You are on page 1of 9

Review

Gestational trophoblastic neoplasia—pathogenesis and


potential therapeutic targets
Ie-Ming Shih

Lancet Oncol 2007; 8: 642–50 Gestational trophoblastic neoplasia comprises a unique group of human neoplastic diseases that derive from fetal
Departments of Pathology, trophoblastic tissues and represent semiallografts in patients. This group is composed of choriocarcinoma, placental-
Oncology, and Gynecology and site trophoblastic tumour, and epithelioid trophoblastic tumour, and many forms are derived from the precursor
Obstetrics, Johns Hopkins
lesions, hydatidiform moles. Although most patients with gestational trophoblastic neoplasia are cured by
Medical Institutions,
Baltimore, MD, USA chemotherapy and tumour resection, some patients suffer from metastatic diseases that are refractory to conventional
(I-M Shih MD) chemotherapy. Therefore, new therapeutic regimens are needed to reduce the toxic effects associated with current
Correspondence to: chemotherapy and to salvage the occasional non-operable patients with recurrent and chemoresistant disease. Until
Dr Ie-Ming Shih the fundamental biology of gestational trophoblastic neoplasia becomes more clearly understood, development of a
shihie@yahoo.com
new treatment will remain empirical. This review will briefly summarise the recent advances in understanding the
molecular aetiology of this group of diseases and highlight the molecules that can be potentially used for therapeutic
targets to treat metastatic gestational trophoblastic neoplasia.

Introduction aberrant placental derivatives; and true gestational


Gestational trophoblastic diseases represent a spectrum trophoblastic neoplasia.2,3
of related disorders including benign trophoblastic The term gestational trophoblastic neoplasia encom-
lesions, premalignant hydatidiform moles, clinically passes a group of interrelated but distinct tumours that
malignant invasive hydatidiform moles, and neoplastic include choriocarcinoma, placental-site trophoblastic
diseases. Hydatidiform moles, especially the invasive tumour (PSTT), and epithelioid trophoblastic tumour
moles, are sometimes clinically considered as gestational (ETT; figure 1). Gestational trophoblastic neoplasms are
trophoblastic neoplasia because they can locally invade unique among human neoplastic diseases because they
and distantly metastasise, but, biologically, they represent are genetically related to fetal tissues (ie, trophoblastic
abnormally formed placental tissues rather than true cells from placentas) and, therefore, represent semi-
neoplasia. Therefore, from the perspectives of patho- allografts in patients.
biology, gestational trophoblastic diseases can be broadly Although there has been substantial interest in
divided into three groups (panel), modified from the exploring the pathogenesis of these neoplasms, studies
2003 WHO classification1: benign trophoblastic lesions on their molecular aspects are challenging for several
(placental-site nodule and exaggerated placental reaction), reasons. First, gestational trophoblastic neoplasms are
which are non-neoplastic lesions; hydatidiform moles rare diseases and the cumulative incidence rate for
(complete, partial, and invasive moles), which are gestational choriocarcinoma, for example, continues to
decline. As a result, tissue specimens, especially those
from fresh tumours, are scarce for molecular analysis.
Second, many metastatic gestational trophoblastic neo-
plasms, which are clinically classified under the rubric of
persistent gestational trophoblastic diseases are not
usually obtained for pathological classification and
characterisation. Therefore, their natures remain intrig-
uing, because whether persistent gestational tropho-
blastic disease represents an invasive hydatidiform mole,
a metastatic choriocarcinoma, PSTT, or ETT is not
known. An absence of such clinicopathological correlation
compromises attempts to further understand the
biological nature of gestational trophoblastic neoplasia.
Third, because of the above reasons, reagents, such as
cell lines and animal models, are limited for mechanistic
studies. For example, only a few choriocarcinoma cell
lines and one PSTT cell line are currently available for
molecular and cell-biology studies.
Despite these challenges, progress has been made in
Figure 1: ETT (arrow) in a hysterectomy specimen of a 32-year-old woman
recent years towards a better understanding of the
ETT was first diagnosed based on endometrial curettage and patient subsequently underwent total hysterectomy.
1 year after surgery, metastatic ETT developed in the lung and was treated with chemotherapy and lung resection. molecular aetiology underlying the development of these
A second recurrent tumour, resistant to prior chemotherapy, developed 1 year later. neoplasms. This review will briefly summarise recent

