Está en la página 1de 9

YGYNO-978608; No.

of pages: 9; 4C:
Gynecologic Oncology xxx (xxxx) xxx

Contents lists available at ScienceDirect

Gynecologic Oncology

journal homepage: www.elsevier.com/locate/ygyno

Epidemiology, diagnosis, and treatment of gestational trophoblastic


disease: A Society of Gynecologic Oncology evidenced-based review
and recommendation☆
N.S. Horowitz a,⁎, R.N. Eskander b, M.R. Adelman c, W. Burke d
a
Brigham & Women's Hospital/Dana Farber Cancer Institute, Boston, MA, USA
b
University of California, San Diego, Moores Cancer Center, La Jolla, CA, USA
c
University of Utah, Salt Lake City, UT, USA
d
Stony Brook Medicine, Long Island, NY, USA

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.1. Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.2. Molar pregnancy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.3. Human chorionic gonadotropin (hCG). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.4. GTN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.5. Low-Risk GTN. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.6. High-risk GTN. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.7. Ultra high-risk disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.8. Surgery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.9. PSTT/ETT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.10. Survivorship issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.11. Recommendations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Author contributions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

1. Introduction can be cured even in the presence of metastatic disease. With the
exception of PSTT and ETT, GTD arises from cytotrophoblasts and
Gestational trophoblastic disease (GTD) is a group of interrelated syncytiotrophoblasts, and produce abundant amounts of human chori-
tumors that arise from abnormal fertilization events and include both onic gonadotropin (hCG), which serves as an excellent tumor marker
benign conditions (complete hydatidiform mole (CM) and partial for diagnosis, monitoring of response, and follow-up to detect recur-
hydatidiform mole (PM), placental site nodule), potentially malignant rence. Given the rarity of this disease, management and treatment
conditions (atypical placental site nodule APSN), as well as malignant recommendations are commonly informed by data from small, retro-
conditions (invasive mole (IM), choriocarcinoma (CA), placental-site spective single institution or collaborative studies, small prospective
trophoblastic tumor (PSTT), and epithelioid trophoblastic tumor studies, meta-analyses, and expert opinion [2]. The purpose of this doc-
(ETT)). These malignancies are collectively referred to as gestational ument is to provide a comprehensive reference for the epidemiology,
trophoblastic neoplasia (GTN). While the majority of GTN occurs after management and outcomes of GTD/GTN. We carried out a systematic
a molar pregnancy (50%), they can arise after any gestational event review of English language studies, identified through a search of
with 25% arising after miscarriage or tubal pregnancy and 25% after MEDLINE/Pubmed, Cochrane Library, and Google Scholar from incep-
term or preterm pregnancies [1]. Unlike most malignancies, GTN tion until June 2021. Using the terms gestational trophoblastic disease,

☆ This practice statement has been endorsed by the American College of Obstetricians and Gynecologists (ACOG).
⁎ Corresponding author at: Brigham & Women's Hospital/Dana Farber Cancer Institute, 75 Francis Street, Dept. of OB/GYN, Division of Gynecologic Oncology, Boston, MA 02115, USA.
E-mail address: nhorowitz@partners.org (N.S. Horowitz).

https://doi.org/10.1016/j.ygyno.2021.10.003
0090-8258/© 2021 Published by Elsevier Inc.

Please cite this article as: N.S. Horowitz, R.N. Eskander, M.R. Adelman, et al., Epidemiology, diagnosis, and treatment of gestational trophoblastic
disease: A Society of Gynecologi..., Gynecologic Oncology, https://doi.org/10.1016/j.ygyno.2021.10.003
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

gestational trophoblastic neoplasia, molar pregnancy, human chorionic imprinted and maternally expressed gene, p57 can differentiate
gonadotropin, chemotherapy, we generated the references for this re- between CM (p57 negative) and either a PM (p57 positive) or non-
view and accompanying recommendations. molar hydropic abortions [25]. These additional diagnostics are
performed on tissue obtained at evacuation, not separate biopsies.
1.1. Epidemiology The initial treatment of hydatidiform moles in women who wish to
preserve fertility is suction dilation and curettage (D&C), preferably per-
Wide variations in the incidence of hydatidiform mole exist formed with the largest cannula that is easily introduced and with
throughout the world with rates ranging from 1 to 2 per 1000 pregnan- uterotonic agents administered as necessary [26,27]. Ultrasound guid-
cies in North America, Europe, and other developed nations, to 2–3 ance may be used at the discretion of the surgeon but may increase
times higher in some Asian countries, Brazil, and India [3]. Extremes of the rate of complete evacuation. Uterine evacuation by medication
reproductive age and prior molar pregnancy have emerged as major only methods is discouraged due to high failure rates, risk of hemor-
risk factors for the development of molar pregnancy. Age < 15 years, rhage, and increased development of GTN requiring chemotherapy
and > 45 years are most strongly associated with the development of [28]. RhD factor is expressed on trophoblastic tissue, therefore RhD im-
molar pregnancy, with the risk being 5–10 times greater for those munoglobulin should be administered at the time of uterine evacuation
over 45 [4,5]. The risk of a second molar pregnancy after a first increases in Rh negative women [3]. Given the earlier gestational age of diagnosis,
to approximately 1–2% (up from 0.1%) and after two molar pregnancies, complications during or after evacuation are uncommon, but surgeons
the risk of a third molar pregnancy is 15–20% [5–7]. This risk is much should be prepared to manage excessive bleeding and pulmonary com-
higher after a complete mole and interestingly, subsequent moles promise. For those who have completed childbearing and have a pre-
tend to be the same histology as the first [8]. sumed molar pregnancy, hysterectomy provides an alternative
Choriocarcinoma is the most common histologic GTN after a normal method of management. In one study, compared to uterine evacuation,
pregnancy. Because of its rapid growth and high propensity for metasta- hysterectomy has a significant advantage in preventing post-molar GTN
tic disease, it is considered the most aggressive form of GTN. The inci- with an approximately 80% reduction in risk [29]. Although hysterec-
dence of choriocarcinoma is approximately 3 per 100,000 deliveries in tomy eliminates any locally invasive disease, some women will have oc-
Europe and North America, compared to approximately 23 per cult metastases at the time of diagnosis and therefore hCG monitoring is
100,000 in Southeast Asia [3,9]. This risk increases with increasing still warranted after hysterectomy [29].
maternal age [10]. Post-evacuation surveillance with hCG is critical in identifying the de-
PSTT and ETT are incredibly rare entities and most are diagnosed velopment of post-molar GTN. Ideally, an assay that can detect all forms
remotely after non-molarpregnancies. The incidence of PSTT is approx- of hCG (beta-hCG, core hCG, C-terminal hCG, nicked-free beta, beta-core,
imately 1 per 100,000 deliveries, and the incidence of ETT is approxi- and hyperglycosylated) equally well should be used and are often re-
mately 0.1 per 100,000 deliveries [3,11]. ferred to as “total hCG assays” [30,31]. hCG is routinely monitored
weekly until normalization or diagnosis of GTN, however, appropriate
1.2. Molar pregnancy duration of hCG surveillance varies considerably by histologic subtype
and the institution performing the surveillance. Current guidelines for
There are two histologically distinct subtypes of hydatidiform moles, the duration of surveillance from various organizations and trophoblastic
complete (CM) and partial (PM). The majority of CM result from fertil- reference centers are noted in Table 1 [32–35]. A recent meta-analysis by
ization of an ovum lacking maternal nuclear DNA, with subsequent du- Albright et al. [35] confirmed that the overall incidence of post-molar
plication of the haploid genome from a single sperm (80%) or dispermic GTN is approximately 20% for CM and 4% for PM. For those patients
fertilization (20%). The resulting karyotypes are diploid, either 46 XX or who normalize their hCG <56 days after evacuation, the risk of develop-
46 XY, and are completely paternally derived [12,13]. PM are almost al- ing GTN is 0.03% following CM and 0.02% after PM. For those that normal-
ways triploid, resulting from dispermic fertilization of an ovum ize ≥56 days the rate is still low, 0.3% and 0.03% respectively. This data is
retaining its maternal DNA [12,13]. Although rare, if a woman has 2 or very reassuring and supports the recommendation that once hCG nor-
3 consecutive moles familial recurrent hydatidiform mole syndrome malization is achieved follow up should continue until 3 months after
(FRHM)) should be suspected [14]. CM related to FRHM are diploid CM and 1 month after PM, while maintaining patient safety [35,36].
and genetically biparental rather than androgenic and result from spo- Despite earlier diagnosis, the incidence of post-molar GTN has
radic mutations in NLRP7 (70% of cases), KHDC3L (5–10%% of cases), remained constant over time [37]. Risk factors for developing
or from gene(s) unidentified to date. [14–16]. If FRHM is confirmed, post-molar GTN include hCG at diagnosis >100,000 IU/L, excessive uter-
egg donation from an unaffected individual is a way to enable a success- ine size, theca lutein cysts, and age > 40 years. Women aged 40–49 with
ful non-molar pregnancy [17]. initial hCG levels exceeding 175,000 mIU/mL represent an especially at
Hydatidiform moles are characterized by unregulated trophoblastic risk group, with 85% developing post-evacuation GTN [38]. In women
proliferation, resulting in increased production of hCG. Elevated hCG older than 50 years, the risk of post-evacuation GTN may be as high as
levels above 100,000 IU/L are commonly observed with CM, but these 60%, regardless of presenting hCG level [39], though population based
levels are rare in PM given the less prominent trophoblastic hyperplasia
[4,18]. Ultrasonographic findings of a vesicular pattern, indicating
Table 1
hydropic villi, and theca lutein cysts are strongly suggestive of the diag- Current guidelines for hCG follow up after molar pregnancy.
nosis of CM [19]. Ultrasonographic features suggestive of a PM include
focal cystic changes in the placenta, presence of a nonviable fetus, and 1. ACOG monthly hCG x 6 months for both complete and partial mole (4).
2. National Comprehensive Cancer Network (NCCN) - 3 consecutive normal hCG
an elevated ratio of the transverse to anteroposterior dimension of the levels followed by 2 hCG assays in 3-month intervals (32)
gestational sac [20]. 3. FIGO - hCG monitoring every 1–2 weeks until normal followed by a single
The use of sensitive hCG assays and early ultrasound have shifted de- confirmatory normal hCG in a month for partial moles and monthly hCG for
tection of molar pregnancy into the first trimester and therefore the 6 months for complete moles (26)
4. United Kingdom (UK) - monthly hCG for six months for both CM starting from
classic presenting symptoms (excessive uterine size, anemia, pre-
the date of evacuation if normalization took ≤56 days or from the date of nor-
eclampsia, hyperemesis, hyperthyroidism, and respiratory failure) are malization if this took >56 days. For PM only one confirmatory normal hCG is
rarely encountered [21–24]. This shift to earlier detection and evacua- needed 1 month after the first normal (34)
tion has necessitated the use of additional diagnostics such as flow cy- 5. New England Trophoblastic Disease Center (NETDC) - After normalization of
tometry to determine ploidy, and immunohistochemistry staining for weekly hCG for partial mole one confirmatory hCG a month after normalization
and 3 monthly hCG for complete moles (36).
the cyclin dependent kinase inhibitor, p57 [17]. As a paternally

