Вы находитесь на странице: 1из 20

REVIEWS

Reproductive health and pregnancy


in women with chronic kidney disease
Kate S. Wiles1,2, Catherine Nelson-Piercy2,3 and Kate Bramham4
Abstract | Chronic kidney disease (CKD) is associated with reduced fertility and an increased risk
of adverse pregnancy outcomes. Rates of pre-eclampsia, fetal growth restriction and preterm
delivery increase incrementally with the severity of CKD and proteinuria. Pre-pregnancy
counselling can facilitate informed decision-making. Safe and effective contraception is required
for women who wish to delay or avoid pregnancy. Pregnancy planning for women who wish to
conceive involves appropriate substitution of known teratogens — including mycophenolate
mofetil, angiotensin blockers and cyclophosphamide — and can aid optimization of disease
control. However, pregnancy, which can occur in women with any stage of CKD, can exacerbate
comorbidities such as anaemia, vitamin D deficiency and hypertension. Increased haemodialysis
provision is associated with improved pregnancy outcomes for women on dialysis. Diagnosis of
pre-eclampsia in women with CKD is complicated in patients with pre-existing hypertension and
proteinuria but can be improved by the use of vasoactive biomarkers as well as placental and fetal
Doppler ultrasound. Pregnancy data for newer drugs used in CKD are limited as pregnancy and
CKD are common exclusion criteria for drug and intervention trials. Although prospective data
may be available for older drugs, the use of most drugs in pregnancy is based on retrospective
data and expert consensus.

Chronic kidney disease (CKD) is estimated to affect 3% Furthermore, a complex interplay with overlapping
of pregnant women in high-income countries1, and the clinical phenotypes exists between CKD and pre-­
prevalence of CKD in this population is predicted to rise eclampsia, with no standardized diagnostic criteria for
owing to increasing maternal age and obesity, highlight- pre-eclampsia super­imposed on CKD. Variation in rates
ing the importance of reproductive health in nephrol- of adverse outcomes is due in part to threshold differ-
ogy. Although CKD is not a barrier to reproduction in ences between studies in the diagnosis and management
most patients, the risk of adverse pregnancy outcomes of superimposed pre-­eclampsia, including iatrogenic
1
Women’s Health Academic — including pre-eclampsia, fetal growth restriction, preterm delivery. Of note, reproductive health issues in
Centre, St Thomas’ Hospital, preterm delivery and accelerated loss of maternal renal relation to renal transplantation are not covered in this
Westminster Bridge Road,
function — is increased in these individuals. Review and are addressed elsewhere2–5.
London SE1 7EH, UK.
2
Guy’s and St Thomas’ NHS In this Review, we outline fertility issues and contra-
Foundation Trust, ceptive options for women with CKD, which are impor- Fertility
Westminster Bridge Road, tant components of women’s health that are frequently Pregnancy rates for women with CKD
London SE1 7EH, UK. neglected in the CKD literature. We also discuss com- Fertility of women with CKD is difficult to precisely
3
Imperial Healthcare NHS
Trust, Du Cane Road,
mon comorbidities (anaemia, vitamin D deficiency evaluate on a population level as pregnancy rates for
London W12 0HS, UK. and hypertension), maternal and neonatal complica- women with CKD can be measured only when both
4
King’s College Hospital NHS tions and therapeutic management of pregnancy in CKD prevalence and the total number of pregnancies
Foundation Trust, Denmark CKD, including drug safety and the use of dialysis. are known. CKD prevalence rates remain an approx-
Hill, London SE5 9RS, UK.
Last, we summarize advances in the identification of imation at best for most women of childbearing age
Correspondence to K.W.  ­pre-eclampsia in the context of CKD. because routine screening for CKD is not under-
and C.N.-P.
kate.wiles@kcl.ac.uk;
Of note, the data on which the information pre- taken. Pregnancy rates can, however, be calculated
Catherine.Nelson-Piercy@ sented in this Review is based are limited by several within specific CKD populations. The UK Obstetric
gstt.nhs.uk factors, including changes in CKD definition over time, Surveillance System6 identified all pregnancies in renal
doi:10.1038/nrneph.2017.187 small study cohorts, disease heterogeneity and insuf- transplant recipients (January 2007 to January 2010)2
Published online 22 Jan 2018 ficient data regarding pre-pregnancy renal function. and in women on dialysis (February 2012 to January

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points indicate that amenorrhoea is present in 37–59% of


women on dialysis13,14. Although the effect of CKD on
• Although chronic kidney disease (CKD) adversely affects fertility, pregnancies can menstruation and fertility is hypothesized to be pro-
occur at all stages of CKD severity portional to the degree of renal dysfunction, the effect
• Safe and effective contraception should be made available for all women with CKD of mild renal dysfunction remains unclear. However,
who do not wish to conceive and those who take teratogenic medications the effects of all stages of CKD on the hypothalamic–­
• CKD increases the risk of adverse pregnancy outcomes, including pre-eclampsia, fetal pituitary–ovarian axis (FIG. 1) are thought to be revers-
growth restriction, preterm delivery and post-partum loss of maternal renal function ible, and normalization of levels of gonadotropins
• Pre-pregnancy hypertension and proteinuria in CKD complicate the diagnosis of (luteinizing hormone (LH) and follicle-stimulating
superimposed pre-eclampsia, which could be improved through vasoactive hormone (FSH)), prolactin and oestrogen is seen
biomarkers as well as placental and fetal Doppler ultrasound ­following renal transplantation15.
• Although data on the use of many drugs in pregnancy are limited, low-dose aspirin, LH and FSH are produced in the anterior pituitary
low‑molecular-weight heparin, labetalol, nifedipine, prednisolone, gland and act synergistically to regulate gonad function,
hydroxychloroquine, azathioprine, ciclosporin and tacrolimus are considered safe
including sex steroid production and gameto­genesis
during pregnancy and breastfeeding
(FIG. 1c). A surge of LH levels at the midpoint of the
menstrual cycle, which is caused by oestrogen-mediated
positive feedback to the hypothalamus and pituitary
2014) in the UK. Combining these data with national gland, is responsible for ovulation. Progesterone —
renal registry data indicated pregnancy rates of 7.6 per released from the corpus luteum and later the placenta
1,000 patients per year for women with a functioning if pregnancy occurs — maintains the endometrium and
renal transplant and 1.4 per 1,000 patients per year prevents menstruation. Prolactin, which is secreted
for women on dialysis compared with national con- by the anterior pituitary gland during pregnancy and
ception rates of 79.1–79.5 per 1,000 women per year 7. lactation, stimulates breast development and milk
Comparable data from Italy reveal a live birth rate of ­production and inhibits ovulation during lactation.
5.5–8.3 per 1,000 women with a functioning transplant Although LH levels are overall increased in women
and 0.7–1.1 per 1,000 women on dialysis (compared receiving dialysis compared with levels in age-matched
with 72.5 per 1,000 women nationally)8. Thus, preg- controls with regular menstrual cycles16, LH levels do
nancy or birth rates among renal transplant recipients not fluctuate in women on dialysis as they do in healthy
and dialysis patients compared with rates in the general individuals17 (FIG. 1a,b). Dysfunctional oestrogen feed-
population are 1:10 and 1:100, respectively. Whether back to the hypothalamus and pituitary in patients with
these data represent a true assessment of fertility or renal dysfunction results in an absence of pre-ovulatory
indicate the impact of CKD on the decision to conceive surges in both oestrogen and LH, which leads to ovula-
remains unknown. tion failure14,16. However, the exact stage and/or degree
of CKD disease severity resulting in dysfunctional LH
Sexual dysfunction in CKD release — and, consequently, suppression of ovulation
Although only limited prevalence data exist, sexual — is currently unknown (FIG. 1d). In addition, renal clear-
dysfunction — an umbrella term used to describe any ance of prolactin in women on dialysis is reduced17,18.
problem experienced during sexual activity, including Reduced prolactin secretion, in conjunction with
reduced libido, difficulty with arousal, dyspareunia and autono­mous secretion that appears resistant to stimu-
anorgasmia — was identified by one study in 81% of 21 latory factors (for example, hypoglycaemia or arginine)
female predialysis patients9. In addition, sexual absti- and suppressive factors (dopamine)16, also contributes to
nence or decreased frequency of intercourse as well as the ­suppression of ovulation.
a negative body image have been described in women
with CKD and are apparent before the need for dialy­ Dialysis provision and fertility
sis10. The aetiology of sexual dysfunction in CKD is Daily haemodialysis is associated with increased patient
likely to be multifactorial, including both biological and survival19, reduced cardiovascular-related hospitaliza-
psychological factors. Although depression may be both tion19, improved blood pressure20 and better quality of
a cause and consequence of sexual dysfunction10–12, the life21 compared with the standard dialysis provision
complex interplay between CKD, sexual dysfunction, (three times per week). The effects of dialysis on fertility
fertility and the perceived risks of pregnancy remains are less clear. One study of seven women showed that
poorly understood. increasing dialysis provision from 16.5 to 28.5 hours
per week was associated with return of menses in two
Mechanistic effects of CKD on fertility out of three previously amenorrhoeic women <40 years
In women with CKD, oligomenorrhoea progresses of age22, and increasing dialysis provision from 12 to
to amenorrhoea as glomerular filtration rate (GFR) 36 hours per week (intensified haemodialysis) has
declines. However, the threshold GFR at which this been used as a tool to maximize fertility in women
progression becomes clinically significant for repro- with end-stage renal disease (ESRD), resulting in con-
ductive health is unknown owing to a lack of data. In ception rates of 15.6%23,24. However, functional assess-
a cohort of 76 women on dialysis aged ≤55 years, 42% ment of the hypothalamic–pituitary–ovarian axis in
Amenorrhoea reported a regular menstrual cycle compared with women r­ eceiving intensified haemodialysis has not
The absence of menstruation. 75% before the start of dialysis13. Small cohort studies been conducted22.

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Physiological menstrual cycle b Menstrual cycle in end-stage CKD

Menstrual Proliferative Menstrual Proliferative


phase phase Midcycle Secretory phase phase phase Midcycle Secretory phase

Ovulation FSH FSH


LH LH
Hormone concentration

Hormone concentration
OE OE
PROG PROG

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28
Day of cycle Day of cycle

c Ovulation and lactation d CKD


Brain
Hypothalamus GnRH
GnRH

• Breast development Anterior


• Milk production pituitary FSH
gland

FSH
↑ Prolactin ↑ LH
Prolactin LH
↓ Renal clearance

Lack of LH
midcycle surge

Uterus Fallopian Anovulation


tube Ovulation

↓ Oestrogen
↑ Oestrogen
Ovary Cyclophosphamide

Figure 1 | Endocrine effects of chronic kidney disease on the hypothalamic–pituitary–ovarian axis. a | During the
Naturefeedback,
midcycle (late follicular) phase of the menstrual cycle, high levels of oestrogen (OE) confer positive Reviews |sensitizing
Nephrology
the pituitary to the effects of gonadotropin-releasing hormone (GnRH) from the hypothalamus. As a result, a surge of
luteinizing hormone (LH) in midcycle occurs, which stimulates ovulation. Following ovulation, progesterone (PROG) is
secreted from the corpus luteum, which acts to prepare the endometrium for implantation. If implantation does not occur,
PROG levels fall and menstruation follows. b | Hypothesized hormone profile during the menstrual cycle in chronic kidney
disease (CKD). Serum LH levels are increased in CKD, but the absence of an LH surge in the late follicular phase of the
menstrual cycle means that ovulation does not occur. Without ovulation and the development of a corpus luteum, PROG
levels do not rise. OE levels are low throughout the cycle. c | The anterior pituitary gland secretes follicle-stimulating
hormone (FSH), LH and prolactin under hypothalamic control. FSH and LH act synergistically to regulate gonad function,
including sex steroid production and gametogenesis in the ovary. At the midpoint of the menstrual cycle, positive OE
feedback leads to a surge in LH, which triggers ovulation. Prolactin production is increased only during pregnancy and
lactation, when it stimulates breast development and lactation while inhibiting ovulation. d | Low OE levels in CKD confer
negative feedback to the hypothalamus–pituitary axis. The absence of an LH surge leads to anovulation. Impaired renal
clearance of prolactin causes inhibition of GnRH secretion from the hypothalamus, also suppressing ovulation.
Cyclophosphamide is gonadotoxic, causing age and dose-dependent premature ovarian failure. The level of CKD at
which these changes occur remains unknown.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Ovarian stimulation Cyclophosphamide and fertility cancer 33 show that LHRH analogues are safe and effec-
The use of drugs to stimulate The alkylating agent cyclophosphamide has a therapeu- tive in the prevention of cyclophosphamide-induced
oocyte development in the tic role in the immunosuppression of rapidly progres- ovarian failure.
ovary before retrieval for sive glomerular disease. However, cyclophosphamide
artificial reproductive
techniques.
has several adverse effects, including dose-dependent Contraception
gonadotoxicity in both men25 and women26, causing Pregnancy planning for women with CKD is impor-
Natural-cycle oocyte oligo­spermia, amenorrhoea and ovarian failure. Toxicity tant because of the increased risk of adverse pregnancy
retrieval is more likely to be irreversible in women, given that outcomes and the common use of teratogenic medica-
Oocyte retrieval from the ovary
the oocyte pool is determined in fetal life and cannot be tions. Although CKD is associated with reduced fer-
following a normal menstrual
cycle, without the use of
regenerated. In addition, older age at initiation of treat- tility (see above), unplanned pregnancies — which are
stimulatory drugs. ment increases the risk of cyclophosphamide-induced associated with an increased risk of obstetric compli-
ovarian failure26,27. cations, even in the absence of comorbidities34 — can
‘Mini pill’ Hence, fertility preservation should be considered occur at all CKD stages and in renal transplant recip-
An oral contraceptive pill that
contains a synthetic
for all women of childbearing age receiving cyclophos- ients. For example, around 33–50% of all pregnancies
progestogen (no oestrogen). phamide. Cryopreservation of oocytes or embryos can among renal transplant recipients are unplanned2,35.
be undertaken before cyclophosphamide treatment Hence, contraceptive counselling should be provided
Intrauterine device is initiated; however, concerns exist regarding ovarian for all women on dialysis who do not want to have chil-
(IUD). A small birth control
stimulation in patients with active systemic lupus ery- dren, particularly those receiving intensified dialysis,
device that is inserted into the
uterus to prevent pregnancy.
thematosus (SLE) owing to the risks of disease exacer- as this method is associated with an increased concep-
May contain a slow-releasing bation and thrombosis, which are hypothesized to be tion rate compared with standard dialysis regimens23.
progestogen (for example, due to increased serum oestrogen concentrations28, and Unfortunately, evidence suggests that few nephrologists
Mirena) or offer contraception treatment delay. Although natural-cycle oocyte retrieval29 discuss fertility issues and contraception with their
without hormonal release
(copper coil).
and cryopreservation of ovarian tissue negate the need patients. Although no published contemporary data
for ovarian stimulation, long-term fertility outcomes exist, questionnaire data from a cohort of 76 women
Subdermal implant remain unclear in women with CKD. Alternatively, with CKD showed that despite 50% being sexually
A small device inserted under LH‑releasing hormone (LHRH) analogues can be used active, only 36% used contraception and only 13% had
the skin. The contraceptive
to inhibit ovarian function for the duration of cyclo- discussed reproductive health issues with their neph-
implant delivers an effective
dose of a synthetic
phosphamide treatment in order to try to preserve rologist 13. Therefore, contraceptive counselling and the
progestogen, providing future fertility. Hypothetical mechanisms by which provision of safe and effective contraception remain
long-acting, reversible LHRH analogues act include the protective inhibition inadequate for many women with CKD. The risks and
contraception. of the hypothalamic–pituitary–ovarian axis and/or a acceptability of different contraceptive methods must be
Progestogen
reduction in ovarian blood flow and therefore expo- balanced against the risks of an unplanned pregnancy.
A synthetic form of sure to circulating cyclophosphamide. A trial of 20 In addition, assessment of contraceptive effectiveness
progesterone. women receiving cyclophosphamide for lupus nephri- should be based on ‘typical use’ rather than presuming
tis showed a reduction in the incidence of premature ‘perfect use’, as discrepancies exist in the failure rate of
ovarian failure in patients receiving an LHRH analogue some contraceptive methods36 (TABLE 1).
compared with that of age-matched and cyclophos-
phamide-dose-matched controls (5% versus 30%)30. Safe and effective methods in chronic kidney
Findings from randomized controlled trials (RCTs) disease
with >250 participants 31,32 and a meta-analysis of Several contraceptive methods exist, but not all are safe
12 RCTs comprising >1,200 patients treated for breast for women with CKD. Progesterone-only contraceptives
— for example, in the form of the ‘mini pill’, an intrauterine
device (IUD) or a subdermal implant — can be used safely
Table 1 | Contraceptive options for women with chronic kidney disease in women with CKD, including patients on dialysis and
transplant recipients.
Contraceptive Perfect-use Typical-use
failure rate (%)* failure rate (%)*
The ability of progesterone-only therapy to inhibit
ovulation varies, depending on the synthetic progestogen
Safe and effective methods used. One study showed that desogestrel provides con-
Progesterone-only pill 0.3 9 sistent inhibition of ovulation (in 102 out of 103 treated
Progesterone intrauterine device 0.2 0.2 subjects) and that this inhibition is maintained even
after 12‑hour delays before re-dosing 37. Therefore, this
Progesterone-only subdermal implant 0.05 0.05
preparation likely confers improved typical-use effec-
Female sterilization 0.5 0.5 tiveness over other progestogens that require re‑dosing
Unsafe and/or ineffective methods within a 3‑hour window each day. The IUD Mirena
Oestrogen-containing methods 0.3 9 (Bayer) and the subdermal implant Nexplanon (Merck,
(pill, patch or ring) Sharp & Dohme) — which are progesterone-only, long-­
Male condom 2 18 acting, reversible contraceptives — provide effective
contraception for 5 and 3 years, respectively. Moreover,
Female condom 5 21
these contraceptives do not rely on daily compliance
No method 85 85 and have a typical-use failure rate comparable to that of
*% of couples experiencing an unplanned pregnancy in the first year of use. Data from REF. 36. ­sterilization36 (TABLE 1).

