Abstract
The extensive metabolic demands of pregnancy require specific physiological and anatomical changes. These changes affect almost all organ systems, including the cardiovascular, respiratory, renal, gastrointestinal, and hematologic system. The placenta adds another layer of complexity. These changes make it challenging for clinicians to understand presenting signs and symptoms, or to interpret laboratory and radiological tests. Furthermore, these physiological alterations can affect the pharmacokinetics and pharmacodynamics of drugs. Postpartum physiology includes the return to pre-pregnancy physiology, and lactation with drug safety concerns, commonly only supported by limited evidence. In addition, the teratogenic effects of medications are often extrapolated from animals, which further adds uncertainties. Unfortunately, pregnant women are only rarely included in clinical drug trials, while doses, regimens, and side effects are often extrapolated from studies conducted in non-pregnant populations.
In this comprehensive review, we present the changes occurring in each system with its effects on the pharmacokinetic variables. Understanding these physiological changes throughout normal pregnancy helps clinicians to optimize the health of pregnant women and their fetuses. Furthermore, the information on pregnancy-related physiology is also critical to guide study design in this vulnerable ‘orphan’ population, and provides a framework to explore pregnancy-related pathophysiology such as pre-eclampsia.
Keywords: Pregnancy, Physiology, Pharmacokinetics, Pharmacology, Anatomy
Introduction
The amount of drug prescriptions and herbal medicine use in pregnancy has substantially increased during recent decades. This change has been more evident during the first trimester, where the average drug usage has increased by 62.5% between 1976 and 2008 [1–4].
The risks of pharmacotherapy in pregnancy are two-fold: for the pregnant woman and her fetus; altered pharmacokinetics and pharmacodynamics may lead to inadequate therapeutic response or maternal toxicity, and maternal-fetal transmission may impair fetal organ formation or cause fetal demise. Studies on the pharmacokinetics and safety of drugs during pregnancy are scarce [4, 5]. Therefore, practitioners need to balance the risks and benefits of each drug before deciding to prescribe.
Pharmacokinetics (PK) of a drug includes the processes of drug absorption, distribution, metabolism, and excretion (ADME), whereas pharmacodynamics (PD) describes the mechanism of action and the pharmacological effect(s) of a drug [6]. Several physiological and anatomical changes that occur during pregnancy may alter some or all of these ADME processes (Table 1). Clinicians often lack the knowledge on how and why drug disposition is altered in pregnancy, and for that reason, determining the “appropriate” doses to result in appropriate target exposure is difficult [7].
Table 1.
The different pharmacokinetic variables and the physiologic changes that affect these variables during pregnancy.
Pharmacokinetic Variable | Physiologic change that affects it | Direction of Change [38] |
---|---|---|
Absorption | Nausea and vomiting | Decreased |
Gastric motility | Delayed absorption | |
Distribution | Increase in body weight | Increased |
Increase in fat deposition | Increased for lipophilic drugs | |
Increase of plasma volume | Increased | |
Decrease of plasma albumin | Increased for unbound drugs | |
Metabolism | Change in hepatic drug metabolizing enzymes | Variable; depends on specific enzyme |
Excretion | Increase of hepatic blood flow | Increased clearance for high excretion ratio drugs |
Increase of renal blood flow | Increased clearance for unchanged drugs | |
Increase of glomerular filtration rate | Increased clearance of unchanged drugs | |
Transport | Placental drug metabolism | Variable |
Placental transport | ||
Accumulation in amniotic fluid |
The increase in plasma volume, weight gain, and change in fat composition, as well as other effects on the renal, cardiovascular, and gastrointestinal systems play a pivotal role in determining the PK variables of a drug. Randomized controlled trials for drugs rarely include pregnant women as clinical investigators encounter barriers that span across medical, logistical, legal, and ethical arenas [5, 8]. The doses of most drugs currently used in pregnancy are extrapolated from studies conducted in men and non-pregnant women. Also, post-marketing surveillance and results from retrospective studies are used to determine the safety profile of drugs [9, 10]. The fact that studies determining teratogenicity of drugs were historically based on animal-model studies, adds complexity to extrapolate the findings to the human species [11]. A recent review showed that less than 0.5% of all registered clinical trials target the use of pharmaceutical drugs among pregnant women. Most of these clinical trials were conducted in the therapeutic areas of anesthesia and analgesia, preterm birth and tocolysis, pre-eclampsia and pregnancy-induced hypertension in descending order [12].
The placenta and potential fetal drug exposure add additional complexity to this situation. Various methods can be used to study whether and to which extent a drug crosses the placenta, such as cell cultures, organ-on-a-chip techniques, ex vivo experiments and in silico models [13]. Ideally, the information obtained from such experiments is integrated in pharmacometric models, such as physiologically based pharmacokinetic (PBPK) models, to improve the understanding of a drug’s PK and PD profile in pregnant women. Yet, although PBPK models have retrospectively demonstrated a good predictive performance, in vivo concentration data from clinical trials are still needed for confirmation and increasing the confidence in these models which requires interdisciplinary collaboration between clinicians and pharmacometricians [14]. In vivo data on fetal exposure are generally limited to single umbilical cord blood sampling at delivery, preferably with paired maternal sampling.
Even if coping well with the logistics of conducting a randomized controlled trial including pregnant women to study mechanisms governing a drug’s PK and PD, the effects of the physiological changes in pregnancy will need to be taken into consideration in the study design, data collection and interpretation. In this review we provide an overview of the systems that are affected in pregnancy and how each can contribute to changing certain variables related to the pharmacokinetics and pharmacodynamics of frequently used drugs during pregnancy.
Cardiovascular system
The cardiovascular system undergoes several significant anatomical and physiological changes during pregnancy. The heart is displaced more laterally and to the upper left of the chest by the effect of the progressively elevated diaphragm. Moreover, the left ventricle muscle wall is enlarged to accommodate the increase in blood volume, and as a result, the ejection fraction remains unchanged, maintaining compliance [15]. In terms of hemodynamic variables, the end-systolic volume and end-diastolic volume are increased, whereas the end-systolic and end-diastolic pressures remain the same [15]. On cardiac exam, there is evidence of changes in physiology, including mild tachycardia, peripheral edema, jugular venous distention, and lateral displacement of cardiac apex [16]. As for heart sounds, a louder first sound with the exaggerated splitting of the second sound and a third heart sound can be heard [16]. Also, systolic murmurs can be heard at the left lateral sternal border in 90% of pregnant women, and this is considered a flow murmur, reflecting the increased blood volume passing through the aortic and pulmonary valve [17].
Cardiac output denotes the amount of work performed by the heart and is the product of stroke volume and heart rate. It is affected by the blood volume, heart rate, and autonomic regulation of the heart and vessels, yet the exact contribution of each remains controversial [18, 19]. Cardiac output increases in a non-linear fashion at the beginning of the pregnancy, reaches its peak by early third trimester which can be up to 45% in a singleton pregnancy, then decreases slightly towards term (Table 2) [20, 21]. That early increase is more pronounced in multiple gestations [22]. During the first trimester, there is a reduction in the systemic vascular resistance (afterload) stimulating the sympathetic nervous system and leading to an increase in heart rate. Thus, a reduction in afterload is responsible for the increase in cardiac output during the first trimester as opposed to the previous attribution to change in stroke volume [20, 23].
Table 2.
Summary of key physiological changes during pregnancy.
System | Parameter | Direction of change (quantitative) [30, 183, 184] |
---|---|---|
Cardiovascular | Cardiac output | Increased (20%–45%) |
Heart rate | Increased (10 to 20 bpm) | |
Stroke volume | Increased (20% to 30%) | |
Plasma volume | Increased (30% to 50%) | |
Blood Pressure | ||
Systolic | No change | |
Diastolic | Decreased (10 mmHg) | |
Respiratory | Respiratory rate | No change |
Residual volume | Decreased | |
Tidal volume | Increased (up to 40%) | |
Minute ventilation | Increased (up to 50%) | |
Functional Residual Capacity | Decreased (20%) | |
Oxygen Consumption | Increased (20%) | |
FEV1 | Unchanged | |
Gastrointestinal | Serum Albumin | Decreased (20% to 40%) |
Gastric Emptying | Prolonged | |
Hepatic artery blood flow | No change | |
Renal | Glomerular filtration rate | Increased (up to 50%) |
Renal Plasma Flow | Increased (up to 50%) | |
Total Body Water | Increased (up to 20%, 75% in ECF and 25% in blood) |
FEV1=Forced Expiratory Volume at end of 1 second; ECF= Extracellular fluid.
During the third trimester, the enlarging size of the gravid uterus compresses the vena cava [19, 24]. In addition, the blood flow to the utero-placental circulation is also at its peak in the third trimester [25]. Both factors contribute to decreasing the preload and might lead to a decrease in cardiac output during the third trimester. However, this is offset by the increase in the resting heart rate. Moreover, there is preferential blood flow to the uterus and placenta (up to 10-fold increase in uterine arterial blood flow) near term [26–29]. During labor, the cardiac output increases again as a result of increased blood volume accompanying contractions, and the phenomenon of “auto-transfusion ” as blood from the utero-placental unit is shunted back to the maternal circulation immediately after delivery [30].
The systolic blood pressure remains the same, but diastolic blood pressures decrease during pregnancy, reaching a nadir at 24–26 weeks and then rises again [23]. Blood pressure variations are common, and it is dependent on the position of the mother and on the systemic vascular resistance which decreases due to the interplay of progesterone, nitric oxide, endothelin, and estrogen [31]. The mean arterial pressure is determined by the systemic vascular resistance and cardiac output and serves as an indicator of organ perfusion. The autoregulation compensates the decrease systemic vascular resistance by increasing the cardiac output. [32]. In the case of pulmonary pressures, the pulmonary vascular resistance is decreased; however, the pulmonary capillary wedge pressure remains the same. This is advantageous because it allows to accommodate the increased cardiac output. In case of a pulmonary vascular disease during pregnancy, the increased resistance would lead to pulmonary artery hypertension which will not tolerate the increased cardiac output and would result in right sided heart failure [33]. Supine hypotension is a known physiological change that occurs in 10% of women during pregnancy as a result of compression of great vessels by the gravid uterus [34] and must not be mistaken for true hypotension.
There is an increase in the plasma volume during pregnancy to accommodate the increased needs of the placenta and the fetus [35, 36]. The plasma volume increase is evident as early as 6–8 weeks, and continues to increase throughout the pregnancy, peaking at 32 weeks [37]. This also causes an increase in the apparent volume of distribution of drugs, which implies that hydrophilic drugs would require a higher initial dose and maintenance dose to obtain plasma concentration levels that are comparable to those in non-pregnant women [38]. In addition, there is a 13% decrease in albumin concentration from non-pregnant levels by 32 weeks [39], which may cause an increase in the unbound drug concentration; this is very important clinically as drugs which are highly bound would have much higher concentrations of their free form during pregnancy (i.e. digoxin and phenytoin) [28, 30]. In addition, the α1-glycoprotein, which is a binding protein for many basic compounds, slightly decreases during healthy pregnancy [40].