2
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

studies place this risk for older women around 30% [10]. For these Table 2
women, or for those in whom reliable hCG follow up cannot be ob- FIGO /WHO criteria for diagnosis of post-molar GTN (26).

tained, hysterectomy should be considered [39–41]. Although use of 1. Plateauing of hCG +/− 10% for 4 consecutive values over 3 weeks (i.e., days 1, 7,
prophylactic chemotherapy at the time of hysterectomy for molar preg- 14, 21)
nancy has been shown to decrease the risk of developing GTN, for those 2. A rise in hCG levels of ≥10% for 3 values over 2-week period (i.e., days 1, 7, 14)
3. Histologic diagnosis of choriocarcinoma or clinical and/or radiologic evidence of
that do develop GTN despite prophylactic chemotherapy, there is often
metastases
a delayed diagnosis, increase drug resistance, exposure to toxicity, and
worse outcome [42,43]. Therefore it is not recommended in this setting.
Development of a new gestation prior to the completion of post- 1.4. GTN
molar surveillance poses a diagnostic dilemma, and as such reliable con-
traception during this period is strongly advised. Importantly, recent Unlike other malignancies, GTN does not necessarily need histologic
data suggests that the use of hormonal contraception during post- confirmation, (although choriocarcinoma is occasionally seen on a D&C
molar follow-up does not increase the risk of developing or the severity or hysterectomy), and is the only malignancy diagnosed based on a
of GTN and does not postpone normalization of hCG [44–46]. serum assay. In 2018, FIGO/WHO updated criteria for diagnosing post-
molar GTN (Table 2) [26]. Prior to 2018, hCG persistence for 6 months
1.3. Human chorionic gonadotropin (hCG) after evacuation was a diagnostic criterion, however, recent data sug-
gests ≥80% of those with a persistently elevated but falling hCG 6 months
Human chorionic gonadotropin (hCG) is part of the glycoprotein after evacuation, who are managed expectantly without initiation of che-
hormone family along with luteinizing hormone (LH), follicle- motherapy, ultimately achieve undetectable levels. For those that do not
stimulating hormone (FSH), and thyroid-stimulating hormone (TSH). achieve undetectable levels, there is no worsening of outcome [52,53].
These hormones are heterodimers, sharing a common alpha subunit For GTN diagnosed after a non-molar pregnancy, a single elevated hCG
and different, but with varying degrees of homology, betasubunits. with exclusion of any other explanation other than GTN is sufficient for
The intact heterodimeric form of hCG is produced almost exclusively diagnosis. This is particularly true if there is unexplained metastatic
by cytotrophoblast and syncytiotrophoblast of the placenta, GTN, disease. Importantly, biopsies should be avoided as the highly vascular
germ cell, and other somatic tumors, such as bladder or lung, with tro- nature of GTN can lead to significant bleeding complications.
phoblastic elements [47]. In addition to the intact heterodimer, there Once criteria are met, patients must be evaluated for extent of dis-
is another isoform, hyperglycosolated hCG (hCG\\H) which contains ease and assigned a FIGO anatomic stage (Table 3) and WHO risk
more sugar residues than regular hCG, is predominately produced by score (Table 4) to determine the likelihood of disease progression
cytotrophoblasts, and promotes trophoblastic growth and invasion and/or resistance to treatment with single agent chemotherapy [54].
[48]. In addition, multiple variants are produced by enzymatic degrada- Evaluation should include a complete physical examination including
tion in the tissue and circulation. The most common variants include a pelvic examination and a chest radiograph as lung and vaginal metas-
free beta subunit, free alpha subunit, nicked hCG, nicked hCG beta, tases are the most common site of disease. FIGO expressly recommends
and hCG beta subunit core fragment [47]. chest radiograph rather than chest computed tomography (CT) for stag-
All common commercially available hCG assays are sandwich-type ing as micrometastases are detected on CT scans in approximately 40%
immunoassays that measure the beta-subunit, the intact hCG, and of patients with negative chest X-rays. These micrometastases do not af-
almost all the major variants described above. These sandwich assays fect overall survival and can unnecessarily increase the use of multi-
are highly sensitive and can detect serum hCG levels as low as agent chemotherapy [55]. If there is evidence of metastatic disease on
1–2 IU/L and in urine as low as 10–50 IU/L [47]. Fortunately, hCG is an the initial evaluation, CT scan of abdomen and pelvis as well as MRI of
exquisitely sensitive tumor marker thus allowing for easy diagnosis, the brain are warranted. Other investigations should include routine
monitoring response to treatment, and identifying recurrence of GTN. laboratory tests (complete blood count, hepatic, renal, thyroid tests,
This is most successful if the same assay is used consistently throughout chemistry, blood group, and hCG) and a pelvic ultrasound to exclude
the course of care as there can be great variation between assays. intrauterine pregnancy, asses uterine size, and estimate intrauterine
False negative and false positive results for hCG can occur tumor size and/or invasion. When calculating WHO score it is important
at the extremes of value. Markedly elevated hCG, usually when to use the hCG at time of GTN diagnosis, not hCG at time of mole evac-
hCG > 1000,000 IU/L can produce a phenomenon known as the “hook uation, as this can lead to an inappropriately elevated WHO score. Addi-
effect”, in which the fixed, solid phase antibodies and the labeled, solu- tionally, WHO risk scores do not apply to PSTT or ETT. Treatment for
ble antibodies are saturated preventing sandwich formation and a neg- these histologies is based on the independent adverse prognostic factors
ative result. If hook effect is suspected, the true value can be obtained FIGO stage (III/IV) and the time from the antecedent pregnancy, with
with serial dilutions of the sample before analysis [49,50]. Additionally, ≥48 months considered high-risk disease warranting chemotherapy
false negative results can occur if the tumor secretes a form of hCG not [56,57].
detected or under-detected by a particular assay. When there is a persis- Women with stage I or stage II/III with a WHO prognostic score < 7
tently low but positive hCG level, defined as an hCG < 1000 IU/L with no are considered to have low-risk disease and are treated with single
more than a two-fold variation over at least a 3-month period and in the agent chemotherapy while those with stage IV disease or WHO score ≥ 7
absence of tumor on imaging, one must consider false positive results, have high-risk disease and receive multi-agent chemotherapy. A sepa-
phantom hCG, or other sources of hCG such as the pituitary [51]. Phan- rate category of ultra- high-risk, defined as WHO score > 12, identifies
tom hCG results from heterophile antibodies that interfere with the im- women at high risk of early death and poor outcome who should be
munoassay. This can be confirmed if hCG elevation is noted only in
serum and not in urine, by using two separate commercial assays in
which the results vary greatly or are negative in repeat assay, or by per- Table 3
sistent elevated hCG despite serial dilution of the serum. The pituitary FIGO staging (54):
gland produces a small amount of hCG which becomes more pro- Stage I Disease confined to uterine corpus
nounced around menopause or in women in whom multi-agent, high Stage II GTN extends outside of uterus, but is limited to the genital structures
dose chemotherapy has temporarily stopped ovarian function. Pituitary (adnexa, vagina, broad ligament)
hCG can be distinguished from real trophoblastic hCG by placing the pa- Stage III GTN extends to the lungs, with or without known genital tract
involvement
tient on higher dose oral contraceptive or GnRH agonist which will sup-
Stage IV All other metastatic sites
press pituitary hCG production [51].

