Summary
Nowadays patients receiving blood components are exposed to much less transfusion-transmitted infectious diseases than three decades before when among others HIV was identified as causative agent for the acquired immunodeficiency syndrome and the transmission by blood or coagulation factors became evident. Since that time the implementation of measures for risk prevention and safety precaution was socially and politically accepted. Currently emerging pathogens like arboviruses and the well-known bacterial contamination of platelet concentrates still remain major concerns of blood safety with important clinical consequences, but very rarely with fatal outcome for the blood recipient. In contrast to the well-established pathogen inactivation strategies for fresh frozen plasma using the solvent-detergent procedure or methylene blue and visible light, the bench-to-bedside translation of novel pathogen inactivation technologies for cell-containing blood components such as platelets and red blood cells are still underway. This review summarizes the pharmacological/toxicological assessment and the inactivation efficacy against viruses, bacteria, and protozoa of each of the currently available pathogen inactivation technologies and highlights the impact of the results obtained from several randomized clinical trials and hemovigilance data. Until now in some European countries pathogen inactivation technologies are in in routine use for single-donor plasma and platelets. The invention and adaption of pathogen inactivation technologies for red blood cell units and whole blood donations suggest the universal applicability of these technologies and foster a paradigm shift in the manufacturing of safe blood.
Keywords: Pathogen inactivation, Blood safety, Dengue virus, Chikungunya virus, West Nile virus, Bacteria, Protozoa, Platelet concentrates, Red blood cells, Randomized clinical trial, Hemovigilance
Introduction
Until now, neither artificial oxygen carriers nor hemostatic agents have had the potential to replace manufactured red blood cell (RBC) units or platelet concentrates donated by healthy volunteers. This ‘natural source’ inherently bears the risk of transmitting blood-borne pathogens, although stringent donor selection criteria and very sensitive and specific blood screening tests have substantially increased the safety margin for blood recipients [1, 2, 3, 4]. In particular, the successful introduction of nucleic acid amplification testing (NAT) for HIV, HBV, and HCV have led to significantly safer blood products resulting in a residual risk of 1:1 million to 1:10 million [1, 5, 6, 7, 8]. Currently, the risk of bacterial contamination, especially in platelet concentrates (1:2,000 to 1:5,000), and transfusion-related bacterial sepsis (1:20,000 to 1:50,000), which is at times fatal (10%), are the most relevant infectious complications [1, 9, 10]. The vast majority of patients receiving platelet concentrates suffer from hematological malignancies, and they are vulnerable to bacteria, leading to higher rates of infectious complications, associated comorbidities, and prolonged hospital stays [11, 12, 13]. Furthermore, transfusion-associated bacterial infections may be underestimated due to the fact that fever is a very common complication in such patients or the used antibiotics mask the symptoms of this transfusion reaction. The capacity to inactivate those potentially contaminating bacteria in platelet concentrates is not in the focus of this article but briefly summarized in the sub-section ‘Inactivation Efficacy of Current Pathogen Inactivation Technologies’.
In addition, it is still impossible to know or reliably predict if and when emerging pathogens will threaten the safety of the blood supply. To this end, the question how to deal with the appearance of new or reemerging pathogens and how novel technological inventions can be used to further improve blood safety for patients in the European Union and elsewhere is the focus of an ongoing discussion [1, 2].
From a social point of view, it is desirable to pursue a zero-risk strategy to completely prevent transfusion-transmitted infections. It is well known from specific industrial sectors, such as aircraft technology and nuclear energy, that the implementation of programs for continuous quality improvement and risk management is essential to approximate the goal of ‘zero-risk’ [14, 15]. The impact of an unfortunate event and the probability of its occurrence are the most important determining factors for quantitative risk assessment and root cause analysis [16, 17]. For transfusion-transmitted infections, the most effective stage of hazard prevention is the elimination of the hazard (pathogen), followed by strategies to mitigate the risk of its transmission, e.g. by donor deferral, pathogen screening, or pathogen inactivation [2, 18, 19, 20].
This review focuses on the exemplified description of the epidemiology and surveillance of arbovirus infections and their potential risk for blood safety due to the fact of an asymptomatic viremic phase. To the author's knowledge NAT assays for dengue virus (DENV), chikungunya virus (CHIKV), and West-Nile virus (WNV) have been developed and are partly commercially available [21]. In this context, the invention, pre-commercial development, and introduction into the market of ‘universal’ pathogen inactivation technologies (PIT) are attractive steps towards the manufacturing of ‘sterile’ cellular blood components from human sources, which have been a global matter of course for cell-free solutions of human origin (albumin, immunoglobulins and coagulation factors) for decades [1, 22, 23, 24, 25].
These PIT imply a proactive, more generalized approach against multiple new and (re-)emerging pathogens, which perpetually challenge the safety of the blood supply and will become an serious alternative to a repetitive implementation of new screening tests (e.g. NAT) [21]. Therefore, this innovation is designated as ‘paradigm shift in transfusion medicine’ [25]. This review summarizes upcoming PIT with a focus on cellular blood components (platelets and RBCs) and highlights their current progress in clinical evaluations [26].
The Emergence of Pathogens
A pathogen emerges either by de novo mutations or by crossing the species barrier to human beings [2, 18]. HIV is an excellent example of a life-threatening emerging infectious disease. AIDS was first diagnosed in patients with Pneumocystis carinii pneumonia and Kaposi's sarcoma, which alerted the US Centers for Disease Control and Prevention to carefully monitor the outbreak of such a new disease for which mainly affected Haitians, homosexuals, hemophiliacs, and i.v. drug users. Two independent research groups (Robert Gallo and Luc Montagnier) published the discovery of a novel retrovirus as the causative agent in Science [27, 28]. It is now widely accepted that HIV originated from simian immunodeficiency virus(SIV)-infected chimpanzees and crossed the animal-human barrier. There, it mutated into HIV and spread throughout society by high-risk transmission channels (www.wikipedia.org). HIV antibody detection assays have been commercially available since the mid-1980s, and they have been implemented in blood donor screenings worldwide to prevent transmission via blood or coagulation factors of human origin [8].
Dengue Virus
The lessons learned with AIDS have led to the well-established concept of emerging infections, which are relevant to transfusion medicine. It is especially relevant for cases in which the donors are potentially infected (e.g. viremic), yet do not have symptoms, and transmission via blood transfusion is probable or proven. This is true for both DENV and CHIKV, which both are transmitted via mosquitos (Aedes aegypti and Aedes albopictus) [2, 29]. Dengue fever is an infectious tropical disease that is commonly self-limiting and is accompanied by flu-like symptoms and a skin rash that is similar to measles. However, in rare instances, life-threatening dengue hemorrhagic fever develops.
Since the 1960s, the incidence of DENV has increased dramatically due to urbanization, population growth, and global warming, with more than 50 to 100 million individuals infected annually in more than 100 endemic countries worldwide [30]. In these countries donor deferral is not an appropriate measure for prevention [2, 30]. Neither an approved vaccine nor a specific antiviral drug is currently available. In addition to the elimination of the mosquito or its habitats, protection from mosquito bites by wearing clothes is the only preventive measure. There are several outbreaks in non-endemic regions such as in Texas, Florida, and Australia [29]. Also in France (n = 2), Croatia (n = 15), and Madeira (n = 2,164), individuals who never left their home country suffer from dengue fever [21, 31]. Furthermore, there is evidence that one of the vectors, Aedes albopictus, now ‘habitats’ (nondengue virus-infected) in Germany, Austria, and Switzerland, stowing away from Italy by commercial transport [32].
DENV can be transmitted by blood transfusions and solid organ transplants in rare cases [30]. Two small outbreaks of transfusion-transmitted DENV in Hong Kong and Singapore have been published, and the incidence of DENV RNA in healthy blood donors was assessed, e.g. in Puerto Rico and Brazil [33, 34, 35, 36]. The rate of asymptomatic DENV RNA-positive individuals was estimated to be 1:250 to 1:1,000 during DENV epidemic outbreaks, and they serve as additional sources of virus dissemination in the community. In Germany, the Robert-Koch-Institut detected more than 600 individuals with dengue fever in 2012, all of whom acquired the infection while traveling (www.rki.de).
Chikungunya Virus
Large CHIKV outbreaks have taken place in the Indian Ocean, India, Southeast Asia, and Europe [2, 37]. CHIKV causes a febrile illness with painful and sometimes long-lasting (up to years) arthralgia. In particular, the outbreak in La Reunion (2005 and 2006) affected nearly one third of the island population (265,000 human cases and 237 deaths) [18, 38, 39, 40]. The collapse of the regional blood supply was prevented by the import of blood components from France and the urgent implementation of INTERCEPT™ as PIT for sufficient platelet support [41].
Air travelling and shipping around the world contributes to the spread of CHIKV and were recognized as a cause for the unexpected CHIKV outbreak in Northern Italy in the summer of 2007 [42, 43]). The outbreak was limited to 205 patients and was caused by an infectious traveler. Currently, a widespread human-mosquito-human transmission cycle vectored by Aedes albopictus is deemed to be unlikely in Europe or the USA because the virus does not persist during cold temperature, mosquito activity decreases in parallel with the virus, there is a well-established socioeconomic framework in place, and there is an international surveillance network [44].
West Nile Virus
The emergence of WNV in the USA can be traced back to first cases, when WNV was imported to New York in 1999 (66 human cases and 22 deaths) [45]. WNV is a mosquito-borne RNA virus whose primary hosts are birds. An epidemic was observed in 2002, with 4,156 individuals affected and a mortality rate of approximately 7% (n = 284) due to severe meningoencephalitis [18]. In 2002, 23 transfusion-transmitted WNV infections were detected, some of which were asymptomatic. In contrast, 6 cases were fatal [46]. Under the leadership of the US Food and Drug Administration (FDA), research organizations, blood donation agencies, and screening test manufacturers build up a task force for the development, industrial scaling-up, and validation of a WNV nucleic acid amplification technology [2]. In summer 2003, WNV NAT test systems with a minipool design were implemented, allowing the screening of viral RNA in a specimen pool of 6–24 donors [47]. Six breakthrough WNV infections have led to a novel concept that provides a seasonal switch to a more sensitive individual donation NAT platform [48, 49]. Occasionally, WNV infections are missed due to a viral titer below the lower limit of detection, as recently reported (Morbidity and Mortality Weekly Report August 9, 2013) by the Center of Disease Control and Prevention (www.cdc.gov) for an immunocompromised patient with non-Hodgkin‘s lymphoma who died by diffuse WNV encephalitis.
WNV epidemics occur every year in USA, one of the most severe outbreaks took place in 2012 with 2,734 neuroinvasive cases and 597 viremic donors which was comparable to WNV outbreaks in 2002/2003 (2,946/2,866 patients with neuroinvasive form and 714 viremic donors, WNV-NAT tested for the first time in 2003 [21]. A phylogeographic model was created based on WNV genome sequences of sampled organisms to reconstruct the spatiotemporal diffusion of WNV in North America [50]. It is speculated that bird movements may be responsible for rapid, long-distance movements (1,000 km/year) of the virus. In the European Union and neighboring countries (in addition to others that border the Mediterranean Sea and Eastern Europe, www.ECDC.europe.eu), an increasing number of autochthonous WNV infections have been documented. The European Network for Arthropod Vector Surveillance for Human Public Health (VBORNET) supported the European Center of Disease Control (ECDC) to take measures for prevention or control of local outbreaks with a panel of experts in the field of entomology (scientific study of insects) and transfusion medicine [21].
For example, 2010 is the third consecutive year that human cases of WNV infections have been identified in Northeastern Italy, with a trend towards a wider geographic area [51]. Therefore, preventive strategies are currently being discussed by the European Union and the Paul-Ehrlich-Institut in Germany to minimize the risk of WNV transmission via blood (and solid organs), such as the temporal deferral of at-risk donors or the implementation of a sensitive NAT screening test (www.pei.de).
Outlook
The emergence of novel pathogens is rather unpredictable, although mathematical models suggest that every 5 years a new transfusion-transmissible infectious agent will emerge [20]. Up-to-date epidemiological data are a prerequisite for proper risk assessment, especially for traveling blood donors who return from endemic areas to Europe [21, 133]. The control of epidemics is of utmost importance and is kept under precautionary surveillance by national and international bodies. Several emerging pathogens are likely transmissible by blood transfusion. Blood safety during the emergence of blood-borne pathogens is a major public health concern, and this is also true for known viruses with genomic mutations susceptible to immune escape mechanisms, new strains of bacteria, or other emerging pathogens or parasites that can spread by international travel or climate change (DENV, Plasmodium falciparum, Trypanosoma cruzi, Babesia necroti) [19, 52, 53]. The highly contagious severe acute respiratory syndrome (SARS) virus in 2002/2003, the H5N1 avian influenza virus (‘bird flu’) since 2003, and, more recently, the Middle East Respiratory Syndrome Coronavirus (MERS-CoV, H7N9) in 2012 are impressive examples of how rapidly pathogens can emerge and spread worldwide [54, 55, 56].
Procedures that address the risk assessment for such emerging infectious diseases that are potentially transmissible by blood and blood products were recently discussed in the USA [20]. Based on strong scientific evidence, policy makers have to carefully consider strategies for optimal risk minimization and for preventing a collapse in the national blood supply. Under- and overestimation of actual risk has undesirable and potentially disastrous consequences. In the absence of available blood screening tests, deferral of blood donors is likely the best alternative to mitigate the risk of dissemination and its amplitude [20]. However, in a case of very high incidence, this strategy implies the disadvantage of paralyzing, at least temporally, the blood supply.
