Next Article in Journal
Improved Understanding of the High Shear Wet Granulation Process under the Paradigm of Quality by Design Using Salvia miltiorrhiza Granules
Next Article in Special Issue
Classification of WHO Essential Oral Medicines for Children Applying a Provisional Pediatric Biopharmaceutics Classification System
Previous Article in Journal
Hyperthermia-Triggered Doxorubicin Release from Polymer-Coated Magnetic Nanorods
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Innovations in Pediatric Drug Formulations and Administration Technologies for Low Resource Settings

1
Global Health and Global Development Divisions, Bill and Melinda Gates Foundation, Seattle, WA 98109, USA
2
Jenny Walsh Consulting Ltd.; Nottingham NG1 1GF, UK
3
School of Pharmacy, University College London, London WC1N 1AX, UK
*
Author to whom correspondence should be addressed.
Pharmaceutics 2019, 11(10), 518; https://doi.org/10.3390/pharmaceutics11100518
Submission received: 22 July 2019 / Revised: 1 October 2019 / Accepted: 1 October 2019 / Published: 8 October 2019
(This article belongs to the Special Issue Paediatric Drug Delivery)

Abstract

:
Despite advances in regulations and initiatives to increase pediatric medicine development, there is still an unmet need for age-appropriate medicines for children. The availability of pediatric formulations is particularly lacking in resource poor areas, due to, for example, area-specific disease burden and financial constraints, as well as disconnected supply chains and fragmented healthcare systems. The paucity of authorized pediatric medicines often results in the manipulation and administration of products intended for adults, with an increased risk of mis-dosing and adverse reactions. This article provides an overview of the some of the key difficulties associated with the development of pediatric medicines in both high and low resource areas, and highlights shared and location specific challenges and opportunities. The utilization of dispersible oral dosage forms and suppositories for low and middle-income countries (LMICs) are described in addition to other platform technologies that may in the future offer opportunities for future pediatric medicine development for low resource settings.

Graphical Abstract

1. Introduction

Pediatric patients are defined as patients aged from birth to less than 16 or 18 years, depending on region [1]. Due to development differences in physiology and cognitive and motor skills compared to adults, this population group often requires medicines that are tailored specifically for them. However, despite almost one third of the world’s population being less than 18 years old [2], there is a global lack of licensed medicines for pediatric use. This is due to factors such as the diversity in age and therapeutic needs of this patient population leading to greater challenges in conducting research into medicines for pediatrics compared to adult products. The availability of pediatric medicines is particularly lacking in resource poor areas because of a paucity of basic healthcare systems and fragmented supply chains, as well as mostly non-temperate and often harsh climatic conditions with a lack of refrigeration limiting formulation options, and area-specific disease burden creating a challenging market potential. This article provides an overview of the current difficulties and potential opportunities associated with the development of pediatric medicines for resource poor areas, with a focus on pharmaceutical development. Insights into the current situation regarding the formulation and administration of age-appropriate medicines for both high and low resource settings are provided to highlight shared and specific key challenges. The application of dispersible dosage forms, as advocated by the World Health Organization (WHO) is described [3], as well as suppositories and other platform technologies that may have the potential to provide opportunities for future development in low and middle-income countries (LMICs). (Defined as those with a gross national income per capita of less than $12,375 in 2018, in accordance with the World Bank [4]).

2. Background

Historically, there has been limited research and development into pediatric medicines, which has been in part due to a lack of market forces and economic benefits for the pharmaceutical industry compared to those for adult products [5]. This has been acknowledged and prompted global initiatives and legislative changes, with the aim of transforming this niche area into an integral part of the drug development process. The focus of these advances has been very much within the domain of high-income countries; regulations in the US and Europe [6,7,8] have made significant changes to the medicine legislature framework, and have led to an increase in the amount of information available on medicine administration to children, increased the number of authorized pediatric products and the number of new pediatric indications for already authorized products [5,9]. Progress is being made in other territories including China, where regulatory reforms have been designed to encourage the development of innovative drugs for pediatric patients in the future [10]. In addition, the Indian drug regulatory authority has acknowledged that pediatric regulation is undoubtedly needed [11,12], and the Indian Council of Medical Research have developed the National Ethical Guidelines for Biomedical Research Involving Children [13], to address ethical issues of conducting research in children. Almost every country in Africa has a national medicines regulatory authority; although they vary in terms of functionality and expertise [14]. Although specific pediatric regulations do not appear to have been adopted, The African Paediatric Fellowship Programme has been developed to provide relevant training for African child health professionals, by Africans, within Africa [15].
In 2007, the Member States of the World Health Organization (WHO) passed a resolution on “Better Medicines for Children” [16] and have since made progress on several fronts with the “Make medicines child size” initiative, including the development and publication of treatment guidelines, and information on the use of essential medicines such as the first Model List of Essential Medicines for Children and the WHO Model Formulary for Children [17]. Additionally, public–private partnerships, for example “Drugs for Neglected Diseases initiative” (DNDi, https://www.dndi.org/), “Medicines for Malaria Venture” (MMV, https://www.mmv.org), and the “TB Alliance” (TBA, https://www.tballiance.org/) are addressing and promoting the development of pediatric formulations for global priority diseases.
Despite these developments, progress is slow and there is still a global scarcity of age-appropriate pediatric formulations, especially in LMICs, with disparities in life expectancy and burden of diseases [18,19]. There is an absence of specific pediatric development regulations in many resource poor countries [12,20] and financial constraints, workforce capacity and limited access to pediatric expertise have made it difficult for these countries to support new developments and maintain the high standards required.
A contributory factor in the lack of pediatric formulations worldwide and in LMICs in particular, may be due to the majority of therapeutic drugs (active pharmaceutical ingredients (APIs)) used to treat priority diseases being off-patent [21], since the investigation of off-patent pediatric medicines is voluntary, with potentially limited financial incentives to conduct this work [22]. Indeed, the paucity of authorized pediatric off-patent products has led to the prescribing of off-label/unlicensed medicines, whereby the medicine may be manipulated by healthcare professionals and caregivers to enable the intended dose to be administered to the patient [23,24,25,26,27,28,29,30]. The impact of such manipulations on the bioavailability and actual dose of API administered is often unknown, which can lead to an increased risk of medication errors and adverse reactions, as well as sub-optimal treatment [31,32,33,34,35].

