03.01.2015 Views

TRACTOCILE VIALS 7.5mg /0.9 ml (Bolus Injection)

TRACTOCILE VIALS 7.5mg /0.9 ml (Bolus Injection)

TRACTOCILE VIALS 7.5mg /0.9 ml (Bolus Injection)

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Product Monograph<br />

Ferring International Center, Kay Fiskers Plads 11, 2300 Copenhagen S, Denmark<br />

Telephone: +45 88 33 88 34 Fax: +45 88 33 88 80 www.ferring.com<br />

The tocolytic with placebo-level<br />

cardiovascular risk 1 atosiban


Product Monograph<br />

Introduction<br />

Preterm birth is a continuing obstetric problem<br />

that contributes significantly to the incidence of<br />

perinatal death and long-term handicap. In this<br />

context, preterm births are believed to account<br />

for between 69% and 83% of neonatal deaths in<br />

various studies. 2-4 Despite this, the incidence of<br />

preterm birth has remained static for many years.<br />

One explanation for this is that the management<br />

of preterm labour has altered very little in the<br />

past 30 years. Strategies aimed at reducing the<br />

incidence of preterm birth include the<br />

identification of risk factors that increase the<br />

likelihood of preterm delivery. Treatment is then<br />

designed to target those risk factors and limit<br />

their effect. Although perinatal mortality has<br />

declined, mostly due to the improved management<br />

of very low birthweight babies rather than<br />

prevention of preterm labour, efforts to prevent<br />

preterm birth have been largely unsuccessful so far<br />

and preterm birth still represents a major<br />

healthcare problem to both developed and<br />

developing countries.<br />

Preterm labour is now known to exist as a<br />

heterogenous syndrome, with many different<br />

aetiologies, some more significant than others.<br />

This may help to explain why treatment strategies<br />

for preterm labour have been relatively<br />

unsuccessful to date.<br />

Pharmacological intervention, through the<br />

administration of tocolytic agents, is the current<br />

mainstay of therapy to suppress preterm labour.<br />

All of the currently available tocolytic agents<br />

share a similar level of efficacy, prolonging<br />

pregnancy for up to 48 hours. Their use is most<br />

effective at gestational ages below 30 weeks, and<br />

it has been shown that an extension of pregnancy,<br />

even of a few days, can have a major impact on<br />

survival and the prevention of morbidity at 24–28<br />

weeks gestation 5 . However, current tocolytic<br />

agents, such as beta-agonists, have significant side<br />

effects in the mother and fetus, which may cause<br />

clinically important complications.<br />

Tractocile ® is the first oxytocin antagonist to be<br />

specifically developed for the treatment of<br />

preterm labour. Tractocile ® has a specific mode<br />

of action, inhibiting oxytocin-induced uterine<br />

contractions by blocking oxytocin receptors in the<br />

uterus. Extensive clinical investigations have<br />

shown Tractocile ® to be at least as effective as<br />

current tocolytic agents. In addition, due to its<br />

novel and specific mode of action, Tractocile ® has<br />

a markedly improved maternal side effect profile<br />

compared with conventional therapies.<br />

This monograph provides an up to date review<br />

of all the data currently available on Tractocile ® .<br />

Information on the chemistry, pharmacology,<br />

clinical and preclinical development of Tractocile ®<br />

is provided.<br />

This monograph is provided as a reference<br />

source for obstetricians treating pregnant<br />

women in preterm labour. Current management<br />

strategies of treating preterm labour are<br />

reviewed and the benefits of using tocolytic<br />

agents are discussed.<br />

2 3


Contents<br />

Introduction ................................................................................3<br />

1 Preterm labour and birth......................................7<br />

Definition ............................................................................8<br />

Extent of the problem ........................................................8<br />

Risk factors ..........................................................................8<br />

Past obstetric history......................................................8<br />

Sociobiological variables ...............................................9<br />

Complications of the current pregnancy....................10<br />

Prediction/diagnosis..........................................................11<br />

Risk scoring systems .....................................................11<br />

Biochemical markers....................................................11<br />

Cervical status...............................................................11<br />

Early diagnosis..............................................................11<br />

2 Management of preterm labour.................13<br />

Diagnosis ...........................................................................14<br />

Objectives ..........................................................................15<br />

Guidelines for managing preterm labour ..................15<br />

Treatment ..........................................................................15<br />

Efficacy...............................................................................16<br />

Tocolytic therapy...............................................................16<br />

Beta-agonists................................................................16<br />

Prostaglandin synthetase inhibitors ...........................18<br />

Magnesium sulphate ...................................................19<br />

Calcium channel blockers ............................................20<br />

Oxytocin antagonists ...................................................21<br />

3 Tractocile ® ........................................................................23<br />

Rationale for development ..............................................24<br />

Mechanism of action ........................................................24<br />

Chemistry of Tractocile ® ....................................................24<br />

Preclinical studies ..............................................................25<br />

Receptor binding: affinity, specificity .........................25<br />

Pharmacological activity In vivo..................................25<br />

In vivo effect on uterine contractility .........................25<br />

Effect on labour duration............................................25<br />

Effect on prostaglandin release ..................................25<br />

Clinical pharmacodynamic studies ...................................26<br />

Clinical pharmacokinetic studies......................................26<br />

4 Clinical efficacy of Tractocile ® ....................29<br />

Phase II clinical experience ...............................................30<br />

Phase III studies .................................................................31<br />

Tractocile ® vs. ritodrine................................................31<br />

Tractocile ® vs. salbutamol ............................................32<br />

Tractocile ® vs. terbutaline............................................32<br />

Pooled analysis.............................................................32<br />

5 Clinical safety of Tractocile ® .........................35<br />

Phase II studies........................................................................36<br />

Phase III studies.......................................................................37<br />

Placebo controlled studies...............................................37<br />

Comparative studies ........................................................37<br />

Pooled analysis .................................................................37<br />

Infant follow up after beta-agonist tocolytic therapy..39<br />

6 Pharmaceutical application<br />

of Tractocile ® .................................................................41<br />

Therapeutic indication......................................................42<br />

Pharmaceutical form.........................................................42<br />

Dosage and administration ..............................................42<br />

Preparation of the initial i.v. solution for injection ...42<br />

Preparation of the diluted Concentrate for Solution<br />

for Infusion.................................................................. 43<br />

Contraindications............................................................. 43<br />

Precautions and warnings ................................................43<br />

Interaction with other medicinal products<br />

and other forms of interaction ........................................43<br />

Pregnancy and lactation...................................................43<br />

Undesirable effects ...........................................................43<br />

Overdose............................................................................43<br />

Storage and packaging information ...............................43<br />

7 Summary of the key benefits of Tractocile ®<br />

in the treatment of preterm labour .........45<br />

Proven efficacy and tolerability .......................................46<br />

Good safety profile ...........................................................46<br />

Absence of tachyphylaxis .................................................46<br />

Specific mode of action ....................................................46<br />

Rapid onset of action........................................................46<br />

Period of use .....................................................................46<br />

Conclusion ......................................................................47<br />

References..............................................................48-51<br />

4 5


6<br />

1<br />

Preterm labour and birth


Chapter 1 Preterm labour and birth<br />

1.1 Definition<br />

The last general consensus defined preterm birth as a<br />

gestational age less than 37 completed weeks or 259 days<br />

from the first day of the last menstrual period. 6 Low infant<br />

birth weight (


Chapter 1 Preterm labour and birth<br />

1.3.c Complications of the current pregnancy<br />

Infection<br />

In some populations, infection is believed to be<br />

responsible for up to 40% of preterm labour. 26 Vaginal<br />

microbial colonisation, vaginosis and even bacteriuria<br />

during the first and second trimester of pregnancy are<br />

known to increase the risk of preterm labour. There are<br />

three sources of evidence to suggest a role for infection<br />

in the onset of preterm labour: 27<br />

• administration of bacteria or bacterial products to<br />

animals results in either abortion or labour<br />

• systemic maternal infections such as pneumonia,<br />

malaria and typhoid fever are associated with the onset<br />

of labour<br />

• localised intrauterine infection is associated with<br />

preterm labour and delivery.<br />

Intrauterine infection is believed to originate in a number<br />

of ways: (1) ascending from the vagina and cervix; (2)<br />

through the placenta, (3) from the peritoneal cavity into<br />

the fallopian tubes; or (4) by iatrogenic means. 28 The most<br />

common pathway is believed to involve ascending<br />

infection from the vagina to the uterus. 29 Microbial<br />

invasion can take place in the absence of clinical or<br />

symptomatic signs of infection, therefore a positive<br />

amniotic fluid culture will be direct evidence of infection.<br />

Infection is thought to trigger preterm labour and<br />

membrane rupture via inflammation and the associated<br />

cytokine cascade (see figure 1). Once it has begun, the<br />

cytokine cascade persists. Antibiotics will therefore be<br />

unable to prevent the onset of birth, however, they can<br />

have an impact when used earlier in pregnancy. Trials<br />

have shown that antibiotic treatment of bacteriuria and<br />

vaginosis reduces the incidence of preterm birth. 30<br />

Figure 1.<br />

Plausible pathway for the initiation of preterm birth<br />

Fetal events:<br />

• Accelerated maturational changes<br />

• Stressors<br />

via CNS<br />

CRH<br />

Oxylacin<br />

Oxylacin<br />

Prostaglandins<br />

Cytokines<br />

Activation of<br />

receptor sites and<br />

calcium uptakes<br />

Myometrium<br />

Contraction<br />

Labour<br />

Cervix<br />

Effacement<br />

and dilation<br />

Maternal events:<br />

• Medical condition e.g. infection<br />

• Lifestyle<br />

• Strain<br />

• Psychological<br />

Oxylacin<br />

Prostaglandins<br />

Cytokines<br />

Maturational<br />

changes<br />

CRH<br />

Oxylacin<br />

Uteroplacental unit<br />

via CNS<br />

Plausible pathway for the initiation of preterm birth<br />

becomes detached from the uterine wall. This has been<br />

shown to be a strong predictive factor of preterm<br />

delivery. 33<br />

paramount importance to be able to predict accurately<br />

those women at risk. Before a decision to intervene in the<br />

pregnancy can be taken, it is essential that preterm<br />

labour has been diagnosed otherwise the mother or baby<br />

may experience unnecessary stress or harm.<br />

Several techniques have been developed in an<br />

attempt to identify women at risk of preterm birth,<br />

however, they are only predictions based on risk analysis.<br />

These techniques include risk-scoring systems,<br />

biochemical markers and cervical status.<br />

1.4.a Risk-scoring systems<br />

Since there are many risk factors associated with preterm<br />

birth, scoring systems have been developed. These<br />

systems have been designed to evaluate overall risk by<br />

assigning weighted scores to each factor’s relative<br />

importance. To be of any clinical use, the scoring systems<br />

should reproduce a high positive predictive value (a high<br />

proportion of true cases among all those with a positive<br />

test result) and be sensitive (able to detect true cases).<br />

Unfortunately, such risk scoring systems are neither<br />

predictive nor sensitive, typically having a positive<br />

predictive value of 17–34% and a sensitivity of


12<br />

2<br />

Management of preterm labour


Chapter 2 Management of preterm labour<br />

Once a clinical diagnosis of true preterm labour has<br />

been made, it is necessary to evaluate the management<br />

options available. A clinical diagnosis has traditionally<br />

been based on the presence of uterine contractions and<br />

cervical dilation. The accuracy of pregnant women to<br />

detect signals of preterm labour is poor with nulliparous<br />

women but improves in multiparous women. 42<br />

A thorough evaluation of a woman presenting with<br />

true preterm labour should include obstetric history,<br />

physical examination, infection screening, toxicology<br />

screening, immunopathology tests and ultrasound<br />

assessment (see table 2).<br />

2.1 Diagnosis<br />

The fundamental issue in the management of preterm<br />

labour is whether the risk of delivery outweighs the risk<br />

of prolonging the pregnancy. It would seem ideal to<br />

prolong pregnancy in all cases to maximise fetal<br />

development, but this is not always possible. The woman<br />

may be already in the later stages of active labour or<br />

serious adverse maternal/fetal factors may dictate the<br />

need for an immediate delivery. 43 Certain criteria have<br />

been proposed for the diagnosis of preterm labour in<br />

order to prevent unnecessary tocolytic therapy. These<br />

include a gestational age assessment, uterine<br />

contractions, membrane status, and cervical assessment.<br />

In some cases when gestational age is unknown it is<br />

important to determine whether a small fetus is preterm<br />

or growth restricted. Measurement of the fetal<br />

Medical history<br />

transcerebellar diameter by ultrasound may help<br />

distinguish them. 44 Regarding uterine contractions, it was<br />

found that at least five contractions per hour after 30<br />

weeks’ gestation is a strong indicator of preterm labour. 45<br />

Uterine contractions alone should not be relied upon,<br />

however, since experience shows that the rate of error<br />

when diagnosing preterm labour using this technique is<br />

more than 50%. 46 If the membranes are intact, a cervical<br />

dilation of at least 2 cm or effacement of 80% with<br />

regular uterine contractions are required for a diagnosis<br />

of preterm labour. Biochemical methods such as the fetal<br />

fibronectin test are also promising an earlier detection<br />

and diagnosis of preterm labour (see table 3).<br />

TABLE 2. CLINICAL APPROACH TO THE EVALUATION OF TRUE PRETERM LABOUR<br />

Physical examination General examination Abdominal examination<br />

Fetal assessment<br />

Sterile speculum examination<br />

Digital vaginal examination<br />

Infection screening<br />

Fetal lung maturity studies<br />

Urine toxicology screen Medications Substance abuse<br />

Immunopathology studies Antinuclear antibody Lupus anticoagulant<br />

Anticardiolipin antibody<br />

Ultrasound assessment<br />

Doppler velocimetry<br />

TABLE 3. CRITERIA FOR THE DIAGNOSIS<br />

OF PRETERM LABOUR<br />

• Gestational age: 20–37 weeks<br />

• Regular uterine contractions: ≥ 5–8 minutes apart,<br />

lasting 30 seconds<br />

• Amniotic membranes: ruptured. If intact, then at least<br />

one of the following is also required:<br />

• Cervical changes (by one observer)<br />

• Cervical effacement > 75%<br />

• Cervical dilation > 2 cm<br />

Post-partum investigations Placental pathology Post-partum infection screening<br />

