systemic administration in neonates, infants and toddlers

Solid dosage forms are intuitively easier to administer than liquids and in terms of its retention within the rectum, hence why suppositories may be more prevalent [Citation49]. Generally, opinion for their use has been regarded as relatively unsafe in pre-term neonates due to unpredictable absorption and also as mucosal damage may occur, consequently causing infection [Citation11,Citation49]. In fact, the suppositories and rectal capsule listed in are licensed from infant age. However, in recent years, studies for rectal administration in this age group have increased and thus also the evidence for their safe use and possible superiority to other dosage form types (mentioned later). There are several recent papers detailing in vivo safety and efficacy studies in neonates of rectal formulations with various APIs for different conditions [Citation127–130]. This said, exposure of the API via the rectal route can be variable due to inter-patient and intra-patient absorption variability within the rectum, especially between the different developmental age groups. This is illustrated in several studies [Citation131–133].

Rectal formulations are gaining special interest for the purposes of (emergency) pre-referral use in resource-limited countries where immediate systemic effects may be needed for life-threatening conditions such as (neonatal) sepsis, malaria, HIV or pneumonia [Citation11,Citation49,Citation134]. There are several recent in vitro formulation studies looking at furthering the development of rectal formulations for such purposes [Citation126,Citation135–138].

There are common limitations associated with rectal drug delivery which include:

- Pre-administration: need for empty bowels and accurate rectal placement to optimize absorption, although the bowels empty more frequently in under 2s.

- Post-administration: possible discomfort for the child particularly if more than one daily dose is needed, premature loss of the dose (by triggered fecal incontinence). Children <2 years might find it a challenge to remain still for insertion, but especially for the short period of time required after.

Lipophilic base suppositories are known to melt at temperatures from 30°C. This poses logistical difficulty (need for refrigeration) in their usability in LMICs where temperatures are often above this [Citation49]. Rectal formulations can be restricted with dose flexibility. As can be seen from , some formulations require splitting of the suppository yet split lines nor proper instructions on how to do to ensure accurate dosing are present. Splitting would also mean the size and shape of the formulation is altered, risking their proper and comfortable insertion [Citation27]. The 2013 EMA Guideline [Citation31] considers shape and size of the formulation an important factor: The age and size of the child should be considered for an appropriate size (diameter and length) of the formulation (and lengths for any required rectal tube delivery device [Citation27]). It is also not recommended for suppositories to be cut for achieving smaller doses unless designed to do so as possible inhomogeneous drug distribution and unreproducible cutting are potential causes for dosing error. The 2006 EMA Reflection paper [Citation27] advises for manufacturers to provide information on drug dispersion uniformity of the product. Doliprane, a paracetamol suppository shows advancement in this regard. The 100 mg dose (for 3–8 kg children; ~term neonate-1 year), has a single score line enabling accurate homogeneous 50 mg doses if required. To the knowledge of the authors Doliprane (Sanofi) may be the only suppository containing a score line.

Compared to oral formulations, rectal formulation development for children <2 years is greatly lacking; and could be explained by the concerns mentioned below, but their limited availability may also be contributing to these concerns too, creating a catch-22 situation. There have only been 6 trials investigating rectal drug delivery in children <2 years in the EU, with only one trial (ongoing) investigating a new pediatric drug formulation (omeprazole rectal capsule), reported in EudraCT since 2004. Unlike with use of oral medications, acceptability for the rectal mode of administration can be seen as more of a challenge than the product itself. However, there are limited supporting studies especially in children for the particular reasons why; as viewpoints are known to differ between geographical location, culture and due to lack of knowledge/familiarity/misconception with the dosage form [Citation49]. For example, the UK and USA are known for their lack of acceptability to rectal administration. Thus, looking at a UK study [Citation139] as a ‘worst case scenario’ the opinions of 150 parents on their perceptions of different routes of analgesic delivery for their children (including from birth) were investigated. 58% thought the rectal route most undesirable when compared to intravenous, intramuscular and oral routes, and was the most unfamiliar with 30% of parents unaware of this route of administration. Only 6% of parents thought this route to allow fast and convenient drug administration. Interestingly, it was slightly more accepted to parents of younger children. With the majority of children and parents not having had any prior experience of rectal administration, it was thought arising parental negativity (such as discomfort, embarrassment, painful, unhygienic, upsetting) was due to lack in knowledge.

Confirmed with a group of older children aged between 8–18 years in Hanning et al., 2020 [Citation140] and showing that there are different challenges for different age groups, 64% said ‘maybe’ or ‘not sure’ to considering taking medicines rectally, with their main concerns being size of the dosage form, how to administer it, if someone else would be required for its administration and potential for misuse. Overall, consensus was that education/increasing awareness and providing direct experience were the best ways to overcome concerns and reduce misconceptions/barriers about rectal delivery.