642 http://oncology.thelancet.com Vol 8 July 2007


Review

According to this model, choriocarcinoma is the most


Panel: Gestational trophoblastic diseases primitive trophoblastic tumour whereas PSTT and ETT
Benign trophoblastic lesions are relatively more differentiated. This hypothesis
Exaggerated placental reaction explains the existence of gestational trophoblastic
Placental-site nodule neoplasia with a mixed histological feature, including
choriocarcinoma, PSTT, and ETT. This new model might
Hydatidiform moles (abnormally formed placentas) also help to explain the findings previously reported by
Complete mole Mazur8 who described ETT that occurred after intense
Partial mole chemotherapy of metastatic choriocarcinomas in the
Invasive mole lung. In the patients described by Mazur, it is plausible
Trophoblastic neoplasia that chemotherapeutic agents allow choriocarcinoma
Choriocarcinoma cells to differentiate into an ETT phenotype, which is
Placental-site trophoblastic tumour more refractory to chemotherapy than the original chorio-
Epithelioid tophoblastic tumour carcinoma counterpart. As a result, ETT predominated
the choriocarcinoma after chemotherapy.
Alongside studies of the histogenesis of gestational
advances and highlight the molecules that could trophoblastic neoplasia, several studies have focused on
potentially be used for therapeutic targets to treat meta- the biology of these neoplasms by characterising the
static gestational trophoblastic neoplasia. expressions of genes that are important in tumorigenesis.
The pathogenesis of each type of neoplasm will now be
Pathogenesis of trophoblastic tumours summarised. Furthermore, advances in the pathogenesis
Previous clinical, pathological, and molecular studies of hydatidiform moles will be briefly highlighted because
have provided fundamental insights into the pathogenesis hydatidiform moles represent the precursor lesions in
of hydatidiform moles,4 but the molecular and cellular some gestational trophoblastic neoplasms and invasive
basis in the development of true gestational trophoblastic hydatidiform moles are, sometimes, clinically considered
neoplasia remain poorly understood. as gestational trophoblastic neoplasia because they can
Gestational trophoblastic neoplasia has long been locally invade and distantly metastasize.
thought to be a homogeneous group of diseases arising
from neoplastic transformation of trophoblastic cells. Choriocarcinoma
However, recent clinicopathological studies have provided Gestational choriocarcinoma is a highly malignant
new evidence that there are at least three distinct types of epithelial tumour that can be associated with any type of
gestational trophoblastic neoplasm including the most gestational event, most often a complete hydatidiform
common type, choriocarcinoma, and the less common mole.4 Several molecular studies have been done to
ones, PSTT and ETT. Molecular analysis of gestational establish the expression of tumour-associated proteins in
trophoblastic neoplasia is largely based on the choriocarcinoma. In the P53 pathway, overexpression of
characterisation of gene-expression profiles in various
types of these neoplasms and the reference of their
unique gene-expression patterns to different trophoblastic Cytotrophoblast
Syncytiotrophoblast
subpopulations in healthy early placentas.2 The main
conclusion from these studies is that, after neoplastic Normal
transformation of trophoblastic stem cells, presumably trophoblast
cytotrophoblast stem cells, specific differentiation progra- Extravillous trophoblast Extravillous trophoblast
mmes dictate the type of trophoblastic tumour that (intermediate trophoblast) (intermediate trophoblast)
at implantation site at chorion laeve
develops (figure 2).5
Of interest, is the fact that these patterns of
differentiation in gestational trophoblastic neoplasia
recapitulate the stages of early placental development.2,3,5–7 Neoplastic
Choriocarcinoma is composed of variable amounts of transformation

neoplastic cytotrophoblast, syncytiotrophoblast, and extra-


villous (intermediate) trophoblast and resembles the
previllous blastocyst, which is composed of a similar Trophoblastic
neoplasia
mixture of trophoblastic subpopulations. By contrast, the
neoplastic cytotrophoblast in PSTT differentiates mainly
Placental-site Epithelial trophoblastic
into extravillous (intermediate) trophoblastic cells in an trophoblastic tumour
Choriocarcinoma
tumour
implantation site whereas the neoplastic cytotrophoblast
in ETT differentiates into chorionic-type extravillous
(intermediate) trophoblastic cells in the chorion laeve.5 Figure 2: Proposed model of pathogenesis for gestational trophoblastic neoplasia

http://oncology.thelancet.com Vol 8 July 2007 643


Review

the P53 protein has been detected in choriocarcinoma,9 The development of a choriocarcinoma might also be
but mutational analysis of TP53 has failed to show related to tumour microenvironment—eg, immune
somatic mutations.10–12 The P53-associated protein, regulation and stromal tissues. HLA-G, a non-classic
MDM2, has also been seen to be overexpressed in chorio- MHC class I molecule, has been shown to participate in
carcinomas.9,13 Because upregulated MDM2 protein is immune regulation and response. The membrane-bound
associated with various human cancers, overexpression and secreted HLA-G molecules are able to inactivate the
of MDM2 in choriocarcinomas might overcome growth local immune response and, therefore, help tumour cells
suppression by wild-type P53 proteins and, therefore, escape from immune surveillance. Trophoblastic cells in
contribute to the development of choriocarcinoma. To normal placentas and gestational trophoblastic neoplasms
show the biological role of MDM2 in choriocarcinoma, have been shown to upregulate HLA-G expression.28,29 In
Wang and co-workers14 used an MDM2 antisense fact, in the human cancers analysed, gestational tropho-
oligonucleotide and showed a synergistic antitumour blastic neoplasms, including choriocarcinoma, contain
effect with DNA-damaging agents in JAR choriocarcinoma the highest levels of HLA-G expression.28 Therefore,
xenografts in mice. HLA-G expressed in choriocarcinomas might assist
By use of a PCR-based subtracted fragmentary cDNA tumour cells to escape from the host immune recognition
library between normal chorionic villi and a chorio- and promote tumour growth.29
carcinoma cell line, Asanoma and colleagues15,16 identified Another explanation for tumour growth could be
a novel homeobox gene designated NECC1 (located on matrix metalloproteinases (MMP) and tissue inhibitors
4q11-q12) that might be involved in the development of of metalloproteinases (TIMPs), which have a key role in
choriocarcinoma. In this study, the expression of NECC1 tumour invasion. MMP is a zinc-dependent proteinase
was lost in all choriocarcinoma cell lines and most that degrades and remodels the extracellular matrix
choriocarcinoma tissues, but healthy adult tissues, surrounding tumour cells. Choriocarcinomas have been
including trophoblastic cells in healthy placentas, shown to express increased concentrations of MMP and
expressed abundant NECC1. The investigators further decreased concentrations of TIMP to help with tumour
showed that engineered expression of NECC1 in invasion and metastasis,30 and this finding could explain
choriocarcinoma cell lines suppressed tumorigenicity why choriocarcinoma is highly metastatic if left
and induced terminal differentiation.15,16 untreated.
Other genes that are potentially involved in the
development of choriocarcinoma include epidermal PSTT and ETT
growth factor receptor (EGFR),17 DOC-2/hDab2 PSTT and ETT are rare gestational trophoblastic
(a candidate tumour suppressor gene),18,19 putative neoplasms and, so far, have not been well studied at the
tumour suppressor loci at chromosomes 7p12-7q11.2320 molecular level, although both tumours have been
and 8p12-p21,21 and the gene for ras GTPase activating described in the published work on diagnostic pathology.2
protein.22 Synergistic upregulation of c-MYC, c-ERB-2, Both forms of these neoplasms can be derived from any
c-FMS, and BCL-2 oncoproteins have also been type of gestational event, including complete and partial
suggested to have an important role in the pathogenesis hydatidiform moles.7,31–33 The origin of both tumours has
of choriocarcinoma.10 Conversely, promoter hyper- puzzled investigators since their discovery because,
methylation of E-cadherin, HIC-1, P16, and TIMP3 has unlike choriocarcinomas, PSTTs and ETTs can develop
been frequently seen in choriocarcinomas, suggesting long after the prior gestational events. The trophoblastic
that downregulation of these genes by promoter nature of PSTT and ETT has been shown by molecular
methylation could be involved in the development of genetic analysis, which shows that they contain new
choriocarcinomas.23 In addition to the candidate (paternal) alleles not present in adjacent healthy uterine
approaches described above, Vegh and colleagues24 tissue.34–36 Additionally, both tumours express trophoblast-
compared the gene-expression pattern between healthy associated markers, including HLA-G and HSD3B1.29 On
trophoblast and choriocarcinoma cell lines using cDNA the basis of published work, most cases of PSTT and ETT
microarrays. In their study, one of the downregulated are thought to be benign, especially for those tumours
genes in choriocarcinoma cells, HSP-27, was selected that are confined to the uterus, but about 15–25% of cases
for characterisation because the HSP-27 protein has are malignant and present with local invasion and distant
been implicated in the development of several human metastasis.37–40 In some cases, recurrent or metastatic
neoplastic diseases.25 The investigators noted that PSTT and ETT can occur in patients long after the initial
choriocarcinoma tissues had a lower level of HSP-27 treatment but, unfortunately, analysis of histopathological
expression compared with trophoblastic cells in early features and molecular changes has not provided reliable
placentas. This downregulation of HSP-27 might markers to predict the long-term clinical behaviour of
contribute to the high sensitivity of choriocarcinomas to PSTT and ETT.41
chemotherapeutic agents because HSP-27 proteins have By contrast to the normal implantation site where
been shown to contribute to drug resistance in invasion of the extravillous (intermediate) trophoblast is
malignant cells.26,27 tightly regulated and confined to the inner third of the