Urinary tract studies<br />

Hysterosalpingogram<br />

2.2 Objectives<br />

The objectives of managing preterm labour are to<br />

minimise perinatal morbidity and mortality while<br />

preserving maternal health. 47 Initial management<br />

options include: (1) immediate delivery, e.g. fetal death<br />

or risk of maternal complications; (2) allow delivery to<br />

proceed naturally, e.g. labour is too advanced to<br />

inhibit; (3) delay delivery; (4) observe with no specific<br />

action; (5) transfer to a specialised tertiary care centre<br />

(see figure 2). 48 Passive management of preterm labour<br />

such as bed rest and sedation has not demonstrated<br />

any significant benefit, however, the addition of<br />

intravenous hydration may be effective short term by<br />

improving uterine blood flow and reducing myometrial<br />

activity.<br />

2.2.a Guidelines for managing preterm labour<br />

The Australian Government has produced the first set of<br />

recommendations for the management of preterm<br />

birth, “Clinical Practice Guidelines for Care Around<br />

Preterm Birth”. 49 The main points taken from these<br />

guidelines are as follows:<br />

• Target audience: healthcare professionals, educational<br />

establishments & consumers<br />

• The guidelines were based on an evidence-rating<br />

system so that informed decisions could be made on<br />

available evidence-based medicine, with Level I<br />

(evidence obtained form a systematic review of all<br />

relevant randomised, controlled trials [Cochrane<br />

Database]) being the ‘gold standard’<br />

• Key factors highlighted by these guidelines included:<br />

– Communication<br />

– Medical indicators of risk<br />

– Social and lifestyle factors<br />

– Risk assessment<br />

– Diagnosis<br />

– Clinical prevention<br />

– Tocolysis<br />

– Pharmacological treatments after PPROM<br />

– Pharmacological treatments to improve neonatal<br />

outcome<br />

– Place of birth and level of care<br />

– Care of the preterm infant<br />

– Support for families<br />

– Care and follow-up after leaving hospital<br />

– Prevention and treatment of specific conditions<br />

Figure 2.<br />

No cervical change<br />

Observe<br />

2.3 Treatment<br />

Threatened<br />

preterm<br />

labour<br />

Cervical<br />

change ie. 2 cm<br />

dilated or 80% effaced<br />

Tocolysis<br />

contraindicated<br />

or>34 completed weeks<br />

Allow<br />

delivery<br />

Diagnostic workup<br />

to determine cause<br />

Advanced labour or<br />

fetal distress<br />

Delivery<br />

(prior to<br />

next pregnancy)<br />

Tocolysis indicated<br />

and gestational age<br />

between 20-34 weeks<br />

Administer tocolytic<br />

and corticosteroids<br />

Clinical algorithm for the treatment of threatened<br />

preterm labour (McNamara and Vintziteos, 1997)<br />

Once the decision to delay delivery has been made, and<br />

providing there are no contraindications, the woman<br />

should receive active management involving tocolytic<br />

therapy. Acute episodes of preterm labour can be<br />

managed actively, e.g. tocolysis, although long-term<br />

monitoring and maintenance tocolytic therapy may be<br />

required in order to avoid a recurrence during the<br />

remainder of the pregnancy. However, there is no<br />

evidence to suggest that maintenance therapy is<br />

clinically effective at preventing a recurrence of<br />

preterm labour.<br />

Treatment for preterm labour consists of several<br />

therapeutic approaches. Tocolysis is regarded as firstline<br />

treatment together with corticosteroids (used to<br />

aid fetal lung maturation) and antibiotics (used as a<br />

prophylactic). Once uterine activity is inhibited or few<br />

contractions are recorded (ie. ≤4 per hour) after<br />

intravenous tocolysis, oral or intravenous maintenance<br />

tocolysis may be initiated, providing maternal and fetal<br />

monitoring is continued. If side effects are excessive,<br />

the infusion rate can be altered (titrated) accordingly.<br />

On the first day following successful tocolytic<br />

treatment, strict bed rest is usually advised and if after<br />

two days contractions are rare or absent, the woman’s<br />

condition is considered to be stable. Women with<br />

abnormal cervical change are usually observed for<br />

longer periods. Other factors leading to a longer<br />

duration of hospitalisation include a complicating<br />

diagnosis (e.g.. urinary tract infection) and low<br />

gestational age. In most uncomplicated cases,<br />

hospitalisation does not exceed 3–4 days.<br />

14 15


Chapter 2 Management of preterm labour<br />

2.4 Efficacy<br />

Evaluating the effectiveness of a therapeutic intervention<br />

has been complicated by the outcome variables used to<br />

define whether treatment is successful. The real impact of<br />

a therapeutic intervention should improve perinatal<br />

outcomes. In the past, clinicians have regarded outcomes<br />

such as small increases in birth weight and length of<br />

gestational age as indicators of success. However, the<br />

widespread use of pharmacological agents has not<br />

significantly reduced the rate of low-birthweight infants.<br />

This may be explained by the fact that only 10–20% of<br />

women at risk for preterm delivery are actually suitable<br />

candidates for the use of tocolytic agents. 9<br />

2.5 Tocolytic therapy<br />

The treatment of preterm labour by reducing or stopping<br />

preterm uterine contractions has been a therapeutic<br />

approach for more than 40 years. Intervention with<br />

pharmacological agents is the preferred first-line<br />

treatment of preterm labour. A number of tocolytic<br />

agents have been used over the last 40 years with varying<br />

levels of success.<br />

2.5.a Beta-agonists<br />

Beta-agonists are the most widely used tocolytic agents,<br />

although their use is primarily associated with treating<br />

asthma. In 1961, the non-selective beta-agonist<br />

isoxsuprine was the first agent proposed for the<br />

treatment of preterm contractions. However, because of<br />

its significant side effects, including maternal hypotension<br />

and fetal bradycardia, its clinical use was limited. The<br />

introduction of ritodrine over 20 years ago heralded the<br />

arrival of the first of the selective beta-agonists, which<br />

also include salbutamol, terbutaline and fenoterol.<br />

Mode of action<br />

Beta 1 and beta 2 receptors are located in most organ<br />

systems in the body. The beta 1 receptors mediate<br />

stimulatory effects and predominate in the heart, small<br />

intestine and adipose tissue. The beta 2 receptors mediate<br />

relaxant/inhibitory effects and are located in the uterus,<br />

blood vessels, bronchioles and liver.<br />

Beta-agonists exert their tocolytic effect by acting<br />

on intramembranous beta 2 receptors in the uterus,<br />

thereby relaxing the smooth muscle of the myometrium.<br />

Stimulation of these receptors activates the enzyme<br />

adenylate cyclase, which leads to an increase in<br />

intracellular cAMP. The cAMP acts as a secondary<br />

messenger, initiating a series of cellular reactions which<br />

ultimately reduce intracellular calcium levels, which in<br />

turn decreases the sensitivity of the contractile unit<br />

within the uterus wall to the effects of calcium and<br />

prostaglandins.<br />

Some beta-agonists have a greater selectivity for<br />

inhibition of uterine contractility than others, but<br />

none of those developed so far are entirely specific to<br />

beta 2 receptors in the uterus. It is the non-selectivity of<br />

beta-agonists that explains their unfavourable safety<br />

profile. The potential to stimulate other organ<br />

systems gives rise to serious systemic side effects to<br />

both the mother and fetus.<br />

Clinical efficacy<br />

All of the randomised, controlled trials of betaagonists<br />

are summarised in table 4. A meta-analysis of<br />

16 methodologically-acceptable trials using a database<br />

of 890 women indicated that beta-agonists delayed<br />

delivery for up to 48 hours, reduced the frequency of<br />

preterm birth and low birthweight babies, but without<br />

a reduction in perinatal mortality or the incidence of<br />

severe neonatal respiratory problems. 50 The Canadian<br />

Preterm Labor Investigators Group reported similar<br />

findings in women treated with oral and intravenous<br />

ritodrine in a placebo-controlled study. 51 More recent<br />

and larger placebo-controlled trials have also raised<br />

uncertainties regarding the use of maintenance betaagonist<br />

therapy. 52,53<br />

Terbutaline is another selective beta 2 -agonist in<br />

which studies have also presented conflicting results<br />

regarding prolonging pregnancy. Comparative trials<br />

with ethanol, 54 ritodrine and magnesium sulphate<br />

demonstrated a similar short-term efficacy in the<br />

treatment of preterm labour. 55 Another commonly<br />

used beta-agonist is salbutamol, although it does not<br />

confer any apparent advantages over the other betaagonists<br />

regarding effectiveness and side effects.<br />

Clinical safety<br />

Since beta-receptors are ubiquitous in body systems, any<br />

pharmacological intervention will be expected to<br />

induce a wide range of side effects. Even though the<br />

rationale for beta-agonist administration is based on<br />

their activity at receptors in the myometrium, they elicit<br />

a number of systemic side effects that are unrelated to<br />

their action in the uterus. Consequently, there are a<br />

variety of serious complications associated with their<br />

use in both mother and fetus.<br />

Maternal adverse effects<br />

The most significant side effects elicited by beta-agonists<br />

affect the cardiovascular system. Beta-agonists produce a<br />

general vasodilation resulting in systolic hypotension,<br />

which induces a compensatory increase in maternal<br />

cardiac output of up to 40–60%. 56 Although a decrease in<br />

peripheral vascular resistance occurs, this is offset by the<br />

large increase in cardiac output, which is further<br />

exacerbated by the antidiuretic effect of beta-agonists<br />

after long-term therapy. The resulting hypertension may<br />

contribute towards an underlying cardiac disease or<br />

TABLE 4. KEY RANDOMISED, CONTROLLED CLINICAL TRIALS OF CURRENT TOCOLYTIC AGENTS<br />

Study drug Control drug Total no. women Outcome parameter Success rate Reference<br />

Ritodrine Placebo 30 Delivery delayed >48 hours 50 vs. 55% [84]<br />

76 Delivery delayed >48 hours 73 vs. 69% [85]<br />

708 Delivery delayed >48 hours 79 vs. 65%*<br />

Mean days gained 28 vs. 25 days [49]<br />

63 Delivery delayed > 7 days 80 vs. 48%* [86]<br />

29 Delivery delayed > 7 days 29 vs. 27% [87]<br />

129 Delivery delayed > 14 days 57 vs. 67% [88]<br />

99 Mean days gained 34 vs. 25 days [89]<br />

100 Mean days gained 20 vs. 15 days [90]<br />

Terbutaline Placebo 38 Delivery delayed > 48 hours 54 vs. 38% [91]<br />

30 Delivery delayed > 7 days 87 vs. 27%* [92]<br />

37 Mean days gained 31 vs. 39 days [93]<br />

Ritodrine 58 Delivery delayed > 48 hours 73 vs. 77% [94]<br />

85 Delivery delayed > 48 hours 45 vs. 69% [53]<br />

99 Delivery delayed > 72 hours 80 vs. 67% [95]<br />

Salbutamol Placebo 144 Delivery 24 hours 94 vs. 22%* [98]<br />

Ritodrine 100 Delivery delayed >48 hours 94 vs. 83% [68]<br />

40 Mean days gained 26 vs. 28 days [99]<br />

Terbutaline 71 Mean days gained 43 vs. 41 days [100]<br />

MgSO4 Placebo 35 Delivery delayed > 48 hours 38 vs. 37% [91]<br />

Ritodrine 120 Delivery delayed > 48 hours 96 vs. 92% [101]<br />

Terbutaline 86 Delivery delayed > 48 hours 70 vs. 45% [53]<br />

Indomethacin 80 Delivery delayed > 48 hours 93 vs. 92% [102]<br />

Nifedipine Placebo 40 Delivery delayed > 48 hours 63 vs. 23%* [77]<br />

Ritodrine 40 Delivery delayed > 48 hours 63 vs. 54% [77]<br />

71 Delivery delayed > 7 days 67 vs. 63% [78]<br />

185 Delivery delayed > 7 days 62 vs. 50 %* [79]<br />

Terbutaline 53 Delivery delayed > 48 hours 68 vs. 67% [80]<br />

* Statistically significant (p


Chapter 2 Management of Preterm labour<br />

Fetal/neonatal adverse effects<br />

Beta-agonists readily cross the placental barrier and<br />

accumulate in the fetal circulation, increasing uteroplacental<br />

blood flow. This is supported by the fact that<br />

fetal tachycardia occurs upon administration of betaagonist<br />

therapy. Despite several reports of fetal<br />

cardiovascular changes, none of them are considered to<br />

be life threatening. Effects on fetal metabolic systems<br />

include hypoglycaemia, believed to be related to<br />

hyperinsulinaemia, and elevated growth hormone levels<br />

induced by beta-agonist stimulation of the fetal<br />

pancreas. Continuous beta-agonist therapy for greater<br />

than six weeks is associated with an increased risk of<br />

reactive hypoglycaemia after birth. 64<br />

Regarding the neonate, there have been no reports<br />

of adverse effects immediately following delivery,<br />

although there have been isolated cases of transient<br />

neonatal tachycardia and tachyarrhythmia reported in<br />

preterm neonates exposed to beta-agonist therapy. 65<br />

Fetal and neonatal adverse events are summarised in<br />

table 6.<br />

Infant follow up studies of up to 9 years have not<br />

indicated any significant difference in outcome between<br />

infants treated with beta-agonists and untreated<br />

preterm controls.<br />

Potential therapeutic benefit<br />

Ritodrine was licensed for use in the United States for<br />

nearly 20 years. However, its licence for preterm labour<br />

has since been rescinded by the US FDA. A review of the<br />

available data suggests perinatal improvements using i.v.<br />

ritodrine have only been documented in women prior to<br />

33 gestational weeks while the efficacy of current oral<br />

dosing remains questionable, with doubts concerning<br />

whether ritodrine can reach adequate serum levels to<br />

achieve tocolysis. 66 One of the problems associated with<br />

the clinical trials to date has been the poor selection<br />

criteria. It has been proposed that maternal and neonatal<br />

adverse effects could be minimised if women most likely<br />

to respond to treatment were included in the studies<br />

together with stricter adherence to the study protocols.<br />

These recommendations also apply to terbutaline<br />

and salbutamol. Newer, more selective beta 2 – agonists<br />

including fenoterol and hexoprenaline have displayed<br />

similar tocolytic effects as well as similar adverse<br />

effects, however they have not been properly<br />

evaluated in a controlled clinical setting.<br />

2.5.b Prostaglandin synthetase inhibitors<br />

Prostaglandin synthetase inhibitors are effective drugs<br />

for inhibiting preterm labour. The most commonly used<br />

prostaglandin synthetase inhibitor is indomethacin,<br />

which has an efficacy at least equivalent to betaagonists<br />

and generally causes fewer maternal side<br />

effects. However, it is less well tolerated by the fetus,<br />

therefore its use should be restricted below 30–32<br />

weeks gestation. Although indomethacin is used in<br />

Europe there are concerns over its safety profile in the<br />

fetus and neonate due to the incidence of significant<br />

cardiovascular side effects.<br />

Mode of action<br />

Prostaglandins have an integral role in the modulation<br />

of uterine contractility during preterm labour.<br />

Prostaglandins activate calcium channels within the<br />

myometrium and function as a secondary messenger,<br />

increasing intracellular calcium levels via the sarcoplasmic<br />

reticulum. Current prostaglandin synthetase inhibitors<br />

target the cyclooxygenase (COX) enzyme, which prevents<br />

the synthesis of prostaglandins from their precursor,<br />

arachidonic acid. There are two forms of the COX<br />

enzyme, an inducible (COX-2) and constitutive<br />

(COX-1) form. The production of COX-1 is relatively<br />

constant throughout pregnancy, whereas COX-2 rises<br />

markedly during labour. COX-1 predominates in fetal<br />

cardiovascular tissue while COX-2 is located in the fetal<br />

membranes and myometrium. Indomethacin affects both<br />

forms of the enzyme hence potential fetal side effects.<br />

Clinical efficacy<br />

Initial observations of women taking high-dose<br />

salicylates showed a greater frequency delivering post-<br />

term, with increases in mean gestational age. Initial<br />

studies with prostaglandin synthetase inhibitors in<br />

women requiring further tocolytic treatment after<br />

initial agents had failed showed encouraging results. 67<br />

Indomethacin was the first prostaglandin synthetase<br />

inhibitor to be used as a tocolytic agent. Placebocontrolled<br />

and comparative studies of indomethacin<br />

demonstrated that indomethacin was more effective<br />

than placebo in delaying preterm delivery during a 24-<br />

hour course of therapy. Regarding the comparative<br />

studies, indomethacin was at least as effective as betaagonists<br />

and magnesium sulphate with respect to delay<br />

of delivery (see table 4).<br />

Clinical safety<br />

Maternal adverse effects<br />

Maternal side effects associated with prostaglandin<br />

synthetase inhibitors are minimal. The most common<br />

include gastro-intestinal irritation and proctitis, but<br />

these are generally tolerated by women. Effects on<br />

platelet function are theoretically reversible, however<br />

there are conflicting reports of altered maternal<br />

bleeding times. 68 Prolonged therapy has been<br />

associated with altered T-cell suppressor activity in the<br />

mother, although the clinical significance of this<br />

remains to be seen. 69<br />

Fetal/neonatal effects<br />

One of the first complications of indomethacin therapy<br />

affects the renal system, resulting in a reduction in<br />

amniotic fluid (oligohydramnios). This is believed to occur<br />

in up to 10–30% of exposed fetuses. 70,71 Both short- and<br />

long-term exposure has been shown to significantly<br />

decrease urine production in the fetus. This was found to<br />

be dose dependent, 72 unrelated to the duration of<br />

therapy 71 and reversible upon cessation of therapy. High<br />

levels in the neonate are also associated with an<br />

increased risk of necrotizing enterocolitis which causes<br />

gastro-intestinal bleeding. 73<br />

There are also significant effects on the cardiovascular<br />

system, with 20–50% of exposed fetuses displaying<br />

vasoconstriction of the ductus arteriosus. This may last up<br />

to 2 hours following each dose, which resolves<br />

completely after 24 hours. 74 Furthermore, the effect is<br />

related to gestational age, with maximum constriction<br />

noted in gestational ages beyond 32 weeks. 75 This is<br />

thought to occur with both short- and long-term<br />

exposure. The exact mechanism by which this constriction<br />

can lead to in utero heart failure remains undetermined,<br />

however chronic constriction may result in hypertrophy<br />

of the fetal pulmonary vasculature leading to<br />

hypertension. Other potential risks associated with<br />

prostaglandin synthetase inhibitors are listed in table 7.<br />

Potential therapeutic benefit<br />

Although the prostaglandin synthetase inhibitors are<br />

regarded as one of the most effective tocolytic agents,<br />

their clinical use should be restricted to short-term<br />

administration only due to their significant and severe<br />

fetal and neonatal side effects. Alternatives to<br />

indomethacin include sulindac, which has the benefit<br />

of being unable to cross the placenta. It is, however, an<br />

inhibitor of both forms of the COX enzyme with the<br />

potential to cause maternal side-effects.<br />

There are also specific inhibitors designed to target<br />

only the COX-2 inducible form. Such drugs are still at an<br />

early stage of development, although an improved<br />

fetal safety profile is anticipated. A recent report on the<br />

use of nimesulide (COX-2 inhibitor) has, however, cast<br />

doubt on the safety of these drugs. A recent case study<br />

reported neonatal irreversible end stage renal failure<br />

after maternal ingestion of nimesulide used as a<br />

tocolytic for 6 weeks. 76<br />

2.5.c Magnesium sulphate<br />

In the USA, the familiarity of administering magnesium<br />

sulphate for the management of preeclampsia and an<br />

apparent lack of side effects helped encourage its use<br />

as a tocolytic agent.<br />

TABLE 5. POTENTIAL MATERNAL COMPLICATIONS ASSOCIATED WITH<br />

BETA-AGONIST ADMINISTRATION [BESINGER & IANNUCCI, 1997]<br />

Cardiovascular Metabolic Neuromuscular Other Multifactorial<br />

maternal death<br />

• Cardiac arrhythmia • Diabetic ketoacidosis • CNS stimulation • Agranulocytosis • Hyperthyroid crisis<br />

