<< Back To Home

CSU 86/2011: INTRADERMAL DELIVERY OF VACCINES: POTENTIAL BENEFITS AND CURRENT CHALLENGES

Friday, 11th of March 2011 Print

Intradermal vaccine delivery looks like a highly technical topic, but it might be the Next Big Thing in vaccination. If solid evidence is available that fractional doses of certain parenterally administered vaccines can be given intradermally without significant differences in seroconversion rates, then – once licensing and other hurdles were overcome – the door would be open to lower costs per fully immunized child. This would be a refreshing change from the last decade, where vaccine costs per fully vaccinated child quintupled in some GAVI beneficiary countries.


INTRADERMAL DELIVERY OF VACCINES: POTENTIAL BENEFITS AND CURRENT CHALLENGES
 
Among the commonly used child vaccines, only BCG is given by the intradermal route. Since intradermal administration can, in some cases, permit dose reduction, the ID route becomes attractive from the economic standpoint, especially for vaccines too costly to give in most developing countries.
 
Good reading.
 
BD

 

JK Hickling,a KR Jones,a M Friede,b D Zehrung,c D Chenc & D

Kristensenc

a Working in Tandem, Cambridge, England.

b Department of Immunization, Vaccines and Biologicals, World Health Organization, Geneva,

Switzerland.

c Program for Appropriate Technology in Health (PATH), PO Box 900922, Seattle, WA, 98109,

United States of America.

Correspondence to Darin Zehrung (e-mail: dzehrung@path.org)

(Submitted: 5 May 2010 – Revised version received: 1 November 2010 – Accepted: 2 November

2010 – Published online:21 January 2010)

 

From www.who.int/bulletin/en/

 

Abstract

Delivery of vaccine antigens to the dermis and/or epidermis of human skin (i.e. intradermal delivery) might be more efficient than injection into the muscle or subcutaneous tissue, thereby reducing the volumes of antigen. This is known as dose-sparing and has been demonstrated in clinical trials with some, but not all, vaccines. Dose-sparing could be beneficial to immunization programmes by potentially reducing the costs of purchase, distribution and storage of vaccines; increasing vaccine availability and effectiveness. The data obtained with intradermal delivery of some vaccines are encouraging and warrant further study and development; however significant gaps in knowledge and operational challenges such as reformulation, optimizing vaccine presentation and development of novel devices to aid intradermal vaccine delivery need to be addressed. Modelling of the costs and potential savings resulting from intradermal delivery should be done to provide realistic expectations of the potential benefits and to support cases for investment. Implementation and uptake of intradermal vaccine delivery requires further research and development, which depends upon collaboration between multiple stakeholders in the field of vaccination.

 

Introduction

Most vaccines are delivered by the intramuscular or subcutaneous routes using a needle and syringe; the intradermal route is only widely used for the administration of Bacille Calmette-Guérin and rabies vaccines. However there is renewed interest in intradermal vaccine delivery, driven by the fact that the dermis and epidermis of human skin are rich in antigen-presenting cells, suggesting that delivery of vaccines to these layers, rather than to muscle or subcutaneous tissue, should be more efficient and induce protective immune responses with smaller amounts of vaccine antigen.1

Clinical trials investigating intradermal delivery and its potential for dose-sparing have been conducted with several different vaccines, with variable results. These have been reviewed in a recent report from the Program for Appropriate Technology in Health (PATH) and the World Health Organization (WHO).2 For some vaccines, there has been a clear demonstration of dose-sparing by intradermal delivery; however, there are several

gaps in knowledge as well as developmental and operational challenges to overcome if the benefits of using intradermal delivery are to be fully realized.

 
Potential benefits

Dose-sparing arising from intradermal delivery of vaccines could be beneficial to immunization programmes, particularly in resource-poor settings, by potentially reducing the per-injection cost (including transport and storage) of vaccines because more doses might be obtained from the existing vaccine presentation.

