You are on page 1of 12

Vox Sanguinis (2003) 85, 171–182

© 2003 Blackwell Publishing


ORIGINAL PAPER

Preclinical safety profile of plasma prepared using the


Blackwell Publishing Ltd.

INTERCEPT Blood System


V. Ciaravino, T. McCullough, G. Cimino & T. Sullivan
Cerus Corporation, Concord, California, USA

Background and Objectives The newly developed INTERCEPT Blood System for
plasma uses the addition of a new psoralen, amotosalen HCl (AMOTOSALEN), followed
by illumination with ultraviolet A light, to inactivate viruses, bacteria, protozoa and
leucocytes that may contaminate fresh-frozen plasma (FFP). Extensive toxicology
studies were performed to characterize the safety of the photochemical treatment
process for its intended use with plasma.
Materials and Methods The studies of general toxicology, safety pharmacology,
phototoxicity, reproductive toxicity and venous irritation, summarized in this review,
provide a comprehensive toxicology profile for photochemically treated 100% plasma.
Results No specific target organ toxicity (based on clinical or histological pathology),
phototoxicity, or reproductive toxicity was observed.
Conclusions The results of an extensive series of studies have demonstrated no toxi-
cologically relevant effects of photochemically treated 100% plasma prepared using
Received: 23 January 2003,
revised 25 June 2003,
the INTERCEPT Blood System for plasma.
accepted 28 June 2003 Key words: amotosalen, INTERCEPT Blood Systems, pathogen inactivation, toxicology.

factor deficiencies may require plasma on a chronic basis


Introduction during their lifetimes. In contrast to severely affected patients,
patients with mild congenital coagulation deficiencies (5–
Clinical uses of fresh-frozen plasma 25% of normal) may be treated intermittently with FFP, as
The demand for fresh-frozen plasma (FFP) has increased as prophylaxis, in preparation for invasive procedures or during
a response to the growing needs of patients who require FFP active bleeding. Correction of acquired complex coagulation-
and cryoprecipitate derived from FFP in support of coagulop- factor deficiencies generally requires short-term treatment
athy, either with or without bleeding, during large-volume with 15 ml/kg (4 units for a small adult, each unit containing
blood transfusion, and for rapid reversal of anticoagulation 250–300 ml from a single blood-donor). As many as 50 FFP
[1]. In addition, FFP is indicated for the treatment of anti- units may be required to treat a single transfusion episode
thrombin III, Protein C or Protein S deficiency when a con- associated with a major surgical procedure, such as liver
centrate is not available, and for therapeutic plasma exchange transplantation [3]. Use of FFP in total plasma exchange
to treat thrombotic thrombocytopenic purpura (TTP) [2]. (TPE) therapy for the treatment of TTP has been reported to
Assessment of preclinical safety requires the design of average 21·5 ± 7·8 l in a single cycle of TPE for an acute
studies to cover the anticipated clinical exposures. Patients episode of TTP [4].
with severe (i.e. < 1% of normal) congenital coagulation
Risk of transfusion-transmitted illness

Correspondence: Tim McCullough, PhD, Cerus Corporation, 2411 Stanwell Despite improvements in donor screening and testing, trans-
Drive, Concord, CA 94520, USA mission of viral diseases by labile blood products cannot be
E-mail: tim_mccullough@cerus.com totally excluded. Current testing is unreliable during the

171
172 V. Ciaravino et al.

Table 1 Test materials used for preclinical safety evaluation programme

Photochemically treated 100% plasma Photochemically treated 35% plasma


a a
With CAD No CAD With CADa No CADa Amotosalen

Acute toxicity X X X X
≤ 1-month toxicity X X X
3-month toxicity X X X X
Safety pharmacology X X X
Reproductive toxicity X X X X
Genotoxicity X X
Carcinogenicity X X X
Vein irritation X X
Phototoxicity X (X)b X (X)b X

a
Compound adsorption device (CAD) used to remove residual amotosalen and free photoproducts.
b
(X) = partial CAD treatment.

window period between infection and seroconversion of a containers. As methylene blue is a planar molecule, it has
donor; however, window periods have been shortened by the affinity for binding to nucleic acids. After activation with
use of genomic testing. In current practice, in many European visible light, methylene blue transfers its energy to molecular
countries where state-of-the-art donor screening and selection oxygen, generating singlet oxygen. Singlet oxygen is the
procedures have been introduced, ‘window’ donations are rare. active virucidal agent in the methylene blue process. It is
The frequencies of seroconversion from repeat donations in known to modify guanosine residues in nucleic acids, as well
Europe have been reported to be 1 : 2·3 × 106 for human as some viral envelopes, through reactions with amino acids.
immunodeficiency virus (HIV), 1 : 0·4 × 106 for hepatitis B These methods are in contrast to Helinx technology, which
virus (HBV) and 1 : 0·62 × 106 for hepatitis C virus (HCV) [5]. results in monoadducts and crosslinks between nucleic acids
Estimates for the risk of viral transmission were higher in and the intercalated amotosalen (see the Materials and
Germany and France (e.g. 1 : 1 × 106 for HIV). Similar, albeit Methods, below).
somewhat higher, values have been found in the United
States [6–9].
INTERCEPT Blood System for plasma
In view of the probable exposure to multiple donors asso-
ciated with the use of plasma in the treatment of congenital More recently, the combination of the new psoralen, amoto-
and acquired coagulopathies, especially in the case of TPE salen, and ultraviolet A (UVA) (320–400 nm) has been shown
therapy for TTP, the risk for a patient to incur a transfusion- to be highly effective for inactivating bacteria, viruses,
transmitted illness increases accordingly. protozoa and passenger leucocytes that may contaminate
Several measures have been adapted to prevent or elimi- plasma [13,14]. This procedure has been developed for single-
nate potential infectious risks in the blood supply and they donor products and has been shown to preserve the thera-
have been introduced in the production process of blood and peutic effectiveness of the plasma product [15–22].
plasma products [10–12]. In the case of FFP, the treatment Addition of chemicals to blood products for the inactiva-
used for viral reduction of pooled plasma products is solvent/ tion of contaminants is a potential cause for concern because
detergent; for single-donor products methylene blue is used of the possible risks associated with the introduction of
[10]. The solvent/detergent method is used to treat pools of chemical agents into therapeutic preparations that will be
plasma products. The pool is initially exposed to chemicals administered to patients. With respect to psoralens [23], the
that denature the coats of enveloped viruses, the chemicals combination of some compounds in this class and UV light
are subsequently removed by chromatography, and then has been associated with genotoxicity, carcinogenicity,
individual plasma units are prepared and allocated from the phototoxicity and other adverse effects [24–30].
pool. This methodology is very effective for lipid-enveloped
viruses, but has limited or no effect on non-enveloped
Preclinical safety
pathogens. Photodynamic viral inactivation of plasma can be
mediated by visible light in the presence of the phenothiazine This article reviews results of the preclinical toxicology
dye, methlyene blue. The commercial procedure is applied to programme carried out in support of the INTERCEPT Blood
individual plasma units, from single donors, in their storage System for plasma (Table 1). The programme consists of