644 http://oncology.thelancet.com Vol 8 July 2007


Review

myometrium, tumour cells of PSTT and ETT are highly usually have the karyotype 46,XX with both sets of
invasive and infiltrate deeply into the myometrium. chromosomes originating from the paternal complement
Although PSTT and ETT share similar clinical features, as a result of haploid genome (23X) duplication. This
careful examination of tumour histology and gene- chromosomal composition is referred to as androgenetic,
expression patterns shows that PSTT and ETT are meaning that genetic material is derived exclusively
composed of different extravillous (intermediate) tropho- from paternal DNA. Partial moles, however, are generally
blastic cells.3,7,38 During neoplastic transformation, the triploid (XXY) with two sets of chromosomes from the
tumour cells in PSTT and ETT are likely to assume paternal complement and a haploid set from the
unique differentiation programmes toward the extra- maternal genetic content. This triploid chromosomal
villous (intermediate) trophoblast in the implantation composition is known as diandric or biparental. Under-
site and the extravillous (intermediate) trophoblast in the standing the cytogenetic constituents that characterise
chorion laeve, respectively (figure 2). complete and partial moles is fundamental for the
Molecularly, PSTT is associated with abnormal future study of how maternally or paternally imprinted
expression of cell-cycle regulatory genes, including those genes contribute to different types of molar pregnancy.
for cyclins, cyclin-dependent kinases, and P53.42 Most Despite the well-established cytogenetic findings
PSTTs are diploid, based on flow cytometric DNA associated with different hydatidiform moles, the
analysis, but PSTTs might also be triploid and, thus, be molecular aetiology underlying the development of
derived from a partial hydatidiform mole.31 ETT expresses molar pregnancy remains elusive. One of the most
markers of epithelial cells, including cytokeratin, exciting studies showed that MALP7 is mutated in
epithelial membrane antigen, E-cadherin, and EGFR, patients with familial and recurrent biparental moles.44
and this finding is compatible with their characteristic MALP7 encodes a member of the CATERPILLER protein
epithelioid growth pattern. family that is potentially involved in inflammatory and
The P63 gene, a transcription factor belonging to the apoptotic processes. More work is needed to elucidate
P53 family, is expressed in ETT but not in PSTT. P63 has the possible mechanisms of mutant NALP7 proteins in
various isoforms that are classified into two groups the pathogenesis of hydatidiform moles, such as during
designated as TAP63 and ∆NP63 isoforms.38 The oogenesis and implantation.
cytotrophoblast on chorionic villi has been shown to Additionally, several oncogenes and tumour-suppressor
express the ∆NP63 isoform, whereas extravillous genes have been studied in complete moles. Similar to
(intermediate) trophoblastic cells in the chorion laeve choriocarcinomas, synergistic upregulation of c-MYC,
and ETT express the TAP63.38 Further studies are needed c-ERB-2, c-FMS, and BCL-2 oncoproteins has been
to delineate the functional role of TAP63 in ETT. suggested in the pathogenesis of complete moles.10
Expression of cyclin E is found in ETT but not in the Mutational analysis of K-ras and P53 did not show
extravillous (intermediate) trophoblastic cells of the mutations in complete moles, but overexpression of P53
chorion laeve, the normal counterpart of ETT.43 This and MDM2 proteins has been recorded.9 Furthermore,
finding suggests that cyclin E probably plays a part in the role of promoter methylation has been studied in
neoplastic transformation of ETT because an oncogenic hydatidiform moles and gestational trophoblastic
role of cyclin E has been shown in other neoplastic neoplasia. Frequent promoter hypermethylation and
diseases. decreased expression of PTEN, CDH1, HIC-1, and
CDKN2A have been found in hydatidiform moles
Hydatidiform moles compared with healthy placentas,23,45 suggesting that
Hydatidiform moles include complete, partial, and inactivating genes that might participate in tumour
invasive moles, the latter representing a hydatidiform suppression by promoter methylation could be one of the
mole that becomes invasive or is exported to a distant mechanisms in the development of molar pregnancy.
anatomical site. Although hydatidiform moles are not Furthermore, promoter hypermethylation of CDKN2A
classed as true gestational trophoblastic neoplasia, alone, or combined with CDH1, was significantly
postmolar gestational trophoblastic neoplasia can be correlated with subsequent development of gestational
seen in many patients with hydatidiform moles, trophoblastic neoplasia.23
especially complete moles. This finding indicates that
hydatidiform moles are the precursor lesions of some New molecular targets with therapeutic potential
gestational trophoblastic neoplasms. Because of the Choriocarcinoma is among the few human cancers that are
close relationship of these moles with gestational highly responsive to chemotherapy and, unlike other
trophoblastic neoplasia, their pathogenesis will be briefly malignant tumours, metastatic choriocarcinomas are
highlighted. potentially curable by combined chemotherapy and
Advances in cytogenetics, and in the molecular and adjuvant surgical procedures.46,47 Despite this fact, a small
immunohistochemical characterisation of hydatidiform but significant proportion of choriocarcinoma patients
moles, have shown a distinct pathogenesis in complete develop recurrent diseases after primary treatment.48,49 By
and partial moles. Most complete moles are diploid and contrast with choriocarcinoma, PSTT and ETT are generally