Dose-sparing might also “stretch” the availability of vaccines in cases where supply is limited by manufacturing capacity. This is probably most relevant for pandemic influenza vaccines where global production capacity limits access to a vaccine at the start of a pandemic.3 In 2009, H1N1 vaccine was not available in most low-income countries until 8 months after WHO’s declaration of the influenza pandemic.4

 Other vaccines with potential supply constraints include yellow fever and inactivated poliovirus vaccines.5,6 The level of demand for inactivated poliovirus vaccine in the period following eradication of wild-type polioviruses and the end of the use of oral poliovirus vaccines is uncertain, but modelling suggests that there could be a “demand spike” and supply shortage of inactivated poliovirus vaccine during this period.6

 

New delivery devices

New devices for easier, more reliable intradermal delivery as alternatives to the currently used Mantoux technique are being developed.2,7 Some of the devices such as disposablesyringe jet injectors are needle-free and could therefore reduce or eliminate needlestick injuries and the costs associated with their treatment, estimated at US$ 535 million per year worldwide.8 Other intradermal delivery devices such as microneedle patches are

likely to occupy less volume than vials or prefilled syringes, thereby reducing demands on cold-chain capacity.

 

Current clinical research

The recent PATH and WHO report reviewed more than 90 clinical trials of intradermal delivery with vaccines against 11 diseases.2 For some vaccines, notably influenza and rabies vaccines, intradermal delivery of reduced doses resulted in equivalent immune responses to the standard dose delivered by the standard route. Data from trials with hepatitis B vaccine were more variable, but also regarded as encouraging.9 Promising data demonstrating dose-sparing have also been obtained with other vaccines including inactivated poliovirus, yellow fever and hepatitis A vaccines.2

Results from clinical trials published since the completion of the report provide further evidence for dose-sparing using the intradermal route. One study compared equivalent doses of modified vaccinia Ankara delivered by subcutaneous, intramuscular and intradermal routes; equivalent immune responses and protection against vacciniavirus challenge were induced with intradermal doses ten-fold lower than those delivered by intramuscular or subcutaneous injection.10,11
 

Two recently published trials of intradermal delivery of reduced (20%) doses of inactivated poliovirus vaccines have reported contrasting results: one trial found that reduced intradermal doses administered to infants aged 2, 4 and 6 months induced similar rates of seroconversion but lower mean antibody titres, compared with intramuscular injection12; however a similar study administered the vaccine at 6, 10 and 14 weeks of age and observed inferior seroconversion rates with reduced intradermal doses.13 Trials of intradermal immunization with seasonal and pandemic influenza

vaccines have also been reported in the past year. Dose-sparing was not observed with a nonadjuvanted, subvirion H5N1 vaccine,14 whereas a 60% intradermal dose of a trivalent seasonal flu vaccine resulted in similar immunogenicity as a standard dose delivered intramuscularly.15

Some rabies vaccines are already delivered using intradermal regimens using needle and syringe.16,17 Recently, a new intradermal injection device (Soluvia,™ Becton Dickinson, Franklin Lakes, NJ, United States of America) was evaluated for intradermal delivery of rabies vaccine; intradermal administration of a 25% dose of rabies vaccine resulted in equivalent antibody titres and seroconversion rates to the full dose delivered

intramuscularly. The same study also evaluated an epidermal vaccination device (Onvax,® Becton Dickinson) that did not induce an immune response in the recipients.18
 

Recent vaccine approvals

A new presentation of a split, trivalent seasonal influenza vaccine, Intanza® (Sanofi pasteur, Lyon, France) has been developed for intradermal immunization. The vaccine is delivered using the Soluvia device, a prefilled syringe with a single needle that is 1.5 mm in length.19,20 Immune responses equivalent to those induced by the standard regimen can be achieved using this device and 60% of the standard dose of influenza vaccine.21,22 A

reduced-dose formulation (9-μg haemagglutinin rather than the standard 15-μg haemagglutinin) for use in healthy adults and a 15-μg haemagglutinin formulation for intradermal delivery in adults aged ³ 60 years have been approved in Australia, Europe and New Zealand.23–25