In the future, universally applicable technologies for robust pathogen inactivation may become acceptable strategies to circumvent such risks. These technologies will also have the potential to replace not only routine gamma-irradiation, bacterial testing, and CMV serology but also to scrutinize the usefulness of other valuable test systems for the detection of viral antigens, antibodies, or nucleic acids.
Current Pathogen Inactivation Technologies
Historically, cell-free blood components, such as human plasma, were pathogen-inactivated with solvent-detergent (SD) for large plasma pools or by the addition of methylene blue (MB) for single plasma products in small-sized blood establishments [1, 22, 23, 24]. In general, pathogen inactivation resulted in reduced factor VIII activity and fibrinogen content of about 70–80% [57, 58]. More surprisingly, results obtained from randomized clinical trials (RCTs) comparing SD plasma / MB plasma with conventional FFP have not been published to a great extent [24]. The above mentioned methods are not transferable to cell-containing blood components as they heavily damage platelets and erythrocytes.
Therefore, this review will focus on novel PIT, which has become increasingly available for cellular blood components by circumventing these disastrous cell killing properties. Notably, INTERCEPT™ (Cerus Corporation, Concord, CA, USA), Mirasol® (Terumo BCT, Lakewood, CO, USA), and THERAFLEX® UV (Macopharma, Mouvaux, France) represent the currently available technological platforms, and they are each at different stages of marketing readiness. In contrast to the SD/MB method, these technologies all have the capacity to treat cellular blood components, whereas with SD/MB only the treatment of plasma is possible.
INTERCEPT technology is based on supplementation with synthetic psoralen S-59, which penetrates cellular and nuclear membranes and reversibly binds to nucleic acids, especially to pyrimidines in single- or double-stranded DNA and RNA [59]. After UV-A light exposure (320–400 nm, 3 J/cm2) it robustly cross-links nucleic acids in an oxygen-independent manner. Consequently, this action is independent of potential cytotoxic reactive oxygen species (ROS). Psoralens are naturally occurring substances found in limes and celery. The mode of action has been carefully investigated, especially the high frequency of covalent cross-links (one adduct is formed in about 1 of 83 bp), which inhibit DNA or RNA replication as well as transcription and repair mechanisms. The unbound S-59 and photoproducts are reduced by an adsorption device. The integrated container set for platelets and the UV-A illuminator are licensed for some time and enable ready-for-use application in the routine workflow of blood banks.
Mirasol technology depends on the addition of vitamin B2 (500 μmol/l riboflavin), resulting in a final concentration of approximately 50 μmol/l riboflavin, plus broad-spectrum UV light (280–400 nm, 6.2 J/ml corresponding to 5 J/cm2) [60]. Riboflavin and its photoproducts are found in natural products (such as foods) and in human blood, albeit at a much lower concentration than in a Mirasol-treated platelet product. Riboflavin serves as a photosensitizer (electron transfer) and promotes the oxidation of nucleic acids, especially guanine residues, without binding to nucleic acids or proteins. This leads to a conversion of riboflavin to lumichrome and other photoproducts. Riboflavin-induced damage is irreversible because both replication processes and RNA/DNA repair mechanisms are strongly inhibited. The frequency of nucleic acid lesions is approximately 1 in 350 bp. However, in context with plasma, a significant role for Mirasol-generated ROS was shown which adversely affected the molecular integrity of avin-labile coagulation factors (FVIII, fibrinogen) and other enzymes like ADAMTS13 [61]. The direct generation of superoxide anion and other ROS may make a reevaluation of the general toxicological assessment necessary.
THERAFLEX UV technology for platelets does not rely on a photodynamic agent and is based exclusively on the force of narrow-bandwidth UV-C light (254 nm, 0.2 J/cm2, irradiation time < 1 min), which induces the formation of pyrimidine dimers [62, 63, 64, 65]. To ensure optimal illumination, the platelets are transferred to a large illumination bag that allows for the homogeneous ‘penetration’ of thin platelet suspensions under frequent agitation (>100/min). Because the addition of a photoactive chemical is unnecessary, there is no need for conventional pharmacokinetic and toxicological assessments. General tolerability and lack of immunogenicity (neoantigen formation) of UV-C-treated platelets have been demonstrated in dogs [66]. In principle, THERAFLEX UV technology is also applicable to FFP. However, a higher UV-C intensity is needed to circumvent the quenching effect of proteins in 100% plasma and to achieve sufficient pathogen reduction rates [25].
Toxicology
The toxicology assessments of all discussed PIT applicable to platelets are summarized in table 1 (modified from [24]). Extensive testing using both in vitro and in vivo assays according to the standards of the International Conference of Harmonization for Drug Development is not suggestive of any toxicologically related safety concerns [60, 67, 68, 69]. Because of the compound absorption device which is integrated into the INTERCEPT Blood System the patient receives only about 1 μg amotosalen per platelet transfusion (LD50 in rats #1,000 mg/kg p.o.), whereas the final riboflavin exposure in adults is much higher (~5 mg) per transfusion [59, 60]. However, in consideration of the LD50 (in mice 50–100 mg/kg i.v.) the safety margin was calculated to be about 650–1,300-fold. Consequently, all above mentioned PIT for platelets have passed the licensing procedure (e.g., CE mark) to introduce each medical device into the market (table 2).
Table 1.
INTERCEPT platelets | Mirasol platelets | Theraflex platelets | ||
---|---|---|---|---|
Phototoxicity | ✓ | ✓*a | ||
Acute toxicology | ✓ | ✓ | ||
Repeated dose | ✓ | ✓ | ||
General pharmacology | ✓ | not applicableb | ||
Reproductive toxicology | ✓ | ✓ | photoreagent-free | |
Genotoxicity | ✓ | ✓ | no toxicological assessment necessaryc | |
Carcinogenicity | ✓ | not applicableb | ||
Neonatal toxicity | ✓ | ✓ | ||
ADME* studies | ✓ | ✓ | ||
Occupational safety | ✓ | ✓ | ||
Neoantigen formation | none | none | none |
Absorption, distribution, metabolism, and excretion.
No phototoxicity of photolyzed riboflavin and lumichrome was observed in acute and genotoxicity studies.
Riboflavin is a food additive and vitamin and a pharmacologic effect of residual riboflavin or its photoproducts is not intended by the manufacturer. In repeated dose toxicity studies as well as in genotoxicity studies there was no evidence of carcinogenicity.
The collateral impact of UVC light on the integrity of plasma proteins should be investigated
Table 2.
PIT | Company | Platelets | Plasma | RBC/whole blood |
---|---|---|---|---|
I. CE mark approval | ||||
S59 + UVA INTERCEPT | Cerus | CE class III | CE class III | not applicable |
May 2002 | November 2006 | |||
S-303 INTERCEPT | Cerus | not applicable | not applicable | not yet |
Riboflavin + UV Mirasol | TerumoBCT | CE class IIb | CE class IIb | not yet |
October 2007 | August 2008 | |||
Theraflex UV | Macopharma | CE class IIb | alternative technology* | not yet |
November 2008 | ||||
II. Clinical evaluation | ||||
S59 + UVA INTERCEPT | Cerus | phase III completed routine use | phase III completed routine use | not applicable |
S-303 INTERCEPT | Cerus | not applicable | not applicable | phase III initiated |
Riboflavin + UV Mirasol | TerumoBCT | phase III ongoing routine use | phase III completed routine use | phase I completed# |
Theraflex UV | Macopharma | phase I completed | alternative technology* | pre-clinical phase |
Based on metdylene blue and visible light.
For escalating UV light energies (22, 33 and 44 J/ml RBC).
Inactivation Efficacy
In addition to the well-known transfusion-transmitted viruses (HIV, HBV, HCV, HTLV) and bacteria, there is a considerable risk of transmitting other pathogens for which blood donations are not commonly tested (e.g., bacteria, Epstein-Barr virus (EBV), Parvo B19) and unknown pathogens that may emerge in the next few years, similar to the appearance of HIV in the early 1980s. It must be noted that the patient's individual burden may be much higher than the estimation of a ‘standard risk’ (e.g., 1:1 million) usually referring to a single blood transfusion may suggest. This is primarily caused by the transfusion frequency during the entire hospital stay or treatment period and is especially true for patients with chronic demand of blood, who require a considerable number of blood transfusions throughout their life (e.g., 100 transfusions corresponding to a drop down of the above mentioned ‘standard risk’ to 1:10,000).
With regard to platelet and plasma products, the pathogen inactivation profiles for viruses, bacteria, and protozoa are summarized in detail in table 3 for each PIT and updated by literature review and personal communication with the companies (Cerus Corporation: Larry Corash; Terumo BCT: Ray Goodrich, Macopharma: Frank Tolksdorf and German Red Cross Blood Service Springe: Axel Seltsam). [24, 59, 60, 62, 63, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79].
Table 3.
INTERCEPT | Mirasol | Theraflex | |
---|---|---|---|
Viruses (enveloped) | |||
HIV-1, cell free | >6.2 | 5.9 | 1.4 |
HIV-1, cell-associated | >6.1 | 4.5 | |
HBV | >5.5 | 2.5a | ≥2.8a |
HCV | >4.5b | >4.1 | >5.0 |
HEV (genotype 3) | >3.0 | ||
HTLV-I | 4.7 | n.t.c | |
HTLV-II | 5.1 | n.t.c | |
CMV, cell free | 2.1d | ongoing | |
CMV, cell-associated | >5.9 | ongoing | |
Bovine viral diarrhea virus | >6.0 | 2.7 | |
WNV | >6.0 | ≥5.1 | 3.5–4.0 |
Chikungunya | >6.4 | 2.2 | ongoing |
Influenza A virus | >5.9 | >5.0 | ≥5.3 |
SARS-CoV | >5.5 | ||
Togavirus | 3.2e | ≥5.3e | |
Rabiesvirus | >6.3f | ≥6.3f | |
Dengue-virus | >5.2 | ongoing | |
La Crosse virus | >3.3 | ||
Crimean-Congo hemorrhagic fever virus | 2.9 | ||
Viruses (non-enveloped) | |||
HAV | 0 | 1.8 | ongoing |
Parvo B19 | >6.2 | >5.0g | 5.0g |
Blue tongue virus | 6.1 | ||
Human adenovirus | >5.9 | ||
Calicivirus | 1.7–2.4 | ||
Picornavirus | 3.2h | >4.0h | |
Bacteria gram-negative | |||
Escherichia coli | >6.4 | 4.4 | >4.0 |
Enterobacter cloacae | 5.9 | >4.3 | |
Klebsiella pneumonia | >5.6 | 2.8 | 4.8 |
Pseudomonas aeruginosa | 4.5 | >4.5 | >4.9 |
Salmonella cholerasuis | >6.2 | ||
Serratia marsescens | >6.7 | 4.0 | >4.9 |
Yersinia enterocolitica | >5.9 | 3.3 | |
Brucella neotomae | 5.4 | ||
Bacteria gram-positive | |||
Bacillus cereus (incl. spores) | 3.6 | 4.3 | |
Bacillus cereus (vegetative) | >6.0 | 2.6 | 4.3 |
Bifidobacterium adolescentis | >6.5 | ||
Clostridium perfringens (vegetative) | >6.7 | >4.7 | |
Corynebacterium minutissimum | >6.3 | ||
Listeria monocytogenes | >6.3 | ||
Proprionobacterium acnes | >6.2 | >2.8 | 4.5 |
Staphylococcus aureus | 6.6 | 4.8 | >4.8 |
Staphylococus epidermidis | >6.6 | 4.6 | >4.9 |
Streptococcus pyogenes | >6.8 | 2.6 | |
Lactobacillus species | >6.9 | ||
Spirochaetes | |||
Borrelia burgdorferi | >6.8 | ||
Treponema pallidum | >6.8 | ongoingi | |
Parasites | |||
Babesia microti | >5.3 | >4.0 | |
Babesia divergens | >5.0 | ||
Leishmania major | >4.3 | >4.0 | |
Leishmania mexicana | >5.0 | ||
Orientia tsutsugamushi | >5.0 | ||
Plasmodium falciparum | >6.0 | >3.2 | >4.9 |
Plasmodium yoelli | >4.4j | ||
Trypanozoma cruzi | >5.3 | >5.0 | 2.8 to 4.2 |
Pseudorabies virus (suides herpes virus).
Chimpanzee transfusion studies.
HIV is used witd some restrictions as model virus also for other retroviruses such as the oncogenic HTLV-I/II.
Bovine rhinotrachitis virus, in addition mouse CMV in vivo mouse transfusion model.
Sindbis virus, HCV model.
Vesicular stomatitis virus.
Porcine parvovirus which suitability as model virus for human Parvo B19 remains questionable.
Porcine encephalomyocarditis virus, HAV model.
In rabbits.
In vivo mouse malaria model.
Especially the bacterial contamination of platelet concentrates (e.g., with Staphylococcus epidermidis and Bacillus cereus) and to less extent of RBCs (e.g., with Yersina enterocolitica and Serratia marcescans) still remains a significant challenge in transfusion medicine with severe impact on the patient's outcome including death [1]. The currently available PIT are able to prevent the majority of transfusion-transmitted bacterial infections. There are some limitations in respect to bacterial spores (e.g., from Bacillus cereus or Clostridium tetani) which are produced as survival strategy under extreme environmental conditions [134]. They are defined as a dormant, non-reproductive structure highly resistant to head, desiccation, freezing, UV irradiation as well as chemical and enzymatic destruction. Some but not all enter into vegetative forms in platelet concentrates as shown by spiking experiments [80]. Furthermore, high titers of bacteria under in vitro conditions are not always inactivated sufficiently (e.g., Klebsiella pneumoniae) [24]. Results obtained from a direct comparison of the Mirasol technology versus bacterial culture testing were in favor for the PIT in platelet concentrates with a low initial contamination level (<20 CFU per product) [72]. Nevertheless, this study has shown in parallel a limited capacity of bacteria elimination. Even though the bacterial contamination occurring during blood donation is believed to be very low, bacteria may start to proliferate rigorously during the entire storage period after an initial lag phase of at least 24–48 h [80]. Therefore, we have to take into account that the current PIT are applied immediately after donation (within the 1st day) so that an exponential bacterial outgrowth until the pathogen inactivation takes place is very unlikely and the limited capacity could be neglected in transfusion practice.