3. Challenges for Progress and Innovations in Developing Pediatric Medicine Formulations

As stated above, pediatric patients have different requirements compared to adults regarding pharmacotherapy [36]. Key formulation parameters for the development of age-appropriate dosage forms regardless of the location of the proposed patient population include correct yet flexible dosing, acceptable excipients, easy product administration, and acceptable palatability. However, there are some differences in other factors between high and LMICs.
It is important that the optimal dose can be accurately administered to the patient, and hence the formulation may require some inherent flexibility to cover a range of doses. Although excipients are generally considered to be inert, new evidence suggests that there may be safety issues with some excipients when used in products for pediatric patients [36]. For example, immature metabolic systems in neonates and infants can lead to the accumulation and toxicity of propylene glycol [37] and benzoic acid/benzoates [38], and so these excipients should be used with caution in very young patients. Flavoring agents may be included to improve the taste of an oral product and facilitate patient adherence, but there are concerns regarding the potential risk of allergy and sensitization with these materials [39].
When formulating a pediatric medicine, it is also necessary to consider the capability of the patient to take the product and the product’s acceptability linked to achieving effective use, the latter being determined by the characteristics of both the product and the user (patient and caregiver) [36]. It is now increasingly acknowledged that the pharmaceutical development of a (pediatric) medicine involves more than its formulation aspects. For example, the recommended dosing frequency, type of packaging, type of medical device, or the comprehensibility of the user instructions may impact the medicine’s “intended use” in clinical and/or domiciliary practice [40]. In addition, the availability and ease of manufacture of the proposed dosage form should be borne in mind, since these will have an impact on cost and patient access. Indeed, patient acceptability, safety, and access must be balanced against each other, and a single “ideal” dosage form is unlikely to exist; in some situations, a compromise may need to be reached when selecting an age-appropriate formulation [41]. Additional challenges of developing pediatric medicines include the ethics and logistics of conducting trials in children. Challenges have been reported to arise from fears of harming children, political and economic influences, lack of resources, and a bureaucratic regulatory framework. In LMICs, there are also issues related to poverty, fear of participation, and mis-trust of sponsors [42].
Product attributes and supply logistics of pediatric medicines for resource-poor settings may differ compared to those of products designed for more economically developed countries, which can lead to additional formulation challenges, as summarized in Table 1. The climate in many LMICs requires products to be stable in high humidity and high temperature storage conditions (ICH Zones III and IV) [43], however, the provision of temperature-controlled storage and transportation in these areas is often limited and unlikely to be available in the near future. The use of solid dosage forms such as tablets is therefore preferred over liquids and semi-solids, since they are typically more stable [44]. Affordability is of key importance, since the majority of the population in developing countries may purchase their medicines through out-of-pocket payments [45]. Indeed, poor affordability can result in lack of availability of medicines in LMICs [46]. Although the cost of a product will depend on numerous factors, including the local pricing strategy applied and the utilization of inexpensive materials (including packaging), simple manufacturing and development processes will facilitate the implementation of low costs and greater patient access. Storage facilities and transportation systems in resource poor areas can be rudimentary and fragmented. Hence, any packaging should ideally be compact with a small footprint, light in weight, and sufficiently robust to withstand ground transportation in rural areas. For example, glass bottles are not a favored primary packaging option for global health products.
In addition to the above considerations, other aspects such as culture and health literacy need to be considered when defining the design criteria for the product. Medicine administration procedures and instructions should be simple to avoid misunderstanding. Caregivers may not understand information provided through healthcare workers, and use informal advice from immediate family members instead [47]. Non-adherence to medication is a global problem that is especially prevalent in LMICs, and factors reported to have a significant negative effect on adherence include poor socioeconomic status, poverty, low level of education, illiteracy, and long distance from treatment center [48]. Data from 2015 show that globally, 844 million people still lacked a basic drinking water service, with 159 million people still collecting drinking water directly from surface water sources, of which 58% lived in sub-Saharan Africa [21]. The potential lack of clean, filtered water can lead to difficulties in the constitution of powders and granules, and so, if this type of dosage form is developed, dispersal in other media such as breast milk may need to be considered.