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171–182


Preclinical safety profile 173

studies that not only evaluated photochemically treated 100%


plasma, but also evaluated the safety of the following test
articles: amotosalen alone and photochemically treated 35%
plasma (conducted to support the safety of the INTERCEPT
Blood System for platelets). In the photochemical treatment
process for platelets, a transfusion unit of platelets is sus-
pended in 35% plasma and 65% PAS III (sodium chloride/
acetate/citrate in phosphate buffer). In the photochemical
treatment process for plasma, a transfusion unit contains 100%
plasma. Studies conducted with photochemically treated 35%
plasma include: single-dose (acute) and multiple-dose (up to
13 weeks) intravenous toxicity, safety pharmacology (central
nervous system [CNS], renal and cardiovascular), male and
female reproductive toxicity, a series of in vitro and in vivo
mutagenicity studies, carcinogenicity in transgenic mice
heterozygous for the p53 tumour-suppressor gene mutation,
vein irritation and phototoxicity. A complete review of the
studies with amotosalen and photochemically treated 35%
plasma has been described in previous publications [31,32].

Materials and methods Fig. 1 The INTERCEPT Blood System for plasma. Collected plasma is mixed
with amotosalen and the mixture is placed in an ultraviolet A (UVA)
illumination device. The contents are then passed through a Compound
Photochemical treatment
Adsorption Device (CAD) for reduction of amotosalen and free
The INTERCEPT Blood System for plasma employs Helinx™ photoproducts. After CAD treatment, the plasma is placed
technology (Fig. 1), which uses amotosalen (a psoralen) and in a final storage container until transfusion.
UVA light, in an ex vivo photochemical treatment process,
to inactivate viruses, bacteria, protozoa and leucocytes in in laboratory animals and in in vitro systems. Study selection
plasma. The structure and photochemistry of amotosalen and design were in accordance with International Conference
has been described previously [33]. The photochemical treat- on Harmonization (ICH) guidelines on technical requirements
ment process begins with the preparation of a plasma unit in for registration of pharmaceuticals for human use. All studies
a plastic storage container (250-ml plasma transfusion unit). were conducted in compliance with Good Laboratory Practices
After addition of amotosalen to a nominal concentration or in accordance with prevailing professional standards. All
of ≈ 50 µg/ml (150 µM), the mixture is illuminated with studies employing animals were perfored in laboratories that
3 Joule/cm2 UVA (320–400 nm) and horizontal agitation fully complied with the requirements of the US Department
is performed concurrently. This causes monoadduct and of Agriculture and the American Association for the Accred-
crosslink formation between nucleic acids and intercalated itation of Laboratory Animal Care.
amotosalen, and the resultant inactivation of pathogens that
depend on nucleic acid replication for proliferation. In the
Establishment of safety margins
absence of UVA light, the amotosalen behaves as a reversible
intercalator and no chemistry occurs. The photochemically Intravenous (i.v.) infusions of photochemically treated
treated plasma is then passed through a Compound plasma, with and without CAD treatment, were carried out to
Adsorption Device (CAD) to remove residual amotosalen and evaluate potential toxicity. Analysis of the plasma level of
unbound photoproducts and then transferred to the final amotosalen was conducted to evaluate toxicokinetic para-
container for storage at −18 °C, according to standard meters in toxicology studies. To establish large safety margins
procedures for FFP. After treatment with the CAD, the amount for amotosalen and photoproduct exposure over clinical
of residual amotosalen and free photoproducts in a 250-ml applications, toxicology studies were designed to increase
plasma transfusion unit are ≈ 60 µg and 170 µg, respectively. the dose volumes administered, increase frequency of admin-
The clinical exposure to amotosalen and its free photo- istration, and to include groups given dose formulations
products for a 1000-ml plasma transfusion would therefore be prepared without CAD treatment. For toxicology studies
≈ 4 µg/kg and 11 µg/kg, respectively, on a body weight basis conducted for the platelet programme, transfusions were
for a 60-kg individual. The toxicology programme support- administered over a 1-h period. For the 100% plasma studies,
ing the INTERCEPT Blood System for plasma was conducted transfusions were administered over a 2-h period.

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171– 182
174 V. Ciaravino et al.

