Platelets

Though operational practices vary centre by centre, the routine use of INTERCEPT™ pathogen inactivation technology for platelets in Europe has demonstrated significant operational gains that translate into cost savings.

  • The replacement of gamma irradiation, cytomegalovirus (CMV) testing, and bacterial detection with pathogen inactivation can result in an estimated savings of €20 to €30 per platelet unit.1,2
  • The ability to prolong platelet storage in some countries with pathogen inactivation enables centres to reduce discard rates. Assuming an improvement of wastage by 10% and an average platelet unit price of €250 to €600, increased shelf life could save €25 to €60 per unit.3,4
  • Pathogen inactivation mitigates the differential risk attained between apheresis and whole-blood derived platelets; thus providing the ability to achieve savings via an optimised production ratio in terms of collection methods.5,6,7,8,14
  • The amortisation of costs for a pathogen inactivation kit over more than one therapeutic unit through double dose apheresis collections decreases costs per kit by 50%.9
  • Significant savings have been modeled for the reduction of acute transfusion reactions, and when using pathogen inactivation as insurance against unknown pathogens.10,11,12,
  • The double dose buffy coat platelet production process can contribute significantly to the affordability of the INTERCEPTTM Blood System for platelets, and in some cases, may allow blood centres to implement pathogen inactivation in a cost-neutral or even a cost-saving way.
Sources:
1.Sigle JP, et al., Comparison of transfusion efficacy of amotosalen-based pathogen-reduced platelet components and gamma-irradiated platelet components, Transfusion (2013); 53:1788-1797.
2.McCullough J, et al., Cost implications of implementation of pathogen-inactivated platelets, Transfusion (2015), doi:10.1111/trf.13149
3.Report of the SaBTO Working Group: Pathogen Inactivation of Platelets
4.Veihola M, et al., Variation of platelet production and discard rates in 17 blood centers representing 10 European countries from 2000 to 2002, Transfusion (2006) 46, 991-995
5.Etablissement Français du Sang Cost Analysis, presented by JP Cazenave at the Consensus Conference, ‘Pathogen Inactivation: Making Decisions About New Technologies’, March 29, 2007 (Toronto, Canada).
6.Andreu G, et al., Use of random versus apheresis platelet concentrates, Transfusion Clinique et Biologique (2007) 14, 514-521
7.Vamvakas EC, Relative safety of pooled whole blood-derived versus single-donor (apheresis) platelets in the United States: a systematic review of disparate risks, Transfusion[JI2]  (2009), Commentary volume 49
8.Lozano ML, et al., Platelet concentrates from whole-blood donations (buffy-coat) or apheresis: which one to use?, Medicina Clínica (Barc.) (2012) 138, 528-533
9.Abedi MR & Doverud AC, Preparation and pathogen inactivation of double dose buffy coat platelet products using the INTERCEPT blood system, JoVE, The Journal of Visualized Experiments (2012) 70, e4414 – Peer-Reviewed Scientific Video Journal
10.Girona-Llobera, et al., Reducing the financial impact of pathogen inactivation technology for platelet components: our experience, Transfusion (2014) 54, 158-168
11.French National Agency for Medicine and Health Product Safety (ANSM), Haemovigilance Activity Report, 2012.
12.SwissMedic Haemovigilance Annual Reports, 2010-2012.
13.Rüesch, et al. Swissmedic Report. Two years’ experience with pathogen inactivation for all platelet concentrates in Switzerland
14.Berger K, et al., Model calculations to quantify clinical and economic effects of pathogen inactivation in platelet concentrates, Onkologie (2013)36, 53-59