Clearly, more acceptability studies need to be undertaken with the support of industry and HCPs to gauge other social aspects e.g. gender, religion, and ethnicity as well as formulation aspects e.g. size, shape, volume. But, inferring from the aforementioned studies regarding age, this route is less of a barrier for the youngest children. The average age to start potty training toddlers is anywhere between 18 months and 2.5 years; this means that parents and carers of under 2s have regular and more natural access to this route of administration. This should decrease the socio-cultural proctology-related taboos or even safeguarding concerns and the reticence of using this route of administration in this age group. As acknowledged, neonates amongst the pediatric population are the age group suffering with very high mortality rates and route of medicine administration plays an important part in their treatment, with rectal formulations arguably offering benefits over other dosage forms. For example: noninvasive emergency (pre-referral) treatment to give fast systemic effects (improving access for LMICs where clean water and access to healthcare like intravenous treatment/trained professionals are limiting factors), no concerns of requirement for water, swallowability, palatability, or extra need for dosing vehicles that would prevent full efficacious doses from being taken.

6. Expert opinion

The pediatric population is a heterogeneous group, and this must be considered in drug product design. Pediatric regulations have helped to bridge the gap between adult and pediatric licensed and age-appropriate medicine availability, but the gap between these two populations still remains, which encourages pediatric OL, OP and UL medicine use as a necessary requirement. Further understanding and advancement in pediatric drug delivery is still required to minimize sub-optimal treatment concerning these issues and enable access to appropriate drug products. This is particularly important for children aged under 2 years [(pre)term-neonates, infants and toddlers]; a very niche subset of the pediatric population with added complexities. Improving understanding of these complexities will enhance progress in this field.

During the first two years of life, rapid anatomical, physiological and behavioral maturation changes create difficulty in correct disease diagnosis and also causes potential changes in drug metabolism. Compared to older children and adults, disease behavior can differ in this age group, and so, for some therapeutic areas such as oncology, a deeper understanding of this would allow the opportunity to explore the use of some drug molecules in alternative therapies for them. This could help to minimize PIP deferrals and waivers over time. However, due to this heterogeneity associated with those aged under 2, recruiting patients for clinical trials is likely to be difficult. Consequently, approval times to license medicine developments may suffer.

The appeal of flexible dosing is understandable for those aged under 2; associated to their heterogeneity. Liquid formulations offer many advantages including good dose flexibility and so remain the gold standard for dosing pediatric patients. However, with concerns of excipient safety alongside mis-dosing risk and reliance on dosing devices, (particularly for neonates born before term), the latest trends in age-appropriate drug delivery for under 2’s have shown a shift toward flexible solid enteral dosage forms. Such formulations include: multiparticulates, minitablets, DTs, ODTs, ODFs and rectal formulations.

Enteral feeding tubes can be common in children under 2 years in intensive care or pre-term neonates who heavily rely on medical care once born. Administration via enteral feeding tubes requires the drug product to be liquidized prior to administration and for it be compatible with the tube. A key theme noted by HCPs is the need for clarification within SmPCs for how a medicine may be properly manipulated and administered through enteral tubes; particularly where there is polypharmacy, potent drug use, and small volumes involved in fluid restricted patients. This would make UL medicine use safer but future oral formulation developments for those aged under 2 should consider enteral tube feasibility studies if anticipated as a requirement. In LMICs, extemporaneous preparations carried out by pharmacists are utilized due to less availability and access to suitable pediatric formulations. Although UL use is not encouraged, it is agreed that the requirement for guidance in standardized preparations and methods of manufacturing or stability testing to minimize mis-dosing and safety issues are needed. Likewise, recent surveys (ongoing UK ‘Parent co-Designed Drug Information for parents and Guardians Taking Neonates home’ (PADDINGToN) survey [Citation19]) show common parental concerns of neonates returning home from NICU to be dosing error and safety. Specifically: lacking detailed knowledge on the significance of the medication, proper storage conditions, experience on preparing and administering medicine (let alone through enteral tubes) and being new to their child’s complex medicine regime. It is understood that instructions for medicines prior to their administration needs to be presented clearly for parents. In cases when OL and UL medicines are used, very little or no information is available, and it can be difficult for parents to seek advice from HCP’s when issues such as vomiting or missed doses arise. The PADDINGToN study [Citation19] shows that supporting medicine administration with technology such as a phone application or QR codes on packaging could help provide easy access to information when required. Dose preparation is a key consideration in dosage form design.

There are many formulation requirements to consider when formulating drug products for children aged under 2; with a desire to develop a drug product design that meets all their requirements. Further studies to enable our understanding of concepts ‘acceptability’ and ‘age-appropriateness’ are fundamental in supporting the choice of the most suitable formulations to develop. Across publications there are discrepancies in how acceptability is understood and evaluated. A more standardized approach for this assessment is also required to allow for a systematic evidence-based analysis to generate this necessary understanding.