 
Knowledge gaps

Despite the number of clinical trials conducted, there is still a lack of certainty whether intradermal delivery offers significant immunological advantages compared with intramuscular or subcutaneous delivery. Very few trials have compared equivalent doses (amounts of antigen) delivered by the different routes and, as such, do not permit direct comparison of relative immunological efficacy of the different methods; the recent studies with modified vaccinia Ankara are notable exceptions.10,11 Most trials have aimed only to determine whether reduced intradermal doses were able to induce a sufficient or non-inferior immune response compared with the standard regimen. In these cases it is possible that a reduced dose delivered by the standard route would also have resulted in an equivalent immune response, as has been reported with some influenza vaccines.26,27 Furthermore, the majority of dose-sparing trials have for reasons of practical simplicity delivered 10% or 20% of the standard dose intradermally. Finer dose-titrations such as

those performed with influenza vaccines have shown that less-dramatic reductions in dose, such as a 40% reduction, might be more realistic.22 To address these concerns, future studies should compare equivalent antigen doses given by the intradermal and standard route and, if possible, establish a dose–response relationship to determine whether any dose-sparing effect observed is simply due to using doses of antigen from the plateau-portion of the dose–response curve.

 

Which vaccines?

For any vaccine, the drivers for changing to the intradermal route, such as the need for dose-sparing to reduce costs and “stretch” the available manufacturing capacity, have to be assessed, but the suitability of the vaccine type and formulation for administration by this route also need to be considered.
 

Live-attenuated vaccines have been successfully delivered intradermally and should be good candidates for intradermal delivery providing that appropriate formulations can be developed.2 Reduced doses of inactivated whole-virion vaccines, such as rabies and inactivated poliovirus vaccines, have also shown satisfactory immunogenicity when delivered intradermally. Inactivated whole-virion influenza vaccines might also be suitable because they have intrinsic immune-stimulating sequences, which might avoid the need for additional adjuvants.28 There are no published clinical data regarding intradermal delivery of polysaccharide conjugate vaccines, which include meningococcal and pneumococcal conjugate vaccines, yet evaluation would be

worthwhile particularly because high cost has limited the introduction of pneumococcal conjugate vaccines in some low-resource countries.29

Vaccines that contain aluminium-based or oil-in-water adjuvants are likely to have unacceptable local reactogenicity following intradermal administration. Clinical trials of intradermal delivery of vaccines formulated with these or novel adjuvants are needed.

 

Potential benefits

Realistic estimates of the cost savings that might be achieved by ID dose-sparing have not been established for most vaccines, especially those that might be relevant to routine immunization in low- and middle-resource countries. Cell-culture-produced rabies vaccines are expensive and are used in complex,

multi-dose schedules for post-exposure prophylaxis. A limited number of cell-culturederived rabies vaccines and two intradermal immunization regimens are already recommended by WHO.16,17 The overall costs involved in administering post-exposure prophylaxis using intramuscular or intradermal regimens in different clinic settings in India have been compared.30 Although significant savings were achieved with intradermal regimens, reducing the dose to 20% of the intramuscular dose did not reduce the overall costs by 80%, instead savings of 15–38% were obtained, depending on the

setting. Intradermal regimens that used 10% rather than 20% of the standard dose resulted in a further reduction in costs of only 9–15%. Furthermore, the estimates did not include costs resulting from vaccine wastage, which were expected to be higher in intradermal regimens.

 
This example reinforces the point that the cost of manufacturing a vaccine is only a proportion of the overall cost of delivering each dose; furthermore, vaccine manufacturers will not necessarily pass on the full savings arising from a reduction in antigen content per dose. Novel devices for intradermal delivery are likely to be more expensive than needles and syringes, and immunization programmes using new devices will also incur the costs of retraining health workers.
 

