首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
To ensure good performance of pathogen inactivation with the INTERCEPT blood system, specific target requirements must be met for platelet dose, volume, plasma content and residual red blood cells (RBCs) prior to photochemical treatment (PCT). A two-arm in vitro study was conducted to compare quality parameters of pooled platelet concentrates (PCs), either treated (test units) or nontreated (control units). PCs meeting European requirements were evaluated with reference to their compliance with INTERCEPT guard bands. Of 50 PCs (25 tests and 25 controls) meeting European quality requirements, 24% (three test and three controls units) did not reach INTERCEPT requirements, particularly in terms of sufficient volumes and RBC contamination. The buffy-coat optimization procedure assessed prior to this study ensured plasma contents well within target limits of 30 to 45%. Due to PCT-related in-process loss of 11% in volume (34.38 +/- 3.94) and in platelet dose (0.41 +/- 0.14), the mean platelet dose was significantly (P < 0.001) lower in test units: 3.1 +/- 0.3 versus 3.6 +/- 0.4 x 10(11). After treatment, six of the overall 25 test units (25%) would not have met the European guideline for platelet dose (3.0 x 10(11)). Before implementation of techniques for pathogen reduction, each centre should optimize processing steps during a validation procedure to ensure PC complying with INTERCEPT targets before and European targets after treatment. Besides buffy-coat optimization for sufficient plasma reduction, centrifugation profiles need to be optimized as well to prevent PC with low volumes and, in particular, with higher than acceptable RBC contamination.  相似文献   

2.
BACKGROUND: The transmission of pathogens via blood transfusion is still a major threat. Expert conferences established the need for a pro-active approach and concluded that the introduction of a pathogen inactivation/reduction technology requires a thorough safety profile, a comprehensive pre-clinical and clinical development and an ongoing hemovigilance program. MATERIAL AND METHODS: The INTERCEPT Blood System utilizes amotosalen and UVA light and enables for the treatment of platelets and plasma in the same device. Preclinical studies of pathogen inactivation and toxicology and a thorough program of clinical studies have been conducted and an active he-movigilance-program established. RESULTS: INTERCEPT shows robust efficacy of inactivation for viruses, bacteria (including spirochetes), protozoa and leukocytes as well as large safety margins. Furthermore, it integrates well into routine blood center operations. The clinical study program demonstrates the successful use for very diverse patient groups. The hemovigilance program shows safety and tolerability in routine use. Approximately 700,000 INTERCEPT-treated products have been transfused worldwide. The system is in clinical use since class III CE-mark registration in 2002. The safety and efficacy has been shown in routine use and during an epidemic. CONCLUSION: The INTERCEPT Blood System for platelets and plasma offers enhanced safety for the patient and protection against transfusion-transmitted infections.  相似文献   

3.
BACKGROUND: A photochemical treatment process has been developed for the inactivation of viruses and bacteria in platelet concentrates. This process is based on the photochemical reaction of a novel psoralen, S- 59, with nucleic acids upon illumination with long-wavelength ultraviolet light (UVA, 320–400 nm). STUDY DESIGN AND METHODS: High levels of pathogens were added to single-donor platelet concentrates containing 3 to 5 × 10(11) platelets in 300 mL of 35-percent autologous plasma and 65-percent platelet additive solution. After treatment with S-59 (150 microM) and UVA (0-3 J/cm2), the infectivity of each pathogen was measured with established biologic assays. In vitro platelet function after photochemical treatment was evaluated during 7 days of storage by using a panel of 14 assays. The in vivo recovery and life span of photochemically treated platelets were evaluated after 24 hours of storage in a primate transfusion model. RESULTS: The following levels of pathogen inactivation were achieved:>10(6.7) plaque-forming units (PFU) per mL of cell-free human immunodeficiency virus (HIV),>10(6.6) PFU per mL of cell-associated HIV,>10(6.8) infectious dose (ID50) per mL of duck hepatitis B virus (a model for hepatitis B virus),>10(6.5) PFU per mL of bovine viral diarrhea virus (a model for hepatitis C virus),>10(6.6) colony-forming units of Staphylococcus epidermidis, and>10(5.6) colony-forming units of Klebsiella pneumoniae. Expression of integrated HIV was inhibited by 0.1 microM S- 59 and 1 J per cm2 of UVA. In vitro and in vivo platelet function were adequately maintained after antiviral and antibacterial treatment. CONCLUSION: Photochemical treatment of platelet concentrates offers the potential for reducing transfusion-related viral and bacterial diseases.  相似文献   

