You are on page 1of 9

Blood Reviews (1997) 11, 169-177

0 1997 Harcourt Brace & Co. Ltd

Haemostasis and thrombosis

Recombinant coagulation factors

l? H. Roddie, C. A. Ludlam

The advent of technology that has allowed production of recombinant proteins on an industrial scale
has revolutionized haemophilia care. Recombinant coagulation factors, as opposed to their plasma-
derived counterparts, have a very low risk for transmission of infectious agents and their use should
eradicate the threat of infection from viruses such as hepatitis C and the human immunodeficiency
virus. This review outlines the manufacturing processesinvolved in the production of the
recombinant coagulation factors and the trials to date that have beenperformed to establish their
safety and efficacy.We also discusssome of the issuesinvolved in the change to useof recombinant
coagulation factors, such as viral safety and potential immunogenicity.

The development of recombinant coagulation factors reported following the use of a monoclonal antibody-
has been a major advance in haemophilia care promis- purified plasma-derived factor VIII concentrate. At
ing almost complete safety of patients from the risk of the present time, two recombinant factor VIII prepa-
transfusion transmitted infections. Although plasma- rations are licensed for clinical use in the treatment of
derived coagulation factor concentrates have a proven haemophilia A: Kogenate (Bayer) and Recombinate
record of efficacy, prior to the current viral inactiva- (BaxtetYHyland). During clinical trials they have been
tion procedures employed in their manufacture and shown to be effective and safe with comparable phar-
less rigorous exclusion of potentially infected donors macokinetic profiles to plasma-derived factor VIII
from the plasma pool, many haemophiliacs became concentrates. A higher incidence of inhibitor develop-
infected with human immunodeficiency virus (HIV) ment has been found in comparison with retro-
and hepatitis C virus in the 1980s. At present, spective trials of plasma-derived factor VIII, but
although plasma-derived concentrates appear safe many of these inhibitors were low level and trans-
from the point of view of HIV infection, concern itory. A further recombinant factor VIII product,
remains over the possibility of other viruses being Refacto (Pharmacia & Upjohn), that differs from the
transmitted. This is because even with the dual viral previous two in that it lacks the B domain of the fac-
inactivation steps of solvent/detergent treatment and tor VIII molecule has reached the stage of phase
heat treatment, human parvovirus B19 remains II/III trials and is not currently licensed. A recombi-
detectable in concentrates and able to infect recipients. nant factor IX, BeneFix (Genetics Institute), has also
In addition, hepatitis C virus transmission has been been manufactured and following clinical trials has
recently obtained its license for clinical use in the
USA. Recombinant FVIIa, NovoSeven (Novo
Nordisk), is a recombinant-derived factor VII that
P. H. Roddie MRCPath, C. A. Ludlam FRCPath, Department of
Haematology, Royal Infirmary of Edinburgh, Lauriston Place, undergoes activation to factor VIIa during the purifi-
Edinburgh EH3 9YW, UK. Tel: +44 (0)131 536 2122; cation process. It is licensed in Europe for use in the
Fax: +44 (0)131 536 2145. treatment of patients with haemophilia A and B and
Correspondence to: Dr C. A. Ludlam inhibitors and for patients with acquired haemophilia.

169
170 Blood Revienx

(B) 9okLhhcavychain .
H2N

ThmmVm cleavage sites


+

Fig. 1 Factor VIII cDNA and protein structure. (A) cDNA for FVIII gene. (B) FVIII molecule in its inactive form circulates as a
heterodimer, the B domain being cleaved during proteolytic processing and activation.