4. Potential Dosage Form Opportunities for Low and Middle-Income Countries

In this age of advanced technologies, there has never been a greater opportunity to develop innovations for children in LMICs who remain vulnerable to the devastating effects of disease. Identifying technology platforms that would meet the needs of this pediatric population is a step towards addressing the lack of age-appropriate medicines. The dosage form examples highlighted below have been chosen to illustrate recognized technology platforms that have the potential to be applied to global health. It is acknowledged that the immunization of children against vaccine-preventable diseases can avert millions of deaths, as well as generate billions of dollars in economic benefits [49,50], and the same product development considerations highlighted above regarding acceptability, stability, cost, and supply should be considered. Specific vaccine technologies are also discussed in detail in various related recent reviews [51,52].

4.1. Oral Dispersible Dosage Forms

For oral products, the development of flexible solid dosage forms, for example tablets that can be dispersed or dissolved in a beverage prior to administration, have been recommended by WHO [44]. They are considered to be suitable for LMICs because they have some of the advantages of solid oral dosage forms, such as superior stability and less bulk compared to liquids, and once dispersed are acceptable for patients of all ages, including those who have difficulty in swallowing tablets. A summary of the potential advantages and disadvantages of dispersible dosage forms is provided in Table 2.
Various API properties need to be considered when developing dispersible dosage forms (see Figure 1).
For example, the dose of API will affect the unit dose quantity of granules or overall size and dimensions of the dispersible tablet, and hence the volume of liquid required for complete dispersion. API physicochemical properties such as particle size and morphology will impact the manufacturing process and choice of excipients. The aqueous solubility of the API will affect how the granules or tablet disperses in liquid, and if a solution or a suspension of the API is formed; if the API fully dissolves, the need to rinse the vessel or add thickeners to the formulation to reduce sedimentation may be avoided. Solubility can affect the bioavailability of an API, and so also has an impact on required dose. APIs with an unpleasant taste are likely to require a sweetener and/or flavor to improve palatability and ensure patient compliance, whilst additional taste-masking techniques may be needed for very aversive APIs [41].
Despite some of the challenges highlighted above, several dispersible tablet formulations have been successfully developed for pediatric patients in LMICs. Examples are provided in Table 3.

4.2. Rectal Forms

Administration of drugs via the rectum offers an alternative to the oral route and is suitable for both local (for example, constipation, hemorrhoids, analgesia) and systemic delivery (for example analgesia, infections, and management of central nervous system disorders), and minimizes hepatic first pass metabolism, since the API is absorbed directly into the systemic circulation, thereby avoiding the liver. Indeed, the extent of rectal absorption has been reported to exceed oral values for some APIs [53]. Dosage forms for the rectal route such as suppositories are suitable for pediatric patients who may have difficulty swallowing tablets and capsules. Furthermore, they do not require taste-masking and can be administered to unconscious or vomiting patients [54]. Suppositories (solid dosage forms that dissolve or melt when inserted into the rectum) constitute most current products administered rectally in clinical practice. They offer several advantages compared to other dosage forms, especially in LMICs. For example, they might be used for the treatment of children in rural areas where parenteral administration is not possible. A summary of the potential advantages and disadvantages of suppositories is provided in Table 4.
There are two main types of suppository bases; fatty bases that melt at the temperature of the rectum, and water-soluble bases that dissolve in the rectum [55]. The selection of base depends on the properties of the API, for example, water-soluble APIs tend to be formulated in fatty bases and lipophilic APIs in water-soluble bases. As with dispersible dosage forms, the required dose of API will impact the size of the suppository. However, age-related changes in the dimensions of the rectum need to be considered to ensure acceptable administration. Although suppositories are considered to be appropriate for children [56], a key constraint in their use appears to be the varying levels of their acceptability around the world and according to patient age. This is an area in which patients and caregivers require further education.
Several new suppository developments are on-going to mitigate some of the disadvantages highlighted above, for example, the use of muco-adhesives and high melting point excipients to improve bioavailability and heat stability, respectively [54]. Despite the above challenges, and potential for regionally specific acceptability, the development of suppositories for LMICs is an area of growing interest [57]. For example, suppositories containing 100 mg of artesunate have been developed and prequalified by WHO for pre-referral treatment for patients aged between six months and six years, with suspected moderate or severe malaria who are unable to take oral medication or obtain injectable antimalarial treatment [58].

5. Other Potential Formulation Technologies for Low and Middle-Income Countries

There are other formulation technologies in addition to dispersible dosage forms and suppositories that may also be in the future applicable to pediatric patients in LMICs. For example, mini tablets (1–3 mm in diameter), like other solid oral dosage forms, have the advantages of better stability and less bulk compared to liquids, but unlike standard tablets, have been reported to be acceptable from birth if uncoated and from six months if coated [59,60]. Mini tablets are a relatively recent innovation, and hence few products appear to have been commercialized, and none specifically for LMICs. Examples of marketed products include Orfiril® long (sodium valproate sustained release), LAMISIL® oral granules (terbinafine hydrochloride), KALYDECO® oral granules (ivacaftor) and Desitin® minitablets (levetiracetam). Oro-dispersible tablets (ODTs) are designed to disperse in the oral cavity without the need for a beverage and are also considered be acceptable for young patients [61], although may require moisture protective packaging. Several ODT products are commercially available, although the majority are only licensed for adult use. Examples of pediatric ODT products include Loratidine ODTs, Calpol® Six Plus Fast Melts (paracetamol), and Nurofen® for Children Meltlets (ibuprofen).
Transdermal delivery, where APIs are delivered through the skin, has several advantages including the avoidance of first pass metabolism, controlled release, and potentially improved patient compliance. However, due to the barrier properties of the skin, only a few APIs can be delivered via this route at therapeutic levels. Microneedles are being developed to address this issue whereby micron sized pores are created through which APIs and vaccines may be administered. The microneedles are long enough to penetrate the dermis but avoid stimulation of dermal nerves and do not puncture the dermal blood vessels [62,63]. This is an emerging technology for which no drug delivery products appear to have yet been marketed, although numerous clinical studies have been conducted or are on-going [64]. Microneedles may offer advantages for LMICs, since it is an easy to use technology which is less invasive and more affordable compared to parenteral delivery [65].

