Abstract
The treatment of immune thrombocytopenia (ITP) in adults has evolved rapidly over the past decade. The second-generation thrombopoietin receptor agonists (TPO-RAs), romiplostim, eltrombopag, and avatrombopag are approved for the treatment of chronic ITP in adults. However, their use in pregnancy is labeled as category C by the United States Food and Drug Administration (FDA) due to the lack of clinical data on human subjects. ITP is a common cause of thrombocytopenia in the first and second trimester of pregnancy, which not only affects the mother but can also lead to thrombocytopenia in the neonatal thrombocytopenia secondary to maternal immune thrombocytopenia (NMITP). Corticosteroids, intravenous immunoglobulins (IVIGs) are commonly used for treating acute ITP in pregnant patients. Drugs such as rituximab, anti-D, and azathioprine that are used to treat ITP in adults, are labeled category C and seldom used in pregnant patients. Cytotoxic chemotherapy (vincristine, cyclophosphamide), danazol, and mycophenolate are contraindicated in pregnant women. In such a scenario, TPO-RAs present an attractive option to treat ITP in pregnant patients. Current evidence on the use of TPO-RAs in pregnant women with ITP is limited. In this narrative review, we will examine the preclinical and the clinical literature regarding the use of TPO-RAs in the management of ITP in pregnancy and their effect on neonates with NMITP.
Keywords: avatrombopag, eltrombopag, immune thrombocytopenia, pregnancy, romiplostim, thrombopoietin receptor agonists
Introduction
Thrombocytopenia, defined as a platelet count less than 150,000/µL, can occur in up to 7–11% of pregnancies due to multiple reasons (Table 1).1–5 Immune thrombocytopenia (ITP), defined as a platelet count less than 100,000/µL, is a common cause of thrombocytopenia in the first and second trimester of pregnancy and accounts for 1–4% of all causes of pregnancy-associated thrombocytopenia.2,3,6–8 Despite significant advances made in the treatment options for adult ITP, the management of pregnant patients presenting with acute ITP remains a challenge. Although all pregnant patients with ITP may not require treatment, when needed, corticosteroids and intravenous immunoglobulins (IVIGs) are the most widely used medications for treating such patients.3,6,9 Steroids take 3–7 days to achieve a response and nearly 2–3 weeks to achieve the maximum effect.10 In pregnant women, prednisone is preferred over dexamethasone due to a lower risk of transfer via the placenta. Also, dexamethasone confers a higher risk of oligohydramnios and facial deformities in the fetus.1,11 IVIG achieves a rapid and effective response in ~80% of patients. However, the response is brief and is lost within a few weeks in most patients.12 Splenectomy is safe during pregnancy; however, it is a more radical option and is rarely pursued.13,14 Intravenous anti-D in Rh(D)-positive women with intact spleen has demonstrated benefit in small pilot studies; however, its use is associated with the risk of maternal and fetal hemolysis.15 Rituximab is labeled as category C by the United States (US) Food and Drug Administration (FDA) for use during pregnancy and is recommended only for very severe cases.14 Likewise, azathioprine is another drug that can be used for ITP during pregnancy; however, it is also labelled category C by the US FDA.14 Vinca alkaloids, mycophenolate, and danazol are teratogenic and are contraindicated in pregnancy.14
Table 1.
Causes of thrombocytopenia in pregnancy | Incidence | Percentage cause of thrombocytopenia in pregnancy |
---|---|---|
Gestational thrombocytopenia | 4.4–11.6% of all pregnancies (2nd/3rd trimester most common) | 75% |
Pregnancy-specific TMA | ||
Pre-eclampsia | 2–8% of all pregnancies (2nd/3rd trimester) | 20% |
HELLP syndrome | 0.5–0.9% of all pregnancies | |
Acute fatty liver of pregnancy | 1 in 5000–10,000 pregnancies | <1% |
Immune thrombocytopenia | 1–2/1000 pregnancies (1st/2nd trimester most common) | 3–4% |
Non-pregnancy-specific TMA | ||
Thrombotic thrombocytopenic purpura | 1 in 200,000 pregnancies | <1% |
Hemolytic uremic syndrome | Extremely rare | |
Atypical hemolytic uremic syndrome | 1 in 25,000 pregnancies | |
Disseminated intravascular coagulation (due to obstetric catastrophes (abruptio placentae, placenta previa) | Uncommon | |
Hereditary thrombocytopenia | Extremely rare | |
Others | ||
Infection | Rare | <1% |
Disseminated intravascular coagulation | ||
Type IIB von Willebrand disease | ||
Paroxysmal nocturnal hemoglobinuria | ||
Bone marrow failure syndromes | ||
Aplastic anemia, MDS, MPN, leukemia, lymphoma, infiltrative disorders |
HELLP, hemolysis, elevated liver enzymes and low platelets; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasm; TMA, thrombotic microangiopathy.
Thrombopoietin receptor agonists (TPO-RAs) act as thrombopoietin (TPO) mimetics to stimulate megakaryopoiesis in the bone marrow and yield an overall response rate of ~80%.16,17 TPO is a cytokine that regulates megakaryocyte and platelet production via the receptor called myeloproliferative leukemia protein (MPL) or CD110.18 Thrombopoietin receptor (TPO-R) is a 635 amino acid long protein with an extracellular, transmembrane, and an intracellular or cytoplasmic domain that is expressed on the surface of megakaryocytes, platelets, hemangioblasts, and hematopoietic stem cells (HSCs).18–20 TPO is produced mainly in the hepatocyte either via the stimulation of the Ashwell–Morell receptor by old de-sialylated platelets;21 or via direct stimulation by interleukin (IL) 6.22 Unlike erythropoietin, hepatocytes keep producing TPO with no regulation of gene expression.23 TPO-R plays a critical role in balancing the levels of TPO by binding, internalizing, and then degrading the excess TPO in circulation.24,25 Platelets and megakaryocytes express the bulk of TPO-R. Their large numbers degrade excessive TPO, hence maintain a negative feedback loop to prevent its action on progenitor stem cells.24,26 In patients with thrombocytopenia, high levels of TPO are noted due to insufficient platelet mass.27 On the contrary, in patients with ITP, the TPO levels remain inappropriately normal or slightly elevated.28–31 Although decreased production of TPO, increased degradation, immune interference with TPO, or increased clearance along with antibody-bearing platelets, or megakaryocytes have been described in the literature, the mechanism for low to normal TPO levels in patients with ITP remains.32
TPO-RA is labeled as ‘category C’ by the US FDA, which means that animal studies have shown adverse effects on the fetus, but no adequate or well-controlled studies are present in humans. As a result, none of the trials exploring the use of TPO-RAs in adult patients with ITP include pregnant patients or lactating mothers. The recent guidelines from the American Society of Hematology and International Working Group also do not promote the use of TPO-RAs in pregnant patients.13,14 The current literature demonstrating the use of TPO-RAs in pregnancy is limited to off-label use in case reports and case series.33–42 Eltrombopag (Promacta), Avatrombopag (Doptelet), and Romiplostim (Nplate) are approved by the US FDA for adult patients with ITP.43,44 Studies of TPO-agonist antibodies [Minibodies (VB22B sc(Fv)2) and domain subclass-converted TPO agonist antibodies (MA01G4G344)] have not been reported as of yet. In this narrative review, we will review the preclinical and clinical literature associated with the use of TPO-RA in pregnancy and its impact on neonatal thrombocytopenia secondary to maternal immune thrombocytopenia (NMITP). A literature search was conducted using the PubMed electronic database from 1950 to 2018. The MeSH heading and/or text words ‘Romiplostim’, ‘Eltrombopag’, ‘Avatrombopag’, ‘Purpura’, ‘thrombocytopenia’, ‘idiopathic’, ‘thrombopoietin receptor agonists,’ ‘Pregnancy’, and ‘Pregnant’ were used. We also searched the Google Scholar database for any additional reports not listed in the PubMed database. In addition to this, the US FDA website and individual drug websites (Nplate, Promacta, Doptelet) were also searched for references. Finally, the bibliographies of all retrieved articles were examined for additional relevant citations.