MOLECULAR BIOLOGY OF FVIII chromatography steps. Impurities which could con-


taminate the product, such as culture medium compo-
The gene for factor VIII was first cloned successfully nents and murine immunoglobulin, are effectively
in 1984.l.’ At that time it was the largest gene to be removed during the purification process. Within the
cloned with a sequence spanning almost 186 Kb and manufacturing process as a whole there is a three-
comprising approximately 0.1% of the X chromo- stage viral safety programme which includes: (i) char-
some. Factor VIII messenger RNA encodes a precur- acterization of the cell bank to ensure it is virus free;
sor protein of 2351 amino acids whilst the mature (ii) scrutiny of incoming biological raw materials and
factor VIII protein is 2332 amino acids long. The (iii) process validation for removal and inactivation of
amino acid sequence shows an organization of three viruses. This includes heat treatment at 40°C for 8 h.
structural domains that occur in the order Human serum albumin is added as a stabilizer to the
Al:A2:B:A3:Cl:C2 (Fig. 1). Factor VIII circulates in purified product which is then filtered, bottled and
the blood in the form of a heterodimer and is stabi- lyophilized.
lized by its interaction with a 50-fold excess of von
Willebrand factor. The B domain is cleaved during
Clinical trials
proteolytic processing and activation and its function,
if any, is unknown.3 Following the successful completion of preclinical
trials in a variety of animal species to demonstrate
safety and efficacy and to assess its pharmacokinetic
RECOMBINANT FACTOR VIII profile, the first clinical trial of Kogenate in patients
with haemophilia A began in 1988. This was con-
Kogenate ducted in three parts. The first part, corresponding to
a phase I trial, was concerned primarily with the
Manufacturing process
pharmacokinetics of Kogenate. The next two parts
rFVII1 (Kogenate) is produced by baby hamster kid- addressed the issues of safety and efficacy.’ Phase I
ney (BHK) cells which have been transfected with the testing employed a cross-over study design in 17
cDNA for the human factor VIII gene. The cells are patients to compare the pharmacokinetics of
cultured in bovine serum free medium and give high Kogenate with the plasma-derived factor VIII, Koate-
yield expression of factor VIII with no requirement HS. No significant differences were observed. This
for von Willebrand factor to stabilize the protein. same group then continued with Kogenate given as
Fermentation takes place in deep tank fermenters prophylaxis three times weekly over a 6-month period.
using a continuous, high cell density cultivation pro- During this time there were no adverse events
cess.4The recombinant factor VIII raw product from reported and no factor VIII inhibitors or antibodies
the fermentation is processed through a multi-step to mouse or hamster proteins developed. Further
purification system resulting in a final product of high proof of safety and efficacy was sought in the phase
purity and specific activity (Fig. 2). The purification II/III style sections of the trial. Fifty-six previously
process includes ion exchange and immunoaffinity treated patients who had completed at least 5 months
Recombinant coagulation factors 17 1

was of the duration of 4.2 years. A factor VIII


Harvest
inhibitor level of > 0.5 BU was considered significant.
Overall incidence of inhibitor development in all
Cell separation
patients was 19.8% occurring at a median of 9
I exposure days. In the patients with severe haemophilia
Anion exchange chromatography the inhibitor incidence was 28% (1 S/64). Of those who
developed an inhibitor just over half were high titre
Virus inactivation (> 10 BU). Five of this group were treated with an
immune tolerance induction protocol using Kogenate
lmmunoabsorption chromatogmphy with good response. In the nine patients with low level
inhibitors all were continued with ‘on-demand’ treat-
Size exclusion chromatography ment with Kogenate and seven have spontaneously
lost their inhibitor whilst two continue to have low
Anion exchange chromatography level inhibitors.’

Buffer exchange formdation


Recombinate

Sterile filtration Munz4f&turing process


The manufacture of Recombinate differs from that for
Lyophilization Kogenate in that the cell lines used to produce the fac-
tor VIII are Chinese hamster ovary (CHO) cells and
these are transfected with cDNA for both human fac-
Fig. 2 The purification process of recombinant FVIII (Kogenate).
tor VIII and von Willebrand factor.8 The transfection
with cDNA for von Willebrand factor is necessary to
of home treatment with Kogenate were evaluable for improve the yield and to stabilize the recombinant
the purposes of this trial. The calculated mean annual factor VIII produced by the CHO cells. The von
consumption of factor VIII was 64 000 units per Willebrand factor is removed during the purification
person. Eighty-two per cent of bleeding episodes process. Within the fermentor cell growth takes place
responded to a single infusion of recombinant factor through a ‘batch-refeed’ process as opposed to the
VIII whilst 12.2% of bleeding episodes required two continuous high cell density cell cultivation employed
infusions. Adverse reactions were predominately mild in the manufacture of Kogenate. Purification of the
and included light-headedness, a metallic taste in the recombinant factor VIII is performed along a number
mouth and localized rashes or discomfort at the infu- of stages which include monoclonal immunoaffinity
sion site. The overall incidence of these was 0.75”/0. column and ion-exchange separation steps. A similar
Kogenate was also used to promote haemostasis dur- viral safety programme is undertaken to that used in
ing major surgery and for the inpatient treatment of the manufacture of Kogenate except that there is no
serious haemorrhage on 32 occasions in 26 patients. viral inactivation step.
Haemostasis was judged to be excellent in all of these
cases. The above study demonstrated that Kogenate
Cliniad trials
was safe and efficacious but further investigation was
necessary to determine the incidence of inhibitor for- The first pilot study in patients of Recombinate was
mation. This was performed by means of a prospec- performed in 1987 whereby its pharmacokinetic profile
tive trial in previously untreated patients. This is the was compared with that of a plasma-derived factor
preferred population for this type of study as previ- VIII, Haemofil-T, in two haemophiliacs.’ Both patients
ously treated patients have already been exposed to exhibited recovery and clearance curves for
foreign protein from factor concentrates which may Recombinate which were comparable with those of the
have induced some degree of immune tolerance and plasma-derived product. They were then continued on
therefore could give a falsely low rate of inhibitor home treatment with Recombinate and during this
development. In the Kogenate previously untreated period had excellent to good responses for the treat-
patient (PUP) study, following the first infusion of ment of bleeding episodes with no adverse events
Kogenate patients were monitored for the develop- reported. A phase II/III trial was then performed to
ment of factor VIII inhibitors by means of a Bethesda gather more pharmacokinetic data and to assesslong-
assay performed at 3-monthly intervals.” Of the 101 term safety and efficacy of the product. Sixty-six
patients who participated in the trial 64 had severe, 16 previously treated patients with severe haemophilia A
moderate and 21 mild haemophilia A. Mean follow-up were enrolled in a prospective trial and of these
172 Blood Reviews