Gastrointestinal System
Heartburn is common during pregnancy. This can be attributed to the decrease in gastric secretion pH, increase in the amount of secretions along with a decrease in the lower esophageal sphincter tone [41]. Lower esophageal sphincter tone decreases as a result of progesterone action on smooth muscle cells and is responsible not only for the heartburn symptom but also contributes to nausea and vomiting [42]. Nausea and vomiting of pregnancy is also common in around 80% of pregnant patients but with variable severity and presentation [43, 44]. It can present as early as the 2nd week and last up until the 2nd trimester, and in some cases until 37 weeks’ gestation or full term [45–47]. This is important to keep in mind as clinicians need to control the symptoms before the absorption of medication and bioavailability of oral drugs is altered. For that reason, it is recommended that patients take their medication at a time when nausea is minimal to avoid decreased bioavailability of the drug due to lower concentrations at the absorption site [38].
There is a controversy as to whether gastric emptying and motility are delayed during pregnancy and if that impacts the peak serum concentration of oral medication. Some studies showed that the delay is a result of increased progesterone and estrogen levels, causing the relaxation of smooth muscle cells in the alimentary tract [48, 49]. This decrease can often lead to constipation and bloating and cause discomfort to patients in addition to its role in augmenting nausea and vomiting during pregnancy resulting theoretically in a delay in the peak concentration of oral drugs in the system. Other studies have noted delayed gastric emptying specifically early in pregnancy [50, 51], compared to those showing no delay in motility throughout pregnancy [50, 52]. Absorption of certain medications, such as acetaminophen, can be affected by delayed gastric emptying during pregnancy. Furthermore, concomitant use of some medications may alter absorption profiles for both pregnant and non-pregnant patients [53]. For example, due to the fact iron supplements and proton pump inhibitors may decrease levothyroxine absorption, patients are instructed not to take them concurrently [54].
The physiological effect of pregnancy on liver enzymes involved in drug metabolism varies [55, 56]. Several phase I enzymatic activities are either enhanced or decreased. For example, activity of the CYP2D6 enzyme is increased resulting in increased clearance of fluoxetine, one of its substrates, which might require dose adjustment to reach a pharmacological effect [57]. Other phase I enzymes, such as CYP2A6, 2C9 and 3A4 appear to be induced as well resulting in decreased plasma concentrations of drugs metabolized via these enzymes, such as cotinine, phenytoin and midazolam [58–60]. Of note, as illustrated for phenytoin[59],a highly protein-bound anticonvulsant, the increase in total drug clearance can be further enhanced by increases in the free fraction which stems itself from reduced serum albumin levels. On the other hand, a decreased activity of drug metabolizing enzyme CYP1A2 will result in a decrease in the clearance of caffeine, olanzapine, and clozapine, and thus dose adjustment is needed to avoid toxic concentrations [57]. The activity of essential enzyme in the phase II metabolism pathway of the liver, including many members of the uridine diphosphate glucuronosyltransferase (UGT) 1 family [61] is also altered. For example, UGT1A1 appears to be induced by increased progesterone concentrations during pregnancy,, while others, such as UGT2B7 appear to be unchanged [62]. Labetalol, an α−1 and β-adrenoreceptor blocker and a substrate of UGT1A1, is an important drug for treating elevated blood pressure in pregnancy (Table 3) [63]. It is believed to have a lower availability due to increased hepatic clearance and first pass excretion, with a bioavailability averaging 20–40%. Its clearance is dependent on phase II metabolism that is enhanced during the second and third trimester compared to the post-partum period, which is similar to metabolism of acetaminophen during pregnancy [62, 64]. As a consequence, labetalol is often dosed using twice or three times daily regimens.
Table 3.
Pharmacokinetic variables of selected clinical drugs commonly used pregnancy.
Drug | Trimester | Comparison group | Overall observed PK changes | References | ||
---|---|---|---|---|---|---|
VD | CL | t1/2 | ||||
Ampicillin | 3rd | Post-partum | ↑38%‡ | ↑50%‡ | ↓12%‡ | [185] |
Cefazolin | 2nd | Post-partum | NR | ↑31‡ | ↓35%‡ | [186] |
Enoxaparin | All | Non-pregnant | ↑48%‡ | ↑49%‡ | ↑13% | [187] |
Fluoxetine | 3rd | Historical controls | NR | NR | NR | [188] |
Lamotrigine | 3rd | Non-pregnant | NR | ↑140‡ | NR | [189] |
Levothyroxine | All | No grouping | NR | no change in dose | NR | [190] |
Labetalol | 1st /term | Post-partum | ↑90% | ↑40%‡&60%‡ respectively | NR | [191] |
Nifedipine | 3rd | Non-pregnant | ↑ (magnitude unspecified) | ↑308% | ↓(magnitude unspecified) | [192] |
Phenytoin | 3rd | Post-partum | NR | ↑256% | NR | [59] |
statistically significant
PK=Pharmacokinetics, NR= not reported, VD= Volume of Distribution, CL=clearance, t1/2=Half-life, IV=intravenous, IM=Intramuscular.
Renal system
During pregnancy, the kidneys are in a state of glomerular hyperfiltration since there is increased plasma volume, increased effective renal plasma flow (eRPF), and decreased renal plasma oncotic pressure. The eRPF increases by about 80% at the end of the first trimester as a result of massive vasodilation from relaxin-mediated nitric oxide release [65, 66]. This is clinically manifested by a decrease in creatinine levels by an average of 0.4 mg/dL reflecting an increased glomerular filtration rate (GFR), which can be up to 50% from baseline [67]. At the beginning of pregnancy, the GFR and eRPF both increase, however, the eRPF declines during late pregnancy while the GFR plateaus from the second trimester till birth and returns to normal levels in the postpartum period [67, 68]. Frequency and urgency, which are common symptoms seen even early in pregnancy, are believed to be due to these changes in GFR and eRPF [69]. Later in pregnancy frequency and urgency are also secondary to the physical compression of the bladder by the growing uterus and fetus. Serum creatinine is a rapid evaluation of glomerular filtration, though a 24-hour urine collection sample is the more accurate assessment of creatinine clearance [70].
Furthermore, this physiological change can be deemed cumbersome when dealing with renally cleared drugs. Lithium, which is prescribed as a mood stabilizer for bipolar patients, is one of the renally cleared drugs of which its clearance is doubled during the third trimester of pregnancy, with a fast return to pre-pregnancy clearance in the first 2 weeks after delivery [71, 72]. Other drugs, among the many that can be affected, include cephalosporins, piperacillin, atenolol, and digoxin [71]. Although the change in GFR is uniform as the pregnancy progresses, it is noted that the renally-excreted drugs are not affected equally, and that is because of the role of renal transporters responsible for secretion and reabsorption in the tubules and changes in the plasma protein binding of some drugs [73].
Urinalysis in pregnancy may show glucosuria resulting from increased GFR and impaired resorption at the proximal tubule level. However, even though it might be physiologic, the finding of glucosuria always warrants investigation for pregnancy related diabetes mellitus [74]. For the same reason, proteinuria is commonly found on urinalysis during pregnancy with values below the threshold of 300 mg/dL, and any value above that would require investigational studies to rule out pregnancy-specific abnormalities. This cut-off is double that of the non-pregnant states [75, 76].
Water and sodium regulation is changed during pregnancy. Despite the rise in all components of the renin-angiotensin system, resistance to pressor effects contribute to a more substantial increase in extracellular volume along with water and sodium retention [77]. As a result, plasma osmolarity is slightly decreased to an average of 280 mOsm/kg with an average value about 270 mOsm/kg and sodium to 136 meq/dL [78]. The body retains sodium despite the increased filtration by the kidneys, through upregulation of mechanisms responsible for reabsorption in the distal tubules.
In addition to the physiological changes, there are several anatomical modifications to be noted in pregnancy. The size of the kidney increases by 1 cm at the end of the pregnancy, as a result of the increase in vasculature and plasma volume [79]. Moreover, the renal calyces and ureters are dilated occurring more than 80% of pregnant women by mid-gestation and more commonly on the right side. Clinically, this is reflected by an increased risk of ascending urinary tract infection from urine stasis and sonographic difficulties in differentiating pathology from normal changes like hydronephrosis [80–82].
Endocrinology system
The several metabolic changes occurring during pregnancy are essential for meeting the demands of the growing fetus and placenta. By the third trimester, the basal metabolic rate is increased by 20% [83]. There exists a state of hyperinsulinism from hyperplasia of islet cells in the pancreas along with a state of peripheral insulin resistance. Human placental lactogen and the human placental growth hormone are thought to contribute to insulin resistance [84, 85]. The former acts as an insulin-like and anti-insulin molecule causing pancreatic cells to secrete insulin, and at the same time inducing peripheral resistance to insulin [84]. This ensures glucose availability for the fetus by keeping the postprandial glucose levels elevated [86, 87]. Also, augmentation of hepatic gluconeogenesis in the third trimester contributes to sustaining the postprandial glucose levels [88]. Patients with pre-existing insulin resistance or diabetes will need to adjust their insulin medication to higher doses as the pregnancy progresses [89].
Maternal lipid levels are increased under the effect of insulin resistance and estrogen [90]. During the first part of pregnancy, lipid synthesis is favored, and stored fat is increased. However, during the third trimester, the fat stored will be used by the mother for energy production [91]. This is important since fatty acids and glycerol serve as the nutritive fuel for the mother, whereas glucose and amino acids are preferentially utilized by the fetus [86]. In addition to the metabolic status, several hormone-secreting organs like the pituitary and thyroid undergo drastic changes. Estrogen induces hyperplasia and hypertrophy of lactotrophs resulting in an enlarged pituitary gland with prolactin levels, subsequently increasing as well [92, 93]. Also, there is a decrease in gonadotrophs from increased progesterone and estrogen. However, the corticotrophs and thyrotrophs remain the same [94]. The placenta also contributes to the hormonal milieu, by secreting its form of the growth hormone synthesized by the syncytiotrophoblast. The placental growth hormone is increased in mid-pregnancy, stimulating the release of insulin-like growth factors which contribute to the acromegalic features of some women during pregnancy [95].
Pregnancy is a state of hypercortisolism. Corticotropin-releasing hormone is synthesized by the placenta and increased until term [96]. This corticotropin-releasing hormone stimulates the release of both placental and pituitary adrenocorticotropic hormone, and characteristically, the former is insuppressible by the low-dose dexamethasone test [97, 98]. Moreover, the hepatic production of cortisol-biding globulins is upregulated under the effect of estrogen, ensuring an increased half-life and decreased hepatic clearance [99]. Consequently, cortisol levels are increased, and mainly, a cortisol surge occurs at labor [100].