4.
BACKGROUND: The INTERCEPT Blood System, a photochemical treatment (PCT) process, has been developed to inactivate pathogens in platelet concentrates. These studies evaluated the efficacy of PCT to inactivate pathogens in plasma and the effect of PCT on plasma function. STUDY DESIGN AND METHODS: Jumbo (600 mL) plasma units were inoculated with high titers of test pathogens and treated with 150 micromol per L amotosalen and 3 J per cm(2) long-wavelength ultraviolet light. The viability of each pathogen before and after treatment was measured with biological assays. Plasma function was evaluated through measurement of coagulation factors and antithrombotic protein activities. RESULTS: The levels of inactivation expressed as log-reduction were as follows: cell-free human immunodeficiency virus-1 (HIV-1), greater than 6.8; cell-associated HIV-1, greater than 6.4; human T-lymphotropic virus-I (HTLV-I), 4.5; HTLV-II, greater than 5.7; hepatitis B virus (HBV) and hepatitis C virus, greater than 4.5; duck HBV, 4.4 to 4.5; bovine viral diarrhea virus, 6.0; severe acute respiratory syndrome coronavirus, 5.5; West Nile virus, 6.8; bluetongue virus, 5.1; human adenovirus 5, 6.8; Klebsiella pneumoniae, greater than 7.4; Staphylococcus epidermidis and Yersinia enterocolitica, greater than 7.3; Treponema pallidum, greater than 5.9; Borrelia burgdorferi, greater than 10.6; Plasmodium falciparum, 6.9; Trypanosoma cruzi, greater than 5.0; and Babesia microti, greater than 5.3. Retention of coagulation factor activity after PCT was expressed as the proportion of pretreatment (baseline) activity. Retention was 72 to 73 percent of baseline fibrinogen and Factor (F)VIII activity and 78 to 98 percent for FII, FV, FVII, F IX, FX, FXI, FXIII, protein C, protein S, antithrombin, and alpha2-antiplasmin. CONCLUSION: PCT of plasma inactivated high levels of a wide range of pathogens while maintaining adequate coagulation function. PCT has the potential to reduce the risk of transfusion-transmitted diseases in patients requiring plasma transfusion support.  相似文献   

5.
BACKGROUND: Contamination of blood products with hepatitis C virus (HCV) can cause infections resulting in acute and chronic liver diseases. Pathogen reduction methods such as photodynamic treatment with methylene blue (MB) plus visible light as well as irradiation with shortwave ultraviolet (UVC) light were developed to inactivate viruses and other pathogens in plasma and platelet concentrates (PCs), respectively. So far, their inactivation capacities for HCV have only been tested in inactivation studies using model viruses for HCV. Recently, a HCV infection system for the propagation of infectious HCV in cell culture was developed. STUDY DESIGN AND METHODS: Inactivation studies were performed with cell culture–derived HCV and bovine viral diarrhea virus (BVDV), a model for HCV. Plasma units or PCs were spiked with high titers of cell culture–grown viruses. After treatment of the blood units with MB plus light (Theraflex MB‐Plasma system, MacoPharma) or UVC (Theraflex UV‐Platelets system, MacoPharma), residual viral infectivity was assessed using sensitive cell culture systems. RESULTS: HCV was sensitive to inactivation by both pathogen reduction procedures. HCV in plasma was efficiently inactivated by MB plus light below the detection limit already by 1/12 of the full light dose. HCV in PCs was inactivated by UVC irradiation with a reduction factor of more than 5 log. BVDV was less sensitive to the two pathogen reduction methods. CONCLUSIONS: Functional assays with human HCV offer an efficient tool to directly assess the inactivation capacity of pathogen reduction procedures. Pathogen reduction technologies such as MB plus light treatment and UVC irradiation have the potential to significantly reduce transfusion‐transmitted HCV infections.  相似文献   