TPO-RAs: preclinical data in pregnancy
Eltrombopag
Eltrombopag, developed under the name SB497115, is a non-peptide TPO mimetic belonging to the bioarylhydrazone class of compounds with an empiric formula of C25H22N4O4 and molecular weight of 445.2 D.45 It has a metal chelation group in the center that binds to the TPO receptor causing phosphorylation and activation of the Janus kinase-2 (JAK-2), signal transducer and activator of transcription 5 (STAT5), mitogen-activated protein kinase (MAPK), and phosphoinositide-3 kinase (PI3K) pathway.45 It is administered orally at a dose of 50–75 mg per day, achieves a peak concentration in 2–6 h, and has a half-life of 26–35 h in patients with ITP. It is excreted primarily in the feces, followed by urine. In clinical trials, eltrombopag showed a dose-dependent increase in platelet count by day 8–10 in healthy subjects.45 Eltrombopag shows species specificity and is active only in humans and chimps.46 Fetal toxicity in animals was seen only at doses whose equivalent doses would be toxic for human beings (Table 2).47 However, no significant maternal–fetal toxicity was noted at doses that were equivalent to doses currently recommended in humans.47–49 Lactation studies in rats showed the continued presence of the drug in the F1 pup even after 22 h of commencing lactation, indicating that it was secreted in the milk.47 A registry study in humans to observe the effects of eltrombopag in lactating mothers was launched but failed to recruit any patients in 5 years and was subsequently withdrawn (NCT01055600).50 Eltrombopag did not affect male fertility in rats even at three times the human equivalent doses.50 In addition to this, eltrombopag did not compete with TPO and hence had additive effects in combination with the native TPO.46 Off-target toxicity in the form of cataracts was noted in young rodents when exposed to a dose that was four times the average human exposure.51 Similar toxicity was also noted in human clinical trials; however, the patients in whom cataracts developed also had numerous other risk factors that predisposed them to develop cataracts (such as prior chronic treatments or use of steroids).48,51
Table 2.
Dose in animals (mg/kg) | Human equivalent dose for ITP | Maternal toxicity | Fetal toxicity | |
---|---|---|---|---|
Eltrombopag | ||||
Embryo-fetal developmental study | ||||
Rats | 10 | 0.8 | None | None |
20 | 2.0 | NOAEL | NOAEL | |
60 | 6.0 | 1. Pre-post transplantation fetal loss | 1. Reduced fetal body weight (6–7%) | |
2. Reduced gravid uterus weight | 2. Few had small cervical ribs | |||
3. Other maternal toxicity | ||||
Rabbits | ||||
30 | 0.04 | None | None | |
80 | 0.3 | None | None | |
150 | 0.5 | None | None | |
Lactation data (pre and postnatal developmental toxicity) | ||||
Rats | ||||
Day 6–20 post gestation | 20 | 2 | No change in reproductive function | Eltrombopag detected in serum of pup |
Romiplostim | ||||
Embryo-fetal developmental study | ||||
Rat (administered every 2nd day) | ||||
10 | 0.07 | None | None | |
30 | 0.8 | None | None | |
100 | 3 | 1. Post implantation loss | 1. Still born pup 14% incidence in dams without drug-neutralizing antibodies | |
2. Less maternal weight gain (by 8%) | ||||
Rabbit (administered every 2nd day) | ||||
10 | 1.2 | None | None | |
30 | 5 | None | None | |
100 | 22 | 1. Reduced weight gain (by 50%) | Multiple malformation in fetus (gastroschisis, ectrodactyly and cutis aplasia) | |
Lactation data (pre and postnatal developmental toxicity) studies not done | ||||
Avatrombopag | ||||
Embryo-fetal developmental study | ||||
Rats | ||||
100 | 53 | Not characterized | Some toxicity observed | |
300 | 80 | 1. Decreased food consumption | 1. Decreased weight gain | |
1000 | 190 | 2. Decreased weight gain | 2. Skeletal variations (extra ribs) | |
3. Maternal mortality | ||||
Rabbits | ||||
100 | 10 | Abortion | None | |
300 | 11 | 1. Abortion | None | |
600 | 35 | 2. Decreased food consumption | ||
3. Decreased weight gain | ||||
Pre and post-natal developmental study (organogenesis through lactation) | ||||
Rats | ||||
100 | 53 | Maternal toxicity at all doses noted | Pup mortality noted at all doses. Maximum mortality between PND 14–21 | |
300 | 80 | |||
600 | 190 | |||
Rats (study 2) | ||||
5 | − | NOAEL | NOAEL | |
15 | − | NOAEL | NOAEL | |
50 | 43 | 1. Decreased body weight gain | ||
2. Delayed sexual maturity | ||||
3. Increased pup mortality from day 4 to 25 | ||||
Juvenile dose range finding postnatal development study (PND 7–34) | ||||
Rats | 10, 100, 300, 600, 1000 mg/kg NOAEL in mother and fetus | |||
Fostamatinib | ||||
Rabbits | ||||
0 | None | None | ||
10 | None | None | ||
20 | None | Malformations | ||
50 | Maternal mortality, Increased post-implantation fetal loss | Malformations, retarded growth | ||
Rats | ||||
0 | None | None | ||
5 | None | None | ||
12.5 | None | Malformations | ||
25 | Decreased birth weight, Increased post implantation loss | Malformations, retarded growth |
ITP, immune thrombocytopenia; NOAEL, no observed adverse effect level; PND, postnatal day.
Romiplostim
Romiplostim, developed under the name of AMG531, is a 60-kDa recombinant protein called ‘peptibody’ with a molecular formula of C1317H2043N361O395S9.45,52 The molecule consists of a ‘peptide domain’ and an ‘Fc domain’.45 The Fc portion of the molecule contains IgG1 heavy chains and kappa light chain constant regions bound by disulfide chains.45 Two identical TPO-peptide sequences are linked to each arm of the Fcγ heavy chain by polyglycine linkers at the C-terminus.45 In the initial stages of development, the 14-amino-acid long TPO peptide had a very short half-life and would have needed frequent administration to have meaningful clinical use. However, when linked with the IgG1-Fc fragment, the half-life of the drug extended dramatically (120–140 h).45 It is administered as a subcutaneous injection at a dose of 1–10 µg/kg once every week. In patients with ITP, romiplostim achieves a peak concentration within 7–50 h (median 14 h).45,53 As with other TPO-RAs, romiplostim also showed maternal and fetal toxicity only at doses that were toxic to both humans and maternal test dams (Table 2).53 However, more severe fetal adverse events such as stillbirth, fetal death, and significant malformations were noted at the toxic doses compared to eltrombopag. Romiplostim binds to the Fc receptor and is expected to be excreted in the milk of lactating mothers.54 Within the western hemisphere, romiplostim (approved by the US FDA for ITP in 2008) has the longest safety record for the management of patients with ITP. In some case, it has been used for more than a decade. Although TPIAO (a TPO drug developed by 3SBio, a Chinese company), was launched in China in 2006 for chemotherapy-induced thrombocytopenia, it was not approved for ITP until 2011. Nearly 80% of patients on treatment with romiplostim either reduced or discontinued treatment and nearly a quarter permanently discontinued treatment.12,17 This finding correlated well with increased function of Tregs and Bregs; and also increased circulation of tumor growth factor-β, which suggested that TPO-RAs may play a key role in restoring immune homeostasis.12,55 Whether it is a direct effect of TPO-RAs or an immunogenic effect of increased platelet turnover is a matter of further debate.12,55–58
Avatrombopag
Avatrombopag, developed under the name of E5501 (formerly called YM477 and AKR 501), is a small molecule TPO-RA with a chemical formula of C29H34Cl2N6O3S2 and a molecular weight of 649.65 g/mol.59,60 Like eltrombopag, it is another non-peptide TPO mimetic with both in vitro and in vivo activity to increase the platelet count.61 It is administered orally, with a dose-dependent response in platelets. It reaches peak concentration in the blood within 5–6 h and has a half-life of approximately 19 h. Like eltrombopag, it is also active only in humans and chimpanzees.61 The clinical review committee approving avatrombopag observed that fetal toxicity was not independent of maternal toxicity and was possibly driven by it.60 In the prenatal and post-natal studies, in which the drug was administered throughout organogenesis, a 15 mg/kg dose in rats appeared to be safe for both the pup and the dam (Table 2). As avatrombopag was not considered for chronic use at the time of clinical review, the committee labeled it as ‘may cause fetal harm’. In a toxico-kinetic study, a radiolabeled dose of avatrombopag did cross over to the fetus by gestational day 18 and was present in the lactation milk by day 10.60
Drugs targeting Fc-FcγR interaction and spleen tyrosine kinase pathway
The Major Histocompatibility Complex (MHC)class I-like Fc receptor, neonatal Fc receptor (FcRn), is involved in a pH-dependent salvage pathway that recycles and subsequently prolongs the half-life of IgG.62 This interaction helps to maintain humoral immunity in humans. However, in patients with autoimmune disorders, this interaction also maintains an abundant quantity of pathogenic IgG, which leads to humoral autoimmunity.62 Drugs that block the interaction of FcRn and IgG are expected to enhance the degradation of the IgG and are being explored as a potential therapy for ITP. Many molecules, like ARGX-113 (efgartigimod), UCB7665 (rozanolixizumab), etc., are currently in clinical trials for adult patients with ITP. Fostamatinib (R788) is an oral spleen tyrosine kinase (syk) inhibitor that converts to its active metabolite R406 (by intestinal alkaline phosphatase) and blocks the downstream effect of Fc receptor activation in the mast cells and B cells.63 It is approved as a second-line treatment for adult patients with ITP based on positive results from two randomized controlled trials.64 Their use in pregnancy is contraindicated based on preclinical data in developmental studies, which show severe urogenital abnormalities in gravid rats and rabbits.65 The US FDA recommends strict contraception before starting the medication, throughout treatment, and for more than one month after stopping the medication.