• Cardiac stimulation • Euglycaemic acidosis • Cerebral vasospasm/ • Altered thyroid function • Pulmonary oedema<br />

• Hypotension • Glucose intolerance ischaemia • Bone marrow suppression • Underlying cardiac<br />

• Myocardial infarction • Hyperglycaemia • Neuromuscular • Cutaneous vasculitis arrythmia<br />

• Myocardial ischaemia • Hyperinsulinaemia alterations • Elevated transaminase levels • Unrecognised<br />

• Peripartum cardiomyopathy • Hyperlactacidaemia • Tremor • Erythema multiforme cardiac disease<br />

• Peripheral vasodilation • Hypocalcaemia • Haemolytic anaemia<br />

• Pulmonary oedema • Hypoglycaemia<br />

• Sodium/water retention • Hypokalaemia<br />

• Tachycardia<br />

• Hypomagnesaemia<br />

TABLE 6. POTENTIAL FETAL/NEONATAL COMPLICATIONS ASSOCIATED WITH<br />

BETA-AGONIST ADMINISTRATION [BESINGER & IANNUCCI, 1997]<br />

Cardiac Metabolic Other<br />

• Altered utero-placental blood flow<br />

• Bradycardia<br />

• Cardiac arrhythmia<br />

• Cardiac stimulation<br />

• Cardiovascular decompensation<br />

• Exacerbation of fetal hypoxia<br />

• Myocardial ischaemia<br />

• Myocardial necrosis<br />

• Peripheral vasodilation<br />

• Septal hypertrophy<br />

• Tachycardia<br />

• Hyperbilirubinaemia<br />

• Hypercholesterolaemia<br />

• Hyperglycaemia<br />

• Hyperinsulinaemia<br />

• Hypocalcaemia<br />

• Hypoglycaemia<br />

• Intraventricular<br />

haemorrhage<br />

• Leukaemoid reaction<br />

• Renal insufficiency<br />

• Retinopathy<br />

18 19


Chapter 2 Management of preterm labour<br />

Mode of action<br />

Magnesium sulphate has been known to reduce uterine<br />

contractility for a long time. The basis of its tocolytic<br />

action is still unknown, although it is presumed to<br />

involve competition with calcium for entry into muscle<br />

cells through voltage-gated calcium channels. Another<br />

theory is that magnesium competitively binds to calcium<br />

storage sites in the myometrial endoplasmic reticulum.<br />

Both mechanisms of action will inhibit the cellular influx<br />

of calcium necessary for smooth muscle contraction.<br />

Clinical safety<br />

A large comparative study reported treatment<br />

discontinuation due to adverse drug effects in 38%<br />

of patients treated with beta-agonists and only<br />

2% treated with magnesium sulphate. 55 However,<br />

magnesium is a non-specific agent and will compete<br />

with calcium throughout the body. Consequently, there<br />

is the potential for a number of widespread side<br />

effects. An increase in the availability of magnesium<br />

sulphate and its use in clinical practice has led to a<br />

greater number of reports of side effects, which has<br />

cast doubt on its continued use. (see table 8 for<br />

summary of maternal/fetal and neonatal effects).<br />

Maternal adverse effects<br />

There is a dramatic difference in the rate and type of<br />

maternal side-effects reported. While initial reports<br />

seemed to be reassuring there are now calls for a<br />

re-evaluation of its use as a tocolytic in clinical practice.<br />

The cardiovascular safety of magnesium sulphate has<br />

recently been challenged. There have been several<br />

cases of pulmonary oedema and myocardial<br />

ischaemia reported, which suggest further potentially<br />

unrecognised myocardial complications. Significant<br />

prolongations in QT intervals may also be indicative of<br />

potential life-threatening arrhythmias.<br />

Fetal/neonatal effects<br />

Significant accumulation of magnesium sulphate in the<br />

fetal circulation may contribute to reports of suppressed<br />

fetal breathing and movement, and neonatal<br />

respiratory depression. Another recent study suggests<br />

an increase in total paediatric mortality due to exposure<br />

to magnesium sulphate. 77 A subsequent meta-analysis<br />

suggested that magnesium sulphate may account for<br />

40% of total paediatric mortality, which translates to an<br />

increase in the absolute number of infant deaths by<br />

about 4800 every year in the USA. 78<br />

Potential therapeutic benefit<br />

The evidence gathered so far for magnesium sulphate is<br />

often inconsistent and unreliable. At low doses, the<br />

side effects are minimal although its efficacy is no better<br />

than beta-agonists. At higher doses, it may be able to<br />

delay delivery for longer but with a subsequent increase<br />

in the severity of side effects. The likeliest role proposed<br />

for magnesium sulphate therapy is for short-term<br />

control of uterine activity, which provides a good<br />

margin of safety.<br />

2.5.d Calcium channel blockers<br />

Calcium channel blockers have proven to be as effective<br />

as beta-agonists and magnesium sulphate in delaying<br />

preterm labour. 79,80 However, the safety of the calcium<br />

channel blockers is still under scrutiny. Since they are<br />

already established in the treatment of several<br />

cardiovascular diseases, such as hypertension and<br />

angina, it is likely that they will cause cardiovascular<br />

side effects. Calcium channel blockers used as tocolytics<br />

include nifedipine and nicardipine.<br />

Mode of action<br />

The calcium channel blockers have a powerful uterine<br />

relaxant effect, which is mediated by inhibiting the<br />

cellular influx of calcium by blocking the voltage-gated<br />

channels present in the myometrium. Animal and<br />

human in vitro models have demonstrated a<br />

suppression of prostaglandin and oxytocin-induced<br />

uterine contractions. Calcium channel blockers have yet<br />

to be approved for use as a tocolytic agent.<br />

Clinical efficacy<br />

Early studies with nifedipine were encouraging, with<br />

subsequent placebo-controlled and comparative<br />

studies revealing an efficacy similar to beta-agonists in<br />

the short and long term. 81,82 (Table 9 summarises<br />

TABLE 7. POTENTIAL MATERNAL AND FETAL/NEONATAL COMPLICATIONS ASSOCIATED WITH<br />

PROSTAGLANDIN SYNTHETASE INHIBITOR ADMINISTRATION [BESINGER & IANNUCCI, 1997]<br />

Maternal complications<br />

• Altered bleeding times<br />

• Altered immune response<br />

• Antipyresis<br />

• Gastro-intestinal irritation<br />

• Platelet dysfunction<br />

• Renal dysfunction<br />

Fetal/neonatal complications<br />

• Altered cerebral blood flow<br />

• Altered immune response<br />

• Constriction of ductus arteriosus<br />

• Cystic brain lesions<br />

• Exacerbation of congenital heart disease<br />

• Fetal hydrops<br />

• Hyperbilirubinaemia<br />

• Intraventricular haemorrhage<br />

• Isolated ileal perforation<br />

• Necrotising enterocolitis<br />

• Oligohydramnios<br />

• Oliguria<br />

• Patent ductus arteriosus<br />

• Persistent pulmonary<br />

hypertension<br />

• Renal dysfunction<br />

maternal/fetal and neonatal side effects). Until larger<br />

clinical trials have been undertaken, these preliminary<br />

findings remain unsupported.<br />

Clinical Safety<br />

Maternal adverse effects<br />

Due to their clinical indication, the cardiovascular side<br />

effects of the calcium channel blockers are well known.<br />

In pregnant women they are used to control<br />

preeclampsia, however they are not recommended as a<br />

first-line therapy for pregnancy-induced hypertension.<br />

Reports of reflex tachycardia and reduced atrioventricular<br />

conduction as a consequence of<br />

hypotension are common. Despite these concerns their<br />

effect on the cardiovascular system appears to be less<br />

severe compared with beta-agonists. Theoretically,<br />

calcium channel blockers may predispose to pulmonary<br />

oedema, although overall clinical experience with<br />

nifedipine in pregnant women suggests a minimal<br />

number of cardiovascular and metabolic side effects.<br />

Fetal/neonatal effects<br />

Fetal cardiovascular function appears to be essentially<br />

unchanged with short-term nifedipine therapy. Of the<br />

limited number of studies conducted so far, few have<br />

demonstrated any significant fetal or neonatal<br />

morbidity. However, there is still concern regarding<br />

safety due to the lack of adequate data. Furthermore,<br />

calcium channel blockers can cross the placenta and<br />

affect fetal oxygen availability and blood flow in animal<br />

studies. 83 This observation is supported by animal<br />

experiments in which nifedipine caused hypoxia and<br />

acidosis in the sheep fetus. The deterioration of blood<br />

gases was out of proportion with the transient decrease<br />

in uroplacental blood flow, demonstrating that another<br />

mechanism exists during nifedipine infusion. 84 Such<br />

observations have led to the suggestion that sublingual<br />

nifedipine should be removed from the market. 85<br />

Potential therapeutic benefit<br />

There is clinical evidence to suggest calcium channel<br />

blockers may have the potential to become an acceptable<br />

tocolytic. However, the safety of this class of drug does<br />

require further scrutiny. It is also worth noting that none<br />

of the tocolytics mentioned so far have undergone<br />

specific evaluation/ development for preterm labour.<br />

(Table 4 summarises the key comparative trials that have<br />

been conducted on the current tocolytics). 86,87<br />

2.5.e Oxytocin antagonists<br />

Until recently, oxytocin was regarded as having a<br />

minor role in the initiation of human labour. However,<br />

increasing evidence suggests that oxytocin is one of<br />

the key components in the initiation of labour. It has<br />

been shown to have a direct role in uterine<br />

contractility – by stimulating the myometrium – and an<br />

indirect role – by increasing the production of prostaglandins<br />

in the decidua. This discovery led to the<br />

development of oxytocin antagonists designed to<br />

suppress the dual effect of oxytocin.<br />

The most promising of the oxytocin antagonists is<br />

atosiban. It is an analogue of oxytocin and has been<br />

rationally designed to compete with endogenous<br />

oxytocin at both the myometrial and decidual oxytocin<br />

receptors. Clinical studies have shown it to be an effective<br />

and safe tocolytic agent.<br />

TABLE 8. POTENTIAL MATERNAL AND FETAL/NEONATAL COMPLICATIONS ASSOCIATED WITH<br />

MAGNESIUM SULPHATE ADMINISTRATION [BESINGER & IANNUCCI, 1997]<br />

Maternal complications<br />

• Altered cardiac conduction<br />

• Cardiac arrest<br />

• Chest tightness<br />

• Dysphagia/ aspiration<br />

• Generalised muscle weakness<br />

• Hypocalcaemia<br />

• Hypothermia<br />

• Hyperkalaemia<br />

• Neuromuscular blockade<br />

• Ophthalmological alterations<br />

• Osmotic diuresis<br />

• Pulmonary oedema<br />

• Respiratory arrest<br />

• Respiratory depression<br />

• Subendocardial ischaemia<br />

• Water retention<br />

Fetal/neonatal complications<br />

• Altered biophysical activities<br />

• Altered heart rate variability<br />

• Amniotic accumulation<br />

• Hypermagnesaemia<br />

• Hypotonia<br />

• Meconium ileus<br />

• Neonatal rickets<br />

• Respiratory depression<br />

TABLE 9. POTENTIAL MATERNAL AND FETAL/NEONATAL COMPLICATIONS ASSOCIATED WITH<br />

CALCIUM CHANNEL BLOCKER ADMINISTRATION [BESINGER & IANNUCCI, 1997]<br />

Maternal complications<br />

• Altered cardiac conduction<br />

• Cutaneous vasodilation<br />

• Drug-induced hepatitis<br />

• Fluid retention<br />

• Hypocalcaemia<br />

• Hypoglycaemia<br />

• Hypotension<br />

• Tachycardia<br />

Fetal/neonatal complications<br />

• Altered uteroplacental<br />

blood flow<br />

• Tachycardia<br />

20<br />

21


3<br />

Tractocile ®<br />

22<br />

23


Chapter 3 Tractocile ® Figure 3. Peptide structures of oxytocin and atosiban<br />