3
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

Table 4
WHO Risk Score (54).

Scores

0 1 2 4

Age (yr) <40 ≥40 –


Antecedent pregnancy Mole Abortion Term
Interval months from index pregnancy <4 4–<7 7–<13 ≥13
Pretreatment serum hCG (International Unit/L) <103 103–<104 104–<105 >105
Largest tumor size (including uterus) 3–<5 cm ≥5 cm
Site of metastases Lung Kidney/spleen Gastrointestinal/liver Brain
Number of metastases – 1–4 5–8 >8
Previous failed chemotherapy – – Single drug 2 or more drugs

Format for reporting to FIGO Annual Report: In order to stage and allot a risk factor score, a patient's diagnosis is allocated to a stage as represented by a Roman numeral I, II, III, and IV. This
is then separated by a colon from the sum of all the actual risk factor scores expressed in Arabic numerals for example, stage II:4, stage IV:9. This stage and score will be allotted for each
patient.
Definition of Risk Categories:
Low Risk - Stage I or Stage II/III with a WHO prognostic score < 7.
High Risk - Stage IV disease or any stage and WHO score ≥ 7.
Ultra-high Risk - Any stage with WHO score > 12.

treated with low dose induction chemotherapy before initiating multi- agent therapy. All women with metastatic choriocarcinoma needed
agent chemotherapy [58]. multi-agent chemotherapy. For those without metastases or choriocar-
cinoma, and hCG was >410,000 IU/L and for those patients with metas-
tases or choriocarcinoma and hCG >150,000 IU/L combination
1.5. Low-Risk GTN
chemotherapy was also necessary to obtain remission [69].
After complete remission is obtained, consolidation therapy, using
Low-risk GTN is primarily treated and cured with one of two single
the last effective regimen should be delivered to prevent relapses.
agent drugs, methotrexate (MTX) or Dactinomycin (Act-D)
Lybol and colleagues showed that 3 rather than 2 cycles of consolidation
[2,26,59–65]. A variety of doses and infusion schedules for these drugs
decreased the risk of relapse (4 vs 8%) [70]. The chemotherapy cycle de-
have been utilized but multi-day MTX (either 8-day or 5-day) and
livered at first hCG normalization is considered the first consolidation
pulse Act-D seem to be the most efficacious (Table 5) [59]. Weekly
cycle.
methotrexate has been used secondarily for ease of schedule and low
Survival for women with low-risk GTN is excellent with rates ap-
toxicity but it is not as effective as multi-day regimens [60]. New data
proaching 100%. To achieve this high rate of success, additional chemo-
suggests that modifying the 8-day MTX regimen to avoid a weekend ad-
therapy regimens or surgical intervention may be necessary. Patients
ministration (treatment on the 8th day rather than 7th) does not com-
can have primary resistant disease (rising hCG after two cycles of treat-
promise oncologic outcome [61]. To date, there is not a clearly superior
ment or hCG plateau (<10% change) over three cycles) or may develop
regimen between these two drugs, with complete remission rates rang-
acquired resistance (hCG plateau over two course or rising hCG over
ing between 75 and 90% and rare grade 3 or 4 toxicities, for both agents
2 weeks after an initial response) [2]. New areas of metastases would
[59–65]. Treatment therefore is often determined by institutional pref-
also qualify as resistance/progression. Depending upon the hCG level
erence. However, in 2016, a Cochrane Review including over 600 pa-
at the time of resistance or if toxicity prevents adequate dosing/sched-
tients in 7 randomized controlled trials showed Act-D appeared to be
ule, switching to the alternate single agent (ActD or MTX) or other sin-
superior to MTX (RR 0.65, 95%CI 0.57–0.75) [59]. Alternative single
gle agent chemotherapy such as Carboplatin should be considered
drug options for low-risk GTN include etoposide or fluorouracil
[71,72]. If there is an inadequate response to the initial single agent,if
[66,67], though these are not typically used outside of Asia.
there are new metastases, and/or if hCG plateaus at a high level
Women with low-risk GTN with scores 5 or 6 are a particularly chal-
(>3000 IU/L), then multi-agent chemotherapy regimens should be em-
lenging group with only approximately 30% responding to single agent
ployed [2]. Additionally, for those with resistant disease, hysterectomy
chemotherapy [68]. A recent multicenter retrospective study identified
or resection of persistent metastatic disease could be considered, espe-
predictors of single agent resistance. In the study approximately 60% of
cially for those who no longer desire to preserve their fertility [41,73].
patients achieved complete remission after first or second line single
Second dilation and curettage has been investigated as an alterna-
tive to chemotherapy for low-risk GTN. In appropriately selected pa-
Table 5 tients, approximately 40% of patients are cured, avoiding toxicity of
Single-agent regimens for low-risk gestational trophoblastic neoplasia (59–65). chemotherapy altogether [74]. Although the ideal candidate for attempt
at second curettage could not be easily defined, patients at extremes of
Methotrexate Regimens Primary remission rates
reproductive age and those with WHO score of 5 or 6 were less likely to
Weekly IM 30–50 mg/m2 49–74%
respond to second curettage. Similarly, for those women with stage I
Multi-day every 2 weeks
5-day IV or IM 0.3–0.5 mg/kg 87–93%
low-risk GTN who are done with child bearing, hysterectomy, with or
8-day IV or IM 1 mg/kg on days 1,3,5,7⁎ 74–90% without a single dose of Act-D at the time of surgery, offers an excellent
8-day IM 50 mg on days 1,3, 5, 7⁎ 70–80% cure rate [41 73].
High dose IV⁎⁎ 69–90% Monitoring hCG during treatment should occur on a weekly basis or
100 mg/m2 over 30 min
with day 1 of each cycle, both are acceptable. Although consolidation
200 mg/m2 over 12 h
chemotherapy starts with the first normal hCG, remission is defined as
Dactinomycin Regimens⁎⁎⁎ three consecutive normal hCG. Posttreatment surveillance consists of
Pulse 1.25 mg/m2 IV every 2 weeks 69–90% monthly hCG for one year. In a recent population-based cohort study,
5- day 10–12 micrograms IV every 5 days 77–94% 73% of recurrences occurred within 1 year of remission and the risk of
⁎ Leucovorin 15 mg PO on days 2, 4, 6,8. recurrence beyond this was <1% per year [75]. If new symptoms such
⁎⁎ Leucovorin 15 mg PO every 12 h × 6 doses beginning 24 h after starting MTX. as vaginal bleeding develop after the year of surveillance, recurrence
⁎⁎⁎ Maximum dose 2 mg (2000 micrograms). needs to be considered and hCG evaluated.