In some instances an animal model virus analogous to the human virus has been used which may be of relevance for drawing significant conclusions as recently shown for porcine Parvo B19. Using a real-time PCR inhibition assay, it could be demonstrated that Mirasol reduces intact human parvovirus B19 DNA in plasma 1.7 log compared to 5 log inhibition of porcine B19 in a tissue culture infectious dose assay [81]. Logarithmic reduction with the prefix ‘greater than’ indicates that a higher infectivity was not given in the in vitro test system applied. The author would like to point out that the virus strain, virus titration method (tissue culture infectious dose), and the calculation platform may differ among the competitors involved.
Therefore, direct side-to-side comparison studies using the same strain of a virus or bacterial species may be desirable to ensure a high degree of comparability and reproducibility. More recently the ISBT Working Party for Transfusion-Transmitted Infectious Diseases published a proof of principle experiment validating the quality, stability and suitability of four PEI bacterial references (Staphylococcus epidermidis, Streptococcus pyogenes, Klebsiella pneumoniae, Escherichia coli) in platelet concentrate low-titer spiking experiments which were performed in 14 laboratories around the world. The aims of this international study were successfully fulfilled [80]. To the author's knowledge the UVC-based Theraflex technology is until today the first PIT using the WHO reference bacteria panel for the generation of inactivation data [65]. In case of bacteria spike experiments with high titers one should be aware that robust pathogen killing in vitro may not affect the problem of endotoxin-induced septic shock in patients. So, from a practical point of view the detection of sterility after applying PIT on blood components with reasonable bacterial contamination (simulating the scenario of contamination during donation) is of more clinical relevance [72]. The pathogen inactivation profiles are increasingly complete for the INTERCEPT and Mirasol with its well-known gap for HAV; however, especially for THERAFLEX UV there are still some data missing, and some transfusion-associated viruses have been found to be relatively resistant to UV-C light exposure – this is primarily true for HIV. PIT for RBCs and whole blood is currently being developed for business success because the patient's individual advantage with regard to blood safety mainly depends on the likelihood (up to 100%) of receiving PIT-treated blood components permanently.
Clinical Evaluation and Hemovigilance Data (Platelets)
In vivo Recovery and Lifespan
The pharmaceutical potency of pathogen-inactivated platelets was principally investigated by radiolabeling studies (111In) that involved re-transfusing autologous platelets initially collected from volunteer donors after 5-day storage. The data were corrected for radiolabeling efficiency and pre-injection elution. The minimal FDA requirements for fresh platelets are a mean recovery rate of 65% and a life span of 8 days. The FDA requirements demand a platelet recovery at day 5 of at least 67% (corresponding to 43.5% of the initial concentration of platelets under fresh conditions) and platelet survival of 58% (corresponding to 4.64 days) compared with fresh reference platelets produced in parallel. In contrast to RBC survival in the blood circulation, it is well known that transfused platelets fill up the spleen pool.
In 2004, a mean recovery of 84% (42.5 ± 8.7% vs. 50.3 ± 7.7%) and a lifespan of 80% (4.8 ± 1.3 vs. 6.0 ± 1.2 days) were published for INTERCEPT-treated compared with non-irra-diated reference platelets suspended in PASIII [82]. Furthermore, AuBuchon et al. [83] have summarized the results obtained for Mirasol-treated platelets with a mean proportion of 75% (50.0 ± 18.9% vs. 66.5 ± 13.4% recovery) and 73% (4.3 ± 1.0 vs. 5.9 ± 1.1 days lifespan). Very recently, the first results (n = 6) of a phase I study for the THERAFLEX UV technology were published and demonstrated a significant decrease in the platelet recovery and survival rates (74% (37.6 ± 6.5% vs. 28.0 ± 8.2%) and 71% (7.3 ± 0.9 vs. 5.2 ± 1.3 days)), which were comparable to other PIT [84]. Furthermore, Thiele et al. [85] presented preliminary data that the transfusion of UV-C-treated autologous platelets in healthy volunteers met the safety and tolerance criteria.
Upon critical review of these data, the pathogen-inactivated platelets have fulfilled the minimal FDA criteria on in vivo quality independent of the technology applied. However, the raw data consistently indicate a slight but significant alteration of in vivo quality between the test and reference groups in each study. This in vivo phenomenon may be in line with in vitro parameters for assessing the functional integrity of platelets, such as the detection of a higher metabolic state or signs of activation [82, 83].
Clinical Evaluation
Depending on the CE approval mark which enables the free movement of goods in the EU member states and the completion of phase III RCTs, the readiness for market introduction varies for the above mentioned PIT (table 2). Medical devices with a higher risk profile are classified into a higher category. The INTERCEPT device is a CE mark class III product (amotosalen) which requires the critical review of clinical data (e.g., inactivation efficacy, 7-day storage) by the Notified Body to demonstrate compliance to the medical device directive and the approval by the respective National Authority. The Mirasol device is a CE mark class IIb product (riboflavin) which needs in principle a self-certification by the manufacturer. To the author's knowledge, the toxicology profile, inactivation efficacy, and clinical data of the Mirasol technology were also reviewed by a Notified Body prior to approval. The most important data of relevant clinical trials which are already published in peer-reviewed journals are summarized in table 4.
Table 4.
PIT | Reference | Study design | Study size (test/reference) | Platelet characteristics (dose in 1011, storage in days) | Primary endpoint (test/reference) | Secondary endpoints (test/reference) | |
---|---|---|---|---|---|---|---|
euroSPRITE Van Rhenen et al. 2003 [86] | INTERCEPT | buffy coat T-sol or plasma, all irradiated | multicenter RCT, ITT (56 days of treatment) 80% power to detect ΔCCI-1h −2,800 or ΔCI 8×109/1 (α=0.05, 2-sided) | 103 patients 52/51 567 products 311/256a | dose 3.9 ± 1.0 / 4.3 ± 1.2 storage 3.5 ± 1.1 / 3.4 ± 1.2 | CCI-1hb 13,100 ± 5,400/14,900 ± 6,200 1,800 ⇒ upper bound 95% CI 4,100 LRAc dose versus CI-1h Δ1.5 × 109/1 ⇒ upper bound 95% CI 6.1 × 109 | CCI-24hb 7,400 ± 5,500/10,600 ± 7,100 LRAc dose versus CI-24h, Δ2.6 × 109 any hemorrhagic event 79%/79% any severe hemorrhagic event 6%/6% post-transfusion hemostatic score: 0.28/0.30 acute transfusion reactions (6 h) 6%/5% platelet transfusion interval (days) 3.0/3.4 number of PC 7.5 ± 5.8 / 5.6 ± 5.9 number of RBC 9.3 ± 5.3 / 8.2 ± 6.3 RBC/day of PC support 0.41 ± 0.35 / 0.56 ± 0.68 |
SPRINT McCullough et al. 2004 [87] | INTERCEPT | apheresis 100% plasma all irradiated | multicenter RCT, ITT (28 days of treatment) non-inferiority design 90% power, α=0.05 Δgrade 2 bleedings (12.5%, 1-sided) Δgrade 3, 4 bleedings (7.0%, 1-sided) | 645 patients 318/327 4,719 products 2678/2041 | dose 3.7/4.0 storage 3.4/3.6 | grade 2 bleeding 58.5%/57.5% | grade 3 or 4 bleeding 4.1%/6.1% CCI-1h 11,100/16,000 CCI-24h 6,700/10,100 acute transfusion reactions (7 days) 3.0%/4.4% platelet transfusion interval (days) 1.9/2.4 number of PC 8.4/6.2 number of RBC 4.8/4.3 RBC/day of PC support 0.31/030 |
Janetzko et al. 2005 [88] | INTERCEPT | apheresis 100% plasma all irradiated | multicenter, RCT, ITT (28 days of treatment) 80% power to detect ΔCI 13×109/1 or ΔCCI-1h 5,250 linear regression analysis | 43 patients 22/21 218 products 103/115 | dose 4.1 ± 1.2/3.8 ± 0.4 storage 3.1 ± 1.0 / 3.2 ± 0.8 | CI-1hb 23,800 ± 18,500/31,200 ± 15,500 Δ7,400 ⇒ upper bound 95% CI 17,900 LRAc dose versus CI-1h Δ7.2 × 109/l | CCI-1h 11,600 ± 7,300/15,100 ± 6,400 CCI-24h 7,300 ± 6,200/10,400 ± 6,500 LRAc dose versus CI-24h, Δ7.4 × 109/l number of PC 4.7 ± 3.3 / 5.5 ± 4.7 platelet transfusion interval (days) 2.4 ± 1.0 / 2.8 ± 1.0 any hemorrhagic event: 64%/71% number of RBC 3.7 ± 3.8 / 5.5 ± 5.6 RBC/dat risk 0.4/0.4 acute transfusion reactions(6 h) 6%/5% |
HOVON Kerkhoffs et al. 2010 [89] | INTERCEPT | buffy coat T-sol or plasma | multicenter RCT ITT non-inferiority design non blindedf 90% power to detect 20% ΔCCI-1h (a 0.025, 1-sided) | 295 patientsg 85/99/(94)h | dose: n.d. storage: equally 4 days | CCI-1h (plasma) 11,400 ± 5,300/17,100 ± 7,300 | CCI-24h 7,900 ± 5,300/12,800 ± 7,800 (plasma) any bleeding patient 32%/19% (plasma) bleedings grade 2 7%/6% (plasma) bleedings grade 3 6%/1% (plasma) platelet transfusion interval (h) 61 ± 47 / 81 ± 47 acute transfusion reactions 7%/13% |
TESSI Lozano et al. 2011 [90] | INTERCEPT | buffy coat (86%) apheresis (14%) T-sol or SSP, partly irradiated | multicenter RCT ITT non-inferiority design 30% margin | 211 patientse 101/98 | dose: equally 4.2 storage: day 7 (80%) or day 6 | CCI-1h 8,200 ±5,400 / 9,400 ± 5,900 | CCI-24h 4,600 ± 3,500 / 6,500 ± 5,200 CI-24h 11,100 ± 8,900 / 15,200 ± 12,200 CI-1h 19,400 ± 13,400 / 21,600 ± 14,600 platelet transfusion interval (days) 2.2/2.3 post-transfusion hemostatic score 84%/77% post-transfusion bleeding (4 days) 23.8%/29.2% RBC transfusion (24 h) 23.8%/21.7% any adverse event (24 h) 31.4%/29.2% |
MIRACLE Cazenave et al. 2010 [108] | Mirasol | buffy coat (30%) apheresis (70%) 100% plasma | multicenter RCT, ITTf non-inferiority design 20% CCI-1h Δ2,940 | 110 patients 56/54 541 products 303/238i | dose: equally 5.4b. storage 2.8 ± 1.1 / 2.6 ± 1.1 | CCI-1hb 11,700 ±1,100 / 16,900 ± 1,100j Δ5,200 failed to show non-inferiority | CCI-24hb 6,700 ± 900/9,900 ± 900, Δ3,200 platelet transfusion interval (days) 2.3 ±1.9 / 2.7 ± 1.4 RBC/patient during treatment: 2.8 ± 1.7/2.6 ± 2.4 any bleedings 59%/43% grade 3-4 bleedings 10.7%/5.6% any adverse event 0.28 ± 0.19 / 0.23 ± 0.19 |
RCT = Randomized clinical trial; ITT=intent to treat study; LRA = longitudinal regression analysis.
Only on-protocol transfusions.
For up to the first 8 transfusions
For all transfusions
Caused by the detection of a significant period-by-treatment interaction the first period was validated primarily, however the power to reject non-inferiority was only 20%.
199 eligible for CCI-1h.
Open labeled.
278 eligible for CCI-1h.
Two references (plasma and T-Sol).
And 65 (18%) / 72 (23%) off-protocol transfusions.
SEM instead of SD.
INTERCEPT
For INTERCEPT-treated platelets, results obtained from several clinical trials enrolling more than 1,000 patients have already been published, and they demonstrate strong evidence for general therapeutic efficacy and safety. The five most relevant phase III studies are summarized in detail in table 4 (euroSPRITE [86], SPRINT [87], Janetzko et al. [88], Kerkhoffs et al. [89], and Lozano et al. [90]). A supplemental ‘pilot’ study [91] and ‘phase I/II’ study [92] were also published, but not listed in table 4 due to limitations in study design and performance status. Several previously discussed issues have been rather controversial, especially the clinical impact of lower CI/CCI values and the correctness of bleeding assessment according to the WHO or Common Terminology Criteria for Adverse Events (CTCAE) classifications.
The results obtained from the RCT published by Kerkhoffs et al. in 2010 (HOVON study) [89] have especially contributed to the confusion ([89, 93, 94]. The lack of blinding and the evaluations of bleeding complications have been the most important critical comments [95, 96, 97]. The observation of grade 2 bleedings below 10% in hematological patients transfused prophylactically is in sharp contrast to previous and recently published data (approximately 60%), making it hard to consider the results (especially bleedings) of this RCT. This must be taken into account when the authors reported a significant increase in all bleedings after the first platelet transfusion (INTERCEPT 32% vs. 19% plasma; p = 0.03) and more grade 3 bleedings (6% vs. 1%; p = 0.044). Interestingly, the authors very recently published data using a rigorous observation and adjudication method to assess bleedings more accurately and consistently [98]. In addition, one other group has developed a new bleeding severity measurement scale to assess bleedings in patients with chemotherapy-induced thrombocytopenia with a higher level of validity [97, 99].