The total triiodothyronine and tetraiodothyronine levels are increased during pregnancy as a result of an estrogen-induced increase in thyroid-binding globulin, decreased hepatic clearance and an increase in production of hormones by the thyroid gland [101, 102]. These hormones play a pivotal role in fetal neurogenesis. In particular, early pregnancy levels of thyroid hormones are positively correlated with cognitive and behavioral development in infancy and childhood [103]. Historically, there has been some controversy about treating subclinical maternal hypothyroidism and improvement of neurodevelopmental outcomes. A recent clinical trial definitively showed that neurodevelopmental outcome through 5 years of age is not significantly improved in the treatment arm [104]. Therefore, the American Congress of Obstetrics and Gynecology along with the American Thyroid Association do not recommend universal screening of thyroid dysfunction during pregnancy [105, 106]
The thyroid gland is increased in size during pregnancy. In addition, an iodine deficiency state ensues because of the active transport of iodine across the placenta to the fetus, increased renal excretion and increased consumption of iodine by the mother’s thyroid [107]. Despite the increase in total hormones, the free-from remains stable, maintaining a euthyroid status in the pregnant woman [108]. In the case of hypothyroidism, the dose of levothyroxine, if required, should be increased by 30% in early pregnancy to meet the maternal demands, and then usually returns to pre-pregnancy doses postpartum to avoid excessive exogenous thyroid hormone exposure [109].
Hematologic changes
As discussed earlier, the plasma volume increases in pregnancy nonlinearly and non-monotonically. It increases by 15% in the first trimester, and more rapidly during the second trimester, continuing at a slower rate during the third trimester till it plateaus in the few weeks before delivery [110, 111]. As a result, there is an average gain of 30 to 50% or more than one liter of blood volume compared to non-pregnant levels [112]. This is advantageous for mother and fetus in response to the increased metabolic demands of pregnancy, and more importantly, it serves as a protective buffer to minimize consequences of large volume blood loss encountered during delivery.
As for blood components, the upregulation of erythropoiesis, induced by erythropoietin hormone from the maternal kidneys, increases red blood cell (RBC) mass. However, this increase lags behind that of the plasma volume resulting in dilutional anemia, or more clinically known as “anemia of pregnancy” [113]. One advantage of this is that the blood viscosity decreases, with increasing carrying capacity of the RBCs, ensuring a more efficient exchange between mother and fetus [114]. The hemoglobin averages at 12.5 g/dl, and despite physiologic anemia, any value below 11 g/dL should be investigated [115]. Iron requirements increase by the latter half of the pregnancy, and it is imperative to start the patient on iron supplements early on in pregnancy especially in the subset of patients who are anemic at the start of their pregnancy to avoid maternal and neonatal morbidity, particularly low birth weight or severe hemorrhage [116–118].
Lymphocytes are higher in pregnancy, averaging around 15,000/mm3, and this value is even higher as we approach labor and delivery [119]. For that reason, diagnosing an infection, especially at the time of delivery like chorioamnionitis using the lab values, may be difficult and the clinician should always support the lab values with the clinical picture. In addition to looking at the number of band cells or immature cells which are not elevated in pregnancy [30]. Although studies have documented changes in platelet count, their level remains within the normal range [120]. It is believed that the drop in platelets is due to hemodilution along with an element of hypersplenism from splenic enlargement, reaching up to 50% during the first trimester, and resulting in platelets sequestration [121–123].
Pregnancy is a hypercoagulable state as evident by upregulation of procoagulation factors like all clotting factors, and especially thrombin (factor II) and fibrinogen (factor I) [124–126]. Moreover, regulatory proteins responsible for anticoagulation such as protein S and anti-thrombin are decreased during pregnancy, and there is increased resistance to activated protein C [127]. The result of the interplay between coagulation and anticoagulation pathway is evident, and this might play a role in decreasing blood loss during delivery and afterwards. However, pregnant women are at increased risk of venous thromboembolism in both pregnancy and postpartum period, and that is a leading cause of maternal death [128–131]. It is maybe that the thrombosis of pelvic veins offers an advantage for facilitating normal postpartum involution, as anti-thrombin falls and reaches a nadir at 12 hours postpartum before rising again and plateauing at 72 hours [132, 133]. Low-molecular-weight-heparin has become the traditional drug of choice of treating venous thromboembolism in pregnancy. Most women go on twice daily dosing for anticoagulation during pregnancy or postpartum, however, a population pharmacokinetic study was able to show efficacy of once-daily dosing, adjusted to weight, in treatment of venous thromboembolism [134]. Often women are continued for low-molecular-weight-heparin for a duration of at least 6 weeks depending on the indication. Further research is needed in the area of optimal anticoagulation dosing especially for women with active thromboembolism who have additional comorbidities such as morbid obesity.
Respiratory System
Increasing progesterone during pregnancy induces a change in the threshold of the respiratory centers of the brain, increasing the sensitivity to carbon dioxide, which is reflected by the slope of the ventilation curve in response to the changes in alveolar carbon dioxide [135, 136]. In addition to that, progesterone also mediates dilation of the respiratory airways and hyperemia and edema of the mucosal surfaces causing nasal congestion, resulting in rhinitis of pregnancy [137–139]. The effect of estrogen is by upregulating the progesterone receptors in the central nervous system, particularly in the medulla and hypothalamus, where the respiratory control center resides [140].
Anatomically, the upward displacement of the diaphragm as a result of the growing gravid uterus leads to a decrease in functional residual capacity [141]. Not only the mechanical effect causes this change, but it has been shown that the displacement occurs earlier in pregnancy when the uterus is not enlarged, and it is attributed to the effect of progesterone and relaxin on inducing the ligamentous attachments of the lower ribs to relax [142]. Other anatomical changes include an increase in the subcostal angle by 50% by the end of pregnancy from 68.5 to 103.5, and the transverse diameter of the rib cage increases by 2 cm [142]. Eventually, the chest wall compliance decreases by the third trimester due to increased abdominal content; however, lung compliance remains the same [143, 144]. Despite the changes in the anatomy, inspiratory, and expiratory maximum pressure values remain preserved throughout pregnancy [141]. Dyspnea is a common presentation during pregnancy as this physiological dyspnea occurs in 50–70% of patients by the third trimester [145].
As for the pulmonary function test, the tidal volume is increased by up to 50% (or 650 ml) of the non-pregnant values, but the respiratory rate remains the same. Consequently, the minute ventilation, which is the product the tidal volume and respiratory rate follow with the trend and increases to up to 50% [146]. The functional residual capacity (FRC) decreases by 20 to 30 percent during pregnancy, as a result of diaphragm elevation decreasing outward recoil of the chest. Pregnant women are at risk of hypoxemia during general anesthesia induction as a result of this decrease in FRC [147, 148]. The expiratory reserve volume and residual volume make up the FRC, and both decrease by about 20% [149]. On the other hand, the inspiratory capacity, which is defined as the maximum inhaled volume from FRC, is increased by 10% and the total lung capacity (a combination of Inspiratory Capacity and FRC) remains the same. These changes do not differ between singleton and twin pregnancy [150].
In pregnancy there is a state of respiratory alkalosis with maternal pH ranging between 7.42–7.46 [151]. Arterial blood gases change as a result of the increase in minute ventilation. The PaO2 is increased from 93 to 106 mmHg, and the PaCO2 is decreased by 37 to 30 mmHg by the end of the third trimester [151–153]. The latter decrease provides an advantageous gradient between mother and fetus, allowing CO2 of the fetus to be transferred to the mother. Fetal acidemia, among other factors, can affect local placental transportation of weakly basic compounds like the anesthetic drugs bupivacaine and meperidine, and can result in the entrapment of these compounds in their ionic form [154, 155]. The renal system compensates by increasing the excretion of bicarbonate, which its serum levels decrease from 23 to 18 mEq/L in late pregnancy [156]. Moreover, the chronic state of respiratory alkalosis in pregnancy would shift the oxyhemoglobin curve to the right, which facilitates the oxygen diffusion across the placenta [157].
Volatile anesthetic agents with minimum alveolar concentration (MAC) values more than 1.5 MAC can lead to dilation of uterine arteries and also reduce uterine tone, and pregnancy has been associated with increased sensitivity to these agents. Most induction agents, opioids, and neuromuscular blockade can be used in pregnancy. Thiopental is the most commonly used agent during rapid-sequence intubation in pregnancy with a lower dose than generally used [158]. More recently, propofol is also used as an alternative agent in early pregnancy after showing no teratogenicity in animal studies [159].
Placenta
The placenta was perceived initially as a protective barrier at the maternal-fetal interface. It was not until fetal malformations were attributed to thalidomide exposure in the early 1960s that physicians started discovering the transport of xenobiotics and other drugs across the placenta from mother to fetus and vice versa [160, 161]. It is well established that uncharged, lipophilic drugs with a relatively low molecular weight can pass readily, and the amniotic fluid surrounding the fetus can also be considered a site for drug distribution and accumulation [6]. However, newer studies are showing evidence of drug transporters in the placenta that can be responsible for active transmission of drugs from the mother to the fetus [162]. In fact, multiple drug transporters, such as P-glycoprotein and the breast cancer resistance protein, are expressed in the membrane of the fetal syncytiotrophoblasts and endothelium at varying levels during gestation [13].
Although the maternal and fetal blood never meet, many nutrients, waste products, and other molecules can and should pass through the brush border (apical) membrane of syncytiotrophoblast cells at the interface [163]. Three types of drug transport categories are recognized: the complete transfer (type 1 drugs), which indicates that a drug can readily cross the placenta, and its concentration will rapidly equilibrate between mother and fetus, or exceeding transfer (type 2 drugs) in which greater concentration is reached in the fetus compared to the mother, and the last is incomplete transfer where the drug is unable to pass through the placenta completely [164].
Like any other molecule, drugs depend on particular mechanisms to pass through the cell membrane. Simple diffusion, like in the cases of midazolam and acetaminophen, is the most common mechanism of drug transport across the placenta, especially for type 1 drugs. Another mechanism is facilitated diffusion like in the case of cephalosporins and corticosteroids, and active transport like the case of norepinephrine or dopamine [164]. However, despite the possibility of drug transport, some medications cannot cross the barrier and reach the fetus. An example of that is doxorubicin, a chemotherapeutic agent involved in several hematologic malignancy treatment protocols [165]. The transfer of molecules across the placenta is also dependent on size, where drugs with a molecular weight > 1000 Da (e.g. low-molecular-weight-heparin and insulin) usually do not cross the placenta [166, 167]. Monoclonal antibodies are a specific group of drugs used to treat oncological or a variety of inflammatory diseases, or to prevent infections in mother or infant (influenza, pertussis).The human placenta has the capacity of active transfer of antibodies from mother to fetus, and this may result in developmental toxicology [168, 169] However, the extent of transfer may also depend on biochemical modifications like PEGylation of the Fc fragment of the antibody. Furthermore, there is some controversy over their use in pregnant women with chronic conditions where the potential benefits outweigh the risks [170].