4
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

1.6. High-risk GTN regimen, TE/TP (paclitaxel and etoposide alternating weekly with pacli-
taxel and cisplatin), with a 70% cure rate, in patients who were
The management of patients with high-risk metastatic or recurrent etoposide and platinum naïve, and reduced hematologic adverse events
GTN remains a clinically difficult scenario. In an effort to cure patients [82]. Alternate salvage treatment programs, with response rates of
with high-risk disease, and salvage those with persistent/recurrent 20–75% are listed in Table 6 80, 83. Uniformly these multi-agent regi-
GTN, several multi-agent chemotherapy regimens have been identified mens have significant hematologic toxicity. Administering granulocyte
(Table 6) [76–83]. However, given the low prevalence of the disease, it is colony stimulating factors (G-CSF) as primary prophylaxis can decrease
unclear which multi-agent regimen is most effective and least toxic, as morbidity, treatment delays and dose reductions [84]. Given the signif-
randomized controlled trials have not been conducted. icant chemosensitivity of GTN, the prognosis for women with stage II or
Developed at Charing Cross in 1979, EMA-CO has become the pre- III high-risk disease remains very good with approximately 70–80%
ferred regimen in the management of patients with high-risk disease achieving complete remission with primary therapy and another
[77]. In a report of 272 consecutive women with high-risk GTN, the cu- 10–15% cured with additional lines of chemotherapy or surgery [77,80].
mulative 5-year survival rate was 86.2%. This was more efficacious in
the chemotherapy naïve population. Seventy-eight percent achieved 1.7. Ultra high-risk disease
complete remission with EMA-CO, 17% developed drug resistance of
whom 70% were salvaged with other chemotherapy, while 11 patients For those patients with ultra high-risk disease (WHO score > 12),
(4%) experienced early deaths [78]. Many other groups have shown experts advocate using induction chemotherapy to reduce the risk of
similar responses to this multi-drug regimen [76,77]. As with low-risk life-threatening complications, predominantly hemorrhage from meta-
disease, chemotherapy for high-risk disease is continued for at least 3 static implants. The use of an induction regimen, with combination low
consolidation courses after the first hCG normalization [76,77]. dose etoposide and cisplatin (EP on days 1 and 2, weekly x 2–3 weeks),
Despite the established efficacy of the EMA-CO regimen, approxi- has been shown to significantly reduce the risk of early death, as re-
mately 30% of women will develop resistance, or experience relapse ported in the Charing Cross experience [58]. When patients present
after remission, necessitating salvage chemotherapy [80]. In these pa- with central nervous system metastatic lesions, in addition to low-
tients, alternate regimens containing etoposide and platinum, with or dose induction EP, the MTX dose in the EMA-CO or EMA-EP regimen
without surgical resection, can usually affect cure. In EMA-CO failures, should be increased to 1 g/m2. Intrathecal (IT) methotrexate
the most commonly employed regimen is EMA-EP (substituting (12.5 mg) can be considered and is typically administered during the
etoposide and cisplatin for cyclophosphamide and vincristine in the CO or EP portion of the multi-agent regimens. Survival rates of 80–85%
EMA-CO regimen), with cure rates ranging from 66 to 85% [79,81]. In have been reported with these regimens and IT MTX [85,86]. Further-
an effort to mitigate the significant hematologic toxicity encountered more, there may be utility to gamma-knife, stereotactic radiotherapy,
using EMA-EP, investigators have also examined a taxane containing and surgical resection to control CNS disease. Whole-brain radiation is
controversial given the long- term cognitive toxicities. For these ultra
high-risk patients, consolidation with 3–4 cycles of therapy should be
Table 6 considered.
Salvage chemotherapy regimens for high-risk, persistent/recurrent, or refractory GTN
(76–83, 87,89, 90).
Active areas of investigation include the utilization of high-dose che-
motherapy (HDC), with stem cell support and use of immune check-
1. EMA-CO - Etoposide with methotrexate, actinomycin D (EMA) and vincristine point inhibitors. Prior investigators have shown sustained complete
and cyclophosphamide (CO)
responses to high-dose chemotherapy with stem cell support in pa-
Day 1 Etoposide 100 mg/m2 IV bolus tients with disease refractory to multiagent regimens. In a review of
Actinomycin-D 500 μg IV push 32 patients treated with HDC at 2 referral centers, 41% (N = 13)
Methotrexate 100 mg/m2 IV push
remained disease free after HDC with or without additional treatment
200 mg/m2 IV over 12 h
Day 2⁎ Actinomycin-D 500 μg IV push [87]. All patients who survived had hCG values ≤12 IU/L before HDC.
Etoposide 100 mg/m2 IV bolus In an effort to capitalize on elevated programmed death ligand
Leucovorin 15 mg PO q 12 h 1 (PD-L1) expression levels identified in GTN specimens [88], use of sin-
Day 8 Vincristine 1.0 mg/m2 gle agent pembrolizumab has been explored. In a small cohort of 4 pre-
Cyclophosphamide 600 mg/m2
viously treated patients (2 with choriocarcinoma and 2 with placental
2. EMA-EP – Etoposide, methotrexate, actinomycin-D, and cisplatin site trophoblastic tumor), 3 experienced durable remissions with single
Day 1 Etoposide 100 mg/m2 IV bolus
agent pembrolizumab, highlighting the potential utility of immunother-
Actinomycin-D 500 μg IV push apy in these disease settings [89]. Furthermore, in a recent small phase II
Methotrexate 100 mg/m2 IV push study, avelumab was shown to cure 8 out of 15 women (53%) with low-
200 mg/m2 IV over 12 h risk GTN whose disease progressed after first line therapy with MTX or
Day 2⁎ Actinomycin-D 500 μg IV push
ActD with minimal and mild toxicity. Importantly, there was also one
Etoposide 100 mg/m2 IV bolus
Leucovorin 15 mg PO q 12 h documented pregnancy after avelumab treatment [90]. The DART trial
Day 8 Etoposide 150 mg/m2 IV bolus (Dual Anti-CTLA-4 and Anti-PD-1 Blockade in Rare Tumors;
Cisplatin 75 mg/m2 IV bolus NCT02834013) is currently examining the utility of dual agent immuno-
3. TE-TP – Paclitaxel + etoposide alternating Paclitaxel + cisplatin therapy in patients with recurrent GTN.
4. ACE – Actinomycin-D, cisplatin, etoposide
5. VIP – Etoposide, ifosphamide cisplatin 1.8. Surgery
6. BEP – Bleomycin, etoposide, cisplatin
7. ICE – Ifosphamide, carboplatin, etoposide
8 Gemcitabine
Despite therapeutic advances and approaches, the importance of
9. Capcetabine or 5 FU containing regimen such as FAEV – floxuridine, surgery in the management of GTN should not be over looked. Hysterec-
dactinomycin, etoposide, and vincristine tomy as well as resection of oligometastatic extra-uterine disease plays
10. Liposomal doxorubicin an important role in the management of those with isolated
11. High dose chemotherapy with stem cell support
chemoresistant disease or those with uterine confined disease who no
12. Immune check point inhibitors
longer desire to preserve fertility [91,92]. Furthermore, salvage hyster-
Methotrexate dose increased to 1 g/m2 if brain metastases.
ectomy, rather than use of multi-agent chemotherapy, should be con-
⁎ Some centers omit day 2 etoposide, Act-D.
sidered in patients with evidence of persistent uterine disease despite