The impact of the revised meta-analysis, which included the RCT of Lozano et al. [90] and excluded the single RCT using Mirasol technology [100] remains questionable [101] because the data published by Kerkhoffs are still part of the analysis. However, the author now discussed that ‘the risk of all bleeding complications is no longer increased’, whereas the assessment of ‘the risk of clinical significant bleeding complications’ is uncertain and strongly depends on the integration or exclusion of a single RCT. In this line, an additional meta-analysis (excluding the HOVON study [89]) did not find any significant differences in bleeding risks [94]. The re-analysis of the SPRINT data has confirmed the equivalency for the prevention of bleedings (grades 2, 3 and 4) and the number of transfused PC and RBC units [102]. In addition to the hemostatic effect, the SPRINT data were further evaluated to assess the adverse event profile. Acute transfusion reactions within 6 h were significantly lower in the INTERCEPT study arm compared to reference platelets suspended in plasma (3.0% vs. 4.4%) [103].
Currently, increasing evidence suggests the clinical non-inferiority of INTERCEPT-treated platelets compared to reference platelets. Post-marketing observational studies confirmed the results which have been obtained by phase III studies [104, 105]. Data from large retrospective studies have also supported the non-inferiority as assessed primarily by these RCTs and have demonstrated a subsequent stable consumption of platelet concentrates in comparable patient cohorts (annual utilization in the same hospital) after INTERCEPT implementation [100, 106, 107, 108]. Furthermore, there was no significant increase in RBC transfusions in recipients transfused with pathogen-inactivated platelets. Osselaer et al. [106, 107] have surveyed the adverse events associated with routine transfusion of INTERCEPT-treated platelet concentrates after implementation of a multicenter active hemovigilance program (Belgium, France and Spain) with 5,106 transfusions in the first period and 7,437 transfusions in the second. In these post-marketing surveillance studies, the majority of patients suffered either a hematological malignancy with chemotherapy / stem cell transplantation or underwent cardiovascular surgery or solid organ transplantation. Children and pregnant women were included to receive pathogen-inactivated platelets in a routine workflow. In the latter period, the authors reported an overall infrequent rate (0.7%) of acute transfusion reactions, mainly of mild severity, and only 5 adverse events were considered severe without a causal link to PIT. Furthermore, Cazenave et al. [100] summarized the data from the Etablissement Francais du Sang Alsace after implementation in 2006, comparing the clinical safety and tolerability of more than 13,000 INTERCEPT-treated PC in approximately 2,000 recipients with reference platelet concentrates suspended in 100% plasma or PASIII. The authors stated a significant reduction in acute transfusion reactions and no evidence of increased RBC consumption in patients receiving INTERCEPT-treated PC. In most of the participating centers, PIT was used to replace gamma-irradiation and anti-CMV serology.
Mirasol
The data volume and level of evidence for the therapeutic efficacy and safety of Mirasol pathogen inactivation technology is less pronounced. Not until 2010, a single multicenter, open-label RCT with non-inferiority design was published by the Etablissement Francais du Sang comparing Mirasol-treated platelet concentrates with conventional products (either apheresis or buffy coat-derived, both suspended in 100% plasma) in hematological patients with thrombocytopenia [109]. Based on the results of the TRAP trial [110], a non-inferiority margin of 20% (equivalent to a mean corrected count increment 1 hour post-transfusion (CCI-1h) difference of 2,940) was defined, and a statistical power of 80% was considered to be sufficient. Six centers enrolled a total of only 118 patients between 2005 and 2007. Four patients in each study arm did not receive any transfusion and were excluded from the intention-to-treat analysis. A total of 678 transfusions were given, 368 in the test group and 310 in the reference group, with a 17.7% and 23.2% frequency of off-protocol transfusions, respectively. The primary endpoint of the study was the CCH-1h on the first 8 transfusions within a 28-day treatment period. The RCT failed to show the hypothesized non-inferiority, with a CCI-1h of 11,725 ± 1,140 (test) versus 16,939 ± 1,149 (reference), resulting in a mean decrease of 5,200 and corresponding to approximately 30%. As secondary outcomes, the CCI24h showed a comparable trend with a mean difference of 3,200 (33%). The difference in the rate of all bleedings did not reach significance (59% vs. 43%; p = 0.127), and the frequency of grade 3 and 4 bleedings was too low (n = 6 (test) vs. n = 3 (reference); p = 0.490) to evaluate the clinical safety profile in terms of bleeding complications as this needs a sufficiently powered study design. Even the authors concluded that further clinical studies are required to determine whether the drop in CCI-1h correlates with an increased risk of bleedings.
So far, only small-sized observational platelet studies have been performed or are still ongoing in Spain, Luxembourg, Lithuania, and Serbia. Among others, Antic et al. [111] have shown no statistically significant difference between test and control group corresponding to a CCI-1h value of 7,500 and CCI-24h of 5,000. Yanez et al. [112] has published a CCI-1h of 12,000 and a CCI-24h of 4,900 based on 26 transfusions in 11 patients, and Coene et al. [113] has also shown comparable CCI-1h and CCI-24h values (9,900 ± 5,700 and 5,900 ± 2,900) calculated from a total of 21 transfusions in 8 patients. Although all authors stated that no adverse events were reported, the validity of this information was insufficient due to the small patient size. Furthermore, a similar hemostatic function of Mirasol-treated platelets was suggested by using thrombelastography as in vivo surrogate system [114]. These single observations did not supersede the need to implement an active hemovigilance program for Mirasol-treated platelet concentrates in routine use and to foster further RCTs. In Italy, one additional multicenter RCT with a calculated size of more than 400 patients (IPTAS) has been initiated using Mirasol-treated PC suspended in PAS to address the concern of clinical relevant bleedings. Furthermore, the PREPAReS study (consortium of The Netherlands, Norway, and Canada) is ongoing and has to recruit more than 600 patients receiving Mirasol-treated platelet concentrates suspended in 100% plasma. Both clinical studies will not be completed before 2015 (personal communication Ray Goodrich).
Outlook
Until 2014, only plasma and platelet concentrates derived from pooled buffy coats or collected by single-donor apheresis pathogen-inactivated using INTERCEPT or Mirasol had received marketing authorization approval by European and Non-European National Bodies, while THERAFLEX UV technology and PIT applied on RBC units or whole blood (see below) are still in the early phase of clinical evaluation. Most clinical experiences are available from more than 2 million post-marketing transfusions using INTERCEPT-treated plasma (ca. 40%) and platelets (ca. 60%) in more than 20 countries (personal communication Larry Corash), for which hemovigilance data have been collected and reviewed systematically as mentioned above. The clinical data also include experiences in neonates and pediatric patients. In addition, national hemovigilance data are also available from France (since 2006) and Switzerland (since 2011). Such data are not available from Germany, although the Paul-Ehrlich-Institut released the first manufacturing authorization in 2007 for the University Hospital Luebeck [105]. In the case of Mirasol, the company announces that over 110,000 Mirasol-treated blood components (ca. 60% plasma, ca. 40% platelets) were applied in participating transfusion centers most frequently in routine use without any severe adverse events or an obviously increased rate of adverse events (personal communication Ray Goodrich). However, systematically collected hemovigilance data post marketing authorization have not yet been published. None of the PIT is currently approved by the FDA for the US market. However, the Cerus Corporation has announced the submission of a premarket approval application for INTERCEPT platelets in the first half of 2014.
Taken together, the ‘inherent’ variability of conventional platelet concentrates (independent of the type of PIT used) must be noted which has been well-accepted over decades. This can be reasonably explained in terms of platelet content by the ‘biological source’ (e.g., volunteer donors with different platelet concentrations in their peripheral blood) and in terms of clinical efficacy by either the natural aging of platelets during storage or non ABO-identical use (e.g. TRAP trial). As reported by Slichter et al. [12] in a large randomized multicenter trial, the Prophylactic Platelet Dose Study (PLADO) with 5,466 prophylactic platelet transfusions in 1,272 patients, the platelet dose does correlate with the CCI after transfusion resulting in a higher transfusion frequency, but not necessarily with higher incidence of bleedings. With this in mind, we must ask to what extent lower efficacy (bearing in mind the non-inferiority margins of about 20%) becomes acceptable for a higher safety profile as a result of using PIT rather than to call efficacy and safety of PIT-treated platelets into question aiming at aspects that were accepted with regard to conventional platelet products in the past (e.g., impaired by storage and ABO non-identical use). Furthermore, the very recently published study by Wandt et al. [13] invites to rethink the prophylactic platelet transfusion strategy in some patients not at risk for increased bleeding complications (e.g. autologous stem cell transplantation) because no increase of major hemorrhages could be observed in these patients when platelets have been transfused only therapeutically. However, quality-of-life aspects remain questionable because most of the patients and physicians are seriously affected by routine monitoring of bleedings as they became aware of a permanent latent danger like a sword of Damocles.
Outlook for Red Blood Cells and Whole Blood
INTERCEPT
The PIT applied on platelets is not transferable to whole blood and RBC units because these optically dense blood components need very high doses of UV light. Therefore, Cerus has developed a novel platform technology (2nd generation) that is based on the addition of the chemical compound S-303 (0.2 mmol/l) and glutathione (GSH; 20 mmol/l) as a quencher [115]. S-303 targets and cross-links nucleic acids via a bis-alkylating group to prevent further replication. After the reaction, the non-reactive, negatively charged breakdown product S-300 is formed and rapidly degraded. To minimize the affinity of S-303 with other nucleophiles, especially proteins, glutathione is added and distributed in the extracellular compartment (plasma) to quench these extracellular reactions, whereas S-303 acts as a ‘pathogen inactivator’ in the intracellular compartment. Treatment solution and breakdown products are removed by centrifugation prior to final storage in approved solutions [116].
During a 1st generation S-303 phase III RCT for chronic anemia patients, two multitransfused participants (of more than 140) developed a low-titer antibody against the acridine moiety of S-303 on the RBC surface that led to positive crossmatch reactions in vitro without any cues regarding its clinical impact, such as acute hemolytic transfusion reactions [117]. Nevertheless, this phase III study and a parallel study in cardiovascular surgery patients were halted prematurely, and Cerus developed a 2nd generation S-303 technology to minimize the acridine moieties on the RBC surface by increasing the concentration of the protecting glutathione during pathogen inactivation process. Subsequently, the inactivation efficacy against bacteria, viruses, and protozoa of the 2nd generation approach as well as the pharmacokinetic and toxicological assessment of S-303 is principally transferable from the 1st generation (tables 5, 6) [115, 118, 119] (personal communication Larry Corash). This cross-reference was accepted by the FDA because of the identity of substances used. A new phase I study was conducted with a total of 26 healthy volunteers receiving autologous S-303-treated RBC transfusions using a dual radiolabeling approach using 51Cr and 99Tc [116]. The 2nd generation of S-303 did not induce antibody formation. The 24-hour recovery at day 35 was excellent, with a mean of 88.0 ± 8.5%, and demonstrated equivalency with the untreated RBC reference arm (90.0 ± 6.9% recovery). The median lifespan was significantly reduced (32.8 vs. 39.5 days), whereas the in vitro evaluation after 35 days of storage has shown equivalency in hemolysis, extracellular potassium, mean corpuscular hemoglobin concentration, and mean corpuscular volume and a marginal loss of hemoglobin during S-303 treatment.
Table 5.
INTERCEPT S-303 RBC | Mirasol whole blood | |
---|---|---|
Phototoxicity | ✓ | ✓ |
Acute toxicology | ✓ | ✓ |
Repeated dose | ✓ | ✓ |
General pharmacology | ✓ | ✓ |
Reproductive toxicology | ✓ | ✓ |
Genotoxicity | ✓ | ✓ |
Carcinogenicity | ✓ | ✓ |
Neonatal toxicity | ✓ | ✓ |
ADME* studies | V | ✓ |
Occupational safety | ✓ | ✓ |
Neoantigen formation | open | ✓ |
Absorption, distribution, metabolism, elimination.
Table 6.
INTERCEPT | Mirasol | ||||
---|---|---|---|---|---|
Viruses (enveloped) | |||||
HIV-1, cell-associated | >6.2 | 4.5 | |||
HIV-1, cell free | >6.5 | ||||
HBV | >6.3a | ongoingb | |||
HCV | ongoing | ||||
HTLV-I | |||||
HTLV-II | |||||
CMV, cell free | |||||
CMV, cell associated | ongoing | ||||
BVDVc | >4.8 | ||||
WNV | |||||
Herpes simplex virus | >6.0 | ||||
Vesicular stomatitis virus | 5.7 | >4.5 | |||
Chikungunya | |||||
Influenza A virus | |||||
SARS-CoV | Dengue-virus | ||||
Viruses (non-enveloped) | |||||
HAV | 1.7 | ||||
Parvo B19 | 4.0d | ||||
Bluetongue virus | >6.0 | 1.5 | |||
Adenovirus type 5 | >7.4 | ||||
Bacteria gram-negative | |||||
Escherichia coli | 7.4 | ||||
Enterobacter cloacae | |||||
Klebsiella pneumonia | |||||
Pseudomonas aeruginosa | 4.5 | ||||
Salmonella cholerasuis | 4.8 | ||||
Serratia marsescens | 4.1 | ||||
Yersinia enterocolitica | 7.4 | ||||
Bacteria gram-positive | |||||
Bacillus cereus (incl. spores) | |||||
Bacillus subtilis (vegetative) | |||||
Bifidobacterium adolescentis | |||||
Clostridium perfringens (vegetative) | |||||
Corynebacterium minutissimum | >6.3 | ||||
Listeria monocytogenes | |||||
Proprionobacterium acnes | >7.1 | ||||
Staphylococcus aureus | >5.1 | ||||
Staphylococus epidermidis | |||||
Streptococcus pyogenes | >6.9 | ||||
Spirochaetes | |||||
Borrelia burgdorferi | |||||
Treponema pallidum | |||||
Parasites | |||||
Babesia microti | >5.5 | >5.0 | |||
Babesia divergens | >7.2 | ||||
Leishmania major | 2.3 | ||||
Plasmodium falciparum | >6.8 | >6.4 | |||
Trypanozoma cruzi | >5.4 | >3.5 |
Duck hepatitis B model.