55 completed at least 18 months of observation whilst Clinical trials


receiving treatment with Recombinate.” Good to excel- Clinical trials of Refacto commenced in 1993. In a
lent haemostasis was achieved in the treatment of phase I/II study 36 previously treated patients with
bleeding episodes and no significant adverse events severe haemophilia A participated in a trial where
occurred. Following on from this was a trial to assess they were split into three groups of 12 patients. One
inhibitor formation in previously untreated patients.” group took part in a cross-over study to compare the
The rate of inhibitor development in 73 patients with pharmacokinetic profile of Refacto with that of a
severe haemophilia A who had received at least one plasma-derived factor VIII, Octonativ-M.‘3 Most
infusion of recombinant factor VIII was 3 1.5% (23/73). pharmacokinetic parameters were similar for Refacto
Five inhibitors were of high titre (BU > 10) and three and the plasma-derived factor VIII except that the
patients were treated with immune tolerance induction volume of distribution and clearance were slightly but
regimes with Recombinate. Seventy-six per cent of all significantly higher after administration of Refacto.
inhibitors were identified within the first 10 exposure This did not lead, however, to any significant differ-
days. In the 15 patients with low responding inhibitors ence in the clinically important factor VIII half-life.
(0.5-10 BU) nine spontaneously lost their inhibitor and Following on from this was a trial of long-term safety
all were being continued on Recombinate. This trial and efficacy in previously treated patients.‘” Eighty-
also confirmed the findings of earlier studies in that seven patients are currently included in the above trial.
Recombinate appeared effective in treating bleeding A total of 778 joint bleeds have been treated at a mean
episodes and was associated with a very low incidence dose of 30 iu/kg per injection. In 94% of joint bleeds
of adverse events. three or fewer injections were required to treat the
haemorrhage. Efficacy was reported as excellent or
good in 87% of cases. Adverse events were rare and
B domain deleted recombinant factor VIII
predominately mild with no serious adverse events
Studies on the function of factor VIII molecules pro- occurring that were considered to be related to the use
duced by deleting various regions of the FVIII cDNA of Refacto. Refacto was also used to promote
showed that lack of the B domain did not affect the haemostasis during surgery in 17 patients undergoing
functional activity of the factor VIII product. Also, 22 procedures requiring at least 1 week of factor VIII
loss of the B domain led to enhanced expression of replacement. Haemostatic efficacy was judged to be
factor VIII which was increased up to 5-fold com- excellent in all cases. A study in previously untreated
pared with the unaltered factor VIII cDNA. The fac- patients to monitor for inhibitor development was
tor VIII molecule so produced appears less prone to begun in October 1994. To date 72 patients with
proteolytic degradation and therefore no human albu- severe haemophilia A have received at least one injec-
min is needed for its stabilization. tion of Refacto. Ten (14%) patients have developed an
inhibitor of which four are high titre. However, as
mean time to inhibitor development was 9.5 days and
Refacto
43 patients in the study had yet to have > 10 exposure
Manufacturing process days the true inhibitor incidence is likely to be higher.
There is currently only one recombinant factor VIII
lacking the B domain, Refacto, that has been pro-
Recombinant factor VIII versus plasma-derived factor
duced and is undergoing clinical trials. Its production
VIII
is achieved by transfecting CHO cells with factor VIII
cDNA lacking the majority of the gene coding for the There are a number of important issues which influ-
B domain. In the gene construct the major part of the ence the decision to change from using plasma-derived
region is a sequence Ser-Glyn-Asn (SQN); the SQ in factor concentrates to using recombinant products
the designated name rFVII1 SQ originates from the (Table 1). Most notable of these is the disastrous con-
amino acid sequence at the fusion site.” The rFVIII- sequences that arose through the transmission of viral
SQ is purified from the conditioned medium of CHO infections such as HIV by plasma-derived coagulation
cells by a series of steps which include immunoaffinity factors to haemophiliacs. Exclusion of potentially
chromatography, ion exchange chromatography and infected individuals from donating plasma and the
size exclusion chromatography. A solvent/detergent viral inactivation methods now used in the manufac-
viral inactivation stage is included within the purifica- ture of plasma-derived concentrates make these prod-
tion process. The final product does not contain any ucts very safe in terms of risk of HIV infection.
von Willebrand factor and albumin is not required for However, it appears likely that it will be impossible to
its stabilization. Its specific activity is 15 000 iu/mg render them completely free from the risk of viral
protein. transmission. The current recommendations from the
Recombinant coagulation factors 173