The maximum dose volumes of photochemically treated test and i.v. administration of large volumes of species-specific
articles employed in the toxicology studies were 25–80 ml/kg homologous plasma was not known. While there is some
for single-dose studies and 25–60 ml/kg in multidose studies. knowledge about canine blood types, very little is known
In the plasma programme, a dose volume of 60 ml/kg was about rodent blood types. Moreover, sterile rodent plasma and
given to rats in a 1-month study. Owing to vehicle effects sterile, blood-typed canine plasma are very difficult to obtain
observed in that study, a lower dose volume was selected for in the quantities needed to conduct toxicology studies. Accord-
subsequent studies. Therefore, depending upon the frequency ingly, the toxicology programme progressed step-wise through
and duration of dosing, 50 ml/kg was selected for use in the 7-, 14-, 28- and 90-day toxicity studies in rats and dogs.
28-day phototoxicity study (where rats were dosed three Based on the clinical use of plasma, the maximum duration
times per week), 50 ml/kg was selected for use in the teratology of toxicity studies for the toxicology programme was set at
study (where rats were dosed daily for 12 days), and 40 ml/kg 3 months. Clinically, plasma units are primarily administered
was selected for daily dosing in the 3-month study. In com- in support of a single medical event. While Cerus Corporation
parison, the plasma volumes of rats, rabbits and monkeys are is not aware of any published comprehensive review of the
≈ 35–40 ml/kg, and the plasma volume of dogs is ≈ 50 ml/ demographics of FFP use, a Cerus survey of the literature
kg. Therefore, the acute doses were approximately twice the indicated that most patients (> 97% of the patients given FFP)
plasma volume of the species used (greater than twice for receive a total of 3–5 units of plasma (600–1000 ml, assum-
rats, and less than twice for dogs), and the daily doses for the ing 200 ml as an average volume for a unit of FFP), given as
multidose studies were ≥ 50% of the plasma volume of the a single dose in support of an acute event such as surgery,
species used in the various studies. The maximum dose trauma, or coumadin reversal (W. Greenmore, personal com-
volumes employed in the toxicology studies were considered munication). There is a small patient population (< 2% of the
the maximum feasible dose volumes of a protein-containing patients given FFP) that receives multiple doses of FFP over
formulation that would not cause undue stress to the renal 2–4 weeks in support of liver transplantation. At two centres
and cardiovascular systems (especially in consideration of surveyed by Cerus, these patients received an average total
the protein load) and thereby compromise the studies. of 30 or 38 units of FFP, respectively (6–8 l). There is also a
very small patient population (< 1% of the patients given
FFP) that receives plasma exchanges (approximately 10 units
Rationale for toxicology studies
of FFP) on several occasions over a 2–4-week period (4–22 l
The following sections summarize the strategy for each study total) for chronic disorders, including TTP, Guillain–Barré
category. syndrome and myasthenia gravis. Of these patients, a very
small number may have recurrent therapy.
Rationale for 100% plasma studies In acute studies, i.v. infusions of up to 80 ml/kg/h of
To support the clinical use of photochemically treated 100% photochemically treated 100% plasma was administered as a
plasma, specific toxicology studies were chosen that were single 2-h dose, and the rats were evaluated for a 14-day
considered critical in the overall evaluation of the programme period. At the end of the 14 days, the rats were killed and
and used 100% plasma, in contrast to the previous studies examined grossly. A set of tissues was preserved but not
conducted with 35% plasma. As a 100% plasma test article processed for histopathological examination. In repeated-
was being administered in the present toxicology studies, an dose studies, i.v. infusions of up to 60 ml/kg of photochemi-
evaluation of potential vein irritation was important. Acute cally treated 100% plasma was administered over 2 h daily
and safety pharmacology studies were performed to evaluate for up to 13 weeks. Clinical observations, body weights, food
overall potential acute systemic toxicity or any effects on intakes and clinical pathology evaluations (haematology,
cardiovascular or renal physiology. Even though no photo- chemistry and urinalysis) were conducted at scheduled inter-
toxicity or reproductive toxicity was elicited with photo- vals. Ophthalmoscopic examinations (indirect and slit lamp)
chemically treated 35% plasma, it was important to evaluate were conducted at scheduled intervals. At the completion of
the higher residual amotosalen levels after photochemical scheduled duration of treatment, animals were killed and
treatment. Teratology was specifically selected because of subjected to gross pathological examinations. In the 13-week
the importance of potential toxicity on the developing study, a subset of the animals were assigned to a 1-month
fetus. Finally, a study of the longest duration, 3 months, observation period and then killed. A set of tissues from each
was selected to evaluate the potential toxicity of prolonged animal was preserved and evaluated histopathologically.
administration of photochemically treated 100% plasma.
Safety pharmacology
Acute and repeated-dose toxicity Safety pharmacology studies were conducted to investigate
Early in the toxicology programme, many technical challenges any potential adverse effects of amotosalen and associated
had to be overcome, as the feasibility of the sterile preparation photoproducts on physiological functions. In the renal safety

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171–182


Preclinical safety profile 175

pharmacology study, a single i.v. infusion (80 ml/kg for 2 h) Chromosome aberration assay. This in vitro assay detects
of a vehicle control (100% rat plasma) or photochemically chromosome damage. The Chinese hamster ovary (CHO) cell
treated 100% plasma with or without CAD treatment, was line, with a well-defined karyotype, was used. The cells were
given to rats. Urine and blood were collected both before assessed for structural chromosome aberrations (chromosome
and after dosing. Creatinine clearance, urinalysis and clin- and chromatid gaps, breaks, rings, deletions, rearrangements,
ical chemistry parameters were measured. The kidneys were etc.) after exposure to the test article and to positive controls.
removed and weighed.
In the cardiovascular study, an i.v. infusion (80 ml/kg for Mouse lymphoma assay. This in vitro assay detects point
for 2 h) of vehicle control (100% dog plasma) or photochem- mutations in the mouse lymphoma L5178Y cell line at the
ically treated 100% plasma, with and without CAD treatment, thymidine kinase (TK) gene. This assay is based on the fact
was given to groups of three male beagle dogs. Each dog that mutant TK cells are resistant to the addition of the pyri-
received control or treated formulation in random order with midine analogue, trifluorothymidine (TFT) and synthesize
1 h between infusions. Electrocardiogram recordings were their DNA de novo. Non-mutant cells die in the presence of TFT.
obtained at predetermined intervals throughout the study. After exposure of cells to the test article or to positive con-
Mean cardiac output, stroke volume, total peripheral resistance trols, the mutation is given time to express and then the cells
and pulmonary wedge pressure were recorded throughout are placed in TFT. Only mutant cells survive and are counted.
the testing period.
In vivo mouse micronucleus assay. This in vivo assay detects
Reproductive toxicology chromosome damage in the cells of the bone marrow of mice.
Clinically, women of childbearing potential and pregnant Polychromatic erythrocytes (PCE) – precursor cells to circu-
women may receive plasma transfusions. In a developmental lating red blood cells – are assessed for the presence of
toxicity (teratology) study, daily intravenous infusions of nuclear material (micronuclei) that may be present after
50 ml/kg plasma, with and without CAD treatment, were given genotoxic interaction with the test article or positive control.
to groups of 25 pregnant rats daily during organogenesis (days These micronuclei represent parts of chromosomes or whole
6 –17 of gestation). On gestation day 21, dams were euthanized chromosomes left behind as the PCE ejects its nucleus in the
and their fetuses were given teratological examinations. normal progression of development to a mature erythrocyte.