3.1 Rationale for development<br />

Oxytocin is now recognised as a potent agent that has a<br />

central role in the initiation of myometrial contractions<br />

during late pregnancy. The development of an analogue<br />

of oxytocin would, in theory, either imitate or block its<br />

effect. The rationale for the development of Tractocile ®<br />

(atosiban), trademark of Ferring BV, The Netherlands<br />

was based on the production of a novel compound<br />

which was highly specific for the uterus, but with a<br />

limited number or an absence of systemic side effects.<br />

The ideal outcome would be to produce a compound<br />

that is effective, safe and well tolerated.<br />

In 1960, Law and du Vigneaud first demonstrated<br />

partial uterotonic antagonism by modifying the oxytocin<br />

molecule at position 2. 88 Further changes to the parent<br />

molecule produced a series of analogues displaying full<br />

oxytocin antagonism. Later studies investigated the<br />

effect of O-alkylation at position 2 of the oxytocin<br />

molecule and the effect such analogues had on pregnant<br />

and non-pregnant human myometrial tissue and in<br />

animal models in vivo. 89 One of these analogues<br />

(a deaminated, O-Ethyl substitution at position 2)<br />

proved to be a full antagonist in vitro, however, showed<br />

only a partial effect when tested in women. This led to<br />

further developments until finally modifications at<br />

positions 1, 2, 4 and 8 of the oxytocin molecule produced<br />

an analogue, atosiban [1-deamino-2-D-Tyr-(O-Et)-4-Thr-<br />

8-Orn]-OT, with a high receptor affinity for the oxytocin<br />

receptor in vitro, and rat uterus in vitro and in vivo<br />

(figure 3). Further studies demonstrated atosiban to be<br />

the most potent analogue developed for inhibiting<br />

uterine contractions initiated by oxytocin in human<br />

pregnant and non-pregnant tissue, and in oestrus rats in<br />

vivo. 90 Atosiban was shown to lack any agonist properties<br />

and was therefore selected for further clinical<br />

investigation into its role as an oxytocin antagonist.<br />

3.2 Mechanism of action<br />

Oxytocin is believed to initiate myometrial contractility by<br />

a direct effect on membrane-bound receptors leading to<br />

an increase in intracellular calcium (see figure 4). Oxytocin<br />

also acts indirectly by stimulating the release of<br />

prostaglandins in decidual and fetal membranes, thereby<br />

contributing further to myometrial contractions and<br />

initiating cervical ripening. Tractocile ® acts by competing<br />

with oxytocin for receptors in the myometrium, and<br />

potentially in the decidual and fetal membranes as well.<br />

This results in a dose-dependent inhibition of uterine<br />

contractility and, furthermore, studies have shown a<br />

reduction in oxytocin-mediated prostaglandin release.<br />

Studies have also shown that Tractocile ® has an equal if<br />

not a greater affinity for vasopressin receptors compared<br />

with oxytocin receptors due to their close chemical<br />

homology. 91,92 This does not represent a clinical problem<br />

regarding vasopressin-like side effects because there is a<br />

H<br />

6<br />

7<br />

8<br />

1 2<br />

Cys<br />

Cys<br />

Pro<br />

Leu<br />

Tyr<br />

substantial increase in the expression of oxytocin<br />

receptors and no change in vasopressin receptor<br />

numbers in the uterus during labour.<br />

Further studies have investigated Tractocile ® on<br />

receptor binding and its effect on intracellular<br />

secondary messengers. Lopez-Bernal et al 93 showed a<br />

dose-dependent inhibition of oxytocin-stimulated<br />

inositol phosphate production, and Thornton et al 94<br />

demonstrated the abolishment of oxytocin-induced<br />

increases in intracellular Ca 2+ and spontaneous<br />

fluctuations in calcium. This final effect suggests that<br />

an additional intracellular process may be attributed<br />

to Tractocile ® following receptor binding.<br />

The utero-specificity of Tractocile ® provides a better<br />

safety profile and an effective alternative to the<br />

current tocolytics responsible for multi-organ side<br />

effects. However, because of its inhibitory effect on<br />

vasopressin receptors, there is the potential to cause a<br />

number of hypothetical secondary effects such as<br />

water resorption by the kidney, vasoconstriction and<br />

stimulation of adrenocorticotrophin hormone. Clinical<br />

studies have failed to demonstrate any of these effects<br />

and, on the contrary, have shown Tractocile ® to be a<br />

well-tolerated tocolytic agent. The efficacy of<br />

Tractocile ® is expected to be at least comparable to<br />

other tocolytic agents due to the multifactorial<br />

aetiologies responsible for preterm labour. Tractocile ®<br />

will affect one, if not more, of the pathways associated<br />

with the initiation of preterm labour.<br />

3.3 Chemistry of Tractocile ®<br />

3<br />

Gin<br />

Asn<br />

4<br />

5<br />

Gly<br />

9<br />

Oxytocin<br />

Ile<br />

NH 2<br />

Tractocile ® contains atosiban, a synthetic cyclic<br />

nonapeptide. Its chemical name is 1-(3-mercaptopropanoic<br />

acid)-2-(O-ethyl-D-tyrosine)-4-L-threonine-8-L ornithineoxytocin,<br />

with a formula C43H67N11O12S2. It has a<br />

molecular mass of 993.5 Daltons and is available as an<br />

acetate salt in the form of a whitish lyophilised amorphous<br />

powder. Potential isomerism can occur since the molecule<br />

contains a total of nine chiral centres. All of the amino acids<br />

H<br />

6<br />

7<br />

8<br />

1 2<br />

Mpa<br />

Cys<br />

Pro<br />

Orn<br />

Tyr<br />

D<br />

3<br />

Thr<br />

Asn<br />

4<br />

5<br />

Gly<br />

9<br />

Atosiban<br />

Ile<br />

NH 2<br />

O<br />

Oxytocin<br />

receptor<br />

are in the L-form, except tyrosine which exists in the<br />

D-form. The molecule can exist as either the Cis or<br />

Trans isomer at the cysteine-proline link, but the<br />

Trans configuration is the most common. Eleven<br />

diastereoisomers with one chiral centre can potentially<br />

be formed during the manufacture of Tractocile ® but it<br />

is possible to produce molecules with multiple chirality.<br />

Tractocile ® is highly soluble in water and is presented<br />

as a solution for parenteral administration at a<br />

concentration of 7.5 mg/<strong>ml</strong>.<br />

3.4 Preclinical studies<br />

O<br />

Ca 2+<br />

Oxytocin<br />

Oxytocin<br />

binding<br />

increases<br />

intracellular<br />

Ca 2+ calcium<br />

Ca 2+<br />

Ca 2+ Increase in<br />

myometrial Ca 2+<br />

contractility<br />

Uterine contractions<br />

Figure 4. Schematic diagram of how Tractocile ®<br />

competitively inhibits oxytocin at its receptor<br />

3.4.a Receptor binding: affinity, specificity<br />

A number of preclinical toxicological and receptor<br />

binding studies were conducted in vitro and in vivo<br />

in animals, and in vitro in human tissue preparations.<br />

The receptor affinities of Tractocile ® , vasopressin<br />

and oxytocin were demonstrated to be similar in<br />

myometrial membrane preparations. 91 Tractocile ® was<br />

also able to displace vasopressin from its receptor site.<br />

In vitro, in human myometrium, Tractocile ® was shown<br />

to completely inhibit oxytocin-induced increases in<br />

intracellular calcium but did not prevent this increase<br />

when it was either potassium- or prostaglandin E 2 -<br />

induced. 94,95 This indicates that Tractocile ® is specific to<br />

the action of oxytocin.<br />

Another observation in rats after chronic treatment<br />

with oxytocin demonstrated a down-regulation in<br />

receptor numbers with continued exposure to<br />

oxytocin, whereas chronic exposure to Tractocile ®<br />

appeared to increase the number of receptors and the<br />

affinity for the receptors. 96<br />

3.4.b Pharmacological activity in vitro<br />

The effect of Tractocile ® has been evaluated in vitro in<br />

isolated preparations from normal or pregnant animal<br />

O<br />

Ca 2+<br />

T<br />

T<br />

Ca 2+<br />

Tractocile ®<br />

Ca 2+<br />

O<br />

Tractocile ® blocks<br />

increase in<br />

intracellular<br />

calcium<br />

Ca 2+<br />

Ca 2+<br />

Inhibition of<br />

myometrial<br />

contractility<br />

Inhibition of contractions<br />

and human uterus. Tractocile ® inhibited oxytocin-induced<br />

contractions in a dose dependent, competitive fashion. 97<br />

The activity of Tractocile ® was also tested in non-pregnant<br />

and term pregnant human myometrial tissue. 90 No effect<br />

was seen on the non-pregnant strips, although the effect<br />

of vasopressin was antagonised. On myometrial strips<br />

obtained following elective Caesarean section from<br />

women at term, Tractocile ® displayed a greater efficacy,<br />

suppressing oxytocin-induced contractions before the<br />

beginning of labour, while during labour the<br />

spontaneous activity of the muscle was more sensitive to<br />

the antagonistic effect of Tractocile ® . 98<br />

3.4.c In vivo effect on uterine contractility<br />

The ability of Tractocile ® to inhibit uterine contractility<br />

has been investigated in the rat, guinea pig and monkey.<br />

In the pregnant monkey, premature contractions and<br />

anticipated deliveries can be induced by continuous<br />

infusion of androstenedione. Tractocile ® (6 µg/kg/min)<br />

inhibited the uterine muscle contractions induced by<br />

androstenedione in all pregnant animals. 99<br />

3.4.d Effect on labour duration<br />

In rats, Tractocile ® (s.c. every 5 mins) given 1 hour after<br />

delivery of the first pup delayed the birth of the second<br />

pup in a dose-dependent fashion. Continuous infusion<br />

(50 µg/min) delayed birth by approximately 1 hour<br />

beyond that observed in the control group. 100<br />

3.4.e Effect on prostaglandin release<br />

Tractocile ® was infused into pregnant ewes to investigate<br />

the effect on circulating maternal and fetal<br />

prostaglandins. The injection of oxytocin induced the<br />

production of maternal PGF 2a . This response was reduced<br />

in a dose-dependent manner when Tractocile ® was<br />

infused two hours earlier. 101<br />

Regarding the safety data from the preclinical studies,<br />

Tractocile ® did not have any significant effect on the CNS,<br />

cardiovascular, pulmonary, urinary or metabolic systems.<br />

Ca 2+<br />

M Y O M E T R I U M<br />

24 25


Chapter 3 Tractocile ®<br />

3.5 Clinical pharmacodynamic studies<br />

There have been several open label, uncontrolled studies<br />

designed to investigate the effect of Tractocile ® on<br />

uterine contractions. Åkerlund et al 102 and Hauksson et<br />

al 103 administered Tractocile ® to healthy, non-pregnant<br />

women at a dose of 10 µg/kg, either as a repeat dose or a<br />

single bolus. Both studies showed an inhibition of uterine<br />

contractions during menstruation and vasopressininduced<br />

contractions. A similar study by Åkerlund 104<br />

showed Tractocile ® (10 µg/kg; single dose) to be capable<br />

of relieving the acute symptoms of dysmenorrhoea<br />

compared to placebo in a randomised, double-blind,<br />

placebo-controlled study. Further studies were aimed at<br />

evaluating the ability of Tractocile ® to inhibit uterine<br />

contractions in women with preterm labour. Åkerlund et<br />

al showed a total inhibition in 46% (6/13) of women<br />

receiving >25 µg/min for >2 hours. 105 This was supported<br />

by results from Andersen et al who showed a total<br />

inhibition of preterm contractions in 50% (6/12) of<br />

women on >25 µg/min for 8–13 hours. 106 Investigations<br />

into effects on lipid or carbohydrate metabolism, in<br />

which a single bolus dose of 10 µg/kg was administered<br />

to healthy, non-pregnant women, showed that<br />

Tractocile ® had no clinically significant effect. 107<br />

3.6 Clinical pharmacokinetic studies<br />

A total of 13 preclinical trials designed to investigate<br />

the pharmacokinetics of Tractocile ® are briefly described.<br />

The optimal dose, route of administration and important<br />

pharmacokinetic parameters were determined (Tables<br />

10–12). The bioavailability of Tractocile ® was compared<br />

between two parenteral routes of administration in early<br />

clinical studies by Åkerlund et al 105 and Lundin et al 108 . The<br />

clearance rate and half-life were evaluated for the i.v.<br />

route and bioavailability determined for i.n. (intranasal)<br />

Tractocile ® . Both studies showed the intravenous route to<br />

be more favourable. In the Åkerlund study, the<br />

bioavailability of i.n. Tractocile ® was estimated to be<br />

5.5%. A maximal plasma concentration was achieved<br />

within 2 minutes via i.v. administration, explaining the<br />

almost immediate inhibition observed in uterine<br />

contractions. The study by Lundin et al measured peak<br />

plasma concentrations in 2–8 minutes after i.v. compared<br />

with 10–45 minutes with i.n. routes. 108 Another study by<br />

Lundin et al evaluated the pharmacokinetic parameters<br />

for i.v Tractocile ® administered as a bolus dose. 109 These<br />

tended to be much higher than the previous studies, due<br />

to a more efficient method, with the half-life estimated<br />

at 39 minutes and the clearance rate approximately<br />

23 l/hr. The bioavailability of Tractocile ® after<br />

subcutaneous administration was also studied in healthy,<br />

non-pregnant female subjects receiving either 7.5 mg i.v.<br />

or s.c. 110 Absorption by the s.c. route was rapid, taking<br />

approximately 30 minutes to reach peak plasma<br />

concentration, and bioavailability was 97%. To observe<br />

any possible differences regarding how the body<br />

processed Tractocile ® in its suggested clinical role,<br />

pharmacokinetic parameters were evaluated in women<br />

with preterm labour. Tractocile ® infused at 300 µg/min<br />

TABLE 10. BASIC PHARMACOKINETIC PROPERTIES OF <strong>TRACTOCILE</strong> ®<br />

Study Aim of study Subjects (no./type) Dose (µg/kg) Effect of Tractocile ®<br />

Åkerlund 1986<br />

Lundin 1986<br />

Lundin 1993<br />

Zinny 1995<br />

Goodwin 1995<br />

Describe plasma<br />

concentratrion and<br />

body clearance with i.v.<br />

or i.n. administration<br />

Describe plasma<br />

concentration and<br />

body clearance with<br />

i.v. or i.n.<br />

Describe plasma<br />

concentration and<br />

body clearance<br />

Describe plasma<br />

concentration and<br />

bioavailability after<br />

s.c. injection<br />

Describe plasma<br />

concentration, body<br />

clearance and<br />

inhibitory activity<br />

11/Healthy,<br />

non-pregnant<br />

11/Healthy,<br />

non-pregnant<br />

8/Healthy,<br />

non-pregnant<br />

18/Healthy<br />

non-pregnant<br />

8/ Preterm<br />

labour<br />

10 i.v., 100 i.n<br />

repeat bolus<br />

10 i.v., 100 i.n.<br />

single bolus<br />

5 mg i.v.<br />

single bolus<br />

7.5 mg i.v., 7.5, 15,<br />

30 mg s.c.<br />

single bolus<br />

300 µg/min i.v.<br />

6–12 hours<br />

Inhibition of contractions<br />


Chapter 3 Tractocile ®<br />

The clinical dose of Tractocile ® for infusion was<br />

proposed as 300 µg/min. Doses higher than this level<br />

600<br />

have not been investigated in clinical trials. This current<br />

rate provides a concentration of 450 ng/<strong>ml</strong> which<br />

500<br />

theoretically should be sufficient to saturate every<br />

oxytocin receptor in the uterus. A bolus dose of 6.75 mg<br />

400<br />

achieves immediate uterine quiescence, which is then<br />

maintained at a steady state concentration during the<br />

infusion period ensuring optimal exposure of the uterus<br />

to the drug (see figure 5).<br />

Valenzuela et al determined the degree of placental<br />

300<br />

200<br />

100<br />

transfer of Tractocile ® to the fetus. 116 This was conducted<br />

in healthy, pregnant women receiving 300 µg/min over<br />

0<br />

3.5 to 8 hours. Fetal plasma concentrations were 12% of<br />

the mother’s levels, and the ratio between fetus and<br />

Time (hours)<br />

mother did not change during infusion.<br />

<strong>Bolus</strong> Infusion<br />

The extent of plasma protein and erythrocyte<br />

Group (mg) (µg/min)<br />

binding was investigated in plasma from fasted,<br />

1<br />

6.75 1300<br />

pregnant women. Labelled Tractocile ® was added to<br />

2<br />

placebo 300<br />

their plasma in vitro at clinically relevant concentrations<br />

and was subsequently measured for free and unbound<br />

levels. Results indicated a moderate degree of protein<br />

binding in the range of 45.7 to 47.5%. Regarding<br />

erythrocyte binding, there was no partitioning into red<br />

blood cells. 117<br />

Finally, regarding pharmacokinetics, two studies<br />

were conducted to investigate the metabolism of<br />

Tractocile ® . Unchanged Tractocile ® and two metabolites,<br />

one major and one minor, were identified in the plasma<br />

and urine of pregnant subjects. The major metabolite<br />

underwent further tests regarding its ratio in maternal<br />

and fetal plasma, with approximately a three-fold<br />

higher concentration in the mother. 118,119 3<br />

4<br />

Dose<br />

2.0<br />

0.6<br />

selection<br />

100<br />

process for Figure 5. Tractocile ®<br />

Atosiban plasma concentration (ng/<strong>ml</strong>)<br />

0 2 4 6 8<br />

Clinical efficacy of Tractocile ®<br />

30<br />

4<br />

28


Chapter 4 Clinical efficacy of Tractocile ®<br />

4.1 Phase II clinical experience<br />

Dose ranging and initial efficacy studies of atosiban were<br />

investigated in a clinical setting in three phase II trials.<br />

The first of this series of studies was aimed at evaluating<br />

the efficacy and safety of atosiban in decreasing or<br />

arresting uterine contractility in women with threatened<br />

preterm labour. 120 A double-blind, placebo-controlled,<br />

randomised trial investigated both primary and<br />

secondary efficacy variables in 118 women (atosiban<br />

n=59, placebo n= 59). The primary variable involved<br />

evaluating the percentage change in the number of<br />

uterine contractions occurring in a one-hour observation<br />

period compared with the subsequent two-hour<br />

treatment period. The secondary efficacy parameters<br />

assessed the proportion of women whose contractions<br />

stopped during treatments, and changes in cervical<br />

dilation and effacement. During treatment 77% of<br />

women receiving atosiban experienced a reduction of<br />

≥40% in the number ofuterine contractions compared<br />

with 32% of placebo-treated women. In women with a<br />

gestational age ≥28 weeks, the response rate in relation<br />

to a reduction of contractions was greater in the<br />

atosiban group and above 31 weeks this difference was<br />

statistically significant (p


Chapter 4 Clinical efficacy of Tractocile ®<br />

(Table 14, Figure 6). Secondary outcomes, such as<br />

reduction in uterine contraction rate and mean<br />

gestational age, were similar in both groups. It was<br />

concluded that Tractocile ® and ritodrine were<br />

comparable with regard to delaying delivery. However,<br />

in terms of tocolytic efficacy and tolerability, Tractocile ®<br />

was superior to ritodrine.<br />

4.2.b Tractocile ® vs. salbutamol<br />

This randomised study enrolled 240 women with<br />

preterm labour who received either Tractocile ® or<br />

salbutamol (2.5–45 µg/min). As a measure of tocolytic<br />

effectiveness, Tractocile ® and salbutamol were<br />

comparable in terms of the number of women<br />

remaining undelivered after 48 hours and 7 days<br />

(Table 15). When the tocolytic efficacy and tolerability<br />

outcome was considered, Tractocile ® was found to be<br />

statistically significantly superior to salbutamol after<br />

7 days of starting treatment. The number of women<br />

remaining undelivered and not requiring an alternative<br />

tocolytic therapy within 7 days was 58.8% in the<br />

Tractocile ® group and 46.3% in the salbutamol group<br />

(p=0.02) (Table 15, Figure 6). Secondary outcomes were<br />

again comparable between treatment groups.<br />

4.2.c Tractocile ® vs. terbutaline<br />

In this comparative study, there were 244 women with<br />

preterm labour randomised to receive either Tractocile ®<br />

or terbutaline. Terbutaline was administered within its<br />

recommended dosage range of 5–20 µg/min. The<br />

primary outcome measuring the number of women<br />

remaining undelivered at 48 hours and 7 days showed<br />

Tractocile ® and terbutaline to be comparable (Table 16).<br />

In terms of tocolytic efficacy and tolerability, Tractocile ®<br />

was regarded as being at least as effective as<br />

terbutaline. The proportion of women remaining<br />

undelivered without requiring alternative tocolytic<br />

therapy after 7 days was 55.6% in the Tractocile ® group<br />

and 43.4% in the terbutaline group (p=0.08) (Table 16,<br />

Figure 6.<br />

Beta-agonist better<br />

Ritodrine<br />

Terbutaline<br />

Salbutamol<br />

Pooled<br />

TABLE 15. TOCOLYTIC EFFECTIVENESS AND TOCOLYTIC EFFICACY & TOLERABILITY<br />

OF <strong>TRACTOCILE</strong> ® AND SALBUTAMOL AT 48 HOURS AND 7 DAYS<br />

Number of women (%)<br />

Tractocile ® Salbutamol Odds 95% CI p value<br />

(n=119) (n=121) ratio<br />

Tocolytic effectiveness<br />

undelivered at 48 h 111 (93.3) 115 (95.0) 0.78 0.24–2.50 0.67<br />

undelivered at 7 days 107 (89.9) 109 (90.1) 1.04 0.39–2.77 <strong>0.9</strong>4<br />

Tocolytic efficacy &<br />

tolerability*<br />

no failure at 48 h 95 (79.8) 91 (75.2) 1.58 0.85–2.95 0.15<br />

no failure at 7 days 70 (58.8) 56 (46.3) 1.89 1.10–3.24 0.02<br />

*Treatment failure = delivery within the time interval or need for alternative tocolysis<br />