5
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

single agent chemotherapy [41,73]. Surgery may also be necessary to completed child bearing. Medically induced evacuations of known
deal with disease-related hemorrhage complications when emboliza- molar pregnancies should be avoided. (Level II-2).
tion is not available or has failed. Weekly hCG monitoring is recommended after evacuation of a com-
plete (CM) or partial mole (PM). Once hCG normalization is achieved,
1.9. PSTT/ETT hCG follow up should continue until 3 months after CM and 1 month
after PM. (Level I).
Given that PSTT and ETT arise from intermediate trophoblast, it is Patients who meet criteria for GTN (Table 1) must undergo staging
not surprising that these histologies behave similarly to each other but per FIGO and WHO Prognostic Scoring System. Staging should include
are different clinically from other GTN and therefore have unique man- physical exam, serum hCG (from diagnosis of GTN not the initial hCG
agement. [56,57]. Surgery is the mainstay of treatment for these dis- value at the time of molar pregnancy diagnosis), pelvic ultrasound,
eases and consists of hysterectomy with lymphadenctomy reserved and chest x-ray. For those with vaginal metastases or pulmonary metas-
for those with grossly enlarged lymph nodes [93]. There are case reports tases on chest x-ray, a CT of the chest, abdomen, pelvis and a brain MRI
of local uterine resection with preservation of fertility [94]. Although should be obtained. (Level II-2).
they are not chemotherapy resistant, compared to choriocarcinoma or Either single agent multi-day MTX or bolus Act-D is recommended
post-molar GTN, they are not as sensitive. First line treatment with for treatment of low-risk GTN. Second curettage is an acceptable alter-
multi-agent regimens like EMA-CO or EMA-EP is often reserved for native for those wishing to avoid chemotherapy. (Level I and II-1).
those that present with advanced stage disease or have ≥48 months For patients with low-risk GTN who develop resistance to or relapse
from the antecedent pregnancy [56,57]. These risk factors are the after single agent chemotherapy treatment with the alternative single
same for both PSTT and ETT. A relatively new entity, atypical placental agent (if hCG ≤3000 IU/L) or with EMA-CO (if hCG >3000 IU/L) is rec-
site nodule (APSN), has been associated with concurrent PSTT/ETT or ommended. (Level II-3).
the development of PSTT/ETT within 16 months of diagnosis in approx- Hysterectomy should be considered for those with stage I choriocar-
imately 10–15% of cases. This is based on small numbers but suggests cinoma or invasive mole who no longer desire fertility preservation.
patients with APSN should have a thorough clinical evaluation and con- Hysterectomy should also be considered as management of complete
sider hysterectomy. Larger series and longer follow up is necessary mole for women aged 40–49 with an initial hCG > 175,000 IU/L or
however in order to best counsel patients [95]. age ≥ 50 with any hCG value. Furthermore, for women with GTN, a
dose of single agent chemotherapy at the time of hysterectomy should
be administered to treat occult metastases. (Level II-2).
1.10. Survivorship issues For patients with high-risk GTN, EMA-CO is the recommended first
line treatment while those with ultra high-risk GTN should receive in-
The majority of women with molar pregnancies or GTN will be cured duction low dose EP followed by either EMA-CO or EMA-EP. (Level I
with treatment, and counseling regarding future pregnancies is critical. and II-1).
History of GTN or use of single agent chemotherapy has not been asso- For those patients with brain metastases, the MTX dose should be in-
ciated with an increased risk of infertility and patients can expect nor- creased to 1 g /m2. Other treatments to consider include neurosurgical
mal reproductive function [96]. Use of multi-agent chemotherapy consultation, radiation, and intrathecal MTX. (Level II-2).
however, was shown to induce early menopause (<45 years) in ap- G-CSF primary prophylaxis should be used in etoposide and plati-
proximately 35% of women and this risk increased with increasing age num containing regimens and as needed to maintain treatment sched-
at the start of treatment [97]. In women who subsequently become ule and dose intensity of other regimens. (Level II-2).
pregnant after molar pregnancy or treatment for GTN, there are good For those whose disease is resistant to or relapses after multi-agent
data to reassure them that the rate of live births, and other pregnancy chemotherapy consider consultation or referral to a high volume tro-
outcomes such as spontaneous abortions and obstetrical complications, phoblastic disease center. Management for these women should include
are no different than the general population, regardless of whether or surgery, when appropriate, and salvage chemotherapy including
not chemotherapy was administered [6]. The one difference is the risk immune checkpoint inhibitors or high dose chemotherapy with stem
of a second molar pregnancy [98]. When patients subsequently cell transplant, in selected patients. (Level II-3).
conceive, obtaining an early ultrasound in the first trimester to confirm Three cycles of consolidation chemotherapy, consisting of the last
a non-molar pregnancy is recommended. Additionally, hCG should be effective regimen, should be given after normalization of the hCG
measured approximately 6–8 weeks after the completion of any gesta- (Level II-2).
tional event (miscarriage, ectopic, term pregnancy) to ensure that it has Surveillance for low and high-risk GTN should consist of monthly
normalized and to exclude the development of choriocarcinoma. hCG x 12 months after completion of consolidation chemotherapy. Fur-
Though the risk of non-molar GTN is incredibly small, an hCG evaluation thermore, for patients with ultra high-risk disease hCG x 24 months
is a simple, inexpensive, and relatively non-invasive test to insure this after completion of chemotherapy is recommended. (Level II-3).
has not developed Examination of the placenta can be considered but For patients with presumed stage I PSTT and ETT hysterectomy with
is not mandatory [2]. removal of suspicious lymph nodes, and cytoreduction for those with
In an analysis of approximately 1900 patients and over 32,000 metastatic disease, should be performed. Furthermore, chemotherapy
person-years of follow up, Savage and his colleagues showed that with EMA-EP should be administered to those diagnosed with advanced
MTX carried no substantial long- term risk of secondary malignancies. stage disease or diagnosis ≥48 months from antecedent pregnancy.
Even after EMA-CO or other alkylating agent containing regimens, the (Level II-3).
risk of secondary malignancies such as leukemia was minimally For those with persistent low level hCG (< 200 IU/L), non-
increased and was associated with prolonged exposure [97]. gestational tumors, false positive hCG, and pituitary hCG should be ex-
cluded by measuring hCG on a different assay and in the urine, by
1.11. Recommendations obtaining a CT of chest, abdomen, pelvis and a brain MRI with pituitary
views, checking menopausal hormones, and trialing high dose oral con-
Women with presumed hydatidiform mole should undergo evacua- traceptive pills or GnRH agonist. (Level II-3).
tion with suction curettage and ultrasound guidance if necessary. For women with a history of GTD, a first trimester ultrasound should
Uterotonics should be administered after evacuation. Given the in- be performed and a hCG should be assessed 6–8 weeks after conclusion
creased risk of GTN, hysterectomy is an acceptable alternative in of any pregnancy event. Examination of the placenta can be considered
women with risk factors for GTN, ≥40 years old or those who have but is not mandatory. (Level II-3).