Summary of physiological changes and model applications
In addition to the previous changes described (Tables 1 & 2), trimester-specific changes are summarized in Figure 1. It is important to note that these percent changes are compared to pre-pregnancy state. These trimester-specific changes may not be valid in disease states like preeclampsia. Pathophysiological changes during preeclampsia result in an additional increase in body weight due to water retention [171], an additional decrease in albumin [172] and impaired GFR (about −15 %) when compared to the normal physiological increase in GFR [173].
Fig 1.
Percentage Change in Physiological Parameters across the Three Trimesters. Percent changes have been calculated at 12, 26 and 38 weeks based on the individual trend lines as described by Dallmann, A. et al. to reflect the first, second and third trimester [193]
HCT=hematocrit; AAG= α−1 acid glycoprotein; GFR= glomerular filtration rate.
During the postpartum period, many of these physiological changes return back to normal and for some this return may be gradual. For example, blood volume decreases rapidly in the first few days under effect of diuresis, and continues to decrease gradually over a period of eight weeks [174]. Labor increases the cardiac output, along with its components (heart rate and stroke volume), to an additional 80% from pre-labor values, and eventually cardiac output will return to normal within 6–8 weeks of pregnancy [175]. Postpartum GFR normalizes to pre-pregnancy estimates within 4 weeks after delivery [173]. Moreover, as uterine size decreases, the FRC also returns to normal within one to two weeks. In cases of chronic medical conditions, such as hypertension, women either resume their pre-pregnancy medications in the postpartum period or commonly stay on labetalol or nifedipine (Table 3) [176]. There is limited data for safety with breastfeeding and use of angiotensin-converting enzyme inhibitors and angiotensin II receptor blocker agents and often these are initiated by a primary care provider at some point beyond the 6 weeks after delivery. Caution is warranted for using thiazide drugs postpartum if they are breastfeeding due to a potential for decreasing breast milk production. The optimal pharmacologic management of diseases in this critical period of recovery from pregnancy is not well studied and it is a new front to discover, as breastfeeding and subsequent maturational changes in intestinal absorption of the infant is an additional factor in the complex model.
Application of mechanistic models for pregnancy
Several modeling tools may help to shed light on changes in pharmacokinetics and pharmacodynamics stemming mainly from the herein discussed physiologic alterations. Mechanistic models, such as PBPK models constitute a strong tool to leverage the accumulated knowledge about physiologic alterations in pregnant women within a biologically plausible framework. For example, previous studies have demonstrated that information obtained from ex vivo perfused placental cotyledons can be integrated in PBPK models to successfully predict and quantify fetal exposure to antiretroviral drugs [177, 178]. Other applications encompass the prediction of a maximum recommended dosage of lithium for pregnant women for treatment of bipolar disorder [179]; the prediction of a worst-case scenario for maternal exposure to the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI) following acetaminophen intake at various stages throughout pregnancy [180]; and the prediction of caffeine pharmacokinetics and pharmacodynamics in the fetus to support the identification of potential cut-offs for caffeine intake during pregnancy [181]. While PBPK models typically focus on pharmacokinetic and pharmacodynamics processes at the organ level, biologically based dose-response (BBDR) models shift the focus to a finer biological scale by incorporating data on biological processes at the cellular and molecular level. These models can be used to study the interactions between compounds and the biological system and to link exposure to a quantifiable biological response. For example, a BBDR model for the thyroid endocrine system was recently used to evaluate perturbations of maternal serum levels of free thyroxin following a range of dietary iodide intake. Through global sensitivity analysis, key parameters of the model were identified that critically drive maternal and fetal iodide kinetics, the thyroid function and their interactions thereby contributing to an improved mechanistic understanding of the thyroidal system during pregnancy [182].
These examples illustrate how modeling approaches can be used to integrate available data and generate new insights into the complex interplay of physiologic alterations in pregnant women and the functioning of the physiological system, and the pharmacokinetic and pharmacodynamics processes of a drug. In combination with clinical data for model development and evaluation, these tools can help to better understand and anticipate clinically relevant changes drug exposure and efficacy. Ultimately, this can lead to a more informed decision-making in the clinical setting and improve therapeutic outcome in pregnant women.
Conclusion
The myriad of changes occurring in pregnancy challenges clinicians to provide the optimal care when prescribing medication in pregnancy. Most notably, changes such that of the gastrointestinal and renal system, along with the increased plasma volume, decreased albumin plasma concentrations and altered enzymatic activity play a key role in determining the pharmacokinetic variables of medication during pregnancy. Understanding these changes is pivotal for the clinician when managing patients that might develop complications such as pre-eclampsia and hemorrhage or have pre-existing medical conditions. To date, pregnant women are still being excluded from many clinical trials studying pharmacokinetic and pharmacodynamic properties of medications. Extrapolating dosages and safety profiles of medications from the non-pregnant population can be misleading or inaccurate and inappropriate dosing foreshadows minor to profound adverse effects. Further pharmacometric and efficacy studies in pregnancy are needed to better understand optimal dosing, so that toxicities are minimized for the mother and the fetus.
Acknowledgement
Funding: The project was funded by NHLBI grant (K23HL141640)
Footnotes
Conflict of Interest: The authors declare that they have no conflict of interest
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.
References
- 1.Mitchell AA, Gilboa SM, Werler MM, et al. (2011) Medication use during pregnancy, with particular focus on prescription drugs: 1976–2008. Am J Obstet Gynecol 205:51.e1–51.e8. 10.1016/j.ajog.2011.02.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Broussard CS, Louik C, Honein MA, Mitchell AA (2010) Herbal use before and during pregnancy. Am J Obstet Gynecol 202:443.e1–443.e6. 10.1016/j.ajog.2009.10.865 [DOI] [PubMed] [Google Scholar]
- 3.Lupattelli A, Spigset O, Twigg MJ, et al. (2014) Medication use in pregnancy: A cross-sectional, multinational web-based study. BMJ Open 4:. 10.1136/bmjopen-2013-004365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Adam MP, Polifka JE, Friedman JM (2011) Evolving knowledge of the teratogenicity of medications in human pregnancy. Am J Med Genet Part C Semin Med Genet 157:175–182. 10.1002/ajmg.c.30313 [DOI] [PubMed] [Google Scholar]
- 5.Sheffield JS, Siegel D, Mirochnick M, et al. (2014) Designing drug trials: Considerations for pregnant women. Clin Infect Dis 59:S437–S444. 10.1093/cid/ciu709 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zhao Y, Hebert MF, Venkataramanan R (2014) Basic obstetric pharmacology. Semin Perinatol 38:475–486. 10.1053/j.semperi.2014.08.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Jeong H (2010) Altered drug metabolism during pregnancy: hormonal regulation of drug-metabolizing enzymes. Expert Opin Drug Metab Toxicol 6:689–699. 10.1517/17425251003677755 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fisk NM, Atun R (2009) Systematic analysis of research underfunding in maternal and perinatal health. BJOG An Int J Obstet Gynaecol 116:347–355. 10.1111/j.1471-0528.2008.02027.x [DOI] [PubMed] [Google Scholar]
- 9.Van der Graaf R, Van der Zande ISE, Den Ruijter HM, et al. (2018) Fair inclusion of pregnant women in clinical trials: An integrated scientific and ethical approach. Trials 19:. 10.1186/s13063-017-2402-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ayad M, Costantine MM (2015) Epidemiology of medications use in pregnancy. Semin Perinatol 39:508–511. 10.1053/j.semperi.2015.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cook MJ, Fairweather FA (1968) Methods Used in Teratogenic Testing. Lab Anim 2:219–228. 10.1258/002367768781082834 [DOI] [Google Scholar]
- 12.Scaffidi J, Mol BW, Keelan JA (2017) The pregnant women as a drug orphan: a global survey of registered clinical trials of pharmacological interventions in pregnancy. BJOG An Int J Obstet Gynaecol 124:132–140. 10.1111/14710528.14151 [DOI] [PubMed] [Google Scholar]
- 13.Dallmann A, Liu XI, Burckart GJ, van den Anker J (2019) Drug Transporters Expressed in the Human Placenta and Models for Studying Maternal-Fetal Drug Transfer. J Clin Pharmacol 59 Suppl 1:S70–S81. 10.1002/jcph.1491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Dallmann A, Mian P, Van den Anker J, Allegaert K (2019) Clinical Pharmacokinetic Studies in Pregnant Women and the Relevance of Pharmacometric Tools. Curr Pharm Des 25:483–495. 10.2174/1381612825666190320135137 [DOI] [PubMed] [Google Scholar]
