You are on page 1of 14

865

Review
ISSN 1462-2416 Pharmacogenomics (2010) 11(6), 865878 10.2217/PGS.10.69 2010 Future Medicine Ltd
Wide-spectrum characterization of trabectedin:
biology, clinical activity and future perspectives
After World War II, the US National Cancer
Institute (MD, USA) carried out a wide-ranging
program of screening marine plant and animals
in order to nd new and promising antineoplas-
tic agents. Among several agents tested in clinical
trials (TABLE 1), extracts from the Caribbean tuni-
cate Ecteinascidia turbinata, later named ectein-
ascidins (ETs), were shown to exert antitumor
activity [1,2]. The small amount of ETs in E. tur-
binata prevented their isolation for over a decade.
The structure of one of these, ET-743, was
nally determined in 1984 and chemically syn-
thesized in 1996 [3,4]. Later, ET-743, or trabect-
edin (Yondelis

, PharmaMar, Madrid, Spain),


was the rst representative of a marine natural
product to receive marketing authorization [5].
Trabectedin has raised a growing interest
in the last few years because of its proven ef-
cacy in the treatment of advanced soft tissue
sarcoma (STS). Given the recent introduction
of this new compound into clinical use, a com-
plete revision of its mechanisms of action (still
not completely understood), the preclinical and
clinical evidences that brought it to its approval
in Europe, and its main side effects and their best
management is needed.
Drug purication, biosynthesis
& structure
As extensively reviewed by Cuevas and
Francesch [5], the diff iculties faced when
develop ing sufcient supplies of trabectedin
have represented a severe limitation on its use
for a long time.
The tunicate normally lives in coastal shal-
low waters (015 m depth) and in lagoons. It
is distributed throughout the Caribbean and
in the temperate regions of the Atlantic and
the Mediterranean.
Before 1997, PharmaMar started both a
wide aquaculture program and a chemical syn-
thesis program, in order to produce enough
ET-743 for preclinical and clinical use.
For the aquaculture program, two main
methodological approaches were developed: sea
farms and inland facilities. In 2004, all of the
Mediterranean aqua farms collectively produced
approximately 100 metric tons of tunicate bio-
mass. However, as a consequence of the isolation
and purication procedures, the nal yields of
ET-743 were of very low concentrations. Thus,
in order to achieve the future commercialization
of the drug, it was necessary to develop a syn-
thetic process to produce ET-743, so as to avoid
the dependence on the natural source.
Since 1996, when the Corey group published
the complete synthesis of ET-743 [4], several
groups have improved the synthesis process,
but the manufacture of ET-743 on an indus-
trial scale was still not possible. Nevertheless,
in 2003, PharmaMar successfully developed a
short and straightforward semisynthetic process
starting from cyanosafracin B, an anti biotic
obtained by fermentation from the bacteria
Pseudomonas uorescens [6,7]. In addition, other
synthetic or semisynthetic approaches have
been reported from a large number of research
groups [816].
Ecteinascidin-743 (trabectedin, Yondelis

; PharmaMar, Madrid, Spain), a 25-year-old antineoplastic


alkylating agent, has recently shown unexpected and interesting mechanisms of action. Trabectedin causes
perturbation in the transcription of inducible genes (e.g., the multidrug resistance gene MDR1) and
interaction with DNA repair mechanisms (e.g., the nucleotide excision repair pathway) owing to drug-
related DNA double strand breaks and adduct formation. Trabectedin was the rst antineoplastic agent
from a marine source (namely, the Caribbean tunicate Ecteinascidia turbinata) to receive marketing
authorization. This article summarizes the mechanisms of action, the complex metabolism, the main
toxicities, the preclinical and clinical evidences of its antineoplastic effects in different types of cancer
and, nally, the future perspectives of this promising drug.
KEYWORDS: adducts n cancer n double strand breaks n ET-743 n MDR1 n NER
n ovarian n sarcomas n therapy n trabectedin
Bruno Vincenzi
1
,
Andrea Napolitano
1
,
Anna Maria Frezza
1
,
Gaia Schiavon
1
,
Daniele Santni
1
&
Giuseppe Tonini
1
1
University Campus Bio-Medico,
Medical Oncology, Via Alvaro del
Portllo 200, 00128 Rome, Italy

Author for correspondence:


Tel.: +39 339 319 9912
b.vincenzi@unicampus.it
For reprint orders, please contact: reprints@futuremedicine.com
Pharmacogenomics (2010) 11(6)
866 future science group
Review Vincenzi, Napolitano, Frezza, Schiavon, Santini & Tonini
Ecteinascidin-743 is composed of three
fused tetrahydroisoquinoline rings (FIGURE 1) [17]:
two of the rings (units A and B) recognize
and bind covalently to the minor groove of
the DNA double helix; the third ring (unit C)
protrudes out of the minor groove and inter-
acts directly with transcription factors, nota-
bly the nucleotide excision repair (NER)
endo nuclease, XPG [18]. Contrasting evidence
currently exists regarding the necessity of this
subunit for the specic biological responses
induced by ET-743 [19,20].
Trabectedin mechanisms of action
n Trabectedin alkylating mechanisms
The antitumor mechanisms of action of ET-743
are various and not entirely understood. One
of ET-743s most studied properties is its abil-
ity to interact with DNA. This property has
been proposed on the basis of biochemistry,
NMR spectro scopy, x-ray crystallography and
molecular modeling data indicating structural
similarities with other antibiotics containing
tetrahydroisoquinoline [2124].
Ecteinascidin-743 contains a carbinolamine
center at the N2 position. The elimination of
the adjacent 21-OH group results in a Schiff
base that is vulnerable to nucleophilic attack.
Therefore, the binding of ET-743 to the exo-
cyclic 2-amino group of guanine in the DNA
minor groove leads to DNA alkylation.
Some important characteristics differenti-
ate ET-743 from other minor groove mono-
alkylating agents: a unique ability to bend
DNA towards the major groove by alkylating
the minor groove, duplex stabilization and
extrahelical protrusion of the C-subunit [25].
Interestingly, the alkylation reaction is not
random but DNA sequence specic (5-PuGC,
5-PyGG) [26]. This specicity seems to be
mainly related to slower rates of the reverse
dealkylation reaction in favored bonding
sequences rather than in nonfavored ones
(5-AGT), probably owing to more stable
conformational changes [27].
A very important point is the specicity
of cellular proteins able to bind to ET-743-
alkylated DNA sequences. Remarkably, the
minor groove alkylation causes an interference
in the DNA-topoisomerase I cleavage sites. At
high concentrations (i.e., micromolar levels),
ET-743 causes a topoisomerase I-induced for-
mation of cleavage complexes enzyme-linked
and -mediated DNA strand breaks which
are the catalytic intermediates of topoisomer-
ization reactions. On the other hand, it traps
and stabilizes these complexes [28], functionally
resembling topo isomerase poisons [29]. Using
pharmacologically reasonable concentrations
of ET-743 (i.e., nanomolar levels) in sensitive
cells, no DNA breakage or DNA/protein cross-
links are seen [30], suggesting only an ancillary
role of topoisomerase I poisoning in ET-743
cytotoxicity [31].
In a similar manner, the DNA binding of
several transcription factors, notably SRF/TCF
and NF-Y, is impaired at concentrations in the
50-M range, which is still much higher than
the concentrations that cause cellular cyto-
toxicity. Nevertheless, the specicity of alkyl-
ated DNAprotein interactions is conrmed by
the evidence that other transcriptional factors
(e.g., MYB and MYC) can bind to the DNA
even at very high concentrations of ET-743
(300 M), as alkylation does not involve the
binding sequences of those specic transcription
factors [32].
The main pharmacological activity of ET-743
is thus mostly independent of an alkylation-
dependent inhibition of the DNA binding of
proteins or transcription factors. In fact, on the
basis of DNAprotein molecular modeling, it
has been shown that ET-743 can target DNA
with two or more molecules simultaneously and
that the DNAET-743 covalent complex are
virtually superimposable to the minor grooves
of DNA when bound to the zinc ngers of
transcription factors [33]. This structure is also
strongly reminiscent of an RNADNA hybrid,
and these striking similarities raise the inter-
esting hypothesis that ET-743 preferentially
targets the minor groove of DNA when bound
to a zinc-nger-containing transcription factor
by the exposition of a more readily accessible
2-amino group of guanine [34].
n Trabectedin & perturbation of
transcription & cell activities
Preincubation of ET-743 with the transcription
factor NF-Y causes inhibition of DNA bind-
ing at lower concentrations compared with the
Table 1. Some Phase II marine natural products.
Compound Source organism Chemical class
Ecteinascidin-743 Ecteinascidia turbinata Tetrahydroisoquinolone alkaloid
Dolastatin 10 Dolabella auricularia
Symploca sp.
Linear peptide
Bryostatin 1 Bugula neritina Macrocyclic lactone
Kahalalide F Elysia rufescens/Bryopsis sp. Cyclic depsipeptide
Squalamine Squalus acanthias Aminosteroid
Dehydrodidemnin B Trididemnum solidum Cyclic depsipeptide
www.futuremedicine.com 867 future science group
Wide-spectrum characterization of trabectedin Review
pre incubation with DNA, suggesting that the
protein rather than the minor groove is the
target [31].
Furthermore, since the inhibition is extremely
rapid (1 min) and the effect is observed at
pharmaco logical concentrations, it is probably
due to the C-subunit or the DNA bending to
the major groove. Transcriptional activation of
NF-Y-dependent genes is likely to be one of the
main cytotoxic mechanisms of ET-743 action [35].
Among the inhibited genes, the most prom-
ising targets code for MDR1 (P-glycoprotein
[P-gp]), HSP79 and collagen I. ET-743 is the
rst pharmacologically relevant agent to prevent
the activation of both MDR1 [36] and HSP79 [34]
transcription by multiple stress inducers (i.e., his-
tone deacetylase inhibitors and UV irradiation),
without affecting their constitutive expression.
ET-743 may thus selectively inhibit activation of
MDR1 expression in tumor cells without affect-
ing constitutive expression in normal cells, but
acting as a specic antitumoral transcriptional
regulator. Preclinical studies have also conrmed
that ET-743 is an effective agent in MDR1-
overexpressing cells [37], despite the hypothesis
that P-gp overexpression contributes to ET-743
resistance in an ovarian cell line [38].