Human.
Bovine viral diarrhea virus (HCV model).
Porcine parvovirus which suitability for human Parvo B19 remains questionable.
The S-303 PIT for RBCs is now ready to run in renewed, middle-sized (n < 100 patients) phase III clinical studies in the European Union, in which the patient's enrollment just began for elective cardiac surgery patients with acute blood loss and for thalassemia patients with chronic transfusion demands (personal communication Larry Corash). These clinical studies focus both on the hemoglobin content and in the latter case on the potential immunogenicity with repeat exposure. Furthermore, the company has announced a Cerus whole blood initiative supported by the Swiss Red Cross Humanitarian Fund to pursue the applicability of the S-303 technology on whole blood in developing countries as a humanitarian effort.
Mirasol
The same riboflavin-based PIT which was successfully evaluated for platelets and plasma is now being further developed for whole blood to achieve the ultimate goal of a single PIT platform that is able to ‘inactivate’ all three blood components simultaneously. Understandably, most of the toxicological assessment and data concerning neoantigenicity and protein binding behavior could be extrapolated to the whole blood system as well [120]. As shown in a baboon study model, riboflavin does not induce a chemical modification on the surface of RBCs such as possessing an immunogenic moiety [121]. Safety and efficacy was assessed in a large animal model (a diffuse, non-surgical bleeding pig model) [122]. The ‘drug-related’ terminology of phase I–III studies is avoided by the fact that the Mirasol system is rather a medical device than a drug (personal communication Ray Goodrich). However, in context with the generation of ROS this classification has to be reconsidered.
Feasibility data from RBCs (after 42 days of storage) separated from whole blood donations (n = 11) and treated by riboflavin and increasing doses of UV light energy (in this case 22, 33 and 44 J/ml) in the USA have shown high variability (ca. 60–80%) in RBC recovery and RBC half-life on day 42 (24 ± 9 days, range 10–36 days) [123]. Five of 11 subjects met the FDA criteria of treatment success as defined by 75% autologous recovery of radiolabeled RBCs after 24 h (lower limit of 95% CI: 70% recovery), which is, however, in the range of gamma-irradiated RBC units. The rate of hemolysis on day 42 (not day 35) is between 1.0 and 1.5%, and the osmotic fragility increases from 0.8 to 4.1% depending on the UV light dose chosen [123]. As recently reported by the company, the current configuration is now based on the exposure of a higher dose of UV light (80 J/ml) to compensate sufficiently for the UV light absorption by hemoglobin [60]. By choosing comparable UV energy doses, the quality of each separated blood component (e.g. rate of hemolysis) has to be maintained during standard storage period, while there is still an acceptable inactivation efficacy of a broad spectrum of pathogens as summarized in table 6. However some of these data are personally communicated and not yet confirmed by original articles in peer-reviewed journals [120, 124, 125, 126, 127, 128, 129, 130]. Riboflavin UV treatment and gamma-irradiation were equally effective in the prevention of transfusion-associated graft-ver-sus-host-disease, resulting in an in vitro 4.7 log reduction of viable T cells which had been subsequently confirmed in immunodeficient murine recipients (NOD-scid IL2rγnull) [131].
The company starts the IMPROVE II RCT (comparable to a phase II study with radiolabeled RBCs) in the USA, obviously to validate the impact of the process optimization and UV light dose fixation (80 J/ml) of Mirasol treatment on the RBC quality and loss of inactivation efficacy. In future, one of the focuses will be the potential clinical application of Mirasol-treated whole blood in military scenarios that allow for the immediate substitution of safe RBCs, platelets, and plasma for cases of life-threatening trauma and trauma-induced coagulopathy (IMPACT). Very interestingly, an additional RCT will take place in Ghana with more than 200 patients to evaluate the prevention of whole blood-associated malaria transmission by Mirasol-based inactivation of whole blood donations [132] (personal communication Ray Goodrich).
Disclosure Statement
PS has received honorarium and travel reimbursement as speaker and chair from CERUS Europe BV.
References
- 1.Lindholm PF, Annen K, Ramsey G. Approaches to minimize infection risk in blood banking and transfusion practice. Infect Disord Drug Targets. 2011;11:45–56. doi: 10.2174/187152611794407746. [DOI] [PubMed] [Google Scholar]
- 2.Dodd RY. Emerging pathogens and their implications for the blood supply and transfusion transmitted infections. Br J Hematol. 2012;159:135–142. doi: 10.1111/bjh.12031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Spahn DR, Goodnough LT. Alternatives to blood transfusion. Lancet. 2013;381:1855–1865. doi: 10.1016/S0140-6736(13)60808-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Engelbrecht S, Wood EM, Cole-Sinclair MF. Clinical transfusion practice update: haemovigilance, complications, patient blood management and national standards. Med J Aust. 2013;199:397–401. doi: 10.5694/mja13.10070. [DOI] [PubMed] [Google Scholar]
- 5.Hourfar MK, Jork C, Schottstedt V, Weber-Schehl M, Brixner V, Busch MP, Geusendam G, Gubbe K, Mahnhardt C, Mayr-Wohlfart U, Pichl L, Roth WK, Schmidt M, Seifried E, Wright DJ German Red Cross NAT Study Group. Experience of German Red Cross blood donor services with nucleic acid testing: results of screening more than 30 million blood donations for human immunodeficiency virus-1, hepatitis C virus, and hepatitis B virus. Transfusion. 2008;48:1558–1566. doi: 10.1111/j.1537-2995.2008.01718.x. [DOI] [PubMed] [Google Scholar]
- 6.Nübling CM, Heiden M, Chudy M, Kress J, Seitz R, Keller-Stanislawski B, Funk MB. Experience of mandatory nucleic acid test (NAT) screening across all blood organizations in Germany: NAT yield versus breakthrough transmissions. Transfusion. 2009;49:1850–1859. doi: 10.1111/j.1537-2995.2009.02212.x. [DOI] [PubMed] [Google Scholar]
- 7.Morel P. Ten years of nucleic acid testing: lessons and prospects (in French) Transfus Clin Biol. 2011;18:133–139. doi: 10.1016/j.tracli.2011.01.007. [DOI] [PubMed] [Google Scholar]
- 8.Roth WK, Busch MP, Schuller A, Ismay S, Cheng A, Seed CR, Jungbauer C, Minsk PM, Sondag-Thull D, Wendel S, Levi JE, Fearon M, Delage G, Xie Y, Jukic I, Turek P, Ullum H, Tefanova V, Tilk M, Reimal R, Castren J, Naukkarinen M, Assal A, Jork C, Hourfar MK, Michel P, Offergeld R, Pichl L, Schmidt M, Schottstedt V, Seifried E, Wagner F, Weber-Schehl M, Politis C, Lin CK, Tsoi WC, O'Riordan J, Gottreich A, Shinar E, Yahalom V, Velati C, Satake M, Sanad N, Sisene I, Bon AH, Koppelmann M, Flanagan P, Flesland O, Brojer E, Ltowska M, Nascimento F, Zhiburt E, Chua SS, Teo D, Stezinar SL, Vermeulen M, Reddy R, Park Q, Castro E, Eiras A, Gonzales Fraile I, Torres P, Ekermo B, Niederhauser C, Chen H, Oota S, Brant LJ, Eglin R, Jarvis L, Mohabir L, Brodsky J, Foster G, Jennings C, Notari E, Stramer S, Kessler D, Hillyer C, Kamel H, Katz L, Taylor C, Panzer S, Reesink HW. International survey on NAT testing of blood donations: expanding implementation and yield from 1999 to 2009. Vox Sang. 2012;102:82–90. doi: 10.1111/j.1423-0410.2011.01506.x. [DOI] [PubMed] [Google Scholar]
- 9.Schrezenmeier H, Walther-Wenke G, Müller TH, Weinauer F, Younis A, Holland-Letz T, Geis G, Asmus J, Bauerfeind U, Burkhart J, Deitenbeck R, Förstemann E, Gebauer W, Höchsmann B, Karakassopoulos A, Liebscher UM, Sänger W, Schmidt M, Schunter F, Sireis W, Seifried E. Bacterial contamination of platelet concentrates: results of a prospective multicenter study comparing pooled whole blood-derived platelets and apheresis platelets. Transfusion. 2007;47:644–652. doi: 10.1111/j.1537-2995.2007.01166.x. [DOI] [PubMed] [Google Scholar]
- 10.Funk MB, Lohmann A, Guenay S, Henseler O, Heiden M, Hanschmann KM, Keller-Stanislawski B. Transfusion-transmitted bacterial infections – haemovigilance data of German blood establishments (1997–2010) Transfus Med Hemother. 2011;38:266–271. doi: 10.1159/000330372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Rebulla P, Finazzi G, Marangoni F, Avvisati G, Gugliotta L, Tognoni G, Barbui T, Mandelli F, Sirchia G. The threshold for prophylactic platelet transfusions in adults with acute myeloid leukemia. Gruppo Italiano Malattie Ematologiche Maligne dell'Adulto. N Engl J Med. 1997;337:1870–1875. doi: 10.1056/NEJM199712253372602. [DOI] [PubMed] [Google Scholar]
- 12.Slichter SJ, Kaufman RM, Assmann SF, McCullough J, Triulzi DJ, Strauss RG, Gernsheimer TB, Ness PM, Brecher ME, Josephson CD, Konkle BA, Woodson RD, Ortel TL, Hillyer CD, Skerrett DL, McCrae KR, Sloan SR, Uhl L, George JN, Aquino VM, Manno CS, McFarland JG, Hess JR, Leissinger C, Granger S. Dose of prophylactic platelet transfusions and prevention of hemorrhage. N Engl J Med. 2010;362:600–613. doi: 10.1056/NEJMoa0904084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wandt H, Schaefer-Eckart K, Wendelin K, Pilz B, Wilhelm M, Thalheimer M, Mahlknecht U, Ho A, Schaich M, Kramer M, Kaufmann M, Leimer L, Schwerdtfeger R, Conradi R, Dölken G, Klenner A, Hänel M, Herbst R, Junghanss C, Ehninger G Study Alliance Leukemia. Therapeutic platelet transfusion versus routine prophylactic transfusion in patients with haematological malignancies: an open-label, multicentre, randomised study. Lancet. 2012;380:1309–1316. doi: 10.1016/S0140-6736(12)60689-8. [DOI] [PubMed] [Google Scholar]
- 14.Myhre BA, McRuer D. Human error-a significant cause of transfusion mortality. Transfusion. 2000;40:879–885. doi: 10.1046/j.1537-2995.2000.40070879.x. [DOI] [PubMed] [Google Scholar]
- 15.Müller MH. Increasing safety by implementing optimized team interaction – experiences of the aviation industry. In: Scharf RE, editor. Progress and Challenges in Transfusion Medicine, Hemostasis and Hemotherapy. Karger: Freiburg; 2008. pp. 1–14. [Google Scholar]
- 16.Leape LL. Errors in medicine. Clin Chim Acta. 2009;404:2–5. doi: 10.1016/j.cca.2009.03.020. [DOI] [PubMed] [Google Scholar]
- 17.Elhence P, Veena S, Sharma RK, Chaudhary RK. Root cause analysis of transfusion error: identifying causes to implement changes. Transfusion. 2010;50:2772–2777. doi: 10.1111/j.1537-2995.2010.02943.x. [DOI] [PubMed] [Google Scholar]
- 18.Dunstan RA, Seed CR, Keller AJ. Emerging viral threats to the Australian blood supply. Aust N Z Public Health. 2008;32:354–360. doi: 10.1111/j.1753-6405.2008.00254.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fournier-Wirth C, Jaffrezic-Renault N, Coste J. Detection of blood-transmissible agents: can screening be miniaturized? Transfusion. 2010;50:2032–2045. doi: 10.1111/j.1537-2995.2010.02678.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gallagher LM, Ganz PR, Yang H, Kessler DA, O'Brien SF, Custer BS, Busch MP, Dodd RY, Stramer SL, Walderhaug MO, Forshee RA, Williams AE, Epstein JS, Anderson SA. Advancing risk assessment for emerging infectious diseases for blood and blood products: proceedings of a public workshop. Transfusion. 2013;53:455–463. doi: 10.1111/j.1537-2995.2012.03808.x. [DOI] [PubMed] [Google Scholar]