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Hypothalamus Systemic changes methods are therefore likely to be contraindicated


• ↓ Sensitivity to RAAS for many women with CKD, particularly given the
• ↑ Water retention ­availability of safer, more effective alternatives.
Renal haemodynamics
• ↓ Plasma osmolality
• 50–85% ↑ RBF Condoms are effective against the transmission of
• 50% ↑ GFR HIV and sexually transmitted diseases. However, these
• Afferent and efferent
arteriole vasodilatation barrier methods are unforgiving of imperfect use —
approximately 1 in 5 women conceive within a year 36
↓ Threshold for
? ? (TABLE 1) — and should therefore not be considered as
vasopressin release
Relaxin Nitric oxide an effective long-term contraceptive option.
Progesterone
Endocrine function Contraceptive failure
• ↑ Erythropoietin
• ↑ RAAS activity The most widely used emergency contraceptives (for
Corpus example, levonorgestrel) do not contain oestrogen
luteum and can therefore be used safely in patients with CKD
to prevent pregnancy within 72 hours of unprotected
Tubular function intercourse. For the medical termination of pregnancy,
• ↑ Glycosuria Structural changes a combination of mifepristone and misoprostol is most
• ↑ Calcium excretion • Pelvicalyceal dilatation
• ↑ Glycosaminoglycans • Hydronephrosis commonly used. However, only limited data are available
• ↓ Reabsorption of • ↑ GBM pore size contributes on the use of both drugs in the context of renal impair-
uric acid to physiological proteinuria ment. Whereas mifepristone is largely metabolized in
the liver, misoprostol is predominantly renally excreted.
Figure 2 | Physiological changes in the renal system in pregnancy. The kidney The peak concentration, half-life and bioavailability of
undergoes structural, haemodynamic, tubular and endocrine changes during pregnancy.
Natureto
Reviews | Nephrology misoprostol are all potentially increased in patients with
In addition, systemic effects in pregnancy are mediated by changes the renin–
angiotensin–aldosterone system (RAAS) and vasopressin. Relaxin, progesterone and nitric
CKD. The clinical relevance of potentially reduced clear-
oxide are hypothesized to play a part in mediating the haemodynamic changes in the ance of misoprostol is unknown, and no dose reduction
kidney. GBM, glomerular basement membrane; GFR, glomerular filtration rate; RBF, renal is recommended, although increased clinical surveillance
blood flow. Adapted by permission from BMJ Publishing Group Limited from REF. 185. should be considered for women with a p ­ re-pregnancy
estimated GFR (eGFR) of <30 ml/min/1.73 m2.

Theoretical concerns exist regarding the use of IUDs Renal physiology during pregnancy
in immunosuppressed women, as immunosuppres- The physiological changes that occur during preg-
sants might reduce their effectiveness owing to inhib­ nancy include haemodynamic, tubular, endocrine and
ition of the uterine inflammation that forms part of ­structural alterations in the kidney (FIG. 2).
their contraceptive mechanism. However, the uterine
milieu is predominantly populated by macrophages, Renal blood flow
whereas immunosuppression used in the management During pregnancy, renal blood flow (RBF) increases by
of immunological renal disease and following transplan- 50–85% with a concurrent increment in GFR43. This
tation acts predominantly via lymphocyte inhibition (see physiological hyperfiltration — mediated by vasodilata-
below), suggesting that different pathways are involved. tion of both afferent and efferent arterioles — occurs in
No evidence of an excess of IUD failures following trans- the absence of glomerular hypertension44 and is there-
plantation has been reported38,39. Furthermore, concern fore different from pathological hyperfiltration associ-
regarding pelvic infection in the context of immunosup- ated with glomerular injury and CKD. Pregnancy and
pression also seems to be unfounded as no correlation multiparity are therefore not associated with a decline in
was found between infective complications and numbers maternal renal function in the absence of CKD. Dextran
‘Combined pill’
of CD4+ T cells in women with HIV-mediated immuno­ clearance modelling suggests that a physio­logical
Contraceptive pill containing
a synthetic oestrogen and suppression40. Similarly, a retrospective study of 11 increase in glomerular membrane pore size occurs with
progestogen. women with renal transplants and a total of 484 months pregnancy, which underlies the finding of gestational
of progesterone–IUD use reported no cases of pelvic proteinuria in the absence of a change in hydrostatic
Transdermal patch infection or unplanned pregnancy 41. force45,46. Amino acid loading during pregnancy can fur-
Contraceptive patch that
delivers synthetic oestrogen
ther increase RBF and GFR, which suggests that extra
and progestogen through Other contraceptive methods filtration capacity — in addition to that already seen in
the skin. Oestrogen-containing contraceptives include the adaptation to pregnancy — exists46.
oestrogen-­containing ‘combined pill’, the transdermal As the above described renal physiological changes are
Vaginal ring
patch and the vaginal ring. All of these methods con- measurable from as early as 8 weeks of gestation, they are
A soft plastic ring worn inside
the vagina that provides fer a risk of hypertension, venous thromboembolism not thought to be mediated by the fetoplacental unit,
contraception via the release (VTE), arterial thrombosis and cervical cancer 42. These which is still underdeveloped at this stage. Instead, the
of synthetic oestrogen and risks are particularly relevant for women with CKD who observed rise in GFR in both the luteal phase of the men-
progestogen. are at increased risk of chronic hypertension, coagulo­ strual cycle and pregnancy may be mediated by relaxin,
Corpus luteum
pathy (due to antiphospholipid antibodies or nephrotic which is released by the corpus luteum47. In rats, both ova-
The remnants of the ovarian syndrome), vascular diseases, and/or neoplasia (in the riectomy and administration of neutralizing relaxin anti-
follicle after ovulation. context of immunosuppression). Oestrogen-containing bodies impaired the renal adaptation to pregnancy48, with

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

evidence from other animal studies that relaxin mediates pregnancy, suggesting an increased sensitivity to RAAS
its effects via matrix metalloproteinase 2 and endothelin in pre-eclampsia, which has been attributed to activat-
receptor type B49. However, other unknown factors — ing angiotensin receptor autoantibodies and angio­
possibly progesterone and nitric oxide50 — also have a genic proteins, including soluble fms-like tyrosine
role, as evidenced by the physiological renal adaptation kinase 1 (sFLT1)64.
to assisted-conception pregnancies in which the corpus Vasopressin is an antidiuretic hormone that is released
luteum is absent and relaxin is not detectable51. by the posterior pituitary gland and stimulates water
reabsorption in the kidney in response to an increase in
Measuring GFR in pregnancy osmolality or a reduction in extracellular fluid volume.
As a result of the dynamic changes in RBF and filtra- The systemic arterial vasodilatation that occurs in preg-
tion fraction that lead to a sustained, augmented GFR nancy is thought to lead to non-osmotic stimulation of
until term52, the serum creatinine concentration falls vasopressin, effectively lowering the osmotic threshold
with physiological renal adaptation to pregnancy. for vasopressin release, contributing to water retention
Modification of Diet in Renal Disease (MDRD)53 and and a reduction in plasma osmolality in pregnancy 65.
CKD-epidemiology (CKD-EPI)54 formulae tend to
underestimate GFR and cannot be used in pregnancy. Outcomes for pregnant women with CKD
Cystatin C provides an alternative surrogate measure Historically, clinicians advised avoidance of pregnancy
of GFR as it is freely filtered at the glomerulus and, in for women with CKD66, a recommendation that — on
pregnancy, synthesis is not thought to be affected by the basis of qualitative assessment — is associated with
the utero-placental unit 55,56. However, serum concen- frustration, anger and regret 67,68. Over the past decades,
trations of maternal cystatin C fail to correlate with the focus of advice has changed owing to advances in
other measures of GFR in pregnancy, including iohexol obstetric surveillance and neonatal care. Nonetheless, we
clearance57, serum creatinine58 and eGFR59. This failure find that many clinicians still desire a general ‘thresh-
to correlate may be caused by an increase in glomerular old’ of renal disease severity above which pregnancy is
negative charge, which reduces the excretion of anionic ill-­advised. Such a threshold cannot be determined by
cystatin C57. Assessment of renal function in pregnancy a single creatinine measurement and does not reflect
is therefore limited to serial monitoring of serum cre- the complex effects of renal dysfunction on pregnancy.
atinine level, and caution should be taken in interpret- Instead, effectively communicated, individualized
ing data from publications that use eGFR equations in pre-pregnancy counselling — based on disease aetiol-
pregnancy 60, which might underestimate pre-pregnancy ogy and severity, rate of GFR decline, obstetric history
CKD severity. and the presence of additional risk factors (see below)
— should enable women with any stage of CKD and
Tubular changes their partners to make an informed choice regarding
During pregnancy, several changes occur that affect pregnancy. Pre-pregnancy counselling is recommended
renal tubular function. Glycosuria frequently occurs for all women with pre-existing medical conditions69.
during pregnancy owing to an increase in filtration In  a recent survey of women with CKD, 66 of 72
in conjunction with reduced tubular reabsorption. (92%) respondents reported that expert pre-pregnancy
Although calcium excretion is increased, leading to ­counselling assisted with decision-making 70.
supersaturation of the urine, simultaneous increases in
urinary glycoproteins are thought to protect the kid- Risk factors
neys against the formation of renal calculi and nephro­ Data from population studies (778 women with CKD)71,
lithiasis61,62. Furthermore, reabsorption of uric acid is large cohort studies (504 women with CKD)60 and a
reduced in pregnancy, resulting in increased renal excre- meta-analysis (23 studies, including 1,514 pregnant
tion. By contrast, serum uric acid is increased in pre-­ women with CKD)72 show that women with CKD have
eclampsia but is insufficiently sensitive for diagnosis63, worse maternal and neonatal outcomes — ­including
with diagnostic specificity reduced still further if there higher rates of pre-eclampsia, preterm delivery,
is coexisting CKD. small-for-gestational-age infants, admission to neonatal
intensive care units (NICUs) and perinatal death — than
Endocrine and systemic changes women without CKD. Absolute rates of adverse out-
Several changes occur during pregnancy, which relate comes vary between studies and are affected by cohort
to the renal synthesis of or renal response to hormones. size, the measure of GFR60 and variance in the thresholds
Pregnancy is a state of profound physiological vasodilata- for both iatrogenic preterm delivery and admission to
tion with mediators (including progesterone, nitric oxide NICUs. However, an increment in all complications with
and prostaglandins) leading to a 5–­10 mmHg reduction worsening renal function exists60 (FIG. 3).
in blood pressure during pregnancy. This change occurs Additional factors associated with adverse preg-
despite evidence of increased renal renin production and nancy outcomes that are independent of renal disease
activation of the systemic renin–angiotensin–­aldosterone severity include pre-existing hypertension60, transplan-
system (RAAS), therefore suggesting that pregnancy is tation status3, lupus nephritis73, proteinuria74 and super­
a state of relative RAAS resistance. By contrast, despite imposed pre-eclampsia75. However, one study reported an
high blood pressure, the RAAS is suppressed in patients increased risk of adverse pregnancy outcomes in women
with pre-eclampsia compared with levels in normal with stage 1 CKD (OR 1.88; 95% CI 1.27–2.79), even in

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Bramham et al.78 b Piccoli et al.60

Pre-eclampsia

SGA <5% SGA <5%

Preterm delivery Preterm delivery


<34/40 <34/40

Preterm delivery Preterm delivery


<37/40 <37/40

NICU admission NICU admission

25% reduction in CKD stage shift


eGFR at 6 months or RRT start
post-partum

0 20 40 60 80 100 0 20 40 60 80 100
Rate of complications (%) Rate of complications (%)