In a similar manner, ET-743 is responsible for
a dose-dependent downregulation of COL1A1
(the gene coding for pro-a1 chain of collagen I)
expression [39]. These data were conrmed by the
particular ET-743 cytotoxicity in mesenchimal
tumors (i.e., sarcomas) and by the ana lysis of a
ET-743-resistant sarcoma cell line, in which pro-
longed exposure to ET-743 caused changes in cell
function through cytoskeleton rearrangement
and/or modulation of collagen levels [40].
Moreover, it has been reported that ET-743
is responsible for a unique microtubule
disorganization in vitro [41].
Further links between transcriptional
inhibition and ET-743 action have not been
investigated. ET-743 has been proved to also
inhibit NF-Y-independent transcription (Sp1-
mediated [42] or SXR-mediated transcription).
SXR inhibition is particularly interesting because
SXR regulates both drug efux (by activating the
expression of the MDR1 gene [43]) and metabo-
lism (by activating the cytochrome P450 (CYP)
CYP3A4 gene [44]). Moreover, gene-expression
ana lysis through microarray technology has
identied selected groups of genes inhibited by
low doses of the drug in different cell lines [45,46].
Remarkably, another important indication of
the unusual antiproliferative effect of ET-743 is
its predominant activity in the G1 phase of the
cell cycle, eventually resulting in a G2/M block,
a different mode of action to other alkylating
agents, [24]. This effect has been conrmed by the
ana lysis of differential ET-743 activity on G1/S
promoters and nucleosomes [47].
The complex antiproliferative activity of
ET-743 involves other mechanisms: an high-
dose transcription-independent effect leading
to rapid apoptosis involving mitochondria, JNK
and caspase-3 [48], and a low-dose transcription-
coupled block of the cell cycle involving the NER
pathway [49].
n Trabectedin & DNA
repair mechanisms
Since mammalian cell lines lacking NER gene
products are resistant to ET-743, a novel mech-
anism of cell-cycle block based on the NER
pathway was recently hypothesized [48,50].
The mechanism of action is probably very
similar to that of the topoisomerase I poison-
ing: ET-743 traps an intermediate in the NER
processing of DNA adducts. This trapped inter-
mediate proteinET-743DNA adduct complex
could be responsible for ET-743 cell toxicity [51].
In particular, the NER protein Rad13 seems to
be crucial for ET-743 cytotoxic effect by the for-
mation of a Rad13DNAET-743 ternary com-
plex, resulting in cell death [22]. The crucial role
of this pathway is demonstrated by the evidence
that restoration of NER function sensitizes cells
to ET-743 treatment [52].
Investigating cell-cycle phase perturbation, the
cells with functional NER appear to overcome
the G1 block more easily, but are then blocked
in the G2/M phase, suggesting that the NER
mechanism itself leads to some sort of persistent
DNA damage [53]. Unrepaired adducts probably
mediate the damage by arresting and degrading
O
O
N
N
O
NH
O H
O
O
O
S
OH
O H
O
Subunit B
Subunit C
Subunit A
Figure 1. Ecteinascidin-743 and its three fused tetrahydroisoquinoline rings.
Pharmacogenomics (2010) 11(6)
868 future science group
Review Vincenzi, Napolitano, Frezza, Schiavon, Santini & Tonini
DNA polymerase II [54] and by forming DNA
double strand breaks (DSBs), monitored by the
activation of g-H2AX and Rad51 foci [55], even
though several other studies showed no DNA
breaks unless at micromolar concentrations of
ET-743 [23,24].
In 2008, a owchart of molecular pathways
involved in ER-743 cytotoxicity was proposed.
According to the authors, there are two types
of DSBs produced by ET-743: trascription-
coupled DSB involving the NER pathways and
replication-coupled DSB involving the ATM
pathway independently from trascription-cou-
pled DSB [56]. Moreover, it has been recently
shown that Von HippelLindau tumor suppres-
sor protein deciency made the cells resistant to
ET-743-induced cell death with a similar effect to
NER deciency. The Von HippelLindau com-
plex could act in promoting DNA polymerase II
degradation in DNA damaged cells [53].
Finally, recent evidence suggests that the
homologous recombination pathway respon-
sible for DNA break repair is essential for
ET-743 cytotoxicity, at least as much as the other
previously mentioned pathways. In fact, cells
lacking the homologous recombination path-
way were extremely sensitive to the drug with
an approximately 100-fold decrease in IC50 [57].
In a similar way, the Fanconi anemia pathway
seems to be very important in the DNA repair
of ET-743 DNA adducts that might functionally
mimic an interstrand DNA crosslink (ICL) [33,58].
Indeed, cells with mutations in any of the tested
Fanconi anemia genes are extremely sensitive to
ET-743 [59], whose behavior mimics that of the
ICL-forming drugs (e.g., mitomycin C) [60].
As previously described, ET-743 has a very
complex mechanism of action via DNA alkyla-
tion and protein interactions. Nevertheless,
ET-743 is an alkylating agent, and its main
pharmacological action seems to be due to both
transcriptional regulation of inducible genes and
to DNA DSB and ICL-like adducts (FIGURE 2).
Myxoid liposarcoma and could lead to the
association of ET743 with compounds acting
on lipogenic pathways.
n Trabectedin & differentiation in
myxoid liposarcoma tumors
Among the different mechanisms of action
ascribed to ET-743, one of the most fascinating
is its potential ability to induce differentiation
in specic histotypes of STS, such as myxoid
liposarcoma. Myxoid liposarcoma is character-
ized by a fusion between the cyclophosphamide,
hydroxydaunorubicin (doxorubicin), Oncovin


(vincristine; Genus Pharmaceuticals, Newbury,
UK) and prednisone/prednisolone (CHOP)
transcription factors and the FUS or EWS genes,
which is probably one of the key points to its
pathogenesis; ET743 has been proven to be par-
ticularly effective in this subtype of sarcoma.
One hypothesis is that the activity of trabectedin
is related to the inactivation of the FUSCHOP
oncogene: Forni et al. reported that trabectedin
causes detachment of the FUSCHOP chimera
from targeted promoters. Reverse transcription
PCR and chromatin immunoprecipitation ana-
lysis in a myxoid liposarcoma line and surgi-
cal specimens of myxoid liposarcoma patients
in vivo show activation of the CAAT/enhancer
binding protein-mediated transcriptional pro-
gram that leads to morphologic changes of
terminal adipogenesis.
Trabectedin metabolism & toxicities
n Trabectedin metabolism
Data regarding ET-743 metabolism are still
limited. In vitro experiments consisting of drug
incubation with a human lymphoblast-expressed
CYP3A4 isoform suggested the presence of at
least three metabolites [301].
With an initial in vivo evaluation, no metab-
olites were observed in urine, serum or bile
samples from patients treated with intravenous
ET-743, whereas deacetylated ET-743 was found
after plasma incubation [61]. Recent evidence
obtained by using [
14
C] ET-743 proved that the
drug is quickly metabolically converted by the
liver to a large number of compounds excreted
in both urine (510% of total) and feces (major
excretory pathway) [62,63].
The presence of O-glucuronidated and
glutat hione-conjugated metabol ites i s
likely [64], although the inhibition of phase II
enzy mes, such as UDP-glucuronosyltransferase,
N-acetyltransferase, sulfotransferase and gluta-
thione S-transferase, did not show any signicant
inuence on the cytotoxicity of ET-743 [65].
The most important ET-743 biotransforma-
tion pathway is the enzymatic conversion by liver
microsomal proteins (~50% of the total conver-
sion). Two main chemical products are formed
during the microsomal breakdown: ET-743S1
and ET-743S2. Two other molecules, ET-743M5
and ET-743M6, are undoubtedly metabolites
of ET-743S2 [301]. ET-743 biotransformation at
the microsomal level is mainly due to CYP3A4,
2C9, 2C19, 2D6 and, to a minor extent, 2E1 [66].
These data have been conrmed in vitro by the
increased cytotoxicity of ET-743 (measured as
IC50 decrease) observed in a HepG2 cell line after
www.futuremedicine.com 869 future science group
Wide-spectrum characterization of trabectedin Review
incubation with CYP inhibitors [63]. Interestingly,
male microsomes showed a signicantly lower
ET-743 biotransformation rate, probably owing
to the different amounts of CYP3A4 and other
CYPs in the microsomal preparations. Typically,
higher quantities of CYP3A4 and faster conver-
sion of ET-743 were observed for female micro-
somes compared with male microsomes, with
CYP3A4 as the major isozyme involved in ET-743
biotransformation (~95%) [64].
Therefore, since CYP3A4 has an important
role in ET-743 metabolism, the risk of in vivo
drugdrug interactions must be considered
when the agent is combined with other CYP3A4
substrates [67].
Finally, given the complexities of ET-743
metabolism, it is unlikely that a single poly-
morphism or the inhibition of a single metabolic
pathway can change its metabolic prole.
n Trabectedin toxicities
The liver is a key organ not only for ET-743
metabolism but also for its collateral effects.
Preclinical studies in animals demonstrated liver
toxicity as an important side effect of ET-743. In
these studies, the female rat was identied as the
species with the highest risk of hepatotoxicity
from ET-743 [68]. Repeated treatment cycles were
able to increase the extent of clinical chemistry
changes, suggesting cumulative toxicity [69].
The unusual form of ET-743 hepatotoxicity in
female rats consists of damaged biliary duct epi-
thelia accompanied by inammation followed by
peribiliary brosis. The pathological alterations
were accompanied by defects of liver function,
such as dramatic elevation of plasma bilirubin lev-
els, moderate increases in plasma levels of alkaline
phosphatase (ALP) and aspartate transaminase
(AST) and a decrease in hepatic CYP activity [70].
In fact, together with dose-limiting myelo-
suppression (i.e., neutropenia and thrombo-
cytopenia), the main ET-743 toxicities registered
during clinical trials were acute (but revers-
ible) transaminitis and subclinical cholangitis,
characterized by increases in ALP and/or
bilirubin [71].