6.
BACKGROUND: Human T-cell leukemia virus Types I and II (HTLV-I and HTLV-II), blood-borne retroviruses found worldwide, can cause leukemia, immunosuppression, and severe neurologic diseases. In most countries, HTLV-I and -II screening is not performed systematically for blood donations. A new photochemical treatment (PCT) with a synthetic psoralen was developed to inactivate most pathogens in platelet (PLT) concentrates or plasma and to improve the safety of blood donations. STUDY DESIGN AND METHODS: Cell-associated HTLV-I or -II (10(6)/mL) was inoculated in full-size fresh PLT concentrates or fresh frozen plasma and treated with 150 micromol per L amotosalen (S-59) and different doses of long-wavelength ultraviolet A (UVA) light. The residual viral titer in the treated samples was assessed by a cocultivation assay on indicator cells. RESULTS: The inactivation obtained at a 3.0 J per cm2 UVA dose was greater than 5.2 log foci-forming units (FFUs) per mL for HTLV-I and 4.6 log FFUs per mL for HTLV-II in presence of human PLT concentrates and greater than 4.5 log FFUs per mL for HTLV-I and 5.7 log FFUs per mL for HTLV-II in the presence of human plasma. The residual infectivity was very low and shown as the limit of detection of the cocultivation assay. CONCLUSION: In human plasma or PLT concentrates, the retroviruses HTLV-I and -II were strongly sensitive to the PCT with 150 micromol per L amotosalen (S-59) and a 3.0 J per cm2 UVA dose. This high efficiency for photoinactivation of these retroviruses opens a possibility of improving the safety of PLTs or plasma transfusion in the future.  相似文献   

7.
Emerging pathogens continue to threaten blood safety, requiring novel safety approaches. INTERCEPT Blood System for platelets (IBSP) inactivates pathogens, aiming at eliminating the risk of transmitting current and emerging pathogens. The objective was to evaluate the incremental cost-effectiveness ratio (ICER) for IBSP in Belgium. A decision model comparing a 'world with IBSP' to a 'world without IBSP' calculates lifetime costs and 'quality adjusted life years' (QALYs) following platelet transfusion in different indications. Disease-specific life expectancy and consequences of transfusion-transmitted infections were obtained from literature. Transfusion safety and costs were obtained from official sources. Hepatitis C virus-like emerging pathogen was simulated. A wide range of ICERs was observed, highly sensitive to the risk of emerging pathogen trans- mission, underlying disease and age. In the most conservative approach, ICER ranged from 3,459,201 Euro/QALY in absence of emerging pathogen to 195,364 Euro/QALY. The mean threshold of emerging infection risk for IBSP dominance (saving money and producing health gains) ranged from 1/1,079 to 1/2,858 transfusions. Considering the high value authorities appear to place on preventing accidental injury, and ICER of recent implementations in transfusion medicine (NAT: up to 2.3 million Euro per lifeyear), IBSP can be considered cost-effective, taking into account the potential risk of emerging pathogens.  相似文献   

8.
Trypanosoma cruzi, the protozoan pathogen that causes Chagas' disease, can be found in the blood of infected individuals for their entire life span. This presents a serious challenge in safeguarding blood products. Transmission of T. cruzi from blood products is a frequent occurrence in Latin America, where Chagas' disease is endemic. This study was designed to determine whether T. cruzi could be inactivated in human platelet concentrates and plasma by a photochemical treatment process with long-wavelength UV A light (UVA, 320 to 400 nm) plus the psoralen amotosalen HCl (Cerus Corporation). Units of platelet concentrates (300 ml) and plasma (300 ml) were intentionally contaminated with approximately 10(6) T. cruzi trypomastigotes, the T. cruzi form found in the bloodstream, per ml. The viability of T. cruzi after photochemical inactivation was determined by their ability to replicate in 3T3 fibroblasts. Controls, including treatment with 150 micro M amotosalen or 3 J/cm(2) UVA alone, did not lead to reduction of the viability of T. cruzi in plasma or platelet concentrates. However, treatment with 150 micro M amotosalen plus 3 J/cm(2) UVA inactivated T. cruzi to undetectable levels in plasma and platelet concentrates. This represented a >5.4-log reduction of T. cruzi in platelet concentrates and >5.0-log reduction of T. cruzi in plasma. We conclude that the amotosalen plus UVA photochemical inactivation technology is effective in inactivating high levels of protozoan pathogens, such as T. cruzi, in platelet concentrates and plasma, as has been previously shown for numerous viruses and bacteria.  相似文献   