Table 1 Issues involved in the decision to change from sources of infectious agents contaminating the pro-
plasma-derived to recombinant FVIII
duction process are: (i) within the production cell line
itself; (ii) from the biological raw materials used in the
Reduced risk of transmission of infectious agents
Incidence of inhibitor development fermentation process, i.e. bovine insulin and; (iii) the
Preservation of immune function chromatographic steps used in the purification process
Cost implications and in the manufacturing process. The production cell
Consequences for plasma fractionation industry line for both of the licensed recombinant factor VIII
Pressure from patients and their families
products undergoes a screening programme for adven-
Potential unlimited supply
titious and endogenous viruses and in addition there is
a final viral removal phase used to treat the bulk factor
Committee for Proprietary Medicinal Products are VIII product. Infections in production cell lines for
that plasma-derived factor VIII undergoes a dual viral recombinant proteins have been reported and are
inactivation process. Human parvovirus B19, how- usually recognized by their cytopathic effect in the cell
ever, is resistant to both solvent/detergent treatment line.‘OIf an infection occurred with a virus that did not
and heating and is therefore capable of being trans- cause either a cytopathic effect or a failure of growth
mitted by plasma-derived concentrates and can cause of the cell line then it could potentially infect the factor
infection in those who have not previously been VIII product, illustrating the importance of continued
exposed to the virus.‘5,‘h This infection, although usu- viral surveillance and in using a viral removal stage in
ally mild, can have serious consequences in certain the manufacturing process. Human albumin is another
groups of patients. Hepatitis A virus, being another potential source of infection as it is used to stabilize
non-lipid enveloped virus, is resistant to solvent/ the final factor VlII product. It has an excellent safety
detergent treatment and an outbreak of hepatitis A in record with no reported incidences of viral infection in
haemophiliacs from several countries in continental recipients attributed to its use. However, there has been
Europe, South Africa and North America was a report of human parvovirus B19 DNA contami-
attributed to a plasma-derived product that had nating vials of both Kogenate and Recombinate which
undergone this form of viral inactivation process.“.” was believed to have originated from the human albu-
Of similar concern is the report of hepatitis C virus min used for stabilization.” The authors of this report
transmitted by a monoclonal antibody-purified heat conclude that although human parvovirus B19 DNA
treated plasma-derived factor VIII.19 These reports was present in the recombinant factor VIII vials it was
taken in conjunction with the ever present threat of not likely to be infective, but that efforts should be
infectious agents, particularly non-lipid coated made to use a non-plasma-derived stabilizer. Refacto
viruses, being transmitted by plasma-derived factor meets that criteria as no human albumin is required
concentrates provides probably the strongest argu- in stabilization of the final factor VIII product and
ment in favour of moving to the use of recombinant therefore it is a safer product as regards potential viral
coagulation factors. It is important to remember, transmission. The recombinant factor IX, BeneFix, is
however, that although recombinant factor VIII prod- produced without using any human- or animal-derived
ucts carry a much higher margin of safety from the proteins and therefore has the highest margin of safety
point of view of transmission of infection there still of all the recombinant coagulation proteins currently
remains a small but definite potential risk. Potential available (Table 2).

Table 2 Production characteristics of recombinant factors VIII and IX

Kogenate Recombinate Refacto BeneFix

Gene VIII VIII-VWF VIII-B domain-less IX-PACE

Cell line BHK CHO CHO CHO

Animal/human Human insulin Bovine albumin Human albumin Recombinant human


proteins used in Human albumin Bovine insulin Murine MoAb insulin
culture and Human transferrin Aprotinin
purification Murine MoAb Murine MoAb

Stabilizer Albumin Albumin None None

Virus inactivation 40°C 8 h None Solvent/detergent None

Specific activity 8-30 2-10 I5 000 200


unitslmg
174 Blood Reviews

Table 3 Results of inhibitor trials in previously untreated patients with severe haemophilia

Plasma derived Kogenate Recombinate Refract0


FVIII (de Bias?)

No. of patients with FVIII < 2% 48 64 73 72*


Mean no. of exposure days to 30 9 10 9.5
inhibitor development
Inhibitors
Total 11 (23%) 18 (28%) 23 (32%) 10 (14%)
< 10BU 2 (4%) 8 (13%) 15 (2lY”) 6 (8%)
> 10BU 9 (19%) 10 (15%) S(ll%) 4 (5%)
Spontaneous loss 0 (0%) 7 (19%) 9 (12%) -

* Forty-three patients in trial have < 10 exposure days.