Current evidence on the use of TPO-RAs in pregnancy
Since all trials of TPO-RAs in adult patients with ITP have excluded pregnant women and lactating mothers, the current literature is restricted to ‘off-label’ use in pregnant women. Till recently, a ‘Nplate Pregnancy Exposure Registry’ (NCT02090088) was open, which was terminated on 6 January 2015, and the results were never published in a journal.66 Four patients, who had romiplostim exposure any time during the pregnancy, were registered during the trial period between March 2014 and January 2015, and all of the pregnancies resulted in live births. Adverse events such as preterm delivery, adrenal insufficiency, phimosis, decreased platelet count, and intraventricular hemorrhage were reported in the registry. As the results were never published formally, it is not possible to draw any conclusions regarding a causal effect between romiplostim and these adverse events. Eltrombopag and avatrombopag are pharmacologically active only in humans and chimpanzees. Hence the preclinical data from rats and rabbits cannot be interpreted conclusively for humans.46,49,60 A registry for eltrombopag use in pregnancy, named ‘Promacta Pregnancy Registry’ (NCT01064336 – February 2010–July 2016), listed only one patient and never reported any results.
Kong et al.67 reported a phase I, prospective study using a novel recombinant thrombopoietin (rhTPO) in pregnant patients with ITP. This was a full length, glycosylated TPO produced by Chinese hamster ovary cells that was almost identical to the endogenous TPO.67 A total of 31 pregnant patients with ITP who were refractory to IVIG and steroids were enrolled in the trial. The patients enrolled in the trial either had a history of bleeding complications due to thrombocytopenia from ITP or had presented with a platelet count of less than 30 × 109/L. Patients in the first trimester of pregnancy were excluded from the trial. A loading dose of 300 U/kg/day was given for 14 days, followed by sequential maintenance. The rhTPO was discontinued if the platelet count was more than 100 × 109/L. After delivery, the dose of rhTPO was changed to 300 U/kg/week from a daily dose. All patients were followed for 24 weeks, and all 31 neonates were followed for 53 weeks after delivery. Three-quarters of all patients had ITP diagnosed before the start of pregnancy. All patients had some bleeding manifestation, out of whom 10% had a severe bleeding manifestation. A complete response (defined as a platelet count more than 100 × 109/L) was observed in 33% (n = 10), and partial response was observed in 44% (n = 13) of subjects by day 14. In 80% of the patients, bleeding events were resolved by day 14. Although, four patients were deemed non-responders, the authors report improvement in bleeding events in these patients as well. No thromboembolic events or any other significant adverse events were noted in the mothers. Serum anti-TPO antibodies were measured at 4 weeks after starting the treatment, at the end of the treatment, and 6 months after stopping the treatment with rhTPO. Although the authors report that none of the mothers developed anti-TPO antibodies, they also acknowledge that the sample size is too small to derive definite conclusions.67
Michel et al.68 reported an international retrospective study of 15 patients (17 pregnancies) who received either eltrombopag (10 pregnancies) or romiplostim (seven pregnancies) for ITP in pregnancy. Eleven patients were diagnosed with ITP prior to the start of pregnancy. Thirteen pregnancies (12 patients) received TPO-RA in the third trimester of pregnancy primarily to prepare for the process of delivery (median 34 weeks of gestation, interquartile range (IQR) 27–39 weeks). Eleven of these 13 pregnancies had minor bleeding manifestations (skin and/or mucosal bleeding). Only one patient had severe gastrointestinal bleeding. The median exposure in these patients was 3 weeks (IQR 1–10 weeks). Three patients receiving eltrombopag for chronic ITP had unexpected pregnancies while on the medication and were exposed to TPO-RAs during the first trimester. Interestingly, it seems from the presented data that these three patients did not receive eltrombopag throughout the pregnancy (median exposure 12 weeks, IQR 9–12 weeks). Two of the three patients received additional treatment with IVIG and corticosteroids, while one patient who was refractory to seven previous lines of treatment did not receive any other treatment. One patient who received romiplostim during the first pregnancy was switched to eltrombopag after delivery of the first neonate and went on to have her second pregnancy while she was on eltrombopag. This was the only patient to receive TPO-RAs throughout the pregnancy (39 weeks). Eight pregnancies reported complete response, six reported partial response, and three pregnancies reported no response. Eleven pregnancies received additional treatment in the form of steroids and IVIG (five reported complete response, four with partial response, and two with no response). No adverse events or thromboembolic events were reported in the mother; however, the data are limited due to the retrospective nature of this study.
Several case reports and small series have reported off-label use of TPO-RAs in pregnant women, which present more granular data regarding the use and adverse events related to TPO-RAs in pregnancy (Table 3). The majority of these patients had ITP (n = 12), and one patient had MYH9 disorder. Except for three patients who started TPO-RA before getting pregnant and chose to stay on it for the entire pregnancy, the medication was started around a median of 30 weeks of gestation (16–36 weeks) and was administered for a median time of 5 weeks (1–24 weeks). All patients who responded to TPO-RA did so within 2–3 weeks of administering the medication. The one patient who was deemed as a non-responder received only a single dose of romiplostim among numerous other agents, including chemotherapy.40 This patient also developed postpartum hemorrhage (PPH) due to persistent thrombocytopenia.40 None of the other patients developed PPH due to low platelet counts. Three patients who started TPO-RA before getting pregnant and chose to continue it throughout the pregnancy developed hypertensive crises and placental infarcts. None of the patients on TPO-RA had an abortion or premature delivery. The decision to continue the medication after delivery was made based on platelet counts, in which a few patients were taken off due to high platelet counts. None of the patients experienced hepatic toxicity, signs of marrow failure, or major vessel thrombosis on either eltrombopag or romiplostim. Currently, lactating mothers are advised either to discontinue breastfeeding or the TPO-RA. However, the fact that oral TPO-RA should not be taken within 4–6 h of consuming food (to prevent chelation) raises the possibility that the high calcium content of breast milk would possibly chelate the TPO-RA as well.49,59,69 As there are no overall data on safety for breastfeeding with the use of TPO-RA, lactating mothers are discouraged to breastfeed if using TPO-RA.
Table 3.