Interestingly, it has been shown that high-
dose dexamethasone administered 24 h before
ET-743 protects rats from hepatotoxicity with-
out compromising antitumor activity. This effect
has been rstly assigned to the dramatic dexa-
methasone-induced reduction of ET-743 hepatic
levels via the induction of the CYP enzyme
CYP3A [72]. Considering the side effects of high-
dose dexamethasone [73,74], the hepatoprotective
effect of other CYP3A inducers have been tested
for a potential use in humans. Indole-3-carbinole
(I3C), a potent inducer of CYP enzymes [75], is
the hydrolysis product of glucosinolates present
in cruciferous vegetables, such as broccoli and
Brussels sprouts. As expected, the food intake of
I3C protected against the detrimental hepatic
effects of ET-743 without interfering with its
antitumor activity in a model of mammary
carci noma. Surprisingly, ingestion of I3C did not
decrease hepatic levels of ET-743 in comparison
with animals who received ET-743 alone, sug-
gesting that CYP induction is not the main hepa-
toprotective event. Thus, the interference with
NF-kB transcriptional activity is the most attrac-
tive candidate mechanism to explain hepatopro-
tection, as it is common to ET-743 antidotes, I3C
and dexamethasone [76]. These data were con-
rmed by the evidence that the protection pro-
vided by a pretreatment with b-naphthoavone
and phenobarbitone, two other CYP inducers,
was not as effective in vivo as dexamethasone.
Pretreatment with N-acetylcysteine, a non-CYP-
inducing hepatoprotective drug, did not protect,
from ET-743-induced liver changes [77]. Recent
studies assessed the capability of sandwich-
cultured primary rat hepatocytes to predict the
hepatoprotective effect of dexamethasone, attrib-
uted, at least in part, to enhanced multidrug
resistance-associated proteins biliary excretion
and increased metabolism by CYP3A1/2 [78].
ET-743 RAD13 NF-Y
DNA breaks
DNA alkylation
and bending
Transcriptional
regulation
Figure 2. Ecteinascidin-743 mechanism of action.
ET-743: Ecteinascidin-743.
Pharmacogenomics (2010) 11(6)
870 future science group
Review Vincenzi, Napolitano, Frezza, Schiavon, Santini & Tonini
There are poor data regarding the mechanisms
of hepatoxicity and hepatoprotection in humans.
A recent meta-ana lysis showed that dexametha-
sone pretreatment increases ET-743 systemic
clearance by 19% by increasing bile ow [79]. A
recent pharmacokinetic/pharmacodynamic ana-
lysis predicted an enhancement of ET-743 safety
and efcacy after administration of dexametha-
sone and a dose-reduction strategy based on the
serum concentration of liver enzymes [80].
As mentioned earlier, the other important and
dose-limiting ET-743 toxicity is represented by
myelosuppression. Reversible dose-dependent
neutropenia was the most frequently reported
toxicity in Phase II studies, with median nadir
values observed at approximately 14 days [81].
ET-743-induced neutropenia is largely dependent
on the intensity of dose administered and inter-
dose intervals, but not the duration of intravenous
infusion [82].
A pharmacokinetic/pharmacodynamic meta-
ana lysis showed that the pharmacological effect
of ET-743 on absolute neutrophil count was
more closely related to drug concentrations in
a hypothetical effect compartment, representa-
tive of bone marrow, rather than from the central
plasma compartment, with ET-743 reducing the
proliferation rate and stimulating the killing rate
of progenitor cells [81]. In mice, ET-743 toxic-
ity was lower in stem cells than in committed
progenitors, suggesting an unlikely long-term
myelosuppression as a consequence of ET-743
treatment [83]. Other in vitro evidence suggests
a direct immuno modulatory effect of ET-743
through inhibited differentiation of monocytes to
macrophages and reduced production of CCL2
and IL-6 [84].
Interestingly, advanced sarcoma patients
receiving dexamethasone before starting ET-743
experienced lower liver and bone marrow toxici-
ties than patients not receiving dexamethasone,
even though any relationship between the two
toxicities remains to be elucidated [85].
Moreover, several case reports described another
rare toxicity of ET-743 treatment: rhabdo myolysis.
In a Phase I trial, one patient experienced grade
4 rhabdomyolysis, renal failure requiring dialy-
sis, grade 4 neutropenia and grade 3 thrombo-
cytopenia. Hepatic toxicity in this patient was not
reported [86]. Two cases of rhabdomyolysis were
reported in a subsequent French Phase II trial [87],
as well as in another European Phase II study [88].
It has been shown that elevated intercycle peaks
of ALP and AST, as well as increased baseline bili-
rubin, are independent predictors of multiorgan
toxicities [89]. For this reason, the differences in
toxicities between European and American trials
may be due to a greater degree of dose adjustment
in the USA based on liver enzymes. However, a
fatal rhabdomyolysis case has been reported by a
US group [90].
Finally, skin and soft tissue damage have been
reported after ET-743 extravasation from a central
venous access device [91].
Preclinical & clinical evidences
The rst preclinical evidence of ET-743 antitu-
mor activity is relatively recent. Approximately
10 years ago, the effect of ET-743 was dem-
onstrated in human tumors explanted from
patients [92], ovarian carcinoma [93,94], melanoma
and non-small-cell lung cancer [93] xenografts. In
human STS cell lines explanted from chemonaive
patients, gene-expression prole ana lysis revealed
the upregulation of 86 genes and the down-
regulation of 244 genes in response to ET-743,
suggesting an important impact in tumor cell
biology [95]. Interestingly, ET-743 promoted dif-
ferentiation in myxoid liposarcoma tumors by
detachment of the pathogenic FUSCHOP
chimera protein from its promoters [96].
Ecteinascidin-743 also showed sequence-
dependent synergistic cytotoxicity with pacli-
taxel (administered before ET-743) in human
breast cancer cell lines in vitro and in vivo [97],
and with paclitaxel (administered before ET-743)
or doxorubicin (administered after ET-743) in
STS cells [98].
An additive or additive-to-synergistic effect
was seen in vitro with ET-743 simultaneously
or after cisplatin [99], irinotecan [100] or doxo-
rubicin [101] in human rabdomyosarcoma or
ovarian cancer cell lines and in a range of
human tumor xenografts, including rhabdo-
myosarcoma, brosarcoma, non-small-cell lung
cancer and ovarian cancer. Relevant effects have
been also seen both in drug-sensitive and drug-
resistant bone tumor cells [102]. Moreover, com-
bination therapy with ET-743 and plasminogen-
related protein B caused increase in human
chondro sarcoma necrosis by antagonizing
tumor-associated microvessel formation [103].
Finally, ET-743 exerts cell line-dependent
radiosensitizing properties. Initial evidence sug-
gested a moderate effect only with cytotoxic con-
centrations of ET-743 [104], but more recent nd-
ings showed a signicant in vitro radiosensitizing
effect and the induction of cell-cycle changes and
apoptosis in several human cancer cell lines in
the presence of pharmacologically appropriate
(within the nanomolar range) concentrations of
the drug [105].
www.futuremedicine.com 871 future science group
Wide-spectrum characterization of trabectedin Review
Several in vitro studies attempted to elucidate
the mechanisms of cellular resistance to ET-743.
MDR1 seems not to be crucial, since ET-743 is
clearly a P-gp substrate, but it makes the cells
resistant to ET-743 only when expressed at very
high levels [106]. Conversely, other pathways
seem to be important, for example IGF-1 recep-
tor for Ewings sarcoma cell line TC-71 [107], and
collagen I for chondrosarcoma cell line CS-1 [39].
A further conrmation of this hypothesis is
that resistance to ET-743 did not correlate with
resistance to doxorubicin [108], indicating that
the two drugs may act through different mecha-
nisms. For example, p53 mutations and deletions
in sarcoma cells correlate with extreme sensitivity
to ET-743 [109], whereas wild-type p53 sensitizes
cells to doxorubicin by downregulating MDR1
expression [110]. Moreover, in vitro ET-743 pre-
vented MDR1 gene expression, showing a syn-
ergistic cytotoxic effect with doxorubicin (a
well-known MDR1 substrate) [98].
Finally, a human chondrosarcoma cell line
resistant to ET-743 exhibited crossresistance to
cisplatin and methotrexate but not to doxoru-
bicin. Zinc nger proteins, especially ZNF93,
are thought to be involved in mechanisms of
resistance, probably because of their role in DNA
damage repair [111].
Following the preclinical studies, a large
number of clinical trials have been carried out
in order to evaluate the in vivo effect of ET-743.
A large number of Phase I trials included patients
with any solid malignancies, without focusing
on specific diseases, who were preclinically
more sensitive to ET-743. TABLE 2 shows maxi-
mum tolerated doses, dose-limiting toxicities
and synergies data.
These results suggested that maximum tol-
erated dose slightly changes with the sched-
ule (11001800 g/m
2
) and the dose-limiting
toxicities are mainly hematological toxicities
(i.e., neutro penia and thrombocytopenia) and
asthenia rather than transient transaminase ele-
vation. It should be highlighted that ET-743 does
not interact with other common chemotherapeu-
tic agents (e.g., doxorubicin, gemcitabine and
cisplatin) from a pharmacodynamic/pharmaco-
kinetic point of view. Therefore, combination
therapies could be suitable and useful in the
management of advanced diseases.
Several Phase II trials have been carried out
and are currently ongoing in patients with cer-
tain tumors, leading to more specic and clini-
cally useful results. ET-743 seems to be a very
promising drug indeed for the treatment of a
wide spectrum of tumors.
Nevertheless, ET-743 failed to show signi-
cant cytotoxicity against gastrointestinal stromal
tumors [112,113] and pretreated advanced colorectal
cancer [114].
However, several studies reported interesting
rates of responses or disease stabilization during
ET-743 therapies with Phase II toxicities over-
lapping Phase I ones, thus conrming the clinical
impact of the drug. The most promising results
have been achieved against STS (especially leio-
myosarcomas and myxoid liposarcoma) and ovar-
ian cancers (TABLE 3). Retrospective studies con-
rmed ET-743 efcacy in advanced pretreated
STSs [115] and advanced pretreated myxoid lipo-
sarcomas [116,117]. A case report showed a durable
objective response lasting at least 8 months in a
case of advanced, recurrent and refractory uter-
ine leiomyosarcoma treated with 1200 g/m
2
intravenous ET-743 over 24 h every 3 weeks [118].