Another important issue in the decision to use the two groups it is known that the level of CD4 posi-
recombinant factor VIII is its potential immunogen- tive lymphocytes is an important predictor of the risk
icity. This is in regard to its ability to stimulate the of developing AIDS. Therefore slowing the progressive
development of factor VIII inhibitors and in its effect decline in CD4 positive lymphocytes, that occurs in
on the immune system of patients receiving it long HIV infected haemophiliacs, by use of high-purity
term. The data from clinical trials of previously factor VIII products would be expected to confer a
untreated patients treated with Kogenate and significant clinical benefit. As the recombinant factor
Recombinate showed inhibitor rates in patients with VIII products are extremely pure it would be hoped
severe haemophilia A of 28% and 3 1.5% respectively. that they would have a minimal impact on immune
Many of these were low level inhibitors which function. A clinical trial performed in HIV seroposi-
developed within a short time of first exposure to tive and seronegative haemophiliacs treated with
recombinant factor VIII and often disappeared spon- Kogenate demonstrated a small but significant
taneously. However, these reported rates were much decrease in the number of CD4 positive lymphocytes
higher in comparison to previous reports on patients in the HIV seropositive individuals over the 35year
treated with intermediate-purity factor VIII concen- study period.24 However, this decline was slower in
trates. There had been concerns with the high-purity comparison to that reported in trials of patients
plasma-derived factor VIII that during the purifica- treated with intermediate-purity products. In the HIV
tion process neoantigen formation may take place and seronegative patients of 6 years of age or older the B,
thus lead to an increase in inhibitor rates. However, microglobulin levels, CD4 positive and CD8 positive
more recent prospective studies on previously lymphocyte counts did not change during the study
untreated patients treated with intermediate and low- period - further supportive evidence that Kogenate
purity concentrates have shown comparable rates of had no deleterious effects on immune function.
inhibitor developmenP (Table 3). It is likely that the An important factor that mitigates against the
higher reported incidence of inhibitors in the recom- immediate widespread use of recombinant factor VIII
binant factor VIII studies was due to the increased is its cost in comparison to the currently available
frequency of surveillance that these patients under- plasma-derived products. Changing patients from
went in comparison to earlier studies and that the their existing plasma-derived factor VIII to a recom-
recombinant products are no more antigenic in regard binant product will have major financial implications.
to inhibitor development than the plasma-derived In addition the move to treat children with prophylac-
concentrates. Continued long-term surveillance will tic factor VIII instead of on-demand treatment will
be necessary in order to definitively establish that this further contribute to this increased cost. As the
is the case but experience thus far is that the majority demand for recombinant factor VIII increases and
of inhibitors occur within 20 exposure days and there- along with it the scale of its production, in combina-
fore late inhibitor development is likely to be negligible. tion with increased competition between different
In terms of the effects on the immune system, it has manufacturers, the cost of recombinant factor VIII
been shown that the use of monoclonal antibody- should fall. In the short term, however, limited finan-
purified plasma-derived factor VIII concentrates cial resources will restrict the number of patients that
appears to halt the decline in numbers of CD4 posi- can be converted to recombinant factor VIII. In the
tive lymphocytes in HIV infected haemophiliacs in United Kingdom the recently published ‘Guidelines
comparison to those treated with intermediate-purity on therapeutic products to treat haemophilia’ pro-
products.23 Although within the context of the study duced by the UK Haemophilia Centre Directors
there was no difference in clinical outcome between Organisation Executive Committee recommend that
Recombinant coagulation factors 175