Author | Age and gravida | Time of diagnosis | Co-morbid conditions | Previous treatment | TPO-RA used and dose | Timing of TPO-RA and weeks on TPO-RA | Platelet response (x109/L | Additional treatment if needed. | Bleeding at presentation | Estimated blood loss at delivery | Mode of delivery and indication | Use of neuraxial anesthesia |
---|---|---|---|---|---|---|---|---|---|---|---|---|
Purushothaman et al.39 | 27 years, G5 | During 2nd pregnancy | None | Steroids, triple therapy for H. pylori | Eltrombopag 25 mg → 50 mg | 29 weeks/ 7 weeks | 20–50 | Platelet transfusion | Spotting per vaginum, petechiae, hemorrhage in conjunctiva | NR, reported as normal | Vaginal at 36 weeks, Obstetric | NR |
Patil et al.35 | 28, G1 | Prior to pregnancy | None | Steroids, IVIG, Rituximab Azathioprine Romiplostim | Romiplostim, 3–4 µg/kg | 5 months before gestation/ 34 weeks | 1 to ~650 (waxing and waning) | IVIG Steroids | Mild epistaxis | 300 ml | Vaginal at ~34 weeks, induced, obstetric | Yes |
Chua et al.38 | 31, G1 | 11 years prior to pregnancy | None | IVIG steroids | Romiplostim 2 → 6 µg/kg | 30 weeks/ 8 weeks | 13–50 | Platelet transfusion | Mild epistaxis | 150 ml | Vaginal, spontaneous, 38 weeks | No |
27, G1 | 28 weeks of gestation | Hypo-thyroid, gestational DM, positive ANA | IVIG steroids | Romiplostim 4 → 3 µg/kg | 33 weeks/ 4 weeks | 4–100 | Platelet transfusion | Gum bleeding, mild epistaxis Spontaneous Bruising | NR | C-section, eclampsia, proteinuria, failure of progression of labor, 37 weeks | General anesthesia | |
Suzuki et al.36 | 25, G3A2 | 2 years prior to pregnancy | 2 abortions in past due to ITP | IVIG steroids Eltrombopag | Continued Eltrombopag, 12.5 mg | 21 months before gestation/entire pregnancy | 19–60 (waxing and waning) | Steroids IVIG platelet transfusion | No bleeding | NR | C-section, severe hypertension, 40 weeks | NR |
Favier et al.34 | 41, G2 | MYH9 disorder | PPH with previous pregnancy | none | Eltrombopag, 50 mg | 36 weeks/ 2 weeks | 30–179 | None | None | Normal | C-section, Breech and PROM, 38 weeks | NR |
Harrington et al.40 | 34, G3A2 (twins) | 29 weeks | 2 abortions in past | IVIG steroids Azathioprine Anti-D | Romiplostim (only one dose) | 32 weeks/ 5 weeks | No response | Azathioprine IVIG plasma exchange | Petechiae ecchymosis | 1700 ml | C-section, twins, 37 weeks | NR |
Payandeh et al.41 | 22, NR | Prior to gestation | Positive ds DNA | IVIG steroids Hydroxy-Chloroquine Splenectomy | Romiplostim, NR | 3 cycles near delivery | Max Platelet count 164 | None | NR | Reported as normal | NR | NR |
Samuelson et al.42 | 32, G2A1 | 5 years prior to gestation | MCTD. First pregnancy terminated, stroke- on coumadin | IVIG steroids Rituximab Romiplostim | Romiplostim 2 → 4 µg/kg | Throughout pregnancy | 369 at delivery (waxing and waning) | IVIG steroids platelet transfusion | None | PPH at 1.5 weeks controlled medically | Vaginal, pre-eclampsia, 35 weeks | Yes |
26, G1 | 10th week | None | IVIG steroids Rituximab | Romiplostim, 1 µg/kg | 32 weeks/ 5 weeks | 5–131 | None | None | 300 ml | Vaginal, 37 weeks | Yes | |
Alkaabi et al.37 | 34, G6 | 27th week | None | IVIG steroids Rituximab Cyclophosphamide Anti-D Eltrombopag | Romiplostim 3 → 6 µg/kg |
~30 weeks/ 4 weeks | 4–91. Then dropped to 52 | None | NR | Reported as normal | Vaginal, 34 weeks | NR |
A, Abortion; G, Gravida; DNA, Deoxyribo nucleic acid; ITP, Immune thrombocytpenia; IVIG, intravenous immunoglobulin; NR, Not reported; MCTD, Mixed connective tissue disorder; MYH-9, Myosine-9 heavy chain; PPH, Post partum hemorrhage; DM, Diabetes mellitus; ANA, Anti-nuclear antibody; TPO-RA, Thrombopoietin receptor agonist; C-section, Cesarean section; PROM, Premature rupture of membranes
Neonatal thrombocytopenia associated with maternal ITP
ITP in the mother can lead to neonatal thrombocytopenia and related complications. Up to 25–50% of neonates born to mothers with ITP develop thrombocytopenia.70–72 Out of these, 5–15% are born with platelet counts less than 50 × 109/L, and 1–5% are born with severe thrombocytopenia (platelet count less than 20 × 109/L).1,71–73 A maternal history of splenectomy (done for treating ITP) or a previous pregnancy with ITP that led to thrombocytopenia in the neonate are some recognized risk factors associated with NMITP.1,72–78 The timing of the diagnosis of ITP in the mother (whether diagnosed before or after pregnancy starts) as a risk factor for NMITP remains controversial.79,80 Passive transfer of maternal antibodies across the placenta is the most widely accepted mechanism behind NMITP. However, it must be noted that up to 40% of neonates will test negative for antiplatelet antibodies.72,81 Although breastfeeding is safe for mothers with ITP, prolonged thrombocytopenia (more than 3 months long) in the neonate has been observed in such cases. The reasons remain anecdotal and are attributed to the passive transfer of antibodies in breast milk.82 A case–control study evaluating prolonged thrombocytopenia in neonates born to mothers with ITP demonstrated the presence of IgA antibodies targeting αIIbβ3 integrin.83 In neonates born to mothers with ITP, a cord blood sample should be drawn to determine the platelet count.1,10 If the cord blood shows thrombocytopenia, then this should be confirmed with a venous blood sample.10,72 The platelet count usually reaches a nadir by postpartum day 1–5.1,2,10,72 IVIG is the drug of choice and is used when the platelet count drops below 30 × 09/L.1,2 Donor platelets are often transfused along with IVIG to raise the platelet counts quickly.1,10,72,80 Steroids are not frequently used in NMITP to prevent neonatal sepsis. However, if used then they are administered in low doses.84 Intracranial hemorrhage (ICH) occurs in less than 1% of neonatres with NMITP.1,85,86 All neonates with NMITP exhibiting neurological manifestations indicative of ICH must be evaluated with a head ultrasound.1 Concurrent NMITP and fetal-neonatal alloimmune thrombocytopenia (FNAIT) must be considered in the differential if a neonate develops ICH and in those born with a platelet count less than 10 × 109/L.1 A full review of FNAIT is done elsewhere.87
Current evidence for the use of TPO-RAs in neonates
Kong et al.67 followed the 31 neonates born during the study period for a median period of 53 weeks (range 39–68 weeks) after delivery. They did not report any difference in the cord blood TPO levels of healthy mothers and mothers with ITP.67 They also did not report any adverse events such as ICH, severe bleeding, or a platelet count less than 10 × 109/L in the neonates after birth or in the follow-up period.67 The incidence of low birth weight and premature labor was similar to those treated with conventional treatment in previous studies.67 It is worth noting that nine out of 31 neonates born to mothers with ITP had a platelet count between 50 and 100 × 109/L, and none was born with a platelet count below 50 × 109/L.67,88 In the retrospective review reported by Michel et al.,68 18 neonates were born from 17 pregnancies (one pregnancy resulted in twins). NMITP was found in six neonates, including the twins from a single mother (median count 14 × 109/L, IQR 4–34 × 109/L). IVIG and platelet transfusions were used for treatment in five out of six neonates with NMITP. One neonate who had thrombocytosis at the time of birth (558 × 109/L) was born to a mother who received a higher than the recommended dose of eltrombopag for 4 weeks (the patient received 100 mg/day; the maximum recommended dose is 75 mg/day). Preterm delivery occurred in five out of 17 pregnancies, and a cesarean section was done in six pregnancies. All the events were attributed to obstetric causes, and none were attributed to TPO-RA exposure in utero. None of the four pregnancies that received TPO-RA during the first trimester reported any adverse outcomes. One death due to genetic malformation (trisomy 8) and one neonate born with pulmonary stenosis had only 1 week’s worth of TPO-RA exposure in utero, that too at the end of the third trimester. On review of all case reports (Table 4), five out of 13 neonates developed thrombocytopenia, and two out of five developed bleeding complications such as intraventricular hemorrhage (IVH) and purpura.35,38 The administration of TPO-RA to the mother did not seem to have any bearing on the prevention of NMITP. IVIG and steroids were used as first-line therapy for NMITP. No babies were reported to have low birth weight, and fetal malformation was reported only in one nenate. The mother of this neonate had a long history of ITP treated with multiple medications (azathioprine, rituximab, steroids, vincristine) and had received romiplostim through her entire pregnancy.35
Table 4.