6
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

Author contributions [16] C.M. Wang, P.H. Dixon, S. Decordova, et al., Identification of 13 novel NLRP7 muta-
tions in 20 families with recurrent hydatidiform mole; missense mutations cluster
in the leucine-rich region, J. Med. Genet. 46 (2009) 569–575.
Dr. Neil Horowitz developed the practice statement outline, contrib- [17] R.A. Fisher, G.J. Maher, Genetics of gestational trophoblastic disease, Best Pract. Res.
uted to the practice statement, reviewed and edited the final draft. Clin. Obstet. Gynecol. 74 (2021) 29–41.
[18] D.R. Genest, O. Laborde, R.S. Berkowitz, et al., A clinicopathologic study of 153 cases
Drs. Ramez Eskander and Marisa Adelman contributed to the prac- of complete hydatidiform mole (1980-1990): histologic grade lacks prognostic sig-
tice statement and reviewed the final draft. nificance, Obstet. Gynecol. 78 (1991) 402–409.
Dr. William Burke reviewed the outline, practice statement draft and [19] C.B. Benson, D.R. Genest, M.R. Bernstein, et al., Sonographic appearance of first tri-
mester complete hydatidiform moles, Ultrasound Obstet. Gynecol. 16 (2000)
oversaw the practice statement development.
188–191.
[20] C. Fine, A.L. Bundy, R.S. Berkowitz, et al., Sonographic diagnosis of partial
Declaration of Competing Interest hydatidiform mole, Obstet. Gynecol. 73 (1989) 414–418.
[21] S.L. Curry, C.B. Hammond, L. Tyrey, et al., Hydatidiform mole: diagnosis, manage-
ment and long-term follow-up of 347 patients, Obstet. Gynecol. 54 (1975) 1–8.
Dr. Burke reports other from Titan Medical, outside the submitted [22] E.I. Kohorn, Molar pregnancy: presentation and diagnosis, Clin. Obstet. Gynecol. 27
work. (1984) 181–191.
Dr. Adelman reports personal fees from AbbVie, outside the submit- [23] A.E. Szulman, U. Surti, The clinicopathological profile of the partial hydatidiform
mole, Obstet. Gynecol. 59 (1982) 597–602.
ted work. [24] V. Soto-Wright, M. Bernstein, D.P. Goldstein, R.S. Berkowitz, The changing clinical
Dr. Horowitz reports personal fees from Up to Date, outside the presentation of complete molar pregnancy, Obstet. Gynecol. 86 (5) (1995) 775–779.
submitted work. [25] D.R. Genest, D.M. Dorfman, D.H. Castrillon, Ploidy and imprinting in hydatidiform
moles: complementary use of flow cytometry and immunohistochemistry of the
Dr. Eskander reports other from AstraZeneca, other from Clovis imprinted gene product p57KIP2 to assist molar classification, J. Reprod. Med. 47
Oncology, other from Eisai, outside the submitted work. (2002) 342–346.
[26] H.Y.S. Ngan, M.J. Seckl, R.S. Berkowitz, et al., Update on the diagnosis and manage-
ment of gestational trophoblastic disease, Int. J. Gynaecol. Obstet. 143 (Supple 2)
Acknowledgements
(2018) 79–85.
[27] L. Padron, F. Rezende, J. Amim, S.Y. Sun, R.C. Charry, I. Maesta, et al., Manual com-
Dr. Horowitz's research is supported by the Donald P. Goldstein, pared with electric vaccum aspiration for treatment of molar pregnancy, Obstet.
Gynecol. 31 (4) (2018) 652–659.
M.D. Trophoblastic Tumor Registry Endowment and The Dyett Family
[28] J.A. Tidy, A.M. Gillespie, N. Bright, et al., Gestational trophoblastic disease: a study of
Trophoblastic Disease Research and Registry Endowment. These mode of evacuation and subsequent need for treatment with chemotherapy,
endowments had no direct role in the generation of the data or the Gynecol. Oncol. 78 (2000) 309–312.
manuscript. [29] P. Zhao, Y. Lu, W. Huang, B. Tong, W. Lu, Total hysterectomy versus uterine evacua-
tion for preventing post-molar gestational trophoblastic neoplasia in patients who
are at least 40 years old: a systemic review and meta-analysis, BMC Cancer 19 (1)
References (2019) 13.
[30] L.A. Cole, Human chorionic gonadotropin and associated molecules, Expert. Rev.
[1] S. Ngan, M.J. Seckl, Gestational trophoblastic neoplasia management: an update, Mol. Diagn. 9 (2009) 51–73.
Curr. Opin. Oncol. 19 (2007) 486–491. [31] C.Y. Muller, L.A. Cole, The quagmire of hCG and hCG testing in gynecologic oncology,
[2] C. Lok, N. van Trommel, L. Massugar, F. Golfier, M. Seckl, Practical clinical guidelines Gynecol. Oncol. 112 (2009) 663–672.
of EOTTD for the treatment and referral of gestational trophoblastic disease, Eur. J. [32] Gestational Trophoblastic Neoplasia, NCCN Guidelines version1. 2022
Cancer 130 (2020) 228–240. [33] C. Coyle, D. Short, L. Jackson, et al., What is the optimal duration of human chorionic
[3] Y.K. Eysbouts, J. Bulten, P.B. Ottevanger, C.M.G. Thomas, M.J. Kate-Booij, A.E. van gonadotropin surveillance following evacuation of a molar pregnancy? A retrospec-
Heraarden, A.G. Siebers, Sweep FCGJ, Massuger LFAG, Trends in incidence for gesta- tive analysis of over 20,000 consecutive patients, Gynecol. Oncol. 148 (2018)
tional trophoblastic disease over the last 20 years in a population-based study, 254–257.
Gynecol. Oncol. 140 (1) (2016) 70–75. [34] J. Tidy, M. Seck, B.W. Hancock, On behalf of the royal college of obstetricians and
[4] R.S. Berkowitz, D.P. Goldstein, Clinical practice. Molar pregnancy, New Engl. J. Med. gynaecologists. Management of Gestational Trophoblastic Disease, BJOG 128
360 (2009) (1639-4). (2021) e1–e27.
[5] N.J. Sebire, M. Foskett, R.A. Fisher, H. Rees, M. Seckl, E. Newlands, Risk of partial and [35] B.B. Albright, J.M. Shorter, S.A. Mastroyannis, E.M. Ko, C.A. Schreiber, S. Sonalkar,
complete hydatidiform molar pregnancy in relation to maternal age, BJOG 109 (1) Obstet. Gynecol. 135 (1) (2020) 12–23.
(2002) 99–102. [36] R.S. Berkowitz, N.S. Horowitz, K.M. Elias, Hydatidiform Mole: Treatment and Follow
[6] R. Vargas, L.M. Barroilhet, K. Esselen, E. Diver, M. Bernstein, D.P. Goldstein, R.S. Up, UpToDate April 2021.
Berkowitz, Subsequent pregnancy outcomes after complete and partial molar preg- [37] S.Y. Sun, A. Melamed, D.P. Goldstein, et al., Changing presentation of complete
nancy, recurrent molar pregnancy, and gestational trophoblastic neoplasia: an up- hydatidiform mole at the New England trophoblastic disease center over the last
date from New England trophoblastic disease center, J. Reprod. Med. 59 (5–6) three decades: does early diagnosis alter risk for gestational trophoblastic neopla-
(2014) 188–194. sia? Gynecol. Oncol. 138 (2015) 46–49.
[38] K.M. Elias, M. Shoni, M.R. Bernstein, et al., Complete hydatidiform mole in women
[7] P.K. Sand, J.R. Lurain, J.I. Brewer, Repeat gestational trophoblastic disease, Obstet.
aged 40–49 years, J. Reprod. Med. 57 (2012) 254–258.
Gynecol. 63 (2) (1984) 140–144.
[39] K.M. Elias, D.P. Goldstein, R.S. Berkowitz, Complete hydatidiform mole in women
[8] N. Eagles, N.J. Sebire, D. Short, P.M. Savage, M.J. Seckl, R.A. Fisher, Risk of recurrent
older than age 50, J. Reprod. Med. (2010) 208–212.
molar pregnancies following complete and partial hydatidiform moles, Hum.
[40] P. Zhao, C. Chen, W. Lu, Comparison of different therapeutic strategies for complete
Reprod. 30 (9) (2015) 2055–2063.
hydatidiform mole in women at least 40 years old: a retrospective cohort study,
[9] H.O. Smith, Gestational trophoblastic disease epidemiology and trends, Clin. Obstet.
BMC Cancer 17 (1) (2017) 733.
Gynecol. 46 (3) (2003) 541–556.
[41] R. Sugrue, O. Foley, K.M. Elias, W.B. Growdon, et al., Outcomes of minimally invasive
[10] P. Savage, M. Winter, V. Parker, V. Harding, A. Sita-Lumsden, R.A. Fisher, R. Harvery, versus open abdominal hysterectomy in patients with gestational trophoblastic
N. Unsworth, N. Sarwar, D. Short, X. Agular, J. Tidy, B. Hancock, R. Coleman, M.J. disease, Gynecol. Oncol. 160 (2) (2021) 445–449.
Seckl, Demographis, natural history and treatment outcomes of non-molar gesta- [42] S. Limpongsanurak, Prophylactic actinomycin D for high-risk complete hydatidiform
tional choriocarcinoma: a UK population study, BJOG 127 (9) (2020) 1102–1107. mole, J. Reprod. Med. 46 (2001) 110–116.
[11] N.S. Horowitz, D.P. Goldstein, R.S. Berkowitz, Placental site trophoblastic tumors and [43] Q. Wang, J. Fu, L. Hu, et al., Prophylactic chemotherapy for hydatidiform mole to
epithelioid trophoblastic tumors: biology, natural history, and treatment modalities, prevent gestational trophoblastic neoplasia, Cochrane Database Syst. Rev. 9 (2017).
Gynecol. Oncol. 144 (1) (2017) 208–214 (Fisher RA, Newlands ES. Gestational tro- [44] M. Stone, K.D. Bagshawe, An analysis of the influences of maternal age, gestational
phoblastic disease: molecular and genetic studies. J Reprod Med 1998;43:81–97). age, contraceptive method, and the mode of primary treatment of patients with
[12] S.D. Lawler, R.A. Fisher, J. Dent, A prospective genetic study complete and partial hydatidiform moles on the incidence of subsequent chemotherapy, Br. J. Obstet.
hydatidiform moles, Am. J. Obstet. Gynecol. 164 (1991) 1270–1277. Gynaecol. 86 (1979) 782–792.
[13] J.M. Lage, S.D. Mark, D.J. Roberts, et al., A flow cytometric study of 137 fresh hydropic [45] P.R.S. Dantas, I. Maesta, J.R. Filho, et al., Does hormonal contraception during molar
placentas: correlation between types of hydatidiform moles and nuclear DNA pregnancy follow-up influence the risk and clinical aggressiveness of gestational
ploidy, Obstet. Gynecol. 79 (1992) 403–410. trophoblastic neoplasia after controlling for risk-factors? Gynecol. Oncol. 147
[14] R.A. Fisher, M.D. Hodges, E.S. Newlands, Familial recurrent hydatidiform mole: a (2017) 364–370.
review, J. Reprod. Med. 49 (2004) 595–601. [46] A. Braga, I. Maesta, D. Short, P. Savage, R. Harvey, M.J. Seckl, Hormonal contraceptive
[15] S. Murdoch, U. Djuric, B. Mazhar, et al., Mutations in NALP7 cause recurrent use before hCG remission does not increase the risk of gestational trophoblastic neo-
hydatidiform moles and reproductive wastage in humans, Nat. Genet. 38 (2006) plasia following complete hydatidiform mole: a historical database review, BJOG
300–302. 129 (8) (2016) 1330–1335.