http://oncology.thelancet.com Vol 8 July 2007 645


Review

more refractory to conventional chemotherapy. Patients


Molecular targets Possible drugs
with PSTT and ETT might develop metastatic diseases and
some patients will have terminal disease.7,37 Therefore, c-MYC Antisense oligonucleotides

new therapeutic regimens are needed to reduce the toxic EGFR Cetuximab, gefitinib, erlotinib
effects associated with current chemotherapy—especially MAPK CI-1040, PD59089
the combination regimens, ie, etoposide, methotrexate, MMP Marimastat
dactinomycin; etoposide, methotrexate, dactinomycin, mTOR AP23573, RAD001, CCI-779
cyclophosphamide, vincristine; and etoposide, cisplatin, MAPK=mitogen-activated protein kinase. mTOR=mammalian target of
etoposide, methotrexate, and dactinomycin—and to salvage rapamycin.
the occasional non-operable patients with metastatic
Table 1: Molecular targets and examples of possible clinical drugs for
chemoresistant diseases. gestational trophoblastic neoplasia
Target-based treatment designed to inactivate molecular
pathways that are essential for tumour-cell growth and
survival has shown clinical promise. Unlike standard oligonucleotides and siRNA that silence c-MYC
chemotherapy, which affects most proliferating cells, expression.54,55 Some of the antisense oligonucleotides
inhibitors that target specific pathways selectively eliminate have successfully completed phase I clinical trials and
tumour cells to achieve maximum therapeutic effects and are at an advanced stage of drug development. Therefore,
minimum adverse side-effects. These new anticancer future patients with metastatic gestational trophoblastic
agents include gefitinib, a small kinase inhibitor that neoplasia could benefit from anti-MYC agents in
targets EGFR, and trastuzumab, which is a humanised combination with conventional treatment.
antibody that targets ERBB2 receptors. Because the merits
of molecular targeting have been shown by clinical studies, EGFR
targeted treatment might also be applicable to treat EGFR is a transmembrane-receptor tyrosine kinase that
metastatic gestational trophoblastic neoplasms. In the belongs to the ERBB-related kinase family. Many
following sections, several molecular targets that are epithelial tumours overexpress EGFR and harbour
expressed in gestational trophoblastic neoplasia tissues somatic mutations, leading to constitutive activation of
and cell lines are discussed (table 1). However, there are the EGFR pathway.56 High levels of EGFR expression
many other inhibitors that target emerging cancer- have been detected in choriocarcinomas, PSTT, and
associated molecules and new therapeutic approaches, ETT.7,17,57 Additionally, the expression levels of EGFR in
such as anti-human chorionic gonadotropin antibody choriocarcinomas and complete hydatidiform moles
treatment and immunotherapy, might also prove useful were significantly higher than those of trophoblastic cells
as new therapeutics in gestational trophoblastic neoplasia in healthy placentas and partial hydratidiform moles.17
in future. Further analysis showed that intense EGFR immuno-
reactivity in the extravillous (intermediate) trophoblastic
c-MYC proto-oncogene cells of a complete mole was seen to be significantly
The activation and overexpression of the c-MYC proto- correlated with the development of a persistent postmolar
oncogene is one of the most frequent molecular changes gestational trophoblastic neoplasm.17 Expression of
in human cancers.50 In trophoblastic lesions, high levels ERBB-3 and ERBB-4, however, did not differ between
of c-MYC expression have been detected in chorio- gestational trophoblastic neoplasms and healthy placental
carcinomas and complete hydatidiform moles, and an tissues.17
increased expression of c-MYC proteins might suppress The anti-EGFR regimen has become the prototype of
differentiation and enhance cellular proliferation in target-based treatment for cancer. Clinical and survival
gestational trophoblastic neoplasia.10 Direct evidence of benefits of anti-EGFR treatment have been shown in
tumour dependence on c-MYC for growth and survival patients with non-small-cell lung cancer and other
comes from elegant studies showing that induction of neoplastic diseases. The repertoire of cancer types that
c-MYC expression results in tumour formation, and that might respond to anti-EGFR treatment is expanding,
the blocking c-MYC activation generally leads to tumour including head and neck and pancreatic carcinomas.
stasis or regression.51,52 Drugs that target EGFR, including cetuximab, gefitinib,
In recent years, the targeting c-MYC oncoproteins has and erlotinib, have emerged as effective treatments
been proposed as a new anticancer regimen.53,54 For against various malignant diseases. Therefore, patients
example, several strategies that aim to inactivate c-MYC with metastatic gestational trophoblastic neoplasia
function and expression are being developed. These might also benefit from anti-EGFR treatment in
approaches use: inhibitors that block c-MYC expression, combination with other anticancer agents.
eg, triple-helix-forming oligonucleotides; molecules that
disrupt MYC-MAX interaction; agents that block the Mitogen-activated protein kinase
functions of c-MYC-regulated genes, eg, cationic Mitogen-activated protein kinase (MAPK) has a role in
porphyrins and ornithine decarboxylase; and antisense signal transduction, and activation (phosphorylation) of