- 21.Paty MC. The expansion of vector-borne diseases and the implications for blood transfusion safety: the case of West Nile Virus, dengue and chikungunya (in French) Transfus Clin Biol. 2013;20:165–173. doi: 10.1016/j.tracli.2013.03.008. [DOI] [PubMed] [Google Scholar]
- 22.Bryant BJ, Klein HG. Pathogen inactivation: the definitive safeguard for the blood supply. Arch Pathol Lab Med. 2007;131:719–733. doi: 10.5858/2007-131-719-PITDSF. [DOI] [PubMed] [Google Scholar]
- 23.Klein HG, Anderson D, Bernardi MJ, Cable R, Carey W, Hoch JS, Robitaille N, Sivilotti ML, Smaill F. Pathogen inactivation: making decisions about new technologies. Report of a consensus conference. Transfusion. 2007;47:2338–2347. doi: 10.1111/j.1537-2995.2007.01512.x. [DOI] [PubMed] [Google Scholar]
- 24.Prowse CV. Component pathogen inactivation: a critical review. Vox Sang. 2013;104:183–199. doi: 10.1111/j.1423-0410.2012.01662.x. [DOI] [PubMed] [Google Scholar]
- 25.Seltsam A, Müller TH. Update on the use of pathogen-reduced human plasma and platelet concentrates. Br J Hematol. 2013;162:442–454. doi: 10.1111/bjh.12403. [DOI] [PubMed] [Google Scholar]
- 26.Kleinman S, Reed W, Stassinopoulos A. A patient-oriented risk-benefit analysis of pathogen-inactivated blood components: application to apheresis platelets in the United States. Transfusion. 2013;53:1603–1618. doi: 10.1111/j.1537-2995.2012.03928.x. [DOI] [PubMed] [Google Scholar]
- 27.Gallo RC, Sarin PS, Gelmann EP, Robert-Guroff M, Richardson E, Kalyanaraman VS, Mann D, Sidhu GD, Stahl RE, Zolla-Pazner S, Leibowitch J, Popovic M. Isolation of human T-cell leukemia virus in acquired immune deficiency syndrome (AIDS) Science. 1983;220:865–8657. doi: 10.1126/science.6601823. [DOI] [PubMed] [Google Scholar]
- 28.Barré-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, Gruest J, Dauguet C, Axler-Blin C, Vézinet-Brun F, Rouzioux C, Rozenbaum W, Montagnier L. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS) Science. 1983;220:868–671. doi: 10.1126/science.6189183. [DOI] [PubMed] [Google Scholar]
- 29.Lanteri MC, Busch MP. Dengue in the context of ‘safe blood’ and global epidemiology: to screen or not to screen? Transfusion. 2012;52:1634–1639. doi: 10.1111/j.1537-2995.2012.03747.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Teo D, Ng LC, Lam S. Is dengue a threat to the blood supply? Transfus Med. 2009;19:66–77. doi: 10.1111/j.1365-3148.2009.00916.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Rogers DJ, Suk JE, Semenza JC. Using global maps to predict the risk of dengue in Europe. Acta Trop. 2014;129:1–14. doi: 10.1016/j.actatropica.2013.08.008. [DOI] [PubMed] [Google Scholar]
- 32.Kampen H, Kronefeld M, Zielke D, Werner D. Further specimens of the Asian tiger mosquito Aedes albopictus (Diptera, Culicidae) trapped in southwest Germany. Parasitol Res. 2013;112:905–907. doi: 10.1007/s00436-012-3128-y. [DOI] [PubMed] [Google Scholar]
- 33.Chuang V, Wong TY, Leung YH, Ma E, Law YL, Tsang O, Chan KM, Tsang I, Que TL, Yung R, Liu SH. Review of dengue fever cases in Hong Kong during 1998 to 2005. Hong Kong Med J. 2008;14:170–177. [PubMed] [Google Scholar]
- 34.Tambyah PA, Koay ES, Poon ML, Lin RV, Ong BK Transfusion-Transmitted Dengue Infection Study Group. Dengue hemorrhagic fever transmitted by blood transfusion. N Engl J Med. 2008;359:1526–1527. doi: 10.1056/NEJMc0708673. [DOI] [PubMed] [Google Scholar]
- 35.Dias LL, Amarilla AA, Poloni TR, Covas DT, Aquino VH, Figueiredo LT. Detection of dengue virus in sera of Brazilian blood donors. Transfusion. 2012;52:1667–1671. doi: 10.1111/j.1537-2995.2012.03729.x. [DOI] [PubMed] [Google Scholar]
- 36.Petersen LR, Tomashek KM, Biggerstaff BJ. Estimated prevalence of dengue viremia in Puerto Rican blood donations, 1995 through 2010. Transfusion. 2012;52:1647–1651. doi: 10.1111/j.1537-2995.2011.03529.x. [DOI] [PubMed] [Google Scholar]
- 37.Fischer D, Thomas SM, Suk JE, Sudre B, Hess A, Tjaden NB, Beierkuhnlein C, Semenza JC. Climate change effects on Chikungunya transmission in Europe: geospatial analysis of vector's climatic suitability and virus’ temperature requirements. Int J Health Geogr. 2013;12:51. doi: 10.1186/1476-072X-12-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Charrel RN, de Lamballerie X, Raoult D. Chikungunya outbreaks – the globalization of vectorborne diseases. N Engl J Med. 2007;356:769–771. doi: 10.1056/NEJMp078013. [DOI] [PubMed] [Google Scholar]
- 39.Renault P, Solet JL, Sissoko D, Balleydier E, Larrieu S, Filleul L, Lassalle C, Thiria J, Rachou E, de Valk H, Ilef D, Ledrans M, Quatresous I, Quenel P, Pierre V. A major epidemic of chikungunya virus infection on Reunion Island, France, 2005–2006. Am J Trop Med Hyg. 2007;77:727–731. [PubMed] [Google Scholar]
- 40.Tsetsarkin KA, Vanlandingham DL, McGee CE, Higgs S. A single mutation in chikungunya virus affects vector specificity and epidemic potential. PLoS Pathog. 2007;3:201. doi: 10.1371/journal.ppat.0030201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rasonglès P, Angelini-Tibert MF, Simon P, Currie C, Isola H, Kientz D, Slaedts M, Jacquet M, Sundin D, Lin L, Corash L, Cazenave JP. Transfusion of platelet components prepared with photochemical pathogen inactivation treatment during a Chikungunya virus epidemic in Ile de La Réunion. Transfusion. 2009;49:1083–1091. doi: 10.1111/j.1537-2995.2009.02111.x. [DOI] [PubMed] [Google Scholar]
- 42.Liumbruno GM, Calteri D, Petropulacos K, Mattivi A, Po C, Macini P, Tomasini I, Zucchelli P, Silvestri AR, Sambri V, Pupella S, Catalano L, Piccinini V, Calizzani G, Grazzini G. The chikungunya epidemic in Italy and its repercussion on the blood system. Blood Transfus. 2008;6:199–210. doi: 10.2450/2008.0016-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Rezza G, Nicoletti L, Angelini R, Romi R, Finarelli AC, Panning M, Cordioli P, Fortuna C, Boros S, Magurano F, Silvi G, Angelini P, Dottori M, Ciufolini MG, Majori GC, Cassone A CHIKV study group. Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet. 2007;370:1840–1846. doi: 10.1016/S0140-6736(07)61779-6. [DOI] [PubMed] [Google Scholar]
- 44.Petersen LR, Stramer SL, Powers AM. Chikungunya virus: possible impact on transfusion medicine. Transfus Med Rev. 2010;24:15–21. doi: 10.1016/j.tmrv.2009.09.002. [DOI] [PubMed] [Google Scholar]
- 45.Nash D, Mostashari F, Fine A, Miller J, O'Leary D, Murray K, Huang A, Rosenberg A, Greenberg A, Sherman M, Wong S, Layton M 1999 West Nile Outbreak Response Working Group. The outbreak of West Nile virus infection in the New York City area in 1999. N Engl J Med. 2001;344:1807–1814. doi: 10.1056/NEJM200106143442401. [DOI] [PubMed] [Google Scholar]
- 46.Pealer LN, Marfin AA, Petersen LR, Lanciotti RS, Page PL, Stramer SL, Stobierski MG, Signs K, Newman B, Kapoor H, Goodman JL, Chamberland ME West Nile Virus Transmission Investigation Team. Transmission of West Nile virus through blood transfusion in the United States in 2002. N Engl J Med. 2003;349:1236–1245. doi: 10.1056/NEJMoa030969. [DOI] [PubMed] [Google Scholar]
- 47.Lai L, Lee TH, Tobler L, Wen L, Shi P, Alexander J, Ewing H, Busch M. Relative distribution of West Nile virus RNA in blood compartments: implications for blood donor nucleic acid amplification technology screening. Transfusion. 2012;52:2012, 447–454. doi: 10.1111/j.1537-2995.2011.03289.x. [DOI] [PubMed] [Google Scholar]
- 48.Montgomery SP, Brown JA, Kuehnert M, Smith TL, Crall N, Lanciotti RS, Macedo de Oliveira A, Boo T, Marfin AA 2003 West Nile Virus Transfusion-Associated Transmission Investigation Team. Transfusion-associated transmission of West Nile virus, United States 2003 through 2005. Transfusion. 2006;46:2038–2046. doi: 10.1111/j.1537-2995.2006.01030.x. [DOI] [PubMed] [Google Scholar]
- 49.Francis RO, Strauss D, Williams JD, Whaley S, Shaz BH. West Nile virus infection in blood donors in the New York City area during the 2010 seasonal epidemic. Transfusion. 2012;52:2664–2670. doi: 10.1111/j.1537-2995.2012.03639.x. [DOI] [PubMed] [Google Scholar]
- 50.Pybus OG, Suchard MA, Lemey P, Bernardin FJ, Rambaut A, Crawford FW, Gray RR, Arinamin-pathy N, Stramer SL, Busch MP, Delwart EL. Unifying the spatial epidemiology and molecular evolution of emerging epidemics. Proc Natl Acad Sci U S A. 2012;109:15066–15071. doi: 10.1073/pnas.1206598109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Barzon L, Pacenti M, Cusinato R, Cattai M, Franchin E, Pagni S, Martello T, Bressan S, Squar-zon L, Cattelan A, Pellizzer G, Scotton P, Beltrame A, Gobbi F, Bisoffi Z, Russo F, Palu G. Human cases of West Nile virus infection in north-eastern Italy, 15 June to 15 November 2010. Euro Surveill. 2011;16:pii19959. [PubMed] [Google Scholar]
- 52.Herwaldt BL, Linden JV, Bosserman E, Young C, Olkowska D, Wilson M. Transfusion-associated babesiosis in the United States: a description of cases. Ann Intern Med. 2011;155:509–519. doi: 10.7326/0003-4819-155-8-201110180-00362. [DOI] [PubMed] [Google Scholar]
- 53.Leiby DA. Transfusion-associated babesiosis: shouldn't we be ticked off? Ann Intern Med. 2011;155:556–557. doi: 10.7326/0003-4819-155-8-201110180-00363. [DOI] [PubMed] [Google Scholar]
- 54.Kuiken T, Fouchier RA, Schutten M, Rimmelzwaan GF, van Amerongen G, van Riel D, Laman JD, de Jong T, van Doornum G, Lim W, Ling AE, Chan PK, Tam JS, Zambon MC, Gopal R, Drosten C, van der Werf S, Escriou N, Manuguerra JC, Stöhr K, Peiris JS, Osterhaus AD. Newly discovered coronavirus as the primary cause of severe acute respiratory syndrome. Lancet. 2003;362:263–270. doi: 10.1016/S0140-6736(03)13967-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Likos AM, Kelvin DJ, Cameron CM, Rowe T, Kuehnert MJ, Norris PJ National Heart, Lung, Blood Institute Retrovirus Epidemiology Donor Study-II (REDS-II) Influenza viremia and the potential for blood-borne transmission. Transfusion. 2007;47:1080–1088. doi: 10.1111/j.1537-2995.2007.01264.x. [DOI] [PubMed] [Google Scholar]
- 56.Qi X, Qian YH, Bao CJ, Guo XL, Cui LB, Tang FY, Ji H, Huang Y, Cai PQ, Lu B, Xu K, Shi C, Zhu FC, Zhou MH, Wang H. Probable person to person transmission of novel avian influenza A (H7N9) virus in Eastern China, 2013:epidemiological investigation. BMJ. 2013;347:f4752. doi: 10.1136/bmj.f4752. doi: 10.1136/bmj.fd4752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Schlenke P, Hervig T, Isola H, Wiesel ML, Kientz D, Pinkoski L, Singh Y, Lin L, Corash L, Cazenave JP. Photochemical treatment of plasma with amotosalen and UVA light: process validation in three European blood centers. Transfusion. 2008;48:697–705. doi: 10.1111/j.1537-2995.2007.01594.x. [DOI] [PubMed] [Google Scholar]
- 58.Hornsey VS, Drummond O, Morrison A, McMillan L, MacGregor IR, Prowse CV. Pathogen reduction of fresh plasma using riboflavin and ultraviolet light: effects on plasma coagulation proteins. Transfusion. 2009;49:2176–2172. doi: 10.1111/j.1537-2995.2009.02272.x. [DOI] [PubMed] [Google Scholar]
- 59.Irsch J, Pathogen Lin L. Inactivation of platelet and plasma blood components for transfusion using the INTERCEPT Blood SystemTM. Transfus Med Hemother. 2011;38:19–31. doi: 10.1159/000323937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Marschner S, Goodrich R. Pathogen reduction technology treatment of platelets, plasma and whole blood using riboflavin and UV light. Transfus Med Hemother. 2011;38:8–18. doi: 10.1159/000324160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Feys HB, Van Aelst B, Devreese K, Devloo R, Coene J, Vanderkerckhoves P, Compermolle V. Oxygen removal during pathogen inactivation with riboflavin and UV light preserves protein function in plasma for transfusion. Vox Sang. 2014;106:307–315. doi: 10.1111/vox.12106. [DOI] [PubMed] [Google Scholar]