Furthermore, medicine preference by healthcare professionals and parents may differ between countries where the norm for types of pharmaceutical treatment/dosage form vary, especially between HMICs and LMICs where there may be an education or cultural element involved. To give an example, rectal drug delivery with its surrounding stigma and perceived complicated administration, is not as widely accepted by parents in the UK compared to in neighboring European countries where it is a more conventional treatment, especially for pre-schoolers. If we can understand medicine use and opinion in different regions, there may be an opportunity to broaden use in other regions through education and shift parental misconception of non-favored/unfamiliar routes and dosage forms.

This inspires the need for a global database citing all globally licensed pediatric medicines and associated details such as licensed age group, route of administration and dose, to be created to harmonize and popularize knowledge globally. With such collaborative information easily accessed, it would incentivize post marketing studies that could capture patient experience and usage of products. A pharmaceutical platform for all children under 2 years, with strong geographical and cultural acceptance is the target. The database would facilitate drug development for the under 2’s by highlighting the types of formulation in use for this age group, but also where and what the needs are. This would help identify when to innovate, re-formulate age-inappropriate OP drugs (as per undersubscribed PUMA requirements) or implement alternative administration routes (e.g. rectal route).

Logistical issues such as manufacturing feasibility, scalability and production costs that may result in high product costs (particularly if utilizing patented technology or developing for orphan diseases), may delay access of newly approved age-appropriate platforms/formulations to market, and especially of their access in LMICs. In HMICs a concern is that physicians and national health services will continue with OL, OP and UL prescribing at the benefit of lower product costs even when a perfectly appropriate licensed product is available. Approaches are needed, with collaboration between innovators, industry and regulators, to enable novel research and new formulations into commercially viable products without greatly increasing costs, to support better and cost-effective pediatric formulations reaching those most in need. It may be misconstrued that innovation is what is always required for advancement in the availability of age-appropriate medicines for the under 2’s, when only the optimization of existing platforms is needed. This is an area where further understanding is needed to best meet their requirements without affecting access.

Article highlights

  • Further understanding into neonatal, infant and toddler therapeutic and pharmaceutical requirements, and advancement in their pharmacotherapy is still required to stop fuelling off-label/off-patent/unlicensed drug use and sub-optimal treatment.

  • Age-appropriate medicines for the under 2’s should encompass good acceptability for them and their carers without affecting pharmaceutical quality, safety and accessibility.

  • Oral solid multiparticulates such as minitablets and sprinkle formulations (granules, pellets, powders) as well as monolithic dispersible formulations, and their administration devices have shown good acceptability in children aged under 2 as an alternative to commonly used liquid formulations.

  • Rectal formulations provide an alternative to patients who cannot take oral medicines; without need for a palatable product or enteral feeding tube administration and can provide fast systemic effects, for emergency use or use in pre-referral treatment in LMICs.

  • Deeper investigation is required into the acceptability of formulations and how it is understood and evaluated in order to produce more globally accepted age-appropriate medicines.

  • Efforts are required to understand how perception of non-favoured administration routes/dosage forms in different geographical locations can be overcome to shift convention and welcome newly developed formulation trends.

  • Understanding is required to identify when innovation or optimisation of existing platforms is needed to ensure access is not affected.

This box summarizes key points contained in the article.

Declaration of interest

TB Ernest is an employee of GlaxoSmithKline and hold shares in the company. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

List of Abbreviations

EU – European Union

PIP – Paediatric Investigation Plan

PDCO - Paediatric Committee

EMA – European Medicines Agency

OL – Off-label

OP – Off-patent

PUMA – Paediatric Use Marketing Authorisation

NICU – Neonatal Intensive Care Unit

ADME – Absorption, Distribution, Metabolism, Excretion

NGT – Nasogastric Tubes

SmPC – Summary of Product Characteristics

PK/PD – Pharmacokinetics and Pharmacodynamics

API - Active Pharmaceutical Ingredient

SEEN - Safe Excipient Exposure in Neonates and Small Children

ESNEE - ▣European Study on Neonatal Exposure to Excipients

UL – Unlicensed

WHO – World Health Organisation

ODT – Orodispersible Tablet

DT – Dispersible Tablet

ODF – Orodispersible Film

LMICs – Low-middle income countries

PIL – Patient Information Leaflet

NTI – Narrow Therapeutic Index

HCP – Healthcare Professionals

PADDINGToN - Parent co-Designed Drug Information for parents and Guardians Taking Neonates home

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was funded by EPSRC grant EP/R513143/1, GSK sponsorship.

References