Controls CKD stage 2 CKD stage 4–5 Controls CKD stage 2 CKD stage 4–5
CKD stage 1 CKD stage 3 CKD stage 1 CKD stage 3

Figure 3 | Adverse pregnancy outcomes according to chronic kidney disease stage. Two cohort studies60,78 show that
worsening of renal function (as assessed by chronic kidney disease (CKD) stage) correlates with Nature Reviews
a consistent | Nephrology
increment in
several pregnancy complications: preterm delivery; pre-eclampsia; neonatal intensive care unit (NICU) admission; loss of
residual maternal renal function; and small-for-gestational-age (SGA) infants (parts a and b).  Variation of absolute rates
of adverse pregnancy outcomes between the studies could be due to differences in cohort size, the use of estimated
glomerular filtration rate (eGFR) in pregnancy and different thresholds for both iatrogenic preterm delivery and NICU
admission. In addition, pre-eclampsia was excluded from the control group in one study78. RRT, renal replacement therapy.

the absence of systemic disease, hypertension and pro- with advanced disease (CKD stages 4–5) remain small.
teinuria, leading to the hypothesis that CKD per se confers Interestingly, one study 60 reported that even women with
risk in pregnancy 60. CKD stage 1 are at risk, with 7.6% (28 out of 370) demon-
strating a post-partum decline in renal function. However,
Progression of maternal renal disease this finding could relate to the use of GFR estimating equa-
Until the past decade, our understanding of CKD progres- tions during pregnancy that might have underestimated
sion in pregnancy was limited to findings from two small the severity of renal disease and misclassified women with
cohort studies76,77. A retrospective study of 70 pregnancies a higher pre-pregnancy stage of disease as CKD stage 1 (see
in 58 women (mean serum creatinine level  168 μmol/l dur- above). A meta-analysis from 2015 that compared renal
ing early pregnancy) reported a 25% reduction in kidney disease progression in 552 pregnant women and 716 non-
function in 43% of women in the 6 weeks after delivery, pregnant women with CKD reported no significant differ-
with 11% of the cohort progressing to ESRD by 6 months ence (OR 0.96; 95% CI 0.69–1.35); however, in that study,
post-partum76. Similarly, a prospective Italian study of 49 progression was crudely defined as a doubling of serum
pregnancies reported that women with a pre-pregnancy creatinine level or a >50% reduction in eGFR, and only
GFR <40 ml/min/1.73 m2 and proteinuria >1 g per day had women with CKD stages 1–3 were included72.
an accelerated decline in renal function (1.17 ± 1.23 ml/min
per month) at 6 months post-partum compared with Specific disease conditions
pre-pregnancy rates (0.21 ± 0.20 ml/min per month)77. Most cohort studies of CKD in pregnancy are hetero­
Comparable rates of loss of maternal renal function geneous and include a variety of renal disease aetiologies.
were also found in studies from 2015 (REF. 60) and 2016 Maternal outcomes for specific disease aetiologies are
(REF. 78), but the sample sizes of pregnancies in women discussed below.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Autosomal dominant polycystic kidney disease. which showed that lupus flares and lupus nephritis occur
Autosomal dominant polycystic kidney disease in 26% and 16% of pregnancies, respectively 91. A pro-
(ADPKD) has an estimated prevalence of 4 in 10,000 spective cohort study comprising 61 pregnant women
individuals79. Although normotensive women with demonstrated that an increase in lupus disease severity
ADPKD reportedly have comparable pregnancy out- score was associated with a proportional and significant
comes to those of unaffected family members, pre-­ increase in the risk of preterm delivery 73. Similarly, the
existing hypertension is a recognized risk factor for presence of lupus nephritis increases the risk of preterm
adverse pregnancy outcomes in ADPKD75. Another delivery among women with SLE90. Other risk factors for
study reported no difference in neonatal risk between adverse pregnancy outcomes in lupus nephritis include
54 women with ADPKD compared with 92 women with black92 and Hispanic93 ethnicity, pre-existing hyper­
simple cysts, although maternal complications, includ- tension89 and severity of proteinuria94. Therefore, delayed
ing hypertension and pre-eclampsia, were increased in conception until 6 months after lupus disease activity is
patients with ADPKD80. Isolated case studies support recommended, and pre-pregnancy renal biopsy might be
the theory that kidney volume is unaffected by preg- indicated to exclude active disease if proteinuria is per-
nancy, and that the condition does not compromise sistent. Given the risks associated with lupus nephritis, in
uterine growth81. conjunction with the clinical difficulties in distinguishing
Through the autosomal dominant nature of ADPKD, active lupus nephritis from pre-eclampsia (TABLE 2), all
offspring have a 50% chance of inheriting the condi- women with current or previous lupus nephritis should
tion; this probability is markedly reduced through the be managed by a specialist team that includes both physi-
use of pre-implantation genetic diagnosis (PGD) to iden- cians and obstetricians with expertise in SLE. For women
tify relevant mutations in embryos before implantation. with lupus nephritis and antiphospholipid antibodies,
However, the ethics of genetic testing for an adult-onset active disease or proteinuria, prophylaxis against VTE
disorder — which is asymptomatic for many years — using low-molecular-weight heparin should be consid-
need to be considered. In a single-centre study of 96 ered; however, the threshold urinary protein:creatinine
women with ADPKD, 50% of patients with CKD and ratio at which the risk of this complication becomes
63% of those with ESRD would have opted for PGD if it clinically relevant remains unknown95. Unless contra­
were available82. Single-embryo transfer is recommended indicated, women with lupus nephritis should be offered
in women with CKD owing to the additive risks associ- low-dose aspirin for pre-eclampsia prophylaxis and
ated with multifetal pregnancy, including p ­ re-eclampsia hydroxychloroquine (see below).
and preterm delivery. Fetal complications in lupus nephritis may arise
through exposure to maternal autoantibodies against
Primary glomerulonephritis. Only limited data on the intracellular ribonucleoproteins Sjogren syndrome
pregnancy outcomes in women with primary glomer- type A antigen (SSA; also known as 52 kDa Ro protein
ulonephritis are available to guide counselling and or TRIM21) and SSB (also known as lupus La protein),
management, as highlighted in a systematic review 83. which undergo placental transfer and confer a 2% risk of
For example, to our knowledge only two studies of congenital heart block (CHB) and a 16% risk of neonatal
pregnancy in women with minimal change disease have cutaneous lupus96. Whereas neonatal cutaneous lupus is
been performed84,85. However, the association of pre-­ benign and resolves with elimination of the maternal
existing hypertension, proteinuria and renal impairment antibody, CHB and endocardial fibroelastosis are associ-
with worse pregnancy outcomes is consistent between ated with increased mortality. Therefore, surveillance
­studies of different CKD aetiologies. Proteinuria has of fetal heart rate and echocardiography for suspected
been found to be an important determinant of outcome cardiac involvement are recommended for women with
in immunoglobulin A (IgA) nephropathy, which is the SSA+ and SSB+ antibodies. Weekly fetal echocardio­
most common primary glomerulonephritis in pregnant graphy is recommended from 16 weeks gestation if
women83. Proteinuria and birthweight are negatively there is a history of a previous affected infant, although
correlated86, and the presence of >1 g per day of uri- the clinical benefits and cost-effectiveness of high-level
nary protein has been shown to be associated with loss surveillance in women without an affected child remain
of residual renal function independent of pregnancy 87. unknown97. Maternal hydroxychloroquine is associated
A comparison of pregnancy outcomes between women with a reduced risk of CHB in women with SLE and
Pre-implantation genetic
diagnosis
with IgA nephropathy and other glomerular diseases has ­previously affected pregnancies98.
(PGD). Examination of the not been carried out.
genetic profile of a gamete or Diabetic kidney disease. Kidney disease is evident in
embryo before implantation. Lupus nephritis. SLE predominantly affects women of 2.5–6.5% of pregnancies in women with type 1 diabe-
childbearing age, with lupus nephritis occurring in ~40% tes mellitus99, although rates of up to 25% are described
Ribonucleoproteins
Protein–RNA complexes. of cases88. Although the risk of adverse pregnancy out- when disease definitions are extended to include
comes is increased in women with active lupus nephritis89, microalbuminuria with a normal eGFR100. Diabetes
Endocardial fibroelastosis even in the context of preserved renal function90, preg- is associated with an increased risk of complications
A disease of the endocardium nancy outcomes are improved when disease remission — including progression of retinopathy, ketoacidosis,
characterized by collagen
deposition, endocardial
is achieved before pregnancy. However, pregnancy can exacerbation of pre-existing cardiovascular disease and
thickening and ventricular trigger disease relapse, as evidenced by findings from a fetal death — independent of whether or not there is
hypertrophy. meta-analysis of 37 studies comprising 2,751 pregnancies coexisting renal disease. A retrospective analysis of 43

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Macrosomia pregnancies in women with diabetic kidney disease who Important comorbidities
A large-for-gestational-age had a median pre-pregnancy serum creatinine level Anaemia, vitamin D deficiency and chronic hyper­
infant. of 68 μmol/l (0.77 mg/dl) reported much higher rates of tension are comorbidities that are associated with
adverse pregnancy outcomes than expected for the level adverse pregnancy outcome and can develop secondary
of renal impairment — including pre-­eclampsia in 42% to underlying CKD.
and preterm delivery in 77% of patients99. Gestational
proteinuria can be substantial and the risk of VTE Anaemia
should be considered, although only limited data regard- Gestational increases in plasma volume in pregnancy
ing the proteinuric threshold for heparin prophylaxis are are proportionally higher than the corresponding
available95. Fetal growth surveillance in patients with dia- increase in red blood cell mass, leading to haemo­
betic kidney disease may be falsely reassuring, as growth dilution and a relative fall in haemoglobin levels.
restriction due to underlying CKD can be masked by Although variation exists in the definition of anaemia
hyperglycaemia-induced m ­ acrosomia. Congenital mal- in pregnancy (haemoglobin concentrations <105 g/l
formation rates increase with poor glycaemic control to <110 g/l, depending on gestation106,107), values that
around the time of conception, but diabetic kidney are <85 g/l in the general obstetric population have
disease confers an additional risk of fetal malforma- been shown to be associated with increased risks
tion compared with women without nephro­p athy of low birthweight (<2,500 g) and preterm delivery
(adjusted OR 2.5; 95% CI 1.1–5.3)101. Some evidence (<37 weeks) by 62% and 72%, respectively, across
supports intensification of RAAS blockade through the ­multiple ethnic groups108.
use of either angiotensin-converting enzyme inhibi- Erythropoiesis is regulated by erythropoietin, which
tors or angiotensin receptor blockers before pregnancy, is produced by fibroblast-like cells within the kidney.
although these need to be discontinued in p ­ regnancy Erythropoietin concentrations increase approximately
because of fetotoxicity (see below). Data from single-­ twofold during pregnancy 109. As women with CKD may
arm studies with 8–24 participants show that pre­ have insufficient capacity for a gestational increase in
conception proteinuria is reduced by RAAS blockade erythropoietin production, supplementation with
with an associated decrease in pregnancy-induced pro- synthetic erythropoietin may be required, even in
teinuria102,103. The use of intensified targets for blood the context of mild renal impairment. For women
pressure (<135/85 mmHg) and proteinuria (<300 mg who required erythropoietin before pregnancy, an
per day) — in conjunction with preconceptual RAAS increased dose should be anticipated during pregnancy.
blockade — is associated with obstetric outcomes that As erythropoietin is a large molecule that does not
are comparable to those of women with diabetes but cross the placental barrier, its use is considered safe in
without proteinuria104. Comprehensive guidelines for the pregnancy and breastfeeding 2,110,111; however, a theor­
management of diabetic kidney disease in pregnancy are etical risk of exacerbating pre-existing or n ­ ew-onset
available elsewhere105. hypertension exists.