9.
BACKGROUND: Pathogen inactivation of platelet (PLT) components (INTERCEPT Blood System, Cerus Europe) was implemented into routine practice at a blood center supporting a tertiary care hospital. Utilization of platelet components (PCs) and red blood cell (RBC) components was analyzed for 3 years before and 3 years after introduction of pathogen inactivation to assess the impact of pathogen inactivation on component use.
STUDY DESIGN AND METHODS: This was a retrospective analysis of prospectively collected data. An electronic database used in routine blood bank hemovigilance to monitor production and use of blood components was analyzed to assess clinical outcomes.
RESULTS: Transfusion records were analyzed for 688 patients supported with conventional PCs and 795 patients supported with pathogen inactivation PCs. Additional analyses were conducted for intensively transfused hematology patients. Patient demographics (age category, sex, and diagnostic category) were not different in the two observation periods. For all patients, mean numbers of PC per patient were not different for conventional PCs and pathogen inactivation PCs (9.9 ± 19.5 vs. 10.1 ± 20.9, p = 0.88). Data for hematology patients (272 conventional PCs and 276 pathogen inactivation PCs) confirmed that days of PLT support were not different (31.6 ± 42.6 vs. 33.1 ± 47.9, p = 0.70) nor was total PLT dose (1011) per patient (87.3 ± 115.4 vs. 88.1 ± 111.6, p = 0.93). RBC use, for all patients and hematology patients, was not different in the two observation periods, either during periods of PLT support or outside periods of PLT transfusion support.
CONCLUSION: Pathogen inactivation of PCs had no adverse impact on component use during a 3-year observation period of routine practice.  相似文献   

10.
Considerable progress has been made in the last decade in producing purer, safer, leucocyte and plasma reduced platelet concentrates (PC) with an extended shelf life. The development of different pathogen inactivation technologies (PIT) has made a substantial contribution to this trend. Preceding platelet PIT (INTERCEPT Blood System/Cerus Corporation, Concord, CA, USA; MIRASOL/Caridian BCT, Lakewood, CO, USA) are based on adding a photosensitive compound to PC. The mixture is then activated by UV light in the UVB and/or UVA spectral regions. A novel procedure, THERAFLEX UV-Platelets (MacoPharma, Mouvaux, France), was recently developed that uses short-wave ultraviolet light (UVC), without addition of any photoactive agent. This technology has proven to be highly effective in sterilising bacteria (the major cause of morbidity/mortality after platelet transfusion) as well as inactivating other transfusion transmitted DNA/RNA containing pathogens and residual leucocytes. Any PIT reflects a balance between the efficacy of pathogen inactivation and preservation of platelet quality and function. A broad spectrum of in vitro tests have become available for the assessment of platelet storage lesion (PSL), aiming to better predict clinical outcome and untoward effects of platelet therapy. Recent paired studies on the release of platelet-derived cytokines, as new platelet performance indicators, revealed a parallel increase in both THERAFLEX UV-treated and control PC throughout storage, supporting the notion that the bioavailability of platelet function is not grossly affected by UVC treatment. This is corroborated by some newer technologies for proteomic analysis, showing that the THERAFLEX UV-Platelets system results in limited disruption of integrin-regulating extracellular disulfide bonds and minimal protein alterations when compared to UVB and gamma irradiation. Moreover, standard in vitro parameters reflecting activation, metabolic activity and function of platelets are useful indicators of the overall performance of processing and storage and may be used as surrogate markers of platelet quality in vivo. However, there is some doubt as to what degree each marker alone or in combination reflects the true clinical outcome of transfused platelets. Therefore, an appropriate clinical programme has been initiated. The preclinical evaluation demonstrated tolerability and immunological safety of THERAFLEX UV-Platelets using an animal model. Additionally, the system has successfully completed two autologous Phase I trials on recovery and survival. Preliminary results suggest that the recovery and survival rates are consistent with other pathogen reduced platelet products that are licensed and in use. The method is currently under evaluation for safety and tolerability of UVC-treated platelets in healthy volunteers. Presently the THERAFLEX UV-Platelets system is the simplest and purest PIT easily adaptable to the existing blood bank setting. In the future, extension of the application range of the THERAFLEX UV-Platelets system is expected, in order to make this new technology compatible with a broad spectrum of collection and processing platforms, and with other blood products.  相似文献   