7
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

[47] Richard Harvey, Measurement of Human Chorionic Gonadotrophin (hCG) in the [71] M.C. Winter, J.A. Tidy, A. Hilts, J. Ireson, S. Gillett, K. Singh, B.W. Hancock, R.E.
management of trophoblastic disease, in: BW Hancock, MJ Seckl, RS Berkowtiz Coleman, Risk adapted single-agent dactinomycin or carboplatin for second-line
(Eds.), Chapter 5 GTD Book. ISSTD Website, 4th., ISSTD, 2015. treatment of methotrexate resistant low-risk gestational trophoblastic neoplasia,
[48] L.A. Cole, Hyerglycosolated hCG, Placenta 31 (8) (2010) 653–664. Gynecol. Oncol. 143 (3) (2016) 565–570.
[49] H.A. Al-Mahdili, G.R. Jones, High-dose hook effect in six automated human chorionic [72] P.A.R. Mora, S.Y. Sun, G.C. Velarde, J.R. Filho, E.H. Uberti, A.P.V. dos Santos Esteves,
gonadotropnin assays, Ann. Clin. Biochem. 47 (pt4) (2010) 383–385. K.M. Elias, N.S. Horowitz, A. Braga, R.S. Berkowitz, Can carboplatin or etoposide re-
[50] A.D. Winder, E. Suarez Mora Am Betty, J.R. Lurain, The “hook effect” causing a neg- place actinomycinod for second-line treatment of methotrexate resistant low-risk
ative pregnancy test in a patient with an advanced molar pregnancy, Gynecol. gestational trophoblastic neoplasia? Gynecol. Oncol. 153 (2) (2019) 277–285.
Oncol. Rep. 21 (2017) 34–36. [73] Y.K. Eysbouts, L.F.A.G. Massuger, J. IntHout, C.A.R. Lok, F.C.G.J. Sweep, F.B.
[51] L.A. Cole, S.A. Khanlian, A. Giddings, S.A. Butler, C.Y. Muller, C. Hammond, E. Kohorn, Ottewanger, The added value of hysterectomy in management of gestational tro-
Gestational trophoblastic diseases: 4. Presentation with persistent low positive phoblastic neoplasia, Gynecol. Oncol. 145 (3) (2017) 536–542.
human chorionic gonadotropin test results, Gynecol. Oncol. 102 (2) (2006) [74] R.J. Osborne, V.L. Filiaci, J.C. Schink, R.S. Mannel, K. Behbackht, et al., Second curet-
165–172. tage for low -risk nonmetastatic gestational trophoblastic neoplasia, Obstet.
[52] A. Braga, B. Torres, M. Burla, I. Maesta, S.Y. Sun, L. Lin, J.M. Madi, E. Uberti, M. Gynecol. 128 (3) (2016) 535–542.
Viggiano, K.M. Elias, R.S. Berkowitz, Is chemotherapy necessary for patients with [75] K. Balachandran, A. Salawu, E. Ghorani, B. Kaur, N.J. Sebire, D. Short, R. Harvey, et al.,
molar pregnancy and human chorionic gonadotropin serum levels raised but falling When to stop human chorionic gonadotrophin (hCG) surveillance after treatment
at 6 months after uterine evacuation? Gynecol. Oncol. 143 (3) (2016) 558–564. with chemotherapy for gestational trophoblastic neoplasia (GTN): a national analy-
[53] R. Agarwal, S. Teoh, D. Short, R. Harvey, P.M. Salvage, M.J. Seckl, Chemotherapy and sis on over 4,000 patients, Gynecol. Oncol. 155 (1) (2019) 8–12.
human chorionic gonadotropin concentrations 6 months after uterine evacuation of [76] J.R. Lurain, D.K. Sing, J.C. Schink, Management of high-risk gestational trophoblastic
molar pregnancy: a retrospective cohort study, Lancet 379 (2012) 130–135. neoplasia: FIGO stages II-IV:risk factor score > or = 7, J. Reprod. Med. 55 (5–6)
[54] Gestational Trophoblastic Neoplasms (Female Reproductive Organs), in: M.B. Amin (2010) 199–207.
(Ed.), AJCC Cancer Staging Manual, Eighth editionSpringer, Chicago, 2017.
[77] M. Alazzam, J. Tidy, R. Osborne, R. Coleman, B.W. Hancock, T.A. Lawrie, Chemother-
[55] M.M. Frijstein, C.A.R. Lok, N.E. van Trommel, M.J. ten Kate-Booij, L.F.A.G. Massuger,
apy for resistant or recurrent gestational trophoblastic neoplasia, Cochrane Database
Lung metastases in low-risk gestational trophoblastic neoplasia: a retrospective co-
Syst. Rev. 13 (1) (2016 Jan), CD008891.
hort study, BJOG 127 (3) (2020) 389–395.
[78] M. Bower, E.S. Newlands, L. Holden, et al., EMA/CO for high-risk gestational tropho-
[56] M.M. Frijstein, C.A.R. Lok, N.E. van Trommel, M.J. Ten Kate-Booji, L.F.A.G. Massuger,
blastic tumours: results from a cohort of 272 patients, J. Clin. Oncol. 15 (1997)
et al., Management and prognostic factors of epithelioid trophoblastic tumors: re-
2636–2643.
sults from the International Society for the Study of trophoblastic diseases database,
Gynecol. Oncol. 152 (2) (2019) 361–367. [79] E.S. Newlands, P.J. Mulholland, L. Holden, M.J. Seckl, G.J. Rustin, Etoposide and cis-
[57] F.E.M. Froeling, R. Ramaswami, P. Papanastasopoulos, B. Kaur, N.J. Sebire, D. Short, platin/etoposide, methotrexate, and actinomycin D (EMA) chemotherapy for pa-
R.A. Fisher, et al., Intensified therapies improve survival and identification of novel tients with high-risk gestational trophoblastic tumors refractory to EMA/
prognostic factors for placental-site and epithelioid trophoblastic tumours, Br. J. cyclophosphamide and vincristine chemotherapy and patients presenting with
Cancer 120 (6) (2019) 587–594. metastatic placental site trophoblastic tumors, J. Clin. Oncol. 18 (4) (2000 Feb)
854–859.
[58] C. Alifrangis, R. Agarwal, D. Short, R.A. Fisher, N.J. Sebire, R. Harvey, et al., EMA/CO for
high-risk gestational trophoblastic neoplasia: good outcomes with induction low- [80] J.R. Lurain, J.C. Schink, Importance of salvage therapy in the management of high-
dose etoposide-cisplatin and genetic analysis, J. Clin. Oncol. 31 (2) (2013 Jan 10) risk gestational trophoblastic neoplasia, J. Reprod. Med. 57 (5–6) (2012) 219–224.
280–286. [81] Y. Mao, X. Wan, W. Lv, X. Xie, Relapsed or refractory gestational trophoblastic neo-
[59] T.A. Lawrie, M. Alazzam, J. Tidy, B.W. Hancock, R. Osborne, First-line chemotherapy plasia treated with the etoposide and cisplatin/etoposide, methotrexate, and actino-
in low-risk gestational trophoblastic neoplasia, Cochrane Database Syst. Rev. 2016 mycin D (EP-EMA) regimen, Int. J. Gynaecol. Obstet. 98 (1) (2007 Jul) 44–47.
(6) (2016). [82] J. Wang, D. Short, N.J. Sebire, I. Lindsay, E.S. Newlands, P. Schmid, et al., Salvage che-
[60] R.J. Osborne, V. Filiaci, J.C. Schink, R.S. Mannel, A. Alvarez Secord, et al., Phase III trial motherapy of relapsed or high-risk gestational trophoblastic neoplasia (GTN) with
of weekly methotrexate or pulsed dactinomycin for low-risk gestational trophoblas- paclitaxel/cisplatin alternating with paclitaxel/etoposide (TP/TE), Ann. Oncol. 19
tic neoplasia: a gynecologic oncology group study, J. Clin. Oncol. 29 (7) (2011) (9) (2008 Sep) 1578–1583.
825–831. [83] J. Yang, Y. Xiang, Wan Xirun, Feng F, Ren Tong. Primary treatment of stage IV gesta-
[61] A. Braga, C. Araujo, P. Mora, A. Paulino Ede Melo, G. Velarde, et al., Comparison of tional trophoblastic noeplasia with floxuridine, dactinomycin, etoposide, and vin-
treatment for low-risk GTN with standard 8-day MTX/FA regimen versus modified cristine (FAEV): a report based on our 10-year clinical experience, Gynecol. Oncol.
MTX/FA regimen without chemotherapy on the weekend, Gynecol. Oncol. 15 (3) 143 (1) (2016) 68–72.
(2020) 598–605. [84] M.J. Kanis, J. Sobecki-Rausch, R. Greendyk, M.E. Dayno, J.R. Lurain, Use of granulocyte
[62] I. Maesta, R. Nitecki, N.S. Horowitz, D.P. Goldstein, et al., Effectiveness and toxicity of colony stimulating factors (G-CSF) with multiagent chemotherapy for gestational
first-line methotrexate chemotherapy in low-risk postmolar gestational trophoblas- trophoblastic neoplasia, J. Reprod. Med. 63 (2018) 209–212.
tic neoplasia: the New England trophoblastic disease center experience, Gynecol. [85] P. Savage, I. Kelpanides, M. Tuthill, D. SHrt, M.J. Seckl, Brain metastases in gestational
Oncol. 148 (1) (2018) 161–167. trophoblastic neoplasia: an update on incidence, management, and outcome,
[63] J.R. Lurain, E.P. Elfstrand, Single-agent methotrexate chemotherapy for the treat- Gynecol. Oncol. 137 (1) (2015) 73–76.
ment of nonmetastatic gestational trophoblastic tumors, Am. J. Obstet. Gynecol. [86] C. Xiao, J. Yang, Y. Xiang, Clinical Management of GTN with brain metastasis: A 20
172 (pt1) (1995) 574–579. year experience in Peking Union Medical College Hospital, China; in Hancock BW,
[64] J.C. Schink, V. Filiaci, H.Q. Huang, J. Tidy, M. Winter, J. Carter, et al., An international Seckl MJ, Berkowitz RS (eds). Chapter 19, GTD Book. ISSTD Website. 4th, ISSTD,
randomized phase III trial of pulse actinomycin-D versus multi-day methotrexate 2015.
for the treatment of low risk gestational trophoblastic neoplasia; NRG/GOG275, [87] M.M. Frijstein, C.A.R. Lok, D. Short, K. Singh, R.A. Fisher, B.W. Hancock, et al., The re-
Gynecol. Oncol. 158 (2) (2020) 354–360. sults of treatment with high-dose chemotherapy and peripheral blood stem cell
[65] G. Mangili, R. Cioffi, S. Danese, L. Frigerio, G. Ferrandina, G. Cormio, E. Rabaiotti, G. support for gestational trophoblastic neoplasia, Eur. J. Cancer 109 (2019 Mar)
Scarfone, A. Gadducci, A. Bergamini, C. Pisano, M. Candiani, Does methotrexate 162–171.
(MTX) dosing in a 8-day MTX/FA regimen for the treatment of low-risk gestational
[88] P.A. Bolze, S. Patrier, J. Massardier, T. Hajri, F. Abbas, A.M. Schott, et al., PD-L1 expres-
trophoblastic neoplasia affect outcome? The MITO-9 study, Gynecol. Oncol. 151 (3)
sion in premalignant and malignant trophoblasts from gestational trophoblastic dis-
(2018) 449–452.
eases is ubiquitous and independent of clinical outcomes, Int. J. Gynecol. Cancer 27
[66] H. Matsui, Y. Itsuka, K. Seki, S. Sekiya, Comparison of chemotherapies with metho-
(3) (2017) 554–561.
trexate, VP-16 and actinomycin D in low risk gestational trophoblastic disease. Re-
[89] E. Ghorani, B. Kaur, R.A. Fisher, D. Short, U. Joneborg, J.W. Carlson, et al.,
mission rates and drug toxicities, Gynecol. Obstet. Investig. 46 (1998) 5–8.
Pembrolizumab is effective for drug-resistant gestational trophoblastic neoplasia,
[67] H.C. Sung, P.C. Wu, H.Y. Yang, Reevaluation of 5-fluorouracil as a single therapeutic
Lancet 390 (10110) (2017 Nov 25) 2343–2345.
agent for gestational trophoblastic neoplasms, Am. J. Obstet. Gynecol. 150 (1984)
69–75. [90] B. You, P.A. Bolze, J.P. Lotz, J. Massardier, L. Gladieff, F. Jody, et al., Avelumab in pa-
tients with gestational trophoblastic tumors with resistance to single-agent chemo-
[68] A. Sita-Lumsden, D. Short, I. Lindsay, N.J. Sebire, D. Adjogatse, M.J. Seckl, P.M. Savage,
therapy: cohort Aof the TROPHIMMUN phase II trial, J. Clin. Oncol. 38 (27) (2020)
Treatment outcomes for 618 women with gestational trophoblastic tumours follow-
3129–3137.
ing a molar pregnancy at Charing cross hospital, 2000-2009, Br. J. Cancer 107 (11)
(2012) 1810–1814. [91] K.M. Doll, J.T. Soper, The role of surgery in the management of gestational tropho-
[69] A. Braga, G. Paiva, E. Ghorani, F. Freitas, G. Guillermo Coca Velande, B. Kour, N. blastic neoplasia, Obstet. Gynecol. Surv. 68 (7) (2013) 533–542.
Unsworth, J. Lozano-Kuehne, A. Viena dos Santos Esteves, Rezende FilhoJ, J. Amim, [92] E. Lehman, D.M. Gershenson, T.W. Burke, C. Levenback, E.G. Silva, M. Morris, Salvage
X. Aguiar, N. Sarwar, K.M. Elias, N.S. Horowitz, R.S. Berkowtiz, M.J. Seckl, Predictors surgery for chemorefractory gestational trophoblastic disease, J. Clin. Oncol. 12
for single-agent resistance in FIGO score 5 or 6 gestational trophoblastic neoplasia: a (1994) 2737–2742.
multicenter retrospective cohort study, Lancet Oncol. 22 (2021) 1188–1198. [93] C. Lan, Y. Li, J. He, J. Liu, Placental site trophoblastic tumor: lymphatic spread and
[70] C. Lybol, F.C. Sweep, R. Harvey, H. Mitchell, D. Short, C.M. Thomas, et al., Relapse possible target markers, Gynecol. Oncol. 116 (3) (2010) 430–437.
rates after two versus three consolidation courses of methotrexate in the treatment [94] X. Shen, Y. Xiang, L. Guo, F. Feng, X. Wan, Y. Xiao, et al., Fertility preserving treatment
of low-risk gestational trophoblastic neoplasia, Gynecol. Oncol. 125 (3) (2012) in young patients with placental site trophoblastic tumors, Int. J. Gynecol. Cancer 22
576–579. (2012) 869–874.