Author | TPO-RA used in first trimester (starting week) | Birth weight (kg) | Gestation age (weeks) | Platelet count at birth (× 109/L) | Developed NMITP | Bleeding manifestation | Platelet nadir, day of gestation | Treatment | Final count | Fetal malformation |
---|---|---|---|---|---|---|---|---|---|---|
Purushothaman et al.39 | No (29) | 1.86 | 36 | 145 | Yes | None | 55, 3rd day | IVIG | 249 | None |
Patil et al.35 | Yes | 1.91 | 33 weeks, 6 days | 70 | Yes | IVH | 33, 8 h | IVIG | 116 | Yes |
Chua et al.38 | No (30) | 2.77 | 38 | 229 | No | None | − | − | − | − |
No (33) | 4.65 | 37 | 53 | Yes | Purpura + IVH (day 15) | 6, Day 4 | IVIG + steroids + platelets | 70 (by 8 weeks) | None | |
Decroocq et al.33 | No (16) | 3.48 | ~40 | 186 | No | None | − | − | − | − |
No (28) | 2.64 | 38 | 90 | Yes | None | 90 | None | Normal by day 5 | None | |
Suzuki et al.36 | Yes | 1.67 | 37 | 416 | No | None | − | − | − | None |
Favier et al.34 | No (36) | 3.145 | ~40 | 62 | No | None | − | − | − | None |
Harrington et al.40 | No (32) | NR | ~37 | NR | Yes | <20 | NR | IVIG | NR | None |
Payandeh et al.41 | No (near delivery) | NR | NR | NR | NR | NR | NR | NR | NR | None |
Samuelson et al.42 | Yes | Normal | ~36 | 255 | No | − | − | − | − | None |
No (32) | NR | 37 | 132 | No | − | − | − | − | None | |
Alkaabi et al.37 | No (~30) | Normal | 34 | NR | No | − | − | − | − | None |
TPO-RA, Thrombopoietin receptor agonist; NMITP, Neonatal-maternal alloimmune thrombocytopenia; IVIG, Intravenous immunoglobulin; IVH, Intraventricular hemorrhage; NR, Not reported
Controversies in the effect of TPO-RA on the fetus and the newborn
The effect of TPO-RA on organogenesis in the human fetus is not clear from the current literature. TPO is known as a critical physiological regulator of hemangioblasts, which eventually differentiate into endothelial and hematopoietic cells.89,90 Excessive TPO exposure has long been speculated as a cause of limb defects in patients with hereditary thrombocythemia, an autosomal dominant disease that occurs either due to the mutation in the TPO gene or the TPO receptor (c-MPL) gene.91 The germline mutation in the TPO gene results in the removal of ‘inhibitory’ signals, which leads to uninhibited translation of the TPO-mRNA and an increase in TPO production.91 Although extremely rare, congenital limb defects have been observed in families with hereditary thrombocythemia.91–94 Graziano et al.91 analyzed the TPO gene of one such patient with hereditary thrombocythemia and congenital unilateral limb defect (proband) and found similar limb defects in his first and third offspring. In 2012, the same group reported another family with hereditary thrombocythemia and limb defects.92 Due to limb defects being rare among families with hereditary thrombocythemia, genetic background is suspected to be at play in addition to the excessive TPO. On the contrary, in patients with congenital thrombocytopenia due to mutations in homeobox genes (such as HOXA11), limb defects (radio-ulnar synostosis syndrome) have been observed, which raises doubts about the relationship between excessive TPO and limb defects.95,96 In addition to this, no limb defects have ever been reported in families with hereditary thrombocythemia, in which the c-MPL gene was mutated, which again does not support the theory of excessive TPO being the cause of limb defects.92 From a clinical aspect, it is a well-established fact that all of the approved TPO-RAs cross the placenta in utero to reach the fetal marrow.88 The current preclinical data suggest fetal harm at doses that were toxic to the maternal dam as well, raising the possibility that the fetal harm may be driven by maternal toxicity rather than the medication itself.49,53 In their phase I study, Kong et al.67 excluded patients in their first trimester of pregnancy hence preventing exposure of rhTPO during organogenesis. In the data from the case reports compiled for this review, one patient who was administered romiplostim throughout the pregnancy delivered a fetus with malformations.35 However, it seems that those patients who received TPO-RA after completion of the first trimester did not have an adverse neonatal outcome.
The management of thrombocytopenia in the neonate aims to prevent catastrophic bleeding, particularly ICH. Platelet transfusion is optimal in the setting of acute bleeding secondary to NMITP. However, several studies have noted that ‘liberal’ transfusion of platelets prophylactically is associated with increased morbidity and mortality in neonates.97–100 In the recently concluded PlaNet-2 trial, a higher mortality and bleeding rate was observed in neonates who received platelet transfusion at a platelet count of 50 × 109/L compared to those who received it at a threshold of 25 × 109/L.101 It is not clear whether the increased mortality is due to the higher platelet counts themselves or transfusion-related issues. Currently, there are no clinical data to support the use of TPO-RA in neonates with thrombocytopenia. In theory, the use of TPO-RA as an alternative to platelet transfusion in neonatal thrombocytopenia seems an attractive option.100,102 However, it usually takes 4–6 days of continuous administration of TPO-RA for the platelets to respond and at least 10–14 days to achieve the maximum response.45,103 Since nearly 80% of severe thrombocytopenia in the neonatal intensive care unit resolve within 14 days,104,105 the use of TPO-RA would be restricted to a minority of neonates who present with ‘persistent thrombocytopenia’ (thrombocytopenia for more than 14 days).100,102 Mahat et al.106 recently published the first report of using romiplostim in a full-term NMITP. Starting on the 34th day of life, the neonate received escalating doses of romiplostim (1–3 µg/kg/week), which led to normalization of platelets by day 69.106 Until further evidence evolves, the use of TPO-RA in neonates should be avoided.
Strengths and limitations
The strength of this paper lies in the comprehensive review of literature pertaining to the use of TPO-RA in pregnant patients with ITP. The preclinical evidence presented here has never been explored in any manuscript and shows that although fetal toxicity was demonstrated in animal models, that is either not applicable to humans (as in the case of eltrombopag and avatrombopag) or does not lead to toxicity at human equivalent doses. However, the lack of prospective data and scarcity of information in the retrospective data is the major limitation of this review.
Conclusion
The treatment options for managing thrombocytopenia in pregnant patients with ITP are limited due to concerns for fetal toxicity. Steroids and IVIG are the most widely used drugs in the treatment of pregnant patients with ITP. Our review of the literature suggests that TPO-RAs can help raise the platelet count within 2–3 weeks in pregnant patients with ITP. In light of the existing literature, it seems that the use of TPO-RAs is safe in the second and third trimesters. However, their use during the first trimester when organogenesis is at its peak must be avoided till further evidence demonstrating fetal and maternal safety becomes available. The current literature does not suggest any untoward effect of using TPO-RAs in late pregnancy on the fetus or the neonate. However, there is no clinical evidence to suggest whether it helps in preventing thrombocytopenia in neonates with NMITP or not. There is a pressing need to conduct a prospective clinical trial or a registry study evaluating the use of TPO-RAs in pregnant patients with ITP.