recombinant factor VIII is the treatment of choice for glutamic acid residues and propeptide cleavage in
all patients.25 However, they state that if prioritization order to become fully functional. The initial develop-
is necessary then patients least exposed to blood ment of recombinant factor IX was hampered by the
products should be given first priority. The order of fact that these post-translational modifications were
priority they suggest in deciding who should be incomplete and led to the production of a function-
treated with recombinant factor VIII is as follows: ally deficient factor IX protein. These problems were
HIV antibody-negative patients; previously untreated eventually overcome and the recombinant factor IX
patients > HCV-negative patients > HCV-positive now produced has full functional activity. Genetics
patients > HIV antibody-positive patients. Institute manufacture BeneFix by co-transfecting the
The financial implications of using recombinant CHO cell line with the cDNA from the factor IX gene
factor VIII are not solely confined to those purchasing and with cDNA encoding an engineered form of the
the product but will also have a considerable impact protease PACE (paired basic amino acid cleaving
on the current major producers of factor VIII, the enzyme). PACE improves the processing efficiency of
plasma fractionation industry. This industry has the profactor IX produced by the CHO cell lines. The
always been driven by the need to produce sufficient purification procedure used to convert the raw prod-
factor VIII to meet demand. The plasma fractiona- uct to high-purity factor IX includes ultrafiltration-
tion companies by stopping their production of factor diafiltration and immunoaffinity chromatography
VIII would save only a small amount in terms of stages. No human plasma- or animal-derived proteins
reduced manufacturing costs but would lose a large are used in the purification and albumin is not
amount of revenue which would probably have to be required for stabilization of the final product.
borne by increasing the price of other plasma prod- Therefore the recombinant factor IX produced is vir-
ucts such as albumin. However, an advantage of using tually free of any risk of transmission of human infec-
recombinant technology to produce factor VIII is that tious agents. The specific activity of the final product
the manufacturing process is not dependent on is 270 u/mg.
obtaining sufficient amounts of plasma for fractiona-
tion and recombinant factor VIII could potentially be Clinical trials
produced in unlimited amounts.‘”
Haemophiliacs and their families are likely to Preclinical animal trials have demonstrated that there
favour recombinant factor VIII for their treatment is comparable pharmacokinetic behaviour between
because of its perceived absence of risk of transmis- BeneFix and the high-purity plasma-derived factor IX,
sion of infectious agents. Mononine.3” This was confirmed in phase I trials in 12
One of the technical considerations which needs to patients with haemophilia B patients using a cross-over
be addressed when switching from using plasma- study design.” In addition, as part of the trial, markers
derived factor VIII concentrates to recombinant of activation of the coagulation system (prothrombin
factor VIII products is how their potencies should be fragments 1 + 2 and fibrinopeptide A) were monitored
assayed as it has been shown that the one-stage factor and these did not become elevated following infusions
VIII assay may underestimate the true potency of of BeneFix, evidence which suggests it is not poten-
recombinant factor VIII products.” This is especially tially thrombogenic. Phase II/III clinical trials to assess
true for the B-domain deleted recombinant factor safety and efficacy in previously treated patients were
VIII, Refacto. It is recommended that the chromo- then initiated. Forty-four patients with moderate or
genic substrate assay method is used to determine severe haemophilia B were started on BeneFix for use
potency of the recombinant factor VIII products. as home treatment. At the most recently reported eval-
uation a total of 693 bleeding episodes had been
treated. Eighty-two per cent of these required a single
RECOMBINANT FACTOR IX infusion of BeneFix to arrest haemorrhage and in 84%
of the infusions given response was rated as excellent or
BeneFix good.‘* There were no serious adverse events that were
deemed to have arisen as a consequence of the use of
1Manufucturingprocess
BeneFix and in particular there were no thrombotic
The factor IX gene and cDNA sequence were cloned events. A single patient developed a low titre inhibitor
in 1982.2R.2’CHO cells were subsequently transfected to factor IX after 39 exposure days. This patient had
with cDNA from the factor IX gene and were able to had previous heavy exposure to plasma-derived factor
express recombinant factor IX. Factor IX is a protein IX. The peak inhibitor titre was 1 BU and it remained
which requires significant post-translational modifica- detectable for 11 months before spontaneously resolv-
tion in the form of gamma carboxylation of terminal ing. In a trial of BeneFix use during surgery a total of