Genotoxicity testing In vivo unscheduled DNA synthesis (UDS) assay. This in vivo
Three in vitro and two in vivo genotoxicity assays were used assay detects any type of DNA damage in hepatocytes in rats.
to assess the genotoxicity of photochemically treated platelets Normally, liver cells do not undergo DNA synthesis or division.
and amotosalen alone; the results have been presented Using a 14C-thymidine label, no significant incorporation of
previously [31,32]. No specific studies were completed using label in liver cells will occur unless the test article or positive
photochemically treated 100% plasma. In vitro genotoxicity control damages the DNA and thus stimulates the repair system.
studies were conducted in the presence and absence of
metabolic activation. This activation involves co-treatment Carcinogenicity
of the test article with a rat liver microsome preparation to The potential carcinogenicity of amotosalen and its photo-
assess whether the test article is metabolized into products products was evaluated in p53 transgenic mice. The p53
resulting in more or less genotoxicity. The studies carried out tumour-suppressor gene has received attention because of its
are described below. frequent alteration in a wide variety of human tumours.
Fifty-per cent or more of all human tumours have detectable
Ames assay. The Ames Salmonella/microsome assay detects p53 genetic lesions [34]. A number of animal models have
point mutations in vitro in a series of histidine-requiring been developed and studied for the role of p53 in mammalian
auxotrophs of Salmonella typhimurium and Escherichia coli. tumorigenesis [35]. These gene-knockout models consist of
Tester strains TA98 and TA1537 are reverted from histidine mice that are heterozygous for the p53 gene. In a study
dependence (auxotrophy) to histidine independence (pro- assessing the spontaneous tumorigenesis in p53-deficient
totrophy) by frameshift mutagens. These two strains have mice, 60 p53 homozygous and 97 heterozygous mice were
different mutations in their histidine genes and thus have examined, twice weekly, for tumours [36]. By the age of
different sensitivities to mutagens. Tester strain TA1535 is 4·5 months, ≈ 50% of the homozygotes had developed
reverted by mutagens that cause base pair (bp) substitutions. tumours and, by 10 months of age, all of the mice had died
Tester strain TA100 is reverted by mutagens that cause both or developed tumours (Fig. 2). In contrast, the p53 heterozy-
frameshift and bp substitutions. E. coli strains WP2 uvrA gotes remained healthy for a longer period of time. By
[pKM101] and WP2 [pKM101] are sensitive to bp substitution 12 months of age, only 8% had tumours. However, after
mutations. Positive controls are included in each study. 12 months of age the rate of tumour development accelerated

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171– 182
176 V. Ciaravino et al.

Fig. 2 Tumour incidence in p53-deficient mice , surviving wild type mice (p53+/+); , heterozygous mice (p53+/–); , homozygous mice (p53–/–). (Taken
from Harvey et al. [36]).

and, by 18 months of age, > 50% of the mice had developed of UV light, including sunlight [27,37,38]. These responses
tumours. Control mice of the wild-type allele did not develop were studied in animals administered photochemically treated
tumours over the 18-month period. Therefore, in this assay, 100% plasma. The phototoxicity studies employed exagger-
heterozygous mice were used between the ages of 7–10 weeks ated conditions in that simulated sunlight exposure was
at the initiation of dosing. At the end of the 26-week dosing intense and was given at about the time of the peak plasma
period, spontaneous tumorigenesis is low, thus avoiding level of the test compound; in addition, albino animals were
interference in the interpretation of the results of the study. employed because they are more sensitive than pigmented
In the assay, designed to assess the potential of amotosalen animals.
to develop tumours, groups of 20 mice/gender were given
high doses of i.v. amotosalen and its photoproducts (up to
Results
250 times the anticipated clinical exposure in a 1-l FFP trans-
fusion) three times weekly for 6 months. The mice were then
Acute and repeated-dose toxicity
euthanized and their tissues examined histologically.
Several acute and repeated-dose studies were conducted in
Vein irritation rats using 100% homologous plasma. A summary of the studies
Because plasma is administered intravenously, information is presented in (Table 2). The results were consistent with,
on the vascular irritation potential of photochemically treated and supplemented the data obtained using, photochemically
100% plasma (without CAD treatment to have maximal treated 35% plasma. Neither acute exposure nor prolonged
exposure to residual amotosalen and its free photoproducts) 90-day daily dosing of photochemically treated 100% plasma
was examined in a conventional model system in the rabbit. resulted in any treatment-related toxicity in the studies.
Vein irritation was assessed by histopathological evaluation
of the vascular tissue.
Safety pharmacology
Phototoxicity Treatment with photochemically treated 100% plasma did
Psoralens are known, in certain circumstances, to be capable not result in any functional or gross pathological changes in
of eliciting dermal and ocular phototoxicity in the presence the renal system of the rat, which was consistent with, and

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171–182


Preclinical safety profile 177

Table 2 Single- and repeated-dose toxicity studies

Test Species Solutions used Dosing Findings

Single-dose Rat Photochemically treated 25 and 80 ml/kg Not toxic, all rats survived, and no treatment-
100% plasma with CAD for 2 h related clinical or gross pathologic changes
treatment were observed after 14 days
28-day Rat Photochemically treated 20 and 60 ml/kg Not toxic, all rats survived, and no treatment-
100% plasma with CAD for 2 h daily related clinical, clinical, gross or
treatment histopathologic changes were observed
13-week Rat Photochemically treated 40 ml/kg for No abnormal findings were observed at the
100% plasma, with and 2 h daily end of dosing or in groups of rats after
without CAD treatment a 1-month observation period

CAD, compound adsorption device (used to remove residual amotosalen and free photoproducts).