Footnotes
Author contributions: NA: conception of the idea, data collection, data analysis, and interpretation, wrote the manuscript.
AM: Project leader, critical revision of the manuscript.
Conflict of interest statement: The authors declare that there is no conflict of interest.
Funding: No financial support was received for this research, authorship, and/or publication of this article.
Availability of data and materials: The dataset supporting the conclusions of this article are included in the article.
ORCID iD: Ankit Mangla https://orcid.org/0000-0002-6789-4315
Contributor Information
Nikki Agarwal, Division of Pediatric Hematology and Oncology, Cleveland Clinic Foundation, Cleveland, OH, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA.
Ankit Mangla, Division of Hematology and Oncology, Seidman Cancer Center, University Hospitals, 11100 Euclid Avenue, Cleveland, OH 44106, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA.
References
- 1. Cines DB, Levine LD. Thrombocytopenia in pregnancy. Hematology Am Soc Hematol Educ Program 2017; 2017: 144–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Lambert MP, Gernsheimer TB. Clinical updates in adult immune thrombocytopenia. Blood 2017; 129: 2829–2835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Fogerty AE. Thrombocytopenia in pregnancy: mechanisms and management. Transfus Med Rev 2018; 32: 225–229. [DOI] [PubMed] [Google Scholar]
- 4. Burrows RF, Kelton JG. Thrombocytopenia at delivery: a prospective survey of 6715 deliveries. Am J Obstet Gynecol 1990; 162: 731–734. [DOI] [PubMed] [Google Scholar]
- 5. Gernsheimer T, James AH, Stasi R. How I treat thrombocytopenia in pregnancy. Blood 2013; 121: 38–47. [DOI] [PubMed] [Google Scholar]
- 6. Stavrou E, McCrae KR. Immune thrombocytopenia in pregnancy. Hematol Oncol Clin North Am 2009; 23: 1299–1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. McCrae KR, Samuels P, Schreiber AD. Pregnancy-associated thrombocytopenia: pathogenesis and management. Blood 1992; 80: 2697–2714. [PubMed] [Google Scholar]
- 8. Crowther MA, Burrows RF, Ginsberg J, et al. Thrombocytopenia in pregnancy: diagnosis, pathogenesis and management. Blood Rev 1996; 10: 8–16. [DOI] [PubMed] [Google Scholar]
- 9. Piatek CI, El-Hemaidi I, Feinstein DI, et al. Management of immune-mediated cytopenias in pregnancy. Autoimmun Rev 2015; 14: 806–811. [DOI] [PubMed] [Google Scholar]
- 10. Sankaran S, Robinson SE. Immune thrombocytopenia and pregnancy. Obstet Med 2011; 4: 140–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Bussel JB, Berkowitz RL, Lynch L, et al. Antenatal management of alloimmune thrombocytopenia with intravenous gamma-globulin: a randomized trial of the addition of low-dose steroid to intravenous gamma-globulin. Am J Obstet Gynecol 1996; 174: 1414–1423. [DOI] [PubMed] [Google Scholar]
- 12. Li J, Sullivan JA, Ni H. Pathophysiology of immune thrombocytopenia. Curr Opin Hematol 2018; 25: 373–381. [DOI] [PubMed] [Google Scholar]
- 13. Neunert C, Terrell DR, Arnold DM, et al. American Society of Hematology 2019 guidelines for immune thrombocytopenia. Blood Adv 2019; 3: 3829–3866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Provan D, Arnold DM, Bussel JB, et al. Updated international consensus report on the investigation and management of primary immune thrombocytopenia. Blood Adv 2019; 3: 3780–3817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Michel M, Novoa MV, Bussel JB. Intravenous anti-D as a treatment for immune thrombocytopenic purpura (ITP) during pregnancy. Br J Haematol 2003; 123: 142–146. [DOI] [PubMed] [Google Scholar]
- 16. Bussel JB, Provan D, Shamsi T, et al. Effect of eltrombopag on platelet counts and bleeding during treatment of chronic idiopathic thrombocytopenic purpura: a randomised, double-blind, placebo-controlled trial. Lancet 2009; 373: 641–648. [DOI] [PubMed] [Google Scholar]
- 17. Kuter DJ, Bussel JB, Lyons RM, et al. Efficacy of romiplostim in patients with chronic immune thrombocytopenic purpura: a double-blind randomised controlled trial. Lancet 2008; 371: 395–403. [DOI] [PubMed] [Google Scholar]
- 18. Varghese LN, Defour JP, Pecquet C, et al. The thrombopoietin receptor: structural basis of traffic and activation by ligand, mutations, agonists, and mutated calreticulin. Front Endocrinol (Lausanne) 2017; 8: 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Debili N, Wendling F, Cosman D, et al. The mpl receptor is expressed in the megakaryocytic lineage from late progenitors to platelets. Blood 1995; 85: 391–401. [PubMed] [Google Scholar]
- 20. Forsberg EC, Prohaska SS, Katzman S, et al. Differential expression of novel potential regulators in hematopoietic stem cells. PLoS Genet 2005; 1: e28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Grozovsky R, Begonja AJ, Liu K, et al. The Ashwell–Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling. Nat Med 2015; 21: 47–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Wolber EM, Jelkmann W. Interleukin-6 increases thrombopoietin production in human hepatoma cells HepG2 and Hep3B. J Interferon Cytokine Res 2000; 20: 499–506. [DOI] [PubMed] [Google Scholar]
- 23. Kuter DJ, Gernsheimer TB. Thrombopoietin and platelet production in chronic immune thrombocytopenia. Hematol Oncol Clin North Am 2009; 23: 1193–1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Fielder PJ, Gurney AL, Stefanich E, et al. Regulation of thrombopoietin levels by c-mpl-mediated binding to platelets. Blood 1996; 87: 2154–2161. [PubMed] [Google Scholar]
- 25. Stoffel R, Wiestner A, Skoda RC. Thrombopoietin in thrombocytopenic mice: evidence against regulation at the mRNA level and for a direct regulatory role of platelets. Blood 1996; 87: 567–573. [PubMed] [Google Scholar]
- 26. Ichikawa N, Ishida F, Shimodaira S, et al. Regulation of serum thrombopoietin levels by platelets and megakaryocytes in patients with aplastic anaemia and idiopathic thrombocytopenic purpura. Thromb Haemost 1996; 76: 156–160. [PubMed] [Google Scholar]
- 27. Gurney AL, Carver-Moore K, de Sauvage FJ, et al. Thrombocytopenia in c-mpl-deficient mice. Science 1994; 265: 1445–1447. [DOI] [PubMed] [Google Scholar]
- 28. Ballem PJ, Segal GM, Stratton JR, et al. Mechanisms of thrombocytopenia in chronic autoimmune thrombocytopenic purpura. Evidence of both impaired platelet production and increased platelet clearance. J Clin Invest 1987; 80: 33–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Kaushansky K. The molecular mechanisms that control thrombopoiesis. J Clin Invest 2005; 115: 3339–3347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Aledort LM, Hayward CP, Chen M-G, et al.; ITP Study Group. Prospective screening of 205 patients with ITP, including diagnosis, serological markers, and the relationship between platelet counts, endogenous thrombopoietin, and circulating antithrombopoietin antibodies. Am J Hematol 2004; 76: 205–213. [DOI] [PubMed] [Google Scholar]
- 31. Emmons RV, Reid DM, Cohen RL, et al. Human thrombopoietin levels are high when thrombocytopenia is due to megakaryocyte deficiency and low when due to increased platelet destruction. Blood 1996; 87: 4068–4071. [PubMed] [Google Scholar]
- 32. Nugent D, McMillan R, Nichol JL, et al. Pathogenesis of chronic immune thrombocytopenia: increased platelet destruction and/or decreased platelet production. Br J Haematol 2009; 146: 585–596. [DOI] [PubMed] [Google Scholar]