In evaluating clinical trials results, it should
be noted that a recent report suggested the inad-
equacy of size-based response criteria (Response
Evaluation Criteria In Solid Tumors) to assess
the efcacy of ET-743 in metastatic sarcoma
patients [119].
More recent trials (recently completed, active
or recruiting) will give further information on
ET-743 cytotoxicity in several types of cancer
(advanced prostate cancer, STS, ovarian cancer,
breast cancer, mesothelioma, endometrial carci-
noma, primary peritoneal cavity cancer and fal-
lopian tube cancer) and on ET-743 potential heart
toxicity (altered ECG QT intervals)[201].
In Europe, ET-743 in monotherapy
(1.5 mg/m
2
as a 24-h continuous infusion every
3 weeks) is currently the only agent approved
as a second-line treatment in the treatment of
advanced STS after the failure of anthra cycline-
and ifosfamide-based regimens. The drug
received orphan drug status from the European
Commission for the treatment of ovarian can-
cer in October 2003. The US FDA granted
ET-743 orphan drug status for both STS and
ovarian cancer.
As for trabectedin hepatic effects, plasma liver
enzymes level (transaminases, bilirubin, alkalin
phosphatases and 5-nucleotidase) should be
checked before each course, because patients
with any baseline alterations have a signicantly
higher probability of developing severe liver tox-
icity. The risk of developing grade 34 toxicities
seems to be strongly reduced through premedi-
cation with dexamethasone (4 mg orally twice
daily 24 h before therapy). A study by Grosso
et al. showed how elevation of transaminases,
neutropenia and thrombocytopenia incidence
Pharmacogenomics (2010) 11(6)
872 future science group
Review Vincenzi, Napolitano, Frezza, Schiavon, Santini & Tonini
are signicantly lower in patients pretreated
with dexamethasone (2%, 2% and 0) then in
not premedicated patients (34, 24 and 25%,
respectively) [84].
Putative predictive factors of activity
As soon as the rst promising effects of ET-743
appeared, strong efforts were made to identify
putative predictive factors of activity and to reli-
ably select subpopulations of patients who may
benet from ET-743 therapy.
The complex ET-743 metabolism seems to
exclude CYP genes as potential candidates.
NER protein levels rather than mRNA levels
seem to be more promising [142]. Moreover, it
has been recently proposed that screening for
mutations in Fanconi anemia genes may facili-
tate the identication of tumors displaying
enhanced sensitivity to ET-743 [59].
No clear putative predictive factors of
ET-743 activity have been identied so far.
Therefore, greater efforts are needed in studies
investigating such aspects.
Conclusion & future perspective
Ecteinascidin-743 is a novel natural anti-
cancer agent, now synthetically produced,
with unique mechanisms of action involving
DNA alkylation, transcriptional repression and
NER/homologous recombination-dependent
cell-cycle blocking. The metabolism of ET-743
is quite complex amd is mainly achieved
through liver microsomal CYPs.
Preclinical and clinical evidence has dem-
onstrated how trabectedin combines a favor-
able tolerability prole with proven efcacy
in the treatment of advanced STS and ovar-
ian cancer. Moreover, ET-743 does not seem
Table 2. Phase I concluded clinical trials.
Drug(s) Tumors Administration Results Ref.
ET-743 Resistant solid
tumors
24 h intravenous q21 MTD: 1800 g/m
2
1500 g/m
2
dose is clinically feasible
DLTs: severe thrombocytopenia and neutropenia
[121,122]
ET-743 STSs 24 h intravenous q21
1500 g/m
2
Risk of severe toxicity enhanced in patients with
hepatic dysfunction
Dexamethasone co-treatment decreased incidence of
severe toxicity and AUC
[123]
ET-743 Solid tumors 72 h intravenous q21 MTD: 1200 g/m
2
DLTs: neutropenia, thrombocytopenia and fatigue
[86]
ET-743 Solid tumors 1 h intravenous q21 vs 3 h
intravenous q21
1 h MTD: 1100 g/m
2
1 h DLTs: thrombocytopenia and fatigue
3 h MTD: 1800 g/m
2
3 h DLTs: pancytopenia and fatigue
1650 g/m
2
dose is clinically feasible
[124]
ET-743 Refractory solid
tumors in children
3 h intravenous q21 Premedication with dexamethasone followed by
1100 g/m
2
dose
[125]
ET-743 Advanced solid
tumors
1 h intravenous for 3 wks
q28 vs 3 h intravenous for
3 wks q28
1 h MTD: 610 g/m
2

3 h MTD: 580 g/m
2

DLTs: febrile neutropenia and fatigue
[126]
PLD plus ET-743 Advanced solid
tumors
1 h intravenous PLD
(30 mg/m
2
), then 3 h
intravenous ET-743 q21
MTD
ET-743
: 1100 g/m
2
ET-743 plus PLD generally well tolerated
[127]
Gemcitabine plus
ET-743
Advanced solid
tumors
Intravenous on days 18-15
of q28 cycle
Study closed for unacceptable frequency of adjustments
to the weekly dosing schedule for hepatic toxicity
No DLTs observed
[128]
Doxorubicin plus
ET-743
STSs 10/15 intravenous
doxorubicin (60 mg/m
2
)
followed by 3 h intravenous
on day 1 q21
MTD
ET-743
: 1100 g/m
2
DLT: neutropenia
[129]
Cisplatin plus
ET-743
Solid tumors Intravenous day 1 and 8 q21 MTD
ET-743
: 600 g/m
2
MTD
CDDP
: 40 mg/m
2
DLT: febrile neutropenia
[130]
Doxorubicin plus
ET-743
Advanced STS and
breast cancer
Intravenous q21 MTD
ET-743
: 800 g/m
2
MTD
DOXO
: 60 mg/m
2
DLTs: febrile neutropenia and asthenia
[131]
AUC: Area under curve; DLT: Dose-limiting toxicity; MTD: Maximum tolerated dose; PLD: Pegylated liposomal doxorubicin; STS: Soft tissue sarcoma; wk: Week.
www.futuremedicine.com 873 future science group
Wide-spectrum characterization of trabectedin Review
to pharmacologically interact with other anti-
neoplastic drugs, demonstrating additive or
additive-to-synergic effects with common
chemotherapeutic agents. Further studies are
needed to assess its activity in combination with
standard chemotherapeutic agents and with
new biological compounds (i.e., monoclonal
antibodies or small-molecule tyrosine kinase
inhibitors) in STS and also in other human
neoplasia of different histological origins.
Financial & competing interests disclosure
The authors have no relevant af liations or nancial
involvement with any organization or entity with a nan-
cial interest in or nancial conict with the subject matter
or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or
options, expert testimony, grants or patents received or
pending, or royalties.
No writing assistance was utilized in the production of
this manuscript.
Table 3. Conducted Phase II clinical trials.
Tumors Administration Results Toxicities Ref.
STS: 25
OS: 3
Ewings sarcoma: 1 PD
at accrual
24 h intravenous q2128
12001800 g/m
2
STS: 2 PRs
OS: 2 PRs
2 MRs, 10 SDs
Transient transaminitis G3 (24%) and G4
(5%), neutropenia G34 (32%) with
concomitant G34 thrombocytopenia
(5.1%), asthenia G23 (21%)
[132]
Advanced pretreated STS:
54 (41% leiomyosarcoma)
24 h intravenous q21
1500 g/m
2
2 PRs, 4 MRs,
9 SDs, 3 CRs
after surgery
Transient transaminitis G34 (50%),
neutropenia G34 (61%), nausea,
vomiting and asthenia
[88]
Advanced anthracycline-
pretreated STS: 54
24 h intravenous q21
1500 g/m
2
3 PRs, 4 MRs
22 SDs
Transient transaminitis G34 (~60%),
neutropenia G34 (~60%)
[87]
Progressive refractory
SFS: 36
24 h intravenous q21
1500 g/m
2
1 CR, 2 PRs,
2 MRs
Neutropenia G34 (34%),
transient transaminitis G34 (26%)
[133]
Advanced STS: 99 24 h intravenous q21
1500 g/m
2
8 PRs, 45 SDs Neutropenia G34 (52%),
transient transaminitis G34 (40%)
[134]
Unresectable advanced
chemonaive STS
24 h intravenous q21
1500 g/m
2
1 CR, 5 PRs, 1 MR Neutropenia G34 (33%),
transient transaminitis G34 (36%)
[135]
Liposarcoma and
leiomyosarcoma: 270
24 h intravenous q21
1500 g/m
2
vs
3 h intravenous every wk for 3
wks q28 580 g/m
2
Reduction in the relative
risk of progression for
patients treated in
the q21
Neutropenia G34 (21% in 24 h arm
vs 2% in 3 h arm), transient transaminitis
G34 (32% in 24 h arm vs 3% in
3 h arm)
[136]
Pretreated advanced
sarcoma: 21
24 h intravenous q21
9001500 g/m
2
3 PRs, 8 SDs Neutropenia G34 (3), transient
transaminitis G34 (8)
[137]
Ovarian carcinoma:
platinum/taxane resistant
(30) or sensitive (29)
3 h intravenous q21
16501300 g/m
2
Platinum sensitive:
1 CR, 9 PRs, 9 SDs
Platinum resistant:
2 PRs, 8 SDs
Transient transaminitis,
asthenia, neutropenia
[138]
Ovarian carcinoma:
platinum resistant (79) or
sensitive (62)
3 h intravenous q28
580 g/m
2
once weekly for
3 wks
Platinum sensitive:
4 CR, 14 PRs, 22 SDs
Platinum resistant:
5 PRs, 36 SDs
Neutropenia G34 (8%),
transient transaminitis G34 (12%)
[139]
Relapsed, platinum-sensitive,
advanced ovarian cancer:
107
Arm A: 24 h intravenous q21
1500 g/m
2
vs Arm B: 3h
intravenous q21 1300 g/m
2
Arm A: ORR 38.9%
Arm B: ORR 35.8%
Neutropenia 55% arm A and
37% arm B, transient transaminitis 55%
arm A and 59% arm B, nausea/vomiting
and asthenia
[140]
Persistent or recurrent
endometrial carcinoma: 50
3 h intravenous q21
1300 g/m
2
1 CR, 18 SDs Transient transaminitis G34 (40%),
neutropenia G34 (13%),
asthenia G34 (14%)
[141]
Pretreated advanced breast
cancer: 21
24 h intravenous q21
1500 g/m
2
4 PRs, 2 MRs, 6 SDs Transient transaminitis
[142]
GIST: 20 24 h intravenous q21
1500 g/m
2
No ORR, 2 SDs Transient transaminitis G3 (10 patients),
asthenia G3 (1 patient)
[113]
GIST: 27 24 h intravenous q21
1500 g/m
2
No ORR, 9 SDs Transient transaminitis G34 (47%),
neutropenia G34 (48%)
[114]
Pretreated advanced
colorectal cancer: 21
3 h intravenous q21
16501100 g/m
2
No ORR, 4 SDs Transient transaminitis G34 (62%),
neutropenia G34 (42.8%)
[115]
CR: Complete response; GIST: Gastrointestinal stromal tumor; MR: Minimal response; ORR: Overall response rate; OS: Overall survival; PD: Progressive disease;
PR: Partial response; SD: Stable disease; STS: Soft tissue sarcoma; wk: Week.