0 1 2 3 4 5<br />

Odds ratio<br />

Homogeneity of centre effects test:<br />

χ 2 =36.29, df=42, p=0.72<br />

Tractocile ® better<br />

Tocolytic efficacy and tolerability outcome<br />

of the CAP-001 studies: odds ratio<br />

p-value<br />

0.029<br />

0.079<br />

0.021<br />

0.0003<br />

Figure 6). Secondary outcomes, as described in the<br />

previous studies, were all comparable between both<br />

treatment groups.<br />

4.2.d Pooled analysis<br />

This was a multinational, multicentre collaboration<br />

carried out in Australia, Canada, Czech Republic,<br />

Denmark, France, Israel, Sweden, and the UK. 124 As the<br />

three studies followed the same protocol (Table 17),<br />

which included strict inclusion/exclusion criteria, it was<br />

possible to provide an overall assessment of the clinical<br />

efficacy by pooling the data. The pooled analysis of the<br />

three CAP-001 studies contained 742 women diagnosed<br />

with preterm labour between 23 and 33 weeks of<br />

gestation. The main outcome of the study, although not<br />

predefined in the protocol, was the assessment of<br />

tocolytic effectiveness in the intention to treat analysis in<br />

terms of the total number of women undelivered at 48<br />

hours and 7 days of starting treatment. Tocolytic efficacy<br />

and tolerability was assessed in terms of the<br />

proportion of women who remained undelivered and<br />

who did not require alternative tocolysis after 7 days<br />

of starting therapy.<br />

For ethical reasons, a composite endpoint was used<br />

as a measure of efficacy (referred to as ‘tocolytic<br />

efficacy and tolerability’) since many of the investigators<br />

were opposed to a protocol that did not allow<br />

administration of alternative tocolysis in the event of the<br />

progression of labour (‘treatment failure’). However,<br />

treatment failure also included women who discontinued<br />

treatment due to adverse events and, consequently, the<br />

efficacy endpoint used in this study was a composite<br />

of both efficacy and tolerability.<br />

Summary of clinical efficacy<br />

Baseline characteristics of the Tractocile ® and betaagonist<br />

groups were comparable prior to the initiation of<br />

treatment. The undelivered rate at 48 hours for<br />

Tractocile ® and beta-agonists was comparable (88.1%<br />

and 88.9%, p=<strong>0.9</strong>9). The proportion of women<br />

undelivered at 7 days, used as a measure of tocolytic<br />

effectiveness, was also comparable between the<br />

Tractocile ® and beta-agonist groups (79.7% vs.. 77.6%,<br />

p=0.28) (Table 18). The proportion of women remaining<br />

undelivered and not requiring alternative tocolysis after 7<br />

days of treatment was significantly higher in the<br />

Tractocile ® group (59.7%) compared with the betaagonist<br />

group (47.4%, p=0.0003) (Table 18, Figure 6), and<br />

TABLE 16. TOCOLYTIC EFFECTIVENESS AND TOCOLYTIC EFFICACY & TOLERABILITY OF <strong>TRACTOCILE</strong> ®<br />

AND TERBUTALINE AT 48 HOURS AND 7 DAYS<br />

Number of women (%)<br />

Tractocile ® Terbutaline Odds 95% CI p value<br />

(n=115) (n=129) ratio<br />

Tocolytic effectiveness<br />

undelivered at 48 h 99 (86.1) 110 (85.3) 1.11 0.52–2.38 0.78<br />

undelivered at 7 days 88 (76.5) 87 (67.4) 1.84 <strong>0.9</strong>6–3.52 0.07<br />

Tocolytic efficacy &<br />

tolerability*<br />

no failure at 48 h 83 (72.2) 88 (68.2) 1.22 0.67–2.22 0.52<br />

no failure at 7 days 64 (55.6) 56 (43.4) 1.62 <strong>0.9</strong>4–2.77 0.08<br />

*Treatment failure = delivery within the time interval or need for alternative tocolysis<br />

• Trial design<br />

TABLE 17. PRINCIPAL COMPONENTS OF THE COMMON STUDY PROTOCOL FOR THE<br />

CAP-001 CLINICAL STUDIES<br />

➞ Randomised, double-blind, double-dummy, beta-agonist-controlled trial<br />

• Population ➞ Women from 23 to 33 weeks gestation with regular uterine contractions (≥8<br />

contractions/h of ≥30 seconds duration), cervix 0–3 cm (nulliparous) or 1–3 cm<br />

(multiparous) dilated & ≥50% change in cervical length<br />

• Administration<br />

of study drugs<br />

• Primary outcomes/<br />

endpoints<br />

➞ Tractocile ® administered as a 6.75 mg i.v. bolus then 300 µg/min i.v. for 3 h<br />

and 100 µg/min i.v. up to 45 h. Beta-agonists administered i.v. according to local<br />

practice guidelines<br />

➞ Proportion of women (%) remaining undelivered >7 days after starting treatment<br />

➞ Proportion of women (%) remaining undelivered and not requiring alternative<br />

tocolysis >7 days after starting treatment<br />

➞ Safety – all adverse events and vital signs reported from start of treatment until<br />

discharge from hospital<br />

32 33


Chapter 4 Clinical efficacy of Tractocile ®<br />

there were also significantly fewer women in the<br />

Tractocile ® group requiring alternative tocolysis<br />

compared with the beta-agonist group (37.1% vs. 46.5%,<br />

p=0.01). Regarding secondary outcomes, Tractocile ® and<br />

beta-agonists were comparable in terms of their effects<br />

on uterine activity, reducing the number of contractions<br />

at a similar rate. Other comparable secondary outcomes<br />

included the number of women requiring retreatment,<br />

mean gestational age at delivery, and mean birth weight.<br />

Problems associated with the inclusion of women into<br />

clinical trials makes the evaluation of tocolytics difficult to<br />

demonstrate. Most women included in trials have already<br />

been transported to a hospital offering specialised care<br />

for preterm birth and may have already received tocolysis<br />

en route. Despite this the CAP-001 studies have<br />

demonstrated that regarding efficacy, Tractocile ® is<br />

comparable to the current standard tocolytics.<br />

TABLE 18. TOCOLYTIC EFFECTIVENESS AND TOCOLYTIC EFFICACY & TOLERABILITY OF <strong>TRACTOCILE</strong> ®<br />

AND BETA-AGONISTS AT 48 HOURS AND 7 DAYS [WORLDWIDE STUDY GROUP 2000].<br />

Number of women (%)<br />

Tractocile ® beta-agonists Odds 95% CI p value<br />

(n=360) (n=371) ratio<br />

Tocolytic effectiveness<br />

undelivered at 48 h 317 (88.1) 330 (88.9) 1.00 0.62–1.62 <strong>0.9</strong>9<br />

undelivered at 7 days 287 (79.7) 288 (77.6) 1.26 0.83–1.90 0.28<br />

5<br />

Clinical safety of Tractocile ®<br />

Tocolytic efficacy &<br />

tolerability*<br />

no failure at 48 h 268 (74.4) 260 (70.1) 1.36 <strong>0.9</strong>7–1.92 0.08<br />

no failure at 7 days 215 (59.7) 176 (47.4) 1.78 1.30–2.43 0.0003<br />

*Treatment failure = delivery within the time interval or need for alternative tocolysis<br />

34


Chapter 5 Clinical safety of Tractocile ®<br />

There have been several phase II and III clinical trials<br />

designed to investigate the efficacy and safety of<br />

Tractocile ® (atosiban). The phase II studies evaluated<br />

atosiban either uncontrolled or by comparison with<br />

placebo, while the phase III studies compared Tractocile ®<br />

with placebo or beta-agonists, the only other class of<br />

drugs licensed for tocolytic therapy.<br />

5.1 Phase II studies<br />

There are three phase II clinical studies that have<br />

investigated the safety profile of atosiban. 115,120,121 The aim<br />

of these studies was to determine the efficacy and safety<br />

of atosiban regarding the reduction of uterine<br />

contractions in women with threatened preterm labour.<br />

Study PAT-U01 was a double-blind, placebocontrolled,<br />

randomised trial. Atosiban (i.v.) was<br />

administered at an infusion rate of 300 µg/min during a<br />

two-hour treatment period. 120 At the end of the study<br />

there were no drop outs reported due to adverse events.<br />

From a total of 56 patients in both study groups, adverse<br />

events were reported in three women from the atosiban<br />

group (nausea, vomiting and diarrhoea) compared with<br />

four from the placebo group (chest pain, headache,<br />

nausea), and a fetal death was reported in the placebo<br />

group. Therefore, it was considered unlikely that any of<br />

the maternal adverse events were associated with<br />

atosiban administration. In addition, maternal heart rate<br />

and blood pressure were not significantly different<br />

between the atosiban and placebo groups.<br />

Post-study follow up revealed that both groups were<br />

similar with respect to the proportion of women<br />

experiencing post-partum complications. Furthermore,<br />

infant assessment revealed no fetal toxicity.<br />

TABLE 19. SUMMARY OF SAFETY RESULTS IN STUDY L91-049<br />

6.5 mg + Placebo + 2 mg + 0.6 mg + Ritodrine<br />

300 µg/min 300 µg/min 100 µg/min 30 µg/min (n=56)<br />

(n=63) (n=59) (n=62) (n=57)<br />

Study drug discontinued<br />

due to adverse events 0 0 0 1 (1.7%) 15 (25.9%)<br />

Commonly reported<br />

maternal adverse events:<br />

Chest pain 1 (1.6%) 1 (1.7%) 1 (1.6%) 2 (3.4%) 9 (15.5%)<br />

Tachycardia 0 2 (3.4%) 0 0 21 (36.2%)<br />

Nausea 6 (9.5%) 3 (5.1%) 2 (3.1%) 3 (5.2%) 12 (20.7%)<br />

Vomiting 0 0 2 (3.1%) 2 (3.4%) 13 (22.4%)<br />

Headache 4 (6.3%) 1 (1.7%) 2 (3.1%) 5 (8.6%) 8 (13.8%)<br />

Neonatal outcome:<br />

RDS* 8 (13.1%) 7 (12.1%) 3 (4.9%) 2 (3.6%) 5 (8.9%)<br />

Death 0 1 1 1 1<br />

*Respiratory distress syndrome<br />

Study PAT-U02 investigated the same objectives as<br />

PAT-U01 except that the dosing period was extended. 121<br />

In this non-controlled study, women received 300 µg/min<br />

of atosiban for a 12-hour period. Atosiban was well<br />

tolerated and no women discontinued treatment.<br />

Maternal adverse events were predominantly mild<br />

and included headache, nausea and vomiting. However,<br />

one case of gallstone pancreatitis was reported.<br />

Regarding the fetus, one serious case of variable heart<br />

rate deceleration was reported, which resolved<br />

spontaneously in hospital. Neither of the serious<br />

maternal or fetal adverse events were considered to be<br />

related to drug treatment. Follow-up data did not<br />

reveal infant abnormalities or complications that could<br />

have been attributed to atosiban.<br />

The final phase II study (L91-049) was a dose-ranging<br />

study designed to find the minimum effective dose of<br />

atosiban. 115 There were four randomised, double-blind<br />

atosiban groups and one open-label ritodrine group.<br />

Adverse events reported with an incidence ≥ 5% in the<br />

atosiban groups were headache and nausea. Almost<br />

26% of women receiving ritodrine discontinued<br />

treatment due to adverse events compared with only<br />

0.4% of women receiving atosiban (Table 19).<br />

Ritodrine was associated with an increase in maternal<br />

pulse rate and fetal heart rate, unlike atosiban where<br />

adverse cardiovascular effects were not reported. There<br />

were also differences between atosiban and ritodrine<br />

regarding metabolic effects, particularly hypokalaemia<br />

and hyperglycaemia, which were reported more<br />

frequently in the ritodrine group.<br />

Six maternal/fetal serious adverse events were<br />

reported in the atosiban groups (including one fetal<br />

death), while there were two cases of serious<br />

maternal/fetal adverse events in the ritodrine group.<br />

One infant death was reported in three of the atosiban<br />

groups and the ritodrine group (Table 19). None of the<br />

deaths or serious adverse events were judged to be<br />

related to treatment.<br />

5.2 Phase III studies<br />

5.2.a Placebo-controlled studies<br />

Two phase III, randomised, double-blind, placebocontrolled<br />

studies (PTL-096 and PTL-098) were carried<br />

out in the USA and South America. The significance of<br />

the safety results obtained from the PTL-096 study has<br />

been scrutinised due to the criteria used to recruit<br />

women into the studies. At randomisation, a<br />

significantly greater number of women below 26 weeks’<br />

gestation and with more severe preterm labour were<br />

randomised to the atosiban group. This contributed<br />

substantially to the greater number of infant deaths<br />

reported in the atosiban group compared with the<br />

placebo group. However, the mortality rate in the<br />

atosiban group was similar to what would be expected<br />

for infants of similar gestational age in the normal<br />

population and consequently infant deaths were<br />

attributed to complications of extreme prematurity.<br />

Despite these methodological limitations, this study was<br />

able to support the safety profile of atosiban, since<br />

maternal and fetal adverse events were similar in the<br />

atosiban and placebo groups.<br />

The second study, PTL-098, investigated the potential<br />

use of atosiban in maintenance therapy in order to<br />

inhibit a second episode of preterm labour. Similar to the<br />

safety results reported in PTL-096, maternal adverse<br />

events and infant outcomes were comparable to placebo<br />

in this study. 125 The only maternal adverse event noted in<br />

the atosiban group were injection site reactions related<br />

to prolonged subcutaneous maintenance therapy.<br />

Infant outcomes after follow-up<br />

Table 20 presents the data from the two placebocontrolled<br />

studies, PTL-096 and PTL-098, after follow-up<br />

at 6, 12 and 24 months after birth. A total of 583 infants<br />

(288 atosiban, 295 placebo) were followed from PTL-096:<br />

73% returned for the 6-month follow-up, 65% for the<br />

12-month follow-up and 55% for the 24-month followup.<br />

From PTL-098, 563 infants (291 atosiban, 272<br />

placebo) were followed: 81% returned for the 6-month<br />

follow-up, 74% for the 12-month follow-up and 55% for<br />

the 24-month follow-up. No unexpected developmental<br />

or neurological outcomes were found at 6-, 12- or 24-<br />

month follow up in the atosiban-treated group. 126,127<br />

5.2.b Comparative studies<br />

Three multinational, multicentre, double-blind,<br />

randomised, controlled trials (CAP-001) were<br />

designed to compare Tractocile ® (atosiban) with the<br />

beta-agonists, ritodrine, salbutamol and terbutaline.<br />

In addition, a pooled analysis incorporated all data<br />

from the three comparative trials. 141 This provided an<br />

overall assessment of the safety and tolerability of the<br />

three beta-agonists in comparison with Tractocile ® . In<br />

each study, the tocolytic agents were administered i.v.<br />

at their clinically recommended doses. In order to<br />

avoid repeating results from each of the three CAP-<br />

001 studies, only the pooled data will be discussed<br />

in detail below.<br />

5.2.c Pooled analysis<br />

A pooled analysis of the three comparative trials was<br />

performed to provide an overall statistical assessment<br />

of the safety data obtained. Overall, there was a total<br />

of 742 women included in the pooled analysis.<br />

TABLE 20. INFANT OUTCOMES OF ATOSIBAN COMPARED WITH PLACEBO<br />

Bayley II assessment of mental and motor development and neurological examination<br />

PTL-096<br />

PTL-098<br />

6 months Atosiban Placebo Atosiban Placebo<br />

Mental Development Index 95 95 100 100<br />

Physical Development Index 93 92 97 96<br />

Neurologically normal 89% 84% 88% 90%<br />

12 months Atosiban Placebo Atosiban Placebo<br />

Mental Development Index 95 97 97 98<br />

Physical Development Index 94 95 97 95<br />

Neurologically normal 90% 90% 93% 94%<br />

24 months Atosiban Placebo Atosiban Placebo<br />

Mental Development Index 84 89 91 91<br />

Physical Development Index 93 94 97 95<br />

Neurologically normal 88% 86% 89% 94%<br />

36 37


Tractocile ® (n=361)<br />

Tractocile ® (n=361)<br />

Chapter 5 Clinical safety of Tractocile ® Figure 9b. Reported neonatal adverse events in the Tractocile ®<br />