supplemented the data obtained with, photochemically treated sample. These human platelets, multiply processed by photo-
35% plasma. chemical treatment without CAD treatment, were genotoxic in
Administration of photochemically treated 100% plasma both the Ames assay (Salmonella typhimurium strain TA1537;
did not result in any electrocardiographic or haemodynamic in the absence of metabolic activation) and in the CHO cell assay.
effects on the cardiovascular system of the dog. These results These effects were attributed solely to the high concentration
were consistent with, and supplemented the data obtained of residual amotosalen and do not reflect clinical exposures.
with, photochemically treated 35% plasma. Amotosalen alone was genotoxic in vitro in the Ames
Although no specific studies were conducted to evaluate assay in Sal. typhimurium strain TA1537, in the mouse lym-
respiratory or gastrointestinal effects, no effects were seen in phoma TK assay, and in the CHO cell assay for chromosome
acute rat toxicity studies following i.v. infusion of 80 ml/kg aberrations. In the Ames assay, amotosalen was not genotoxic
of photochemically treated 100% plasma, with or without in any other strain tested, including Sal. typhimurium TA98,
CAD treatment. TA100 and TA1535 and E. coli WP2 uvrA (pKM101) and WP2
(pKM101). These are the expected results for an intercalator
in an in vitro assay (intercalation is the mechanism for amo-
Reproductive toxicity
tosalen’s specificity for targeting nucleic acids).
Administration of photochemically treated 100% plasma did Metabolic activation with a hepatic S9 fraction reduced the
not cause maternal or fetal developmental toxicity. These genotoxic potential of both amotosalen and the multiply
results were consistent with, and supplemented the teratology processed photochemically treated human platelets without
study conducted with, photochemically treated 35% plasma CAD treatment, in all in vitro genotoxicity studies. These
[29,30]. Additionally, for the photochemically treated 35% results are consistent with those obtained from in vivo
plasma studies, a complete series of reproductive toxicology genotoxicity assays (the hepatic unscheduled DNA synthesis
studies was conducted, which included male and female assay in rats and the micronucleus assay in mice), in which
fertility studies in rats, an additional teratology study in the mammalian metabolic pathways were intact and functional,
rabbit, and a peri- postnatal reproductive development study and high doses of i.v. amotosalen were not genotoxic.
in the rat, none of which resulted in any reproductive toxicity The ratios between the doses of amotosalen that were
as a result of administration of photochemically treated 35% genotoxic in a given assay and expected clinical peak plasma
plasma administered at 25 ml/kg. levels (12 ng/ml) were very high in all the assays carried out
(Table 4). Among the in vitro assays, in which a positive result
was obtained, the lowest ratios were found in the CHO cell
Genotoxicity testing
studies (> 167-fold without metabolic activation and > 2000-
Photochemically treated human platelets processed as in clin- fold with metabolic activation). The in vivo assay results were
ical use, but without CAD treatment, indicated no genotoxicity negative; the ratio of the highest non-genotoxic dose to the
in any in vitro or in vivo assay (Table 3). In these studies, the expected clinical exposure level were ≥ 3000-fold.
residual amotosalen levels were far below the threshold con-
centrations for the positive effects observed with amotosalen
Carcinogenicity
alone (≥ 5 µg/ml). To increase residual amotosalen and photo-
products in a platelet concentrate unit, amotosalen addition There was no evidence of an increased incidence of tumours in
and UVA illumination were repeated 25 times in a single any of the mice treated with amotosalen or photochemically

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171– 182
178 V. Ciaravino et al.

Table 3 Genotoxicity studies

Test Test system Solutions used Findings

Ames assay Salmonella typhimurium Photochemically treated human Not genotoxic at the highest dose tested:
and Escherichia coli a platelets without CAD treatment 2–2·5 µg/ml residual amotosalen and
(± S9) 8–10 µg/ml photoproducts

Ames assay Salmonella typhimurium Photochemically treated human In the presence of metabolic activation;
and Escherichia coli a platelets exposed to 25 cycles not genotoxic at the highest dose tested:
of amotosalen addition and 62 µg/ml residual amotosalen
illumination (± metabolic activation) In the absence of metabolic activation; positive
in strain TA1537 at mean concentrations of
residual amotosalen of 35 µg/ml

Ames assay Salmonella typhimurium Psoralen amotosalen In the presence of metabolic activation;
and Escherichia coli a positive for strain TA1527 at 44 µg/ml

Chromosome aberration CHOb cells Photochemically treated human In the absence of metabolic activation;
assay platelets without CAD treatment positive at 103 µg/ml
(± metabolic activation) Not genotoxic at the highest dose tested; 2–
2·5 µg/ml residual amotosalen and 8–10 µg/ml
photoproducts

Chromosome aberration CHOb cells Psoralen amotosalen In the presence of metabolic activation;
assay positive at averag econcentrations of 46 µg/ml
In the absence of metabolic activation;
positive at ≥ 5 µg/ml

Chromosome aberration CHOb cells Photochemically treated human In the presence of metabolic activation;
assay platelets exposed to 25 cycles of positive at > 15 µg/ml residual amotosalen
amotosalen addition and In the absence of metabolic activation;
illumination (± metabolic activation) positive at 7·5 µg/ml residual amotosalen

Mouse lymphoma assay L1578Y/TK+/– cells Photochemically treated human Not genotoxic at the highest dose tested;
platelets without CAD treatment 2–2·5 µg/ml residual amotosalen and
(± metabolic activation) 8–10 µg/ml photoproducts

Mouse lymphoma assay L1578Y/TK+/– cells Psoralen amotosalen In the presence of metabolic activation;
positive at > 7·5 µg/ml
In the absence of metabolic activation;
negative at > 65 µg/ml

In vivo UDSc assay Rat Photochemically treated human Not genotoxic at 200 µg/kg residual amotosalen
platelets without CAD treatment

In vivo UDSc assay Rat Psoralen amotosalen Not genotoxic at doses as high as 34 mg/kg

In vivo micronucleus assay Mouse Photochemically treated human Not genotoxic at 200 µg/kg residual amotosalen
platelets without CAD treatment

In vivo micronucleus assay Mouse Psoralen amotosalen Not genotoxic at doses as high as 66 mg/kg

a
Salmonella strains TA98, TA100, TA1535 and TA1537; Escherichia coli strains WP2 uvrA [pKM101] and WP2 [pKM101].
b
Chinese hamster ovary cells.
c
Unscheduled DNA synthesis.
CAD, compound adsorption device (used to remove residual amotosalen and free photoproducts).

treated 35% plasma [31]. These results, like those of the


Venous irritation
genotoxicity studies, support the conclusion that plasma
prepared using the INTERCEPT Blood System does not In a specific study designed to assess vascular irritation
present a carcinogenic risk to patients. potential, photochemically treated 100% plasma, without