- 15.Kametas NA, McAuliffe F, Hancock J, et al. (2001) Maternal left ventricular mass and diastolic function during pregnancy. Ultrasound Obstet Gynecol 18:460–466. 10.1046/j.0960-7692.2001.00573.x [DOI] [PubMed] [Google Scholar]
- 16.Davies GAL, Herbert WNP (2007) Assessment and Management of Cardiac Disease in Pregnancy. J Obstet Gynaecol Canada 29:331–336. 10.1016/S17012163(16)32432-X [DOI] [PubMed] [Google Scholar]
- 17.Northcote RJ, Knight P V., Ballantyne D (1985) Systolic murmurs in pregnancy: Value of echocardiographic assessment. Clin Cardiol 8:327–328. 10.1002/clc.4960080604 [DOI] [PubMed] [Google Scholar]
- 18.Desai DK, Moodley J, Naidoo DP (2004) Echocardiographic assessment of cardiovascular hemodynamics in normal pregnancy. Obstet Gynecol 104:20–29. 10.1097/01.AOG.0000128170.15161.1d [DOI] [PubMed] [Google Scholar]
- 19.van Oppen AC, Stigter RH, Bruinse HW (1996) Cardiac output in normal pregnancy: A critical review. Obstet Gynecol 87:310–318. 10.1016/00297844(95)00348-7 [DOI] [PubMed] [Google Scholar]
- 20.Meah VL, Cockcroft JR, Backx K, et al. (2016) Cardiac output and related haemodynamics during pregnancy: A series of meta-analyses. Heart 102:518–526. 10.1136/heartjnl-2015-308476 [DOI] [PubMed] [Google Scholar]
- 21.Hunter S, Robson SC (1992) Adaptation of the maternal heart in pregnancy. Heart 68:540–543. 10.1136/hrt.68.12.540 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ghi T, Degli Esposti D, Montaguti E, et al. (2015) Maternal cardiac evaluation during uncomplicated twin pregnancy with emphasis on the diastolic function. Am J Obstet Gynecol 213:376.e1–376.e8. 10.1016/j.ajog.2015.05.003 [DOI] [PubMed] [Google Scholar]
- 23.Mahendru AA, Everett TR, Wilkinson IB, et al. (2014) A longitudinal study of maternal cardiovascular function from preconception to the postpartum period. J Hypertens 32:849–856. 10.1097/HJH.0000000000000090 [DOI] [PubMed] [Google Scholar]
- 24.Clapp JF, Capeless E (1997) Cardiovascular function before, during, and after the first and subsequent pregnancies. Am J Cardiol 80:1469–1473. 10.1016/S0002-9149(97)00738-8 [DOI] [PubMed] [Google Scholar]
- 25.Thaler I, Manor D, Itskovitz J, et al. (1990) Changes in uterine blood flow during human pregnancy. Am J Obstet Gynecol 162:121–125. 10.1016/00029378(90)90834-T [DOI] [PubMed] [Google Scholar]
- 26.Nelson DB, Stewart RD, Matulevicius SA, et al. (2015) The Effects of Maternal Position and Habitus on Maternal Cardiovascular Parameters as Measured by Cardiac Magnetic Resonance. Am J Perinatol 32:1318–1323. 10.1055/s-0035-1563719 [DOI] [PubMed] [Google Scholar]
- 27.Sanghavi M, Rutherford JD (2014) Cardiovascular Physiology of Pregnancy. Circulation 130:1003–1008. 10.1161/CIRCULATIONAHA.114.009029 [DOI] [PubMed] [Google Scholar]
- 28.Frederiksen MC (2001) Physiologic changes in pregnancy and their effect on drug disposition. Semin Perinatol 25:120–123. 10.1053/sper.2001.24565 [DOI] [PubMed] [Google Scholar]
- 29.Flo K, Wilsgaard T, Vårtun A, Acharya G (2011) A Longitudinal Study of the Relationship Between Maternal Cardiac Output Measured by Impedance Cardiography and Uterine Artery Blood Flow in the Second Half of Pregnancy. Obstet Anesth Dig 31:178–179. 10.1097/01.aoa.0000400323.42088.c2 [DOI] [PubMed] [Google Scholar]
- 30.Pacheco LD, Costantine MM, Hankins GD V, Cohen MS (2013) Physiologic Changes During Pregnancy In: Mattison DR (ed) Clinical Pharmacology During Pregnancy. Academic Press, pp 5–16 [Google Scholar]
- 31.Carbillon L, Uzan M, Uzan S (2000) Pregnancy, vascular tone, and maternal hemodynamics: A crucial adaptation. Obstet Gynecol Surv 55:574–581. 10.1097/00006254-200009000-00023 [DOI] [PubMed] [Google Scholar]
- 32.Melchiorre K, Sharma R, Thilaganathan B (2012) Cardiac structure and function in normal pregnancy. Curr Opin Obstet Gynecol 24:413–421. 10.1097/GCO.0b013e328359826f [DOI] [PubMed] [Google Scholar]
- 33.Pieper PG, Hoendermis ES (2011) Pregnancy in women with pulmonary hypertension. Netherlands Hear J 19:504–508. 10.1007/s12471-0110219-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kinsella SM, Lohmann G (1994) Supine hypotensive syndrome. Obstet Gynecol 83:774–788 [PubMed] [Google Scholar]
- 35.Chesley LC (1972) Plasma and red cell volumes during pregnancy. Am J Obstet Gynecol 112:440–450. 10.1016/0002-9378(72)90493-0 [DOI] [PubMed] [Google Scholar]
- 36.Pritchard JA (1965) Changes in the Blood Volume During Pregnancy and Delivery. Anesthesiology 26:393–399. 10.1097/00000542-196507000-00004 [DOI] [PubMed] [Google Scholar]
- 37.Hytten FE, Paintin DB (1963) Increase in Plasma Volume During Normal Pregnancy. BJOG An Int J Obstet Gynaecol 70:402–407. 10.1111/j.14710528.1963.tb04922.x [DOI] [PubMed] [Google Scholar]
- 38.Feghali M, Venkataramanan R, Caritis S (2015) Pharmacokinetics of drugs in pregnancy. Semin Perinatol 39:512–519. 10.1053/j.semperi.2015.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Murphy MM, Scott JM, McPartlin JM, Fernandez-Ballart JD (2002) The pregnancy-related decrease in fasting plasma homocysteine is not explained by folic acid supplementation, hemodilution, or a decrease in albumin in a longitudinal study 1–3. Am J Clin Nutr 76:614–619 [DOI] [PubMed] [Google Scholar]
- 40.Masatoshi H, Yoshihiko U, Kazunori H, et al. (2002) Changes in urinary excretion of six biochemical parameters in normotensive pregnancy and preeclampsia. Am J Kidney Dis 39:392–400 [DOI] [PubMed] [Google Scholar]
- 41.Richter JE (2005) Review article: The management of heartburn in pregnancy. Aliment Pharmacol Ther 22:749–757. 10.1111/j.13652036.2005.02654.x [DOI] [PubMed] [Google Scholar]
- 42.Law R, Maltepe C, Bozzo P, Einarson A (2010) Treatment of heartburn and acid reflux associated with nausea and vomiting during pregnancy. Can Fam Physician 56:143–144 [PMC free article] [PubMed] [Google Scholar]
- 43.O’Brien B, Zhou Q (1995) Variables Related to Nausea and Vomiting During Pregnancy. Birth 22:93–100. 10.1111/j.1523-536X.1995.tb00566.x [DOI] [PubMed] [Google Scholar]
- 44.Pepper G V, Roberts SC (2006) Rates of nausea and vomiting in pregnancy and dietary characteristics across populations. Proc R Soc B Biol Sci 273:2675–2679. 10.1098/rspb.2006.3633 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Brandes JM (1967) First-trimester nausea and vomiting as related to outcome of pregnancy. Obstet Gynecol 30:427–31 [PubMed] [Google Scholar]
- 46.Quinlan JD, and Hill D (2003) Nausea and Vomiting of Pregnancy. Am Fam Physician 68:121–128 [PubMed] [Google Scholar]
- 47.Klebanoff MA, Koslow PA, Kaslow R, Rhoads GG (1985) Epidemiology of vomiting in early pregnancy. Obs Gynecol 66:612–6 [PubMed] [Google Scholar]
- 48.Ali RAR, Egan LJ (2007) Gastroesophageal reflux disease in pregnancy. Best Pract Res Clin Gastroenterol 21:793–806. 10.1016/j.bpg.2007.05.006 [DOI] [PubMed] [Google Scholar]
- 49.Parry E, Shields R, Turnbull AC (1970) Transit Time in the Small Intestine in Pregnancy. BJOG An Int J Obstet Gynaecol 77:900–901. 10.1111/j.1471-0528.1970.tb03423.x [DOI] [PubMed] [Google Scholar]
- 50.Macfie AG, Magides AD, Richmond MN, Reilly CS (1991) Gastric emptying in pregnancy. Br J Anaesth 67:54–57. 10.1093/bja/67.1.54 [DOI] [PubMed] [Google Scholar]
- 51.Levy DM, Williams OA, Magides AD, Reilly CS (1994) Gastric emptying is delayed at 8–12 weeks’ gestation. Br J Anaesth 73:237–238. 10.1093/bja/73.2.237 [DOI] [PubMed] [Google Scholar]
- 52.Sandhar BK, Elliott RH, Windram I, Rowbotham DJ (1992) Peripartum changes in gastric emptying. Anaesthesia 47:196–198. 10.1111/j.13652044.1992.tb02116.x [DOI] [PubMed] [Google Scholar]
- 53.Galinksy RE, Levy G (1984) Absorption and metabolism of acetaminophen shortly before parturition. Drug Intell Clin Pharm 18:977–979. 10.1177/106002808401801205 [DOI] [PubMed] [Google Scholar]
- 54.Skelin M, Lucijanić T, Amidžić Klarić D, et al. (2017) Factors Affecting Gastrointestinal Absorption of Levothyroxine: A Review. Clin Ther 39:378–403. 10.1016/j.clinthera.2017.01.005 [DOI] [PubMed] [Google Scholar]
- 55.Anderson GD, Carr DB (2009) Effect of pregnancy on the pharmacokinetics of antihypertensive drugs. Clin Pharmacokinet 48:159–168. 10.2165/00003088-200948030-00002 [DOI] [PubMed] [Google Scholar]
- 56.Tracy TS, Venkataramanan R, Glover DD, Caritis SN (2005) Temporal changes in drug metabolism (CYP1A2, CYP2D6 and CYP3A Activity) during pregnancy. Am J Obstet Gynecol 192:633–639. 10.1016/j.ajog.2004.08.030 [DOI] [PubMed] [Google Scholar]
- 57.Hodge LS, Tracy TS (2007) Alterations in drug disposition during pregnancy: Implications for drug therapy. Expert Opin Drug Metab Toxicol 3:557–571. 10.1517/17425225.3.4.557 [DOI] [PubMed] [Google Scholar]
- 58.Dempsey D, Jacob P III, Benowitz NL (2002) Accelerated metabolism of nicotine and cotinine in pregnant smokers. J Pharmacol Exp Ther 301:594–598. 10.1124/jpet.301.2.594 [DOI] [PubMed] [Google Scholar]
- 59.Tomson T, Lindbom U, Ekqvist B, Sundqvist A (1994) Disposition of Carbamazepine and Phenytoin in Pregnancy. Epilepsia 35:131–135. 10.1111/j.15281157.1994.tb02922.x [DOI] [PubMed] [Google Scholar]
- 60.Hebert MF, Easterling TR, Kirby B, et al. (2008) Effects of pregnancy on CYP3A and P-glycoprotein activities as measured by disposition of midazolam and digoxin: A University of Washington specialized center of research study. Clin Pharmacol Ther 84:248–253. 10.1038/clpt.2008.1 [DOI] [PubMed] [Google Scholar]