6. Conclusions

The development of pediatric medicines has greater challenges compared to the development of medicines intended for adult patients, which has led to a significant unmet medical need. Additional considerations need to be applied to the development of pediatric medications for LMICs, including those linked to cost, acceptability, usability, heat stability, healthcare provider training, health policy, and local regulatory requirements. Although progress is being made, there is still a paucity of available age-appropriate pediatric medicines in LMICs. A number of technology platforms including emerging technologies which may offer potential additional solutions to mitigate some of the challenges associated with the development and supply of pediatric products for LMICs. The global health community should maximize opportunities to address the disparity in access to medicines that meet the needs of neonates, infants and children, where there is a great opportunity on long term health impact.

Author Contributions

Conceptualization, S.E.G., N.B., J.W., S.S., S.H.; writing—original draft preparation, J.W., S.S.; writing—review and editing, S.E.G., N.B., J.W., S.S.

Funding

This research was funded by Bill and Melinda Gates Foundation (BMGF).

Conflicts of Interest

S.E.G., N.B., and S.H. are employees of BMGF; J.W. and S.S. provide consultancy support to BMGF. The company had no role in the the writing of the manuscript.

References

  1. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). Addendum to ICH E11: Clinical Investigation of Medicinal Products in the Pediatric Population E11 (R1). 2017. Available online: https://database.ich.org/sites/default/files/E11_R1_Addendum.pdf (accessed on 4 October 2019).
  2. Estimated from World Population Figures for 2015 Derived from the United Nations DESA Population Division, World Population Prospects 2019. Available online: https://population.un.org/wpp/dataquery/ (accessed on 4 October 2019).
  3. World Health Organization (WHO). Development of Paediatric Medicines: Points to Consider in Formulation; Technical Report Ser. 2012; WHO: Geneva, Switzerland, 2012; pp. 197–225. [Google Scholar]
  4. Turner, M.A.; Catapano, M.; Hirschfeld, S.; Giaquinto, C. Paediatric drug development: The impact of evolving regulations. Adv. Drug Deliv. Rev. 2014, 73, 2–13. [Google Scholar] [CrossRef]
  5. World Bank Country and Lending Groups. Available online: https://datahelpdesk.worldbank.org/knowledgebase/articles/906519 (accessed on 4 October 2019).
  6. European Council (EC). Regulation (EC) No 1901/2006 of the European Parliament and of the Council of 12 December 2006 on medicinal products for paediatric use and amending; Regulation (EEC) No. 1768/92, Directive 2001/20/EC, Directive 2001/83/EC and Regulation (EC) No 726/2004. Off. J. Eur. Communities 2006, L378/1–L378/19. [Google Scholar]
  7. European Council (EC). Regulation (EC) No 1902/2006 of the European Parliament and of the Council of 12 December 2006 amending Regulation 1901/2006 on medicinal products for paediatric use. Off. J. Eur. Communities 2006, L378/20–L378/21. [Google Scholar]
  8. Food and Drug Administration (FDA). Food and Drug Administration Safety and Innovation Act (FDASIA); 126 Stat. 993; FDA: Washington, DC, USA, 2012. [Google Scholar]
  9. European Medicines Agency (EMA). 10-Year Report to the European Commission General Report on the Experience Acquired as a Result of the Application of the Paediatric Regulation; EMA/231225/2015; EMA: Amsterdam, The Netherlands, 2016. [Google Scholar]
  10. Wang, B.; Davidson, A. An overview of major reforms in China’s regulatory environment. Regul. Rapp. 2017, 14, 5–9. [Google Scholar]
  11. Jain, A.V.M.; Pramod Jumar, T.M.; Naveen Kumar, S. Regulation for paediatric drug development in India: Need of hour. J. Clin. Sci. 2014, 6, 14–17. [Google Scholar]
  12. Rani, V.; Nanda, N.B. A comprehensive study on the regulation of paediatrics in US, EU and India: Present status and future prospective. Adv. Chem. Biochem. Sci. 2015, 2, 1–12. [Google Scholar]
  13. Indian Council of Medical Research, National Ethical Guidelines for Biomedical Research Involving Children. 2017. Available online: https://icmr.nic.in/sites/default/files/guidelines/National_Ethical_Guidelines_for_BioMedical_Research_Involving_Children_0.pdf (accessed on 4 October 2019).
  14. Ndomondo-Sigonda, M.; Miot, J.; Naidoo, S.; Dodoo, A.; Kaale, E. Medicines regulation in Africa: Current state and opportunities. Pharm. Med. 2017, 31, 383–397. [Google Scholar] [CrossRef]
  15. Wilmshurst, J.M.; Morrow, B.; du Preez, A.; Githanga, D.; Kennedy, N.; Zar, H.J. The African Pediatric Fellowship Program: Training in Africa for Africans. Pediatrics 2016, 137, e20152741. [Google Scholar] [CrossRef]