Pharmacogenomics (2010) 11(6)
874 future science group
Review Vincenzi, Napolitano, Frezza, Schiavon, Santini & Tonini
Bibliography
Papers of special note have been highlighted as:
n
of interest
nn
of considerable interest
1 Simmons TL, Andrianasolo E, McPhail K,
Flatt P, Gerwick WH: Marine natural
products as anticancer drugs. Mol. Cancer
Ther. 4(2), 333342 (2005).
2 Food-Drugs From The Sea (Proceedings of the
Food-Drugs from the Sea Symposium).
Kingston, RI, USA, 2427 August, 1969).
Youngken HW Jr (Ed.). Marine Technology
Society, Washington, DC, USA (1970).
3 Rinehart KL: Antitumor compounds from
tunicates. Med. Res. Rev. 20(1), 127
(2000).
4 Corey EJ, Gin DY, Kania RS:
Enantioselective total synthesis of
ecteinascidin 743. J. Am. Chem. Soc.
118(38), 92029203 (1996).
5 Cuevas C, Frascesch A: Development of
Yondelis

(trabectedin, ET-743).
A semisyntetic process solves the supply
problem. Nat. Prod. Rep. 26, 322337
(2009).
nn
Describes the isolation of ecteinascidin 743
from the natural source and steps leading
to the semisynthetic process of supplying.
6 Ikeda Y, Idemoto H, Hirayama F et al.:
Safracins, new antitumor antibiotics. I.
Producing organism, fermentation and
isolation. J. Antibiot. 36, 12791283 (1983).
7 Menchaca R, Martinez V, Rodriguez A
et al.: Synthesis of natural ecteinascidins
(ET-729, ET-745, ET-759B, ET-736,
ET-637, ET-594) from cyanosafracin B.
J. Org. Chem. 68(23), 88598866 (2003).
8 Chen J, Chen X, Bois-Choussy M, Zhu J:
Total synthesis of ecteinascidin 743. J. Am.
Chem. Soc. 128, 8789 (2006).
9 Saito N, Tachi M, Seki R, Kamayachi H,
Kubo A: A practical synthesis of the ABC
ring model of ecteinascidins. A. Chem.
Pharm. Bull. 48, 15491557 (2000).
10 Zhou B, Guo J, Danishefsky SJ: Studies
directed to the total synthesis of ET 743 and
analogues thereof: an expeditious route to
the ABFGH subunit. Org. Lett. 4, 4346
(2002).
11 Jin W, Metobo S, Williams RM: Synthetic
studies on ecteinascidin-743: constructing a
versatile pentacyclic intermediate for the
synthesis of ecteinascidins and saframycins.
Org. Lett. 5, 20952098 (2003).
12 Magnus P, Matthews KS, Lynch V:
New strategy for the synthesis of
tetrahydroisoquinoline alkaloids. Org. Lett.
5, 21812184 (2003).
13 Chen X, Chen J, De Paolis M, Zhu J:
Synthetic Studies toward ecteinascidin 743.
J. Org. Chem. 70, 43974408 (2005).
14 Zheng S, Chan C, Furuuchi T et al.:
Stereospecic formal total synthesis of
ecteinascidin 743. Angew. Chem. Int. Ed.
Engl. 45, 17541759 (2006).
15 Fishlock D, Williams RM: Synthetic studies
on ET-743. Asymmetric, stereocontrolled
construction of the tetrahydroisoquinoline
core via radical cyclization on a glyoxalimine.
Org. Lett. 8(15), 32993301 (2006).
16 Fishlock D, Williams RM. Synthetic studies
on Et-743. Assembly of the pentacyclic core
and a formal total synthesis. J. Org. Chem.
73(24), 95949600 (2008).
17 Marco E, David-Cordonnier MH, Bailly C,
Cuevas C, Gago F: Further insight into the
DNA recognition mechanism of trabectedin
from the differential afnity of its
demethylated analogue ecteinascidin ET729
for the triplet DNA binding site CGA.
J. Med. Chem. 49(23), 69256929 (2006).
18 Herrero AB, Martin-Castellanos C, Marco E,
Gago F, Moreno S: Cross-talk between
nucleotide excision and homologous
recombination DNA repair pathways in the
mechanism of action of antitumor
trabectedin. Cancer Res. 66, 81558162
(2006).
19 David-Cordonnier MH, Gajate C, Olmea O
et al.: DNA and non-DNA targets in the
mechanism of action of the antitumor drug
trabectedin. Chem. Biol. 12(11), 12011210
(2005).
20 Erba E, Cavallaro E, Damia G et al.:
The unique biological features of the marine
product Yondelis (ET-743, trabectedin) are
shared by its analog ET-637, which lacks the
C ring. Oncol. Res. 14(1112), 579587
(2004).
Executive summary
Trabectedin in the past: from discovery to industrial synthesis
Since the late 1960s, extracts from the Caribbean tunicate Ecteinascidia turbinata have shown promising anticancer activity.
In 1984, the structure of Ecteinascidin (ET)-743 was nally determined.
Since 1996, several chemical processes for synthesis have been achieved.
Pleiotropic & complex mechanisms of action
ET-743 acts as a DNA-alkylating drug, but functionally resembling classical topoisomerase poisons.
ET-743 inhibits the transcription of inducible genes (notably MDR1) via different pathways (e.g., NF-Y mediated, Sp1 mediated and
SXR mediated).
ET-743 interacts with DNA-repairing pathways (notably nucleotide excision repair and homologous recombination) owing to its ability to
form adducts and induce DNA breaks.
Metabolism & toxicities
The liver is the key organ for both metabolism and specic (i.e., nonhematological) toxicities.
In vivo, dexamethasone reduces both liver and hematological toxicities, probably via interference with NF-kB transcriptional activity.
Preclinical/clinical evidences
ET-743 shows pronounced antineoplastic activity in very different cancer line cells, both alone and in combination with common
chemotherapeutic agents.
ET-743 in monotherapy (1.5 mg/m
2
as a 24-h continuous infusion every 3 weeks) is the only agent approved as a second-line therapy in
the treatment of advanced soft tissue sarcoma (Europe).
ET-743 has received the orphan drug status for the treatment of ovarian cancer (Europe and USA) and for soft tissue sarcoma (USA).
The risk of developing grade 34 toxicities seems to be strongly reduced after premedication with dexamethasone.
Trabectedin in the future
Further studies are needed to understand specic predictive factors of response to trabectedin.
Future studies are needed to study trabectedin-based schedules in combination with chemotherapeutic and biological agents
(i.e., monoclonal antibodies and small inhibitors).
www.futuremedicine.com 875 future science group
Wide-spectrum characterization of trabectedin Review
875 www.futuremedicine.com
21 Guan Y, Sakai R, Rinehart KL, Wang AH:
Molecular and crystal structures of
ecteinascidins: potent antitumor compounds
from the Caribbean tunicate Ecteinascidia
turbinata. J. Biomol. Struct. Dyn. 10(5),
793818 (1993).
22 Sakai R, Rinehart KL, Guan Y, Wang AH:
Additional antitumor ecteinascidins from a
Caribbean tunicate: crystal structures and
activities in vivo. Proc. Natl Acad. Sci. USA
89(23), 1145611460 (1992).
23 Pommier Y, Kohlhagen G, Bailly C,
Waring M, Mazumder A, Kohn KW:
DNA sequence- and structure-selective
alkylation of guanine N2 in the DNA minor
groove by ecteinascidin 743, a potent
antitumor compound from the Caribbean
tunicate Ecteinascidia turbinata. Biochemistry
35(41), 1330313309 (1996).
24 Seaman FC, Hurley L: Interstrand
cross-linking by bizelesin produces a
WatsonCrick to Hoogsteen base-pairing
transition region in d(CGTAATTACG)2.
Biochemistry 32(47), 1257712585 (1993).
25 Zewail-Foote M, Hurley LH:
Ecteinascidin-743: a minor groove alkylator
that bends DNA toward the major groove.
J. Med. Chem. 42(14), 24932497 (1999).
26 Pommier Y, Kohlhagen G, Kohn KW,
Leteurtre F, Wani MC, Wall ME: Interaction
of an alkylating camptothecin derivative with
a DNA base at topoisomerase I-DNA cleavage
sites. Proc. Natl Acad. Sci. USA 92(19),
88618865 (1995).
27 Zewail-Foote M, Hurley LH: Differential
rates of reversibility of ecteinascidin 743-DNA
covalent adducts from different sequences lead
to migration to favored bonding sites. J. Am.
Chem. Soc. 123(27), 64856495 (2001).
28 Takebayashi Y, Poourquier P, Yoshida A,
Kohlhagen G, Pommier Y: Poisoning of
human DNA topoisomerase I by ecteinascidin
743, an anticancer drug that selectively
alkylates DNA in the minor groove. Proc. Natl
Acad. Sci. USA 96, 71967201 (1999)
29 Pommier Y, Pourquier P, Fan Y, Strumberg D:
Mechanism of action of eukaryotic DNA
topoisomerase I and drugs targeted to the
enzyme. Biochim. Biophys. Acta 1400(13),
83105 (1998).