- 61.Tukey RH, Strassburg CP (2002) Human UDP-Glucuronosyltransferases: Metabolism, Expression, and Disease. Annu Rev Pharmacol Toxicol 40:581–616. 10.1146/annurev.pharmtox.40.1.581 [DOI] [PubMed] [Google Scholar]
- 62.Jeong H, Choi S, Song JW, et al. (2008) Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination. Xenobiotica 38:62–75. 10.1080/00498250701744633 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sibai BM, Mabie WC, Shamsa F, et al. (1990) A comparison of no medication versus methyldopa or labetalol in chronic hypertension during pregnancy. Am J Obstet Gynecol 162:960–967. 10.1016/0002-9378(90)91297-P [DOI] [PubMed] [Google Scholar]
- 64.Allegaert K, Peeters MY, Beleyn B, et al. (2015) Paracetamol pharmacokinetics and metabolism in young women. BMC Anesthesiol 15:. 10.1186/s12871015-0144-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Conrad KP, Davison JM (2014) The renal circulation in normal pregnancy and preeclampsia: Is there a place for relaxin? Am J Physiol Ren Physiol 306:F1121–35. 10.1152/ajprenal.00042.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Danielson LA, Conrad KP (1995) Acute blockade of nitric oxide synthase inhibits renal vasodilation and hyperfiltration during pregnancy in chronically instrumented conscious rats. J Clin Invest 96:482–490. 10.1172/JCI118059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Dunlop W (1981) Serial Changes in Renal Haemodynamics During Normal Human Pregnancy. BJOG An Int J Obstet Gynaecol 88:1–9. 10.1111/j.14710528.1981.tb00929.x [DOI] [PubMed] [Google Scholar]
- 68.Davison JM, Dunlop W (1984) Changes in renal hemodynamics and tubular function induced by normal human pregnancy. Semin Nephrol 4:198–207 [Google Scholar]
- 69.Frederice CP, Amaral E, De Oliveira Ferreira N (2013) Urinary symptoms and pelvic floor muscle function during the third trimester of pregnancy in nulliparous women. J Obstet Gynaecol Res 39:188–194 [DOI] [PubMed] [Google Scholar]
- 70.Nguyen MT, Maynard SE, Kimmel PL (2009) Misapplications of commonly used kidney equations: Renal physiology in practice. Clin J Am Soc Nephrol 4:528–534. 10.2215/CJN.05731108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Anderson GD (2005) Pregnancy-induced changes in pharmacokinetics: A mechanistic-based approach. Clin Pharmacokinet 44:989–1008. 10.2165/00003088-200544100-00001 [DOI] [PubMed] [Google Scholar]
- 72.Wesseloo R, Wierdsma AI, Van Kamp IL, et al. (2017) Lithium dosing strategies during pregnancy and the postpartum period. Br J Psychiatry 211:31–36. 10.1192/bjp.bp.116.192799 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Allegaert K, van Mieghem T, Verbesselt R, et al. (2009) Cefazolin pharmacokinetics in maternal plasma and amniotic fluid during pregnancy. Am J Obstet Gynecol 200:170.e1–170.e7. 10.1016/j.ajog.2008.08.067 [DOI] [PubMed] [Google Scholar]
- 74.Davison JM, Hytten FE (1975) the Effect of Pregnancy on the Renal Handling of Glucose. BJOG An Int J Obstet Gynaecol 82:374–381. 10.1111/j.14710528.1975.tb00652.x [DOI] [PubMed] [Google Scholar]
- 75.Roberts J, August P, Bakris G, et al. (2013) Hypertension in Pregnancy. Obstet Gynecol 122:1122–1131. 10.1097/01.AOG.0000437382.03963.88 [DOI] [PubMed] [Google Scholar]
- 76.Kattah A, Milic N, White W, Garovic V (2017) Spot urine protein measurements in normotensive pregnancies, pregnancies with isolated proteinuria and preeclampsia. Am J Physiol - Regul Integr Comp Physiol 313:R418–R424. 10.1152/ajpregu.00508.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Barron WM, Lindheimer MD (1985) Renal Function and Volume Homeostasis during Pregnancy In: Gleicher N, Buttino L, Elkayam U (eds) Principles of Medical Therapy in Pregnancy, 3rd editio. Appleton and Lange, Stanford, CT, pp 779–790 [Google Scholar]
- 78.Davison JM, Vallotton MB, Lindheimer MD (1981) Plasma Osmolality and Urinary Concentration and Dilution During and After Pregnancy: Evidence That Lateral Recumbency Inhibits Maximal Urinary Concentrating Ability. BJOG An Int J Obstet Gynaecol 88:472–479. 10.1111/j.1471-0528.1981.tb01019.x [DOI] [PubMed] [Google Scholar]
- 79.Bailey RR, Rolleston GL (1971) Kidney length and ureteric dilatation in the puerperium. J Obstet Gynaecol Br Commonw 78:55–61 [DOI] [PubMed] [Google Scholar]
- 80.Hertzberg BS, Carroll BA, Bowie JD, et al. (1993) Doppler US assessment of maternal kidneys: Analysis of intrarenal resistivity indexes in normal pregnancy and physiologic pelvicaliectasis. Radiology 186:689–692. 10.1148/radiology.186.3.8430175 [DOI] [PubMed] [Google Scholar]
- 81.Brown MA (1991) Urinary tract dilatation in pregnancy. Am J Obstet Gynecol 164:642–643. 10.1016/S0002-9378(11)80039-6 [DOI] [PubMed] [Google Scholar]
- 82.Rasmussen PE, Nielsen FR (1988) Hydronephrosis during pregnancy: a literature survey. Eur J Obstet Gynecol Reprod Biol 27:249–259. 10.1016/00282243(88)90130-X [DOI] [PubMed] [Google Scholar]
- 83.Berggren EK, Presley L, Amini SB, et al. (2015) Are the metabolic changes of pregnancy reversible in the first year postpartum? Diabetologia 58:1561–1568. 10.1007/s00125-015-3604-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Brelje TC, Scharp DW, Lacy PE, et al. (1993) Effect of homologous placental lactogens, prolactins, and growth hormones on islet b-cell division and insulin secretion in rat, mouse, and human islets: Implication for placental lactogen regulation of islet function during pregnancy. Endocrinology 132:879–887. 10.1210/endo.132.2.8425500 [DOI] [PubMed] [Google Scholar]
- 85.Ryan EA, Enns L (1988) Role of Gestational Hormones in the Induction of Insulin Resistance. J Clin Endocrinol Metab 67:341–347. 10.1210/jcem-67-2341 [DOI] [PubMed] [Google Scholar]
- 86.Butte NF (2000) Carbohydrate and lipid metabolism in pregnancy: Normal compared with gestational diabetes mellitus. Am J Clin Nutr 71: [DOI] [PubMed] [Google Scholar]
- 87.Butler AE, Cao-Minh L, Galasso R, et al. (2010) Adaptive changes in pancreatic beta cell fractional area and beta cell turnover in human pregnancy. Diabetologia 53:2167–2176. 10.1007/s00125-010-1809-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Angueira AR, Ludvik AE, Reddy TE, et al. (2015) New insights into gestational glucose metabolism: Lessons learned from 21st century approaches. Diabetes 64:327–334. 10.2337/db14-0877 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Callesen NF, Ringholm L, Stage E, et al. (2012) Insulin requirements in type 1 diabetic pregnancy: Do twin pregnant women require twice as much insulin as singleton pregnant women? Diabetes Care 35:1246–1248. 10.2337/dc11-2467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Herrera E, Ortega-Senovilla H (2014) Lipid Metabolism During Pregnancy and its Implications for Fetal Growth. Curr Pharm Biotechnol 15:24–31. 10.2174/1389201015666140330192345 [DOI] [PubMed] [Google Scholar]
- 91.Herrera E (2002) Lipid metabolism in pregnancy and its consequences in the fetus and newborn. Endocrine 19:43–55. 10.1385/ENDO:19:1:43 [DOI] [PubMed] [Google Scholar]
- 92.Tyson JE, Hwang P, Guyda H, Friesen HG (1972) Studies of prolactin secretion in human pregnancy. Am J Obstet Gynecol 113:14–20. 10.1016/00029378(72)90446-2 [DOI] [PubMed] [Google Scholar]
- 93.Molitch ME (2011) Prolactinoma in pregnancy. Best Pract Res Clin Endocrinol Metab 25:885–896. 10.1016/j.beem.2011.05.011 [DOI] [PubMed] [Google Scholar]
- 94.Scheithauer BW, Sano T, Kovacs KT, et al. (1990) The Pituitary Gland in Pregnancy: A Clinicopathologic and Immunohistochemical Study of 69 Cases. Mayo Clin Proc 65:461–474. 10.1016/S0025-6196(12)60946-X [DOI] [PubMed] [Google Scholar]
- 95.Feldt-Rasmussen U, Mathiesen ER (2011) Endocrine disorders in pregnancy: Physiological and hormonal aspects of pregnancy. Best Pract Res Clin Endocrinol Metab 25:875–884. 10.1016/j.beem.2011.07.004 [DOI] [PubMed] [Google Scholar]
- 96.Stalla GK, Bost H, Stalk J, et al. (1989) Human corticotropin-releasing hormone during pregnancy. Gynecol Endocrinol 3:1–10. 10.3109/09513598909152447 [DOI] [PubMed] [Google Scholar]
- 97.Sasaki A, Shinkawa O, Yoshinaga K (1989) Placental corticotropin-releasing hormone may be a stimulator of maternal pituitary adrenocorticotropic hormone secretion in humans. J Clin Invest 84:1997–2001. 10.1172/JCI114390 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Rees LH, Burke CW, Chard T, et al. (1975) Possible placental origin of ACTH in normal human pregnancy. Nature 254:620–622. 10.1038/254620b0 [DOI] [PubMed] [Google Scholar]
- 99.Demey-Ponsart E, Foidart JM, Sulon J, Sodoyez JC (1982) Serum CBG, free and total cortisol and circadian patterns of adrenal function in normal pregnancy. J Steroid Biochem 16:165–169 [DOI] [PubMed] [Google Scholar]
- 100.Carr BR, Parker CR, Madden JD, et al. (1981) Maternal plasma adrenocorticotropin and cortisol relationships throughout human pregnancy. Am J Obstet Gynecol 139:416–422. 10.1016/0002-9378(81)90318-5 [DOI] [PubMed] [Google Scholar]
- 101.Glinoer D, Gershengorn MC, Dubois A, Robbins J (1977) Stimulation of thyroxine-binding globulin synthesis by isolated rhesus monkey hepatocytes after in vivo βestradiol administration. Endocrinology 100:807–813. 10.1210/endo100-3-807 [DOI] [PubMed] [Google Scholar]
- 102.Ain KB, Mori Y, Refetoff S (1987) Reduced clearance rate of thyroxine-binding globulin (TBG) with increased sialylation: A mechanism for estrogen-induced elevation of serum TBG concentration. J Clin Endocrinol Metab 65:689–696. 10.1210/jcem-65-4-689 [DOI] [PubMed] [Google Scholar]