646 http://oncology.thelancet.com Vol 8 July 2007


Review

MAPK results in the regulation of downstream effectors This glycoprotein is co-localised with an adhesion
to coordinate various cellular activities, including molecule, called carcinoembryonic antigen-related cell
proliferation, differentiation, apoptosis, angiogenesis, adhesion molecule 1 (CEACAM1), in the extravillous
and migration. Aberrant molecular changes that result (intermediate) trophoblast of a healthy placenta and also,
in the constitutive activation of MAPK contribute to probably, in choriocarcinoma cells.68 The expression of
tumour development, with activation being caused by osteopontin has been shown to enhance the invasion of
either a genetic or epigenetic event. For example, CAECAM1-expressing trophoblastic cells.68 Likewise,
activating mutations in KRAS and BRAF, the upstream expression of highly phosphorylated osteopontin activates
genes of MAPK, have been found in various human the PI3K/mTOR/P70S6 kinase pathway and triggers
carcinomas.58,59 Additionally, MAPK activation can result migration of CAECAM1-expressing trophoblastic cells in
from growth-factor stimulation in the tumour micro- choriocarcinoma cells grown in culture. Therefore, this
environment.60 tumour phenotype could be suppressed by the
Kobel and co-workers61 studied MAPK activation in compounds LY294002 and rapamycin, which inhibit
PSTT tissues using immunohistochemistry. They PI3K and mTOR, respectively.66
showed that the activated (phosphorylated) form of Over the past years, several mTOR inhibitors have been
MAPK was expressed in 84% of cases, but not in healthy developed and tested in preclinical tumour models,69,70
extravillous (intermediate) trophoblastic cells, the and, more importantly, several of these inhibitors,
normal counterparts of PSTT.61 However, because the including AP23573, RAD001, and CCI-779, have been
total concentration of MAPK was similar between PSTT tested in clinical trials as single agents or in combination
and healthy extravillous (intermediate) trophoblastic with other drugs in patients with solid or haematological
cells, the investigators proposed that hyperactive MAPK neoplastic diseases.71,72 Ongoing clinical studies involving
was not likely to be the result of MAPK-gene upreg- mTOR inhibitors will, hopefully, identify the efficacy of
ulation, but rather a result of other epigenetic events these drugs in treating patients with cancer and provide
that lead to its activation. To characterise the biological the foundation for future use of mTOR inhibitors in
role of MAPK activation in PSTT, they established the patients with gestational trophoblastic neoplasia.
first PSTT cell line, implantation-site trophoblastic cells,
in which the cells were highly motile and invasive in Matrix metalloproteinase (MMP)
vitro. Treatment with the mitogene inhibitors, CI-1040 As previously discussed, the upregulation of MMP and
and PD59089, which prevent activation of MAPK, downregulation of TIMP in choriocarcinomas suggest
significantly reduced the motility and invasion of IST-2 that patients with metastatic, chemoresistant gestational
cells in vitro.61 These findings suggest a functional role trophoblastic neoplasia might benefit from MMP
of MAPK activation in the motility and invasion of PSTT inhibitors. Marimastat is an MMP inhibitor and
cells. Because CI-1040 and other mitogen inhibitors that preclinical studies in animal-tumour models have shown
prevent MAPK activation have already been tested in that MMP inhibitors restrict the growth of solid tumours,
clinical trials,62–64 these inhibitors could also be assessed inhibit metastatic potential, and block tumour angio-
for treating patients with metastatic PSTT who have not genesis. This inhibitor is an orally bioavailable drug that
responded to conventional treatment. Whether or not has also been assessed in clinical trials of patients with
inhibitors of the MAPK pathway can reduce the invasive- advanced non-small-cell lung cancer, relapsed prostate
ness and metastatic potential of PSTT would be intere- cancer, glioblastoma multiforme, and several other types
sting to learn. of cancer. In general, modest toxic effects have been
reported, on the basis of several phase I clinical trials,73
Mammalian target-of-rapamycin (mTOR) and marimastat has been shown to delay progression in
mTOR is a member of the phosphatidyl inositol 3´ kinase patients with biochemically relapsed prostate cancer.74
(PI3K) family and is one of the key molecules that Further studies need to be done to establish the efficacy
participate in the cellular signalling network. On of marimastat and other new MMP inhibitors in treating
stimulation by miscellaneous growth signals, a cascade metastatic choriocarcinomas, and to find out if the
of signalling occurs from PI3K, Akt, and mTOR to P70S6 clinical outcome can be improved in patients with
kinase, resulting in a variety of cellular responses, choriocarcinoma who do not respond to conventional
including cellular migration and proliferation. Osteo- chemotherapy.
pontin, a cell-surface phosphorylated glycoprotein, repre-
sents one of the signals that trigger activation of the Promises and challenges of target-based
PI3K/mTOR/P70S6 pathway and has been known to treatment
have an important role during implantation by aiding The molecules discussed above have been shown to be
attachment of the trophectoderm of a blastocyst to the expressed in neoplastic trophoblastic cells and, thus, their
surface epithelium of the endometrium.65 In gestational inhibitors represent attractive new therapeutics in
trophoblastic neoplasia, osteopontin is highly expressed chemoresistant gestational trophoblastic neoplasia.
in choriocarcinoma tissues and the derived cell lines.66,67 However, there are several challenges ahead before