A major benefit of changing to needle-free devices is a reduction in the direct health-care and societal costs resulting from needlestick injuries, although this is difficult to quantify in financial terms.8,31 A reduction in the use of sharps and their associated costs could also be achieved, possibly more easily, by using needle-free delivery devices such as disposable-syringe jet injectors, without changing the route or dose of vaccine

administered.
 

The incremental costs associated with changing some routine childhood accinations in Brazil, India and South Africa to intradermal delivery of reduced (20%) doses have been modelled.32 For India and South Africa, the model indicated that intradermal delivery of reduced doses of hepatitis B vaccine using disposable-syringe jet injectors would increase the overall costs per fully-immunized child by US$ 0.45 and by US$ 0.76, respectively; hepatitis B vaccine is only one of five or six infant vaccines routinely administered in these two countries. In contrast, in Brazil, the model indicated

that intradermal reduced doses of hepatitis B and yellow fever vaccines would reduce overall costs per fully-immunized child by US$ 0.11. Although antigen content of the vaccines delivered intradermally was reduced by 80%, the model showed that prices w re only reduced by 20%. The model did not include costs associated with treatment of bloodborne infections transmitted by sharps.

 
PATH has modelled the potential incremental costs of routine immunization with inactivated poliovirus vaccines in India. This analysis assumed that the full cost savings resulting from reduced antigen content were passed on by manufacturers and included estimates for costs due to transmission of bloodborne infection. In this case, delivery of a 20% intradermal dose with either needle and syringe or disposable-syringe jet injectors
could result in cost savings of 71–73% per immunized child.33
 
In applications where it is warranted, significant investment by vaccine manufacturers and other stakeholders will be required to change the route of vaccine delivery. It is important, therefore, to model accurately the costs involved to determine the potential benefits and to support the case for investment. Decisions should be based on cost modelling and economic analysis, with continual improvement of inputs and assumptions.
 

Challenges

Changing the delivery route and dose for a vaccine is not a straightforward task, and several challenges and operational issues need to be addressed.

 

Demonstration of efficacy

The applicability and benefits of intradermal administration will vary among vaccines; it cannot be assumed that reduced intradermal doses will be efficacious in all cases. Even when there is dose-sparing, it might not be with a dose as low as 20% or 10% of the standard dose. The degree of dose-sparing that can be achieved will need to be determined for each vaccine in non-inferiority trials. It will also be important to maintain a “margin of safety” to ensure adequate immunogenicity across the whole target population and to ensure the potency of the reduced dose is sufficient to induce protection at the end of the vaccine’s shelf life.

 

Reformulation

Intradermal doses of 20% of a standard 0.5 ml dose or 10% of a 1.0 ml dose might not require reformulation; 0.1 ml of the existing formulation could easily be administered, as is the usual practice in clinical trials of intradermal delivery. If more antigen per dose is required, then the vaccine will need to be concentrated so that it can be delivered in the smaller volume. In the development of Intanza®, which contains 60% of the standard amount of antigen in 20% of the volume, there were challenges filling the device with a small volume as well as adjusting vaccine concentration and viscosity.34Even if there is no need to adjust the concentration of the vaccine, it might be necess ry to incorporate a preservative into the formulation if dose-sparing means that more than one dose can be obtained per vial.

 

Vaccine presentation

One consequence of delivering a smaller volume per dose is that a single-dose vial presentation would effectively become a multi-dose vial, yielding perhaps 5 or 10 intradermal doses. Liquid vaccines with multi-dose vial presentations can contain preservatives so that unused vaccine can be stored more safely beyond the end of an immunization session.35 Single-dose vials do not typically contain thiomersal or other preservatives. If dose-sparing allows multiple doses to be drawn from preservative-free vaccines in single-dose vials, unused vaccine would have to be discarded at the end of the

immunization session. Computer modelling has shown that matching the number of doses per vial with the immunization session’s size has a significant economic impact in terms of minimizing vaccine wastage as well as supply, storage and disposal costs.36 For some vaccines and settings, fewer than five doses per vial would be the most cost-effective option. Therefore, alternative, smaller-volume presentations might be required, otherwise

intradermal delivery might not yield the expected cost savings in settings where immunization session-sizes are small.