- 103.Henrichs J, Ghassabian A, Peeters RP, Tiemeier H (2013) Maternal hypothyroxinemia and effects on cognitive functioning in childhood: How and why? Clin Endocrinol (Oxf) 79:152–162. 10.1111/cen.12227 [DOI] [PubMed] [Google Scholar]
- 104.Casey BM, Thom EA, Peaceman AM, et al. (2017) Treatment of subclinical hypothyroidism or hypothyroxinemia in pregnancy. N Engl J Med 376:815–825. 10.1056/NEJMoa1606205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.(2015) Practice Bulletin No. 148: Thyroid disease in pregnancy. Obstet Gynecol 125:996–1005. 10.1097/01.AOG.0000462945.27539.93 [DOI] [PubMed] [Google Scholar]
- 106.Alexander EK, Pearce EN, Brent GA, et al. (2017) 2017 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease during Pregnancy and the Postpartum. Thyroid 27:315–389 [DOI] [PubMed] [Google Scholar]
- 107.Moleti M, Trimarchi F, Vermiglio F (2014) Thyroid physiology in pregnancy. Endocr Pract 20:589–596. 10.4158/EP13341.RA [DOI] [PubMed] [Google Scholar]
- 108.Glinoer D (1999) What happens to the normal thyroid during pregnancy? Thyroid 9:631–635. 10.1089/thy.1999.9.631 [DOI] [PubMed] [Google Scholar]
- 109.Alexander EK, Marqusee E, Lawrence J, et al. (2004) Timing and magnitude of increases in levothyroxine requirements during pregnancy in women with hypothyroidism. N Engl J Med 351:241–249+310. 10.1056/NEJMoa040079 [DOI] [PubMed] [Google Scholar]
- 110.Bernstein IM, Ziegler W, Badger GJ (2001) Plasma Volume Expansion in Early Pregnancy. Obstet Gynecol 97:669–672 [DOI] [PubMed] [Google Scholar]
- 111.Lund CJ, Donovan JC (1967) Blood volume during pregnancy. Significance of plasma and red cell volumes. Am J Obstet Gynecol 98:393–403. 10.1016/00029378(67)90160-3 [DOI] [PubMed] [Google Scholar]
- 112.de Haas S, Ghossein-Doha C, van Kuijk SMJ, et al. (2017) Physiological adaptation of maternal plasma volume during pregnancy: a systematic review and meta-analysis. Ultrasound Obstet Gynecol 49:177–187. 10.1002/uog.17360 [DOI] [PubMed] [Google Scholar]
- 113.Goonewardene M, Shehata M, Hamad A (2012) Anaemia in pregnancy. Best Pract Res Clin Obstet Gynaecol 26:3–24. 10.1016/j.bpobgyn.2011.10.010 [DOI] [PubMed] [Google Scholar]
- 114.Huisman A, Aarnoudse JG, Huisjes HJ, et al. (1987) Whole blood viscosity during normal pregnancy. BJOG An Int J Obstet Gynaecol 94:1143–1149. 10.1111/j.1471-0528.1987.tb02313.x [DOI] [PubMed] [Google Scholar]
- 115.(2008) ACOG practice bulletin no. 95: Anemia in pregnancy. Obstet Gynecol 112:201–207. 10.1097/AOG.0b013e3181809c0d [DOI] [PubMed] [Google Scholar]
- 116.Haider BA, Olofin I, Wang M, et al. (2013) Anaemia, prenatal iron use, and risk of adverse pregnancy outcomes: Systematic review and meta-analysis. BMJ 347:. 10.1136/bmj.f3443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Peña-Rosas J, De-Regil L, Dowswell T, Viteri F (2015) Daily oral iron supplementation during pregnancy. Cochrane Database Syst Rev N.PAG-N.PAG. 10.1002/14651858.CD004736.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ahmadzia HK, Phillips JM, James AH, et al. (2018) Predicting peripartum blood transfusion in women undergoing cesarean delivery: A risk prediction model. PLoS One 13:. 10.1371/journal.pone.0208417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kühnert M, Strohmeier R, Stegmüller M, Halberstadt E (1998) Changes in lymphocyte subsets during normal pregnancy. Eur J Obstet Gynecol Reprod Biol 76:147–151. 10.1016/S0301-2115(97)00180-2 [DOI] [PubMed] [Google Scholar]
- 120.Boehlen F, Hohlfeld P, Extermann P, et al. (2000) Platelet count at term pregnancy: A reappraisal of the threshold. Obstet Gynecol 95:29–33. 10.1016/S00297844(99)00537-2 [DOI] [PubMed] [Google Scholar]
- 121.Han L, Liu X, Li H, et al. (2014) Blood coagulation parameters and platelet indices: Changes in normal and preeclamptic pregnancies and predictive values for preeclampsia. PLoS One 9:. 10.1371/journal.pone.0114488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Maymon R, Zimerman AL, Strauss S, Gayer G (2007) Maternal Spleen Size Throughout Normal Pregnancy. Semin Ultrasound, CT MRI 28:64–66. 10.1053/j.sult.2006.10.005 [DOI] [PubMed] [Google Scholar]
- 123.Kenny LC, McCrae KR, Gary Cunningham F (2014) Platelets, coagulation, and the liver In: Taylor RN, Roberts JM, Cunningham FG, Lindheimer MDBT-CHD in P (Fourth E (eds) Chesley’s Hypertensive Disorders in Pregnancy, Fourth Edition, 4th editio. Academic Press, San Diego, pp 379–396 [Google Scholar]
- 124.Uchikova EH, Ledjev II (2005) Changes in haemostasis during normal pregnancy. Eur J Obstet Gynecol Reprod Biol 119:185–188. 10.1016/j.ejogrb.2004.06.038 [DOI] [PubMed] [Google Scholar]
- 125.McLean KC, Bernstein IM, Brummel-Ziedins KE (2012) Tissue factor-dependent thrombin generation across pregnancy. Am J Obstet Gynecol 207:135.e1–135.e6. 10.1016/j.ajog.2012.05.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Sharief LT, Lawrie AS, Mackie IJ, et al. (2014) Changes in factor XIII level during pregnancy. Haemophilia 20:. 10.1111/hae.12345 [DOI] [PubMed] [Google Scholar]
- 127.Gary Cunningham F, Nelson DB (2015) Disseminated intravascular coagulation syndromes in obstetrics. Obstet Gynecol 126:999–1011 [DOI] [PubMed] [Google Scholar]
- 128.Hellgren M (1996) Hemostasis During Pregnancy And Puerperium. Pathophysiol Haemost Thromb 26:244–247. 10.1159/000217305 [DOI] [PubMed] [Google Scholar]
- 129.Hayes M, Bourjeily G, Rosene-Montella K (2009) Venous thromboembolic disease and pregnancy [10]. N Engl J Med 360:639. [PubMed] [Google Scholar]
- 130.Cantwell R, Clutton-Brock T, Cooper G, et al. (2011) Saving Mothers’ Lives: Reviewing maternal deaths to make motherhood safer: 2006–2008. The Eighth Report of the Confidential Enquiries into Maternal Deaths in the United Kingdom. BJOG 118 Suppl:1–203. 10.1111/j.1471-0528.2010.02847.x [DOI] [PubMed] [Google Scholar]
- 131.James AH, Jamison MG, Brancazio LR, Myers ER (2006) Venous thromboembolism during pregnancy and the postpartum period: Incidence, risk factors, and mortality. Am J Obstet Gynecol 194:1311–1315. 10.1016/j.ajog.2005.11.008 [DOI] [PubMed] [Google Scholar]
- 132.James AH (2012) The natural history of pelvic vein thrombosis: The natural history of involution? Am J Obstet Gynecol 206:276–277. 10.1016/j.ajog.2012.01.013 [DOI] [PubMed] [Google Scholar]
- 133.James AH, Rhee E, Thames B, Philipp CS (2014) Characterization of antithrombin levels in pregnancy. Thromb Res 134:648–651. 10.1016/j.thromres.2014.07.025 [DOI] [PubMed] [Google Scholar]
- 134.Patel JP, Green B, Patel RK, et al. (2013) Population pharmacokinetics of enoxaparin during the antenatal period. Circulation 128:1462–1469. 10.1161/CIRCULATIONAHA.113.003198 [DOI] [PubMed] [Google Scholar]
- 135.Jensen D, Duffin J, Lam YM, et al. (2008) Physiological mechanisms of hyperventilation during human pregnancy. Respir Physiol Neurobiol 161:76–86. 10.1016/j.resp.2008.01.001 [DOI] [PubMed] [Google Scholar]
- 136.Lyons H, Antonio R (1959) The sensitivity of the respiratory center in pregnancy and after administration of progesterone. Trans Assoc Am Physicians 72:173–180 [Google Scholar]
- 137.LoMauro A, Aliverti A (2015) Respiratory physiology of pregnancy. Breathe 11:297–301. 10.1183/20734735.008615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ellegrad E (2006) Pregnancy rhinitis. Immunol Allergy Clin N Am 26:119–135 [DOI] [PubMed] [Google Scholar]
- 139.Toppozada H, Michaels L, Toppozada M, et al. (1982) The human respiratory nasal mucosa in pregnancy. An electron microscopic and histochemical study. J Laryngol Otol 96:613–26 [DOI] [PubMed] [Google Scholar]
- 140.Weinberger SE, Weiss ST, Cohen WR, et al. (1980) Pregnancy and the lung. Am Rev Respir Dis 121:559–581. 10.1164/arrd.1980.121.3.559 [DOI] [PubMed] [Google Scholar]
- 141.Gilroy RJ, Mangura BT, Lavietes MH (1988) Rib cage and abdominal volume displacements during breathing in pregnancy. Am Rev Respir Dis 137:668–672. 10.1164/ajrccm/137.3.668 [DOI] [PubMed] [Google Scholar]
- 142.Goldsmith LT, Weiss G, Steinetz BG (1995) Relaxin and its role in pregnancy. Endocrinol Metab Clin North Am 24:171–186 [PubMed] [Google Scholar]
- 143.Marx GF, Murthy PK, Orkin LR (1970) Static compliance before and after vaginal delivery. Br J Anaesth 42:1100–1104. 10.1093/bja/42.12.1100 [DOI] [PubMed] [Google Scholar]
- 144.Gee JB, Packer BS, Millen JE, Robin ED (1967) Pulmonary mechanics during pregnancy. J Clin Invest 46:945–952. 10.1172/JCI105600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Tenholder MF, South-Paul JE (1989) Dyspnea in pregnancy. Chest 96:381–388. 10.1378/chest.96.2.381 [DOI] [PubMed] [Google Scholar]
- 146.Prowse CM, Gaensler EA (1965) Respiratory and Acid-Base Changes During Pregnancy. Anesthesiology 26:381–392. 10.1097/00000542196507000-00003 [DOI] [PubMed] [Google Scholar]
- 147.Crapo RO (1996) Normal cardiopulmonary physiology during pregnancy. Clin Obstet Gynecol 39:3–16 [DOI] [PubMed] [Google Scholar]
- 148.Costantine MM (2014) Physiologic and pharmacokinetic changes in pregnancy. Front Pharmacol 5 APR: 10.3389/fphar.2014.00065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Cugell DW, Frank NR, Gaensler EA, Badger TL (1953) Pulmonary function in pregnancy. I. serial observations in normal women. Obstet Gynecol Surv 8:658–662 [DOI] [PubMed] [Google Scholar]
- 150.Siddiqui AH, Tauheed N, Ahmad A, Mohsin Z (2014) Pulmonary function in advanced uncomplicated singleton and twin pregnancy. J Bras Pneumol 40:244–249. 10.1590/s1806-37132014000300007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Templeton A, Kelman GR (1976) Maternal blood-gases, (PAO2-PaO2), physiological shunt and VD/VT in normal pregnancy. Br J Anaesth 48:1001–1004. 10.1093/bja/48.10.1001 [DOI] [PubMed] [Google Scholar]