Maternal adverse events<br />

The most frequently reported maternal adverse event<br />

following beta-agonist treatment was tachycardia,<br />

which occurred at a similar rate across all three study arms<br />

(approx. 75%). Maternal tachycardia was defined as a<br />

maternal heart rate >120 bpm. Clinically important<br />

adverse events included a case of myocardial ischaemia<br />

and two cases of pulmonary oedema in the beta-agonist<br />

group. A third case of pulmonary oedema was reported<br />

in the Tractocile ® group after the woman had<br />

subsequently been given a beta-agonist as rescue<br />

therapy for 7 days following atosiban administration.<br />

The incidence of at least one maternal cardiovascular<br />

side effect was 8.3% in the atosiban group and 81.2% in<br />

the beta-agonist group. Maternal cardiovascular side<br />

effects included pulmonary oedema, chest pain,<br />

myocardial ischaemia, dyspnœa, palpitation, tachycardia,<br />

hypotension and syncope (Figure 7a). Constitutional<br />

adverse events such as vomiting, nausea and headache<br />

were also more frequently reported after beta-agonist<br />

administration (Figure 7b).<br />

Regarding those women who discontinued treatment<br />

due to adverse events, there were four (1.1%) from the<br />

Tractocile ® group and 56 (15.4%) from the beta-agonist<br />

group (p=0.0001) (Figure 8). The mean ± SD heart rate<br />

(bpm) in women when the study was discontinued was<br />

86.2±13.9 in the atosiban group and 124.7±19.6 in the<br />

beta-agonist group (p=0.0001).<br />

% Incidence<br />

75<br />

70<br />

65<br />

60<br />

55<br />

50<br />

45<br />

40<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

*<br />

Pulmonary oedema<br />

Myocardial ischaemia<br />

Tractocile ® (n=361)<br />

Beta-agonists (n=372)<br />

Chest pain<br />

Dyspnœa<br />

Palpitation<br />

Tachycardia<br />

Hyperglycæmia<br />

Hypokalæmia<br />

* this single patient case occurred<br />

after switch to beta-agonist therapy<br />

% Incidence<br />

% Incidence<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Figure 8.<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Frequency of treatment discontinuation due to<br />

maternal side effects according to the allocated<br />

study medication<br />

Nausea<br />

Beta-agonists (n=372)<br />

Maternal<br />

cardiovascular<br />

side effects<br />

Tractocile ® (n=406)<br />

Beta-agonists (n=432)<br />

Vomiting<br />

Headache<br />

Discontinuations<br />

due to side effects<br />

Tremor<br />

Hypertension<br />

Hypotension<br />

% Incidence<br />

Fetal adverse events<br />

The most frequently reported fetal adverse event was<br />

fetal tachycardia in the beta-agonist group (28%)<br />

compared with only 3% in the Tractocile ® group (Figure<br />

9a). Fetal tachycardia was defined as a fetal heart rate<br />

>170 bpm. Other events, such as fetal distress and<br />

bradycardia, were reported to a similar extent in both<br />

the Tractocile ® and beta-agonist groups.<br />

Neonatal adverse events<br />

Both treatment groups were considered to be<br />

comparable regarding neonatal morbidity (Figure 9b).<br />

Admissions to specialised intensive care units and the<br />

period of time spent in the unit were similar in both<br />

treatment groups (31% Tractocile ® vs. 30% betaagonists).<br />

Major congenital anomalies were reported<br />

in seven (1.7%) infants in the atosiban group and four<br />

infants (<strong>0.9</strong>%) in the beta-agonist groups.<br />

There were 18 fetal/infant deaths over the course of<br />

the studies. Six were reported in the Tractocile ® group<br />

(4 singleton and 2 twins) compared with 12 from the<br />

beta-agonist groups (5 singletons and 7 twins).<br />

Consequently, the perinatal mortality rate was 14.7 per<br />

1000 in the atosiban group and 27.7 per 1000 in the<br />

beta-agonist group. The indicated causes of death<br />

were complications associated with prematurity, such<br />

as infection, respiratory distress syndrome, necrotising<br />

enterocolitis and intraventricular haemorrhage. Of<br />

these deaths, three were intrauterine deaths, two in<br />

the beta-agonist group and one in the atosiban group.<br />

None of the deaths were regarded by the investigators<br />

to be related to the tocolytic agent.<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Tachycardia<br />

Tractocile ® (n=361)<br />

Beta-agonists (n=372)<br />

Bradycardia<br />

Fetal distress<br />

Fetal death<br />

Asphyxia<br />

Hypoxia<br />

% Incidence<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

RDS<br />

Cerebral haemorrhage<br />

Beta-agonists (n=372)<br />

Apnoea<br />

and beta-agonist groups<br />

Bradycardia<br />

Arrhythmia<br />

Hypotension<br />

Summary<br />

The principal difference between Tractocile ® and<br />

beta-agonists in terms of safety outcomes was the<br />

incidence of maternal cardiovascular side effects<br />

(8.3% vs. 81.2%, p


Chapter 5 Clinical safety of Tractocile ®<br />

TABLE 21. INFANT OUTCOMES FOLLOWING TOCOLYTIC TREATMENT WITH BETA-AGONISTS<br />

Study Design Outcome<br />

Freysz et al 1977<br />

Polowczy et al 1984<br />

Hadders-Algra et al 1986<br />

Laros et al 1991<br />

Forty two infants from preterm women<br />

treated with ritodrine (60-80 mg/day)<br />

during a period from 3–93 days were<br />

matched with control infants.<br />

Twenty infants exposed to ritodrine<br />

during management of preterm labour<br />

were examined at 7–9 years of life and<br />

compared with matched controls.<br />

A group of 78 six-year-old infants<br />

exposed to ritodrine during pregnancy<br />

for an average of 28 days were matched<br />

with control groups.<br />

201 infants exposed to either isoxsuprine,<br />

ritodrine, terbutaline or a combination,<br />

compared with 130 controls. Follow up<br />

was analysed at 1, 3 and 4 years.<br />

There were no statistically significant differences<br />

between the two groups for any of the variables<br />

of development studied.<br />

No significant differences were detected<br />

regarding growth, neurological findings<br />

and psychometric testing.<br />

No significant differences were found regarding<br />

urinalysis, body length, weight, head<br />

circumference, neurological findings. However,<br />

school performances of ritodrine-treated infants<br />

were considered to be poorer.<br />

No significant differences were found in growth<br />

or development. Observations of time-related<br />

events did however suggest some evidence of<br />

greater mortality and trauma in<br />

the terbutaline group.<br />

6<br />

Pharmaceutical application<br />

of Tractocile ®<br />

40


Chapter 6 Pharmaceutical application of Tractocile ®<br />

6.1 Therapeutic indication<br />

Tractocile ® (atosiban) is indicated to delay imminent<br />

preterm birth in pregnant women with:<br />

• a gestational age between 24 and 33 completed weeks<br />

• regular uterine contractions of at least 30 seconds,<br />

duration at a rate of ≥4 per 30 minutes<br />

• age ≥18 years<br />

• a cervical dilation of 1–3 cm (0–3 cm for nulliparous<br />

women) and effacement of ≥50%<br />

• normal fetal heart rate.<br />

6.2 Pharmaceutical form<br />

Tractocile ® is available at a concentration of 7.5 mg/<strong>ml</strong> of<br />

atosiban in:<br />

• <strong>0.9</strong> <strong>ml</strong> vial for i.v. bolus injection (‘Solution for <strong>Injection</strong>’)<br />

• 5 <strong>ml</strong> vial for i.v. infusion (‘Concentrate for Solution<br />

for Infusion’)<br />

6.3 Dosage and administration<br />

The duration of treatment should not exceed 48 hours<br />

and the total dose administered should preferably not<br />

exceed 330 mg atosiban during a full course of Tractocile ®<br />

therapy. In the Phase III clinical trials, the majority of<br />

women were administered Tractocile ® for a total of 18<br />

hours (one vial of <strong>0.9</strong> <strong>ml</strong> plus four vials of 5 <strong>ml</strong>).<br />

Treatment with Tractocile ® should be initiated and<br />

maintained by a physician experienced in the treatment<br />

of preterm labour. Tractocile ® is administered i.v. in three<br />

stages (figure 10 and table 22):<br />

1. An initial bolus injection of 6.75 mg corresponding to<br />

7.5 mg/<strong>ml</strong> is recommended as soon as possible after<br />

preterm labour has been diagnosed<br />

2. Immediately followed by a continuous high-dose<br />

infusion of 300 µg/min for 3 hours, known as the<br />

‘loading infusion’<br />

3. Followed by a lower dose infusion of 100 µg/min for<br />

a maximum of 45 hours, known as the ‘subsequent<br />

infusion’.<br />

If episodes of preterm labour recur after uterine<br />

quiescence has been achieved, the three-stage regimen<br />

above can be repeated. However, it is worth noting that<br />

a maximum of three re-treatments were administered to<br />

women during clinical trials. Alternative therapy should<br />

be considered in case of persistent uterine contractions.<br />

Step II - 'Loading infusion'<br />

Step I - 'Initial bolus i.v. injection'<br />

<strong>0.9</strong> <strong>ml</strong> of Tractocile ® Solution<br />

for injection (7.5 mg/<strong>ml</strong>)<br />

Continuous infusion Tractocile ® Concentrate<br />

for Solution for infusion. Infusion rate of<br />

24 <strong>ml</strong>/hour=300 µg/min for 3 hours<br />

Step III - 'Subsequent infusion'<br />

Follow by a lower dose<br />

of Tractocile ® Concentrate for<br />

Solution for infusion<br />

Reduce infusion rate<br />

0f 8 <strong>ml</strong>/hour=100 µg/min<br />

for up to 45 hours<br />

TABLE 22. STANDARD DOSING REGIMEN FOR <strong>TRACTOCILE</strong> ®<br />

Withdraw 10 <strong>ml</strong> solution from<br />

a 100 <strong>ml</strong> infusion bag and<br />

discard. Replace it with<br />

(2 x 5 <strong>ml</strong> vials) Tractocile ®<br />

iv to pregnant woman<br />

Prepare a new 100 <strong>ml</strong> bag by<br />

withdrawing 10 <strong>ml</strong> solution<br />

from a 100 <strong>ml</strong> infusion bag<br />

and discard. Replace it with<br />

(2 x 5 <strong>ml</strong> vials) Tractocile ®<br />

i.v. to pregnant woman<br />

Figure 10. Visual representation of the administration<br />

of Tractocile ®<br />

6.3.a Preparation of the initial i.v. Solution for <strong>Injection</strong><br />

The initial bolus should be administered as a <strong>0.9</strong> <strong>ml</strong><br />

injection, equating to a dose of 6.75 mg Tractocile ® .<br />

Withdraw <strong>0.9</strong> <strong>ml</strong> of a <strong>0.9</strong> <strong>ml</strong> labelled vial of Tractocile ®<br />

7.5 mg/<strong>ml</strong> Solution for <strong>Injection</strong> and administer slowly<br />

as an i.v bolus over one minute. The Tractocile ® 7.5<br />

mg/<strong>ml</strong> Solution for <strong>Injection</strong> must be used immediately<br />

after opening.<br />

6.3.b Preparation of the diluted Concentrate for Solution<br />

for Infusion<br />

For the i.v. infusion, following the bolus dose, Tractocile ®<br />

7.5 mg/<strong>ml</strong> Concentrate for Solution for Infusion should<br />

be diluted in one of the following solutions:<br />

Stage Regimen Dose Rate Duration<br />

1 <strong>0.9</strong><strong>ml</strong> i.v. injection 6.75 mg <strong>Bolus</strong> 1 min<br />

2 i.v. infusion 18 mg/h 24 <strong>ml</strong>/h 3 h<br />

3 i.v. infusion 6 mg/h 8 <strong>ml</strong>/h Up to 45 h<br />

• <strong>0.9</strong>% (w/v) isotonic normal saline solution<br />

• Ringer’s lactate solution<br />

• 5% (w/v) isotonic glucose solution.<br />

Dilution must be performed immediately after opening.<br />

To prepare the infusion solution in a 100 <strong>ml</strong> infusion<br />

bag, withdraw 10 <strong>ml</strong> from the infusion bag and discard.<br />

This should be replaced by 10 <strong>ml</strong> Tractocile ® 7.5 mg/<strong>ml</strong><br />

Concentrate for Solution for Infusion from two 5 <strong>ml</strong> vials,<br />

resulting in a concentration of 75 mg Tractocile ® in<br />

100 <strong>ml</strong> (figure 10). The loading infusion is delivered at a<br />

rate of 24 <strong>ml</strong>/hour, equivalent to a dose of 18 mg/hour<br />

(300 µg/min), for 3 hours. After 3 hours, this is reduced to<br />

a rate of 8 <strong>ml</strong>/hour, equivalent to a dose of 6 mg/hour<br />

(100 µg/min), for the remainder of the infusion.<br />

Prepare new 100 <strong>ml</strong> infusion bags in the same way as<br />

described above to allow the infusion to be continued<br />

uninterrupted. If an infusion bag with a different<br />

volume is used, a proportional calculation should be<br />

made for the preparation. The diluted Tractocile ®<br />

7.5 mg/<strong>ml</strong> Concentrate for Solution for Infusion must be<br />

used within 24 hours of preparation.<br />

To ensure accurate dosing, a controlled infusion<br />

device is recommended to adjust the rate of flow in<br />

drops/minute when required. An i.v. microdrip chamber<br />

can provide a convenient range of infusion rates within<br />

the recommended dose levels for Tractocile ® .<br />

6.4 Contraindications<br />

Tractocile ® should not be used in the following<br />

conditions:<br />

• gestational age below 24 or over 33 completed weeks<br />

• antepartum uterine haemorrhage requiring<br />

immediate delivery<br />

• eclampsia and severe preeclampsia requiring delivery<br />

• intrauterine fetal death<br />

• suspected intrauterine infection<br />

• placenta praevia<br />

• abruptio placenta<br />

• any other conditions of the mother or fetus, in which<br />

continuation of pregnancy is hazardous<br />

• known hypersensitivity to the active substance or any<br />

of the excipients<br />

• premature rupture of the membranes >30 weeks’<br />

gestation<br />

• intrauterine growth retardation and abnormal fetal<br />

heart rate.<br />

6.5 Precautions and warnings<br />

During administration of Tractocile ® it is advisable to<br />

monitor maternal uterine contractions and fetal heart<br />

rate at regular intervals. It is also necessary to evaluate<br />

the benefit of delaying delivery against the potential<br />

risk of chorioamnionitis in women with suspected<br />

PROM. In women with intrauterine growth retardation,<br />

the decision whether to continue or restart treatment<br />

with Tractocile ® will be dependent upon the assessment<br />

of fetal maturity. There is no experience with<br />

Tractocile ® treatment in women with impaired kidney or<br />

liver function.<br />

As an antagonist of oxytocin, Tractocile ® may<br />

theoretically facilitate uterine relaxation and<br />

postpartum bleeding. Therefore, blood loss after<br />

delivery should be monitored. However, inadequate<br />

uterine contraction postpartum was not observed<br />

during the Phase III clinical trials.<br />

6.6 Interaction with other medicinal products<br />

and other forms of interaction<br />

Drug interaction studies have been performed<br />

investigating betamethasone and Labelatol. The<br />

studies conclude that co-administration of atosibanbetamethasone<br />

and atosiban–labelatol had no clinically<br />

relevant influence on drug bioavailability. A manuscript<br />

has been submitted and is pending publication. Data<br />

on file and available on request.<br />

6.7 Pregnancy and lactation<br />

No toxic effects of Tractocile ® have been reported in<br />

embryotoxicity studies. Small amounts of a metabolite<br />

of atosiban have been shown to pass from plasma<br />

into the breast milk of lactating women.<br />

6.8 Undesirable effects<br />

The undesirable maternal adverse reactions reported in<br />

clinical studies were generally of a mild severity and<br />

included nausea (14%), headache, vomiting, flushing,<br />

dizziness, tachycardia, hypotension, injection site reaction<br />

and hyperglycaemia (1–10%).<br />

For the newborn, the Phase III clinical trials did not<br />

reveal any specific undesirable effects of Tractocile ® .<br />

Infant outcomes were in the range of normal variation<br />

and were comparable to both placebo and beta-agonists.<br />

6.9 Overdose<br />

Few cases of Tractocile ® overdosing were reported<br />

during Phase III clinical trials and these cases occurred<br />

without any specific signs or symptoms. There is no<br />

known specific treatment in case of an overdose.<br />

6.10Storage and packaging information<br />

Tractocile ® will be available in single packs of <strong>0.9</strong> <strong>ml</strong> and<br />

5 <strong>ml</strong> vials. The shelf life of Tractocile ® is 2 years when<br />

stored in the original container at a temperature of<br />

2–8°C. Once opened, Tractocile ® must be used<br />

immediately (<strong>0.9</strong> <strong>ml</strong>) or diluted immediately (5 <strong>ml</strong>).<br />

Dilutions should be used within 24 hours of preparation.<br />

42 43


7<br />

Summary of the key benefits of<br />

Tractocile ® in the treatment<br />

of preterm labour


Chapter 7 Summary of the key benefits of Tractocile ® in the treatment of preterm labour<br />