11.
BACKGROUND: The epidemic of West Nile virus (WNV) in the US resulted in cases of transfusion-transmitted WNV. Effective pathogen reduction methods could have removed this infectious agent from the blood supply We have evaluated the efficacy of photodynamic treatment of fresh frozen plasma (FFP) with methylene blue (MB), a decontamination method applied in several European countries. STUDY DESIGN AND METHODS: FFP units (300 ml each) were spiked with WNV. MB was added, and the units were illuminated with white or monochromatic yellow light. WNV infectivity was determined by bioassay. WNV-RNA was quantitated by real-time PCR. The inactivation of WNV was investigated under standard and under suboptimal conditions, respectively. In addition, rechallenge experiments with multiple addition of WNV at maximal load (approx. 105 CFU/ml) and repeated illumination without replenishing MB were performed. RESULTS: Complete inactivation of WNV was achieved by MB (0.8-1 mmol/l) and illumination with white light (30,000-45,000 Lux) within 2 min. White yellow light 20-40 J/cm(2) (2.5-5 min) were sufficient for inactivation by 5.75 log10-steps. The rechallenge experiments revealed the substantial reserve capacity of the procedure to inactivate WNV. Quantitative PCR indicated that the viral RNA was rapidly destroyed. CONCLUSION: All experimental data demonstrate the enormous potency of phototreatment with MB to inactivate WNV in plasma.  相似文献   

12.
BACKGROUND: The Intercept Blood System (IBS) for platelets has been developed to reduce pathogen transmission risks during transfusions. OBJECTIVE: This study was a comprehensive economic analysis of the cost-effectiveness of using the IBS for single-donor apheresis platelets (AP) and random-donor pooled platelet concentrates (PC) versus AP and PC without the IBS in the United States in patient populations in which platelets are commonly transfused. METHODS: All data used in this analysis were summarized from existing published sources (primarily indexed in MEDLINE) and data on file at Baxter Healthcare Corporation (Chicago, Illinois) and Cerus Corporation (Concord, California). A literature-based decision-analytic model was developed to assess the economic costs and clinical outcomes associated with the use of AP and PC treated with the IBS for several conditions and procedures that account for a considerable proportion of the platelet usage in the United States: acute lymphocytic leukemia, non-Hodgkin's lymphoma, coronary artery bypass graft, and hip arthroplasty Risks of infection with HIV, hepatitis C virus (HCV), hepatitis B virus, human T-cell lymphotropic virus type 1, or bacterial agents were incorporated into the model. Possible benefits of reduction of the risk of emerging HCV like pathogens and elimination of the need for gamma irradiation were explored in sensitivity analyses. RESULTS: The incremental cost per quality-adjusted life-year gained by using AP + IBS versus untreated AP ranged from 1,308,833 dollars to 4,451,650 dollars (without bacterial testing) and 4,759,401 dollars to 22,968,066 dollars (with bacterial testing). Corresponding figures for PC + IBS versus untreated PC ranged from 457,586 dollars to 1,816,060 dollars. Inclusion of emerging HCV like virus and the elimination of the need for gamma irradiation improved the cost-effectiveness to a range of 177,695 dollars to 1,058,127 dollars for AP without bacterial testing, 176,572 dollars to 1,330,703 dollars for AP with bacterial testing, and 22,888 dollars to 153,564 dollars for PC. The model was most likely to be affected by mortality from bacterial contamination, IBS effect on platelet utilization, and the inclusion of potential benefits (ie, gamma irradiation and/or emergent HCV-like virus). The model was relatively insensitive to changes in the IBS price and viral transmission risks. CONCLUSIONS: The cost-effectiveness of pathogen inactivation via the IBS for platelets is comparable to that of other accepted blood safety interventions (eg, nucleic acid amplification technology). The IBS for platelets may be considered a desirable strategy to increase the safety of platelet transfusions and a potential insurance against the threat of emerging pathogens.  相似文献   