Table 2 | Clinical overlap between normal pregnancy, CKD, active lupus and pre-eclampsia
Clinical features Normal pregnancy CKD Lupus flare Pre-eclampsia
Hypertension No Yes Yes Yes
Proteinuria <300 mg per day Yes Yes Yes
Haematuria No Yes Yes No
Increased serum Physiological Not above pre-pregnancy Yes Yes
creatinine creatinine level
Rate of change in Third trimester None unless disease Can be rapid (hours or Can be rapid
serum creatinine progression or days) (hours or days)
superimposed acute injury
Skin Hyperpigmentation, No Malar rash, discoid lupus, No
striae acute cutaneous lupus
Joints Mechanical pain No Arthritis No
Hair loss Yes (post-partum No Yes No
telogen effluvium)
Haemoglobin Iron-deficiency Erythropoietin deficiency Anaemia of chronic Haemolysis
anaemia disease, haemolysis
Platelets Gestational Normal Thrombocytopenia Thrombocytopenia
thrombocytopenia
Other blood tests ↑ ESR ↓ eGFR prior to pregnancy • ↓ Complement • Transaminitis
• ↑dsDNA • ↓ PLGF
• ↑sFLT1
CKD, chronic kidney disease; dsDNA, double-stranded DNA; eGFR, estimated glomerular filtration rate; ESR, erythrocyte
sedimentation rate; PLGF, placenta growth factor; sFLT1, soluble fms-like tyrosine receptor kinase 1.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The most common cause of anaemia in pregnancy (IU) per week until replete for women with a serum
is iron deficiency, which is estimated to affect >40% of calcifediol <20 ng/ml (50 nmol/l). This treatment is
pregnancies112. Hence, pregnant women with CKD may followed by a maintenance daily dose of 400–1,000 IU,
need both iron and erythropoietin supplementation. depending on ethnicity and body mass index 121.
Markers of iron deficiency in nonpregnant patients Although calcifediol is the major circulating form of
with CKD include ferritin <100 μg/l, transferrin satura- vitamin D, it has low biological activity until converted
tion (<20%), >6% hypochromic red cells and reticulo­ to calcitriol (also known as 1,25‑dihydroxyvitamin D3).
cyte haemoglobin content equivalent to <25 pg 113; Serum calcitriol levels are approximately threefold
however, the specificity and sensitivity of these markers higher in the first trimester and five to six times higher
in assessing iron status in pregnancy are unknown and in the third trimester than those in nonpregnant
should be investigated further. Oral iron is inexpen­ women125. To what extent this increase is dependent on
sive and accessible, although the intravenous route may the capacity of the kidney to augment ­1α‑hydroxylase
provide more effective replacement in CKD owing to enzyme activity is unknown126, as 1α‑hydroxylase activ-
better bioavailability 114. Parenteral iron is generally ity is also present in the colon, skin, macrophages and
considered safe in pregnancy and breastfeeding 5,115–118, the placenta127. In the absence of better evidence, active
although a rare risk of an allergic reaction exists. vitamin D analogues (such as alphacalcidol and calcit-
Vitamin B12 (also known as cobalamin) deficiency riol) should be continued throughout pregnancy at the
is a rare cause of anaemia during pregnancy but may appropriate pre-pregnancy dose.
need to be considered for patients on low-protein diets. In nonpregnant women with CKD, assessment of
Protein prescription in patients with CKD who are not bone health includes both vitamin D status and para-
pregnant requires a balance between the potential for thyroid hormone levels. Hyperparathyroidism can occur
a reduction in renal disease progression and optimal in CKD secondary to both vitamin D deficiency and
nutritional status. However, in pregnancy, nutritional impaired renal clearance of phosphate. Of note, as
status predominates and low-protein diets should not pregnancy does not cause an increase in parathyroid
be recommended. Of note, assessment of vitamin B12 hormone levels, they can be interpreted as for nonpreg-
status during pregnancy is inherently difficult, as nant individuals. However, treatment of hyperpara­
physiological lowering of plasma vitamin B12 ­levels thyroidism with calcimimetics is not recommended
by up to 30% contributes to misdiagnosis of defi- owing to i­ nadequate safety data (see below).
ciency when nonpregnant reference ranges are used
for interpretation119. Chronic hypertension
Hypoxia-inducible factor (HIF) activators are an Chronic hypertension affects 20–50% of pregnant
emerging class of drugs that might have a therapeutic women with CKD, depending on the severity of renal
role in the management of renal anaemia via stimula- disease60. A meta-analysis comprising >750,000 women
tion of erythropoietin production. However, the small shows that chronic hypertension is associated with
size of these molecules potentially enables placen- superimposed pre-eclampsia (relative risk (RR) 7.7;
tal transfer, and HIF has multiple direct and indirect 95% CI 5.7–10.1), preterm delivery (RR 2.7; 95% CI
effects that have the potential to modulate almost any 1.9–3.6), low birthweight (RR 2.7; 95% CI 1.9–3.8),
developmental, physiological process120. No formal rec- NICU admission (RR 3.2; 95% CI 2.2–4.4) and peri­
ommendation has been made for the use of this class of natal death (RR 4.2; 95% CI 2.7–6.5)128. Control of
drug in pregnancy, but on the basis of their molecular hypertension is a key component of nonpregnant CKD
characteristics, we would not recommend their use at management. However, a historical, theoretical concern
conception or during pregnancy. that aggressive blood pressure control during preg-
nancy could impair fetal growth and contribute to fetal
Vitamin D deficiency morbidity exists. In the 2015 Control of Hypertension
Vitamin  D deficiency affects 13–64% of pregnant in Pregnancy Study (CHIPS) trial, in which 987
women121 and is associated with increased incidences of patients with hypertension were randomly assigned
pre-eclampsia and gestational diabetes mellitus. Meta- to either tight (85 mmHg) or less tight (100 mmHg)
analyses report that oral vitamin D supplementation diastolic blood pressure targets129, tight blood pres-
is associated with reduced risks of pre-eclampsia, low sure control (mean 133/85 mmHg) was not associated
birthweight and preterm birth, although inconsistencies with an increase in adverse pregnancy outcomes but
exist in the findings of individual studies122,123. was associated with a reduced risk of severe maternal
Vitamin  D status in pregnancy is assessed by hypertension compared with less tight control (mean
measuring serum levels of calcifediol (also known as 139/90 mmHg). Although the CHIPS trial excluded
25‑hydroxyvitamin D, a precursor of active vitamin D), women with proteinuria, increasing expert consen-
Hyperparathyroidism
Increase in parathyroid although optimal serum calcifediol levels (variably sus is that hypertension in pregnant women with
hormone levels, which can be defined as a >20–30 ng/ml (50–75 nmol/l)124), as well as CKD should be treated to a target blood pressure of
primary due to pathology optimal doses of colecalciferol or ergocalciferol, remain <140/90 mmHg (REFS 83,130). Nevertheless, no com-
within the parathyroid gland unknown. According to expert consensus, serum cal- prehensive studies of chronic hypertension specific
or secondary due to
hypocalcaemia or
cifediol levels are to be checked in pregnancy for all to pregnant women with CKD have been carried out to
hyperphosphataemia (both of pregnant women with CKD, and replacement colecalci­ date, and published data are insufficient to establish
which can be caused by CKD). ferol can be given at a dose of 20,000 international units evidence-based targets.

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Therapeutic management to the timing of intercourse and the regularity of the


As many pregnant women with CKD require medi- menstrual cycle and should be undertaken at least once
cation for control of hypertension, inflammatory glo- per month for those with irregular cycles.
merular disease or prevention of transplant rejection, Labetalol, nifedipine, captopril and enalapril are
an understanding of drug compatibility with pregnancy compatible with lactation133,134. Methyldopa is substi-
and breastfeeding is essential before any prescription tuted post-partum because of the risk of exacerbating
in all women of childbearing age. However, manufac- postnatal depression.
turers and medication administrative bodies (such as
the US Food and Drug Administration (FDA)) have Immunosuppressant drugs
understandably taken a cautionary approach to med- Corticosteroids. Steroids are used to inhibit immune
ication use in pregnancy. As a result, very few drugs responses in inflammatory and autoimmune condi-
have a licence for use in pregnancy, and pregnant and tions and to prevent transplant rejection. Prednisolone
lactating women are often specifically excluded from is the preferred agent for use in pregnancy as placen-
­studies of safety and efficacy. Recommendations regard- tal metabolism of prednisolone results in only 10% of
ing safety are usually based on data from retrospective the maternal dose reaching the fetal circulation135,136.
case series, isolated case reports and animal studies. An However, as corticosteroid use is associated with an
attempt by the FDA to translate available drugs into increased risk of gestational diabetes mellitus, hyper-
a letter-based category (A–D plus X; corresponding tension, urinary tract and other infections as well
to increasing levels of evidence of harm) is simplistic as preterm delivery 137, all corticosteroids should be
and has led to false assumptions about drug safety. administered at the lowest possible therapeutic dose.
For example, on the basis of isolated case reports and Screening for gestational diabetes mellitus is recom-
supratherapeutic dosing in animals, azathioprine is clas- mended at 28 weeks gestation or sooner if other risk
sified by the FDA as category D and therefore unsafe factors such as increased body mass index and ethnicity
for use in pregnancy; however, data in humans indi- are present. Although historical, retrospective studies
cate that the drug is safe in clinical practice (see below). suggested an association between first-trimester corti-
Although FDA categorization was changed in 2015 to a costeroid exposure and an increase in the incidence of
Pregnancy and Lactation Labelling Rule (PLLR) aimed cleft lip and palate138,139, this link has not been substan-
at improving the available information, the revision of tiated in prospective case–control studies140–142, and a
labelling for existing drugs may take up to 3 years. An cohort study with >50,000 steroid-exposed pregnancies
overview of medications commonly used in nephrology failed to show any risk of oral facial clefts143. Parenteral
practice and their safety in relation to their use during steroids to cover the physiological stress of delivery
conception, pregnancy and ­breastfeeding is provided should be considered for women receiving >7.5 mg
in TABLE 3. prednisolone per day for >2 weeks during pregnancy.
The concentration of prednisolone in breastmilk is
Antihypertensive drugs estimated to be 0.015–0.074% of the maternal dose133. No
The safety of antihypertensive drugs in pregnancy is adverse effects have been reported in breastfed infants
based on historical, retrospective analyses and favours with maternal use of any corticosteroid, and evidence
older classes of drugs, including the adrenergic recep- suggests that neonatal adrenal suppression ­following
tor agonists labetalol and methyldopa and the calcium in utero or lactation exposure does not occur 144.
channel blocker nifedipine. These drugs are considered
safe in the management of both pre-existing and gesta- Azathioprine. Azathioprine is a precursor to immuno­
tional hypertension in pregnancy. Although the thera- suppressive antagonists of purine metabolism. The drug
peutic effect of different classes of antihypertensives is undergoes degradation to 6‑mercaptopurine before
known to vary with ethnicity in nonpregnant patients, enzymatic conversion to active thioguanine nucleo-
no equivalent robust data are currently available to tides by a number of enzymes, including thio­purine
guide drug selection in pregnancy; therefore, the choice S-methyltransferase (TPMT). The fetus is likely to be
of drug is determined by its availability, licensing, local protected to some extent from the clinical effects of
protocols as well as the experience and preference of azathioprine and 6‑mercaptopurine as the immature
the physician. fetal liver is not thought to express inosine triphosphate
Angiotensin-converting enzyme inhibitors and pyrophosphatase, which is another of the enzymes
angiotensin receptor blockers are able to cross the pla- involved in conversion to active metabolites, although
centa. These classes of drugs are fetotoxic in the second further studies of the metabolic pathways in mother,
and third trimesters — causing oligohydramnios and placenta and fetus are needed. Outcome s­ tudies in
neonatal renal failure — but not in the first trimester, women receiving thiopurines for renal disease, inflam-
when population data are corrected for confounding matory bowel disease145 and connective tissue disease
variables, including maternal demography, and comor- do not demonstrate an increased risk of preterm deliv-
bidities such as diabetes and hypertension131,132. Thus, ery, congenital malformations or childhood neoplasia
RAAS blockers can be continued for nephroprotection when data are corrected for underlying maternal dis-
Supratherapeutic dosing
Administering a drug dose that
during the period of time taken to conceive, provided ease. Azathioprine is therefore considered safe to use
is higher than that needed to they are discontinued at an early diagnosis of preg- throughout pregnancy 146–148. If azathioprine treatment is
achieve therapeutic effects. nancy. Pregnancy testing is recommended according started during pregnancy, it is advisable to check TPMT

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 3 | Considerations for drugs commonly used in nephrology during conception, pregnancy and lactation
Drug Conception Pregnancy Lactation Refs
Overall Maternal Fetal considerations
considerations
Antihypertensive drugs
Labetalol Safe Safe Licenced for pregnancy. No association with Safe (also 133,166
Consider other congenital abnormalities metoprolol and
comorbidities (for atenolol)
example, asthma)
Nifedipine Safe Safe None No association with Safe 133,166
congenital abnormalities
Methyldopa Safe Safe None No association with Avoid owing to 133,166
congenital abnormalities risk of postnatal
depression
ACE inhibitors No apparent Unsafe None Fetotoxic in second Safety data 131–134,
increase in risk and third trimesters, available for 166
with first-trimester leading to fetal and captopril and
use when correct neonatal renal failure, enalapril
for underlying bone and aortic
hypertension. arch malformations,
Continue until oligohydramnios and
conception if required pulmonary hypoplasia
for nephroprotection
Angiotensin Insufficient data Unsafe None Fetotoxic in second No data 133,166
receptor blockers on exposure in and third trimesters,
early pregnancy. leading to fetal and
Discontinue in neonatal renal failure,
advance of pregnancy bone and aortic
arch malformations,
oligohydramnios and
pulmonary hypoplasia
Immunosuppressant drugs
Corticosteroids Safe Safe Potential risks: Fetus exposed to <10% Safe. Small amounts 133,
diabetes; hypertension; maternal dose owing to in breast milk. 135–144
pre-eclampsia; placental deactivation. Consider timing
infection; preterm No evidence of increased feeds to 4 hours
rupture of membranes; congenital abnormalities after administration
aim for minimum if high dose given
maintenance dose (for example,
methylprednisolone
induction) and
monitor infant
Azathioprine Safe Safe Recommend checking Placental transfer. Safe. Low 133,
TPMT status before No association with concentration in 145–149
dosing congenital abnormalities breast milk
Mycophenolate Unsafe. Effective Unsafe None Placental transfer. Excreted in breast 133,150
contraception during Teratogenic, causing milk. Avoid use
treatment and for ear, heart, eye, lip during lactation
minimum 6 weeks and/or palate, kidney
after treatment. and bone abnormalities,
Ensure disease tracheoesophageal
stability prior to fistula and congenital
conception diaphragmatic hernia.
Increased rate of
miscarriage
Ciclosporin Safe Safe Monitor pre-dose levels. Placental transfer. Safe 133,152,
May need higher dose in No association with 154
pregnancy congenital abnormalities.
Increased risk of
gestational diabetes
Tacrolimus Safe Safe Monitor pre-dose levels. Placental transfer. Safe 133,
May need a higher dose No association with 153–155
in pregnancy congenital abnormalities.
Increased risk of
gestational diabetes

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 3 (cont.) | Considerations for drugs commonly used in nephrology during conception, pregnancy and lactation
Drug Conception Pregnancy Lactation Refs
Overall Maternal Fetal considerations
considerations
Immunosuppressant drugs (cont.)
Hydroxychloroquine Safe Safe Withdrawal may Placental transfer. No Safe 73,98,133,
precipitate lupus flare. increase in miscarriage or 157–160
Indicated throughout congenital abnormality.
pregnancy if patient May reduce risk of CHB
has a history of lupus with anti-SSA+ and/or
nephritis anti-SSB+
Cyclophosphamide Unsafe. Effective Unsafe None Placental transfer. Excreted in breast 133,161
contraception during Teratogenic. Congenital milk. Discontinue
and for 3 months abnormalities of the skull, breastfeeding
after treatment. ear, face, limb and visceral during and for
Dose-related and organs. Increased risk of 36 hours after
age-related risk of miscarriage treatment
infertility
Rituximab Unclear (limited data Unclear If indicated for severe Active placental transfer Unclear (limited 133,162,
available). Treatment (limited disease, aim to give during second and third data available). 163
decision depends data dose before, or in early, trimester. Potential Possible excretion of
on indication and available) pregnancy to minimize risk of neonatal B cell trace amounts, but
alternative options the risk of neonatal depletion. Avoid unless neonatal absorption
B cell depletion potential benefit to unlikely
mother outweighs
risk. Long-term effects
unknown
Eculizumab Unclear (limited data Unclear Morbidity of underlying Active placental transfer Unclear (limited 133,164,
available). Treatment (limited condition may mean in second and third data available). 165
decision depends data treatment required in trimester. No congenital Possible excretion of
on indication and available) pregnancy. Monitor abnormality reported trace amounts, but
alternative options for increased dosage in 20 infants. Long-term neonatal absorption
requirements effects unknown unlikely
Other drugs
Aspirin (75–150 mg) Safe Safe Decreases risk of No association with Safe 133,
pre-eclampsia. No congenital abnormalities 166–170
evidence of maternal
haemorrhagic
complications
Low-molecular- Safe Safe Used for VTE No placental transfer Safe 95,133
weight heparin prophylaxis. Threshold
for use in proteinuria is
unknown
Allopurinol Unclear (limited data Unclear None Placental transfer. No Therapeutic levels in 133,173
available) (limited increase in congenital breastfed infants
data abnormality in 31
available) infants. Cautionary
use in first trimester
recommended
Colchicine Safe Safe None Associated with Infants receive an 133,174
preterm delivery and estimated 10% of
lower birthweight, maternal dose with
but contribution of no reported adverse
underlying disease is effects
unclear
Iron Safe Safe Intravenous None Safe 114,118,
preparations may offer 133
better bioavailability
in CKD
Erythropoietin Safe Safe Increased gestational No placental transfer Safe 109‑111,
requirement. Monitor 133
blood pressure
ACE, angiotensin-converting enzyme; CHB, congenital heart block; CKD, chronic kidney disease; SSA, Sjogren syndrome type A antigen; SSB, Sjogren syndrome
type B antigen; TPMT, thiopurine methyltransferase; VTE, venous thromboembolism.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