- 62.Mohr H, Steil L, Gravemann U, Thiele T, Hammer E, Greinacher A, Müller TH, Völker U. A novel approach to pathogen reduction in platelet concentrates using short-wave ultraviolet light. Transfusion. 2009;49:2612–2624. doi: 10.1111/j.1537-2995.2009.02334.x. [DOI] [PubMed] [Google Scholar]
- 63.Mohr H, Gravemann U, Bayer A, Müller TH. Sterilization of platelet concentrates at production scale by irradiation with short-wave ultraviolet light. Transfusion. 2009;49:1956–1963. doi: 10.1111/j.1537-2995.2009.02228.x. [DOI] [PubMed] [Google Scholar]
- 64.Seltsam A, Müller TH. UVC irradiation for pathogen reduction of platelet concentrates and plasma. Transfus Med Hemother. 2011;38:43–54. doi: 10.1159/000323845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Seghatchian J, Struff WG, Reichenberg S. Main properties of the THERAFLEX MB-plasma system for pathogen reduction. Transfus Med Hemother. 2011;38:55–64. doi: 10.1159/000323786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Pohler P, Lehmann J, Veneruso V, Tomm J, von Bergen M, Lambrecht B, Kohn B, Weingart C, Müller TH, Seltsam A. Evaluation of the tolerability and immunogenicity of ultraviolet C-irradiated autologous platelets in a dog model. Transfusion. 2012;52:2414–2426. doi: 10.1111/j.1537-2995.2012.03583.x. [DOI] [PubMed] [Google Scholar]
- 67.Ciaravino V, McCullough T, Cimino G. The role of toxicology assessment in transfusion medicine. Transfusion. 2003;43:1481–1492. doi: 10.1046/j.1537-2995.2003.00544.x. [DOI] [PubMed] [Google Scholar]
- 68.Ciaravino V, Hanover J, Lin L, Sullivan T, Corash L. Assessment of safety in neonates for transfusion of platelets and plasma prepared with amotosalen photochemical pathogen inactivation treatment by a 1-month intravenous toxicity study in neonatal rats. Transfusion. 2009;49:985–994. doi: 10.1111/j.1537-2995.2008.02076.x. [DOI] [PubMed] [Google Scholar]
- 69.Reddy HL, Dayan AD, Cavagnaro J, Gad S, Li J, Goodrich RP. Toxicity testing of a novel riboflavin-based technology for pathogen reduction and white blood cell inactivation. Transfus Med Rev. 2008;22:133–153. doi: 10.1016/j.tmrv.2007.12.003. [DOI] [PubMed] [Google Scholar]
- 70.Ruane PH, Edrich R, Gampp D, Keil SD, Leonard RL, Goodrich RP. Photochemical inactivation of selected viruses and bacteria in platelet concentrates using riboflavin and light. Transfusion. 2004;44:877–885. doi: 10.1111/j.1537-2995.2004.03355.x. [DOI] [PubMed] [Google Scholar]
- 71.Stramer SL, Hollinger FB, Katz LM, Kleinman S, Metzel PS, Gregory KR, Dodd RY. Emerging infectious disease agents and their potential threat to transfusion safety. Transfusion. 2009;49(suppl 2):1S–29S. doi: 10.1111/j.1537-2995.2009.02279.x. [DOI] [PubMed] [Google Scholar]
- 72.Goodrich RP, Gilmour D, Hovenga N, Keil SH. A laboratory comparison of pathogen reduction technology treatment and culture of platelet products for addressing bacterial contamination concerns. Transfusion. 2009;49:1205–1216. doi: 10.1111/j.1537-2995.2009.02126.x. [DOI] [PubMed] [Google Scholar]
- 73.Dupuis K, Arnold DJ, Sawyer L. High titers of dengue virus in platelet concentrates are inactivated by amotosalen and UVA light (abstract) Transfusion. 2012;52(3 suppl 225A.) [Google Scholar]
- 74.Arslan Ö, Ozukul A, Dupuis K, Sahin F, Stassinopoulos A. Inactivation of Crimean-Congo hemorrhagic fever virus (CCHFV) in full units of human plasma using amotosalen and UVA. Vox Sang. 2013;105(suppl 1):155. [Google Scholar]
- 75.Castro E, González LM, Rubio R, Gironés N, Montero E. The efficacy of UVC pathogen inactivation on the reduction of Babesia divergens in buffy coat derived platelets. Vox Sang. 2013;105(suppl 1):134. [Google Scholar]
- 76.Castro E, Marín-García P, Bautista JM, Montero E, Runio JM. The efficacy of UVC pathogen inactivation on the reduction of Plasmodium falciparum in buffy coat derived platelets. Vox Sang. 2013;105(suppl 1):133. [Google Scholar]
- 77.Gravemann U, Schmidt JP, Tolksdorf F, Sumian C, Müller TH, Bauer G, Biczysko S, Keppler O, Seltsam A, Rabenau HF. Influenza A virus H3N2 is efficiently inactivated by the Theraflex UV-Platelets System. Vox Sang. 2013;105(suppl 1):134. [Google Scholar]
- 78.Vanlandingham DL, Keil SD, Horne KM, Pyles R, Goodrich RP, Higgs S. Photochemical inactivation of chikungunya virus in plasma and platelets using the Mirasol pathogen reduction technology system. Transfusion. 2013;53:284–290. doi: 10.1111/j.1537-2995.2012.03717.x. [DOI] [PubMed] [Google Scholar]
- 79.Steinmann E, Gravemann U, Friesland M, Doerrbecker J, Müller TH, Pietschmann T, Seltsam A. Two pathogen reduction technologies – methylene blue plus light and shortwave ultraviolet light – effectively inactivate hepatitis C virus in blood products. Transfusion. 2013;53:1010–1018. doi: 10.1111/j.1537-2995.2012.03858.x. [DOI] [PubMed] [Google Scholar]
- 80.Störmer M, Arroyo A, Brachert J, Carrero H, Devine D, Epstein JS, Gabriel C, Gelber C, Goodrich R, Hanschmann KM, Heath DG, Jacobs MR, Keil S, de Korte D, Lambrecht B, lee CK, Marcelis J, Marschner S, McDonald C, McGuane S, McKee M, Müller TH, Muthivhi T, Pettersson A, Radziwon P, Ramirez-Arcos S, Reesink HW, Rojo J, Rood I, Schmidt M, Schneider CK, Seifried E, Sicker U, Wendel S, Wood EM, Yomtovian RA, Montag T. Establishment of the first international repository for transfusion-relevant bacteria reference strains: ISBT working party transfusion-transmitted infectious diseases (WP-TTID), subgroup bacteria. Vox Sang. 2012;102:22–31. doi: 10.1111/j.1423-0410.2011.01510.x. [DOI] [PubMed] [Google Scholar]
- 81.Bakkour S, Chafets DM, Wen L, Montalvo L, Busch MP, Lee T. Evaluation of Mirasol® treatment of parvovirus B19 in plasma using real-time PCR inhibition assays. Transfusion. 2012;52(suppl):224A–225A. [Google Scholar]
- 82.Snyder E, Raife T, Lin L, Cimino G, Metzel P, Rheinschmidt M, Baril L, Davis K, Buchholz DH, Corash L, Conlan MG. Recovery and life span of 111indium-radiolabeled platelets treated with pathogen inactivation with amotosalen HCl (S-59) and ultraviolet A light. Transfusion. 2004;44:1732–1740. doi: 10.1111/j.0041-1132.2004.04145.x. [DOI] [PubMed] [Google Scholar]
- 83.AuBuchon JP, Herschel L, Roger J, Taylor H, Whitley P, Li J, Edrich R, Goodrich RP. Efficacy of apheresis platelets treated with riboflavin and ultraviolet light for pathogen reduction. Transfusion. 2005;45:1335–1341. doi: 10.1111/j.1537-2995.2005.00202.x. [DOI] [PubMed] [Google Scholar]
- 84.Bashir S, Cookson P, Wiltshire M, Hawkins L, Sonoda L, Thomas S, Seltsam A, Tolksdorf F, Williamson LM, Cardigan R. Pathogen inactivation of platelets using ultraviolet C light: effect on in vitro function and recovery and survival of platelets. Transfusion. 2013;53:990–1000. doi: 10.1111/j.1537-2995.2012.03854.x. [DOI] [PubMed] [Google Scholar]
- 85.Thiele T, Pohler P, Kohlmann T, Sümnig A, Aurich K, Selleng K, Westphal A, Müller T, Seltsam A, Greinacher A. Open phase I clinical trial on safety and tolerance of autologous UVC-treated single donor platelet concentrates in healthy volunteers – dose escalation study (abstract) Transfus Med Hemother. 2013;40(suppl 1):50. [Google Scholar]
- 86.van Rhenen D, Gulliksson H, Cazenave J-P, Pam-philon D, Ljungman P, Klueter H, Vermeij H, Kappers-Klune M, de Greef G, Laforet M, Lioure B, Davis K, Marblie S, Mayaudon V, Flament J, Conlan J, Conlan M, Lin L, Metzel P, Buchholz D, Corash L. Transfusion of pooled buffy coat platelet components prepared with photochemical pathogen inactivation treatment: the euroSPRITE trial. Blood. 2003;101:2426–2433. doi: 10.1182/blood-2002-03-0932. [DOI] [PubMed] [Google Scholar]
- 87.McCullough J, Vesole D, Benjamin R, Slichter S, Pineda A, Snyder E, Stadtmauer E, Lopez-Plaza I, Coutre S, Strauss RG, Goodnough LT, Fridey JL, Raife T, Cable R, Murphy S, Howard IVF, Davis K, Lin JS, Metzel P, Corash L, Koutsoukos A, Lin L, Buchholz D, Conlan M. Therapeutic efficacy and safety of platelets treated with a photochemical process for pathogen inactivation: the SPRINT trial. Blood. 2004;104:1534–1541. doi: 10.1182/blood-2003-12-4443. [DOI] [PubMed] [Google Scholar]
- 88.Janetzko K, Cazenave JP, Klüter H, Kientz D, Michel M, Beris P, Lioure B, Hastka J, Marblie S, Mayaudon V, Lin L, Lin JS, Conlan MG, Flament J. Therapeutic efficacy and safety of photochemically treated apheresis platelets processed with an optimized integrated set. Transfusion. 2005;45:1443–1452. doi: 10.1111/j.1537-2995.2005.00550.x. [DOI] [PubMed] [Google Scholar]
- 89.Kerkhoffs JL, van Putten WL, Novotny VM, Te Boekhorst PA, Schipperus MR, Zwaginga JJ, van Pampus LC, de Greef GE, Luten M, Huijgens PC, Brand A, van Rhenen DJ Dutch-Belgian HOVON cooperative group. Clinical effectiveness of leucoreduced, pooled donor platelet concentrates, stored in plasma or additive solution with and without pathogen reduction. Br J Hematol. 2010;150:209–217. doi: 10.1111/j.1365-2141.2010.08227.x. [DOI] [PubMed] [Google Scholar]
- 90.Lozano M, Knutson F, Tardivel R, Cid J, Maymó RM, Löf H, Roddie H, Pelly J, Docherty A, Sherman C, Lin L, Propst M, Corash L, Prowse C. A multi-centre study of therapeutic efficacy and safety of platelet components treated with amotosalen and ultraviolet A pathogen inactivation stored for 6 or 7 d prior to transfusion. Br J Hematol. 2011;153:393–401. doi: 10.1111/j.1365-2141.2011.08635.x. [DOI] [PubMed] [Google Scholar]
- 91.Slichter SJ, Raife TJ, Davis K, Rheinschmidt M, Buchholz DH, Corash L, Conlan MG. Platelets photochemically treated with amotosalen HCl and ultraviolet A light correct prolonged bleeding times in patients with thrombocytopenia. Transfusion. 2006;46:731–740. doi: 10.1111/j.1537-2995.2006.00791.x. [DOI] [PubMed] [Google Scholar]
- 92.Simonsen AC, Johansson PI, Conlan MG, Jacquet M, Lin JS, Junge K, Lin L, Sørensen H, Borregaard N, Flament J. Transfusion of 7-day-old amotosalen photochemically treated buffy-coat platelets to patients with thrombocytopenia: a pilot study. Transfusion. 2006;46:424–433. doi: 10.1111/j.1537-2995.2006.00739.x. [DOI] [PubMed] [Google Scholar]
- 93.Vamvakas EC. Meta-analysis of the randomized controlled trials of the hemostatic efficacy and capacity of pathogen-reduced platelets. Transfusion. 2011;51:1058–1071. doi: 10.1111/j.1537-2995.2010.02925.x. [DOI] [PubMed] [Google Scholar]
- 94.Cid J, Escolar G, Lozano M. Therapeutic efficacy of platelet components treated with amotosalen and ultraviolet A pathogen inactivation method: results of a meta-analysis of randomized controlled trials. Vox Sang. 2012;103:322–330. doi: 10.1111/j.1423-0410.2012.01614.x. [DOI] [PubMed] [Google Scholar]
- 95.Corash L, Sherman CD. Evaluation of platelet transfusion clinical trials. Br J Hematol. 2011;153:529–531. doi: 10.1111/j.1365-2141.2010.08413.x. [DOI] [PubMed] [Google Scholar]
- 96.Kerkhoffs JLH. Response to Corash and Sherman. Br J Hematol. 2011;153:531–532. [Google Scholar]
- 97.Sherman CD, Corash LM. Assessment of hemostasis in platelet transfusion clinical trials with differential follow-up. Vox Sang. 2012;103:179. doi: 10.1111/j.1423-0410.2012.01593.x. [DOI] [PubMed] [Google Scholar]