176 Blood Reviews

13 surgical procedures were performed (10 major, 3 factor VII is purified via a number of steps which
minor). A continuous infusion of BeneFix was given include ion exchange and immunoaffinity chromatog-
for three of the major surgical procedures. The raphy. Included within this purification process is a
response to treatment was excellent to good in all cases. solvent/detergent viral inactivation step. During ion
Clinical trials in previously untreated patients to study exchange chromatography the recombinant factor
in particular the incidence of inhibitor development VII undergoes autoactivation to the final product,
started in October 1995. recombinant factor VIIa. Nine of the ten glutamic
acid residues on the protein are fully gamma carboxy-
lated and the remaining one is partially gamma car-
RECOMBINANT FACTOR VIIA
boxylated. There is no requirement for any stabilizing
protein in the final product and in fact no human-
The development of recombinant factor VIIa was
derived proteins are used at any stage during the
motivated by the fact that there are a limited number
manufacturing process.
of therapeutic options available to treat haemophiliac
patients who have inhibitors. For many patients these
therapies may be ineffective or associated with an Clinical trials
unacceptable risk of side-effects. With high titre
Preclinical animal trials to assess potential thrombo-
inhibitors it is not usually possible to give sufficient
genicity did not demonstrate any evidence of systemic
quantities of factor VIII to overcome the inhibitor
activation of the coagulation cascade following infu-
and achieve measurable factor VIII levels. In these sit-
sion of recombinant factor VIIa.33 These were
uations although porcine factor VIII may be used
followed by clinical trials. The Compassionate Use
often the human factor VIII antibodies are cross-reac-
Programme started in 1988. Its purpose was to evalu-
tive with the porcine factor VIII making this therapy
ate the treatment of bleeding episodes with
similarly ineffective. Activated prothrombin complex
NovoSeven in patients with haemophilia and
concentrates work in around 60-70% of bleeding
inhibitors when all other therapeutic options had been
episodes. This product contains a variety of partially
exhausted or patients required essential surgery and
activated clotting factors, including factors IXa and
no alternative treatment was possible. In the early
Xa, and as such carries a risk of systemic activation of
stages of this study many patients were treated with 60
the coagulation system and thrombosis and therefore
yg/kg or less of NovoSeven but it became apparent
must be used with caution. Factor VIIa was chosen as
that a higher dose was required to ensure adequate
a potential bypassing agent because, in the absence of
dosing in all patients and it was therefore recom-
tissue factor, it has no proteolytic effect despite being
mended that the initial dose should be 90 yg/kg with
in an activated form and therefore does not have the
an interval between subsequent doses of 2-3 h. This
propensity to cause systemic activation of coagula-
recommendation was based upon the results of pre-
tion. Following administration it binds to tissue factor,
clinical and laboratory testing and from the experi-
the resulting complex being able to convert factor X to
ence of the clinical trials. It appears that in order to
factor Xa without any requirement for factors VIII or
achieve haemostasis levels of factor VII:coagulation
IX. Tissue factor is normally only expressed by dam-
activity (FVII:C) should be maintained at > 6 u/ml.‘”
aged endothelium localizing the action of the factor
The results of the Compassionate Use Programme are
VIIa to the site of tissue injury.
as follows: for the treatment of joint and muscle bleeds
Initially factor VIIa was produced by purification
global evaluation of response in 494 bleeding episodes
from normal plasma and was successfully used to
in 111 patients was judged to be excellent/effective in
treat bleeding episodes in haemophiliac patients with
79% of joint bleeds, 62% of muscle bleeds and 69% of
inhibitors. Because the concentration of factor VII in
tense muscle bleeds/compartment syndrome? Central
human plasma is very low (0.5 rig/l) large scale manu-
nervous system bleeds are one of the most serious
facture from human plasma would not have been fea-
and life threatening complications of haemophilia. In
sible and so attention turned to producing FVIIa by
22 episodes of central nervous system bleeding
means of recombinant technology.
in patients with haemophilia A and inhibitors
NovoSeven was effective in 83% of cases with only
NovoSeven one fatality.36 Results in the surgical setting are simi-
larly encouraging. NovoSeven was used to promote
Manufacturing process
haemostasis in 12 patients requiring essential or life
cDNA from the factor VII gene was transfected into saving surgery for a total of 13 surgical procedures. In
a BHK cell line. The culture medium containing one case a global evaluation of the end treatment
the secreted FVII is harvested and the recombinant result was not reported but in the other 12 cases the
Recombinant coagulation factors 177