  16. World Health Organization (WHO). Sixtieth World Health Assembly WHA60.20, Better Medicines for Children. 2007. Available online: http://apps.who.int/medicinedocs/documents/s21455en/s21455en.pdf (accessed on 8 May 2019).
  17. Ivanovska, V.; Rademaker, C.M.; van Dijk, L.; Mantel-Teeuwisse, A.K. Pediatric drug formulations: A review of challenges and progress. Pediatrics 2014, 134, 361–372. [Google Scholar] [CrossRef]
  18. Bassat, Q.; Tanner, M.; Guerin, P.J.; Stricker, K.; Hamed, K. Combating poor-quality anti-malarial medicines: A call to action. Malar. J. 2016, 15. [Google Scholar] [CrossRef]
  19. Beggs, S.A.; Cranswick, N.F.; Reed, M.D. Improving drug use for children in the developing world. Arch. Dis. Child 2005, 1091–1093. [Google Scholar] [CrossRef] [PubMed]
  20. Nsabagasani, X.O.-O.J.; Mbonye, A.; Ssengooba, F.; Nantanda, R.; Muyinda, H.; Holme Hansen, E. The “child size medicines” concept: Policy provisions in Uganda. J. Pharm. Policy Pract. 2015, 8, 8–12. [Google Scholar] [CrossRef] [PubMed]
  21. World Health Organization (WHO). UNICEF Progress on Drinking Water, Sanitation and Hygiene. 2017. Available online: http://apps.who.int/iris/bitstream/10665/258617/1/9789241512893-eng.pdf?ua=1 (accessed on 8 May 2019).
  22. Wimmer, S.R.W.; McCarthy, S.; Neubert, A. The EU paediatric regulation: Still a large discrepancy between therapeutic needs and approved paediatric investigation plans. Paediatr. Drugs 2014, 16, 397–406. [Google Scholar] [CrossRef] [PubMed]
  23. Abdulah, R.-O.; Khairinisa, M.A.; Pratiwi, A.A.; Barliana, M.I.; Pradipta, I.S.; Halimah, E.; Diantini, A.; Lestari, K. Off-label paediatric drug use in an Indonesian community setting. J. Clin. Pharm. Ther. 2015, 40, 409–412. [Google Scholar] [CrossRef] [PubMed]
  24. Ankrah, D.N.A.; Turkson, J.T.; Boateng, E.B.; Amegavie, F.T.T.; Bruce, E. Insufficient access to oral paediatric medicines in Ghana: A descriptive study. BMC Health Serv. Res. 2016, 16, 198. [Google Scholar] [CrossRef] [PubMed]
  25. Bastiaans, D.E.; Cressey, T.R.; Vromans, H.; Burger, D.M. The role of formulation on the pharmacokinetics of antiretroviral drugs. Expert Opin. Drug Metab. Toxicol. 2014, 10, 1019–1037. [Google Scholar] [CrossRef] [PubMed]
  26. Bjerknes, K.B.S.; Kristensen, S.; Brustugun, J.; Wang, S. Manipulating tablets and capsules given to hospitalised children in Norway is common practice. Acta Paediatr. 2017, 106, 503–508. [Google Scholar] [CrossRef] [PubMed]
  27. Magalhaes, J.; Rodrigues, A.T.; Roque, F.; Figueiras, A.; Falcao, A.; Herdeiro, M.T. Use of off-label and unlicenced drugs in hospitalised paediatric patients: A systematic review. Eur. J. Clin. Pharmacol. 2014, 71, 1–13. [Google Scholar] [CrossRef]
  28. Nakamura, H.; Ishikawa, Y. How do Japanese children take their medicines, and what are pharmacists and paediatricians doing about it? Int. J. Pharm. 2014, 469, 249–250. [Google Scholar] [CrossRef]
  29. Oshikoya, K.A.O.I.; Godman, B.; Fadare, J.; Orubu, S.; Massele, A.; Senbanjo, I.O. Off-label prescribing for children with chronic diseases in Nigeria; Findings and implications. Expert Opin. Drug Saf. 2017, 16, 981–988. [Google Scholar] [CrossRef]
  30. Richey, R.H.; Hughes, C.; Craig, J.V.; Shah, U.U.; Ford, J.L.; Barker, C.E.; Peak, M.; Nunn, A.J.; Turner, M.A. A systematic review of the use of dosage form manipulation to obtain required doses to inform use of manipulation in paediatric practice. Int. J. Pharm. 2016, 518, 155–166. [Google Scholar] [CrossRef] [PubMed]
  31. Bellis, J.R.; Kirkham, J.J.; Thiesen, S.; Conroy, E.J.; Bracken, L.E.; Mannix, H.L.; Bird, K.A.; Duncan, J.C.; Peak, M.; Turner, M.A.; et al. Adverse drug reactions and off-label and unlicensed medicines in children: A nested case-control study of inpatients in a pediatric hospital. BMC Med. 2013, 1, 238. [Google Scholar]
  32. Cuzzolin, L.; Atzei, A.; Fanos, V. Off-label and unlicensed prescribing for newborns and children in different settings: A review of the literature and a consideration about drug safety. Expert Opin. Drug Saf. 2006, 5, 703–718. [Google Scholar] [CrossRef] [PubMed]
  33. Cuzzolin, L. Off-label drug in the newborn. J. Pediatr. Neonat. Individual Med. 2014, 3, e030224. [Google Scholar]
  34. Mason, J.; Pirmohamed, M.; Nunn, T. Off-label and unlicensed medicine use and adverse drug reactions in children: A narrative review of the literature. Eur. J. Clin. Pharmacol. 2012, 68, 21–28. [Google Scholar] [CrossRef] [PubMed]
  35. Neubert, A.; Dormann, H.; Weiss, J.; Egger, T.; Criegee-Rieck, M.; Rascher, W.; Brune, K.; Hinz, B. The impact of unlicensed and off-label drug use on adverse drug reactions in paediatric patients. Drug Saf. 2004, 27, 1059–1067. [Google Scholar] [CrossRef]
  36. European Medicines Agency (EMA). Committee for Medicinal Products for Human Use (CHMP), Paediatric Committee (PDCO), Guideline on Pharmaceutical Development of Medicines for Paediatric Use; EMA/CHMP/QWP/805880/2012 Rev. 2, 2013; EMA: Amsterdam, The Netherlands, 2013. [Google Scholar]