13.
BACKGROUND: During the Chikungunya virus (CHIKV) epidemic on Ile de La Réunion, France, more than 30% of 750,000 inhabitants were infected. Local blood donation was suspended to prevent transfusion-transmitted infection (TT-CHIKV). To sustain the availability of platelet (PLT) components, the Établissement Français du Sang implemented universal pathogen inactivation (INTERCEPT, Cerus Europe BV) of PLT components (CPAs). The study assessed the safety of PLT components treated with pathogen inactivation transfused in routine clinical practice.
STUDY DESIGN AND METHODS: This was a retrospective observational study using patient medical records and the AFSSAPS hemovigilance database (eFIT) to identify TT-CHIKV and adverse events (AEs) classified as acute transfusion reactions (ATRs) to PLT components prepared with pathogen inactivation.
RESULTS: During 1 year, 1950 INTERCEPT-CPAs were transfused to 335 adult, 51 pediatric, and 41 infant patients. Nineteen AEs were observed in 15 patients and 10 were classified as ATRs. Eight ATRs occurred in 6 pediatric hematology-oncology patients. No ATRs were observed in infants. The most frequently reported signs and symptoms were Grade 1 urticaria, itching, chills, fever, and anxiety. No cases of transfusion-related acute lung injury, TT-sepsis, or TT-CHIKV were detected.
CONCLUSIONS: INTERCEPT-CPAs were well tolerated in a broad range of patients, including infants. ATR incidence was low and when present ATRs were of mild severity.  相似文献   

14.
Besides the current efforts devoted to microbial risk reduction, pathogen inactivation technologies promise reduction of the residual risk of known and emerging infectious agents. A novel pathogen reduction process for platelets, the THERAFLEX UV-Platelets system, has been developed and is under clinical evaluation for its efficacy and safety. In addition, proof of principle has been shown for UVC treatment of plasma units. The pathogen reduction process is based on application of UVC light of a specific wavelength (254 nm) combined with intense agitation of the blood units to ensure a uniform treatment of all blood compartments. Due to the different absorption characteristics of nucleic acids and proteins, UVC irradiation mainly affects the nucleic acid of pathogens and leukocytes while proteins are largely preserved. UVC treatment significantly reduces the infectivity of platelet units contaminated by disease-causing viruses and bacteria. In addition, it inactivates residual white blood cells in the blood components while preserving platelet function and coagulation factors. Since no photoactive compound needs to be added to the blood units, photoreagent-related adverse events are excluded. Because of its simple and rapid procedure without the need to change the established blood component preparation procedures, UVC-based pathogen inactivation could easily be implemented in existing blood banking procedures.  相似文献   

15.
SUMMARY: TWO PREPARATIONS ARE AVAILABLE FOR PLATELET TRANSFUSION: single-donor apheresis platelet concentrates (APC) and pooled platelet concentrates (PPC) prepared from 4-6 whole blood units. Clear advantages of APC over PPC are a markedly reduced donor exposure of recipients, and easier logistics when attempting a complete supply with ABO-identical and Rh-compatible platelet concentrates. Regulations should aim at complete ABO-identical platelet transfusions because major and minor ABO-incompatible platelet transfusions are probably associated with significantly increased morbidity and mortality. The main advantage of PPC is lower costs. Preparation of PPC is however inevitably accompanied by substantial wastage of plasma and red cells. Only major supraregional blood transfusion centers can guarantee full-coverage supply with ABO-identical and Rh-compatible PPC. Whether APC are more effective than PPC and associated with fewer septic platelet transfusion reactions as shown in some but not all studies, has to be examined in future prospective controlled trials.  相似文献   