http://oncology.thelancet.com Vol 8 July 2007 647


Review

molecular target-based treatment becomes a reality for


this group of diseases. First, the expression frequency of Search strategy and selection criteria
the target genes in a large series of neoplastic tissues is Published and unpublished data for this review were
not yet known, because such clinical samples are rare for identified by searching PubMed. Key words used to identify
study. Furthermore, studies of gestational trophoblastic papers included, “trophoblast”, “gestation”, “neoplasm”,
diseases have primarily focused on hydatidiform moles “metastasis”, “target”, “therapy”, “pathogenesis”,
and choriocarcinoma. PSTT and ETT have rarely been “choriocarcinoma”, “placenta”, “mole”, and “epithelioid”. The
studied so knowledge of these forms of gestational search strategy was not limited by year of publication, but
trophoblastic neoplasia is limited. Second, it is crucial to only articles published in English were selected.
question whether the anticancer effects seen in preclinical
models using gestational trophoblastic neoplasia cell
lines can be translated to treatment efficacy in vivo. light on the pathogenesis of gestational trophoblastic
Third, the clinical usefulness of applying molecular neoplasia but also provide an array of candidate molecules
inhibitors in gestational trophoblastic neoplasia depends with therapeutic potential. Characterisation of the
on careful correlation studies between clinical response molecular changes that have been targeted to treat several
and the genetic and molecular changes of genes and the other human cancers in gestational trophoblastic neo-
pathways they control, respectively. For example, plasia would open an exciting avenue for target-based
mutational analysis of EGFR and members of the PI3K treatment of metastatic forms of this disease that are
and MAPK pathways would be important, in addition to refractory to conventional treatment.
the assessment of their gene-expression levels. The Conflicts of interest
results of such genotype–phenotype correlation studies The author declared no conflict of interest.
would be essential for the development of tailored References
treatment in order to keep potential side effects to a 1 Genest DR, Berkowitz RS, Fisher RA. Gestational trophoblastic
disease. Lyon: IARC press, 2003.
minimum and optimise therapeutic effects. Fourth, given
2 Shih IeM, Kurman RJ. Molecular basis of gestational trophoblastic
the fact that gestational trophoblastic neoplasia is rare diseases. Curr Mol Med 2002; 2: 1–12.
and most patients can be cured by the current treatment, 3 Shih IM, Kurman RJ. The pathology of intermediate trophoblastic
recruitment of patients with this disease in clinical trials tumors and tumor-like lesions. Int J Gynecol Pathol 2001; 20: 31–47.
4 Soper JT. Gestational trophoblastic disease. Obstet Gynecol 2006;
can be challenging because only a limited number of 108: 176–87.
patients qualify and are available for such studies. 5 Mao TL, Kurman RJ, Huang CC, Lin MC, Shih IM.
Nevertheless, drugs that inhibit the expression or function Immunohistochemistry of choriocarcinoma: an aid in differential
diagnosis and in elucidating pathogenesis. Am J Surg Pathol 2007;
of the molecular targets have been assessed in both (in press).
preclinical and clinical trials of various cancer types. 6 Shih IM, Seidman JD, Kurman RJ. Placental site nodule and
With results from ongoing studies soon to emerge and characterization of distinctive types of intermediate trophoblast.
new inhibitors in the development pipeline, future Hum Pathol 1999; 30: 687–94.
7 Shih IM, Kurman RJ. Epithelioid trophoblastic tumor: a neoplasm
patients with gestational trophoblastic neoplasia should distinct from choriocarcinoma and placental site trophoblastic
benefit from new waves of target-based treatment that tumor simulating carcinoma. Am J Surg Pathol 1998;
have been primarily developed to treat other types of 22: 1393–403.
8 Mazur MT. Metastatic gestational choriocarcinoma. Unusual
cancer. Effective multicentre collaborative networks pathologic variant following therapy. Cancer 1989; 63: 1370–77.
should be developed or better orchestrated to aid with 9 Fulop V, Mok SC, Genest DR, Gati I, Doszpod J, Berkowitz RS. p53,
these clinical studies and to share valuable clinical p21, Rb and mdm2 oncoproteins. Expression in normal placenta,
partial and complete mole, and choriocarcinoma. J Reprod Med
specimens, in order to characterise emerging targets 1998; 43: 119–27.
with therapeutic potential in gestational trophoblastic 10 Fulop V, Mok SC, Genest DR, Szigetvari I, Cseh I, Berkowitz RS.
neoplasia. With this continuous effort, we will be well- c-myc, c-erbB-2, c-fms and bcl-2 oncoproteins. Expression in
positioned to further establish the pathogenesis of this normal placenta, partial and complete mole, and choriocarcinoma.
J Reprod Med 1998; 43: 101–10.
rare but intriguing disease and offer a better treatment 11 Chen CA, Chen YH, Chen TM, et al. Infrequent mutation in tumor
alternative for patients. suppressor gene p53 in gestational trophoblastic neoplasia.
Carcinogenesis 1994; 15: 2221–23.
12 Shi Y-F, Xie X, Zhao C-L, et al. Lack of mutation in tumour-
Conclusion suppressor gene p53 in gestational trophoblastic tumours.
Recent advances in molecular studies of trophoblastic Br J Cancer 1996; 73: 1216–19.
cells in healthy placentas and in gestational trophoblastic 13 Landers JE, Haines DS, Strauss JF 3rd, George DL. Enhanced
translation: a novel mechanism of mdm2 oncogene overexpression
neoplasms have indicated that these neoplasms recap- identified in human tumor cells. Oncogene 1994; 9: 2745–50.
itulate normal trophoblast differentiation seen in early 14 Wang H, Zeng X, Oliver P, et al. MDM2 oncogene as a target for
developing placentas. Relating gestational trophoblastic cancer therapy: an antisense approach. Int J Oncol 1999;
15: 653–60.
neoplasia with normal trophoblastic differentiation
15 Asanoma K, Kato H, Inoue T, Matsuda T, Wake N. Analysis of a
programmes highlights the fundamental nature of this candidate gene associated with growth suppression of
group of diseases and provides the framework for future choriocarcinoma and differentiation of trophoblasts. J Reprod Med
molecular studies. These new findings not only shed 2004; 49: 617–26.