 

Regulatory issues

Intradermal delivery of fractional doses of an existing vaccine formulation intended for subcutaneous/intramuscular injection would have to be undertaken on an “off-label” basis. A licensing amendment between the relevant national regulatory authority and the vaccine producer would be needed to authorize official “on-label” use of fractional doses.37 Alternatively, marketing approval for the new intradermal formulation and

presentation of the vaccine would be needed.
 

For many existing vaccines, there might not be a sufficiently strong commercial incentive for manufacturers to undertake the expensive processes of reformulation, production of new presentations and application for marketing authorizations for a new delivery route. Novel vaccines might, therefore, be more likely candidates for intradermal delivery. This route should be evaluated early in research and development to avoid expensive retesting and redevelopment at a later stage. Unless there are strong drivers to use the intradermal route, vaccine manufacturers are likely to continue to use standard

presentations and routes of delivery even for new vaccines, thereby reducing risk from the development process.

 
Device development

Novel devices for simple, reliable and reproducible intradermal delivery of vaccines will be needed if this route is to be widely used. There are several different types of intradermal delivery devices in development.1,2,7 However they cannot be developed in isolation; the device inventors require access to vaccines and collaboration with vaccine manufacturers to enable development, testing and approval of device/vaccine combinations.

 

In the shorter term, delivery technologies could include simple adapters fitted to existing needles and syringes to control the depth and angle of delivery, syringe-mounted arrays of hollow microneedles and needle-free disposable-syringe jet injectors.2 Each of these approaches is compatible with existing liquid and lyophilized formulations and presentations and so should be simpler to introduce into immunization programmes than some other technologies.

 
Devices that are in earlier stages of development include skin-patches covered in microneedles coated with, or composed of, vaccine. Encouraging preclinical data have been obtained with several formats of this type of device.38–40 Challenges remain in producing solid formulations of different types of vaccine antigens, developing methods for coating sufficient antigen onto the microneedles and controlling the reproducibility of antigen delivery. If these can be overcome, microneedle patches could provide several benefits in addition to dose-sparing, including small-packaged volumes and simplicity of use.

 

Conclusion

Data from clinical trials indicate that intradermal delivery of reduced doses of some, although not all, vaccines results in equivalent immune responses to the standard regimen. An increasing number of trials are producing data that support dose-sparing. However, there are significant operational challenges, such as reformulation, changing from a single- to a multiple-dose presentation, development of intradermal delivery

devices and training health workers. Economic studies and modelling exercises have shown that cost savings should be achievable with intradermal immunization with some vaccines. These analyses are needed to help guide and support development and implementation of intradermal vaccine delivery as well as provide an indication of potential savings.

 

Research and development in all of these disciplines is required if the promise of intradermal delivery is to be realized; this will require collaboration between academic and clinical researchers, vaccine manufacturers, device developers, regulatory authorities, national immunization programmes and nongovernmental global health organizations. A careful evaluation of all aspects of this route of delivery for vaccines should continue to

be a priority for global public health.

 

Acknowledgements

We thank Courtney Jarrahian and Kristina Lorensen for reviewing the manuscript and

Emily Griswold for proofreading.

 

Funding:

Work on this article by PATH staff members and consultants was supported by funding

from the Bill & Melinda Gates Foundation. The authors had full control of all primary

data.

Competing interests:

None declared.

 

References

1. Lambert PH, Laurent PE. Intradermal vaccine delivery: will new delivery

systems transform vaccine administration? Vaccine 2008;26:3197-208.

PMID:18486285 doi:10.1016/j.vaccine.2008.03.095

2. Intradermal delivery of vaccines: a review of the literature and potential for

development for use in low- and middle-income countries. Seattle:

Program for Appropriate Technology in Health (PATH); 2009. Available

from: http://www.path.org/files/TS_opt_idd_review.pdf [accessed 16

December 2010].