levels as a dose reduction is required for those with an has been used in women with SLE during pregnancy
inherited deficiency of TPMT to prevent maternal for over 3 decades with reassuring safety data, and a
toxicity. As for nonpregnant patients with CKD, mon- meta-analysis reported no increase in the risk of con-
itoring of leukocyte count and serum transaminases is genital malformation, preterm delivery, still birth or
recommended because of possible adverse effects of low-birthweight infants158. Of note, the association of
myelotoxicity and liver injury. Monitoring is required hydroxychloroquine with a higher spontaneous abor-
fortnightly at drug initiation, reducing to once per tri- tion rate may be confounded by the earlier presentation
mester when a stable dose is achieved. Azathioprine is of women taking hydroxychloroquine to health services,
compatible with breastfeeding 146,149. which improves the documentation of early pregnancy
loss compared with controls. This association also lacks
Mycophenolate mofetil. Mycophenolate mofetil biological plausibility, as hydroxychloroquine has been
(MMF) is used in nonpregnant patients for the treat- associated with evidence of improved placentation.
ment of lupus nephritis and for ongoing immuno­ Pregnant women with SLE who stop hydroxychloro-
suppression following renal transplantation. However, quine treatment have a higher requirement for antenatal
the compound is teratogenic, causing a typical clinical corticosteroids to suppress disease activity than those
syndrome that includes hypoplastic nails, shortened fin- who continue treatment 159. In addition, hydroxychloro-
gers, diaphragmatic hernia, microtia, micrognathia, cleft lip quine use is linked to reductions in both growth restric-
and palate and congenital heart defects150,151. The risk tion and the recurrence of CHB73,98. Counselling and
of these outcomes is estimated to be 20–25%, but this screening for hydroxychloroquine-related retinopathy
approximation may be influenced by reporting bias. In is recommended regardless of pregnancy 160. Although
addition, the relative risks of withdrawal during organo- hydroxychloroquine is excreted into breast milk, meas-
genesis in the first trimester, followed by continued use ured levels are 0.25–2% of the maternal dose, with no
in the second and third trimester, are difficult to quan- adverse effects reported133.
tify. Women receiving MMF are therefore advised to
switch to a nonteratogenic immunosuppressant agent Cyclophosphamide. Cyclophosphamide, which is used
(such as azathioprine or a calcineurin inhibitor) at in the management of rapidly progressive glomerulo-
least 3 months before conception. For the rare cases in nephritis, is a potent immunosuppressant that causes
which MMF is the only drug that can achieve disease or lymphocyte cytotoxicity via inhibition of DNA replica-
transplant stability, very careful counselling regarding tion. The drug is teratogenic and fetotoxic, causing skull
the risks of continuing MMF treatment during preg- abnormalities, ear and craniofacial malformations, limb
nancy is required. There is an absence of data regarding and visceral organ abnormalities and growth restriction.
MMF and lactation, and treatment is not recommended In addition, suppression of haematopoiesis and neuro-
­during breastfeeding. logical impairment occurs with exposure after the first
trimester 161. Cyclophosphamide is excreted into breast
Calcineurin inhibitors. Calcineurin inhibitors (such as milk, with cytopenia reported in exposed infants. Thus,
ciclosporin and tacrolimus) target signalling pathways the use of cyclophosphamide is contraindicated in
that regulate T cell activation. Although neither ciclo- pregnancy and lactation. Although pregnancy should
sporin nor tacrolimus are teratogenic152,153, both drugs be avoided in the context of progressive renal disease
are diabetogenic154; therefore, screening for gestational and active vasculitis, plasma exchange and rituximab
diabetes mellitus should be undertaken at 28 weeks of (see below) can be considered as alternative treatment
gestation or earlier if other risk factors are present. options in the context of unplanned ­pregnancy or for a
Owing to pregnancy-related changes in protein bind- new diagnosis made during pregnancy.
ing, the volume of distribution, and drug clearance, ther-
apeutic drug-level monitoring of calcineurin inhibitors Rituximab. Rituximab, which is a monoclonal antibody
is essential, and higher doses may be needed. According that is increasingly used to treat primary and second-
to expert consensus, however, the lower end of the tac- ary glomerular diseases, opsonizes and destroys B cells
rolimus pre-pregnancy dosing range is recommended in by binding to the CD20 antigen. Despite its half-life of
pregnant patients because of the possibility of high free
~22 days, B cell depletion occurs for up to 6 months162;
drug levels during pregnancy, which are not detectable hence, rituximab administration, even before preg-
Diaphragmatic hernia
Congenital defect in the
with the use of standard assays155. nancy, may lead to intrauterine exposure. However,
diaphragm that allows The use of tacrolimus is considered safe during continued rituximab administration during pregnancy
movement of abdominal breastfeeding. Babies of mothers taking tacrolimus are may be n ­ ecessary for disease remission.
viscera into the chest. born with therapeutic drug levels, but these fall progres- Rituximab freely crosses the placenta, but data
sively in both breastfed and bottle-fed infants156. Fewer
on the effects of fetal exposure are limited: one study
Microtia
A congenital abnormality in lactation data for ciclosporin exist, but no adverse effects
reported B cell depletion in 11 of 90 exposed infants but
which the pinna (external ear) have been reported. no increase in congenital malformations163. Neonatal
is underdeveloped. effects are minimized with use in the first trimester,
Hydroxychloroquine. Hydroxychloroquine has diverse when placental transfer is passive, rather than during
Micrognathia
A congenital abnormality
immunomodulatory effects, including a reduction the second and third trimesters, when active placen-
in which the jaw is in inflammatory cytokines, inhibition of antigen-­ tal transfer of antibodies occurs. Neonatal B cell count
underdeveloped. presenting cells and antioxidant activity 157. The drug and rituximab levels can be checked after delivery

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

if indicated (for example, in neonatal sepsis). Delay growth restriction in pregnant women with CKD60.
of neonatal vaccination with live vaccines (such as Low-dose aspirin can be continued during breastfeed-
Bacillus Calmette-Guérin (BCG), rotavirus or varicella ing if indicated, as transfer into milk is undetectable
zoster virus) is advisable until the neonate is older than (with levels of the metabolite salicylic acid probably
6 months following in utero exposure to rituximab. being subclinical to infants170).
Long-term infant outcomes following in utero exposure
to rituximab remain unknown. Cinacalcet. Cinacalcet, which is used in CKD for the
Limited data indicate that rituximab is transported treatment of refractory secondary hyperparathy-
into breast milk in trace concentrations, although roidism, is a calcimimetic that increases the sensitivity
absorption is considered unlikely due to its destruction of the calcium-­sensing receptor to extracellular calcium,
in the gastrointestinal tract of the infant 133. Therefore, thereby inhibiting secretion of parathyroid hormone.
­rituximab is not a contraindication to lactation. The few available studies that have described the use
of cinacalcet in pregnancy 171,172 have reported success-
Eculizumab. Eculizumab is a monoclonal antibody that ful outcomes. However, in the absence of sufficient
targets active complement protein C5. The antibody is safety data, current practice is to stop c­ inacalcet before
used for the treatment of paroxysmal nocturnal haemo­ ­pregnancy and during breastfeeding.
globinuria as well as the induction and maintenance
treatment of atypical haemolytic uraemic syndrome Allopurinol. Allopurinol crosses the placenta, and its
(aHUS), which can be triggered or relapse during preg- use in the first trimester is cautioned, although mal-
nancy. Although published data are limited, eculizumab formation rates were not increased in a cohort of 31
use in pregnancy may be necessary to reduce morbid- women173. A therapeutic allopurinol dose is received by
ity, including ESRD associated with aHUS164. In 75 breastfed infants, and although no adverse effects have
pregnancies from 61 women with paroxysmal noc- been reported, infants should be monitored for adverse
turnal haemoglobinuria, no increased risk of congen- effects such as rash and gastrointestinal symptoms133.
ital abnormality was reported with use of eculizumab,
although the antibody was detected in seven of 20 cord Colchicine. Colchicine is an anti-inflammatory agent
blood samples165. Long-term effects of in utero exposure used in the management of rheumatological disease and
to eculizumab remain unknown. As pregnancy affects gout. A meta-analysis of 550 pregnancies exposed to col-
the pharmacokinetics of this agent, an increase in dos- chicine doses of 1–2 mg per day showed no association
age or frequency of treatment may be required165. No with miscarriage or malformation. Although there was
antibody was detected in the breast milk of ten patients a measured association between colchicine and reduc-
with p­ aroxysmal nocturnal haemoglobinuria165. tions in gestational age and birthweight in a small num-
ber of heterogeneous studies, the clinical significance
Other drugs of these data is unclear, with possible confounding due
Aspirin. Aspirin is an antiplatelet agent used in preg- to the underlying condition174. An estimated 10% of
nancy from 12 weeks of gestation to reduce the risk the dose is received from breast milk, with no adverse
of pre-eclampsia in high-risk women 166. A meta-­ effects reported133.
analysis shows that low-dose aspirin (60–150 mg)
confers a reduction in pre-eclampsia (RR 0.76; 95% CI Dialysis in pregnancy
0.62–0.95), preterm birth (RR 0.86; 95% CI 0.76–0.98) Haemodialysis. A dramatic increase in the number of
and intrauterine growth restriction (RR 0.8; 95% CI pregnancies has been reported in women requiring renal
0.65–0.99) as well as an absence of evidence of harm167. replacement therapy. Systematic review data reported
An RCT in women diagnosed as being at high risk of 90 cases between 2000 and 2008, which increased to
pre-eclampsia on the basis of a maternal physiological 574 cases only 6 years later 175. Augmentation of hae-
and biomarker profile performed at 11–13 weeks of modialysis dose is associated with fewer fetal deaths176,
gestation showed a 62% reduction (OR 0.38; 95% CI and a meta-analysis shows a continuous inverse corre-
0.20–0.74) of preterm pre-eclampsia in women taking lation between the number of haemodialysis hours per
150 mg per day aspirin compared with those taking pla- week and rates of preterm birth (P = 0.044; r2 = 0.22) and
cebo168. Interestingly, a similar risk reduction was not small-for-gestational-­age infants (P = 0.017; r2 = 0.54)175.
seen in this study for women with chronic hypertension In a contemporary cohort of 17 women including 22
(OR 1.30; 95% CI 0.33–5.12)169, despite the hypothesis pregnancies, intensive haemodialysis (defined as an
that pre-eclampsia risk, and therefore bene­f it from average of 43 hours per week) conferred an 86% live
aspirin prophylaxis, would be proportionally greater birth rate at a mean gestational age of 36 weeks com-
than in normotensive women. However, this finding is pared with a live birth rate of 61% at a mean gestational
limited by the possibility of an underpowered subgroup age of 27 weeks in 70 women receiving haemodialysis
analysis and the difficulty in making a diagnosis of for a mean of 17 hours per week177. Provision of dialysis
superimposed pre-eclampsia in the absence of standard for 48 hours per week optimizes maternal physiology,
blood pressure parameters. Given the limitations of this with patients achieving gestational blood pressures of
single study, all women with CKD should be offered 112/72 to 122/81 mmHg with minimal use of antihy-
low-dose aspirin in pregnancy owing to the increased pertensive agents, and predialysis urea concentrations
risks of pre-eclampsia, preterm delivery and fetal of 8–14 mmol/l, although increased dialysate provision

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

of potassium, calcium and phosphate may be required24. decision to undergo pregnancy should be weighed
Nonetheless, complication rates remain high even in carefully with options for transplantation within the
women receiving intensive haemodialysis, and >75% of window of childbearing age. Maternal indications for
such women require preterm delivery 175,177. Pregnancy initiating haemodialysis in pregnancy mirror indica-
on haemodialysis remains a high-risk option, and the tions outside of pregnancy and include fluid overload,
hyperkalaemia and acidosis that are resistant to medi-
cal management. However, the most common trigger
Box 1 | Reproductive health issues for women with CKD for dialysis initiation in pregnancy is the fetotoxicity
of urea, although the maternal serum urea threshold
Pre-pregnancy at which dialysis should be commenced in pregnancy
Fertility continues to be a subject of ongoing debate. Historically,
• Reduced in chronic kidney disease (CKD), but pregnancy can occur at all CKD stages. the threshold of urea for dialysis initiation in pregnancy
• Recommend pursuit of fertility preservation if cyclophosphamide is administered. was set at a concentration of 17–20 mmol/l owing to
Contraception the correlation between higher urea levels with deliv-
• Risk of adverse outcomes to unplanned pregnancy at all CKD stages. ery <32 weeks and birthweight <1,500 g, both of which
• Progesterone-only methods (including the ‘mini pill’, intrauterine devices and are recognized to adversely affect neonatal prognosis178.
subdermal implants) are safe and effective in women with CKD. However, given the positive correlation between hours
• By contrast, barrier methods have high failure rates with typical use. of haemo­dialy­sis provision and pregnancy outcomes,
the emerging consensus is that dialysis initiation should
Conception
be considered at lower levels of urea (15 mmol/l), par-
• Pre-pregnancy counselling before conception is strongly recommended. ticularly for women with progressive renal dysfunc-
• Remove or substitute teratogens before conception. tion, with dialysis provision matched to residual
• Consider risks and/or benefits to remaining on angiotensin-converting enzyme renal function23.
inhibitors until conception.
• Ensure disease and/or transplant stability before conception. Peritoneal dialysis. The incidence of pregnancy in
Pregnancy women on peritoneal dialysis is lower than for haemo-
Risk management dialysis175, which has been attributed to factors such as
• Incremental risk of adverse outcomes with increasing CKD severity. high dialysate osmolality and fallopian tube injury sec-
• Increased obstetric surveillance (including growth scans and Doppler ultrasound) ondary to peritoneal infection. Systematic review data
required. reveal a higher rate of small-for-gestational-age infants
• Venous thromboembolism prophylaxis for women with substantial proteinuria, lupus in women undergoing peritoneal dialysis (67%) than in
flare and antiphospholipid antibodies. those undergoing haemodialysis (31%), which could be
• Increased dialysis provision improves outcomes in stage 5 CKD. due to placental development being adversely affected
• Gestational diabetes mellitus screening in women receiving steroids and/or
by peritoneal dialysis175. However, possible limitations
calcineurin inhibitors (CNIs). of cohort size and publication bias should be taken
into account.
• Therapeutic drug monitoring for women taking CNIs.
Hypertension Biomarkers in CKD and pre-eclampsia
• Labetalol, nifedipine and methyldopa are safe to use during pregnancy. In the past 5 years, prospective research has sought
• No increase in adverse fetal outcome when treated to a diastolic blood pressure to improve the diagnosis of superimposed pre-­
of 85 mmHg. eclampsia in women with CKD. Diagnostic criteria
• Treatment target of <140/90 mmHg (according to expert consensus). for pre-­e clampsia in the absence of CKD are based
Pre-eclampsia on the development of de novo hypertension and pro-
• Low-dose aspirin (75–150 mg) prophylaxis recommended for all pregnant women teinuria after 20 weeks of gestation. Hence, the pres-
with CKD. ence of these signs before pregnancy in women with
• Diagnosis of pre-eclampsia is complicated by pre-existing hypertension and CKD complicates the diagnosis of superimposed pre-­
proteinuria. eclampsia. Moreover, hypertension and proteinuria can
Anaemia also occur de novo in later pregnancy in the absence
of pre-eclampsia owing to gestation-related physio-
• Erythropoietin and iron supplementation may be required.
logical changes. As a result, the distinction between
Post-partum physiological gestational change, CKD, lupus disease
Lactation flare and superimposed pre-eclampsia is challenging
• Captopril and enalapril are safe to use in breastfeeding. (TABLE 2), leading to difficult decisions with regard to
• Neonatal tacrolimus clearance in breastfed infants is comparable to that of delivery and iatrogenic prematurity. Superimposed
bottle-fed infants. pre-eclampsia contributes to preterm delivery and
• Up-to-date drug safety data in lactation are available103. small-for-gestational-age infants among patients with
Drugs CKD. Heightened obstetric surveillance is therefore
• Review CNI dose if dose was increased during pregnancy. warranted for all women with CKD to enable early
detection of impaired fetal growth and to assess the
Renal function
umbilical artery through the use of Doppler ultrasound,
• Risk of loss of residual renal function during pregnancy and post-partum.
a  technique by which impending fetal distress is