7.1 Proven efficacy and tolerability<br />

The efficacy of Tractocile ® is demonstrated most clearly<br />

in the comparative studies with beta-agonists. As a<br />

tocolytic, this group of drugs are currently the most<br />

popular in clinical practice, therefore, the results from<br />

the CAP-001 studies are very encouraging for<br />

Tractocile ® . As demonstrated in the pooled analysis,<br />

Tractocile ® was statistically superior to beta-agonist<br />

treatment in terms of its efficacy and tolerability.<br />

There was a higher proportion of women remaining<br />

undelivered and not requiring alternative tocolytic<br />

treatment within 7 days in the Tractocile ® group<br />

compared with the beta-agonists. In addition,<br />

Tractocile ® demonstrated statistical superiority in terms<br />

of an increased delay in delivery from the start of<br />

treatment or the requirement of alternative tocolytic<br />

therapy. Regarding tocolytic effectiveness, defined as<br />

the proportion of women undelivered at 7 days,<br />

Tractocile ® was comparable to the beta-agonists. There<br />

was also no difference between the two treatments<br />

regarding secondary outcomes such as mean<br />

gestational age at delivery and mean birth weight.<br />

7.2 Superior safety profile<br />

Tractocile ® has proven to be superior to current<br />

standard tocolytic therapy and shown to be well<br />

tolerated by women with preterm labour and by the<br />

fetus. Tractocile ® is associated with a placebo-level<br />

incidence of cardiovascular adverse events. The few<br />

adverse events that have been reported were mild<br />

or moderate in severity and were more likely to result<br />

from iatrogenic causes such as the delivery process and<br />

the consequences of prematurity rather than<br />

the drug itself. Tractocile ® is the only tocolytic with<br />

proven short-term and long-term safety for mother<br />

and baby.<br />

7.3 Absence of tachyphylaxis<br />

There have been no reports of tachyphylaxis with<br />

maintenance Tractocile ® therapy, however, this<br />

phenonomen is commonly associated with the use<br />

of beta-agonists.<br />

7.4 Specific mode of action<br />

Tractocile ® contains the oxytocin analogue atosiban,<br />

which is designed to compete with oxytocin at receptor<br />

sites in the myometrium and decidua of the uterus.<br />

Atosiban itself does not elicit a response once it<br />

occupies a receptor, therefore it acts as an antagonist<br />

blocking the activation of oxytocin receptors in the<br />

uterus. This blockade causes an inhibition of uterine<br />

contractility. Oxytocin receptors are predominantly<br />

found in the uterus, therefore, atosiban is organ<br />

specific, which explains the paucity of side effects.<br />

7.5 Rapid onset of action<br />

Tractocile ® has a rapid onset of action, inhibiting<br />

preterm uterine contractions at a comparable rate to<br />

beta-agonists.<br />

7.6 Period of use<br />

Studies have indicated an acceptable therapeutic<br />

window of between 24 and 33 gestational weeks.<br />

This is comparable to other tocolytic agents such as<br />

beta-agonists.<br />

Conclusion<br />

Although there have been improvements<br />

in neonatal survival after preterm birth this is<br />

associated with significant costs. The<br />

prevention of preterm birth is the ideal<br />

outcome in particular before 30 weeks’<br />

gestation, below which time the mortality and<br />

morbidity are disproportionately high. Due to<br />

its heterogenous nature, preventative<br />

strategies for preterm birth are unlikely to be<br />

effective on a global scale since there will be<br />

more than one definitive treatment. Delaying<br />

preterm delivery will however help gain time<br />

to administer steroids, transfer the mother in<br />

utero to a tertiary centre and provide other<br />

measures to improve pregnancy outcome.<br />

Tocolytic success should be measured in<br />

terms of each day gained rather than a direct<br />

measure of gestational age at delivery. It is<br />

clear that some clinical benefit in the shortterm<br />

prolongation of pregnancy can be<br />

achieved using tocolytic therapy, however,<br />

the safety of these pharmacological agents<br />

remains a concern. It is the issue of safety<br />

that restricts tocolysis to gestational ages<br />

below 34 weeks.<br />

The introduction of oxytocin antagonists as<br />

a safe alternative to beta-agonists potentially<br />

allows for tocolysis at later gestational ages.<br />

Improved diagnostic techniques will<br />

inevitably result in the earlier identification of<br />

preterm labour and the more accurate<br />

selection of women in whom tocolysis will be<br />

beneficial. Together with improved<br />

approaches in treatment, the ultimate goal of<br />

prolonging pregnancy to an extent that<br />

provides substantial benefit for the infant<br />

and mother may soon be achievable.<br />

46 47


References<br />

1. Romero R, Sibai BM, Sanchez-Ramos L, Valenzuela GJ, Veille J-C,<br />

Tabor B, Perry KG, Varner M, Goodwin TM, Lane R, Smith J,<br />

Shangold G, Creasy GW. An oxytocin receptor antagonist<br />

(atosiban) in the treatment of preterm labor: a randomized,<br />

double-blind, placebo- controlled trial with tocolytic rescue. Am<br />

J Obst Gyn 2000; 182:1173-1183.<br />

2. Copper RL, Goldenberg RL, Creasy RK et al. A multicentre<br />

study of preterm birth weight and gestational age-specific<br />

neonatal mortality. Am J Obstet Gynecol 1993; 168: 78–84.<br />

3. Lopez-Bernal A, Watson SP, Phaneuf S, Europe-Finner GN.<br />

Biochemistry and physiology of preterm labour and delivery.<br />

Bailliers Clin Obstet Gynaecol 1993; 7 523–552.<br />

4. Morrison JC. Preterm birth: a puzzle worth solving. Obstet<br />

Gynecol 1990; 76: 5S–12S.<br />

5. Goldenberg RL, Nelson KG, Davis RO, Koski J. Delay in delivery:<br />

influence of gestational age and the duration of delay on<br />

perinatal outcome. Obstet Gynecol 1984; 64: 480-484.<br />

6. WHO. International statistical classification of diseases<br />

and related health problems. 10th revision. Vol. 2. Geneva:<br />

WHO: 1993.<br />

7. Keirse MJNC. New perspectives for the effective treatment of<br />

preterm labour. Am J Obstet Gynecol 1995; 173: 618–28.<br />

8. Rush RW, Keirse MJNC, Howart P, Baum JD, Anderson ABM,<br />

Turnbull AC. Contribution of preterm delivery to perinatal<br />

mortality. BMJ 1976; 2: 965–968.<br />

9. Villar J, Ezcurra EJ. Preterm delivery syndrome: the unmet<br />

need. New perspectives for the effective treatment of<br />

preterm labour – an international consensus. Res Clin Forums<br />

1994; 16: 9–38.<br />

10. Tucker JM, Goldenberg RL, Davies RO et al. Etiologies of<br />

preterm birth in an indigent population: is prevention a<br />

logical expectation Obstet Gynecol 1991; 77: 343–387.<br />

11. Elder MG. New perspectives for the effective treatment of<br />

preterm labour: an international consensus. Res Clin Forums<br />

1994; 16(3): 189–199.<br />

12. Romero R, Mazor M, Munoz H, Gomez R, Galasso M, Sherer<br />

DM. The preterm labor syndrome. Ann N Y Acad Sci 1994;<br />

734: 414–29.<br />

13. Hall MH, Danielian P, Lamont RF. The importance of preterm<br />

birth. In: Elder MG, Lamont RF, Romero R, editors. Preterm<br />

Labor. New York: Churchill Livingstone: 1997 p. 1–28.<br />

14. Carr-Hill RA, Hall MH. The repetition of spontaneous preterm<br />

labour. Br J Obstet Gynaecol 1985; 92: 921–928.<br />

15. Bakketeig LS, Hoffman HJ. Epidemiology of preterm birth. In:<br />

Preterm labour. Elder MG, Hendricks CH, editors. Butterworth’s<br />

International Medical Reviews: London; 1981, p17.<br />

16. Wang X, Zuckerman B, Coffman GA, Corwin MJ. Familial<br />

aggregation of low birthweight among whites and blacks in the<br />

United States. N Engl J Med 1995; 333: 1744.<br />

17. Hoffman HJ, Bakketeig LS. Risk factors associated with the<br />

occurrence of preterm birth. Clin Obstet Gynecol 1984; 27:<br />

539–552.<br />

18. Cnattingius S, Forman MR, Berendes H, Graubard BI, Isotalo L.<br />

Effect of age, parity and smoking on pregnancy outcome: a<br />

population-based study. Am J Obstet Gynecol 1993; 168: 16–21.<br />

19. Kristensen J, Langhoff-Roos J, Kristensen FB. Idiopathic preterm<br />

deliveries in Denmark. Obstet Gynecol 1995; 85: 549–552.<br />

20. Hickey CA, Cliver SP, McNeal SF, Hoffman HJ, Goldenberg RL.<br />

Prenatal weight gain patterns and spontaneous preterm birth<br />

among non-obese black and white women. Obstet Gynecol<br />

1995; 85: 909.<br />

21. Lu<strong>ml</strong>ey J. The epidemiology of preterm birth. Baillieres Clin<br />

Obstet Gynaecol 1993; 7: 477–98.<br />

22. DiFronza JR, Lew RA. Effect of maternal cigarette smoking on<br />

pregnancy complications and sudden infant death syndrome.<br />

J Fam Pract 1995; 40: 385.<br />

23. Chasnoff I, Griffeth D, MacGregor S et al. Temporal patterns<br />

of cocaine use in pregnancy. JAMA 1989; 261: 1714–1724.<br />

24. Villar J, Farnot U, Barros F, Victoria C, Langer A, Belzian JM.<br />

A randomized trial of psychosocial support during high-risk<br />

pregnancies. N Engl J Med 1992; 327: 1266–1271.<br />

25. Tafari N, Naeye RL, Gobezie A. Effects of maternal<br />

undernutrition and heavy physical work during pregnancy on<br />

birth weight. Br J Obstet Gynaecol 1980; 87: 222–226.<br />

26. Lettieri L, Anthony M, Vintzileos M et al. Does “idiopathic”<br />

preterm labor resulting in preterm birth exist Am J Obstet<br />

Gynecol 1993; 160: 1480–1485.<br />

27. Gomez R, Romero R, Mazor M, Ghezzi F, David C and Yoon<br />

BH. The role of infection in preterm labor and delivery. In:<br />

Elder MG, Lamont RF, Romero R, editors. Preterm Labor. New<br />

York: Churchill Livingstone: 1997 p. 85–125.<br />

28. Blanc W. Pathways of fetal and early neonatal infection: viral<br />

placentitis, bacterial and fungal chorioamnionitis. J Paediatr<br />

1964; 59: 473.<br />

29. Romero R, Mazor M. Infection and preterm labour. Clin Obstet<br />

Gynecol 1988; 31: 553.<br />

30. Smaill F. Antibiotic versus no treatment for asymptomatic<br />

bacteriuria. In: Enkikn MW, Keirse MJNC, Renfrew MJ,<br />

Neilson JP, eds. Pregnancy and childbirth module. Cochrane<br />

database of systematic reviews, review no. 03170, 22 April<br />

1993. Cochrane updates on disk. Oxford: Update Software,<br />

1994; disk issue 1.<br />

31. Arias F, Tomich P. Etiology and outcome of low birth weight<br />

and preterm infants. Obstet Gynecol 1982; 60: 277–281.<br />

32. Savitz DA, Blackmore CA, Thorp JM. Epidemiologic<br />

characteristics of preterm delivery: etiologic heterogeneity.<br />

Am J Obstet Gynecol 1991; 164: 343–387.<br />

33. Williams MA, Mittendorf R, Lieberman E, Monson RR.<br />

Adverse infant outcomes associated with first-trimester<br />

vaginal bleeding. Obstet Gynecol 1991; 78: 14–18.<br />

34. MacLennan AH. Multiple gestation: clinical characteristics and<br />

management. In: Creasy RK, Resnik R (eds). Maternal fetal<br />

medicine, 3rd ed. WB Saunders, Philadelphia, 1994, p. 591.<br />

35. Lipitz S, Frenkel Y, Watts C et al. High order multifetal<br />

gestation management and outcome. Obstet Gynecol<br />

1990; 76: 215–218.<br />

36. Wenstrom KD, Gall SA. Incidence, morbidity and mortality,<br />

and diagnosis of twin gestations. Clin Perinatol 1988; 15: 1.<br />

37. McLean M, Walters WAW, Smith R. Prediction and early<br />

diagnosis of preterm labor: a critical review. Obstet Gynecol Surv<br />

1993; 48: 209–25.<br />

38. Lockwood CJ, Wein R, Lapinski R. The presence of cervical<br />

and vaginal fetal fibronectin predict preterm delivery in an<br />

inner-city obstetric population. Am J Obstet Gynecol 1993;<br />

169: 798–804.<br />

39. McGregor JA, Jackson M, Lachelin GC et al. Salivary oestriol<br />

as risk assessment for preterm labor: a prospective trial. Am J<br />

Obstet Gynecol 1995; 173: 1337–1342.<br />

40. Newman RB, Godsey RK, Ellings JM et al. Quantification of<br />

cervical change: relationship to preterm delivery in multifetal<br />

gestation.Am J Obstet Gynecol 1991; 165: 264–271.<br />

41. Tongsong T, Kamprapanth P, Srisomboon J et al. Single<br />

transvaginal sonographic measurement of cervical length<br />

early in the third trimester as a predictor of preterm delivery.<br />

Obstet Gynecol 1995; 86: 184–187.<br />

42. Alexander S, Slater C. Labouring under linguistic delusions:<br />

the impact of linguistic factors in international studies of<br />

preterm labour. Language Comm 1987; 7: 179–185.<br />

43. Amon E. Premature labor. In: Reece EA, Hobbins JC, Mahoney<br />

MJ, Petrie RH, (eds). Medicine of the fetus and mother.<br />

JB Lippincott: Philadelphia 1992; 1398–1429.<br />

44. Lee W, Barton S, Comstock CH et al. Transverse cerebellar<br />

diameter (TCD): a useful predictor of gestational age for<br />

fetuses with asymmetric growth retardation. Am J Obstet<br />

Gynecol 1991; 165: 1044–1050.<br />

45. Copper RL, Goldenberg RL, Davis RO et al. Warning<br />

symptoms, uterine contractions, and cervical examination<br />

findings in women at risk of preterm delivery. Am J Obstet<br />

Gynecol 1990; 162: 748–754.<br />

46. Creasy RK. Early detection of preterm labour. In: Queenan,<br />

JT, Hobbins JC (eds) Protocols for high risk pregnancies.<br />

3rd Edition. Blackwell Science, London, 1996, p. 535–538.<br />

47. Maresh M. The obstetric management of preterm labour.<br />

Curr Obstet Gynaecol 1992; 2: 199–206.<br />

48. McNamara HM, Vintzileos AM. The clinical approach to<br />

preterm labour. In: Elder MG, Lamont RF, Romero R, (eds).<br />

Preterm labor. New York, Churchill Livingstone; 1997 p.<br />

207–242.<br />

49. National Health and Medical Research Council (Australia).<br />

Clinical practice guidelines for care around pretrem birth.<br />

Australian Government Publishing Service, GPO Box 9848,<br />

Canberra, ACT 2601, Australia.<br />

50. King JF, Grant A, Keirse MJNC, Chalmers I. Beta-mimetics in<br />

preterm labour: an overview of the randomised controlled<br />

trials. Br J Obstet Gynaecol 1988; 95: 211–222.<br />

51. Canadian Preterm Labor Investigators Group. Treatment of<br />

preterm labor with the beta-adrenergic agonist ritodrine.<br />

N Engl J Med 1992; 327: 308–312.<br />

52. Hudgens DR, Conradi SE. Sudden death association with<br />

terbutaline sulfate infusion. Am J Obstet Gynecol 1993; 169:<br />

210–221.<br />

53. Hankins GD. Complications of beta-sympathomimetic<br />

tocolytic agents. In: Clark SL, Cotton DB, Hankins GDV,<br />

Phelan JP (eds) Critical Care Obstetrics. 2nd Edition. Boston;<br />

Blackwell Scientific Publications, 1991.<br />

54. Caritis SN, Carson D, Greebon D et al. A comparison of<br />

terbutaline and ethanol in the treatment of preterm labour.<br />

Am J Obstet Gynecol 1982; 142: 183–190.<br />

55. Beall MH, Edgar BW, Paul RH, Smith-Wallace T. A comparison<br />

of ritodrine, terbutaline, and magnesium sulfate for the<br />

suppression of preterm labor. Am J Obstet Gynecol 1985;<br />

153: 854–862.<br />

56. Schwarz R, Retzke U. Cardiovascular effects of terbutaline<br />

in pregnant women. Acta Obstet Gynecol Scand 1983;<br />

62: 419–424.<br />

57. Ingemarsson I, Arulkumaran S, Kottegoda SR. Complications<br />

of beta-mimetic therapy in preterm labour. Aust NZ J Obstet<br />

Gynaecol 1985; 25: 182–189.<br />