  37. European Medicines Agency (EMA). Questions and Answers on Propylene Glycol and Esters in the Context of the Revision of the Guideline on ‘Excipients in the Label and Package Leaflet of Medicinal Products for Human Use’ (CPMP/463/00 Rev. 1); Technical Report EMA/CHMP/704195/2013; EMA: Amsterdam, The Netherlands, 2014. [Google Scholar]
  38. European Medicines Agency (EMA). Questions and Answers on Benzoic acid and Benzoates in the Context of the Revision of the Guideline on ‘Excipients in the Label and Package Leaflet of Medicinal Products for Human Use’; (CPMP/463/00), EMA/CHMP/508189/2013; EMA: Amsterdam, The Netherlands, 2014. [Google Scholar]
  39. Walsh, J.; Cram, A.; Woertz, K.; Breitkreutz, J.; Winzenburg, G.; Turner, R.; Tuleu, C.; European Formulation Initiative. Playing hide and seek with poorly tasting paediatric medicines: Do not forget the excipients. Adv. Drug Deliv. Rev. 2014, 73, 14–33. [Google Scholar] [CrossRef] [Green Version]
  40. Van Riet-Nales, D.A. Child Friendly Medicines: Availability, Pharmaceutical Design, Usability and Patient Outcomes. Ph.D. Thesis, Utrecht University, Utecht, The Netherlands, 2014. [Google Scholar]
  41. Walsh, J.; Ranmal, S.R.; Ernest, T.B.; Liu, F. Patient acceptability, safety and access: A balancing act for selecting age-appropriate oral dosage forms for paediatric and geriatric populations. Int. J. Pharm. 2017, 536, 547–562. [Google Scholar] [CrossRef]
  42. Joseph, P.D.; Craig, J.C.; Tong, A.; Caldwell, P.H. Researchers’, regulators’, and sponsors’ views on pediatric clinical trials: A multinational study. Pediatrics 2016, 138, e20161171. [Google Scholar] [CrossRef]
  43. The World by Income and Region. World Bank, 2019. Available online: https://datatopics.worldbank.org/world-development-indicators/the-world-by-income-and-region.html (accessed on 4 October 2019).
  44. World Health Organization (WHO). Report of the Informal Expert Meeting on Dosage Forms of Medicines for Children. 2008. Available online: https://www.who.int/selection_medicines/committees/expert/17/application/paediatric/Dosage_form_reportDEC2008.pdf (accessed on 8 May 2019).
  45. World Health Organization (WHO). Guideline on Country Pharmaceutical Pricing Policies. 2013. Available online: https://www.ncbi.nlm.nih.gov/books/NBK258631/ (accessed on 8 May 2019).
  46. Ewen, M.; Zweekhorst, M.; Regeer, B.; Laing, R. Baseline assessment of WHO’s target for both availability and affordability of essential medicines to treat non-communicable diseases. PLoS ONE 2017, 12, e0171284. [Google Scholar] [CrossRef]
  47. McKinn, S.; Thuy Linh, D.; Foster, K.; McCaffery, K. Distributed health literacy in the maternal health context in Vietnam. HLRP 2019, 3, e31–e42. [Google Scholar] [CrossRef] [PubMed]
  48. World Health Organization (WHO). Adherence to Long Term Therapies. Evidence for Action. 2003. Available online: https://apps.who.int/iris/bitstream/handle/10665/42682/9241545992.pdf?sequence=1 (accessed on 27 September 2019).
  49. World Health Organization (WHO). Global Vaccine Action Plan 2011–2020 (2012). Available online: https://www.who.int/immunization/global_vaccine_action_plan/GVAP_doc_2011_2020/en/ (accessed on 27 September 2019).
  50. World Health Organization (WHO). Estimates of Disease Burden and Cost-Effectiveness. 2019. Available online: https://www.who.int/immunization/monitoring_surveillance/burden/estimates/en/ (accessed on 27 September 2019).
  51. Wallis, J.; Shenton, D.P.; Carlisle, R.C. Novel approaches for the design, delivery and administration of vaccine technologies. Clin. Exp. Immunol. 2019, 196, 205–214. [Google Scholar] [CrossRef] [PubMed]
  52. Miquel-Clopes, A.; Bentley, E.G.; Stewart, J.P.; Carding, S.R. Mucosal vaccines and technology. Clin. Exp. Immunol. 2019, 196, 189–204. [Google Scholar] [CrossRef] [PubMed]
  53. Van Hoogdalem, E.; de Boer, A.G.; Breimer, D.D. Pharmacokinetics of rectal drug administration, Part, I. General considerations and clinical applications of centrally acting drugs. Clin. Pharmacokinet 1991, 21, 11–26. [Google Scholar] [CrossRef] [PubMed]
  54. Jannin, V.; Lemagnen, G.; Gueroult, P.; Larrouture, D.; Tuleu, C. Rectal route in the 21st Century to treat children. Adv. Drug Deliv. Rev. 2014, 73, 34–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Aulton, M.; Taylor, K. (Eds.) Aulton’s Pharmaceutics. The Design and Manufacture of Medicines, 4th ed.; Elsevier Ltd.: Amsterdam, The Netherlands, 2013. [Google Scholar]
  56. European Medicines Agency (EMA). Reflection Paper: Formulations of Choice for the Paediatric Population, 2006; EMEA/CHMP/PEG/194810/2005; EMA: Amsterdam, The Netherlands, 2005. [Google Scholar]