8
N.S. Horowitz, R.N. Eskander, M.R. Adelman et al. Gynecologic Oncology xxx (xxxx) xxx

[95] B. Kaur, D. Short, R.A. Fisher, P.M. Savage, M. Seckl, N.J. Sebire, Atypical placental site [97] P. Savage, R. Cooke, J. O’Nions, J. Krell, A. Kwan, M. Camarata, et al., Effects of single-
nodule (APSN) and association with malignant gestational trophoblastic disease; a agent and combination chemotherapy for gestational trophoblastic tumors on risk
clinicopathologic study of 21 cases, Int. J. Gynecol. Pathol. 34 (2) (2015) 152–158. of second malignancy and menopause, J. Clin. Oncol. 33 (5) (2015) 472–478.
[96] U. Joneborg, L. Coopmans, N. van Trommel, M.J. Seckl, C.A.R. Lok, Fertility and preg- [98] N.J. Sebire, R.A. Fisher, M. Foskett, et al., Risk of recurrent hydatidiform mole and
nancy outcome in gestational trophoblastic disease, Int. J. Gynecol. Cancer 31 (3) subsequent pregnancy outcome following complete or partial hydatidiform molar
(2021) 399–411. pregnancy, BJOG 110 (2003) 22–26.

También podría gustarte