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Doppler ultrasound predicted through detection of progressive changes in Conclusions


The use of sound waves to waveform. In addition, placental insufficiency due to Fertility, family planning and pregnancy considerations
detect movement. This pre-eclampsia can be predicted at 20 weeks gestation are of increasing importance to both nephrologists and
technique is used in pregnancy through assessment of uterine artery vascular resist- obstetricians owing to trends in women’s health and
to examine the vascular
waveform in uterine and
ance. One prospective cohort comprising 15 women renal disease. CKD in women of childbearing age con-
umbilical arteries to predict or showed that elevated uterine artery Doppler pres- fers a risk of reduced fertility and adverse pregnancy out-
diagnose pathology. sure index has 100% specificity for the prediction of comes. Safe and effective contraceptive methods should
superimposed pre-eclampsia in women with CKD78, be available for all women of reproductive age with CKD
but this finding requires further validation in a larger to allow pregnancy to be planned, with individualized and
cohort including women with advanced CKD. Another detailed risk counselling undertaken before conception
study demonstrated that uterine and umbilical artery (BOX 1). Anaemia, vitamin D deficiency and hypertension
Doppler ultrasound could be used to differentiate pre-­ are complicating comorbidities in pregnancy that may
eclampsia from CKD in the absence of pre-­eclampsia, develop as a consequence of underlying CKD and require
with normal uterine and umbilical waveforms pre- tailored surveillance and management. The prescription
dicting CKD, whereas abnormal waveforms were of drugs in pregnancy is always a balance of risk versus
­associated with pre-eclampsia179. benefit. For many drugs used in CKD, the benefit of ther-
In terms of biomarkers, an increase in the antiangi- apy during pregnancy is greater than any associated risk,
ogenic marker sFLT1 and a decrease in the angiogenic provided known teratogens and fetotoxic drugs (RAAS
marker placenta growth factor (PLGF; also known as blockers, MMF and cyclophosphamide) are avoided. For
PGF) have been demonstrated to be useful tools for the women requiring dialysis in pregnancy, increasing the
prediction of pre-eclampsia180 and the need for delivery number of hours of haemodialysis ­provision is associated
within 2 weeks181. A similar pattern of biomarkers has with better outcomes.
been reported to discriminate between women with Future research is required into the mechanisms and
CKD with and without superimposed pre-eclampsia78, management of impaired fertility in CKD, preferential
including women with lupus nephritis182 and women on choices and optimal dosage of therapeutic agents for
dialysis183. However, larger cohort studies are required the management of hypertension, and the pathophysiol-
to confirm the diagnostic utility of these markers in ogy and prevention of progressive renal deterioration in
pregnant women with CKD. Research into the thera- pregnancy. Furthermore, proteinuric thresholds for VTE
peutic utility of removing antiangiogenic factors such as prophylaxis as well as vasoactive biomarker and Doppler
sFLT1 from the circulation in pre-eclampsia currently validation in the prediction and diagnosis of s­ uperimposed
excludes women with underlying CKD184. pre-eclampsia warrant further investigation.

1. Piccoli, G. B. et al. Pregnancy and chronic kidney in patients with chronic kidney disease. Nat. Clin. 22. van Eps, C. et al. Changes in serum prolactin, sex
disease: a challenge in all CKD stages. Clin. J. Am. Soc. Pract. Nephrol. 3, 200–207 (2007). hormones and thyroid function with alternate nightly
Nephrol. 5, 844–855 (2010). 11. Steele, T. E. et al. Sexual experience of the chronic nocturnal home haemodialysis. Nephrology 17,
2. Bramham, K. et al. Pregnancy in renal transplant peritoneal dialysis patient. J. Am. Soc. Nephrol. 7, 42–47 (2012).
recipients: a UK national cohort study. Clin. J. Am. 1165–1168 (1996). 23. Hladunewich, M. & Schatell, D. Intensive dialysis
Soc. Nephrol. 8, 290–298 (2013). 12. Solak, Y. et al. Effects of sildenafil and vardenafil and pregnancy. Hemodial. Int. 20, 339–348
3. Piccoli, G. B. et al. Outcomes of pregnancies after treatments on sleep quality and depression in (2016).
kidney transplantation: lessons learned from CKD. hemodialysis patients with erectile dysfunction. 24. Barua, M. et al. Successful pregnancies on nocturnal
A comparison of transplanted, nontransplanted Int.J. Impot. Res. 23, 27–31 (2011). home hemodialysis. Clin. J. Am. Soc. Nephrol. 3,
chronic kidney disease patients and low-risk 13. Holley, J. L., Schmidt, R. J., Bender, F. H., Dumler, F. 392–396 (2008).
pregnancies: a multicenter nationwide analysis. & Schiff, M. Gynecologic and reproductive issues in 25. Meistrich, M. L., Wilson, G., Brown, B. W.,
Transplantation 101, 2536–2544 (2017). women on dialysis. Am. J. Kidney Dis. 29, 685–690 da Cunha, M. F. & Lipshultz, L. I. Impact of
4. Webster, P., Lightstone, L., McKay, D. B. & (1997). cyclophosphamide on long-term reduction in sperm
Josephson, M. A. Pregnancy in chronic kidney disease 14. Lim, V. S., Henriquez, C., Sievertsen, G. & count in men treated with combination chemotherapy
and kidney transplantation. Kidney Int. 91, Frohman, L. A. Ovarian function in chronic renal for ewing and soft tissue sarcomas. Cancer 70,
1047–1056 (2017). failure: Evidence suggesting hypothalamic anovulation. 2703–2712 (1992).
5. Hladunewich, M. A., Melamad, N. & Bramham, K. Ann. Intern. Med. 93, 21–27 (1980). 26. Boumpas, D. T. et al. Risk for sustained amenorrhea in
Pregnancy across the spectrum of chronic kidney 15. Wang, G. C. et al. Measurements of serum pituitary- patients with systemic lupus erythematosus receiving
disease. Kidney Int. 89, 995–1007 (2016). gonadal hormones and investigation of sexual and intermittent pulse cyclophosphamide therapy.
6. Knight, M., Kurinczuk, J. J., Tuffnell, D. & reproductive functions in kidney transplant recipients. Ann. Intern. Med. 119, 366–369 (1993).
Brocklehurst, P. The UK Obstetric Surveillance System Int. J. Nephrol. 2010, 612126 (2010). 27. Ioannidis, J. P., Katsifis, G. E., Tzioufas, A. G. &
for rare disorders of pregnancy. BJOG 112, 263–265 16. Palmer, B. F. & Clegg, D. J. Gonadal dysfunction in Moutsopoulos, H. M. Predictors of sustained
(2005). chronic kidney disease. Rev. Endocr. Metab. Disord. amenorrhea from pulsed intravenous
7. [No authors listed.] Statistical bulletin: conceptions 18, 17–30 (2017). cyclophosphamide in premenopausal women with
in England and Wales 2015. Office for National 17. Holley, J. L. & Schmidt, R. J. Changes in fertility and systemic lupus erythematosus. J. Rheumatol. 29,
Statistics https://www.ons.gov.uk/ hormone replacement therapy in kidney disease. 2129–2135 (2002).
peoplepopulationandcommunity/ Adv. Chron. Kidney Dis. 20, 240–245 (2013). 28. Bellver, J. & Pellicer, A. Ovarian stimulation for
birthsdeathsandmarriages/conceptionandfertilityrates/ 18. Yavuz, D., Topçu, G., Ozene, r C., Akalin, S. & Sirikçi, O. ovulation induction and in vitro fertilization in
bulletins/conceptionstatistics/2015#main-points Macroprolactin does not contribute to elevated levels patients with systemic lupus erythematosus and
(2017). of prolactin in patients on renal replacement therapy. antiphospholipid syndrome. Fertil. Steril. 92,
8. Piccoli, G. B. et al. The children of dialysis: live-born Clin. Endocrinol. 63, 520–524 (2005). 1803–1810 (2009).
babies from on‑dialysis mothers in Italy — an 19. Weinhandl, E. D., Liu, J., Gilbertson, D. T., 29. Elizur, S. E. et al. Fertility preservation treatment for
epidemiological perspective comparing dialysis, Arneson, T. J. & Collins, A. J. Survival in daily home young women with autoimmune diseases facing
kidney transplantation and the overall population. hemodialysis and matched thrice-weekly in‑center treatment with gonadotoxic agents. Rheumatology
Nephrol. Dial. Transplant. 29, 1578–1586 hemodialysis patients. J. Am. Soc. Nephrol. 23, 47, 506–1509 (2008).
(2014). 895–904 (2012). 30. Somers, E. C., Marder, W., Christman, G. M.,
9. Basok, E. K. et al. Assessment of female sexual 20. Rocco, M. V. et al. The effects of frequent nocturnal Ognenovski, V. & McCune, W. J. Use of a
function and quality of life in predialysis, peritoneal home hemodialysis: the frequent hemodialysis network gonadotropin-releasing hormone analog for
dialysis, hemodialysis, and renal transplant patients. nocturnal trial. Kidney Int. 80, 1080–1091 (2011). protection against premature ovarian failure
Int. Urol. Nephrol. 41, 473–481 (2009). 21. Finkelstein, F. O. et al. At‑home short daily during cyclophosphamide therapy in women with
10. Finkelstein, F. O., Shirani, S., Wuerth, D. & hemodialysis improves the long-term health-related severe lupus. Arthritis Rheum. 52, 2761–2767
Finkelstein, S. H. Therapy insight: sexual dysfunction quality of life. Kidney Int. 82, 561–569 (2012). (2005).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 17