30 Takebayashi Y, Goldwasser F, Urasaki Y,
Kohlhagen G, Pommier Y: Ecteinascidin-743
induces protein-linked DNA breaks in human
colon carcinoma HCT116 cells and is
cytotoxic independently of topoisomerase I
expression. Clin. Cancer Res. 7(1),185191
(2001).
31 Erba E, Bergamaschi D, Bassano L et al.:
Ecteinascidin-743 (ET-743), a natural marine
compound, with a unique mechanism of
action. Eur. J. Cancer 37(1), 97105 (2001).
32 Bonfanti M, La Valle E, Fernandez Sousa
Faro JM et al.: Effect of ecteinascidin-743 on
the interaction between DNA binding
proteins and DNA. Anticancer Drug Des.
14(3), 179186 (1999).
33 Garca-Nieto R, Manzanares I, Cuevas C,
Gago F: Increased DNA binding specicity
for antitumor ecteinascidin 743 through
proteinDNA interactions? J. Med. Chem.
43(23), 43674369 (2000).
34 Marco E, Garca-Nieto R, Mendieta J,
Manzanares I, Cuevas C, Gago F: A 3.
(ET743)-DNA complex that both resembles
an RNADNA hybrid and mimicks zinc
nger-induced DNA structural distortions.
J. Med. Chem. 45(4), 871880 (2002).
35 Minuzzo M, Marchini S, Broggini M,
Faircloth G, DIncalci M, Mantovani R:
Interference of transcriptional activation by
the antineoplastic drug ecteinascidin-743.
Proc. Natl Acad. Sci. USA 97(12), 67806784
(2000).
36 Jin S, Gorfajn B, Faircloth G, Scotto KW:
Ecteinascidin-743, a transcription-targeted
chemotherapeutic that inhibits MDR1
activation. Proc. Natl Acad. Sci. USA 97(12),
67756779 (2000).
37 Kanzaki A, Takebayashi Y, Ren XQ et al.:
Overcoming multidrug drug resistance in
P-glycoprotein/MDR1-overexpressing cell
lines by ecteinascidin 743. Mol. Cancer Ther.
1(14), 13271334 (2002).
38 Erba E, Bergamaschi D, Bassano L et al.:
Isolation and characterization of an IGROV-1
human ovarian cancer cell line made resistant
to Ecteinascidin-743 (ET-743). Br. J. Cancer.
82(10), 17321739 (2000).
39 Louneva N, Saitta B, Herrick DJ, Jimenez SA:
Transcriptional inhibition of type I collagen
gene expression in scleroderma broblasts by
the antineoplastic drug ecteinascidin 743.
J. Biol. Chem. 278(41), 4040040407 (2003).
40 Shao L, Kasanov J, Hornicek FJ, Morii T,
Fondren G, Weissbach L: Ecteinascidin-743
drug resistance in sarcoma cells:
transcriptional and cellular alterations.
Biochem. Pharmacol. 66(12), 23812395
(2003).
41 Garcia-Rochal M, Garcia-Gravalos MD, Avila
J: Characterisation of antimitotic products
from marine organisms that disorganise the
microtubule network: ecteinascidin 743,
isohomohalichondrin-B and LL-15.
Br. J. Cancer 73(8), 875883 (1996).
42 Friedman D, Hu Z, Kolb EA, Gorfajn B,
Scotto KW: Ecteinascidin-743 inhibits
activated but not constitutive transcription.
Cancer Res. 62(12), 33773381 (2002).
n
Provides an overview of the specic
mechanism of inhibition of activated
transcription by ecteinascidin 753.
43 Synold TW, Dussault I, Forman BM:
The orphan nuclear receptor SXR
coordinately regulates drug metabolism and
efux. Nat. Med. 7(5), 584590 (2001).
44 Xie W, Barwick JL, Downes M et al.:
Humanized xenobiotic response in mice
expressing nuclear receptor SXR. Nature
406(6794), 435439 (2000).
45 Martinez EJ, Corey EJ, Owa T: Antitumor
activity- and gene expression-based proling
of ecteinascidin ET 743 and phthalascidin PT
650. Chem. Biol. 8(12), 11511160 (2001).
46 Marchini S, Marrazzo E, Bonomi R et al.:
Molecular characterisation of two human
cancer cell lines selected in vitro for their
chemotherapeutic drug resistance to ET-743.
Eur. J. Cancer. 41(2), 323333 (2005).
47 Minuzzo M, Ceribelli M, Pitarque-Mart M
et al.: Selective effects of the anticancer drug
Yondelis (ET-743) on cell-cycle promoters.
Mol. Pharmacol. 68(5), 14961503 (2005).
48 Gajate C, An F, Mollinedo F: Differential
cytostatic and apoptotic effects of
ecteinascidin-743 in cancer cells.
Transcription-dependent cell cycle arrest and
transcription-independent JNK and
mitochondrial mediated apoptosis. J. Biol.
Chem. 277(44), 4158041589 (2002).
49 Takebayashi Y, Pourquier P, Zimonjic DB,
et al.: Antiproliferative activity of
ecteinascidin 743 is dependent upon
transcription-coupled nucleotide-excision
repair. Nat. Med. 7(8), 961966 (2001).
50 Damia G, Silvestri S, Filiberti L, Broggini M,
DIncalci M: Importance of DNA repair
mechanisms for the sensitivity of tumor cells
to ET-743. Proceedings of the 1999
AACRNCIEORTC International Conference
on Molecular Targets and Cancer Therapeutics.
Washington DC, USA, 1619 November
1999 (Abstract 3872).
51 Zewail-Foote M, Li VS, Kohn H, Bearss D,
Guzman M, Hurley LH: The inefciency of
incisions of ecteinascidin 743-DNA adducts
by the UvrABC nuclease and the unique
structural feature of the DNA adducts can be
used to explain the repair-dependent toxicities
of this antitumor agent. Chem. Biol. 8(11),
10331049 (2001).
52 Damia G, Silvestri S, Carrassa L et al.:
Unique pattern of ET-743 activity in different
cellular systems with dened deciencies in
DNA-repair pathways. Int. J. Cancer 92,
583588 (2001).
53 Tavecchio M, Natoli C, Ubezio P, Erba E,
DIncalci M: Dynamics of cell cycle phase
perturbations by trabectedin (ET-743) in
nucleotide excision repair (NER)-decient
and NER-procient cells, unravelled by a
novel mathematical simulation approach.
Cell Prolif. 40, 885904 (2007).
Pharmacogenomics (2010) 11(6)
876 future science group
Review Vincenzi, Napolitano, Frezza, Schiavon, Santini & Tonini
54 Aune GJ, Takagi K, Sordet O et al.:
Von HippelLindau coupled and
transcription-coupled nucleotide excision
repair dependent degradation of RNA
polymerase II in response to trabectedin.
Clin. Cancer Res. 14(20), 64496455 (2008)
55 Grazziotin Soares D, Escargueil AE,
Poindessous V et al.: Replication and
homologous recombination repair regulate
DNA double-strand break formation by the
antitumor alkylator ecteinascidin 743. Proc.
Natl Acad. Sci. USA 104(32), 1306213067
(2007).
56 Guirouilh-Barbat J, Redon C, Pommier Y:
Transcription-coupled DNA double-strand
breaks are mediated via the nucleotide
excision repair and the Mre11Rad50Nbs1
complex. Mol. Biol. Cell. 19(9), 39693981
(2008).
57 Tavecchio M, Simone M, Erba E et al.:
Role of homologous recombination in
trabectedin-induced DNA damage.
Eur. J. Cancer 44(4), 609618 (2008).
n
Describes the crosstalk of DNA repair
pathways in trabectedin-induced
DNA damage.
58 Marco E, Gago F: DNA structural similarity
in the 2:1 complexes of the antitumor drugs
trabectedin (Yondelis) and chromomycin A3
with an oligonucleotide sequence containing
two adjacent TGG binding sites on opposing
strands. Mol. Pharmacol. 68, 15591567
(2005).
59 Casado JA, Ro P, Marco E et al.:
Relevance of the Fanconi anemia pathway in
the response of human cells to trabectedin.
Mol. Cancer Ther. 7(5), 13091318 (2008).
60 Andreassen PR, Ho GP, DAndrea AD:
DNA damage responses and their many
interactions with the replication fork.
Carcinogenesis 27, 883892 (2006).
61 Sparidans RW, Rosing H, Hillebrand MJX
et al.: Search for metabolites of ecteinascidin
743, a novel, marine-derived, anti-cancer
agent, in man. Anticancer Drugs 12, 653666
(2001).
62 Beumer JH, Rademaker-Lakhai JM,
Rosing H et al.: Metabolism of trabectedin
(ET-743, Yondelis) in patients with advanced
cancer. Cancer Chemother. Pharmacol. 59,
825837 (2007).
n
Review of the specic methabolic
pathways of ecteinascidin 743 and its
biological derivatives
63 Beumer JH, Rademaker-Lakhai JM, Rosing
H, Lopez-Lazaro L, Beijnen JH, Schellens
JH: Trabectedin (Yondelis, formerly ET-743),
a mass balance study in patients with
advanced cancer. Invest. New Drugs 23(5),
429436 (2005).
64 Reid JM, Kuffel MJ, Ruben SL et al.: Rat and
human liver cytochrome P-450 isoform
metabolism of ecteinascidin 743 does not
predict gender-dependent toxicity in humans.
Clin. Cancer Res. 8, 29522962 (2002).
65 Brandon EFA, Meijerman I, Klijn JS et al.:
In-vitro cytotoxicity of ET-743 (trabectedin,
Yondelis), a marine anti-cancer drug, in the
Hep G2 cell line: inuence of cytochrome
P450 and phase II inhibition, and cytochrome
P450 induction. Anticancer Drugs 16,
935943 (2005).