648 http://oncology.thelancet.com Vol 8 July 2007


Review

16 Asanoma K, Matsuda T, Kondo H, et al. NECC1, a candidate 38 Shih IM, Kurman RJ. p63 expression is useful in the distinction of
choriocarcinoma suppressor gene that encodes a homeodomain epithelioid trophoblastic and placental site trophoblastic tumors by
consensus motif. Genomics 2003; 81: 15–25. profiling trophoblastic subpopulations. Am J Surg Pathol 2004;
17 Tuncer ZS, Vegh GL, Fulop V, Genest DR, Mok SC, Berkowitz RS. 28: 1177–83.
Expression of epidermal growth factor receptor-related family 39 Shih IeM, Kurman RJ. Molecular basis of gestational trophoblastic
products in gestational trophoblastic diseases and normal placenta diseases. Curr Mol Med 2002; 2: 1–12.
and its relationship with development of postmolar tumor. 40 Shih I-M, Mazur MT, Kurman RJ. Gestational trophoblastic disease.
Gynecol Oncol 2000; 77: 389–93. In: Kurman RJ, ed. Blaustein’s pathology of the female genital tract,
18 Fulop V, Colitti CV, Genest D, et al. DOC-2/hDab2, a candidate 5th edn. New York: Springer-Verlag, 2002: 1193–247.
tumor suppressor gene involved in the development of gestational 41 Shih IM, Kurman RJ. Placental site trophoblastic tumor--past as
trophoblastic diseases. Oncogene 1998; 17: 419–24. prologue. Gynecol Oncol 2001; 82: 413–14.
19 Fulop V, Mok SC, Berkowitz RS. Molecular biology of gestational 42 Ichikawa N, Zhai YL, Shiozawa T, et al. Immunohistochemical
trophoblastic neoplasia: a review. J Reprod Med 2004; 49: 415–22. analysis of cell cycle regulatory gene products in normal trophoblast
20 Matsuda T, Sasaki M, Kato H, et al. Human chromosome 7 carries and placental site trophoblastic tumor. Int J Gynecol Pathol 1998;
a putative tumor suppressor gene(s) involved in choriocarcinoma. 17: 235–40.
Oncogene 1997; 15: 2773–81. 43 Mao TL, Seidman JD, Kurman RJ, Shih IeM. Cyclin E and p16
21 Burke B, Sebire NJ, Moss J, et al. Evaluation of deletions in 7q11.2 immunoreactivity in epithelioid trophoblastic tumor–an aid in
and 8p12-p21 as prognostic indicators of tumour development differential diagnosis. Am J Surg Pathol 2006; 30: 1105–10.
following molar pregnancy. Gynecol Oncol 2006; 103: 642–48. 44 Murdoch S, Djuric U, Mazhar B, et al. Mutations in NALP7 cause
22 Stahle-Backdhal M, Inoue M, Zedenius J, et al. Decreased recurrent hydatidiform moles and reproductive wastage in humans.
expression of Ras GTPase activating protein in human trophoblastic Nat Genet 2006; 38: 300–02.
tumors. Am J Pathol 1995; 146: 1073–78. 45 Chen H, Ye D, Xie X, Lu W, Zhu C, Chen X. PTEN promoter
23 Xue WC, Chan KY, Feng HC, et al. Promoter hypermethylation of methylation and protein expression in normal early placentas and
multiple genes in hydatidiform mole and choriocarcinoma. hydatidiform moles. J Soc Gynecol Investig 2005; 12: 214–17.
J Mol Diagn 2004; 6: 326–34. 46 Cagayan MS, Lu-Lasala LR. Management of gestational
24 Vegh GL, Fulop V, Liu Y, et al. Differential gene expression pattern trophoblastic neoplasia with metastasis to the central nervous
between normal human trophoblast and choriocarcinoma cell lines: system: a 12-year review at the Philippine General Hospital.
downregulation of heat shock protein-27 in choriocarcinoma in J Reprod Med 2006; 51: 785–92.
vitro and in vivo. Gynecol Oncol 1999; 75: 391–96. 47 Lurain JR, Singh DK, Schink JC. Primary treatment of metastatic
25 Ciocca DR, Oesterreich S, Chamness GC, McGuire WL, high-risk gestational trophoblastic neoplasia with EMA-CO
Fuqua SA. Biological and clinical implications of heat shock chemotherapy. J Reprod Med 2006; 51: 767–72.
protein 27,000 (Hsp27): a review. J Natl Cancer Inst 1993; 48 Ngan HY, Tam KF, Lam KW, Chan KK. Relapsed gestational
85: 1558–70. trophoblastic neoplasia: a 20-year experience. J Reprod Med 2006;
26 Richards EH, Hickey E, Weber L, Master JR. Effect of 51: 829–34.
overexpression of the small heat shock protein HSP27 on the heat 49 Newlands ES. The management of recurrent and drug-resistant
and drug sensitivities of human testis tumor cells. Cancer Res 1996; gestational trophoblastic neoplasia (GTN). Best Pract Res Clin Obstet
56: 2446–51. Gynaecol 2003; 17: 905–23.
27 Garrido C, Ottavi P, Fromentin A, et al. HSP27 as a mediator of 50 Dang CV, O’Donnell KA, Zeller KI, Nguyen T, Osthus RC, Li F.
confluence-dependent resistance to cell death induced by anticancer The c-Myc target gene network. Semin Cancer Biol 2006;
drugs. Cancer Res 1997; 57: 2661–67. 16: 253–64.
28 Shih IM. Application of human leukocyte antigen-G expression in 51 Jonkers J, Berns A. Oncogene addiction: sometimes a temporary
the diagnosis of human cancer. Hum Immunol 2007; 68: 272–76. slavery. Cancer Cell 2004; 6: 535–38.
29 Singer G, Kurman RJ, McMaster M, Shih IeM. HLA-G 52 Yu D, Dews M, Park A, Tobias JW, Thomas-Tikhonenko A.
immunoreactivity is specific for intermediate trophoblast in Inactivation of Myc in murine two-hit B lymphomas causes
gestational trophoblastic disease and can serve as a useful marker dormancy with elevated levels of interleukin 10 receptor and
in differential diagnosis. Am J Surg Pathol 2002; 26: 914–20. CD20: implications for adjuvant therapies. Cancer Res 2005;
30 Vegh GL, Selcuk Tuncer Z, Fulop V, Genest DR, Mok SC, 65: 5454–61.
Berkowitz RS. Matrix metalloproteinases and their inhibitors in 53 Vita M, Henriksson M. The Myc oncoprotein as a therapeutic target
gestational trophoblastic diseases and normal placenta. for human cancer. Semin Cancer Biol 2006; 16: 318–30.
Gynecol Oncol 1999; 75: 248–53. 54 Ponzielli R, Katz S, Barsyte-Lovejoy D, Penn LZ. Cancer
31 Palmieri C, Fisher RA, Sebire NJ, et al. Placental site trophoblastic therapeutics: targeting the dark side of Myc. Eur J Cancer 2005;
tumour arising from a partial hydatidiform mole. Lancet 2005; 41: 2485–501.
366: 688. 55 Nilsson JA, Keller UB, Baudino TA, et al. Targeting ornithine
32 Felemban AA, Bakri YN, Alkharif HA, Altuwaijri SM, Shalhoub J, decarboxylase in Myc-induced lymphomagenesis prevents tumor
Berkowitz RS. Complete molar pregnancy. Clinical trends at King formation. Cancer Cell 2005; 7: 433–44.
Fahad Hospital, Riyadh, Kingdom of Saudi Arabia. J Reprod Med 56 Mendelsohn J, Baselga J. Epidermal growth factor receptor
1998; 43: 11–13. targeting in cancer. Semin Oncol 2006; 33: 369–85.
33 Moore-Maxwell CA, Robboy SJ. Placental site trophoblastic tumor 57 Muller-Hocker J, Obernitz N, Johannes A, Lohrs U. P53 gene
arising from antecedent molar pregnancy. Gynecol Oncol 2004; product and EGF-receptor are highly expressed in placental site
92: 708–12. trophoblastic tumor. Hum Pathol 1997; 28: 1302–06.
34 Oldt RJ 3rd, Kurman RJ, Shih IeM. Molecular genetic analysis of 58 Singer G, Oldt R 3rd, Cohen Y, et al. Mutations in BRAF and KRAS
placental site trophoblastic tumors and epithelioid trophoblastic characterize the development of low-grade ovarian serous
tumors confirms their trophoblastic origin. Am J Pathol 2002; carcinoma. J Natl Cancer Inst 2003; 95: 484–86.
161: 1033–37.
59 Cohen Y, Xing M, Mambo E, et al. BRAF mutation in papillary
35 Fisher RA, Paradinas FJ, Newlands ES, Boxer GM. Genetic evidence thyroid carcinoma. J Natl Cancer Inst 2003; 95: 625–27.
that placental site trophoblastic tumours can originate from a
60 Satyamoorthy K, Li G, Gerrero MR, et al. Constitutive mitogen-
hydatidiform mole or a normal conceptus. Br J Cancer 1992;
activated protein kinase activation in melanoma is mediated by both
65: 355–58.
BRAF mutations and autocrine growth factor stimulation.
36 Arima T, Imamura T, Sakuragi N, et al. Malignant trophoblastic Cancer Res 2003; 63: 756–59.
neoplasms with different modes of origin. Cancer Genet Cytogenet
61 Kobel M, Pohl G, Schmitt WD, Hauptmann S, Wang TL, Shih IeM.
1995; 85: 5–15.
Activation of mitogen-activated protein kinase is required for
37 Chang YL, Chang TC, Hsueh S, et al. Prognostic factors and migration and invasion of placental site trophoblastic tumor.
treatment for placental site trophoblastic tumor- report of 3 cases Am J Pathol 2005; 167: 879–85.
and analysis of 88 cases. Gynecol Oncol 1999; 73: 216–22.