58. Leveno KJ, Cunningham FG. Beta-adrenergic agonists for<br />

preterm labour. N Engl J Med 1992; 327: 349–351.<br />

59. Benedetti TJ, Caritis SN. Treatment of preterm labor with<br />

beta-adrenergic agonist ritodrine. N Engl J Med 1992; 327:<br />

1758–1760.<br />

60. Besinger RE, Iannucci TA. Tocolytic therapy. In: Elder MG,<br />

Lamont RF, Romero R, (eds). Preterm labor. New York, Churchill<br />

Livingstone; 1997 p. 243–97.<br />

61. Bax A, Middledorp AM, Harinck B, Holleboom C, Roosmalen<br />

JV. Unilateral pulmonary edema as a life-threatening<br />

complication of ritodrine. Acta Obstet Gynecol Scand 1999;<br />

78: 915-916.<br />

62. Wolff F, Cartens V, Fisher H, Behrenbeck D, Bolte A.<br />

Cardiopulmonary effects of betamimetic tocolytic and<br />

glucocorticoid therapy in pregnant women. Arch Gynecol<br />

1986; 239: 49–58.<br />

63. Price PH. Review of betamimetic drugs for tocolysis. Report<br />

tospecial FDA public meeting on re-evaluation of ritodrine<br />

labelling. 28th October 1992.<br />

64. Musci MN, Abbasi S, Otis C, Bolegnese RJ. Prolonged ritodrine<br />

exposure and immediate neonatal outcome. J Perinatol 1988;<br />

8: 27–32.<br />

65. Kazzi NJ, Gross TL, Kazzi GM, Williams TG. Neonatal<br />

complications following in utero exposure to intravenous<br />

ritodrine. Acta Obstet Gynecol Scand 1987; 66: 65–69.<br />

66. Schiff E, Sivan E, Terry S et al. Currently recommended oral<br />

regimens for ritodrine tocolysis result in extremely low<br />

plasma levels. Am J Obstet Gynecol 1993; 169: 1059–1064.<br />

67. Zuckerman H, Reiss U, Rubinstein I. Inhibition of human<br />

premature labor by indomethacin. Obstet Gynecol 1974; 44:<br />

787–792.<br />

68. Lunt CC, Satin AJ, Barth WH, Hankins GD. The effect of<br />

indomethacin tocolysis on maternal coagulation status. Obstet<br />

Gynecol 1994; 84: 820–822.<br />

69. Durandy A, Brami C, Griscelli C. The effects of indomethacin<br />

administration during pregnancy on womens’ and<br />

newborns’ T-suppressor lymphocyte activity and on HLA class<br />

II expression by newborns’ leukocytes. Am J Reprod Immunol<br />

Microbiol 1985; 8: 94–101.<br />

70. Morales WJ, Smith SG, Angel JL et al. Efficacy and safety of<br />

indomethacin versus ritodrine in the management of<br />

preterm labor: a randomized study. Obstet Gynecol 1989; 74:<br />

567–573.<br />

71. Besinger RE. A systematic review of adverse events<br />

documented in the use of currently available treatment of<br />

preterm labor. Res Clin Forums 1994; 16: 89–126.<br />

72. Goldenberg RL, Davis RO, Baker RC. Indomethacin-induced<br />

oligohydramnios. Am J Obstet Gynecol 1989; 160: 1196–1197.<br />

73. Niebyl JR, Blake DA, White RD et al. The inhibition of<br />

premature labor with indomethacin. Am J Obstet Gynecol<br />

1980; 136: 1014–1019.<br />

74. Moise KJ, Huhta JC, Sharif DS et al. Indomethacin in the<br />

treatment of preterm labor. Effects on the fetal ductus<br />

arteriosus. N Engl J Med 1988; 319: 327–331.<br />

75. Moise KJ. Effect of advancing gestational age on the<br />

frequency of fetal ductal constriction in association with<br />

maternal indomethacin use. Am J Obstet Gynecol 1993; 168:<br />

1350–1353.<br />

76. Peruzzi L, Gianoglio B, Porcellini MG, Coppo R. Neonatal endstage<br />

renal failure associated with maternal ingestion of<br />

cyclo-oxygenase-type-1 selective inhibitor nimesulide as<br />

tocolytic. Lancet 1999; 354: 1615 (letter).<br />

77. Mittendorf R, Covert R, Boman J, Khoshnood B, Lee KS,<br />

Siegler M. Is tocolytic magnesium sulphate associated with<br />

increased total paediatric mortality Lancet 1997; 250:<br />

1517–1518.<br />

78. Mittendorf R, Pryde P, Khoshnood B, Lee KS. If tocolytic<br />

magnesium sulfate is associated with excess total pediatric<br />

mortality, what is its impact Obstet Gynecol 1998; 92: 308–311.<br />

48 49


79. Kupfermine M, Lessing JB, Yaron Y, Peyser MR. Nifedipine<br />

versus ritodrine for suppression of preterm labour. Br J<br />

Obstet Gynaecol 1993; 100: 1090–4.<br />

80. Papatsonis DN, van Geijn HP, Ader HJ, Lange FM, Bleker OP,<br />

Dekker GA. Nifedipine and ritodrine in the management of<br />

preterm labor: a randomised multicenter trial. Obstet<br />

Gynecol 1997; 90: 230–4.<br />

81. Read MD, Wellby DE. The use of a calcium antagonist<br />

(nifedipine) to suppress preterm labour. Br J Obstet Gynaecol<br />

1986; 93: 933–937.<br />

82. Smith CS, Woodland MB. Clinical comparison of oral<br />

nifedipine and subcutaneous terbutaline for initial tocolysis.<br />

Am J Obstet Gynecol 1993; 170: 280–4.<br />

83. Leonardi MR, Hankins GDV. What’s new in tocolytics. Prev<br />

Prem 1992; 19: 367–384.<br />

84. Blea CW, Barnard JM, Magness RR, Phernetton TM, Hendricks<br />

SK. Effect of nifedipine on fetal and maternal hemodynamics<br />

and blood gases in the pregnant ewe. Am J Obstet Gynecol<br />

1997; 176: 922–30.<br />

85. Waisman GD, Mayorga LM, Camera MI, Vignolo CA,<br />

Martinotti A. Magnesium plus nifedipine: potentiation of<br />

hypotensive effect in preeclampsia Am J Obstet Gynecol<br />

1988; 159: 308–309.<br />

86. Christensen KK, Ingemarsson I, Leideman T, Solum H,<br />

Svenningson N. Effect of ritodrine on labor after premature<br />

rupture of the membranes. Obstet Gynecol 1980;55:187–90.<br />

87. Morales WJ, Madhav H. Efficacy and safety of indomethacin<br />

compared with magnesium sulfate in the management of<br />

preterm labor: a randomized study. Am J Obstet Gynecol<br />

1993;169:97–102.<br />

88. Law HD, du Vigneaud V. Synthesis of 2-pmethoxyphenylalanine<br />

oxytocin (O-Methyl-oxytocin) and<br />

some observations on its pharmacological behaviour. J Am<br />

Chem Soc 1960; 82: 4579–4581.<br />

89. Melin P, Vilhardt H, Lindeberg G, Larsson L-E, Akerlund M.<br />

Inhibitory effect of O-alkylated analogues of oxytocin and<br />

vasopressin on human and rat myometrial activity. J<br />

Endocrinol 1981; 88: 173–180.<br />

90. Melin P, Trojnar J, Johansson B, Vilhardt H, Akerlund M.<br />

Synthetic antagonists of the myometrial response to<br />

vasopressin and oxytocin. J Endocrin 1986; 111: 125–131.<br />

91. Ryden G, Andersson RGG, Berg G, Karlsson S-G, Oscarsson Y.<br />

Binding of four oxytocin analogues to myometrial oxytocin<br />

and arginine-vasopressin binding sites in pregnant women.<br />

Gynecol Obstet Invest 1990; 29: 6–7.<br />

92. Maggi M, Fantoni G, Baldi E, Cioni A, Rossi S, Vannelli GB,<br />

Melin P, Akerlund M, Serio M. Antagonists for the human<br />

oxytocin receptor: an in vitro study. J Reprod Fert 1994; 101:<br />

345–352.<br />

93. Lopez-Bernal A, Phipps SL, Rosevear SK, Turnbull AC.<br />

Mechanism of action of the oxytocin antagonist 1-deamino-<br />

2-D-Tyr(OEt)-4-Thr-8-Orn-oxytocin. Br J Obstet Gynaecol<br />

1989; 96: 1108–1110.<br />

94. Thornton S, Gillespie JI, Anson LC, Greenwell JR, Melin P,<br />

Dunlop W. The effect of the oxytocin antagonists, CAP 476<br />

and F327, on calcium mobilisation in single cultured human<br />

myometrial cells. Br J Obstet Gynaecol 1993; 100: 581–586.<br />

95. Phaneuf S, Asboth G, MacKenzie IZ, Melin P, Lopez Bernal A.<br />

Effect of oxytocin antagonists on the activation of human<br />

myometrium in vitro: atosiban prevents oxytocin-induced<br />

desensitisation. Am J Obstet Gynecol 1994; 171: 1627–1634.<br />

96. Engstrom T, Atke A, Vilhardt H. Oxytocin receptors and<br />

contractile response of the myometrium after long term<br />

infusion of prostaglandin F2a, Indomethacin, oxytocin and an<br />

oxytocin antagonist in rats. Regulat Peptides 1988; 20: 65–72.<br />

97. Hageman WE, Demarest KT, Tolman EL, Peterson PA. in vitro<br />

uterine contractility studies with RWJ-22164-000 (Atosiban).<br />

Internal report no. 419965:2 DD-96016 August 1996.<br />

98. Kinsler VA, Thornton S, Ashford MLJ, Melin P, Smith SK. The<br />

effect of the oxytocin antagonists F314 and F792 on the in<br />

vitro contractility of human myometrium. Br J Obstet<br />

Gynaecol 1996; 103: 373–375.<br />

99. Giussani DA, Jenkins SL, Mecenas CA, et al. The oxytocin<br />

antagonist atosiban prevents androstenedione-induced<br />

myometrial contractions in the chronically instrumented<br />

pregnant rhesus monkey. Endocrinology 1996; 137: 3302–3307.<br />

100. Hahn DW, McGuire JL, Vanderhoof M, Ericson E, Pasquale SA.<br />

Evaluation of drugs for arrest of premature labor in a new<br />

animal model. Am J Obstet Gynecol 1984; 148: 775–778.<br />

101. Jenkin G. Oxytocin and prostaglandin interactions in<br />

pregnancy and at parturition. J Reprod Fertil 1992; 45: 97–111.<br />

102. Akerlund M, Hauksson A, Lundin S, Melin P, Trojnar J.<br />

Vasotocin analogues which competitively inhibit vasopressin<br />

stimulated uterine activity in healthy women. Br J Obstet<br />

Gynaecol 1986; 93: 22–27.<br />

103. Hauksson A, Akerlund M, Melin P. Uterine blood flow and<br />

myometrial activity at menstruation, and the action of<br />

vasopressin and a synthetic antagonist. Br J Obstet Gynaecol<br />

1988; 95: 898–904.<br />

104. Akerlund M. Can primary dysmenorrhea be alleviated by<br />

avasopressin antagonist Results of a pilot study. Acta<br />

Obstetrica Scandinavica 1987; 66: 459–461.<br />

105. Akerlund M, Stromberg P, Hauksson A, Andersen LF, Lyndrup<br />

J, Trojnar J et al. Inhibition of uterine contractions of<br />

premature labour with an oxytocin analogue. Results from a<br />

pilot study. Br J Obstet Gynaecol 1987; 94: 1040–44.<br />

106. Andersen LF, Lyndrup J, Akerlund M, Melin P. Oxytocin<br />

receptor blockade: a new principle in the treatment of<br />

preterm labor Am J Perinatol 1989; 6: 196–199.<br />

107. Eriksson H, Bjorgell P, Akerlund M, Hauksson A, Melin P. Effects<br />

of a tocolytic oxytocin analogue on lipid and carbohydrate<br />

metabolism. Gynecol Obstet Invest 1990; 29: 97–100.<br />

108. Lundin S, Akerlund M, Fagerstrom PO, Hauksson A, Melin P.<br />

Pharmacokinetics in the human of a new synthetic<br />

vasopressin and oxytocin uterine antagonist. Acta Endocrinol<br />

1986; 112: 465–472.<br />

109. Lundin S, Broeders A, Melin P. Pharmacokinetic properties of<br />

the tocolytic agent [Mpa1, D-Tyr(Et)2, Thr4, Orn8]-oxytocin<br />

(Antocin) in healthy volunteers. Clin Endocrinol 1993; 39:<br />

369–374.<br />

110. Zinny M. A probe study to determine the bioavailability, dose<br />

proportionality, and safety of subcutaneous atosiban<br />

administrations compared with intravenous atosiban in normal<br />

female subjects. (Protocol M92–020). Internal report of the<br />

R.W. Johnson Pharmaceutical Research Institute, document no.<br />

354869:1 dated 18 July 1995.<br />

111. Goodwin TM, Millar L, North L, et al. The pharmacokinetics<br />

of the oxytocin antagonist atosiban in pregnant women with<br />

preterm uterine contractions. Am J Obstet Gynecol 1995;<br />

173: 913–917.<br />

112. Kahn J. Rising dose tolerance and safety evaluation of<br />

atosiban (RWJ 22164) in normal female subjects (Protocol<br />

I88-012). Internal report of the R.W. Johnson Pharmaceutical<br />

Research Institute, document no. 20870:1, dated 1 July 1993.<br />

113. Kahn J. Safety and tolerance evaluation of a 12 hour infusion<br />

of atosiban (RWJ 22164) in normal female volunteers<br />

(Protocol I88-084). Internal report of the R.W. Johnson<br />

Pharmaceutical Research Institute, document no. 200769:1,<br />

dated 12 October 1993.<br />

114. Lasseter KC. A double-blind pharmacokinetic and safety study<br />

of120 hours of infusion of Antocin ® (RWJ 22164) in normal<br />

female volunteers (Protocol N93–046). Internal report of the<br />

R.W. Johnson Pharmaceutical Research Institute, document no.<br />

354474:1, dated 30 August 1995.<br />

115. Goodwin TM, Valenzuela GJ, Silver H, Creasy G and the Atosiban<br />

Study Group. Dose ranging study of the oxytocin antagonist<br />

atosiban in the treatment of preterm labor. Obstet Gynecol<br />

1996; 88: 331–336.<br />

116. Valenzuela GJ, Craig J, Bernhardt MD, Holland ML. Placental<br />

passage of the oxytocin antagonist atosiban. Am J Obstet<br />

Gynecol 1995; 172: 1304–1306.<br />

117. Christopher RJ. Determination orf the in vitro plasma protein<br />

binding of atosiban (RWJ-22164-021) and distribution into<br />

human erythrocytes (DM92-04). Scientific memo of the R.W.<br />

Johnson Pharmaceutical Research Institute, document no.<br />

397084:1, dated 16 July 1996.<br />

118. McKown LA, Wu WN. Metabolism of atosiban following a bolus<br />

and i.v infusion dose to female volunteers (study DM95350).<br />

Internal report of the R.W. Johnson Pharmaceutical Research<br />

Institute, document no. 393955:1, dated 21 February 1996.<br />

119. McKown LA, Wu WN. Relative plasma concentrations of the<br />

atosiban M1 metabolite at steady-state following intravenous<br />

bolus and infusion doses to normal female volunteers, and in<br />

maternal and fetal plasma samples following intravenous<br />

infusion to healthy pregnant women (Study DM96318).<br />

Internal report of the R.W. Johnson Pharmaceutical Research<br />

Institute, document no. 408949:1, dated 25 June 1996.<br />

120. Goodwin TM, Paul RP, Silver H, et al. The effect of the<br />

oxytocin antagonist atosiban on preterm uterine activity in<br />

the human. Am J Obstet Gynecol 1994; 170: 474–478.<br />

121. Goodwin TM, Valenzuela G, Silver H, et al. Treatment of<br />

preterm labor with the oxytocin antagonist atosiban. Am J<br />

Perinatol 1996; 13: 143–146.<br />

122. Sanchez-Ramos L et al. A double-blind placebo controlled trial of<br />

oxytocin receptor antagonist (antocin) maintenance therapy in<br />

patients with preterm labor. Am J Obstet Gynecol 1997; 176: SPO<br />

abstr 69, S30.<br />

123. Moutquin J-M, Sherman D, Cohen H, Mohide PT, Hochner-<br />

Celnikier D, Fejgin M, Liston RM, Dansereau J, Mazor M, Shalev<br />

E, Boucher M, Glezerman M, Zimmer EZ. Double-blind,<br />

randomized, controlled trial of atosiban and ritodrine in the<br />

treatment of preterm labor: a multicenter effectiveness and<br />

safety study. Am J Obst Gyn 2000, vol 182, 5.<br />

124. BJOG, accepted for publication.<br />

125. Silver H, Valenzuela G, Sanchez-Ramos L. et al. Maternal side<br />

effects and safety of the oxytocin receptor antagonist<br />

antocin. Am J Obstet Gynecol 1997; 176: SPO abstr 124, S45.<br />

126. Goodwin TM, Randall H, Perry K et al. A report on infant<br />

outcomes at 6 and 12 months after the use of atosiban in the<br />

management of preterm labor. Presented at the ACOG<br />

meeting, New Orleans, USA, May 1998.<br />

127. Data on file. The RW Johnson Pharmaceutical Research Institute.<br />

128. Laros RK jr, Kitterman JA, Heilbron DC, Cowan RM, Tooley<br />

WH. Outcome of very-low-birth-weight infants exposed to<br />

beta-sympathomimetics in utero. Am J Obstet Gynecol 1991;<br />

164: 1657–64.<br />

129. Freysz H, Willard D, Lehr A, Messer J, Boog G. A long term<br />

evaluation of infants who received a beta-mimetic drug while in<br />

utero . J Perinatol Med 1977; 5: 94–99.<br />

130. Polowzcyk D, Tejani N, Lauersen N, Siddiq F. Evaluation of 7<br />

to 9 year old children exposed to ritodrine in utero. Obstet<br />

Gynecol 1984;64: 485–488.<br />

131. Hadders-Algra M, Touwen BC, Huisjes HJ. Long term followup<br />

of children prenatally exposed to ritodrine. Br J Obstet<br />

Gynaecol 1986; 93: 156–61.<br />

50 51

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!