66 Brandon EFA, Sparidans RW, Guijt K et al.:
In vitro characterization of the human
biotransformation and CYP reaction
phenotype of ET-743 (Yondelis, trabectedin),
a novel marine anticancer drug. Invest. New
Drugs 24, 314 (2006).
67 Levy RH, Thummel KE, Trager WF,
Hansten PD, Eichelbaum M: Metabolic Drug
Interactions. Lippincott Williams and
Wilkins, PA, USA (2000).
68 Mirsalis JC, Schindler-Horvat JE,
Tomaszewski JE et al.: Preclinical toxicology
studies of ecteinascidin 743. Proc. Am. Assoc.
Cancer Res. 37, 375 (1996).
69 Mirsalis JC, Fairchild DG, Smith S et al.:
Toxicity of ecteinascidin 743 in female
Fischer-344 rats administered i.v. in a
multiple-dose regimen. Proc. Am. Assoc.
Cancer Res. 38, 309 (1997).
70 Donald S, Verschoyle RD, Edwards R et al.:
Hepatobiliary damage and changes in hepatic
gene expression caused by the antitumor drug
ecteinascidin-743 (ET-743) in the female rat.
Cancer Res. 62(15), 42564262 (2002).
71 Valoti G, Nicoletti MI, Pellegrino A et al.:
Ecteinascidin-743, a new marine product with
potent antitumor activity on human ovarian
carcinoma xenografts. Clin. Cancer Res. 4,
19771983 (1998)
72 Donald S, Verschoyle RD, Greaves P et al.:
Complete protection by high-dose
dexamethasone against the hepatotoxicity of
the novel antitumor drug Yondelis (ET-743)
in the rat. Cancer Res. 63, 59025908 (2003).
73 Hari P, Srivastava RN: Pulse corticosteroid
therapy with methylprednisolone or
dexamethasone. Ind. J. Pediatr. 65, 557560
(1998).
74 Sprung CL, Caralis PV, Marcial EH et al.:
The effects of high-dose corticosteroids in
patients with septic shock. A prospective,
controlled study. N. Engl. J. Med. 311,
11371143 (1984).
75 Leibelt DA, Hedstrom OR, Fischer KA,
Pereira CB, Williams DE: Evaluation of
chronic dietary exposure to indole-3-carbinol
and absorption-enhanced 3,3
-diindolylmethane in SpragueDawley rats.
Toxicol. Sci. 74, 1021 (2003).
76 Donald S, Verschoyle RD, Greaves P et al.:
Dietary agent indole-3-carbinol protects
female rats against the hepatotoxicity of the
antitumor drug ET-743 (trabectedin) without
compromising efcacy in a rat mammary
carcinoma. Int. J. Cancer 111(6), 961967
(2004).
77 Donald S, Verschoyle RD, Greaves P, Orr S,
Jimeno J, Gescher AJ: Comparison of four
modulators of drug metabolism as protectants
against the hepatotoxicity of the novel
antitumor drug Yondelis (ET-743) in the
female rat and in hepatocytes in vitro. Cancer
Chemother. Pharmacol. 53, 305312 (2004).
78 Lee JK, Leslie EM, Zamek-Gliszczynski MJ,
Brouwer KL: Modulation of trabectedin
(ET-743) hepatobiliary disposition by
multidrug resistance-associated proteins
(MRPs) may prevent hepatotoxicity. Toxicol.
Appl. Pharmacol. 228(1), 1723 (2008).
79 Perez-Ruizo JJ, Zannikos P, Hirankarn S
et al.: Population pharmacokinetic meta-
analysis of trabectedin (ET-743, Yondelis) in
cancer patients. Clin. Pharmacokinet. 46(10),
867884 (2007).
80 Fetterly GJ, Owen JS, Stuyckens K et al.:
Semimechanistic pharmacokinetic/
pharmacodynamic model for hepatoprotective
effect of dexamethasone on transient
transaminitis after trabectedin (ET-743)
treatment. Cancer Chemother. Pharmacol.
62(1), 135147 (2008).
81 Villalona-Calero MA, Eckhardt SG, Weiss G
et al.: A phase I and pharmacokinetic study of
ecteinascidin-743 on a daily 5 schedule in
patients with solid malignancies. Clin. Cancer
Res. 8(1), 7585 (2002).
82 Hing J, Perez-Ruixo JJ, Stuyckens K,
Soto-Matos A, Lopez-Lazaro L, Zannikos P:
Mechanism-based pharmacokinetic/
pharmacodynamic meta-analysis of
trabectedin (ET-743, Yondelis) induced
neutropenia. Clin. Pharmacol. Ther. 83(1),
130143 (2008).
83 Gmez SG, Bueren JA, Faircloth GT, Jimeno
J, Albella B: In vitro toxicity of three new
antitumoral drugs (trabectedin, aplidin, and
kahalalide F) on hematopoietic progenitors
and stem cells. Exp. Hematol. 31(11),
11041111 (2003).
84 Allavena P, Signorelli M, Chieppa M et al.:
Anti-inammatory properties of the novel
antitumor agent Yondelis (trabectedin):
inhibition of macrophage differentiation and
cytokine production. Cancer Res. 65(7),
29642971 (2005).
85 Grosso F, Dileo P, Sanlippo R et al.: Steroid
premedication markedly reduces liver and
bone marrow toxicity of trabectedin in
advanced sarcoma. Eur. J. Cancer 42(10),
14841490 (2006).
www.futuremedicine.com 877 future science group
Wide-spectrum characterization of trabectedin Review
877 www.futuremedicine.com
86 Ryan DP, Supko JG, Eder JP et al.: Phase I
and pharmacokinetic study of ecteinascidin
743 administered as a 72-hour continuous
intravenous infusion in patients with solid
malignancies. Clin. Cancer Res. 7, 231242
(2001)
87 Brain EG: Safety and efcacy of ET-743:
The French experience. Anticancer Drugs 13,
S11S14 (2002).
88 Yovine A, Riofrio M, Blay JY et al.: Phase II
study of ecteinascidin-743 in advanced
pretreated soft tissue sarcoma patients. J. Clin.
Oncol. 22, 890899 (2004).
89 Gomez J, Lopez Lazaro L, Guzman C et al.:
Identication of biochemical parameters that
predict the onset of severe toxicities in
patients treated with ET-743. Presented at:
2000 ASCO Annual Meeting. New Orleans,
LA, USA, 2023 May (2000).
90 Skorupa A, Beldner M, Kraft A, Montero AJ:
Fatal rhabdomyolysis as a complication of
ET-743 (Yondelis) chemotherapy for sarcoma.
Cancer Biol. Ther. 6(7), 10151017 (2007).
91 Theman TA, Hartzell TL, Sinha I et al.:
Recognition of a new chemotherapeutic
vesicant: trabectedin (ecteinascidin-743)
extravasation with skin and soft tissue
damage. J. Clin. Oncol. 27(33), E198E200
(2009).
92 Izbicka E, Lawrence R, Raymond E et al.:
In vitro antitumor activity of the novel marine
agent, ecteinascidin-743 (ET-743,
NSC-648766) against human tumors
explanted from patients. Ann. Oncol. 9(9),
981987 (1998).
93 Valoti G, Nicoletti MI, Pellegrino A et al.:
Ecteinascidin-743, a new marine natural
product with potent antitumor activity on
human ovarian carcinoma xenografts. Clin.
Cancer Res. 4(8), 19771983 (1998).
94 Hendriks HR, Fiebig HH, Giavazzi R,
Langdon SP, Jimeno JM, Faircloth GT: High
antitumour activity of ET743 against human
tumour xenografts from melanoma,
non-small-cell lung and ovarian cancer. Ann.
Oncol. 10(10), 12331240 (1999).
95 Martinez N, Sanchez-Beato M, Carnero A
et al.: Transcriptional signature of
Ecteinascidin-743 (Yondelis, trabectedin) in
human sarcoma cells explanted from
chemo-naive patients. Mol. Cancer Ther. 4(5),
814823 (2005).
96 Forni C, Minuzzo M, Virdis E et al.:
Trabectedin (ET-743) promotes
differentiation in myxoid liposarcoma tumors.
Mol. Cancer Ther. 8(2), 449457 (2009).
97 Takahashi N, Li W, Banerjee D et al.:
Sequence-dependent synergistic cytotoxicity
of ecteinascidin-743 and paclitaxel in human
breast cancer cell lines in vitro and in vivo.
Cancer Res. 62(23), 69096915 (2002).
98 Takahashi N, Li WW, Banerjee D,
Scotto KW, Bertino JR: Sequence-dependent
enhancement of cytotoxicity produced by
ecteinascidin 743 (ET-743) with doxorubicin
or paclitaxel in soft tissue sarcoma cells.
Clin. Cancer Res. 7(10), 32513257 (2001).
99 DIncalci M, Colombo T, Ubezio P et al.:
The combination of Yondelis and cisplatin is
synergistic against human tumor xenografts.
Eur. J. Cancer 39(13), 19201926 (2003).
100 Riccardi A, Meco D, Ubezio P et al.:
Combination of trabectedin and irinotecan is
highly effectivein a human
rhabdomyosarcoma xenograft. Anticancer
Drugs 16, 811815 (2005).
101 Meco D, Colombo T, Ubezio P et al.:
Effective combination of ET-743 and
doxorubicin in sarcoma: preclinical studies.
Cancer Chemother. Pharmacol. 52(2),
131138 (2003).
102 Scotlandi K, Perdichizzi S, Manara MC
et al.: Effectiveness of ecteinascidin-743
against drug-sensitive and -resistant bone
tumor cells. Clin. Cancer Res. 8(12),
38933903 (2002).
103 Morioka H, Weissbach L, Vogel T et al.:
Antiangiogenesis treatment combined with
chemotherapy produces chondrosarcoma
necrosis. Clin. Cancer Res. 9(3), 12111217
(2003).
104 Simoens C, Korst AE, De Pooter CM et al.:
In vitro interaction between ecteinascidin
743 (ET-743) and radiation, in relation to its
cell cycle effects. Br. J. Cancer 89(12),
23052311 (2003).
105 Romero J, Zapata I, Crdoba S et al.: In vitro
radiosensitisation by trabectedin in human
cancer cell lines. Eur. J. Cancer 44(12),
17261733 (2008).