- 152.Liberatore SM, Pistelli R, Patalano F, et al. (1984) Respiratory function during pregnancy. Respiration 46:145–150. 10.1159/000194683 [DOI] [PubMed] [Google Scholar]
- 153.Awe RJ, Brooke Nicotra M, Newsom TD, Viles R (1979) Arterial oxygenation and alveolar-arterial gradients in term pregnancy. Obstet Gynecol 53:182–186 [PubMed] [Google Scholar]
- 154.Ueki R, Tatara T, Kariya N, et al. (2009) Comparison of placental transfer of local anesthetics in perfusates with different pH values in a human cotyledon model. J Anesth 23:526–529. 10.1007/s00540-009-0815-7 [DOI] [PubMed] [Google Scholar]
- 155.Johnson RF, Herman NL, Johnson HV, et al. (1996) Effects of fetal pH on local anesthetic transfer across the human placenta. Anesthesiology 85:608–615. 10.1097/00000542-199609000-00021 [DOI] [PubMed] [Google Scholar]
- 156.Lucius H, Gahlenbeck H, Kleine HO, et al. (1970) Respiratory functions, buffer system, and electrolyte concentrations of blood during human pregnancy. Respir Physiol 9:311–317. 10.1016/0034-5687(70)90088-5 [DOI] [PubMed] [Google Scholar]
- 157.Tsai CH, de Leeuw NKM (1982) Changes in 2, 3-diphosphoglyeerate during pregnancy and puerperium in normal women and in β-thalassemia heterozygous women. Am J Obstet Gynecol 142:520–523. 10.1016/00029378(82)90754-2 [DOI] [PubMed] [Google Scholar]
- 158.Gin T, Mainland P, Chan MTV, Short TG (1997) Decreased thiopental requirements in early pregnancy. Anesthesiology 86:73–78. 10.1097/00000542199701000-00011 [DOI] [PubMed] [Google Scholar]
- 159.Nejdlova M, Johnson T (2012) Anaesthesia for non-obstetric procedures during pregnancy. Contin Educ Anaesth Crit Care Pain 12:203–206. [Google Scholar]
- 160.Koren G, Ornoy A (2018) The role of the placenta in drug transport and fetal drug exposure. Expert Rev Clin Pharmacol 11:373–385. 10.1080/17512433.2018.1425615 [DOI] [PubMed] [Google Scholar]
- 161.Vargesson N (2015) Thalidomide-induced teratogenesis: History and mechanisms. Birth Defects Res Part C - Embryo Today Rev 105:140–156. 10.1002/bdrc.21096 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Joshi AA, Vaidya SS, St-Pierre M V., et al. (2016) Placental ABC Transporters: Biological Impact and Pharmaceutical Significance. Pharm Res 33:2847–2878. 10.1007/s11095-016-2028-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Desforges M, Sibley CP (2010) Placental nutrient supply and fetal growth. Int J Dev Biol 54:377–390. 10.1387/ijdb.082765md [DOI] [PubMed] [Google Scholar]
- 164.Pacifici GM, Nottoli R (1995) Placental Transfer of Drugs Administered to the Mother. Clin Pharmacokinet 28:235–269. 10.2165/00003088-199528030-00005 [DOI] [PubMed] [Google Scholar]
- 165.Cardonick E, Iacobucci A (2004) Use of chemotherapy during human pregnancy. Lancet Oncol 5:283–291. 10.1016/S1470-2045(04)01466-4 [DOI] [PubMed] [Google Scholar]
- 166.Andrew M, Boneu B, Cade J, et al. (1985) Placental transport of low molecular weight heparin in the pregnant sheep. Br J Haematol 59:103–108. 10.1111/j.1365-2141.1985.tb02969.x [DOI] [PubMed] [Google Scholar]
- 167.Jovanovic L, Pettitt DJ (2007) Treatment with insulin and its analogs in pregnancies complicated by diabetes. Diabetes Care 30:. 10.2337/dc07-s220 [DOI] [PubMed] [Google Scholar]
- 168.Pentsuk N, Van Der Laan JW (2009) An interspecies comparison of placental antibody transfer: New insights into developmental toxicity testing of monoclonal antibodies. Birth Defects Res Part B - Dev Reprod Toxicol 86:1–17 [DOI] [PubMed] [Google Scholar]
- 169.Tun GSZ, Lobo AJ (2015) Evaluation of pharmacokinetics and pharmacodynamics and clinical efficacy of certolizumab pegol for Crohn’s disease. Expert Opin Drug Metab Toxicol 11:317–327. 10.1517/17425255.2015.995166 [DOI] [PubMed] [Google Scholar]
- 170.Clowse MEB, Wolf DC, Förger F, et al. (2015) Pregnancy outcomes in subjects exposed to certolizumab pegol. J Rheumatol 42:2270–2278. 10.3899/jrheum.140189 [DOI] [PubMed] [Google Scholar]
- 171.Staelens AS, Vonck S, Molenberghs G, et al. (2016) Maternal body fluid composition in uncomplicated pregnancies and preeclampsia: a bioelectrical impedance analysis. Eur J Obstet Gynecol Reprod Biol 204:69–73. 10.1016/j.ejogrb.2016.07.502 [DOI] [PubMed] [Google Scholar]
- 172.Kreepala C, Kitporntheranunt M, Sangwipasnapaporn W, et al. (2018) Assessment of preeclampsia risk by use of serum ionized magnesium-based equation. Ren Fail 40:99–106. 10.1080/0886022X.2017.1422518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Lopes van Balen VA, Gansewinkel TAG, Haas S, et al. (2019) Maternal kidney function during pregnancy: systematic review and meta analysis. Ultrasound Obstet Gynecol 54:297–307. 10.1002/uog.20137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Datta S, Kodali BS, Segal S (2010) Maternal Physiological Changes During Pregnancy, Labor, and the Postpartum Period In: Obstetric Anesthesia Handbook: Fifth Edition. Springer New York, New York, NY, pp 1–14 [Google Scholar]
- 175.Robson SC, Dunlop W, Moore M, Hunter S (1987) Haemodynamic changes during the puerperium: a Doppler and M mode echocardiographic study. BJOG An Int J Obstet Gynaecol 94:1028–1039. 10.1111/j.1471-0528.1987.tb02286.x [DOI] [PubMed] [Google Scholar]
- 176.Rachael JP, Nelson-Piercy C (2004) Management of hypertension before, during, and after pregnancy. Heart 90:1499–1504 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Schalkwijk S, Buaben AO, Freriksen JJM, et al. (2018) Prediction of Fetal Darunavir Exposure by Integrating Human Ex-Vivo Placental Transfer and Physiologically Based Pharmacokinetic Modeling. Clin Pharmacokinet 57:705–716. 10.1007/s40262-017-0583-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Liu XI, Momper JD, Rakhmanina N, et al. (2019) Physiologically Based Pharmacokinetic Models to Predict Maternal Pharmacokinetics and Fetal Exposure to Emtricitabine and Acyclovir. J Clin Pharmacol. 10.1002/jcph.1515 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Horton S, Tuerk A, Cook D, et al. (2012) Maximum recommended dosage of lithium for pregnant women based on a PBPK model for lithium absorption. Adv Bioinformatics 2012:. 10.1155/2012/352729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Mian P, van den Anker JN, van Calsteren K, et al. (2019) Physiologically Based Pharmacokinetic Modeling to Characterize Acetaminophen Pharmacokinetics and N-Acetyl-p-Benzoquinone Imine (NAPQI) Formation in Non-Pregnant and Pregnant Women. Clin Pharmacokinet. 10.1007/s40262-019-00799-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Darakjian LI, Kaddoumi A (2019) Physiologically Based Pharmacokinetic/Pharmacodynamic Model for Caffeine Disposition in Pregnancy. Mol Pharm 16:1340–1349. 10.1021/acs.molpharmaceut.8b01276 [DOI] [PubMed] [Google Scholar]
- 182.Lumen A, McNally K, George N, et al. (2015) Quantitative global sensitivity analysis of a biologically based dose-response pregnancy model for the thyroid endocrine system. Front Pharmacol 6:. 10.3389/fphar.2015.00107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Jarvis S, Nelson-Piercy C (2014) Common symptoms and signs during pregnancy. Obstet Gynaecol Reprod Med 24:245–249. 10.1016/j.ogrm.2014.05.006 [DOI] [Google Scholar]
- 184.Tan EK, Tan EL (2013) Alterations in physiology and anatomy during pregnancy. Best Pract Res Clin Obstet Gynaecol 27:791–802. 10.1016/j.bpobgyn.2013.08.001 [DOI] [PubMed] [Google Scholar]
- 185.Philipson A (1977) Pharmacokinetics of ampicillin during pregnancy. J Infect Dis 136:370–376. 10.1093/infdis/136.3.370 [DOI] [PubMed] [Google Scholar]
- 186.Philipson A, Stiernstedt G, Ehrnebo M (1987) Comparison of the Pharmacokinetics of Cephradine and Cefazolin in Pregnant and Non-Pregnant Women. Clin Pharmacokinet 12:136–144. 10.2165/00003088-198712020-00004 [DOI] [PubMed] [Google Scholar]
- 187.Dungan JS (2009) Changes in Enoxaparin Pharmacokinetics During Pregnancy and Implications for Antithrombotic Therapeutic Strategy. Yearb Obstet Gynecol Women’s Heal 2009:88–89. 10.1016/s1090-798x(09)79028-9 [DOI] [PubMed] [Google Scholar]
- 188.Heikkinen T, Ekblad U, Palo P, Laine K (2003) Pharmacokinetics of fluoxetine and norfluoxetine in pregnancy and lactation. Clin Pharmacol Ther 73:330–337. 10.1016/S0009-9236(02)17634-X [DOI] [PubMed] [Google Scholar]
- 189.Pennell PB (2010) Erratum: The impact of pregnancy and childbirth on the metabolism of lamotrigine (Neurology (2004) 62 (292–295)). Neurology 74:2028 10.1212/WNL.0b013e3181e78c1d [DOI] [PubMed] [Google Scholar]
- 190.Chopra IJ, Baber K (2003) Treatment of primary hypothyroidism during pregnancy: is there an increase in thyroxine dose requirement in pregnancy? Metabolism 52:122–128. 10.1053/meta.2003.50019 [DOI] [PubMed] [Google Scholar]
- 191.Fischer JH, Sarto GE, Hardman J, et al. (2014) Influence of gestational age and body weight on the pharmacokinetics of labetalol in pregnancy. Clin Pharmacokinet 53:373–383. 10.1007/s40262-013-0123-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Ter Laak MA, Roos C, Ouw DJ, et al. (2015) Pharmacokinetics of nifedipine slowrelease tablets during sustained tocolysis. Int J Clin Pharmacol Ther 53:84–91. 10.5414/CP202215 [DOI] [PubMed] [Google Scholar]
- 193.Dallmann A, Ince I, Meyer M, et al. (2017) Gestation-Specific Changes in the Anatomy and Physiology of Healthy Pregnant Women: An Extended Repository of Model Parameters for Physiologically Based Pharmacokinetic Modeling in Pregnancy. Clin Pharmacokinet 56:1303–1330. 10.1007/s40262-017-0539-z [DOI] [PubMed] [Google Scholar]