3. Kieny MP, Costa A, Hombach J, Carrasco P, Pervikov Y, Salisbury D, et al. A

global pandemic influenza vaccine action plan. Vaccine 2006;24:6367-70.

PMID:17240560 doi:10.1016/j.vaccine.2006.07.021

4. Partridge J, Kieny MP. Global production of seasonal and pandemic (H1N1)

influenza vaccines in 2009-2010 and comparison with previous estimates

and global action plan targets. Vaccine 2010;28:4709-12. PMID:20488262

doi:10.1016/j.vaccine.2010.04.083

5. Yellow fever initiative: providing an opportunity of a lifetime. Geneva: World

Health Organization; 2010. Available from:

http://www.who.int/csr/disease/yellowfev/YFIbrochure.pdf [accessed 16

December 2010].

6. Oliver Wyman Inc. Global post-eradication IPV supply and demand

assessment: integrated findings. Seattle: Bill & Melinda Gates Foundation;

2009. Available from:

http://www.polioeradication.org/content/general/March%202009%20OW%

20IPV%20Effort%20Report.pdf [accessed 16 December 2010].

7. Weniger BG, Papania MJ. Alternative vaccine delivery methods. In: Plotkin

SA, Orenstein WA, Offit PA, eds. Vaccines, 5th ed. Amsterdam: Elsevier;

2008:1357-92.

8. Miller MA, Pisani E. The cost of unsafe injections. Bull World Health Organ

1999;77:808-11. PMID:10593028

9. Sangaré L, Manhart L, Zehrung D, Wang CC. Intradermal hepatitis B

vaccination: a systematic review and meta-analysis. Vaccine

2009;27:1777-86. PMID:19200451 doi:10.1016/j.vaccine.2009.01.043

10. Wilck MB, Seaman MS, Baden LR, Walsh SR, Grandpre LE, Devoy C, et al.

Safety and immunogenicity of modified vaccinia Ankara (ACAM3000):

effect of dose and route of administration. J Infect Dis 2010;201:1361-70.

PMID:20350191 doi:10.1086/651561

11. Seaman MS, Wilck MB, Baden LR, Walsh SR, Grandpre LE, Devoy C, et al.

Effect of vaccination with modified vaccinia Ankara (ACAM3000) on

subsequent challenge with Dryvax. J Infect Dis 2010;201:1353-60.

PMID:20350190 doi:10.1086/651560

12. Mohammed AJ, AlAwaidy S, Bawikar S, Kurup PJ, Elamir E, Shaban MMA,

et al. Fractional doses of inactivated poliovirus vaccine in Oman. N Engl J

Med 2010;362:2351-9. PMID:20573923 doi:10.1056/NEJMoa0909383

13. Resik S, Tejeda A, Lago PM, Diaz M, Carmenates A, Sarmiento L, et al.

Randomized controlled clinical trial of fractional doses of inactivated

poliovirus vaccine administered intradermally by needle-free device in

Cuba. J Infect Dis 2010;201:1344-52. PMID:20350164

doi:10.1086/651611

14. Patel SM, Atmar RL, El Sahly HM, Cate TR, Keitel WA. A phase I evaluation

of inactivated influenza A/H5N1 vaccine administered by the intradermal

or the intramuscular route. Vaccine 2010;28:3025-9. PMID:19931380

doi:10.1016/j.vaccine.2009.10.152

15. Chi RC, Rock MT, Neuzil KM. Immunogenicity and safety of intradermal

influenza vaccination in healthy older adults. Clin Infect Dis 2010;50:1331-

8. PMID:20377407 doi:10.1086/652144

16. WHO expert consultation on rabies (WHO technical report series 931).

Geneva: World Health Organization; 2005. Available from:

http://www.who.int/rabies/trs931_%2006_05.pdf [accessed 16 December

2010].