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171–182


Preclinical safety profile 179

Table 4 Relationship of clinical exposure to exposures in genotoxicity


Phototoxicity
studies
Two studies were conducted to evaluate the phototoxic
Multiples of potential of photochemically treated 100% plasma: a single-
Study type clinical exposurea
dose study and a 1-month study. In the single-dose study,
male and female rats were administered 2-h i.v. infusions
Photochemically treated 35% plasma
of 20 or 80 ml/kg/dose of photochemically treated 100%
Ames
Non-activated/activated (all strains) > 208b
plasma. The control group was given 2-h infusions of 80 ml/
Ames (multidose illuminate) kg/dose of 100% plasma that had not received photochemical
Non-activated (TA 1537) 2000 treatment. At ≈ 15 min after dosing (control) or at ≈ 15 min,
Non-activated (all other strains) > 5167b 6 h or 24 h after dosing, a depilated, dorsal skin site and both
Activated (all strains) > 5167b eyes were exposed to an estimated 0·5 minimal erythema
Mouse lymphoma dose (MED) of ultraviolet radiation (UVR) provided by a
Non-activated/activated > 167b xenon-arc solar simulator. No evidence of dermal or ocular
CHO cell phototoxicity occurred in the study.
Non-activated/activated > 167b
In the 1-month study, up to 50 ml/kg photochemically
CHO Cell (multidose illuminate)
treated 100% plasma, administered over 2 h, was given three
Non-activated 250
times/week for 4 weeks to male and female rats. The rats were
Activated > 1083c
Rat hepatocyte UDS (i.v. bolus) > 50d
exposed to simulated sunlight following the third dose each
Mouse micronucleus (i.v. bolus) > 50d week. Additionally, groups of rats were administered photo-
Amotosalen chemically treated 100% plasma that was partially CAD treated,
Ames such that residual amotosalen levels were as high as 10 µM,
Non-activated (TA1537) 1417 10 times higher than the clinical concentration of ≈ 1 µM in
Activated (TA1537) 3750 a plasma unit. No evidence of dermal or ocular phototoxicity
Non-activated/activated > 595 250 occurred in the study. In both studies, evidence of dermal photo-
(all other strains) toxicity was present in the positive control, 8-methoxypsoralen
Mouse lymphoma
(8-MOP)-treated females and males (10 µg/kg). 8-MOP is cur-
Non-activated 417
rently used for therapy of chronic refractory psoriasis and
Activated > 5417b
cutaneous T-cell lymphoma.
CHO cell
Non-activated 167
Activated 2000 Neoantigenicity
Rat hepatocyte UDS (i.v. bolus) > 3000b
Mouse micronucleus (i.v. bolus) > 3000b The photochemical treatment of plasma leads to the formation
of covalent adducts with plasma macromolecules. Because
a
Ratio of highest non-genotoxic amotosalen dose level (in vitro studies) or the adducts are primarily associated with lipids instead of
peak amotosalen plasma level (in vivo studies) to the clinical peak plasma proteins, their neoantigenic potential is considered low.
level of amotosalen (12 ng/ml) after transfusion of 1-l of fresh-frozen
Nevertheless, the neoantigenic potential of photochemically
plasma (FFP).
b treated plasma has been extensively studied in preclinical
Negative at the highest doses tested in all assays.
c and clinical studies. Toxicology studies (especially the 1-
Negative at the highest dose tested in one or more assays.
d
Negative at the highest feasible intravenous (i.v.) dose level. and 3-month toxicity studies) also provided information
relevant to neoantigenicity. In these toxicity studies, there
was no clinical evidence of sensitization resulting in his-
CAD treatment, was non-irritating to the ear vein of the tamine release. Histopathologically, there was no evidence
rabbit when administered intravenously by infusion for 2 h in any study of lymphocyte proliferation, immune-tissue
at a dose volume of 20 ml/kg. stimulation or immune-complex deposition. There was also
In a 13-week toxicity study in rats, there was no evidence no evidence of any immunological response to photochemi-
of infusion-site venous irritation when photochemically cally treated plasma or coagulation factors, as activated
treated 100% plasma, with or without CAD treatment, was partial thromboplastin time (aPPT) and prothrombin time
administered intravenously via the tail vein at 40 ml/kg for (PT) values were unaffected in animals that were dosed
2 h daily for 13 weeks. daily for up to 3 months with photochemically treated
In these single- and repeat-dose studies, the daily infusion 100% plasma. These data further support the absence of a
volume in all species was up to a 3·5-fold multiple of the neoantigenic response to photochemically treated 100%
clinical infusion volume of 1 l (≈ 17 ml/kg body weight). plasma.

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171– 182
180 V. Ciaravino et al.

Fig. 3 Safety margins in toxicology studies obtained with the INTERCEPT Blood System for plasma, presented as multiples of the clinical exposure of amotosalen
(4 µg/kg).