  57. Ham, A.S.; Buckheit, R.W. Designing and developing suppository formulations for anti-HIV drug delivery. Ther. Deliv. 2017, 8, 805–817. [Google Scholar] [CrossRef]
  58. Rectal artesunate (RAS), Medicines for Malaria Venture. Available online: https://www.mmv.org/access/products-projects/rectal-artesunate-ras (accessed on 4 October 2019).
  59. Klingmann, V.; Seitz, A.; Meissner, T.; Breitkreutz, J.; Moeltner, A.; Bosse, H.M. Acceptability of uncoated mini-tablets in neonates—A Randomized Controlled Trial. J. Pediatr. 2015, 167, 893. [Google Scholar] [CrossRef]
  60. Klingmann, V.; Spomer, N.; Lerch, C.; Stoltenberg, I.; Frömke, C.; Bosse, H.M.; Breitkreutz, J.; Meissner, T. Favorable acceptance of mini-tablets compared with syrup: A randomized controlled trial in infants and preschool children. J. Pediatr. 2013, 163, 1728. [Google Scholar] [CrossRef]
  61. Rance, F.; Deslandes, B.; Decosta, P. Acceptability and tolerance of prednisolone metasulfobenzoate in orally dispersing tablets in 2 to 12-years-old children. Arch. Pediatr. 2004, 11, 1127–1130. [Google Scholar] [CrossRef]
  62. Ita, K. Transdermal delivery of drugs with microneedles-potential and challenges. Pharmaceutics 2015, 7, 90–105. [Google Scholar] [CrossRef]
  63. Larrañeta, E.; Lutton, R.E.M.; Woolfson, A.D.; Donnelly, R.F. Microneedle arrays as transdermal and intradermal drug delivery systems: Materials science, manufacture and commercial development. Mater. Sci. Eng. R 2016, 104, 1–32. [Google Scholar] [CrossRef] [Green Version]
  64. Sharma, S.; Hatware, K.; Bhadane, P.; Sindhikar, S.; Mishra, D.K. Recent advances in microneedle composites for biomedical applications: Advanced drug delivery technologies. Mater. Sci. Eng. C 2019, 103, 109717. [Google Scholar] [CrossRef] [PubMed]
  65. Gonzalez-Vazquez, P.; Larraneta, E.; McCrudden, M.T.C.; Jarrahian, C.; Rein-Weston, A.; Quintanar-Solares, M.; Zehrung, D.; McCarthy, H.; Courtenay, A.J.; Donnelly, R.F. Transdermal delivery of gentamicin using dissolving microneedle arrays for potential treatment of neonatal sepsis. J. Control Release 2017, 265, 30–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Map of factors of active pharmaceutical ingredients (API) affecting dispersible tablet formulation approaches.
Figure 1. Map of factors of active pharmaceutical ingredients (API) affecting dispersible tablet formulation approaches.
Pharmaceutics 11 00518 g001
Table 1. Exemplative comparison of potential considerations for pediatric medicine, pharmaceutical development, and supply requirements for high income economies compared to low- and middle-income economies.
Table 1. Exemplative comparison of potential considerations for pediatric medicine, pharmaceutical development, and supply requirements for high income economies compared to low- and middle-income economies.
PropertyTraditional Pharma Drug DevelopmentGlobal Health Drug DevelopmentImpact on Development Opportunities for Low Resource Settings
Target population0–18 years0–18 yearsNone
Excipients (safety)Acceptable for proposed patientsAcceptable for proposed patientsNone
AcceptabilityPalatable, non-irritantPalatable, non-irritantMinimal. Potential cultural differences (e.g., flavors) may need to be considered
Dose preparationMinimal manipulation/preparation, oral products may be mixed with water/food/beverageMinimal manipulation, preferably no preparationAvoid requirement for mixing if possible. If required, consider readily available vehicles, e.g., breast milk
Administration Easy to administer, use of administration device if necessaryEasy to administer, preferably no administration device neededConsider alternative options for dosing (no device)
Storage conditions25 °C–30 °C/60–65% RH, refrigerated accepted (2–8 °C)30 °C/75% RHReady to use liquids and semi-solids less favorable
PackagingVarious, no restrictionsCompact, light in weight, robustSelect light/compact container closure (preferably not glass)
CostLow overall cost ideal but not necessaryLow – standard non-complex manufacturing with low cost raw materialsSelect only routine processes and commonly available, non-specialist, low cost excipients
Supply chainGenerally well-developed and efficientPoorly developed and fragmentedLonger shelf life may be required, robust packaging
RegulatoryMature and well-recognized regulatory requirementsDisparate regulatory requirementsTailor regulatory strategy to each country/market
Table 2. Advantages and disadvantages of oral dispersible dosage forms.
Table 2. Advantages and disadvantages of oral dispersible dosage forms.
AdvantagesDisadvantages
Dosing
Once dispersed in liquid is easy to swallow; suitable for the whole pediatric population from birth upwardsLimited dose flexibility, although a break line may be introduced to sub-divide tablets; granules generally provided in unit-dose packs (e.g., sachet); more than one dose strength may be needed to cover the required dose range