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

31. Moore, H. C. et al. Goserelin for ovarian protection 55. Kristensen, K. et al. Temporal changes of the plasma 76. Jones, D. C. & Hayslett, J. P. Outcome of pregnancy in
during breast-cancer adjuvant chemotherapy. levels of cystatin C, beta-trace protein, women with moderate or severe renal insufficiency.
N. Engl. J. Med. 372, 923–932 (2015). beta2‑microglobulin, urate and creatinine during N. Engl. J. Med. 335, 226–232 (1996).
32. Lambertini, M. et al. Ovarian suppression with pregnancy indicate continuous alterations in the renal 77. Imbasciati, E. et al. Pregnancy in CKD stages 3 to 5:
triptorelin during adjuvant breast cancer filtration process. Scand. J. Clin. Lab Invest. 67, fetal and maternal outcomes. Am. J. Kidney Diseases.
chemotherapy and long-term ovarian function, 612–618 (2007). 49, 753–762 (2007).
pregnancies, and disease-free survival: a randomized 56. Kristensen, K., Strevens, H., Lindström, V. & Grubb, A. 78. Bramham, K. et al. Diagnostic and predictive
clinical trial. JAMA 314, 2632–2640 (2015). Wide-Swensson, D. Increased plasma levels of biomarkers for pre-eclampsia in patients with
33. Lambertini, M. et al. Ovarian suppression using beta2‑microglobulin, cystatin C and beta-trace protein established hypertension and chronic kidney disease.
luteinizing hormone-releasing hormone agonists in term pregnancy are not due to utero-placental Kidney Int. 89, 874–885 (2016).
during chemotherapy to preserve ovarian function and production. Scand. J. Clin. Lab Invest. 68, 649–653 79. Willey, C. J. et al. Prevalence of autosomal dominant
fertility of breast cancer patients: a meta-analysis of (2008). polycystic kidney disease in the European Union.
randomized studies. Ann. Oncol. 26, 2408–2419 57. Strevens, H., Wide-Swensson, D., Torffvit, O. & Nephrol. Dial. Transplant. 32, 1356–1363 (2017).
(2015). Grubb, A. Serum cystatin C for assessment of 80. Wu, M. et al. Pregnancy outcomes in autosomal
34. Shah, P. S. et al. Intention to become pregnant and glomerular filtration rate in pregnant and non- dominant polycystic kidney disease: a case-control
low birth weight and preterm birth: a systematic pregnant women. Indications of altered filtration study. J. Matern. Fetal Neonatal Med. 29, 807–812
review. Maternal Child Health J. 15, 205–216 (2009). process in pregnancy. Scand. J. Clin. Lab Invest. 62, (2016).
35. Yildirim, Y. & Uslu, A. Pregnancy in patients with 141–147 (2002). 81. Jung, J. H. et al. Successful pregnancy in a patient with
previous successful renal transplantation. 58. Bramham, K., Makanjuola, D., Hussein, W., Cafful, D. autosomal dominant polycystic kidney disease on long-
Int. J. Gynaecol. Obstet. 90, 198–202 (2005). & Shehata, H. Serum cystatin is not a marker of term hemodialysis. J. Kor. Med. Sci. 29, 301–304
36. Trussell, J. Contraceptive failure in the United States. glomerular filtration rate in pregnancy. Obstet. Med. (2014).
Contraception 83, 397–404 (2011). 2, 121–122 (2009). 82. Swift, O., Vilar, E., Rahman, B., Side, L. & Gale, D. P.
37. Korver, T. et al. Maintenance of ovulation inhibition 59. Larsson, A., Palm, M., Hansson, L. O. & Axelsson, O. Attitudes in patients with autosomal dominant
with the 75‑microg desogestrel-only contraceptive Cystatin C and modification of diet in renal disease polycystic kidney disease toward prenatal diagnosis
pill (Cerazette) after scheduled 12‑h delays in tablet (MDRD) estimated glomerular filtration rate differ and preimplantation genetic diagnosis. Genet. Test.
intake. Contraception 71, 8–13 (2005). during normal pregnancy. Acta Obstet. Gynecol. Mol. Biomarkers 20, 741–746 (2016).
38. Estes, C. M. & Westhoff, C. Contraception for the Scand. 89, 939–944 (2010). 83. Blom, K., Odutayo, A., Bramham, K. &
transplant patient. Semin. Perinatol. 31, 372–377 60. Piccoli, G. B. et al. Risk of adverse pregnancy Hladunewich, M. A. Pregnancy and glomerular
(2007). outcomes in women with CKD. J. Am. Soc. Nephrol. disease: a systematic review of the literature with
39. Krajewski, C. M., Geetha, D. & Gomez-Lobo, V. 26, 2011–2022 (2015). management guidelines. Clin. J. Am. Soc. Nephrol. 12,
Contraceptive options for women with a history of 61. Gambaro, G. et al. Increased urinary excretion of 1862–1872 (2017).
solid-organ transplantation. Transplantation 95, glycosaminoglycans in pregnancy and in diabetes 84. Jungers, P. et al. Chronic kidney disease and
1183–1186 (2013). mellitus: a protective factor against nephrolithiasis. pregnancy. Adv. Nephrol. Necker Hosp. 15, 103–141
40. Morrison, C. S. et al. Is the intrauterine device Nephron 50, 62–63 (1988). (1986).
appropriate contraception for HIV‑1 infected women? 62. Butler, E. L., Cox, S. M., Eberts, E. G. & 85. Abe, S., et al. The influence of antecedent renal
BJOG 108, 784–790 (2001). Cunningham, F. G. Symptomatic nephrolithiasis disease on pregnancy. Am. J. Obstet. Gynecol. 153,
41. Ramhendar, T. & Byrne, P. Use of the levonorgestrel- complicating pregnancy. Obstet. Gynecol. 96, 508–514 (1985).
releasing intrauterine system in renal transplant 753–756 (2000). 86. Packham, D., Whitworth, J. A., Fairley, K. F. &
recipients: a retrospective case review. Contraception 63. Williams, K. P. & Galerneau, F. The role of serum uric Kincaid-Smith, P. Histological features of IgA
86, 288–289 (2012). acid as a prognostic indicator of the severity of glomerulonephritis as predictors of pregnancy
42. Brynhildsen, J. Combined hormonal contraceptives: maternal and fetal complications in hypertensive outcome. Clin. Nephrol. 30, 22–26 (1988).
prescribing patterns, compliance, and benefits versus pregnancies. J. Obstet. Gynaecol. Can. 24, 628–632 87. Limardo, M. et al. Pregnancy and progression of IgA
risks. Ther. Adv. Drug Saf. 5, 201–213 (2014). (2002). nephropathy: results of an Italian multicenter study.
43. Davison, J. M. & Dunlop, W. Renal hemodynamics 64. Verdonk, K., Visser, W., Van Den Meiracker, A. H. & Am. J. Kidney Dis. 56, 506–512 (2010).
and tubular function in normal human pregnancy. Danser, A. H. The renin-angiotensin-aldosterone 88. Hoover, P. J. & Costenbader, K. H. Insights into the
Kidney Int. 18, 152–161 (1980). system in pre-eclampsia: the delicate balance between epidemiology and management of lupus nephritis
44. Helal, I., Fick-Brosnahan, G. M., Reed-Gitomer, B. good and bad. Clin. Sci. 126, 537–544 (2014). from the US rheumatologist’s perspective. Kidney Int.
& Schrier, R. W. Glomerular hyperfiltration: 65. Davison, J. M. & Lindheimer, M. D. Volume 90, 487–492 (2016).
definitions, mechanisms and clinical implications. homeostasis and osmoregulation in human pregnancy. 89. Imbasciati, E. et al. Pregnancy in women with
Nat. Rev. Nephrol. 8, 293–300 (2012). Baillieres Clin. Endocrinol. Metab. 3, 451–472 (1989). pre-existing lupus nephritis: predictors of fetal and
45. Roberts, M., Lindheimer, M. D. & Davison, J. M. 66. [No authors listed.] Pregnancy and renal disease. maternal outcome. Nephrol. Dial. Transplant. 24,
Altered glomerular permselectivity to neutral dextrans Lancet 306, 801–802 (1975). 519–525 (2009).
and heteroporous membrane modelling in human 67. Tong, A., Brown, M. A., Winkelmayer, W. C., 90. Bramham, K. et al. Pregnancy outcomes in systemic
pregnancy. Am. J. Physiol. 270, F338–F343 (1996). Craig, J. C. & Jesudason, S. Perspectives on pregnancy lupus erythematosus with and without previous
46. Milne, J. E. C., Lindheimer, M. D. & Davison, J. M. in women with CKD: a semistructured interview study. nephritis. J. Rheumatol. 38, 1906–1913 (2011).
Glomerular heteroporous membrane modeling in third Am. J. Kidney Dis. 66, 951–961 (2015). 91. Smyth, A. et al. A systematic review and meta-
trimester and postpartum before and during amino 68. Tong, A., Jesudason, S., Craig, J. C. & analysis of pregnancy outcomes in patients
acid infusion. Am. J. Physiol. Renal Physiol. 282, Winkelmayer, W. C. Perspectives on pregnancy in with systemic lupus erythematosus and lupus
F170–F175 (2002). women with chronic kidney disease: systematic review nephritis. Clin. J. Am. Soc. Nephrol. 5, 2060–2068
47. Conrad, K. P. & Davison, J. M. The renal circulation in of qualitative studies. Nephrol. Dial. Transplant. 30, (2010).
normal pregnancy and preeclampsia: is there a place 652–661 (2015). 92. Chakravarty, E. F. et al. Factors that predict
for relaxin? Am. J. Physiol. Renal Physiol. 306, 69. Knight, M. et al. Saving Lives, Improving Mothers’ prematurity and preeclampsia in pregnancies that
F1121–F1135 (2014). Care — Surveillance of maternal deaths in the UK are complicated by systemic lupus erythematosus.
48. Novak, J. et al. Relaxin is essential for renal 2012–2014 and lessons learned to inform maternity Am. J. Obstet. Gynecol. 192, 1897–1904 (2005).
vasodilation during pregnancy in conscious rats. care from the UK and Ireland Confidential Enquiries 93. Buyon, J. P. et al. Predictors of pregnancy outcomes in
J. Clin. Invest. 107, 1469–1475 (2001). into Maternal Deaths and Morbidity 2009–2014. patients with lupus: a cohort study. Ann. Intern. Med.
49. Jeyabalan, A. et al. Essential role for vascular (National Perinatal Epidemiology Unit, University of 163, 153–163 (2015).
gelatinase activity in relaxin-induced renal Oxford, 2016). 94. Moroni, G. et al. Maternal outcome in pregnant
vasodilation, hyperfiltration, and reduced myogenic 70. Wiles, K. S. et al. Pre-pregnancy counselling in chronic women with lupus nephritis: a prospective multicenter
reactivity of small arteries. Circ. Res. 93, 1249–1257 kidney disease: a retrospective analysis of nine years’ study. J. Autoimmun. 74, 194–200 (2016).
(2003). experience. BMC Nephrol. 16, 28 (2015). 95. Royal College of Obstetricians and Gynaecologists.
50. Cadnapaphornchai, M. A. et al. Chronic NOS inhibition 71. Kendrick, J. et al. Kidney disease and maternal and Reducing the risk of venous thromboembolism in
reverses systemic vasodilation and glomerular fetal outcomes in pregnancy. Am. J. Kidney Dis. 66, pregnancy and the puerperium. Green Top
hyperfiltration in pregnancy. Am. J. Physiol. Renal 55–59 (2015). Guideline 37a https://www.rcog.org.uk/globalassets/
Physiol. 280, F592–F598 (2001). 72. Zhang, J. J. et al. A systematic review and meta- documents/guidelines/gtg-37a.pdf (2015).
51. Conrad, K. P. & Baker, V. L. Corpus luteal contribution analysis of outcomes of pregnancy in CKD and CKD 96. Cimaz, R., Spence, D. L., Hornberger, L. &
to maternal pregnancy physiology and outcomes in outcomes in pregnancy. Clin. J. Am. Soc. Nephrol. 10, Silverman, E. D. Incidence and spectrum of neonatal
assisted reproductive technologies. Am. J. Physiol. 1964–1978 (2015). lupus erythematosus: a prospective study of infants
Regul. Integr. Comp. Physiol. 304, R69–R72 (2013). 73. Moroni, G. et al. Fetal outcome and recommendations born to mothers with anti‑Ro autoantibodies.
52. Odutayo, A. &, Hladunewich, M. Obstetric of pregnancies in lupus nephritis in the 21st century. J. Pediatr. 142, 678–683 (2003).
nephrology: renal hemodynamic and metabolic A prospective multicenter study. J. Autoimmun. 74, 97. Andreoli, L. et al. EULAR recommendations for
physiology in normal pregnancy. Clin. J. Am. Soc. 6–12 (2016). women’s health and the management of family
Nephrol. 7, 2073–2080 (2012). 74. Piccoli, G. B. et al. Maternal-foetal outcomes in planning, assisted reproduction, pregnancy and
53. Smith, M. C., Moran, P., Ward, M. K. & Davison, J. M. pregnant women with glomerulonephritides. Are all menopause in patients with systemic lupus
Assessment of glomerular filtration rate during glomerulonephritides alike in pregnancy? erythematosus and/or antiphospholipid syndrome.
pregnancy using the MDRD formula. BJOG 115, J. Autoimmun. 79, 91–98 (2017). Ann. Rheum. Dis. 6, 476–485 (2017).
109–112 (2008). 75. Chapman, A. B., Johnson, A. M. & Gabow, P. A. 98. Izmirly, P. M. et al. Maternal use of
54. Alper, A. B. et al. Performance of estimated Pregnancy outcome and its relationship to progression hydroxychloroquine is associated with a reduced risk
glomerular filtration rate prediction equations in of renal failure in autosomal dominant polycystic of recurrent anti-SSA/Ro‑antibody-associated cardiac
preeclamptic patients. Am. J. Perinatol. 28, 425–430 kidney disease. J. Am. Soc. Nephrol. 5, 1178–1185 manifestations of neonatal lupus. Circulation 126,
(2010). (1994). 76–82 (2012).