showed a half-life of amotosalen ranging from 6 to 9·1 h


Discussion and conclusions (from studies conducted with 100% and 35% plasma) com-
The toxicology studies conducted with photochemically pared to 8·9 h in humans (determined in a phase 1 clinical
treated 100% and 35% plasma used the same amotosalen con- trial). Safety margins in the toxicology studies are shown in
centration (150 µM) and UVA exposure (3 J/cm2). Analytical Fig. 3 and are represented by the types of studies conducted
characterization high pressure liquid chromatography (HPLC) and the multiples of clinical exposure in these studies. No
and liquid chromatography/mass spectroscopy/mass spectro- specific target-organ toxicity (clinical pathology or patho-
scopy (LC/MS/MS) of 100% and 35% plasma after amotosalen logy) occurred in any study conducted with 100% or 35%
photochemical treatment indicated that the same residual com- plasma. The principal findings for amotosalen alone were
pounds (amotosalen and free photoproducts) were present in CNS observations (used at concentrations 6250-fold than the
both preparations, and no materials were characterized in estimated clinical exposure of 4 µg/kg), electrocardiographic
one preparation but not in the other. Toxicology studies with observations (10 000-fold), and phototoxicity (250-fold). With
photochemically treated 35% plasma are therefore applicable respect to mutagenicity, the ratios of the highest non-mutagenic
to the preclinical safety evaluation of photochemically treated dose levels to clinical exposures were 167- and 2000-fold
100% plasma. To supplement the studies carried out with 35% higher for the in vitro studies, without and with metabolic
plasma, the following toxicology studies were conducted activation, respectively. For in vivo assays (which were all
with photochemically treated 100% plasma: acute systemic negative), the ratios were at least 3000-fold. Based on the ext-
toxicity; 1- and 3-month toxicity; safety pharmacology remely large safety margins (up to 10 000 times the estimated
(a cardiovascular study in dogs and a renal study in rats); clinical exposure to amotosalen), the CNS and electrocardio-
developmental toxicity; vein irritation; and single-dose graphic observations are not considered to be of any physio-
and 4-week multiple-dose phototoxicity studies. Toxicology logical relevance for the proposed use of the CAD-treated,
studies completed with photochemically treated 100% plasma, photochemically treated 100% plasma. Based on the absence
as well as photochemically treated 35% plasma and amoto- of any phototoxicity with photochemically treated 100%
salen alone, are presented in Table 1. plasma, and the large safety margin for amotosalen alone
The potential toxicity of amotosalen and photochemically (1000-fold), photochemically treated 100% plasma is not
treated 35% plasma was extensively studied in > 80 studies, considered to be phototoxic for its proposed use. Based on
using the same approach as a new pharmaceutical study and the mechanism for in vitro mutagenicity, the extremely large
according to ICH guidelines [31,32]. In toxicology studies con- ratios for in vitro mutagenicity and the absence of any muta-
ducted with illuminated 100% plasma, the levels of amoto- genicity for in vivo assays, the in vitro mutagenicity findings
salen administered were well above the exposure a patient are not considered to be of any toxicological significance for
would receive in clinical applications (≈ 4 µg/kg of residual the proposed use of photochemically treated 100% plasma.
amotosalen in a 1-l FFP transfusion based on a body weight Furthermore, the possible concern regarding genotoxicity
of 60 kg). Pharmacokinetic data from toxicology studies relates to the potential for a carcinogenic risk. No evidence

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171–182


Preclinical safety profile 181

of carcinogenicity occurred in a p53 carcinogenicity assay, 13 Alfonso R, Lin C, Dupuis K, Corten L, Reames A, Londe H, Chen
which is a highly sensitive model for genotoxic carcinogens. C, Yip J, Cook D, Smyers J, Clarke S, Wages D, Narayan D, Hei D,
In that study, p53 transgenic mice were dosed intravenously Wiesehan G, Corash L: Inactivation of viruses with preservation
for 6 months with very high doses of amotosalen and its of coagulation function in fresh frozen plasma. Blood 1996;
889:526a
photoproducts, the highest dose being 250-fold the clinical
14 Corten L, Wiesehan G, Smyers J, Murthy KK, McClure HM, Alter
amotosalen exposure.
H: Photochemical inactivation of hepatitis B (HBV) and hepatitis
The results reviewed in this article show that treatment C (HCV) viruses in human plasma as assessed in a chimpanzee
with the combination of the psoralen, amotosalen, plus UVA infectivity model. Blood 2000; 98:60a
light, in the INTERCEPT Blood System for plasma, yields 15 Alfonso R, Lin C, Dupuis K, Corten L, Reames A, Londe H, Chen
plasma preparations with a very low potential for toxicity in C, Yip J, Cook D, Smyers J, Clarke S, Wages D, Narayan D, Hei D,
patients. Findings from previous studies have demonstrated Wiesehahn G, Corash L: Inactivation of viruses with preserva-
the ability of the INTERCEPT Blood System for plasma to tion of coagulation function in fresh frozen plasma. Blood 1996;
inactivate a wide range of viruses, bacteria and protozoa, as 88:526a
well as leucocytes, in platelet concentrates [13,14], without 16 Wages D, Smith D, Walsh J, Smyers J, Chen C, Moore C, Jue C,
compromising haemostatic efficacy [15–22] thus potentially Davis K, Metzel P, Corash L, Cimino G, Hei D, Shafer S,
Wiesehahn G, Buchholz DH: Transfusion of therapeutic doses of
increasing transfusion safety. The results of the preclinical
virally inactivated fresh frozen plasma in healthy subjects. Blood
safety studies indicate that there are no toxicologically relevant
1997; 90:409a
effects associated with plasma prepared using this new system. 17 Wages D, Radu-Radurescu L, Adams M, Hambleton J, MacKenzie
M, Shafer S, Lee M, Smyers J, Metzel P, Buchholz D, Wiesehahn G,
References Corash L: Quantitative analysis of coagulation factors in response
to transfusion of S-59 phototchemically treated fresh frozen
1 Sullivan MT, McCullough J, Schreiber GB, Wallace EL: Blood plasma (S-59 FFP) and standard FFP. Blood 1998; 92:503a
collection and transfusion in the United States in 1997. Transfu- 18 Wages D, Bass N, Keefe E, Viele M, Menozzi D, Maslow L, Khalili M,
sion 2002; 42:1253–1260 MacKenzie M, Porter L, Smyers J, Lau M, Wiesehahn G, Corash L:
2 Development Task Force of the College of American Pathologists: Treatment of acquired coagulopathy by transfusion of fresh
Practice parameters for the use of fresh frozen plasma, cryo- frozen plasma (FFP) prepared using a novel, single unit photo-
precipitate, and platelets. J Am Med Assoc 1994; 271:777–781 chemical pathogen inactivation (P.I.) process. Blood 1999;
3 Laine E, Steadman R, Calhoun L, Blackall D, Levin P, Braunfeld M, 94:247a
Nourmand H, Neelakanta G, Ting L, Gornbein J, Busuttil R, Petz L. 19 Wages D, Smith D, Walsh J, Smyers J, Chen C, Moore C, Jue C,
Comparison of RBCs and FFP with whole blood during liver Davis K, Metzel P, Corash L, Cimino G, Hei D, Shafer S,
transplant surgery. Transfusion 2003; 43:322–327 Wiesehahn G, Buchholz DH: Virally-inactivated fresh frozen
4 Rock G: Comparison of plasma exchange with plasma infusion plasma transfusion in normal volunteers. Vox Sang 1998;
in the treatment of thrombotic thrombocytopenic purpura. N 74:1290
Engl J Med 1991; 325:393–397 20 Mintz P, Steadman R, Blackall D, Petz L, Bass N, Streiff M,
5 Müller-Breitkreutz K: Results of viral marker screening of Ness P, McCullough J, Ramies D, Wages D, Valentine ME,
unpaid donations and probability of window donations. Vox Ziomek T, Corash L: Pathogen inactivated fresh frozen plasma
Sang 2000; 78:149–157 prepared using Helinx™ technology is efficacious and well
6 Pillonel J: Screening of viral markers for HIV, HBV and HCV tolerated in the treatment of end-stage liver disease patients –
infections in blood donors in France and residual risk of viral The Step AC trial. Blood 2001; 98:709a
transmission by blood transfusion. Eurosurveillance 1998; 21 Pinkoski L, Amir S, Smyers J, Corash L, Ramies Wiesehahn G:
3:76–79 Photochemically treated INTERCEPT plasma retains protein C,
7 Glück D: Infektionsmarker bei blutspenden. Aus der studie zur protein S, and antithrombin activities. Transfus Clin Biol 2001;
epidemiologie von HIV – und hepatitisinfektionen bei blut- 8:100s
spenden. Infusionsther Transfusionsmed 1997; 24:167–170 22 deAlarcon P, Benjamin R, Shopnick R, Dugdale M, Smith P,
8 Aubuchon JP: Safety of the blood supply in the United States; Abshire T, Ortiz I, Matthew P, Cohen A, Konkle B, Streiff M,
opportunities and controversies. Ann Int Med 1997; 127:904– Ramies D, Wages D, Pinoski L, Arroyo A, Valentine ME,
909 Lazott R, Corash L, Kessler C, Hambleton J: Hemostatic response
9 Schreiber GB: The risk of transfusion-transmitted viral infec- in congenital coagulation factor-deficient patients transfused
tions. N Engl J Med 1996; 334:1685–1690 with fresh frozen plasma (FFP) prepared by Helinx™ pathogen
10 Burnouf T, Radosevich M: Reducing the risk of infection from inactivation technology – The Step CC Trials. Transfusion 2001;
plasma products: specific preventative strategies. Blood Rev 41:89S
2000; 14:94–110 23 Wagner SJ, White R, Wolf L, Chapman J, Robinette D, Lawlor TE,
11 Hoots WK: History of plasma-product safety. Transfus Med Rev Dodd RY: Determination of residual 4′-aminomethyl-4,5′,8-
2001; 2:3–10 trimethylpsoralen and mutagenicity testing following psoralen
12 Morgenthaler JJ: Securing viral safety for plasma derivatives. plus UVA treatment of platelet suspensions. Photochem Photo-
Transfus Med Rev 2001; 15:224–233 biol 1993; 57:819–824