end result was considered excellent (11) or efficient (l).,’ 18. Normann A, Graff J, Gerritzen A et al. Detection of hepatitis
A virus RNA in commercially available factor VIII
There was no laboratory evidence of systemic activa- preparation. Lancet 1992; 340: 123221233.
tion of coagulation detected. A Home Treatment trial 19. Berntrop E, Nilsson IM, Ljung R, Wide11 A. Hepatitis C virus
(started in 1994) is underway which will evaluate the transmission by monoclonal antibody purified factor VIII
concentrate. Lancet 1990; 335: 1531-1532.
safety and efficacy of NovoSeven given to control 20. Rabenau H, Ohlinger V, Anderson J et al. Contamination of
joint, muscle and mucocutaneous bleeds in the home genetically engineered CHO cells by epizootic haemorrhagic
setting. Sixty patients have been enrolled and the aim disease virus (EHDV). Biologicals 1993: 21: 207.-214.
21. Eis-Hiibinger A, Sasowski U, Brackmann H, Kaiser R, Matz
is to accumulate 120 bleeding episodes.ix B, Schneweis K. Parvovirus B19 DNA is frequently present in
The Compassionate Use Programme has closed recombinant coagulation factor VIII products. Thromb
and NovoSeven is now licensed in Europe for the Haemost 1996; 786: 1118.
22. de Biasi R, Rocino A, Papa ML, Salerno E, Mastrullo L, De
treatment of bleeding in patients with haemophilia Blasi D. Incidence of factor VIII inhibitor development in
and inhibitors and in patients with acquired haemophilia A patients treated with less pure plasma derived
haemophilia due to factor VIII or IX inhibitors. concentrates. Thromb Haemost 1994; 71: 544547.
23. de Biasi R. Rocino A, Miraglia E, Mastrullo L. Quirino AA.
The impact of a very high purity factor VIII concentrate on
REFERENCES the immune system of human immunodeficiency virus-
infected haemophiliacs: a randomised prospective two year
I. Wood WI, Capon DJ, Simonsen CC et al. Expression of active comparison with an intermediate purity concentrate. Blood
human factor VIII from recombinant DNA clones. Nature 1991; 78: 191881922.
1984; 312: 330-336. 24. Manucci PM, Brettler DB, Aldeort LM et al. Immune status
2. Toole JJ, Knopf JL, Wozney JM et al. Molecular cloning of a of human immunodeficiency virus seropositive and
cDNA encoding human antihemophiliac factor. Nature 1984; seronegative haemophiliacs infused for 3.5 years with
3 12: 342~-347. recombinant factor VIII. Blood 1994; 83: 195881962.
3. Kaufman RJ. Expression and structure-function properties of 25. United Kingdom Haemophilia Centre Directors Organisation
recombinant factor VIII. Trans Med Rev 1992; 6: 2355246. Executive Committee. Guidelines on therapeutic products to
4. Boedeker BGD. The manufacturing of recombinant factor treat haemophilia and other hereditary coagulation disorders.
VIII, Kogenate. Trans Med Rev 1992; 6: 256-260. Haemophilia 1997; 3: 63377.
5. Schwartz RS, Abildgaard CF, Aldeort LM et al. Human 26. Limentani SA. Will recent innovations in therapy save
recombinant DNA-derived antihaemophiliac factor (factor perceived deficiencies in self-sufficiency policies. Blood
VIII) in the treatment of haemophilia A. N Engl J Med 1990; Coagulation and Fibrinolysis 1994; S(suppl4): S8 llS84.
323: 1800-1805. 27. Oswaldsson U, Frank L, Sandberg H, Mikaelsson M.
6. Lusher JM, Arkin S, Abildgaard CF et al. Recombinant Comparison of factor VIII: C methods and reagents in the
factor VIII for the treatment of previously untreated patients assay of factor VIII concentrates and post-injection patient
with haemophilia A. N Engl J Med 1993; 328: 453459. plasma samples. XXI Int Congr Wld Fed Haemophilia,
7. Lusher JM. Recombinant clotting factor concentrates. Mexico City, 1994 (abstract).
Balliere’s Clinical Haematology 1996; 9: 291-303. 28. Choo GH, Gould KG, Rees JC, Brownlee GG. Molecular
8. Gomperts ED, Lundblad R, Adamson R. The manufacturing cloning of the gene for human anti-haemophiliac factor IX.
process of recombinant factor VIII, Recombinate. Trans Med Nature 1982; 299: 178180.
Rev 1992; 6: 247-25 1. 29. Kurachi K, Davie EW. Isolation and characterisation of a
9. White GC, McMillan CW, Kingdon HS, Shoemaker CB. Use cDNA encoding for human factor IX. Proc Nat Acad Sci
of recombinant antihemophiliac factor in the treatment of USA 1982; 79: 6461-6464.
two patients with classical haemophilia. N Engl J Med 1989; 30. Keith JC. Ferranti TJ, Misra B et al. Evaluation of
320: 166-170. recombinant human factor IX; pharmacokinetic studies in the
10. Bray GL for the Recombinate Study Group. Current status of rat and dog. Thromb Haemost 1995; 73: 101 105.
clinical studies of recombinant factor VIII (Recombinate) in 31. White GC, Shapiro AD, Ragni MV et al. A pharmacokinetic
patients with haemophilia. Trans Med Rev 1992; 6: 2522255. evaluation of recombinant human factor IX (rhFIX) in
11. Bray GL, Gomperts ED, Courter S et al. A multicentre study patients with severe haemophilia B. Thromb Haemost 1995;
of recombinant factor VIII (Recombinate): safety, efficacy, 73: 1015.
and inhibitor risk in previously untreated patients with 32. White GC, Beebe A, Neilson B. Recombinant factor IX.
haemophilia A. Blood 1994; 83: 24282435. Thromb Haemost 1997: 78: 261-265.
12. Mikaelsson M, Eriksson B, Lind P et al. Manufacturing and 33. Diness V, Bregengaard C, Erhardtsen E, Hedner U.
characterization of a new B-domain deleted recombinant Recombinant human factor VIIa (rVIIa) in a rabbit stasis
factor VIII, r-VIII SQ, Thromb Haemost 1993; 69: 1205. model. Thromb Res 1992; 67: 233-241.
13. Fijnvandraat K, Berntrop E, ten Cate JW et al. Recombinant 34. Hedner U. Dosing and monitoring NovoSeven treatment.
B-domain deleted FVIII (rFVIII-SQ): pharmacokinetics and Haemostasis 1996; 26(suppl 1): 102-108.
initial safety aspects in haemophilia A patients. Thromb 35. Molskov Beth R. Recombinant factor VIIa in joint and
Haemost 1997; 77: 298302. muscle bleeding episodes. Haemostasis 1996; 26(suppl 1):
14. Berntrop E. Second generation, B-domain deleted 1355138.
recombinant factor VIII. Thromb Haemost 1997; 78: 256-260. 36. Rice KM. Savidge GE NovoSeven (recombinant factor VIIa)
15. Azzi A, Ciappi S, Zakvrzewska K, Morlini M, Mariani G, in central nervous system bleeds. Haemostasis 1996; 26
Manucci PM. Human parvovirus B19 infection in haemophiliacs (suppl 1): 13lll34.
first infused with two high-purity virally attenuated factor-VIII 37. Ingerslev J, Freidman D, Gastineau D et al. Major surgery in
concentrates. Am J Haematol 1992: 39: 228-230. haemophiliac patients with inhibitors using recombinant
16. Prowse C. Ludlam CA, Yap PL. Human parvovirus B19 and factor VIIa. Haemostasis 1996; 26(suppl 1): 118123.
blood products. VOX Sanguinis 1997; 72: l-1 0. 38. American experience with home use of NovoSeven
17. Manucci PM. Outbreak of hepatitis A among Italian patients recombinant factor VIIa in haemophiliacs with inhibitors.
with haemophilia. Lancet 1992; 339: 819. Haemostasis 1996: 26(suppl 1): 1433149.

You might also like