106 Beumer JH, Buckle T, Ouwehand M et al.:
Trabectedin (ET-743, Yondelis) is a substrate
for P-glycoprotein, but only high expression
of P-glycoprotein confers the multidrug
resistance phenotype. Invest. New Drugs
25(1), 17 (2007).
107 Manara MC, Perdichizzi S, Serra M et al.:
The molecular mechanisms responsible for
resistance to ET-743 (trabectedin; Yondelis)
in the Ewings sarcoma cell line, TC-71. Int.
J. Oncol. 27(6), 16051616 (2005).
108 Duan Z, Choy E, Jimeno JM, Cuevas Cdel
M, Mankin HJ, Hornicek FJ: Diverse
cross-resistance phenotype to ET-743 and
PM00104 in multi-drug resistant cell lines.
Cancer Chemother. Pharmacol. 63(6),
11211129 (2009).
109 Moneo V, Serelde BG, Fominaya J et al.:
Extreme sensitivity to Yondelis (trabectedin,
ET-743) in low passaged sarcoma cell lines
correlates with mutated p53. J. Cell. Biochem.
100(2), 339348 (2007).
110 Zhan M, Yu D, Lang A, Li L, Pollock RE:
Wild type p53 sensitizes soft tissue sarcoma
cells to doxorubicin by down-regulating
multidrug reisistance-1 expression.
Cancer. 92, 15561566 (2001).
111 Duan Z, Choy E, Harmon D et al.:
ZNF93 increases resistance to ET-743
(trabectedin; Yondelis) and PM00104
(Zalypsis) in human cancer cell lines. PLoS
One 4(9), E6967 (2009).
112 van Kesteren C, Cvitkovic E, Taamma A
et al.: Pharmacokinetics and
pharmacodynamics of the novel marine-
derived anticancer agent ecteinascidin 743
in a Phase I dose-nding study. Clin.
Cancer Res. 6(12), 47254732 (2000).
113 Taamma A, Misset JL, Riofrio M et al.:
Phase I and pharmacokinetic study of
ecteinascidin-743, a new marine compound,
administered as a 24-hour continuous
infusion in patients with solid tumors.
J. Clin. Oncol. 19, 12561265 (2001).
114 Puchalski TA, Ryan DP,
Garcia-Carbonero R et al.:
Pharmacokinetics of ecteinascidin 743
administered as a 24-h continuous
intravenous infusion to adult patients with
soft tissue sarcomas: associations with
clinical characteristics, pathophysiological
variables and toxicity. Cancer Chemother.
Pharmacol. 50(4), 309319 (2002).
115 van Kesteren C, Twelves C, Bowman A
et al.: Clinical pharmacology of the novel
marine-derived anticancer agent
Ecteinascidin-743 administered as a 1- and
3-h infusion in a Phase I study. Anticancer
Drugs 13(4), 381393 (2002).
116 Lau L, Supko JC, Blaney S et al.:
A Phase I and pharmacokinetic study of
ecteinascidin-743 (Yondelis) in children
with refractory solid tumors. a childrens
oncology group study. Clin. Cancer Res.
11(2 Pt 1), 672677 (2005).
117 Forouzesh B, Hidalgo M, Chu Q et al.:
Phase I and pharmacokinetic study of
trabectedin as a 1- or 3-hour infusion
weekly in patients with advanced solid
malignancies. Clin. Cancer Res. 15(10),
35913599 (2009).
118 von Mehren M, Schilder RJ, Cheng JD
et al.: A Phase I study of the safety and
pharmacokinetics of trabectedin in
combination with pegylated liposomal
doxorubicin in patients with advanced
malignancies. Ann. Oncol. 19(10),
18021809 (2008).
119 Messersmith WA, Jimeno A, Ettinger D
et al.: Phase I trial of weekly trabectedin
(ET-743) and gemcitabine in patients with
advanced solid tumors. Cancer Chemother.
Pharmacol. 63, 181188 (2008).
Pharmacogenomics (2010) 11(6)
878 future science group
Review Vincenzi, Napolitano, Frezza, Schiavon, Santini & Tonini
120 Blay JY, von Mehren M, Samuels BL et al.:
Phase I combination study of trabectedin and
doxorubicin in patients with soft tissue
sarcoma. Clin. Cancer Res. 14(20),
66566662 (2008).
121 Sessa C, Cresta S, Noberasco C et al.:
Phase I clinical and pharmacokinetic study of
trabectedin and cisplatin in solid tumours.
Eur. J. Cancer 45(12), 21162122 (2009).
122 Sessa C, Perotti A, Noberasco C et al.:
Phase I clinical and pharmacokinetic study of
trabectedin and doxorubicin in advanced soft
tissue sarcoma and breast cancer. Eur. J.
Cancer 45(7), 11531161 (2009).
123 Huygh G, Clement PM, Dumez H et al.:
Ecteinascidin-743: evidence of activity in
advanced, pretreated soft tissue and bone
sarcoma patients. Sarcoma 2006, 56282
(2006).
124 Grosso F, Jones RL, Demetri GD et al.:
Efcacy of trabectedin (ecteinascidin-743) in
advanced pretreated myxoid liposarcomas:
a retrospective study. Lancet Oncol. 8(7),
595602 (2007).
125 Grosso F, Sanlippo R, Virdis E et al.:
Trabectedin in myxoid liposarcomas (MLS):
a long-term analysis of a single-institution
series. Ann. Oncol. 20(8), Phase I944
(2009).
126 Tewari D, Saffari B, Cowan C, Wallick AC,
Koontz MZ, Monk BJ: Activity of trabectedin
(ET-743, Yondelis) in metastatic uterine
leiomyosarcoma. Gynecol. Oncol. 102(3),
421424 (2006).
127 Hollebecque A, Adenis A, Taieb S,
Lebedinsky C, Penel N: Inadequacy of
size-based response criteria to assess the
efcacy of trabectedin among metastatic
sarcoma patients. Invest. New Drugs
DOI: 10.1007/s10637-009-9262-4 (2009)
(Epub ahead of print).
128 Delaloge S, Yovine A, Taamma A et al.:
Ecteinascidin-743: a marine-derived
compound in advanced, pretreated sarcoma
patients preliminary evidence of activity.
J. Clin. Oncol. 19(5), 12481255 (2001).
129 Garcia-Carbonero R, Supko JG, Manola J
et al.: Phase II and pharmacokinetic study of
ecteinascidin 743 in patients with progressive
sarcomas of soft tissues refractory to
chemotherapy. J. Clin. Oncol. 22(8),
14801490 (2004).
130 Le Cesne A, Blay JY, Judson I et al.: Phase II
study of ET-743 in advanced soft tissue
sarcomas: a European Organisation for the
Research and Treatment of Cancer (EORTC)
soft tissue and bone sarcoma group trial.
J. Clin. Oncol. 23(3), 576584 (2005).
131 Garcia-Carbonero R, Supko JG, Maki RG
et al.: Ecteinascidin-743 (ET-743) for
chemotherapy-naive patients with advanced
soft tissue sarcomas: multicenter Phase II and
pharmacokinetic study. J. Clin. Oncol.
23(24), 54845492 (2005).
132 Demetri GD, Chawla SP, von Mehren M
et al.: Efcacy and safety of trabectedin in
patients with advanced or metastatic
liposarcoma or leiomyosarcoma after failure of
prior anthracyclines and ifosfamide: results of
a randomized phase ii study of two different
schedules. J. Clin. Oncol. 27(25), 41884196
(2009).
133 Roylance R, Seddon B, McTiernan A,
Sykes K, Daniels S, Whelan J: Experience of
the use of trabectedin (ET-743, Yondelis) in
21 patients with pre-treated advanced sarcoma
from a single centre. Clin. Oncol. (R. Coll.
Radiol.) 19(8), 572576 (2007).
134 Sessa C, De Braud F, Perotti A et al.:
Trabectedin for women with ovarian
carcinoma after treatment with platinum and
taxanes fails. J. Clin. Oncol. 23(9),18671874
(2005).
135 Krasner CN, McMeekin DS, Chan S et al.:
A Phase II study of trabectedin single agent in
patients with recurrent ovarian cancer
previously treated with platinum-based
regimens. Br. J. Cancer 97(12), 16181624
(2007).
136 Del Campo JM, Roszak A, Bidzinski M et al.:
Phase II randomized study of trabectedin
given as two different every 3 weeks dose
schedules (1.5 mg/m2 24 h or 1.3 mg/m2 3 h)
to patients with relapsed, platinum-sensitive,
advanced ovarian cancer. Ann. Oncol. 20(11),
17941802 (2009).
137 McMeekin DS, Lisyanskaya A, Crispens M
et al.: Single-agent trabectedin as second-line
therapy of persistent or recurrent endometrial
cancer: results of a multicenter Phase II study.
Gynecol. Oncol. 114(2), 288292 (2009).
138 Zelek L, Yovine A, Brain E et al.: A Phase II
study of Yondelis (trabectedin, ET-743) as a
24-h continuous intravenous infusion in
pretreated advanced breast cancer.
Br. J. Cancer 94(11), 16101614 (2006).
139 Ryan DP, Puchalski T, Supko JG et al.:
A Phase II and pharmacokinetic study of
ecteinascidin 743 in patients with
gastrointestinal stromal tumors. Oncologist
7(6), 531538 (2002).
140 Blay JY, Le Cesne A, Verweij J et al.:
A Phase II study of ET-743/trabectedin
(Yondelis) for patients with advanced
gastrointestinal stromal tumours. Eur. J.
Cancer 40(9), 13271331 (2004).
141 Paz-Ares L, Rivera-Herreros F, Daz-Rubio E
et al.: Phase II study of trabectedin in
pretreated patients with advanced colorectal
cancer. Clin. Colorectal Cancer 6(7), 522528
(2007).
142 Stevens EV, Nishizuka S, Antony S et al.:
Predicting cisplatin and trabectedin drug
sensitivity in ovarian and colon cancers.
Mol. Cancer Ther. 7(1), 1018 (2008).
n Website
201 Ongoing and unpublished clinical trials
www.clinicaltrials.gov
n Patent
301 Metabolites of Ecteinascidin-743
WO 99/58125 (1999).

You might also like