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171– 182
182 V. Ciaravino et al.

24 Granstein RD, Morison WL, Kripke ML: Carcinogenicity of 31 Ciaravino V, McCullough T, Dayan AD: Pharmacokinetic and
combined ultraviolet B radiation and psoralen plus ultraviolet A toxicology assessment of INTERCEPT (S-59 and UVA treated)
irradiation treatment of mice. Photodermatol Photoimmunol platelets. Human Exp Toxicol 2001; 20:533–550
Photomed 1992–93; 9:198–202 32 Ciaravino V: Preclinical Safety of a nucleic acid-targeted Helinx™
25 Chiou CC, Yang JL: Mutagenicity and specific mutation spec- compound: a clinical perspective. Semin Hematol 2001; 38:12–19
trum induced by 8-methoxypsoralen plus a low dose of UVA in 33 Wollowitz S: Fundamentals of psoralen-based Helinx™ tech-
the hprt gene in diploid human fibroblasts. Carcinogenesis 1995; nology for inactivation of infectious pathogens and leucocytes
16:1357–1362 in platelets and plasma. Semin Hematol 2001; 38:4–11
26 Bredberg A, Nachmansson N: Psoralen adducts in a shuttle 34 Dass SB, Bucci TJ, Heflich RH, Casciano DA: Evaluation of
vector plasmid propagated in primate cells: high mutagenicity transgenic p53+/– mouse for detecting genotoxic liver carcino-
of DNA cross-links. Carcinogenesis 1987; 8:1923–1927 gens in a short-term bioassay. Cancer Lett 1999; 143:81–85
27 Chandler R, Diawara MM, Caprioglio H, Chavez KJ, Williams DE: 35 Sanders JM, Burka LT, Chanas B, Matthews HB: Comparative
Examination of psoralens-induced photodermatitis in Wistar rats xenobiotic metabolism between Tg. AC and p53+/– genetically
under scanning electron microscopy. J Nat Toxins 1999; 8:167–177 altered mice and their respective wild types. Toxicol Sci 2001;
28 Lindelof B: Risk of melanoma with psoralen/ultraviolet A 61:54–61
therapy for psoriasis. Do the known risks now outweigh the 36 Harvey M, McArthur MJ, Montgomery CA Jr, Butel JS,
benefits? Drug Safety 1999; 20:289–297 Bradley A, Donehower LA: Spontaneous and carcinogen-
29 Lagatolla C, Dolzani L, Granzotto M, Monti-Bragadin C: Geno- induced tumorigenesis in p53-deficient mice. Nat Genet 1993;
toxic activity of 4,4′,5′-trimethylazapsoralen on plasmid DNA. 5:225–229
Teratog Carcinog Mutagen 1998; 18:239–248 37 Snellman E, Rantanen T: Concentration-dependent phototoxicity
30 Nataraj AJ, Black HS, Ananthaswamy HN: Signature p53 muta- in trimethylpsoralen bath psoralen ultraviolet A. Br J Dermatol
tion at DNA cross-linking sites in 8-methoxypsoralen and ultra- 2001; 144:490–494
violet A (PUVA)-induced murine skin cancers. Proc Natl Acad 38 Morison WL: Subacute phototoxicity caused by treatment with
Sci USA 1996; 93:7961–7965 oral psoralen plus UV-A. Arch Dermatol 1997; 133:1609

© 2003 Blackwell Publishing Ltd. Vox Sanguinis (2003) 85, 171–182

You might also like