16.
The greatest transfusion-transmitted disease risk facing a transfusion recipient is that of bacterial sepsis. The prevalence of bacterial contamination in platelets and red blood cells is approximately 1 in 3,000 units transfused. The available data indicate that transfusion-associated sepsis develops after 1 in 25,000 platelet transfusions and 1 in 250,000 red blood cell transfusions. One of the most widely used strategies for decreasing bacterial sepsis risk is bacterial detection. A roundtable meeting of experts was convened during the XXVIII Annual Congress of the International Society of Blood Transfusion (Edinburgh, UK, July 2004) to provide a forum for experts to share their experiences in the routine bacterial detection of platelet products. This article summarizes the presentations, discussions, and recommendations of the panel. The data presented indicate that some of the current bacterial screening technology is useful for blocking the issuance of platelet units that contain relatively high levels of contaminating bacteria. Platelet units are usually released based on a test-negative status, which often become test-positive only upon longer storage. These data thus suggest that bacterial screening may not prevent all transfusion-transmitted bacterial infections. Two transfusion-transmitted case reports further highlighted the limitation of the routine bacterial screening of platelet products. It was felt that newer technologies, such as pathogen inactivation, may represent a more reliable process, with a higher level of safety. The panel thus recommended that the Transfusion Medicine community may need to change its thinking (paradigm) about bacterial detection, toward the possibility of the pathogen inactivation of blood products, to deal with the bacterial contamination issue. It was suggested, where permitted by regulatory agencies, that blood centers should consider adopting first-generation pathogen inactivation systems as a more effective approach to reducing the risk of transfusion-associated sepsis than some of the approaches currently available.  相似文献   

17.
INTRODUCTION: The Intercept Blood System, using InterSol as additive solution, is used for inactivation of contaminating pathogens in PCs, thus reducing the risk for transfusion transmitted infection and making it possible to prolong the storage period. This study aimed at investigating the ability of Intercept treated platelets to induce clot formation, as measured by coagulation time using free oscillation rheometry (FOR), and to compare with that of platelets in concentrates with the additive solution T-Sol or plasma. METHODS: Seventy-four single-donor platelet units were diluted in InterSol (n=27) or T-Sol (n=47) to a mean plasma concentration of 38%. The Intercept treatment was performed by addition of amotosalen HCl to the InterSol PCs followed by UVA irradiation and treatment with a compound adsorption device (CAD). Forty-six units were collected and stored in 100% plasma for comparison. Clotting time was measured by FOR in fresh PCs (within 26h after collection) after stimulation by a platelet activator. Soluble P-selectin was analysed as a marker of platelet activation in the Intercept and T-Sol PCs. RESULTS: The clotting time was shorter for Intercept treated platelets compared to platelets in T-Sol and plasma (p<0.05). There was no difference in clotting time between T-Sol and plasma PCs. Soluble P-selectin was higher for Intercept platelets than platelets in T-Sol (p<0.05). CONCLUSIONS: The platelets treated with the Intercept procedure had good clot promoting capacity.  相似文献   

18.

Background

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is unlikely to be a major transfusion-transmitted pathogen; however, convalescent plasma is a treatment option used in some regions. The risk of transfusion-transmitted infections can be minimized by implementing Pathogen Inactivation (PI), such as THERAFLEX MB-plasma and THERAFLEX UV-Platelets systems. Here we examined the capability of these PI systems to inactivate SARS-CoV-2.

Study Design and Methods

SARS-CoV-2 spiked plasma units were treated using the THERAFLEX MB-Plasma system in the presence of methylene blue (~0.8 μmol/L; visible light doses: 20, 40, 60, and 120 [standard] J/cm2). SARS-CoV-2 spiked platelet concentrates (PCs) were treated using the THERAFLEX UV-platelets system (UVC doses: 0.05, 0.10, 0.15, and 0.20 [standard] J/cm2). Samples were taken prior to the first and after each illumination dose, and viral infectivity was assessed using an immunoplaque assay.