17. World Health Organization. Rabies vaccines. WHO position paper. Wkly

Epidemiol Rec 2007;82:425-35. PMID:18064757

18. Laurent PE, Bourhy H, Fantino M, Alchas P, Mikszta JA. Safety and efficacy

of novel dermal and epidermal microneedle delivery systems for rabies

vaccination in healthy adults. Vaccine 2010;28:5850-6. PMID:20600481

doi:10.1016/j.vaccine.2010.06.062

19. Alarcon JB, Hartley AW, Harvey NG, Mikszta JA. Preclinical evaluation of

microneedle technology for intradermal delivery of influenza vaccines. Clin

Vaccine Immunol 2007;14:375-81. PMID:17329444

doi:10.1128/CVI.00387-06

20. Laurent PE, Bonnet S, Alchas P, Regolini P, Mikszta JA, Pettis R, et al.

Evaluation of the clinical performance of a new intradermal vaccine

administration technique and associated delivery system. Vaccine

2007;25:8833-42. PMID:18023942 doi:10.1016/j.vaccine.2007.10.020

21. Arnou R, Eavis P, Pardo JDJ, Ambrozaitis A, Kazek M, Weber F.

Immunogenicity, large-scale safety and lot consistency of an intradermal

influenza vaccine in adults aged 18–60 years: Randomized, controlled,

phase III trial. Hum Vaccin 2010;6:346-54. PMID:20372053

doi:10.4161/hv.6.4.10961

22. Beran J, Ambrozaitis A, Laiskonis A, Mickuviene N, Bacart P, Calozet Y, et

al. Intradermal influenza vaccination of healthy adults using a new

microinjection system: a 3-year randomised controlled safety and

immunogenicity trial. BMC Med 2009;7:13. PMID:19341446

doi:10.1186/1741-7015-7-13

23. Committee for Medical Products for Human Use. Summary of product

characteristics (Intanza 9microgram). Available from:

http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-

_Product_Information/human/000957/WC500033852.pdf [accessed 16

December 2010].

24. Arnou R, Icardi G, De Decker M, Ambrozaitis A, Kazek M, Weber F, et al.

Intradermal influenza vaccine for older adults: a randomized controlled

multicenter phase III study. Vaccine 2009;27:7304-12. PMID:19849996

doi:10.1016/j.vaccine.2009.10.033

25. Australian Drug Evaluation Committee. ADEC 262nd meeting resolutions;

resolution 9254. Commonwealth of Australia Gazette 2009;11. Available

from: http://www.tga.gov.au/docs/html/adec/adec0262.htm [accessed 16

December 2010].

26. Treanor J, Keitel W, Belshe R, Campbell J, Schiff G, Zangwill K, et al.

Evaluation of a single dose of half strength inactivated influenza vaccine in

healthy adults. Vaccine 2002;20:1099-105. PMID:11803070

doi:10.1016/S0264-410X(01)00440-6

27. Belshe RB, Newman FK, Wilkins K, Graham IL, Babusis E, Ewell M, et al.

Comparative immunogenicity of trivalent influenza vaccine administered

by intradermal or intramuscular route in healthy adults. Vaccine

2007;25:6755-63. PMID:17692438 doi:10.1016/j.vaccine.2007.06.066

28. Geeraedts F, Goutagny N, Hornung V, Severa M, de Haan A, Pool J, et al.

Superior immunogenicity of inactivated whole virus H5N1 influenza

vaccine is primarily controlled by Toll-like receptor signalling. PLoS Pathog

2008;4:e1000138. PMID:18769719

29. World Health Organization. Worldwide progress in introducing pneumococcal

conjugate vaccine, 2000–2008. Wkly Epidemiol Rec 2008;83:388-92.