http://oncology.thelancet.com Vol 8 July 2007 649


Review

62 Rinehart J, Adjei AA, Lorusso PM, et al. Multicenter phase II study 69 Lane HA, Lebwohl D. Future directions in the treatment of
of the oral MEK inhibitor, CI-1040, in patients with advanced non- hormone-sensitive advanced breast cancer: the RAD001
small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol (Everolimus)-letrozole clinical program. Semin Oncol 2006;
2004; 22: 4456–62. 33 (2 suppl 7): S18–25.
63 Lorusso PM, Adjei AA, Varterasian M, et al. Phase I and 70 Nathan CA, Amirghahari N, Rong X, et al. Mammalian target of
pharmacodynamic study of the oral MEK inhibitor CI-1040 in rapamycin inhibitors as possible adjuvant therapy for microscopic
patients with advanced malignancies. J Clin Oncol 2005; residual disease in head and neck squamous cell cancer. Cancer Res
23: 5281–93. 2007; 67: 2160–68.
64 Thompson N, Lyons J. Recent progress in targeting the Raf/MEK/ 71 Smolewski P. Recent developments in targeting the mammalian
ERK pathway with inhibitors in cancer drug discovery. target of rapamycin (mTOR) kinase pathway. Anticancer Drugs 2006;
Curr Opin Pharmacol 2005; 5: 350–56. 17: 487–94.
65 Martin PM, Sutherland AE. Exogenous amino acids regulate 72 Okuno S. Mammalian target of rapamycin inhibitors in sarcomas.
trophectoderm differentiation in the mouse blastocyst through an Curr Opin Oncol 2006; 18: 360–62.
mTOR-dependent pathway. Dev Biol 2001; 240: 182–93. 73 Goffin JR, Anderson IC, Supko JG, et al. Phase I trial of the matrix
66 Al-Shami R, Sorensen ES, Ek-Rylander B, Andersson G, Carson metalloproteinase inhibitor marimastat combined with carboplatin
DD, Farach-Carson MC. Phosphorylated osteopontin promotes and paclitaxel in patients with advanced non-small cell lung cancer.
migration of human choriocarcinoma cells via a p70 S6 kinase- Clin Cancer Res 2005; 11: 3417–24.
dependent pathway. J Cell Biochem 2005; 94: 1218–33. 74 Rosenbaum E, Zahurak M, Sinibaldi V, et al. Marimastat in the
67 Briese J, Oberndorfer M, Schulte HM, Loning T, Bamberger AM. treatment of patients with biochemically relapsed prostate cancer:
Osteopontin expression in gestational trophoblastic diseases: a prospective randomized, double-blind, phase I/II trial. Clin Cancer
correlation with expression of the adhesion molecule, CEACAM1. Res 2005; 11: 4437–43.
Int J Gynecol Pathol 2005; 24: 271–76.
68 Briese J, Oberndorfer M, Patschenik C, et al. Osteopontin is
colocalized with the adhesion molecule CEACAM1 in the
extravillous trophoblast of the human placenta and enhances
invasion of CEACAM1-expressing placental cells. J Clin Endocrinol
Metab 2005; 90: 5407–13.

650 http://oncology.thelancet.com Vol 8 July 2007

You might also like