- 33. Decroocq J, Marcellin L, Le Ray C, et al. Rescue therapy with romiplostim for refractory primary immune thrombocytopenia during pregnancy. Obstet Gynecol 2014; 124 (Suppl. 1): 481–483. [DOI] [PubMed] [Google Scholar]
- 34. Favier R, De Carne C, Elefant E, et al. Eltrombopag to treat thrombocytopenia during last month of pregnancy in a woman with MYH9-related disease: a case report. A A Pract 2018; 10: 10–12. [DOI] [PubMed] [Google Scholar]
- 35. Patil AS, Dotters-Katz SK, Metjian AD, et al. Use of a thrombopoietin mimetic for chronic immune thrombocytopenic purpura in pregnancy. Obstet Gynecol 2013; 122: 483–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Suzuki N, Hiraga J, Hariyama Y, et al. A low birth weight infant with no malformations delivered by a primary immune thrombocytopenia patient treated with eltrombopag. Int J Hematol 2018; 108: 109–111. [DOI] [PubMed] [Google Scholar]
- 37. Alkaabi JK, Alkindi S, Riyami NA, et al. Successful treatment of severe thrombocytopenia with romiplostim in a pregnant patient with systemic lupus erythematosus. Lupus 2012; 21: 1571–1574. [DOI] [PubMed] [Google Scholar]
- 38. Chua SJ, Morton MR, Svigos J, et al. Use of romiplostim in pregnancy for refractory idiopathic thrombocytopenic purpura: two case reports with maternal and fetal outcomes and literature review. Obstet Med. Epub ahead of print 29 May 2018. DOI: 10.1177/1753495X18773960 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Purushothaman J, Puthumana KJ, Kumar A, et al. A case of refractory immune thrombocytopenia in pregnancy managed with elthrombopag. Asian J Transfus Sci 2016; 10: 155–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Harrington P, Nelson-Piercy C, Williamson C, et al. Refractory severe immune thrombocytopenia in a twin pregnancy. Obstet Med 2018; 11: 35–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Payandeh M, Karami A, Karami N, et al. Romiplostim: successful treatment of a pregnant woman with refractory immune thrombocytopenia. Biomed Res Ther 2018; 5: 2565–2571. [Google Scholar]
- 42. Samuelson BT, Kreuziger LB, Gernsheimer T. Use of romiplostim for refractory primary immune thrombocytopenia during pregnancy. Clin Obstet Gynecol Reprod Med 2017; 3: 1–3. [Google Scholar]
- 43. Rosa K. Supplemental application for avatrombopag in ITP accepted by FDA. Targeted Oncology. https://www.targetedonc.com/view/supplemental-application-for-avatrombopag-in-itp-accepted-by-fda
- 44. Wang L, Gao Z, Chen XP, et al. Efficacy and safety of thrombopoietin receptor agonists in patients with primary immune thrombocytopenia: a systematic review and meta-analysis. Sci Rep 2016; 6: 39003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Kuter DJ. Biology and chemistry of thrombopoietic agents. Semin Hematol 2010; 47: 243–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Erickson-Miller CL, Delorme E, Tian SS, et al. Preclinical activity of eltrombopag (SB-497115), an oral, nonpeptide thrombopoietin receptor agonist. Stem Cells 2009; 27: 424–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Nieto M, Calvo G, Hudson I, et al. The European Medicines Agency review of eltrombopag (Revolade) for the treatment of adult chronic immune (idiopathic) thrombocytopenic purpura: summary of the scientific assessment of the Committee for Medicinal Products for Human Use. Haematologica 2011; 96: e33–e40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Cheng G. Eltrombopag, a thrombopoietin-receptor agonist in the treatment of adult chronic immune thrombocytopenia: a review of the efficacy and safety profile. Ther Adv Hematol 2012; 3: 155–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Australian Government. Department of Health and Ageing Therapeutic Goods Administration. Australian public assessment report for eltrombopag olamine. Woden: Australia, 2010. [Google Scholar]
- 50. ClinicalTrials.gov. Milk-only lactation study for patients on eltrombopag. NCT01055600 (2015). https://clinicaltrials.gov/ct2/show/NCT01055600 (accessed 28 February 2021).
- 51. Gonzalez-Porras JR, Bastida JM. Eltrombopag in immune thrombocytopenia: efficacy review and update on drug safety. Ther Adv Drug Saf 2018; 9: 263–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Stasi R, Evangelista ML, Amadori S. Novel thrombopoietic agents: a review of their use in idiopathic thrombocytopenic purpura. Drugs 2008; 68: 901–912. [DOI] [PubMed] [Google Scholar]
- 53. Australian Government. Department of Health and Ageing Therapeutic Goods Administration Australian Public Assessment Report (AUSPAR) for Romiplostim. Woden: Australia, 2018. [Google Scholar]
- 54. Kuter DJ. Romiplostim. Cancer Treat Res 2011; 157: 267–288. [DOI] [PubMed] [Google Scholar]
- 55. Zufferey A, Kapur R, Semple JW. Pathogenesis and therapeutic mechanisms in immune thrombocytopenia (ITP). J Clin Med 2017; 6: 16. [Google Scholar]
- 56. Semple JW, Italiano JE, Jr, Freedman J. Platelets and the immune continuum. Nat Rev Immunol 2011; 11: 264–274. [DOI] [PubMed] [Google Scholar]
- 57. Li C, Li J, Li Y, et al. Crosstalk between platelets and the immune system: old systems with new discoveries. Adv Hematol 2012; 2012: 384685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018; 131: 1777–1789. [DOI] [PubMed] [Google Scholar]
- 59. DOPTELET. Avatrombopag (package insert). Durham, NC: Dova Pharmaceuticals, Inc., 2018. [Google Scholar]
- 60. DOPTELET (Avatrombopag). Durham, NC: Dova Pharmaceuticals, Inc., 2018. [Google Scholar]
- 61. Fukushima-Shintani M, Suzuki K, Iwatsuki Y, et al. AKR-501 (YM477) a novel orally active thrombopoietin receptor agonist. Eur J Haematol 2009; 82: 247–254. [DOI] [PubMed] [Google Scholar]
- 62. Petkova SB, Akilesh S, Sproule TJ, et al. Enhanced half-life of genetically engineered human IgG1 antibodies in a humanized FcRn mouse model: potential application in humorally mediated autoimmune disease. Int Immunol 2006; 18: 1759–1769. [DOI] [PubMed] [Google Scholar]
- 63. Connell NT, Berliner N. Fostamatinib for the treatment of chronic immune thrombocytopenia. Blood 2019; 133: 2027–2030. [DOI] [PubMed] [Google Scholar]
- 64. Bussel JB, Arnold DM, Boxer MA, et al. Long-term fostamatinib treatment of adults with immune thrombocytopenia during the phase 3 clinical trial program. Am J Hematol 2019; 94: 546–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Clemens GR, Schroeder RE, Magness SH, et al. Developmental toxicity associated with receptor tyrosine kinase Ret inhibition in reproductive toxicity testing. Birth Defects Res A Clin Mol Teratol 2009; 85: 130–136. [DOI] [PubMed] [Google Scholar]
- 66. Nplate® pregnancy exposure registry. Thousand Oaks, CA: Amgen, 2014. [Google Scholar]
- 67. Kong Z, Qin P, Xiao S, et al. A novel recombinant human thrombopoietin therapy for the management of immune thrombocytopenia in pregnancy. Blood 2017; 130: 1097–1103. [DOI] [PubMed] [Google Scholar]
- 68. Michel M, Ruggeri M, Gonzalez-Lopez TJ, et al. Use of thrombopoietin receptor agonists for immune thrombocytopenia in pregnancy: results from a multicenter study. Blood 2020; 136: 3056–3061. [DOI] [PubMed] [Google Scholar]