PMID:18949860

30. Goswami A, Plun-Favreau J, Nicoloyannis N, Sampath G, Siddiqui MN,

Zinsou J. The real cost of rabies post-exposure treatments. Vaccine

2005;23:2970-6. PMID:15811642 doi:10.1016/j.vaccine.2004.12.008

31. Ekwueme DU, Weniger BG, Chen RT. Model-based estimates of risks of

disease transmission and economic costs of seven injection devices in

sub-Saharan Africa. Bull World Health Organ 2002;80:859-70.

PMID:12481207

32. Griffiths UK, Santos AC, Nundy N, Jacoby E, Matthias D. Incremental costs

of introducing jet injection technology for delivery of routine childhood

vaccinations: Comparative analysis from Brazil, India and South Africa.

Vaccine 2010 Nov 27 [Epub ahead of print]. Available from:

http://www.ncbi.nlm.nih.gov/pubmed/21115059 [accessed 16 December

2010].

33. Improving the affordability of inactivated poliovirus vaccines (IPV) for use in

low- and middle-income countries: an economic analysis of strategies to

reduce the cost of routine IPV immunization. Seattle: Program for

Appropriate Technology in Health (PATH); 2010. Available from:

http://www.path.org/publications/details.php?i=1809 [accessed 16

December 2010].

34. Picot V. Intradermal immunization: an alternative route for vaccine

administration. Articles as per sessions meeting report. Vaccine 2008;26

Suppl 9;S1-5. PMID:19268138 doi:10.1016/j.vaccine.2008.10.092

35. The use of opened multi-dose vials of vaccine in subsequent immunization

sessions. Geneva: World Health Organization; 2000. Available from:

http://www.who.int/vaccines-documents/DocsPDF99/www9924.pdf

[accessed 16 December 2010].

36. Lee BY, Norman BA, Assi T, Chen S, Bailey RR, Rajgopal J, et al. Single

versus multi-dose vaccine vials: an economic computational model.

Vaccine 2010;28:5292-300. PMID:20566395

doi:10.1016/j.vaccine.2010.05.048

37. Report of a meeting on priorities for pneumococcal and Haemophilus

influenzae type b (Hib) vaccine development, February 1999. Geneva:

World Health Organization; 2001. Available from:

http://www.who.int/vaccines-documents/DocsPDF01/www530.pdf

[accessed 16 December 2010].

38. Kim YC, Quan F, Compans RW, Kang S, Prausnitz MR. Formulation and

coating of microneedles with inactivated influenza virus to improve vaccine

stability and immunogenicity. J Control Release 2010;142:187-95.

PMID:19840825 doi:10.1016/j.jconrel.2009.10.013

39. Sullivan SP, Koutsonanos DG, Del Pilar Martin M, Lee JW, Zarnitsyn V, Choi

S, et al. Dissolving polymer microneedle patches for influenza vaccination.

Nat Med 2010;16:915-20. PMID:20639891 doi:10.1038/nm.2182 Available

from: http://www.ncbi.nlm.nih.gov/pubmed/20639891 [accessed 16

December 2010].

40. Fernando GJP, Chen X, Prow TW, Crichton ML, Fairmaid EJ, Roberts MS, et

al. Potent immunity to low doses of influenza vaccine by probabilistic

guided micro-targeted skin delivery in a mouse model. PLoS ONE

2010;5:e10266. PMID:20422002 doi:10.1371/journal.pone.0010266



--

To subscribe or unsubscribe from these Child Survival Updates, pls contact kidsurvival@gmail.com. If you unsubscribe, indicate from which E mail address you are receving these updates.

 

When subscribing, write from your most permanent E-mail address, not always that of your current employer.

 
Do not subscribe on behalf of friends or colleagues; forward updates to them for their decision.

Those wishing to read only malaria updates should subscribe at
kidsurvivalmalaria@gmail.com

 


Those wishing to read only vaccination updates should subscribe at kidsurvivalvaccination@gmail.com

 

READER COMMENTS

 

If you have a comment you want posted, send to rdavis@africamail.com


 

WEBPAGE

 

These updates are also available at www.childsurvival.net  

 

40757765