Results

Treatment of spiked plasma with the THERAFLEX MB-Plasma system resulted in an average ≥5.03 log10 reduction in SARS-CoV-2 infectivity at one third (40 J/cm2) of the standard visible light dose. For the platelet concentrates (PCs), treatment with the THERAFLEX UV-Platelets system resulted in an average ≥5.18 log10 reduction in SARS-CoV-2 infectivity at the standard UVC dose (0.2 J/cm2).

Conclusions

SARS-CoV-2 infectivity was reduced in plasma and platelets following treatment with the THERAFLEX MB-Plasma and THERAFLEX UV-Platelets systems, to the limit of detection, respectively. These PI technologies could therefore be an effective option to reduce the risk of transfusion-transmitted emerging pathogens.  相似文献   

19.
BACKGROUND: Treatment with psoralens and UVA (PUVA) has been shown to be efficacious in eliminating the risk of virus transmission by platelet concentrates (PCs). It has previously been demonstrated that, during the inactivation of cell-free vesicular stomatitis virus (VSV) by aminomethyltrimethylpsoralen (AMT) and UVA in PCs, platelet function could be protected either by oxygen removal before irradiation or by inclusion of a type I free radical quencher, such as mannitol. STUDY DESIGN AND METHODS: Under previous PUVA treatment conditions for PCs (25 micrograms/mL AMT; 30 min UVA at 7 mW/cm2; 2 mM [2 mmol/L] mannitol), more than 6 log10 of added cell-free VSV was completely inactivated. In the current study, various PUVA conditions are evaluated for efficacy in inactivating other viral forms that could be present in PCs. Maintenance of platelet integrity (i.e., platelet number, solution pH, and aggregation response during initial storage after treatment) and kill of cell-associated VSV are examined. RESULTS: While cell-free viruses were inactivated efficiently under previous PUVA conditions, cell-associated VSV and the non-lipid-enveloped bacteriophage M13 were not. Effective inactivation of these viruses was achieved by raising the concentration of AMT to 50 micrograms per mL and extending the period of irradiation to 90 minutes (39 J/cm2). However, for maintenance of platelet integrity under these conditions, the prior removal of oxygen or the inclusion of compounds known to quench both type I and type II photoreactants (e.g., flavonoids such as rutin) was required. CONCLUSION: These findings suggest that the viral safety of PCs may be enhanced through treatment with AMT and UVA in the presence of flavonoids, and that flavonoid use may prove beneficial in other systems where oxygen-mediated damage occurs.  相似文献   

20.
Primary cytomegalovirus (CMV) infection is usually asymptomatic in immunocompetent patients but can cause serious life-threatening complications in immunocompromised CMV-seronegative patients, including patients receiving a bone marrow or peripheral blood stem cell transplant, recipients of some solid-organ transplants, and low-birth-weight neonates. Current recommendations for preventing transfusion-transmitted CMV (TT-CMV) infection in these patients include exclusive use of CMV-seronegative and/or leukoreduced cellular blood components (red blood cells and platelets) for transfusion. However, breakthrough cases of TT-CMV still occur. Despite improving the safety of blood components, testing remains a reactive approach to blood safety. In contrast, pathogen inactivation technologies offer a proactive approach with the potential to further improve blood safety. To reduce the risks associated with platelet transfusions, a photochemical treatment (PCT) process using a combination of the psoralen amotosalen HCl and long-wavelength UV light has been developed and introduced into clinical practice in Europe. PCT has been shown to result in greater than 5.9-log reductions in infectivity of human CMV in platelet concentrates and to prevent the transfusion transmission of murine CMV in a mouse transfusion model. Thus, PCT pathogen inactivation may play a role in further reducing the incidence of TT-CMV infection in patients who are at risk for serious CMV disease. Because PCT is a technology that targets nucleic acids, it also offers a proactive process for the inactivation of a broad range of viral, bacterial, and protozoan pathogens in addition to CMV.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号