- 98.Ypma PF, Kerkhoffs JL, van Hilten JA, Middelburg RA, Coccoris M, Zwaginga JJ, Beckers EM, Fijnheer R, van der Meer PF, Brand A. The observation of bleeding complications in haematooncological patients: stringent watching, relevant reporting. Transfus Med. 2012;22:426–431. doi: 10.1111/j.1365-3148.2012.01193.x. [DOI] [PubMed] [Google Scholar]
- 99.Webert KE, Arnold DM, Lui Y, Carruthers J, Arnold E, Heddle NM. A new tool to assess bleeding severity in patients with chemotherapy-induced thrombocytopenia. Transfusion. 2012;52:2466–2474. doi: 10.1111/j.1537-2995.2012.03634.x. [DOI] [PubMed] [Google Scholar]
- 100.Cazenave JP, Isola H, Waller C, Mendel I, Kientz D, Laforêt M, Raidot JP, Kandel G, Wiesel ML, Corash L. Use of additive solutions and pathogen inactivation treatment of platelet components in a regional blood center: impact on patient outcomes and component utilization during a 3-year period. Transfusion. 2011;51:622–629. doi: 10.1111/j.1537-2995.2010.02873.x. [DOI] [PubMed] [Google Scholar]
- 101.Vamvakas EC. Meta-analysis of the studies of bleeding complications of platelets pathogen-reduced with the Intercept system. Vox Sang. 2012;102:302–316. doi: 10.1111/j.1423-0410.2011.01555.x. [DOI] [PubMed] [Google Scholar]
- 102.Murphy S, Snyder E, Cable R, Slichter SJ, Strauss RG, McCullough J, Lin JS, Corash L, Conlan MG SPRINT Study Group. Platelet dose consistency and its effect on the number of platelet transfusions for support of thrombocytopenia: an analysis of the SPRINT trial of platelets photochemically treated with amotosalen HCl and ultraviolet A light. Transfusion. 2006;46:24–33. doi: 10.1111/j.1537-2995.2005.00671.x. [DOI] [PubMed] [Google Scholar]
- 103.Snyder E, McCullough J, Slichter SJ, Strauss RG, Lopez-Plaza I, Lin JS, Corash L, Conlan MG SPRINT Study Group. Clinical safety of platelets photochemically treated with amotosalen HCl and ultraviolet A light for pathogen inactivation: the SPRINT trial. Transfusion. 2005;45:1864–1875. doi: 10.1111/j.1537-2995.2005.00639.x. [DOI] [PubMed] [Google Scholar]
- 104.Infanti L, Stebler C, Job S, Ruesch M, Gratwohl A, Irsch J, Lin L, Buser A. Pathogen-inactivation of platelet components with the INTERCEPT Blood SystemTM a cohort study. Transfus Apher Sci. 2011;45:175–181. doi: 10.1016/j.transci.2011.07.013. [DOI] [PubMed] [Google Scholar]
- 105.Schlenke P, Hagenah W, Irsch J, Sundin D, Corash L, Lin L, Kirchner H, Wagner T. Safety and clinical efficacy of platelet components prepared with pathogen inactivation in routine use for thrombocytopenic patients. Ann Hematol. 2011;90:1457–1465. doi: 10.1007/s00277-011-1222-3. [DOI] [PubMed] [Google Scholar]
- 106.Osselaer JC, Cazenave JP, Lambermont M, Garraud O, Hidajat M, Barbolla L, Tardivel R, Defoin L, Waller C, Mendel I, Raidot JP, Kandel G, De Meuter R, Fabrigli P, Dehenau D, Arroyo JL, Padrón F, Gouezec H, Corral M, Jacquet M, Sundin D, Lin L, Corash L. An active haemovigilance programme characterizing the safety profile of 7437 platelet transfusions prepared with amotosalen photochemical treatment. Vox Sang. 2008;94:315–323. doi: 10.1111/j.1423-0410.2007.01035.x. [DOI] [PubMed] [Google Scholar]
- 107.Osselaer JC, Messe N, Hervig T, Bueno J, Castro E, Espinosa A, Accorsi P, Junge K, Jacquet M, Flament J, Corash L. A prospective observational cohort safety study of 5106 platelet transfusions with components prepared with photochemical pathogen inactivation treatment. Transfusion. 2008;48:1061–1071. doi: 10.1111/j.1537-2995.2008.01643.x. [DOI] [PubMed] [Google Scholar]
- 108.Cazenave JP, Waller C, Kientz D, Mendel I, Lin L, Jacquet M, Propst M, Liu W, Corash L, Sundin D, Defoin L, Messe N, Osselaer JC. An active hemovigilance program characterizing the safety profile of 7483 transfusions with plasma components prepared with amotosalen and UVA photochemical treatment. Transfusion. 2010;50:1210–1219. doi: 10.1111/j.1537-2995.2009.02579.x. [DOI] [PubMed] [Google Scholar]
- 109.Cazenave JP, Folléa G, Bardiaux L, Boiron JM, Lafeuillade B, Debost M, Lioure B, Harousseau JL, Deconinck E, Tabrizi R, Cahn JY, Michallet M, Ambruso D, Tissot JD, Sensebé L, Kondo T, McCullough J, Rebulla P, Escolar G, Mintz P, Heddle NM, Goodrich RP, Bruhwyler J, Le C, Cook RJ, Stouch B, Fletcher D, Deberdt L Mirasol Clinical Evaluation Study Group. A randomized controlled clinical trial evaluating the performance and safety of platelets treated with MIRASOL pathogen reduction technology. Transfusion. 2010;50:2362–2375. doi: 10.1111/j.1537-2995.2010.02694.x. [DOI] [PubMed] [Google Scholar]
- 110.The Trial to Reduce Alloimmunization to Platelets Study Group. Leukocyte reduction and ultraviolet B irradiation of platelets to prevent alloimmunization and refractoriness to platelet transfusions. N Engl J Med. 1997;337:1861–1869. doi: 10.1056/NEJM199712253372601. [DOI] [PubMed] [Google Scholar]
- 111.Antic A, Stanojkovic Z, Jelic M, Stanojkovic M. Clinical efficacy of buffy coat platelets treated with riboflavin and ultraviolet light (Mirasol PRT system) Vox Sang. 2011;101(suppl 1):178. [Google Scholar]
- 112.Yanez M, Garcia de Coca A, Medina L, Galvez FJ, Martin Vaca MC, Martinez Alvarez M, Prieto Arija L, Blanco Peris L. Clinical evaluation of platelets treated with riboflavin and UV light in the presence of platelet additive solution in thrombocytopenic patients. Vox Sang. 2011;101(suppl 1):178. [Google Scholar]
- 113.Coene GC, De Schrijver E, Sabot B, Deeren D, Reynaerts I, Vandekerckhove P. In vitro and in vivo evaluation of Mirasol pathogen reduction technology treated platelets stored in SSP+ Vox Sang. 2011;101(suppl 1):188–189. [Google Scholar]
- 114.Johansson PI, Simonsen AC, Brown PN, Ostrowski SR, Deberdt L, Van Hoydonck P, Yone-mura SS, Goodrich RP. A pilot study to assess the hemostatic function of pathogen-reduced platelets in patients with thrombocytopenia. Transfusion. 2012;53:2043–2052. doi: 10.1111/trf.12055. [DOI] [PubMed] [Google Scholar]
- 115.Henschler R, Seifried E, Mufti N. Development of the S-303 pathogen inactivation technology for red blood cell concentrates. Transfus Med Hemother. 2011;38:33–42. doi: 10.1159/000324458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Cancelas JA, Dumont LJ, Rugg N, Szczepiorkowski ZM, Herschel L, Siegel A, Pratt PG, Worsham DN, Erickson A, Propst M, North A, Sherman CD, Mufti NA, Reed WF, Corash L. Stored red blood cell viability is maintained after treatment with a second-generation S-303 pathogen inactivation process. Transfusion. 2011;53:529–534. doi: 10.1111/j.1537-2995.2011.03163.x. [DOI] [PubMed] [Google Scholar]
- 117.Benjamin RJ, McCullough J, Mintz PD, Snyder E, Spotnitz WD, Rizzo RJ, Wages D, Lin JS, Wood L, Corash L, Conlan MG. Therapeutic efficacy and safety of red blood cells treated with a chemical process (S-303) for pathogen inactivation: a phase III clinical trial in cardiac surgery patients. Transfusion. 2005;45:1739–1749. doi: 10.1111/j.1537-2995.2005.00583.x. [DOI] [PubMed] [Google Scholar]
- 118.Mufti NA, Erickson AC, North AK, Hanson D, Sawyer L, Corash LM, Lin L. Treatment of whole blood (WB) and red blood cells (RBC) with S-303 inactivates pathogens and retains in vitro quality of stored RBC. Biologicals. 2010;38:14–19. doi: 10.1016/j.biologicals.2009.10.019. [DOI] [PubMed] [Google Scholar]
- 119.North A, Ciaravino V, Mufti N, Corash L. Pre-clinical pharmacokinetic and toxicology assessment of red blood cells prepared with S-303 pathogen inactivation treatment. Transfusion. 2011;51:2208–2218. doi: 10.1111/j.1537-2995.2011.03132.x. [DOI] [PubMed] [Google Scholar]
- 120.Goodrich RP, Doane S, Reddy HL. Design and development of a method for the reduction of infectious pathogen load and inactivation of white blood cells in whole blood products. Biologicals. 2010;38:20–30. doi: 10.1016/j.biologicals.2009.10.016. [DOI] [PubMed] [Google Scholar]
- 121.Goodrich RP, Murthy KK, Doane SK, Fitzpatrick CN, Morrow LS, Arndt PA, Reddy HL, Buytaert-Hoefen, Garratty G. Evaluation of potential immune response and in vivo survival of riboflavin-ultraviolet light-treated red blood cells in baboons. Transfusion. 2009b;49:64–74. doi: 10.1111/j.1537-2995.2008.01940.x. [DOI] [PubMed] [Google Scholar]
- 122.Okoye OT, Reddy H, Wong MD, Doane S, Resnick S, Karamanos E, Skiada D, Goodrich R, Inaba K. Large animal evaluation of the safety and efficacy of whole blood treated with Mirasol system in a diffuse, non-surgical bleeding model. Transfusion. 2013;53(suppl 82A.) doi: 10.1111/trf.12894. [DOI] [PubMed] [Google Scholar]
- 123.Cancelas JA, Rugg N, Fletcher D, Pratt PG, Worsham DN, Dunn SK, Marschner S, Reddy HL, Goodrich RP. In vivo viability of stored red blood cells derived from riboflavin plus ultraviolet light-treated whole blood. Transfusion. 2011;51:1460–1468. doi: 10.1111/j.1537-2995.2010.03027.x. [DOI] [PubMed] [Google Scholar]
- 124.Keil SD, Rapaport R, Doane SK, Young R, Marschner S, Campbell TB. Viral reduction of intracellular HIV using the Mirasol® system for whole blood. Vox Sang. 2012;103(suppl 1):144. [Google Scholar]
- 125.Tonnetti L, Thorp AM, Reddy HL, Goodrich RP, Leiby DA. Evaluation of the reduction of T. cruzi with the Mirasol® system for whole blood. Transfusion. 2010;50(suppl 209A.) doi: 10.1111/j.1537-2995.2009.02538.x. [DOI] [PubMed] [Google Scholar]
- 126.Tonnetti L, Thorp AM, Reddy HL, Keil SD, Goodrich RP, Leiby DA. Evaluation of Babesia microti reduction with the Mirasol® system for whole blood. Transfusion. 2011;51(suppl 27A.) [Google Scholar]
- 127.Tonnetti L, Thorp AM, Reddy HL, Keil SD, Doane S, Goodrich RP, Leiby DA. Validating the Mirasol® system for whole blood: impact on Leishmania donovani infectivity. Transfusion. 2012;52(suppl 212A.) doi: 10.1111/trf.12820. [DOI] [PubMed] [Google Scholar]
- 128.Doane S, Keil SD, Gilmour H, Reddy H, Wilkinson S, Hovenga N, Goodrich R. Treatment of whole blood with riboflavin and UV light: effectiveness against clinically relevant bacterial contamination. Transfusion. 2013;53(suppl 203A.) [Google Scholar]
- 129.El Chaar ME, Atwal S, Freimanis GL, Dinko B, Sutherland CJ, Allain J-P. Inactivation of Plasmodium falciparum in whole blood by riboflavin plus irradiation. Transfusion. 2013;53:3174–3183. doi: 10.1111/trf.12235. [DOI] [PubMed] [Google Scholar]
- 130.Reddy HL, Doane SK, Keil SD, Marschner S, Goodrich RP. Development of a riboflavin and ultraviolet light-based device to treat whole blood. Transfusion. 2013;53(suppl):131S–136S. doi: 10.1111/trf.12047. [DOI] [PubMed] [Google Scholar]
- 131.Fast LD, Nevola M, Tavares J, Reddy HL, Goodrich RP, Marschner S. Treatment of whole blood with riboflavin plus ultraviolet light, an alternative to gamma irradiation in the prevention of transfusion-associated graft-versus-host disease? Transfusion. 2013;53:373–381. doi: 10.1111/j.1537-2995.2012.03715.x. [DOI] [PubMed] [Google Scholar]
- 132.Owusu-Ofori S, Owusu-Ofori A, Freimanis G, Keil SD, Allain J. Riboflavin and UV illumination treatment of whole blood units in a Ghanaian blood center. Transfusion. 2013;53(suppl):202A–203A. [Google Scholar]
- 133.Oei W, Janssen MP, van der Poel CL, van Steenbergen JE, Rehmet S, Kretzschmar ME. Modeling the transmission risk of emerging infectious diseases through blood transfusion. Transfusion. 2013;53:1421–1428. doi: 10.1111/j.1537-2995.2012.03941.x. [DOI] [PubMed] [Google Scholar]
- 134.Störmer M, Vollmer T, Kleesiek K, Dreier J. Spore-forming organisms in platelet concentrates: a challenge in transfusion bacterial safety. Transfus Med. 2008;18:371–376. doi: 10.1111/j.1365-3148.2008.00895.x. [DOI] [PubMed] [Google Scholar]