- 69. Al-Samkari H, Kuter DJ. Optimal use of thrombopoietin receptor agonists in immune thrombocytopenia. Ther Adv Hematol 2019; 10: 2040620719841735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Melekoglu NA, Bay A, Aktekin EH, et al. Neonatal outcomes of pregnancy with immune thrombocytopenia. Indian J Hematol Blood Transfus 2017; 33: 211–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Webert KE, Mittal R, Sigouin C, et al. A retrospective 11-year analysis of obstetric patients with idiopathic thrombocytopenic purpura. Blood 2003; 102: 4306–4311. [DOI] [PubMed] [Google Scholar]
- 72. van der Lugt NM, van Kampen A, Walther FJ, et al. Outcome and management in neonatal thrombocytopenia due to maternal idiopathic thrombocytopenic purpura. Vox Sang 2013; 105: 236–243. [DOI] [PubMed] [Google Scholar]
- 73. Kelton JG. Idiopathic thrombocytopenic purpura complicating pregnancy. Blood Rev 2002; 16: 43–46. [DOI] [PubMed] [Google Scholar]
- 74. Goldman BG, Hehir MP, Yambasu S, et al. The presentation and management of platelet disorders in pregnancy. Eur J Haematol 2018; 100: 560–566. [DOI] [PubMed] [Google Scholar]
- 75. Hachisuga K, Hidaka N, Fujita Y, et al. Can we predict neonatal thrombocytopenia in offspring of women with idiopathic thrombocytopenic purpura? Blood Res 2014; 49: 259–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Koyama S, Tomimatsu T, Kanagawa T, et al. Reliable predictors of neonatal immune thrombocytopenia in pregnant women with idiopathic thrombocytopenic purpura. Am J Hematol 2012; 87: 15–21. [DOI] [PubMed] [Google Scholar]
- 77. Loustau V, Debouverie O, Canoui-Poitrine F, et al. Effect of pregnancy on the course of immune thrombocytopenia: a retrospective study of 118 pregnancies in 82 women. Br J Haematol 2014; 166: 929–935. [DOI] [PubMed] [Google Scholar]
- 78. Vadasz B, Chen P, Yougbare I, et al. Platelets and platelet alloantigens: lessons from human patients and animal models of fetal and neonatal alloimmune thrombocytopenia. Genes Dis 2015; 2: 173–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Rottenstreich A, Israeli N, Roth B, et al. Risk factors associated with neonatal thrombocytopenia in pregnant women with immune thrombocytopenic purpura. J Matern Fetal Neonatal Med. Epub ahead of print 4 November 2018. DOI: 10.1080/14767058.2018.1523891 [DOI] [PubMed] [Google Scholar]
- 80. Kawaguchi K, Matsubara K, Takafuta T, et al. Factors predictive of neonatal thrombocytopenia in pregnant women with immune thrombocytopenia. Int J Hematol 2014; 99: 570–576. [DOI] [PubMed] [Google Scholar]
- 81. Sainio S, Joutsi L, Jarvenpaa AL, et al. Idiopathic thrombocytopenic purpura in pregnancy. Acta Obstet Gynecol Scand 1998; 77: 272–277. [PubMed] [Google Scholar]
- 82. Gernsheimer TB. Congenital and acquired bleeding disorders in pregnancy. Hematology Am Soc Hematol Educ Program 2016; 2016: 232–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Hauschner H, Rosenberg N, Seligsohn U, et al. Persistent neonatal thrombocytopenia can be caused by IgA antiplatelet antibodies in breast milk of immune thrombocytopenic mothers. Blood 2015; 126: 661–664. [DOI] [PubMed] [Google Scholar]
- 84. Bussel JB, Sola-Visner M. Current approaches to the evaluation and management of the fetus and neonate with immune thrombocytopenia. Semin Perinatol 2009; 33: 35–42. [DOI] [PubMed] [Google Scholar]
- 85. Bussel JB, Zabusky MR, Berkowitz RL, et al. Fetal alloimmune thrombocytopenia. N Engl J Med 1997; 337: 22–26. [DOI] [PubMed] [Google Scholar]
- 86. Kaplan C. Foetal and neonatal alloimmune thrombocytopaenia. Orphanet J Rare Dis 2006; 1: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Bussel JB, Primiani A. Fetal and neonatal alloimmune thrombocytopenia: progress and ongoing debates. Blood Rev 2008; 22: 33–52. [DOI] [PubMed] [Google Scholar]
- 88. Bussel JB, Lee EJ. TPO for ITP in pregnancy. Blood 2017; 130: 1073–1074. [DOI] [PubMed] [Google Scholar]
- 89. Perlingeiro RC, Kyba M, Bodie S, et al. A role for thrombopoietin in hemangioblast development. Stem Cells 2003; 21: 272–280. [DOI] [PubMed] [Google Scholar]
- 90. Zambidis ET, Oberlin E, Tavian M, et al. Blood-forming endothelium in human ontogeny: lessons from in utero development and embryonic stem cell culture. Trends Cardiovasc Med 2006; 16: 95–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Graziano C, Carone S, Panza E, et al. Association of hereditary thrombocythemia and distal limb defects with a thrombopoietin gene mutation. Blood 2009; 114: 1655–1657. [DOI] [PubMed] [Google Scholar]
- 92. Graziano C, David M, Magini P, et al. An additional family with association of hereditary thrombocytosis and transverse limb deficiency: confirmation of a rare clinical spectrum. Am J Med Genet A 2012; 158A: 3211–3213. [DOI] [PubMed] [Google Scholar]
- 93. Hussein K, Percy M, McMullin MF, et al. Clinical utility gene card for: hereditary thrombocythemia. Eur J Hum Genet. Epub ahead of print 5 June 2014. DOI: 10.1038/ejhg.2013.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Stockklausner C, Echner N, Klotter AC, et al. Hereditary thrombocythemia caused by a thrombopoietin (THPO) gain-of-function mutation associated with multiple myeloma and congenital limb defects. Ann Hematol 2012; 91: 1129–1133. [DOI] [PubMed] [Google Scholar]
- 95. Horvat-Switzer RD, Thompson AA. HOXA11 mutation in amegakaryocytic thrombocytopenia with radio-ulnar synostosis syndrome inhibits megakaryocytic differentiation in vitro. Blood Cells Mol Dis 2006; 37: 55–63. [DOI] [PubMed] [Google Scholar]
- 96. Thompson AA, Nguyen LT. Amegakaryocytic thrombocytopenia and radio-ulnar synostosis are associated with HOXA11 mutation. Nat Genet 2000; 26: 397–398. [DOI] [PubMed] [Google Scholar]
- 97. Baer VL, Lambert DK, Henry E, et al. Severe thrombocytopenia in the NICU. Pediatrics 2009; 124: e1095–e1100. [DOI] [PubMed] [Google Scholar]
- 98. Ballmaier M, Germeshausen M. Advances in the understanding of congenital amegakaryocytic thrombocytopenia. Br J Haematol 2009; 146: 3–16. [DOI] [PubMed] [Google Scholar]
- 99. Garcia MG, Duenas E, Sola MC, et al. Epidemiologic and outcome studies of patients who received platelet transfusions in the neonatal intensive care unit. J Perinatol 2001; 21: 415–420. [DOI] [PubMed] [Google Scholar]
- 100. Cremer M, Sallmon H, Kling PJ, et al. Thrombocytopenia and platelet transfusion in the neonate. Semin Fetal Neonatal Med 2016; 21: 10–18. [DOI] [PubMed] [Google Scholar]
- 101. Curley A, Stanworth SJ, Willoughby K, et al. Randomized trial of platelet-transfusion thresholds in neonates. N Engl J Med 2019; 380: 242–251. [DOI] [PubMed] [Google Scholar]
- 102. Sallmon H, Gutti RK, Ferrer-Marin F, et al. Increasing platelets without transfusion: is it time to introduce novel thrombopoietic agents in neonatal care? J Perinatol 2010; 30: 765–769. [DOI] [PubMed] [Google Scholar]
- 103. Kuter DJ. New thrombopoietic growth factors. Blood 2007; 109: 4607–4616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Murray NA, Howarth LJ, McCloy MP, et al. Platelet transfusion in the management of severe thrombocytopenia in neonatal intensive care unit patients. Transfus Med 2002; 12: 35–41. [DOI] [PubMed] [Google Scholar]
- 105. Sparger KA, Ramsey H, Lorenz V, et al. Developmental differences between newborn and adult mice in response to romiplostim. Platelets 2018; 29: 365–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Mahat U, Talati R, Kodish E. Comment on: use of thrombopoietin receptor agonist (romiplostim) in neonatal autoimmune thrombocytopenia due to maternal immune thrombocytopenia. Pediatr Blood Cancer 2019; 66: e27706. [DOI] [PubMed] [Google Scholar]