18 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

99. Klemetti, M. M. et al. Obstetric and perinatal outcome 122. De‑Regil, L. M., Palacios, C., Lombardo, L. K. & and biologic disease modifying anti-rheumatic drugs
in type 1 diabetes patients with diabetic nephropathy Peña-Rosas, J. P. Vitamin D supplementation for and corticosteroids. Rheumatology 55, 1693–1697
during 1988–2011. Diabetologia 58, 678–686 women during pregnancy. Cochrane Database Syst. (2016).
(2015). Rev. 2, CD008873 (2016). 147. Götestam Skorpen, C. et al. The EULAR points to
100. Piccoli, G. B. et al. Type 1 diabetes, diabetic 123. Palacios, C., De‑Regil, L. M., Lombardo, L. K. & consider for use of antirheumatic drugs before
nephropathy, and pregnancy: a systematic review and Peña-Rosas, J. P. Vitamin D supplementation during pregnancy, and during pregnancy and lactation.
meta-study. Rev. Diabet. Stud. 10, 6–26 (2013). pregnancy: updated meta-analysis on maternal Ann. Rheum. Dis. 75, 795–810 (2016).
101. Bell, R., Glinianaia, S. V., Tennant, P. W., Bilous, R. W. outcomes. J. Steroid Biochem. Mol. Biol. 164, 148. Ostensen, M. et al. Pregnancy and reproduction in
& Rankin, J. Peri-conception hyperglycaemia and 148–155 (2016). autoimmune rheumatic diseases. Rheumatology 50,
nephropathy are associated with risk of congenital 124. World Health Organization. Vitamin D 657–664 (2011).
anomaly in women with pre-existing diabetes: a supplementation in pregnancy. http://www.who.int/ 149. Sau, A. et al. Azathioprine and breastfeeding: is it
population-based cohort study. Diabetologia 55, nutrition/publications/micronutrients/guidelines/ safe? BJOG 114, 498–501 (2007).
936–947 (2012). vit_d_supp_pregnant_women/en/ (2012). 150. Sifontis, N. M. et al. Pregnancy outcomes in solid
102. Hod, M. et al. Diabetic nephropathy and pregnancy: 125. Tamblyn, J. A. et al. Dysregulation of maternal and organ transplant recipients with exposure to
the effect of ACE inhibitors prior to pregnancy on placental vitamin D metabolism in preeclampsia. mycophenolate mofetil or sirolimus. Transplantation
fetomaternal outcome. Nephrol. Dialysis Transplant. Placenta 50, 70–77 (2017). 82, 1698–1702 (2006).
10, 2328–2333 (1995). 126. Turner, M., Barré, P. E., Benjamin, A., Goltzman, D. & 151. Perez-Aytes, A. et al. In utero exposure to
103. Bar, J. et al. Pregnancy outcome in patients with Gascon-Barré, M. Does the maternal kidney contribute mycophenolate mofetil: a characteristic phenotype?
insulin dependent diabetes mellitus and diabetic to the increased circulating 1,25‑dihydroxyvitamin D Am. J. Med. Genet. A 146A, 1–7 (2008).
nephropathy treated with ACE inhibitors before concentrations during pregnancy? Miner. Electrolyte 152. Bar Oz, B., Hackman, R., Einarson, T. & Koren, G.
pregnancy. J. Pediatr. Endocrinol. Metab. 12, Metab. 14, 246–252 (1988). Pregnancy outcome after cyclosporine therapy during
659–666 (1999). 127. Hewison, M., Zehnder, D., Chakraverty, R. & pregnancy: a meta-analysis. Transplantation 71,
104. Nielsen, L. R., Damm, P. & Mathiesen, E. R. Improved Adams, J. S. Vitamin D and barrier function: a novel 1051–1055 (2001).
pregnancy outcome in type 1 diabetic women with role for extra-renal 1 alpha-hydroxylase. Mol. Cell 153. Kainz, A., Harabacz, I., Cowlrick, I. S., Gadgil, S. D.
microalbuminuria or diabetic nephropathy: effect of Endocrinol. 215, 31–38 (2004). & Hagiwara, D. Review of the course and outcome of
intensified antihypertensive therapy? Diabetes Care. 128. Bramham, K. et al. Chronic hypertension and 100 pregnancies in 84 women treated with tacrolimus.
32, 38–44 (2009). pregnancy outcomes: systematic review and meta- Transplantation. 70, 1718–1721 (2000).
105. Bramham, K. Diabetic nephropathy and pregnancy. analysis. BMJ 348, g2301 (2014). 154. Chakkera, H. A., Kudva, Y. & Kaplan, B. Calcineurin
Semin. Nephrol. 37, 362–369 (2017). 129. Magee, L. A. et al. Less-tight versus tight control of inhibitors: pharmacologic mechanisms impacting both
106. World Health Organization. WHO recommendations hypertension in pregnancy. N. Engl. J. Med. 372, insulin resistance and insulin secretion leading to
on antenatal care for a positive pregnancy experience. 407–417 (2015). glucose dysregulation and diabetes mellitus.
http://apps.who.int/iris/bitstream/10665/ 130. Bramham, K., Hall, M., Lightstone, L. & Clin. Pharmacol. Ther. 101, 114–120 (2017).
250796/1/9789241549912-eng.pdf?ua=1 Nelson-Piercy, C. Renal Disease in Pregnancy 2nd edn 155. Zheng, S. et al. Pharmacokinetics of tacrolimus
(2016). (Cambridge Univ. Press, in press). during pregnancy. Ther. Drug Monit. 34, 660–670
107. Pavord, S. et al. UK guidelines on the management of 131. Li, D. K., Yang, C., Andrade, S., Tavares, V. & (2012).
iron deficiency in pregnancy. Br. J. Haematol. 156, Ferber, J. R. Maternal exposure to angiotensin 156. Bramham, K., Chusney, G., Lee, J., Lightstone, L. &
588–600 (2012). converting enzyme inhibitors in the first trimester and Nelson-Piercy, C. Breastfeeding and tacrolimus: serial
108. Steer, P., Alam, M. A., Wadsworth, J. & Welch, A. risk of malformations in offspring: a retrospective monitoring in breast-fed and bottle-fed infants.
Relation between maternal haemoglobin cohort study. BMJ 343, d5931 (2011). Clin. J. Am. Soc. Nephrol. 8, 563–567 (2013).
concentration and birth weight in different ethnic 132. Bateman, B. T. et al. Angiotensin-converting enzyme 157. Wallace, D. J., Gudsoorkar, V. S., Weisman, M. H. &
groups. BMJ 310, 489–491 (1995). inhibitors and the risk of congenital malformations. Venuturupalli, S. R. New insights into mechanisms of
109. McMullin, M. F., White, R., Lappin, T., Reeves, J. & Obstet. Gynecol. 129, 174–184 (2017). therapeutic effects of antimalarial agents in SLE.
MacKenzie, G. Haemoglobin during pregnancy: 133. US National Library of Medicine. Drugs and Lactation Nat. Rev. Rheumatol. 8, 522–533 (2012).
relationship to erythropoietin and haematinic status. Database (LactMed). TOXNET https://toxnet.nlm.nih. 158. Kaplan, Y. C., Ozsarfati, J., Nickel, C. & Koren, G.
Eur. J. Haematol. 71, 44–50 (2003). gov/newtoxnet/lactmed.htm (2017). Reproductive outcomes following hydroxychloroquine
110. Sienas, L., Wong, T., Collins, R. & Smith, J. 134. Nice, F. J. et al. Medications and breast-feeding: a use for autoimmune diseases: a systematic review and
Contemporary uses of erythropoietin in pregnancy: guide for pharmacists, pharmacy technicians, and meta-analysis. Br. J. Clin. Pharmacol. 81, 835–848
a literature review. Obstet. Gynecol. Surv. 68, other healthcare professionals part II. J. Pharmacy (2016).
594–602 (2013). Technol. 20, 85–95 (2004). 159. Clowse, M.,E., Magder, L., Witter, F. & Petri, M.
111. Sanchez-Gonzalez, L. R. et al. Efficacy and safety of 135. Benediktsson, R., Calder, A. A., Edwards, C. R. & Hydroxychloroquine in lupus pregnancy. Arthritis
adjuvant recombinant human erythropoietin and Seckl, J. R. Placental 11 beta-hydroxysteroid Rheum. 54, 3640–3647 (2006).
ferrous sulfate as treatment for iron deficiency dehydrogenase: a key regulator of fetal glucocorticoid 160. Marmor, M. F., Kellner, U., Lai, T. Y., Melles, R. B. &
anemia during the third trimester of pregnancy. exposure. Clin. Endocrinol. 46, 161–166 (1997). Mieler, W. F. American Academy of Ophthalmology
Eur. J. Obstet. Gynecol. Reprod. Biol. 205, 32–36 136. van Runnard Heimel, P. J., Schobben, A. F., recommendations on screening for chloroquine and
(2016). Huisjes, A. J., Franx, A. & Bruinse, H. W. The hydroxychloroquine retinopathy (2016 revision).
112. de Benoist, B., McLean, E., Egll, I. & Cogswell, M. transplacental passage of prednisolone in pregnancies Ophthalmology 123, 1386–1394 (2016).
Worldwide prevalence of anaemia 1993–2005. complicated by early-onset HELLP syndrome. Placenta 161. Zemlickis, D. et al. Fetal outcome after in utero
WHO Global Database on Anaemia (World Health 26, 842–845 (2005). exposure to cancer chemotherapy. Arch. Intern. Med.
Organization, 2008). 137. Laskin, C. A. et al. Prednisone and aspirin in women 152, 573–576 (1992).
113. Gaweda, A. E. Markers of iron status in chronic with autoantibodies and unexplained recurrent fetal 162. United States Food and Drug Administration. Rituxan
kidney disease. Hemodial. Int. 21 (Suppl. 1), S21–27 loss. N. Engl. J. Med. 337, 148–153 (1997). final labeling text https://www.accessdata.fda.gov/
(2017). 138. Park-Wyllie, L. et al. Birth defects after maternal drugsatfda_docs/label/2010/103705s5311lbl.pdf
114. Albaramki, J., Hodson, E. M., Craig, J. C. & exposure to corticosteroids: prospective cohort study (2010).
Webster, A. C. Parenteral versus oral iron therapy for and meta-analysis of epidemiological studies. 163. Chakravarty, E. F., Murray, E. R., Kelman, A. &
adults and children with chronic kidney disease. Teratology 62, 385–392 (2000). Farmer, P. Pregnancy outcomes after maternal
Cochrane Database Syst Rev. 1, CD007857 (2012). 139. Carmichael, S. L. et al. Maternal corticosteroid use exposure to rituximab. Blood 117, 1499–1506
115. Tariq, N., Ayub, R., Khan, W. U., Ijaz, S. & Alam, A. Y. and orofacial clefts. Am. J. Obstet. Gynecol. 197, (2011).
Parenteral iron therapy in the treatment of iron 585.e1–585.e7 (2007). 164. Bruel, A. et al. Hemolytic uremic syndrome in
deficiency anemia during pregnancy: a randomized 140. Källén, B. Maternal drug use and infant cleft lip/palate pregnancy and postpartum. Clin. J. Am. Soc. Nephrol.
controlled trial. J. Coll. Physicians Surg. Pak. 25, with special reference to corticoids. Cleft Palate http://dx.doi.org/10.2215/CJN.00280117 (2017).
193–197 (2015). Craniofac. J. 40, 624–628 (2003). 165. Kelly, R. J. et al. Eculizumab in pregnant patients with
116. Kriplani, A. et al. Intravenous iron sucrose therapy 141. Czeizel, A. E. & Rockenbauer, M. Population-based paroxysmal nocturnal hemoglobinuria. N. Engl. J. Med.
for moderate to severe anaemia in pregnancy. case-control study of teratogenic potential of 373, 1032–1039 (2015).
Indian J. Med. Res. 138, 78–82 (2013). corticosteroids. Teratology 56, 335–340 (1997). 166. National Institute for Health and Care Excellence.
117. Al, R. A. et al. Intravenous versus oral iron for 142. Tata, L. J. et al. Effect of maternal asthma, Hypertension in pregnancy: the management of
treatment of anemia in pregnancy: a randomized trial. exacerbations and asthma medication use on hypertensive disorders in pregnancy. (RCOG Press,
Obstet. Gynecol. 106, 1335–1340 (2005). congenital malformations in offspring: a UK 2011).
118. al‑Momen, A. K. et al. Intravenous iron sucrose population-based study. Thorax 63, 981–987 (2008). 167. Henderson, J. T. et al. Low-dose aspirin for the
complex in the treatment of iron deficiency anemia 143. Hviid, A. & Mølgaard-Nielsen, D. Corticosteroid use prevention of morbidity and mortality from
during pregnancy. Eur. J. Obstet. Gynecol. Reprod. during pregnancy and risk of orofacial clefts. CMAJ preeclampsia: a systematic evidence review for the U.
Biol. 69, 121–124 (1996). 183, 796–804 (2011). S. Preventive Services Task Force. Ann. Intern. Med.
119. Moll, R. &. Davis, B. Iron, vitamin B12 and folate. 144. Norwood, F. et al. Myasthenia in pregnancy: best 160, 695–703 (2014).
Clin. Sci. 45, 198–203 (2017). practice guidelines from a UK multispecialty working 168. Rolnik, D. L. et al. Aspirin versus placebo in
120. Maxwell, P. H. & Eckardt, K. U. HIF prolyl hydroxylase group. J. Neurol. Neurosurg. Psychiatry 85, 538–543 pregnancies at high risk for preterm preeclampsia.
inhibitors for the treatment of renal anaemia and (2014). N. Engl. J. Med. 377, 613–622 (2017).
beyond. Nat. Rev. Nephrol. 12, 157–168 (2016). 145. Francella, A. et al. The safety of 6‑mercaptopurine 169. Poon, L. C. et al. Aspirin for evidence-based
121. Royal College of Obstetricians and Gynaecologists. for childbearing patients with inflammatory bowel preeclampsia prevention trial: effect of aspirin in
Vitamin D in Pregnancy. Scientific Impact Paper disease: a retrospective cohort study. prevention of preterm preeclampsia in subgroups of
Number 43 https://www.rcog.org.uk/globalassets/ Gastroenterology 124, 9–17 (2003). women according to their characteristics and medical
documents/guidelines/scientific-impact-papers/ 146. lint, J. et al. BSR and BHPR guideline on prescribing and obstetrical history. Am. J. Obstet. Gynecol. 217,
vitamin_d_sip43_june14.pdf (2014). drugs in pregnancy and breastfeeding-part I: standard 585.e1–585.e5 (2017).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 19


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

170. Datta, P., Rewers-Felkins, K., Kallem, R. R., a Canadian and United States cohort comparison. in preeclampsia. J. Am. Soc. Nephrol. 27, 903–913
Baker, T. & Hale, T. W. Transfer of low dose aspirin J. Am. Soc. Nephrol. 25, 1103–1109 (2014). (2016).
into human milk. J. Hum. Lact. 33, 296–299 178. Asamiya, Y. et al. The importance of low blood urea 185. Williams, D. & Davison, J. Chronic kidney disease in
(2017). nitrogen levels in pregnant patients undergoing pregnancy. BMJ 336, 211 (2008).
171. Edling, K. L. et al. A pregnant dilemma: primary hemodialysis to optimize birth weight and gestational
hyperparathyroidism due to parathyromatosis in age. Kidney Int. 75, 1217–1222 (2009). Acknowledgements
pregnancy. Endocr. Pract. 20, e14–e17 (2014). 179. Piccoli, G. B. et al. Pre-eclampsia or chronic kidney The authors acknowledge the US National Institute for Health
172. Nadarasa, K. et al. The use of cinacalcet in pregnancy disease? The flow hypothesis. Nephrol. Dial. Transplant. Research (NIHR) Rare Diseases Translational Research
to treat a complex case of parathyroid carcinoma. 28, 1199–1206 (2013). Collaboration as well as the Biomedical Research Centre at
Endocrinol. Diabetes Metab. Case Rep. 2014, 180. Agrawal, S., Cerdeira, A., S., Redman, C. & Vatish, M. Guy’s and St. Thomas’ UK National Health Service (NHS)
140056 (2014). Meta-analysis and systematic review to assess the role Foundation Trust and King’s College London for funding K.W.
173. Hoeltzenbein, M., Stieler, K., Panse, M., Wacker, E. of soluble fms-like tyrosine recpetor kinase and under the terms of a doctoral research fellowship. The views
& Schaefer, C. Allopurinol use during pregnancy — placental growth factor ratio in prediction of expressed are those of the authors and not necessarily those
outcome of 31 prospectively ascertained cases and preeclampsia: the SaPPhirE study. Hypertension http:// of the NHS, the NIHR or the UK Department of Health.
a phenotype possibly indicative for teratogenicity. dx.doi.org/10.1161/HYPERTENSIONAHA.117.10182
PLoS ONE 8, e66637 (2013). (2017). Author contributions
174. Indraratna, P. L., Virk, S., Gurram, D. & Day, R. O. 181. Chappell, L. C. et al. Diagnostic accuracy of placental K.W., C.N.P. and K.B. contributed equally to the conception,
Use of colchicine in pregnancy: a systematic review growth factor in women with suspected preeclampsia: design, drafting and revision of this article.
and meta-analysis. Rheumatology http://dx.doi.org/ a prospective multicenter study. Circulation 128,
10.1093/rheumatology/kex353 (2017). 2121–2131 (2013). Competing interests statement
175. Piccoli, G. B. et al. Pregnancy in dialysis patients in 182. Kim, M. Y. et al. Angiogenic factor imbalance early in The authors declare no competing interests.
the new millennium: a systematic review and meta- pregnancy predicts adverse outcomes in patients with
regression analysis correlating dialysis schedules and lupus and antiphospholipid antibodies: results of the Publisher’s note
pregnancy outcomes. Nephrol. Dial. Transplant. 31, PROMISSE study. Am. J. Obstet. Gynecol. 214, 108. Springer Nature remains neutral with regard to jurisdictional
1915–1934 (2016). e1–108.e14 (2016). claims in published maps and institutional affiliations.
176. Hou, S. H. Frequency and outcome of pregnancy in 183. Akbari, A. et al. Circulating angiogenic factors in
women on dialysis. Am. J. Kidney Dis. 23, 60–63 a pregnant woman on intensive hemodialysis: Reviewer information
(1994). a case report. Can. J. Kidney Health Dis. 3, 7 (2016). Nature Reviews Nephrology thanks G. Lipkin and the other,
177. Hladunewich, M. A. et al. Intensive hemodialysis 184. Thadhani, R. et al. Removal of soluble fms-like anonymous reviewers for their contribution to the peer review
associates with improved pregnancy outcomes: tyrosine kinase‑1 by dextran sulfate apheresis of this work.

20 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Вам также может понравиться