Administration
Generally non-complex and simple method of administration, with no need for measuring device (e.g., dosing cup, spoon, or oral syringe)Requires dispersion in water or other beverage prior to administration; whole volume of liquid dispersion must be taken; rinsing of vessel may be required to ensure all residue (if any) is taken
Excipient Safety
Do not require the inclusion of preservatives; many excipients commonly used in dispersible dosage forms have an acceptable safety profile in pediatric patientsMay require sweetener and/or flavor to ensure acceptable palatability
Stability
Better stability than liquids or semi-solidsMay need moisture protective packaging; in-use stability once dispersed likely to be limited; compatibility with dispersing vehicle should be confirmed
Manufacture and Supply Chain
Non-complex development process; standard manufacturing and packaging equipment may be used; low bulk/footprint; easy to store and transportHumidity control may be required during manufacture
Table 3. Examples of Dispersible Products for Low to Middle-Income Countries.
Table 3. Examples of Dispersible Products for Low to Middle-Income Countries.
Product NameAPI and StrengthIndication
Coartem® DispersibleArtemether 20 mg/Lumefantrine 120 mgUncomplicated malaria due to Plasmodium falciparum.
SPAQ-COAmodiaquine 150 mg Sulfadoxine-Pyrimethamine 500 mg/25 mgSeasonal malaria chemoprevention
Paracetamol Dispersible tabletsParacetamol 100 mg and 250 mgPain
Zinc Dispersible tabletsZinc 20 mgDiarrhea
Amoxicillin Dispersible tabletsAmoxicillin 125 mg and 250 mgPneumonia
Sulfamethoxazole/Trimethoprim Dispersible tablets (cotrimoxazole)Sulfamethoxazole 100 mg/Trimethoprim 20 mgPneumocystis pneumonia, prophylaxis against infections in HIV patients
Lamivudine/Nevirapine/Zidovudine 30/50/60 mg dispersible tabletsLamivudine 30 mg/Nevirapine 50 mg/Zidovudine 60 mgTreatment of HIV-1
Lamivudine/Stavudine/NevirapineLamivudine 30 mg/Stavudine 6 mg/Nevirapine 50 mg and Lamivudine 60 mg/Stavudine 12 mg/Nevirapine 100 mgTreatment of HIV
API: Active Pharmaceutical Ingredient; HIV: Human Immunodeficiency Virus.
Table 4. Advantages and disadvantages of suppositories.
Table 4. Advantages and disadvantages of suppositories.
AdvantagesDisadvantages
Dosing
Suitable for pediatric patients from one month and unconscious or vomiting patients; able to deliver high doses of API; generally avoids first pass metabolism; can be used for local or systemic delivery; suitable for APIs that are gastro-irritant or prone to degradation in the stomachMay be associated with variable API absorption; potentially reduced API absorption if rectum is not empty; not recommended in preterm neonates due to risk of trauma and resulting infection; limited dose flexibility and more than one dose strength/size may be needed to cover the required dose range
Administration
Generally non-complex method of administration, although training may be required to ensure correct insertion; no need for administration device although some devices are available (e.g., suppository inserter)Route of administration may not be acceptable to some patients/caregivers (social/cultural reasons), leading to non- compliance; suppository may be expelled (involuntarily or via defecation)
Excipient Safety
Do not require the inclusion of sweeteners or flavor; many excipients commonly used in suppositories are well tolerated by the rectal mucosa and have an acceptable safety profile in pediatricsSome excipients may cause mucosal irritation
Stability
Can melt at temperatures above 30 °C
Manufacture and Supply Chain
Relatively low-cost excipients; lower bulk/footprint compared to liquids; easier to transport than liquidsManufacture more difficult than other common dosage forms (tablets, liquids); may need temperature-controlled storage (depending on melting point); humidity control may be required during manufacture

Share and Cite

MDPI and ACS Style

Gerrard, S.E.; Walsh, J.; Bowers, N.; Salunke, S.; Hershenson, S. Innovations in Pediatric Drug Formulations and Administration Technologies for Low Resource Settings. Pharmaceutics 2019, 11, 518. https://doi.org/10.3390/pharmaceutics11100518

AMA Style

Gerrard SE, Walsh J, Bowers N, Salunke S, Hershenson S. Innovations in Pediatric Drug Formulations and Administration Technologies for Low Resource Settings. Pharmaceutics. 2019; 11(10):518. https://doi.org/10.3390/pharmaceutics11100518

Chicago/Turabian Style

Gerrard, Stephen E., Jennifer Walsh, Niya Bowers, Smita Salunke, and Susan Hershenson. 2019. "Innovations in Pediatric Drug Formulations and Administration Technologies for Low Resource Settings" Pharmaceutics 11, no. 10: 518. https://doi.org/10.3390/pharmaceutics11100518

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop