ArticlePDF AvailableLiterature Review

From the Sea to Anticancer Therapy

Authors:

Abstract and Figures

Discovery, isolation, characterisation and pre-clinical and clinical trials of plant- or animal-derived drugs displaying pharmacological activities continue to develop and enlarge. Cancer chemotherapy is one of the most promising areas for these drugs. Since a very long time, nature has been an attractive source of potential medicinal agents for human use. The deep sea is becoming a novel and potently appealing source for new drugs, as well as shallow waters. This interest is mainly related to the terrific chemical diversity found in the vast number of plants and animal species, as well as in the microbial world. During the evolution, a rich source of biologically active compounds is developed in the depths of the sea, often reflecting ecological adaptation. Most of them (toxins) are developed to allow survival and flourishing acting against predators and parasites. Recent progress in Scuba diving, hi-tech/biotechnological and procedural advances in structure clarification, organic synthesis and biological assay determined the characterisation and preclinical/clinical evaluation of novel anticancer drugs. The aim of this review is to provide a description of their discovery, mode of action and clinical application.
Content may be subject to copyright.
Current Medicinal Chemistry, 2011, 18, 3551 -3562 3551
0929-8673/11 $58.00+.00 © 2011 Bentham Science Publishers Ltd.
From the Sea to Anticancer Therapy
P. Russo*,1, C. Nastrucci1 and A. Cesario1,2
1IRCCS "San Raffaele Pisana", Rome, Italy
2Thoracic Surgery Unit, Catholic University, Rome, Italy
Abstract: Discovery, isolation, characterisation and pre-clinical and clinical trials of plant- or animal-derived drugs displaying pharma-
cological activities continue to develop and enlarge. Cancer chemotherapy is one of the most promising areas for these drugs. Since a
very long time, nature has been an attractive source of potential medicinal agents for human use. The deep sea is becoming a novel and
potently appealing source for new drugs, as well as shallow waters. This interest is mainly related to the terrific chemical diversity found
in the vast number of plants and animal species, as well as in the microbial world. During the evolution, a rich source of biologically
active compounds is developed in the depths of the sea, often reflecting ecological adaptation. Most of them (toxins) are developed to
allow survival and flourishing acting against predators and parasites. Recent progress in Scuba diving, hitech/biotechnological and
procedural advances in structure clarification, organic synthesis and biological assay determined the characterisation and
preclinical/clinical evaluation of novel anticancer drugs. The aim of this review is to provide a description of their discovery, mode of
action and clinical application.
Keywords: Anti-cancer therapy, clinical trial, FDA-approval, EMEA-approval, marine compound, mechanism of action, pre-clinical trial,
synthetic compound.
"The great depths of the ocean are entirely unknown to us.
Soundings cannot reach them. What passes in those remote depths-
what beings live, or can live, twelve or fifteen miles beneath the
surface of the waters-what is the organisation of these animals, we
can scarcely conjecture". Jules Verne, "Twenty Thousand Leagues
Under the Sea" (PART 1 - CHAPTER II, PRO AND CON) [1].
INTRODUCTION
The potential therapeutic effects of natural products have been
recognised since ancient times. Thus, plants and their extracts have
been employed for the treatment of different human diseases for
millennia, and their use has been documented in most ancient ar-
chaeological sources. When in September 1991, in the Tyrolean
Alps (Italy), the well-preserved body of Ötzi, the Iceman (the Simi-
laum man old of 5,300 years) was discovered, some medicinal
herbs were found in his personal effects. These herbs probably were
used to treat the parasites found in his intestines [2]. Although mor-
phine was extracted from opium only in 1817 by Frederick Ser-
turner (1783–1841), ancient civilisations in the Persia, Egypt, and
Mesopotamia cultivated the poppy plant [3]. Thus, in the oldest
medical text, written in Mesopotamia around 2,600 years before
modern era, Papaver somniferum was included in a list of approxi-
mately 1,000 plants and plant-derived substances [4]. The isolation
of morphine from opium - the first isolation of a natural product -
was a seminal event in the development of pharmacology as an
independent discipline. The modern oncology utilizes different
important plant-derived anticancer drugs, such as taxol or vincris-
tine [5]. The newest tendencies in natural drug discovery highlight
the marine ecosystem as a source of novel chemical entities for new
leads in the treatment of cancer [4]. With the ocean covering 70%
of the Earth’s surface, and with the uniqueness of the environmental
conditions present in the oceans, the ocean can be considered a very
promising source of natural drugs – or synthetic derivatives– for the
future [6]. Marine life, during evolution, developed a large number
of organic compounds that are not directly involved in their normal
growth, development or reproduction. Essentially, the functions of
these compounds (toxins) are predator deterrence, prevention of
fouling, inhibition of overgrowth, and protection from ultraviolet
radiation [7]. The richest sources of these toxins are soft-bodied and
mainly sessile organisms, such as sponges, cnidarians, sea slugs,
*Address correspondence to this author at the IRCCS "San Raffaele Pisana", Via di
Valcannuta, 247, I-00166 Roma, Italy; Tel: +39 348 3339704; Fax: +39 06 52255668;
E-mails: patrizia_russo@hotmail.it, patrizia.russo@sanraffaele.it
and tunicates, that lack physical defence against their predators, and
use, for their chemical defence, some cytotoxic secondary metabo-
lites [6]. Since these compounds are released into the water and
therefore rapidly diluted, they have to be characterized by an out-
standing potency to retain their efficacy. According to a recent re-
view the described chemical diversity of the sea totalled 18,552
marine natural products (MNPs), from 27 marine phyla, dominated
by sponge (35.6% total) and cnidarians-derived (18.6% total) com-
pounds [8].
The discovery/isolation of C-nucleosides from the Caribbean
sponge, Cryptotheca crypta, provided the basis for the synthesis of
cytarabine [1- -arabinofuranosylcytosine (Ara-C)], the first marine-
derived anticancer agent for clinical use that is one of the most
effective chemotherapeutic agents in the treatment of acute myeloid
leukaemia (AML) since the 1960s [9]. Moreover, one of its fluori-
nated derivatives, gemcitabine, is used, for an example, as first line,
in combinations with other drugs, in the advanced non-small cell
lung cancer (NSCLC) [10].
Starting to the fifties the isolation and identification of marine
natural products with anti-cancer properties flourished [11]. This
"explosion" is mainly related to the progress made by the develop-
ment and implementation of Scuba diving techniques and sub-
mersibles that allow the “pharmacological exploration” of the seas
to collect the source organisms.
There are currently two Food and Drug Administration (FDA)-
approved drugs in the U.S. Pharmacopeia, namely cytarabine [Cy-
tosar-U1, Depocyt1, (Jun 17 1969) [11]) and Eribulin [Halaven
[12], recently approved also by the European Agency for the
Evaluation of Medicinal Products (EMEA) [13] in cancer treatment.
Trabectedin (Yondelis1) is approved by EMEA in cancer treatment
[14], and is completing key Phase III studies in the U.S. for ap-
proval.
It was estimated that 118 MNPs are in preclinical trials, 22
MNPs in clinical trials and 3 MNPs in the market. MNPs in pre-
clinical trials derived from 16 phyla, namely Porifera (39.0% total),
Chordata (9.7%) and Ascomycota (9.3%). MNPs in clinical trials
were derived from 8 phyla, namely Porifera (34.1% total), Mol-
lusca (18.2%), Chordata (13.6%) and Proteobacteria (13.6%).
MNPs in the market were derived from 2 phyla , with 2 compounds
(Cytarabine and Halaven) from Porifera and 1 compound (Yon-
delis™) from Cho rdata [8].
This review will briefly provide details of two FDA-approved
drugs and for the one EMEA- approved, information concerning
their discovery, mode of action and clinical application. Moreover,
3552 Current Medicinal Chemistry, 20 11 Vol. 18, No. 23 Russo et al.
the reviewed drugs currently under Phase II or III such as Plitidep-
sin (Aplidin), Eribulin mesylate (E7389), ILX651 (synthadotin or
tasidotin), an orally active third-generation Dolastatin 15 analogue,
and Bryostatin-1 have been reviewed. Briefly details of the recently
discovered compounds, such as Spisulosine, Discodermolide, Sali-
nosporamide, or a marine lipid extract from the New Zealand
green-lipped mussel (Perna canaliculus), which entered in Phase I
studies, will be reported. Moreover, two compounds, such as
Neovastat and Zicotinide will also be reviewed here.
CYTARABINE (ARA-C, CYTOSAR-U®;TARABINE PFS)
In 1945 Werner Bergmann, a young organic chemist, collected
some unidentified sponges species in the shallow waters off Elliot
Key, Florida [15]. The same species of sponge was also found in
the waters off Bimini Islands in the Bahamas (3-60 m under water).
The taxonomist de Laubenfels named the sponges Cryptotethia
crypta [15]. After boiling in acetone, a crystalline material, similar
to but not thymidine, was isolated. It was named spongothymidine,
but, according to its chemical structures, its scientific name is "3-
beta-D-arabofuranosylthymine [16]. Spongothymidine differs
slightly from thymidine, a DNA base, thus there is a hydroxyl (-
OH, alcohol) group instead of a hydrogen (-H) at the C-2 carbon
position of the molecule (Fig. 1). At the time of Bergmann's dis-
covery the study of DNA structure and replication was in the initial
stages (in 1953 Watson and Crick published the DNA structure
[17]) as well as the anticancer therapy (in 1955 the United States
Congress created a National Cancer Chemotherapy Service Center
at the National Cancer Institute (NCI) [18]). Having observed that
malignant cells grow out of control, it was supposed that a strategy
to kill them might be blocking their cell (DNA) replication. There-
fore, some scientists started to design or find compounds able to
interfere with the replication of DNA. At that time, the approach
involved changing in the base part of the nucleoside, whereas the
sugar (deoxyribose) remained intact. Then, spongothymidine was
discovered and captivatingly contained nucleosides with a modified
sugar instead of a modified base. This led medical researchers to
design nucleosides with changed sugar moieties. One such nucleo-
side was cytosine arabinoside (cytarabine or Ara- C; Pfizer U.S.
Pharmaceuticals Group) [19].
The seminal work of Seymour Cohen highlighted its antitumor
activity, thus, one of his papers is considered a Citation Classic
[20]. Then, Howard Skipper translated Ara-C from the laboratory
bench to the bed treating childhood ALL [21-23]. Recently FDA
approved intrathecal liposomal cytarabine (ITlipAC) in relapsed or
refractory infant and paediatric leukaemia [24]. More analogues
have been designed, synthesized, and found to be active against
cancer cells. A difluorinated analogue of deoxy-cytidine, namely
gemcitabine (Gemzar®, Fig. 1) [25] is currently considered as the
first line treatment for advanced pancreatic [26] and gastrointestinal
cancer [27] as well as in advanced NSCLC [28] and advanced
breast cancer [29].
Discodermolide (XAA296A)
The light sensitive compound, Discodermolide discovered in
1987 at the Harbor Branch Oceanographic Institution by the
Gunasekera’s group was isolated from the Caribbean marine
sponge Discodermia dissoluta living at 140 m. (330ft) in the sea
[30]. This drug is a macrolide (polyhydroxylated lactone) and a
member of the polyketides’ class of compounds, binding and stabi-
lizing the microtubules of target cells and Salisporamide A, induc-
ing the G2/M stage in the cell cycle, thus arresting cell division and
causing apoptosis in many cancer cell lines [31]. In addition to
anticancer properties, discodermolide possesses immunosuppresive
and cytotoxic activity. It was licensed by Novartis Pharma AG for
commercial development in 1998 and underwent Phase I human
clinical trials, at the beginning showing promising anticancer prop-
erties, particularly for pancreatic cancer and other multi-drug resis-
tant cancers [32]. Treatment with discodermolide provokes late
activation of caspase-3 and -8 and a cleavage of poly(ADP-
ribose)polymerase (PARP) in NSLCS cells [33].
Moreover, this treatment causes an efflux of cytochrome c from
the mitochondria, however, neither Bcl-2 overexpression nor
FADD-negative cells or inhibition of caspases managed to prevent
Fig. (1). Spongothymidine and its derivatives.
Adapted from: [136] sponge; [137] chemical structure thymidine, spongothymidine, Ara-C; [138] chemical structure Gemtamicine.
From the Sea to Anticancer Therapy Current Medicinal Chemistry, 2011 Vol. 18, No. 23 3553
cells from apoptosis [33]. Additionally, release of cathepsin b (not
caspase activation) appears to cause cell death induced by dis-
codermolide because the inhibition of cathepsin b prevents NSLCS
cells from apoptosis [33]. These apoptotic features suggest that an
apoptotic cascade is involved in mediating the cytotoxic effects of
discodermolide. The effects of discodermolide were promising in
murine tumour models, however the pharmaceutical company No-
vartis withdrew it from Phase I trials because of lack of efficacy
and cytotoxicity (mild-to-moderate toxicity from 0.6 mg/m2 to 19.2
mg/m2) in humans [34], nevertheless it is still considered poten-
tially useful in combinatory drug therapy.
Salinosporamide A (NPI-0052)
Another molecule Salinosporamide was isolated in 1991 by
Fenicle and Jensens at the Scripps Institution of Oceanography
(SIO; University of California, San Diego) [35], being discovered
from a cultured “deep water” actinomycete, Salinispora tropica, the
first to be identified as a marine bacteria seawater-requiring [35]. It
shows cytotoxic effects in tumour cells and induces apoptosis in
cells resistant to protease inhibitors, such as bortezomib. Salinospo-
ramide A is a proteasome inhibitor, which inhibits the activity of
proteosomal enzymes like chymotrypsin and trypsin but also acti-
vates caspase-8 and 9. It seems, however that Salinosporamide A
apoptotic pathway differs from the bortezomid pathway because it
does not require signalling via Bax and Bak [36]. The authors be-
lieve that the differences observed in the induction of apoptosis
may be a reason why bortezomib-resistant cells are sensitive to
treatment with Salinosporamide A [36]. In normal lymphocytes and
stem cells derived from bone marrow, Salinosporamide A also
showed a lower cytotoxicity when compared to bortezomib [36].
Finally, Salinosporamide A is undergoing Phase I clinical trials for
the treatment of relapsed and refractory multiple myeloma [37].
The material used for preclinical and clinical evaluation against
drug resistant multiple myelomas, to date is obtained by fermenta-
tion and not by chemical synthesis [reviewed by 35, 38].
Bryostatin 1
The bryostatins belong to a group of 20 novel macrocyclic lac-
tones originally isolated from a Californian population of the foul-
ing community marine bryozoan Bugala neritina (Bugulidae) by
the group of Pettit in 1983 [39] and it is one of the best studied
compounds of this kind. It appears to have a range of properties,
including anti-cancer, anti-tumour, immunostimulant bioactivity
(including immunomodulation and stimulation of haematopoietic
progenitor cells and to activate T-cells). The production of Bry-
ostatin 1 is carried out to date by cultivating Bugala neritina, as a
complete chemical synthesis has not been accomplished. The mo-
lecular site of action was discovered and Bryostatin 1 was found to
bind to protein kinase C with high affinity, perhaps the key for the
anticancer and immunostimulating activities observed. In particular,
Bryostatins’ mode of action consists in binding to the regulatory
domain of the protein kinase C (PKC) competing with natural PKC
ligands like phorbol esters [40], then bryostatin 1 induces PKC
activation by autophosphorylation and translocation to the cell
membrane [41]. Bryostatin 1 appears to prompt different effects on
tumour cell lines, such as inhibiting proliferation, inducing final
differentiation and causing apoptosis [42].
In particular, Bryostatins were tested in combination with other
cytotoxic agents, particularly because Phase II studies involving
single agents have shown minimal activity and as PKC activation
could cause chemoresistance. Bryostatin 1 induces phosphorylation
of the anti-apoptotic protein Bcl-2, thereby inactivating it [43],
taken as a single compound which shows weak apoptotic effects,
but in a combination therapy becomes an interesting agent. For
example, Bryostatin 1 combined with paclitaxel was found to syn-
ergistically increase the upregulation of caspases induced by pacli-
taxel, even in cells overexpressing the anti-apoptotic protein Bcl-xL
[44]. Several Phase II studies have been occurred to date, in particu-
lar, Phase II study of bryostatin-1 (NSC 339555) and paclitaxel in
patients with metastatic or unresectable locally advanced adenocar-
cinoma of the stomach or gastroesophageal junction, the combina-
tion of bryostatin-1 and paclitaxel (weekly) was found to be active
in patients with gastric and gastroesophageal adenocarcinoma (sup-
ported by NCI -National Cancer Institute- Contract No. T99-0103)
[45]. Also in another Phase II trial, in patients with untreated, ad-
vanced gastric or gastroesophageal junction adenocarcinoma, se-
quential paclitaxel followed by bryostatin-1 resulted in a better
response rate than expected by paclitaxel alone [46]. However, a
phase II study of bryostatin-1 and paclitaxel in patients with ad-
vanced non-small cell lung cancer gave disappointing results, the
drugs combination showed no significant clinical response but was
associated with reproducible toxicity, particularly myalgia with no
elevated serum cytokines, suggesting a non-inflammatory etiology
of this toxicity [47]. It consistently appears, following several inde-
pendent studies on the action of Bryostatin 1 for the treatment of
intractable diseases, such as cancers, like leukaemia, melanomas,
adenocarcinima, etc., with or without other compounds for a com-
bined treatment whose most common side effect is myalgia i.e.
muscle pain, but taken as a single therapeutic agent also demon-
strates little efficacy in humans.
Neovastat (AE-941)
Neovastat is a standardized aqueous shark cartilage extract. Al-
though this compound is not strictly derived from the deep sea, it is
classified as a naturally occurring multifunctional antiangiogenic
agent, blocking the formation of blood vessels. Essentially Neovas-
tat prevents tumours form the formation of new capillaries in tu-
mours, thereby depriving them of oxygen and the nutrients they
need to grow, in fact both tumour growth and metastasis are de-
pendent on angiogenesis [48]. Neovastat, administered orally, was
evaluated for safety and efficacy in phase I and II clinical trials in
patients with advanced lung cancer refractory to treatment, at the
beginning with promising results for possible survival benefits,
showing a dose-dependent increase in the median survival time
[49]. More recently, however, a randomized Phase III trial, on pa-
tients with unresectable stage II NSCLC was published [50]. In this
randomized, double-blinded, placebo-controlled Phase III clinical
study, no activity of Neovastat is reported.
Ziconotide (Prialt)
Ziconotide is a synthetic form of peptide extracted from the
venom of predatory tropical cone snails Conus magus, living under
2000 m in the sea [51]. Generally, the pharmacological properties
of conopeptides are interesting, among those the -conotoxin
MVIIA (Ziconitide, Prialt®) has obtained an approval as an analge-
sic drug. Essentially, Ziconotide, binding to presynaptic N-type
calcium channels, reversibly blocks their activity reducing the re-
lease of excitatory neurotransmitter [52]. Ziconotide received FDA
approval in December 2004 (Prialt1 is marketed by Elan Corpora-
tion, PLC [53] as an infusion into the cerebrospinal fluid through an
intrathecal pump system and is currently used against severe
chronic pain in patients with cancer or AIDS [54]. Ziconotide has
also been approved by the EMEA [Doc. rif.: EMEA/772122/2009
EMEA/H/C/551]. Although the therapeutical importance of Zicoti-
nide as anti-pain is undoubtful, it is however beyond the objective
of the review (antineoplastic activity); thus its use in cancer therapy
is mainly for palliative care and therefore included.
Eribulin Mesylate (E7389)
Eribulin mesylate (E7389, Halaven, Eisai Europe, Ltd.), a struc-
turally simplified, synthetic analogue of the MNP Halichondrin B
was recently approved by the U.S. FDA (November 15, 2010) to
3554 Current Medicinal Chemistry, 20 11 Vol. 18, No. 23 Russo et al.
treat patients with metastatic breast cancer who have prior received
at least two chemotherapy regimens for late-stage disease, including
both anthracycline- and taxane-based chemotherapies [12, 55].
Very recently, on 20 January 2011, the EMEA approved Eribulin
mesylate (Halaven) in monotherapy for the following indication:
treatment of patients with locally advanced or metastatic breast
cancer who have progressed after at least two chemotherapeutic
regimens for advanced disease. Prior therapy should have included
an anthracycline and a taxane unless patients were not suitable for
these treatments" [13]. As highlighted by “Nature” in 2010:... It
represents a hard-won victory for the total synthesis of natural
products, a field of chemistry that, although still popular in acade-
mia, had gone out of fashion for many in the pharmaceutical indus-
try" [56].
Complex marine natural products of the Halichondrin class
were isolated by Uemura and Hirata from the western Pacific
sponge Halichondria okadai [57], and later by Pettit et al. from the
Comoros marine sponge Axinella-carteri [58]. Unfortunately, only
miniscule amounts of Halichondrin B were obtained from its ma-
rine source. Thus, Halichondrin B was synthesized de novo [59],
and this work led to the syntheses of multiple “truncated” analogues
including the macrocyclic ketone analogue E7389 [60] (Fig. 2).
E7389 is more potent than the parental compounds in interacting
with tubulin. Both two compounds bind to tubulin and inhibited its
assembly showing to be non-competitive inhibitors of vinblastine or
Dolastatin 10 to tubulin, however, they did not induce an aberrant
tubulin assembly reaction, which is the characteristic of both vin-
blastine (tight spirals) and Dolastatin 10 (aggregated rings and spi-
rals) [61]. It is most likely, as molecular modelling studies sug-
gested, that both drugs form highly unstable, small aberrant tubulin
polymers, rather than the massive stable structures observed with
vinca alkaloids and antimitotic peptides. Moreover, the binding
models explained why the truncated form (E7389) is moderately
more potent than the larger natural product [62]. Recently, it was
reported that E7389 (Eribulin) is more potent than ER-076349 a
similar synthetic analogue (Fig. 2) [63]. It was suggest that Eribu-
lin's in vivo superiority is related to its ability to induce irreversible
mitotic blockade, correlated to persistent drug retention and Bcl-2
phosphorylation. Similar results arose with irreversible vincristine
and reversible vinblastine, indicating persistent cellular retention as
a constituent of irreversibility [63]. Eribulin entered in Phase II-III
clinical trials. It seems particularly promising for pretreated pa-
tients, with several chemotherapic regimen, affected by locally
advanced, recurrent, or metastatic breast cancer (Table 1) [64-67].
Trabectedin ( YondelisTM, Formerly E T-743)
Since 1986, the Developmental Therapeutics Program of the
NCI acquired plants and marine organisms, through collection con-
tracts performed in over 25 tropical and subtropical countries
worldwide [68]. The project found that around 4% of the marine
species examined (mainly animals) contained antitumour com-
pound(s), a number close to that for terrestrial species (mainly
plants). As a part of this program, an extract from the sea squirt
Ecteinascidia turbinata was found, which in 1969, revealed anti-
cancer activity [69]. Trabectedin (also known as ecteinascidin 743
or ET-743 or YondelisTM, PharmaMar in partnership with Johnson
& Johnson Pharmaceutical Research & Development, L.C.C. -
J&JPRD-) is a tetrahydroisoquinoline alkaloid isolated from the
marine tunicate Ecteinascidia turbinate (Fig. 3) with an exclusive
mechanism of action, being able to bind at the minor groove of
DNA and alkylate guanine at the N2 position, whereas most alky-
lating agents bind guanine at position N7 or O6 in the major groove
[for a recent review see ref. 70]. As a consequence Trabectedin
causes perturbation in the transcription of inducible genes (e.g., the
multidrug resistance gene MDR1, HSP70 promoter, type I colla-
gen) and interaction with DNA repair mechanisms (e.g., the nucleo-
tide excision repair pathway) [71]. The transcription-coupled nu-
cleotide excision repair-(TC-NER)-dependent activity of Trabecte-
din is similar to other anticancer agents that cause DNA-binding
enzymes to kill cells. Interestingly, there are evidence of a
Fig. (2). Halichondrin-B and its derivatives.
Adapted from: [139] sponge, [140] Chemical structure E7389, [141] Chemical structure ER-076349, [142] Chemical structure Halichondrin-
B.
From the Sea to Anticancer Therapy Current Medicinal Chemistry, 2011 Vol. 18, No. 23 3555
Table 1. Eribulin in Phase II-III Clinical Trials
Tumour Phase Schedules Evaluation Conclusion Ref.
Extensively pretreated patients with locally
recurrent or metastatic breast cancer.
Phase III
(762 patients randomized 2:1)
1.4 mg/m2i.v.
infusion on days 1 and 8 of
a 21-day cycle
Active
Recommended dose reductions
[64]
Extensively pretreated patients with locally
recurrent or metastatic breast cancer.
Phase III
(1102 patients randomized 1:1 for
Eribulin or capecitabine
1.4 mg/m2i.v.
infusion on days 1 and 8 of
a 21-day cycle
Ongoing [64]
Metastatic breast cancer previously treated
with an anthracycline and a taxane.
Phase II
(87 patients)
1.4 mg/m2i.v.
infusion on days 1 and 8 of
a 28-day cycle
Activity with manageable toler-
ability
[65]
Locally advanced or metastatic breast cancer
previously treated with an anthracycline, a
taxane, and capecitabine.
Phase II
(269 patients)
1.4 mg/m2i.v.
infusion on days 1 and 8 of
a 21-day cycle
Efficacious in a population of
extensively pretreated patients
[66]
Metastatic or recurrent squamous cell carci-
noma of the head and neck.
Phase II
(29 male, 11 female. Thirty-three
patients (83%) with metastatic dis-
ease)
1.4 mg/m2on
days 1 and 8 i.v. every 21-
day cycle.
No clinically significant
activity
[67]
Fig. (3). Yondelis.
Adapted from: [143] Chemical structure Yondelis, [144] Tunicate.
correlation between mutations at the TC-NER system and the ten-
dency to develop cancer [72]. The TC-NER machinery, like some
chemotherapeutic agents, is implicated in the repair of adducts de-
rived by alkylating agents. Trabectedin requires an efficient TC-
NER system to induce cytotoxicity in human cancer cell lines [70]
and this correlation suggests a potential for combined therapy with
platinum salts [70]. Trabectedin is specifically active against cells
that are in G1 phase, making it a unique drug since other DNA-
alkylating agents are typically active on cells undergoing DNA
synthesis [73]. This property makes Trabectedin a good candidate
for antineoplastic activity. The Committee for Medicinal Products
for Human Use concluded that Yondelis’s benefits are greater than
its risks and recommended that it be given marketing authorisation.
Yondelis has been authorised under ‘Exceptional Circumstances’.
Because the numbers of patients with soft-tissue sarcoma and ovar-
ian cancer are low, the diseases are considered ‘rare’, and Yondelis
was designated by EMEA an ‘orphan medicine’ (a medicine used in
rare diseases) on 30 May 2001 (for soft-tissue sarcoma) and on 17
October 2003 (for ovarian cancer) [74]. Also the U.S. FDA on Oc-
tober 7, 2004, granted orphan drug status to Yondelis to treat soft
tissue sarcomas [75]. A recent work states that Trabectedin is a
potentially cost-effective treatment of metastatic soft tissue sar-
coma) patients [76]. Development in patients with other malignan-
cies, such as breast and ovarian carcinoma, is currently ongoing
[77-79]. However, in July 2009 FDA rejected Yondelis (in combi-
nation) to treat ovarian cancer [80].
AplidinTM (Plitidepsin)
Plitidepsin (AplidinTM, PharmaMar USA Inc.) is a cyclic dep-
sipeptide isolated from the marine tunicate Aplidium albicans cur-
rently obtained by chemical synthesis as a didemnin second-
generation (Fig. 4).
Different studies elucidated the mechanism of action of Pliti-
depsin. Briefly it causes modulation of:
i. epidermal growth factor receptors
ii. non-receptor protein-tyrosine kinase SRC
iii. the serine/threonine kinases JNK, and p38 MAPK [81-83]
Moreover induces:
iv. cell cycle arrest at G1-S and inhibition of protein synthesis
[84-85]
v. inhibition of the signal transduction process targeting the
membrane-bound enzyme, palmitoyl thioesterase [86]
vi. inhibition of the vascular endothelial growth factor secre-
tion [87-88]
3556 Current Medicinal Chemistry, 20 11 Vol. 18, No. 23 Russo et al.
vii. inhibition of low molecular weight protein tyrosine phos-
phates [89]
viii. early induction of oxidative stress, activation of Rac1 small
GTPase, Rac1 translocation to cholesterol-rich membrane
domains and the later down-regulation of MAPK phospha-
tase 1 [90].
Phase I programs reported that Aplidin induced neuromuscular
toxicity, asthenia, skin toxicity, and diarrhoea, whereas no haemato-
logical toxicity was observed [91-92]. Since neuromuscular toxicity
appeared similar to that described in the adult form of carnitine
palmitoyl transferase deficiency type 2, a genetic disease treated
with L-carnitine [93], it was concluded that Aplidin interacts with
the carnitine system. Thus, L-carnitine was included to Aplidin
trials to alleviate the neuromuscular toxicity and to allow a further
dose escalation [94]. Table 2 reports Aplidin activity in Phase II
trials. Briefly, Aplidin needs further evaluation in selected patients
whereas it was completely ineffective in advanced or metastatic
urothelium transitional cell carcinoma, relapsed or progressed
SCLC and locally advanced or metastatic NSCLC [95-101].
ILX651 (Synthadotin or Tasidotin)
ILX651 (Genzyme Corp., San Antonio, TX) is an orally active
third-generation Dolastatin 15 analogue with microtubule-targeted
antimitotic activity. Dolastatin 15, a seven-subunit depsipeptide
derived from Dolabella auricularia, common name "wedge sea
hare", is a species of large sea slug, a marine opisthobranch gastro-
pod mollusc in the family Aplysiidae [102]. In tasidotin the car-
boxyl-terminal ester group of Dolastatin 15 has been replaced by a
carboxy-terminal tert-butyl amide (Fig. 5). Tasidotin, as well as its
major metabolite tasidotin C-carboxylate (also called N-
desbenzylamino-cemadotin [103-104], inhibits mitosis without
significantly depolymerising or disorganizing the spindle microtu-
bules. Moreover, tasidotin inhibits weakly tubulin polymerization
into microtubules in vitro and in contrast with other microtubule-
targeted drugs it did not inhibit the growth rate. In contrast to stabi-
lizing plus ends, tasidotin robustly suppresses dynamic instability at
microtubule plus ends, whilst it weakly enhanced microtubule dy-
namic instability at minus ends. Interestingly, tasidotin C-
carboxylate is >10 times more potent than tasidotin. Thus, tasidotin
may be considered a rather weak prodrug, while tasidotin C-
carboxylate, may be the more active intracellular form of the com-
pound. The results imply that the principal mechanism of tasidotin
is suppressing spindle microtubule dynamics [105]. Briefly, tasi-
dotin induces a G2-M block suppressing spindle microtubule in
treated cells that ultimately results in apoptosis [106]. At least three
Phase I studies are being conducted [107-109]. Tasidotin displays a
more favourable toxicity profile compared with other antitubulin
agents (particularly the lack of severe cumulative neuropathy, pe-
Fig. (4). Aplidin.
Adapted from: [145] tunicate, [146] chemical structure Aplidin.
Table 2. Plitidepsin (Aplidin) in Phase II Trials
Tumour Schedules Conclusion Evaluation Ref.
Relapsed-refractory multiple myeloma 5 mg/m2 3 h i.v. infusion every 2 weeks +/-
20 mg/day on days 1 to 4 of oral dexametha-
sone every 2 weeks
Single-agent has limited but reproducible activity in relapsed/
refractory multiple myeloma patients
Activity observed after dexamethasone addition merits further
study
[95]
Advanced or metastatic urothelium transi-
tional cell carcinoma
5 mg/m2 3 h continuous i.v. infusion every 2
weeks
No objective tumour responses [96]
Unresectable advanced medullary thyroid
carcinoma
5 mg/m2 3 h i.v. infusion every 2 weeks Limited clinical activity [97]
Advanced malignant melanoma 5 mg/m2 3 h continuous i.v. infusion every 2
weeks
A minor degree of antitumor activity
Further evaluation in combination schedules may be warranted.
[98]
Unresectable advanced renal cell carcinoma 5 mg/m2 3 h continuous i.v. infusion every 2
weeks +/- L-carnitine
Some antitumor activity in selected patients [99]
Relapsed or progressed SCLC 3.2 mg/m2 1 h weekly i.v. infusion Absence of antitumor activity [100]
Locally advanced or metastatic NSCLC 5 mg/m2 3 h continuous i.v. infusion every 2
weeks +/- L-carnitine
Absence of antitumor activity [101]
From the Sea to Anticancer Therapy Current Medicinal Chemistry, 2011 Vol. 18, No. 23 3557
ripheral enema, and fatigue). Moreover, evidence for antitumour
activity (a minor response in one patient with NSCLC, and a stable
disease, lasting 11 months, in one patient with hepatocellular carci-
noma) warrants further disease-directed evaluations. Phase II stud-
ies, conducted in melanoma, NSCLC, and hormone-refractory pros-
tate cancer did not show sufficient efficacy to warrant further single
agent development using intravenously (iv) route. Tasidotin will be
investigated as an orally administered antineoplastic agent (as the
hydrochloride salt) in patients with advanced, refractory cancer on
the basis of new preclinical data [110].
ES-285 (Spisulosine)
ES-285 (PharmaMar ES-285, PharmaMar USA Inc.), a natural
analogue of the membrane phospholipid sphingosine isolated from
the marine mollusc Spisula polynyma, is a sphingoid-type base
which presents a long unsaturated alkyl chain (C18) and a 1,2-
aminoalcohol motif in an anti relationship (Fig. 6) [111]. Its pri-
mary mechanism of action has not yet been completely revealed.
Since ES-285 induces cytoskeletal abnormalities and lost of actin
stress fibres in exposed cells leading cell shape changes (from a
bipolar and spindle-shaped appearance to a rounded contour [112],
it was postulated that RhoA, a small GTP binding protein regulating
actin stress fibres, was its main target. cDNA microarrays revealed
that ES-285 treatment significantly alters the expression of a large
number of genes including those involved in apoptosis, cell cycle,
signal transduction and the actin cytoskeleton [113]. Moreover,
Spisulosine, in prostate cells, induced apoptosis independently of
the stress-related MAP kinases as well as of PPAR receptor,
PI3K/Akt and classical PKCs, but, interestingly, induced de novo
synthesis of ceramide, that in turn determines activation of PKC!
[114]. Until now only two Phase I studies have been completed.
The first evaluated the safety, pharmacokinetics, pharmacogenom-
ics, and efficacy of ES-285. In this phase I trial, ES-285 was admin-
istered as an i.v. infusion over 24 hours every 3 weeks. The maxi-
mum tolerated dose (MTD) was determined at 256 mg/m2, since
128 mg/m2 was considered the best tolerated. In this population of
pretreated 28 patients, no objective tumour responses and no reduc-
tions in tumour markers were observed. However, stable disease
was recorded in nine patients. Gene expression profiling by cDNA
microarray in paired patient surrogate tissue samples (blood and
skin biopsies), before and after 24-hour infusion of ES-285, showed
that cell cycle genes were the most significantly overrepresented
[115]. The second study, including thirty patients, considered as the
MTD the concentration of 80 - 100 mg/m2/day with the liver en-
zyme elevations as dose limiting. Low antitumour activity was also
observed [116].
GLMLE (Green-Lipped Mussel Lipid Extract, Lyprinol®)
GLMLE (Lyprinol®; Pharmalink Marketing Services Pty. Ltd,
Burleigh Heads, Queensland, Australia) is a marine lipid extract
from the New Zealand green-lipped mussel (Perna canaliculus)
(Fig. 7) obtained by supercritical fluid extraction [for more details
of the technique see Ref. 117] of the stabilised mussel powder using
liquefied carbon dioxide [118]. GLMLE contains a different num-
ber of lipids including sterol esters, triglycerides, free fatty acids
(saturated and unsaturated), sterols and polar lipids. Free fatty acids
(54%) and triglycerides (27%) constitute the predominant lipid
classes of Lyprinol. Further investigation of Lyprinol by silver-ion
thin layer chromatography and gas chromatography-mass spec-
trometry revealed a unique mixture of polyunsaturated fatty acids
(PUFA) containing omega-3. Lyprinol exhibited strong inhibition
of both COX-1 and COX-2, apparently, without selectivity of the
two enzymes [119]. Lyprinol is also a potent inhibitor of the
lipoxygenase (LOX) pathways, in particular, the 5-and 12-LOX
enzymes [120] and induced apoptosis in cultured cell lines [121].
Despite the lack of clinical evidence, there was a significant media
and public interest, supported only by preclinical results, that led to
a considerable use by different patients [122]. Based on the public-
ity, the American Cancer Society continues an online information
page on Lyprinol [123]. In 2002, an Australian study showed that
Lyprinol, administered to thirteen patients with advanced prostate
and breast cancer, did not shrink the tumours or reduce the levels of
pain [124]. Recently (2010), a Phase I study in patients with ad-
vanced breast and prostate cancers did not reach the MTD and no
objective tumour responses were observed [125]. This drug was
included in the review to highlight, once more, the need of con-
trolled human clinical trials for potential therapeutic agents, before
widespread handling, based only on media conjectures.
Fig. (5). Halichondrin-B and its derivatives.
Adapted from: [147] chemical structure Synthadotin, [148] chemical structure Dolastin-15, [149] mollusk.
3558 Current Medicinal Chemistry, 20 11 Vol. 18, No. 23 Russo et al.
Fig. (6). ES-285.
Adapted from: [150] Mollusk, [151] chemical structure ES-285.
Fig. (7). Lyprinol is a marine lipid extract from the New Zea-
land green-lipped mussel (Perna canaliculus)
Adapted from: [152].
CONCLUSION
Since 1945, marine organisms supplied natural products with a
rich source of unusual metabolites [6]. The drug examples reported
in this review showed that biologically different organisms from the
sea continue to provide new small-molecule organic compounds
with effective or potential anticancer activity. The introduction of
some active agents such as Cytarabine, Eribulin mesylate or Yon-
delis helped to change the natural history of some types of human
cancer. Moreover, these compounds offered a great opportunity to
evaluate new and potentially relevant mechanisms of action (i.e.
Yondelis). The understanding of the cellular/molecular pathway(s)
supporting their antitumour activity and the analysis of their phar-
macodynamic(s) might generate valuable information of the genes
engaged in the sensitivity/resistance to these drugs. This approach
might guide to the identification of patterns allowing adapted thera-
pies. Although the mechanisms of action of these compounds are
different, the ultimate antitumour effects are due to induction of
apoptosis in cancer cells.
Finding a novel and potent bioactive natural product by random
screening is, in itself, a big success in drug discovery. Indeed, a
majority of these organisms live below the intertidal zones and
often in the deep layer of the ocean under the thermocline at a depth
of 1000 fathoms (1800 m) or more. These areas, called the benthic
zone, are difficult to access and characterized by poor visibility,
cold water and high pressure. Thus, it is extremely difficult to local-
ize and identify the source of the compounds of interest. Moreover,
the development of a new drug as well as the elucidation and
evaluation of the molecule, usually requires extensive optimisation
of conditions and large quantities of pure compounds exceeding
those that are feasible to collect from a marine environment without
affecting the natural population [126]. Furthermore, information on
the ecological niche(s) of these organisms can be used to rapidly
discover additional natural sources of the same or very similar
compounds. Indeed, the employment of marine eukaryotes for
large-scale production of drugs features several difficulties. They
are largely due to the fact, that, in many cases, the eukaryotic or-
ganism is killed in the process of obtaining the bioactive material,
and the majority of these eukaryotes cannot be cultured in labora-
tory, but need to be hand-picked by Scuba divers [127]. Finally, the
sustainability of these organisms in nature might be compromised.
The challenges of natural product research have resulted in a search
for an alternative to bioactive product development. For instance
high-throughput screening (HTS) of synthetic chemical libraries
and the combinatorial chemistry might be used. Marine natural
product extracts also show numerous problems for a modern drug
discovery program such as : (a) presence of large quantity of inor-
ganic salts, (b) chemical diversity reflecting unlike and seldom
opposite pharmacological activities, (c) major presence of a non-
selective compound possibly covering the activity of minor selec-
tive compounds, (d) concentrations below detection thresholds of
minor compounds present in crude extracts. Fractionation strategies
could effectively split the organic components from the inorganic
salts and it would be useful to concentrate the active principal
components. By having partially purified libraries, daughter plates
can be easily generated for new screening programs [128].
However, in contrast to chemical libraries, marine compounds have
been characterised by diversity and structural complexity that allow
maximum selectivity and interaction with the target [129-130]. On
the other hand, this complexity makes their chemical synthesis
extremely difficult [131-133]. New analytical methods such as
Ultra High Performance Liquid Chromatography (UPLC) coupled
to high resolution mass spectrometers (MS), capillary probe nuclear
magnetic resonance spectrometers, and Desorption Electrospray
Ionisation Mass Spectrometry (DESI-MS) technique allow the
process of natural product discovery from a limited samples [134].
In the area of cancer therapeutics, E. J. Corey and coworkers
published the first enantioselective total synthesis of Ecteinascidin-
743 (Fig. 3) in 1996. Thus, Corey’s brief total synthesis allowed the
From the Sea to Anticancer Therapy Current Medicinal Chemistry, 2011 Vol. 18, No. 23 3559
1996. Thus, Corey’s brief total synthesis allowed the use of this
potent marine drug into advanced clinical trials [135]. Halaven
(Eribulin mesylate) is the result of nearly 25 years of fights in the
laboratory; it symbolizes a great victory in the total synthesis of
natural products, a field of chemistry that, although well-liked by
academia research, was considered unfashionable for many phar-
maceutical industries.
In conclusion, the importance of products from marine sources
might be attributed to the various structures, intricate carbon skele-
tons, different and peculiar mechanisms of action and the ease in
which human bodies would accept these molecules with minimal
manipulation. The current tendency is to discover new precursor
molecules from synthetic molecules, since it is more cost-effective
and a less impactant option for the marine environment. The future
will imply a close collaboration between academic research and the
pharmaceutical industry to develop new drugs from chemical struc-
tures isolated from marine sources. Last but not least, environ-
mental and conservation issues must be considered, as it is our duty
to protect the sea, an invaluable source of living organisms and
novel bioactive compounds, with countless properties, including
those medically important, from inordinate human actions, such as
disrupting fishing practices, like bottom trawling and from over
exploitation and degradation of the coral reefs, as well as to point
out that national and international laws should be able to protect the
biodiversity and ecosystems of the seas. The development of new
drug derived from the marine environment must always include a
plan to supply enough compound for preclinical and clinical phases,
especially when these drugs are found from living animals, inverte-
brates or algae that are found below the intertidal zone.
ACKNOWLEDGEMENTS
We apologize to the many contributors in this field whose work
could not be cited here for space restrictions.
ABBREVIATIONS
Ara-C = 1- -arabinofuranosylcytosine
ALL = Acute Lymphoblastic Leukemia
AML = Acute myeloid leukemia
EMEA = European Agency for the Evaluation of Medicinal
Products
FADD = Fas-Associated protein with Death Domain
FDA = Food and Drug Administration
iv = Intravenously
LOX = Lipoxygenase
MNPs = Marine natural products
MTD = Maximum tolerated dose
NCI = National Cancer Institute
NSCLC = Non-small cell lung cancer
PKC! = Protein kinase C zeta
SCLC = Small Cell Lung Cancer
TC-NER = Transcription-coupled nucleotide excision repair
REFERENCES
[1] http://jv.gilead.org.il/martin/20000_1-02.html (Accessed February 12, 2011).
[2] http://www.archaeologiemuseum.it/it/oetzi-uomo-venuto-dal-ghiaccio (Ac-
cessed February 12, 2011).
[3] Lockermann, G.; Serturner, F.W. The discoverer of morphine. J. Chem.
Educ., 1951, 28, 277.
[4] Newman, D.J. Natural products as leads to potential drugs: an old process or
the new hope for drug discovery? J. Med. Chem., 200 8, 51, 2589–2599.
[5] Cragg, G.M.; Newman, D.J. Plants as a source of anticancer agents; in Eth-
nopharmacology; Elisabetsky, Etkin, Eds. in Encyclopedia of Life Support
Systems (EOLSS), Developed under the Auspices of the UNESCO, Eolss
Publishers, Oxford ,UK, [http://www.eolss.net](Accessed February 12,
2011).
[6] Faulkner, D.J. Marine pharmacology. Antonie Van Leeuwenho ek, 20 00, 77,
135–145.
[7] Scheuer, P.J. Some marine ecological phenomena – chemical basis and
biomedical potential. Science ,1990, 248, 173–177.
[8] Kinghorn, A.D.; Chin, Y.W.; Swanson, S.M. Discovery of natural product
anticancer agents from biodiverse organisms. Curr. Op in. Drug Discov. De-
vel., 2009 , 12, 189-196.
[9] Robak, T.; Wierzbowska, A. Current and emerging therapies for acute mye-
loid leukemia. Clin. Ther., 2009, 31, 2349-2370.
[10] Goffin, J.; Lacchetti, C.; Ellis, P.M.; Ung, Y.C.; Evans, W.K.. Lung Cancer
Disease Site Group of Cancer Care Ontario's Program in Evidence-Based
Care. First-line systemic chemotherapy in the treatment of advanced non-
small cell lung cancer: a systematic review. J. Thorac. Oncol.,2010, 5, 260-
274.
[11] Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over
the last 25 years. J. Nat. Prod.,2007, 70, 461-477.
[12] www.fiercebiotech.com/fda_approval (Accessed February 12, 2011).
[13] http://www.ema.europa.eu/docs/en_GB/document_library/Summary_of_
opinion_-_Initial_authorisation/human/002084/WC500101044.pdf. (Ac-
cessed February 12, 2011).
[14] http://www.prnewswire.co.uk (Accessed February 12, 2011).
[15] Bergmann, W.; Feeney, R.J. Contributions to the Study of Marine Products.
XXXII. The Nucleosides of Sponges. I. J. Org. Chem., 1951, 16, 981-987.
[16] Bergmann, W.; Feeney, R J. The Isolation of a New Thymine Pentoside from
Sponges. J. Am. Chem. Soc., 1950, 72, 2809.
[17] Watson, J.D.; Crick, F. Molecular Structure of Nucleic Acids: A Structure
for Deoxyribose Nucleic Acid. Nature, 171, 1953, 4356, 737-738.
[18] http://pushpanjalihealthcare.co (Accessed February 12, 2011).
[19] Secrist, J.A. 3rd. Nucleosides as anticancer agents: from concept to the
clinic. Nucleic Acids Symp. Ser., Oxford UK, 2005, 49, 15-16.
[20] Cohen, S.S.; Flaks, J.G.; Barner, H.D.; Loeb, M.R.; Lichtenstein, J. The
mode of action of 5-fluorouracil and its derivatives. Proc. Nat. Acad. Science
(USA), 1958, 44, 1004-1012.
[21] Freireich, E.J.; Gehan, E.A.; Rail, D. P.; Schmidt, L.H.; Skipper, H.E. Quanti-
tative Comparison of Toxicity of Anticancer Agents in Mouse, Rat, Hamster,
Dog, Monkey, and Man. Cancer Chemother. Rep., 1966, 50, 219-244.
[22] Skipper, H.E. Criteria associated with destruction of leukemia and solid
tumor cells in animals. Cancer Res., 1967, 27, 2636-2645.
[23] Skipper, H.E.; Perry, S. Kinetics of normal and leukemic leukocyte popula-
tions and relevance to chemotherapy. Cancer Res., 1970; 30, 1883-1897.
[24] Seif, A.E.; Reilly, A.F.; Rheingold, S.R. Intrathecal liposomal cytarabine in
relapsed or refractory infant and pediatric leukemias: the Children's Hospital
of Philadelphia experience and review of the literature. J. Pediatr. Hematol.
Oncol., 2010, 32:, e349-352.
[25] Hertel, L.W.; Kroin, J.S.; Grossman, C.S.; Grindey, G.B.; Dorr, A.F.; Storni-
olo, A.M.V.; Plunkett, W.; Gandhi, V.; Huang, P. Synthesis and Biological
Activity of 2',2'-Difluorodeoxycytidine (Gemcitabine) in Biomedical Fron-
tiers of Fluorine Chemistry Chapter 19, pp 265–278 ACS S ymposium Series,
Vol. 639 August 13, 1996.
[26] Merl, M.Y.; Abdelghany, O.; Li, J.; Saif, M.W. First-line treatment of metas-
tatic pancreatic adenocarcinoma: can we do better? Highlights from the
"2010 ASCO Annual Meeting". Chicago, IL, USA. June 4-8, 2010. J. Pan-
creas (Online), 2010, 11, :317-320.
[27] Javle, M., Hsueh, C.T. Recent advances in gastrointestinal oncology--updates
and insights from the 2009 annual meeting of the American society of clini-
cal oncology. J. Hematol. Oncol., 2010 ,3, 11.
[28] Ramalingam, S.; Belani, C. Systemic chemotherapy for advanced non-small
cell lung cancer: recent advances and future directions. On cologist, 2008, 13
Suppl., 1, 5-13.
[29] Mauri, D.; Polyzos, N.P.; Salanti, G.; Pavlidis, N.; Ioannidis, J.P. Multiple-
treatments meta-analysis of chemotherapy and targeted therapies in advanced
breast cancer. J. Natl. Cancer Inst., 2008, 100, 1780-1791.
[30] Gunasekera, S. P.; Gunasekera, M.; Longley, R. E.; Schulte, G. K. Dis-
codermolide: a new bioactive polyhydroxylated lactone from the marine
sponge Discodermia dissoluta. J. Org. Chem., 1990,55, 4912-4915.
[31] Jordan, M.A. Mechanism of action of antitumor drugs that interact with
microtubules and tubulin. Curr .Med. Chem .An ticancer Agents, 2002, 2, 1-
17.
[32] Kuppens, I.E.Current state of the art of new tubulin inhibitors in the clinic.
Curr. Clin. Pharmacol., 2006, 1, 57-70.
[33] von Schwarzenberg. K.; Vollmar, A.M. Targeting apoptosis pathways by
natural compounds in cancer: Marine compounds as lead structures and
chemical tools for cancer therapy. Cancer Lett ., 2010 Jul 29. Epub ahead of
print
[34] Molinski, T.F.; Dalisay, D.S.; Lievens, S.L.; Saludes, J.P. Drug development
from marine natural products, Nat. Rev. Drug Disco v., 2009, 8, 69-85.
[35] Williams, P.G.; Buchanan, G.O.; Feling, R.H.; Kauffman, C.A.; Jensen, P.R.;
Fenical, W. New cytotoxic salinosporamides from the marine Actinomycete
Salinispora tropica. J. Org. Chem., 2005, 70, 6196-203.
3560 Current Medicinal Chemistry, 20 11 Vol. 18, No. 23 Russo et al.
[36] Chauhan, D.; Catley, L.; Li, G.; Podar, K.; Hideshima, T.; Velankar, M.;
Mitsiades, C.; Mitsiades, N.; Yasui, H.; Letai, A.; Ovaa, H.; Berkers, C.;
Nicholson, B.; Chao, T.H.;Neuteboom, S.T.; Richardson, P.; Palladino,
M.A.; Anderson, K.C. A novel orally active proteasome inhibitor induces
apoptosis in multiple myeloma cells with mechanisms distinct from Borte-
zomib. Cancer Cell, 2005, 8 (), 40719.
[37] Chauhan, D. ; Hideshima, T.; Anderson, K.C. A novel proteasome inhibitor
NPI-0052 as an anticancer therapy. B r. J. Cancer, 2006, 95, 961-965.
[38] Tsueng, G.; Lam, K.S. A low-sodium-salt formulation for the fermentation
of salinosporamides by Salinispora tropica strain NPS21184, Appl. Micro-
biol. Biotechnol., 2008, 78, 821-826.
[39] Pettit, G.R. Progress in the discovery of biosynthetic anticancer drugs. J. Nat.
Prod., 1996, 59, 812-821.
[40] Wender, P.A.; Cribbs, C.M.; Koehler, K.F.; Sharkey, N.A.; Herald, C.L.;
Kamano, Y.; Pettit, G .R.; Blumberg, P.M. Modeling of the bryostatins to the
phorbol ester pharmacophore on protein kinase C. Proc. Natl. Acad. Sci.(
USA), 1988, 85, 7197-7201.
[41] Isakov, N.; Galron, D.; Mustelin, T.; Pettit, G.R.; Altman, A. Inhibition of
phorbol ester-induced T cell proliferation by bryostatin is associated with
rapid degradation of protein kinase C. J. Immunol., 1993, 150, 1195-1204.
[42] Kortmansky, J.; Schwartz, G.K. Bryostatin-1: a novel PKC inhibitor in
clinical development. Cancer Invest , 2003, 21, 924-936.
[43] Kornblau, S.M. ; Konopleva, M.; Andreeff, M.Apoptosis regulating proteins
as targets of therapy for haematological malignancies. Exp ert. Opin. Investig.
Drugs, 1999, 8, 2027-2057.
[44] Wang, S.; Wang, Z.; Boise, L.H.; Dent, P.; Grant, S. Bryostatin 1 enhances
paclitaxel-induced mitochondrial dysfunction and apoptosis in human leu-
kemia cells (U937) ectopically expressing Bcl-xL. Leukemia, 1999, 13,
1564-1573.
[45] http://www.asco.org/ascov2/Meetings/Abstracts?&vmview=abst_detail_view
&confID=23&abstractID=102344.
[46] Ajani. J.A.; Jiang, Y.; Faust, J.; Chang, B.B. ; Ho, L.; Yao, J.C.; Rousey, S.;
Dakhil, S.; Cherny, R.C.; Craig, C.; Bleyer, A., A multi-center phase II study
of sequential paclitaxel and bryostatin-1 (NSC 339555) in patients with un-
treated, advanced gastric or gastroesophageal junction adenocarcinoma. In-
vest. New Drugs, 2006, 24, 353-357.
[47] Winegarden, J.D.; Mauer, A.M.; Gajewski, T.F.; Hoffman, P.C.; Krauss, S.;
Rudin, C.M.; Vokes, E.E.A phase II study of bryostatin-1 and paclitaxel in
patients with advanced non-small cell lung cancer. Lu ng Cancer, 2003, 39,
191-196.
[48] Folkman, J, ; Shing, Y. Angiogenesis. J Biol Chem, 1992, 267, 10931-10934.
[49] Latreille, J.; Batist, G.; Laberge, F.; Champagne, P.; Croteau, D.; Falardeau,
P.; Levinton, C.; Hariton, C.; Evans, W,K.; Dupont, E. Phase I/II trial of the
safety and efficacy of AE-941 (Neovastat) in the treatment of non-small-cell
lung cancer. Clin. Lung Cance r, 2003, 4, 231-236.
[50] Lu, C.; Lee, J.J.; Komaki, R.; Herbst, R.S.; Feng, L.; Evans, W.K.; Choy, H.;
Desjardins, P.; Esparaz, B.T.; Truong, M.T.; Saxman, S.; Kelaghan, J.;
Bleyer, A.; Fisch, M.J. Chemoradiotherapy with or without AE-941 in stage
III non-small cell lung cancer: a randomized phase III trial. J. Natl. Cancer
Inst., 2010, 102, 859-865.
[51] Olivera, B.M. w-Conotoxin MVIIA: from marine snail venom to analgesic
drug. In Drugs from the Sea (Fusetani, N., ed.), 2000 pp. 75-85, Karger.
[52] Molinski, T.F.; Dalisay, D.S.; Lievens, S.L.; Saludes, J.P. Drug development
from marine natural products. Nat. Rev. Drug Disco v., 2009, 8:69-85.
[53] http:// www.elan.com/therapies/products/prialt.asp
[54] Garber, K. Peptide leads new class of chronic pain drugs. Nature Biotech.,
2005, 23, 399.
[55] Traynor, K. Eribulin approved for advanced breast cancer. Am. J. Health
Syst. Pharm., 2011, 68, 6.
[56] Ledford, H. Complex synthesis yields breast-cancer therapy. Nature, 2010,
468, 608-609.
[57] Hirata, Y.; Uemura, D. Halichondrins - antitumor polyether macrolides from
a marine sponge. Pure Appl. Chem., 1986, 58, 701–710.
[58] Pettit, G.R.; Herald, C.L.; Boyd, M.R.; Leet, J.E., Dufresne, C., Doubek,
D.L; Schmidt, J.M.; Cern y, R.L.; Hooper, J.N.A., Rutzler, K.C. Antineoplas-
tic agents. 219. Isolation and structure of the cell growth inhibitory constitu-
ents from the western Pacific marine sponge Axinella sp. J. Med. Chem.,
1991, 34, 3339–3340.
[59] Aicher, T.D.; Buszek, K.R.; Fang, F.G.; Forsyth, C.J.; Jung, S.H.; Kishi, Y.;
Matelich, M.C.; Scola, P.M.; Spero, D.M.; Yoon, S.K. Total Synthesis of
Halichondrin B and Norhalichondrin B. .J. Am. Chem. Soc., 1992, 114, 3162.
[60] Seletsky, B.M.; Wang, Y.; Hawkins, L.D.; Palme, M.H.; Habgood, G.J.;
DiPietro, L.V.; Towle, M.J.; Salvato, K.A.; Wels, B.F.; Aalfs, K.K.; Kishi,
Y.; Littlefield, B.A.; Yu, M.J. Structurally simplified macrolactone ana-
logues of halichondrin B. Bioorg. Med. Chem. Lett., 2 004, 14, 5547-5550.
[61] Jimeno, A. Eribulin: rediscovering tubulin as an anticancer target. Clin.
Cancer Res., 2009, 15, 3903-3905.
[62] Dabydeen, D.A.; Burnett, J.C.; Bai, R.; Verdier-Pinard, P.; Hickford, S.J.;
Pettit, G.R.; Blunt, J.W.; Munro, M.H.; Gussio, R.; Hamel, E. Comparison of
the activities of the truncated halichondrin B analog NSC 707389 (E7389)
with those of the parent compound and a proposed binding site on tubulin.
Mol. Pharmacol., 2006, 70, 1866-1875.
[63] Towle, M.J.; Salvato, K.A.; Wels, B .F.; Aalfs, K.K.; Zheng, W.; Seletsky,
B.M.; Zhu, X.; Lewis, B.M.; Kishi, Y.; Yu, M.J.; Littlefield, B.A. Eribulin
induces irreversible mitotic blockade: Implications of cell-based pharma-
codynamics for In vivo efficacy under intermittent dosing conditions. Cancer
Res., 2011, 71, 496-505.
[64] Twelves, C.; Cortes, J.; Vahdat, L.T.; Wanders, J.; Akerele, C.; Kaufman,
P.A. Phase III trials of eribulin mesylate (E7389) in extensively pretreated
patients with locally recurrent or metastatic breast cancer. Clin. Breast Can-
cer, 2010, 10, 160-163.
[65] Vahdat, L.; Pruitt, B.; Fabian, C.J.; Rivera, R.R.; Smith, D.A.; Tan-Chiu, E.;
Wright, J.; Tan, A.R.; Dacosta, N.A.; Chuang, E.; Smith, J.; O'Shaughnessy,
J.; Shuster, D.E.; Meneses, N.L.; Chandrawansa, K.; Fang, F.; Cole, P.E.;
Ashworth, S.; Blum, J.L. Phase II study of eribulin mesylate, a halichondrin
B analog, in patients with metastatic breast cancer previously treated with an
anthracycline and a taxane. J. Clin. Oncol., 2009, 27, 2954-2961.
[66] Cortes, J.; Vahdat, L.; Blum, J.L.; Twelves, C.; Campone, M.; Roché, H.;
Bachelot, T.; Awada, A.; Paridaens, R.; Goncalves, A.; Shuster, D.E.; Wan-
ders, J.; Fang, F.; Gurnani, R.; Richmond, E.; Cole, P.E.; Ashworth, S.; Alli-
son, M.A. Phase II study of the halichondrin B analog eribulin mesylate in
patients with locally advanced or metastatic breast cancer previously treated
with an anthracycline, a taxane, and capecitabine. J. Clin. Oncol., 2010, 28,
3922-3928.
[67] Arnold, S.M.; Moon, J.; Williamson, S.K.; Atkins, J.N.; Ou, S.H.; Leblanc,
M.; Urba, S.G.; Phase II evaluation of eribulin mesylate (E7389, NSC
707389) in patients with metastatic or recurrent squamous cell carcinoma of
the head and neck: Southwest Oncology Group trial S0618. Invest. New
Drugs, 2009, Nov 25 [Epub ahead of print].
[68] http://dtp.nci.nih.gov/ (Accessed February 12, 2011).
[69] Rinehart, K.L. Antitumor compounds from tunicates. Med. Res. Rev. 2000,
20, 1-27.
[70] D'Incalci, M.; Galmarini, C.M. A review of trabectedin (ET-743): a unique
mechanism of action. Mol. Cancer The r., 2010, 9, 2157-2163.
[71] Vincenzi, B.; Napolitano, A.; Frezza, A.M.; Schiavon, G.; Santini, D.; Ton-
ini, G. Wide-spectrum characterization of trabectedin: biology, clinical activ-
ity and future perspectives. Pharmacogenomics, 2010, 11, 865-878.
[72] de Boer, J.; van Steeg, H.; Berg, R.J.; Garssen, J.; de Wit, J.; van Oostrum,
C.T.; Beems, R.B.; van der Horst, G.T.; van Kreijl, C.F.; de Gruijl, F.R.;
Bootsma, D.; Hoeijmakers, J.H.; Weeda, G. Mouse model for the DNA re-
pair/basal transcription disorder trichothiodystrophy reveals cancer predispo-
sition. Cancer Res., 1999, 59, 3489-3494.
[73] Erba, E.; Bergamaschi, D.; Bassano, L.; Damia, G.; Ronzoni, S.; Faircloth,
G.; D'Incalci, M. Ecteinascidin-743 (Yondelis), a natural marine compound,
with a unique mechanism of action. Eu r. J. Cancer, 2001, 37, 97-105.
[74] http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human
/medicines/000773/human_med_001165.jsp&murl=menus/medicines/medici
nes.jsp&jsenabled=true Doc. Ref.: EMEA/706735/2009 EMEA/H/C/773
(Accessed February 12, 2011).
[75] http://rarediseases.about.com/b/2004/10/10/yondelis-granted-fda-orphan-
drug-status.htm. (Accessed February 12, 2011).
[76] Soini, E.J.; García San Andrés, B.; Joensuu, T. Trabectedin in the treatment
of: cost-effectiveness, cost-utility and value of information. Ann. Oncol.,
2011, 22, 215-223.
[77] Brüning, A.; Mylonas, I. New emerging drugs targeting the genomic integ-
rity and replication machinery in ovarian cancer. Arch. Gynecol. Obstet.,
2010, Nov 17. [Epub ahead of print].
[78] Demetri, G.D.; Chawla, S. P.; von Mehren, M.; Ritch, P.; Baker, L.H.; Blay,
J.Y.; Hande, K.R.; Keohan, M.L.; Samuels, B.L.; Schuetze, S.; Lebedinsky,
C.; Elsayed, Y.A.; Izquierdo, M.A.; Gómez, J.; Park, Y.C.; Le Cesne, A. Ef-
ficacy and safety of trabectedin in patients with advanced or metastatic
liposarcoma or leiomyosarcoma after failure of prior anthracyclines and ifos-
famide: results of a randomized phase II study of two different schedules. J.
Clin. Oncol., 2009, 27, 4188-4196.
[79] McMeekin, D.S.; Lisyanskaya, A.; Crispens, M.; Oza, A.M.; Braly, P.;
Doering, D.; Bayever, E.; Michiels, B.; Markman, M. Single-agent trabecte-
din as second-line therapy of persistent or recurrent endometrial cancer: re-
sults of a multicenter phase II study. Gynecol. Oncol, 2009, 114, 288-292.
[80] http://www.fda.gov/downloads/AdvisoryCommittees/ CommitteesMeeting-
Materials/Drugs/OncologicDrugsAdvisoryCommittee/UCM171149.pdf.
(Accessed February 12, 2011).
[81] Cuadrado, A.; Garcia-Fernandez, L.F.; Gonzalez, L.; Suarez, Y.; Losada, A.:
Alcaide, V.; Martinez, T.; Fernandez-Sousa, J.M.; Sanchez-Puelles, J.M.;
Munoz, A. Aplidin induces apoptosis in human cancer cells via glutathione
depletion and sustained activation of the epidermal growth factor receptor,
Src, JNK, and p38 MAPK.. J. Biol. Chem., 2003, 278, 241-250.
[82] Garcia-Fernandez, L.F.; Losada, A.: Alcaide, V.; Alvarez, A.M.; Cuadrado,
A.; González, L.; Nakayama, K.; Nakayama, K.I.; Fernandez-Sousa, J.M.;
Muñoz, A.: Sanchez-Puelles, J.M. Aplidin induces the mitochondrial apop-
totic pathway via oxidative stress-mediated JNK and p38 activation and pro-
tein kinase C delta. Oncogene, 2 002, 21, 7533-7544.
[83] Cuadrado, A.; Gonzalez, L.; Suarez, Y.; Martinez, T.; Munoz, A. JNK acti-
vation is critical for Aplidin-induced apoptosis. Oncogene, 2004, 23, 4673-
4680.
[84] Geldof, A.A.; Mastbergen, S.C.; Henrar, R.E.; Faircloth, G.T. Cytotoxicity
and neurocytotoxicity of new marine anticancer agents evaluated using in vi-
tro assays. Cancer Chemother. Pharmaco l., 1999, 44, 312-318.
[85] Erba, E.; Bassano, L.; Di Liberti, G.; Muradore, I.; Chiorino, G.; Ubezio, P.;
Vignati, S .; Codegoni, A.; Desiderio, M.A.; Faircloth, G.; Jimeno, J.;
From the Sea to Anticancer Therapy Current Medicinal Chemistry, 2011 Vol. 18, No. 23 3561
D'Incalci, M. Cell cycle phase perturbations and apoptosis in tumour cells
induced by aplidine. Br. J. Cance r, 2002, 86, 1510-1517.
[86] Crews, C.M.; Lane, W.S.; Schreiber, S.L. Didemnin binds to the protein
palmitoyl thioesterase responsible for infantile neuronal ceroid lipofuscino-
sis. Proc. Natl. Acad .Sci. (USA), 1996 , 93, 4316-4319.
[87] Broggini, M.; Marchini, S.V.; Galliera, E., Borsotti, P.; Taraboletti, G.; Erba,
E.; Sironi, M.; Jimeno, J.; Faircloth, G.; Giavazzi, R.; D'Incalci, M. Aplidine,
a new anticancer agent of marine origin, inhibits vascular endothelial growth
factor (VEGF) secretion and blocks VEGF-VEGFR-1 (flt-1) autocrine loop
in human leukaemia cells MOLT-4. Leukemia, 20 03, 17, 52-59.
[88] Erba, E.; Serafini, M.; Gaipa, G.; Tognon, G.; Marchini, S.; Celli , N.;
Rotilio, D.; Broggini, M.; Jimeno, J.; Faircloth, G.; Biondi, A.; D'Incalci, M.
Effect of Aplidin in acute lymphoblastic leukaemia cells. Br. J. Cancer,
2003, 89, 763-773.
[89] Taddei, M.L.; Chiarugi, P.; Cuevas, C.; Ramponi, G.; Raugei, G. Oxidation
and inactivation of low molecular weight protein tyrosine phosphatase by the
anticancer drug Aplidin. Int. J. Cance r, 2006, 118, 2082-2088.
[90] Gonzalez-Santiago, L.; Suarez, Y.; Zarich, N.; Munoz-Alonso, M.J; Cuad-
rado, A.; Martinez, T.; Goya, L.; Iradi, A.; Saez-Tormo, G.; Maier, J.V.;
Moorthy, A.; Cato, A.C.; Rojas, J.M.; Muñoz; A. Aplidin induces JNK-
dependent apoptosis in human breast cancer cells via alteration of glutathione
homeostasis, Rac1 GTPase activation, and MKP-1 phosphatase downregula-
tion. Cell Death Differ., 2006, 13, 1968–1981.
[91] Jimeno, J.; Lopez-Martin, J.A.; Ruiz-Casado, A.; Izquierdo, M.A.; Scheuer,
P.J.; Rinehart, K. Progress in the clinical development of new marine-derived
anticancer compounds. Anticancer Drugs, 2004, 15, 321-329.
[92] Le Tourneau, C.; Raymond, E.; Faivre, S. Aplidine: a paradigm of how to
handle the activity and toxicity of a novel marine anticancer poison. Cur r.
Pharm. Des., 2007, 13, 3427-3439.
[93] Wieser, T.; Deschauer, M.; Olek, K.; Hermann, T.; Zierz, S. Carnitine palmi-
toyltransferase II deficiency: molecular and biochemical analysis of 32 pa-
tients. Neurology, 2003, 60, 1351-1353.
[94] Le Tourneau, C.; Faivre, S.; Ciruelos, E.; Domínguez, M.J.; López-Martín,
J.A.; Izquierdo, M.A.; Jimeno, J.; Raymond, E. Reports of clinical benefit of
plitidepsin (Aplidine), a new marine-derived anticancer agent, in patients
with advanced medullary thyroid carcinoma. Am. J. Clin. Oncol., 2010, 33,
132-136.
[95] Mateos, M. V.; Cibeira, M. T.; Richardson, P.G.; Prosper, F.; Oriol, A.; de la
Rubia, J.; Lahuerta, J.J.; García-Sanz, R.; Extremera, S.; Szyldergemajn, S.;
Corrado, C.; Singer, H.; Mitsiades C.; Anderson, K.C.; Bladé J.; San Miguel,
J. Phase II clinical and pharmacokinetic study of plitidepsin 3-hour infusion
every two weeks alone or with dexamethasone in relapsed and refractory
multiple myeloma. Clin. Cancer Res., 201 0, 16, 3260-3269.
[96] Dumez, H.; Gallardo, E.; Culine, S.; Galceran, J.C.; Schöffski, P.; Droz, J. P.;
Extremera, S.; Szyldergemajn, S.; Fléchon, A. Phase II study of biweekly
plitidepsin as second-line therapy for advanced or metastatic transitional cell
carcinoma of the urothelium. Ma r. Drugs; 2009, 16, 451-463.
[97] Baudin, E.; Droz, J.P.; Paz-Ares, L.; van Oosterom, A.T.; Cullell-Young, M.;
Schlumberger, M. Phase II study of plitidepsin 3-hour infusion every 2
weeks in patients with unresectable advanced medullary thyroid carcinoma.
Am. J. Clin. Oncol., 2010, 33, 83-88.
[98] Eisen T, Thomas J, Miller WH Jr, Gore M, Wolter P, Kavan P, Martín JA,
Lardelli P.Phase II study of biweekly plitidepsin as second-line therapy in
patients with advanced malignant melanoma. Melanoma Res., 2009, 19, 185-
192.
[99] Schöffski, P.; Guillem, V.; Garcia, M.; Rivera, F.; Tabernero, J.; Cullell, M.;
Lopez-Martin, J.A.; Pollard, P.; Dumez, H.; del Muro, X.G.; Paz-Ares, L.
Phase II randomized study of Plitidepsin (Aplidin), alone or in association
with L-carnitine, in patients with unresectable advanced renal cell carcinoma.
Mar. Drugs, 2009, 7, 57-70.
[100] Eisen, T.; Thatcher, N.; Leyvraz, S.; Miller, W.H. Jr.; Couture, F.; Lorigan,
P.; Lüthi, F.; Small, D.; Tanovic, A.; O'Brien, M. Phase II study of weekly
plitidepsin as second-line therapy for small cell lung cancer. Lun g Cancer,
2009, 64, 60-65.
[101] Peschel, C.; Hartmann, J.T.; Schmittel, A.; Bokemeyer, C.; Schneller, F.;
Keilholz, U.; Buchheidt, D.; Millan S.; Izquierdo, M.A.; Hofheinz; R.D.
Phase II study of plitidepsin in pretreated patients with locally advanced or
metastatic non-small cell lung cancer. Lung Can cer, 2008, 60, 374-380.
[102] Bai, R .; Friedman, S.J.; Pettit, G.R.; Hamel, E. Dolastat in 15, a potent an-
timitotic depsipeptide derived from Dolabella auricularia. Interaction with
tubulin and effects of cellular microtubules. B iochem. Pharmacol., 1 992, 23,
43, 2637-2645.
[103] de Arruda, M.; Cocchiaro, C.A.; Nelson, C.M.; Grinnell, C.M.; Janssen, B.;
Haupt, A.; Barlozzari, T. LU103793 (NSC D-669356): a synthetic peptide
that interacts with microtubules and inhibits mitosis. Ca ncer Res., 1995, 55,
3085–3092.
[104] Newman, R.; Fuentes, A.; Covey, J.; Benvenuto, J. Preclinical pharmacology
of the natural marine product dolastatin 10 (NSC 376128). Drug Metab. Dis-
pos., 1994, 22, 428–432.
[105] Ray, A.; Okouneva, T.; Manna, T.; Miller, H.P.; Schmid, S.; Arthaud, L.;
Luduena, R.; Jordan, M.A.; Wilson, L. Mechanism of action of the microtu-
bule-targeted antimitotic depsipeptide tasidotin (formerly ILX651) and its
major metabolite tasidotin C-carboxylate. Cancer Res., 2007, 67, 3767-3776.
[106] Garg, V.; Zhang, W.; Gidwani, P.; Kim, M.; Kolb, E.A. Preclinical analysis
of tasidotin HCl in Ewing's sarcoma, rhabdomyosarcoma, synovial sarcoma,
and osteosarcoma. Clin. Cancer Res., 2007, 13, 5446-5454.
[107] Mita, A.C.; Hammond, L.A.; Bonate, P.L.; Weiss, G.; McCreery, H.; Syed,
S.; Garrison, M.; Chu, Q.S.;, DeBono, J.S.; Jones, C.B.; Weitman, S.; Row-
insky, E.K. Phase I and pharmacokinetic study of tasidotin hydrochloride
(ILX651), a third-generation dolastatin-15 analogue, administered weekly for
3 weeks every 28 days in patients with advanced solid tumors. Clin. Cancer
Res., 2006, 12, 5207-5215.
[108] Cunningham, C.; Appleman, L.J.; Kirvan-Visovatti, M.; Ryan, D.P.; Regan,
E.; Vukelja, S.; Bonate, P.L.; Ruvuna, F.; Fram, R.J.; Jekunen, A.; Weitman,
S.; Hammond, L.A.; Eder, J. P. Jr. Phase I and pharmacokinetic study of the
dolastatin-15 analogue tasidotin (ILX651) administered intravenously on
days 1, 3, and 5 every 3 weeks in patients with advanced solid tumors. Clin.
Cancer Res., 2005, 11, 7825-7833.
[109] Ebbinghaus, S.; Rubin, E.; Hersh, E.; Cranmer, L.D.; Bonate, P.L.; Fram,
R.J.; Jekunen, A.; Weitman, S.; Hammond, L.A. A phase I study of the do-
lastatin-15 analogue tasidotin (ILX651) administered intravenously daily for
5 consecutive days every 3 weeks in patients with advanced solid tumors.
Clin. Cancer Res., 2005, 11, 7807-7816.
[110] http://www.genzymeoncology.com/onc/research/onc_p_tasidotinHydrochl-
oride.asp. (Accessed February 12, 2011).
[111] Faircloth, G.; Cuevas, C. Kahalalide F and ES285: potent anticancer agents
from marine molluscs. Prog. Mol. Subcell. Biol., 2006, 43, 363–379.
[112] Cuadros, R.; Montejo de Garcini, E.; Wandosell, F.; Faircloth, G.; Fernan-
dez-Sousa, J.M.; Avila, J. The marine compound Spisulosine, an inhibitor of
cell proliferation, promotes the disassembly of actin stress fibers. Cancer
Lett., 2000, 152, 23–29.
[113] Baird, R.D.; Clarke, P.A.; Workman, P. ES-285, a novel marine anticancer
agent: investigation of mechanism of action using gene expression microar-
rays. Proc. Am. Assoc. Cancer Res., 2005, 46, Abstract #4111.
[114] Sánchez, A.M.; Malagarie-Cazenave, S.; Olea, N.; Vara, D.; Cuevas, C.;
Díaz-Laviada, I. Spisulosine (ES-285) induces prostate tumor PC-3 and
LNCaP cell death by de novo synthesis of ceramide and PKCzeta activation.
Eur. J. Pharmacol., 2008, 584, 237-245.
[115] Baird, R.D.; Kitzen, J.; Clarke, P.A.; Planting, A.; Reade, S.; Reid, A.;
Welsh, L.; López Lázaro, L.; de las Heras, B.; Judson, I.R.; Kaye, S.B.; Es-
kens, F.; Workman, P.; deBono, J.S.; Verweij, J. Phase I safety, pharmacoki-
netic, and pharmacogenomic trial of ES-285, a novel marine cytotoxic agent,
administered to adult patients with advanced solid tumors. Mol. Cancer
Ther., 2009, 8, 1430-1437.
[116] Vilar, E.; Grünwald, V.; Schöffski, P.; Singer, H.; Salazar, R.; Iglesias, J.L.;
Casado; E.; Cullell- Young, M.; Baselga, J.; Tabernero, J. A phase I dose-
escalating study of ES-285, a marine sphingolipid-derived compound, with
repeat dose administration in patients with advanced solid tumors. Invest.
New Drugs, 2010, Sep 7 [Epub ahead of print].
[117] http://www.cyberlipid.org/extract/extr0008.htm.
[118] Macrides, T.A.; Kalafatis, N. Super-critical lipid extract from mussels having
anti inflammatory activity. U.S, Patent 6,083,536, July 4, 2000.
[119] McPhee, S.; Hodges, L.D.; Wright, P.F.; Wynne, P.M.; Kalafatis, N.; Har-
ney, D.W.; Macrides, T.A. Anti-cyclooxygenase effects of lipid extracts
from the New Zealand green-lipped mussel, Perna canaliculus. Comp . Bio-
chem. Physiol. B Biochem. Mol. Biol., 2007, 146, 346-356.
[120] Whitehouse, M.W.; Macrides, T.A.; Kalafatis, N.; Betts, W.H.; Haynes,
D.R.; Broadbent J. Anti-inflammatory activity ofa lipid fraction (Lyprinol)
from the NZ green-lipped mussel. Inflammopharmacology, 1997, 5, 237–
246.
[121] Zhang, C.; Betts, W.; Chai, F.; Betts, W.H.; Zalewski, P.D. Lyprinol, a lipid
extract from the New Zealand green-lipped mussel, is a lipoxygenase inhibi-
tor that induces apoptosis in human tumour cells. J. Nutr., 2001, 131 (Suppl),
196s (Abstr 1).
[122] BBC News | Health | Mussel offers cancer hope - [Sunday, August 1, 1999
Published at 11:07 GMT 12:07 UK, http://news.bbc.co.uk/2/hi/health/
409147.stm]. (Accessed February 12, 2011).
[123] http://www.cancer.org/docroot/ETO/content/ETO_5_3X_Lyprinol.asp.
(Accessed February 12, 2011).
[124] Dickson, J.; Pittman, K.; Patterson, K.; Price, T.; Norman, J.; Moldovan, S.;
Moretti, K.; Miller, J.; Burfield, G.; Betts, H. A phase 1/2 study of Lyprinol
in advanced hormone refractory prostate cancer, and hormone and chemo-
therapy refractory breast cancer. Proc. Am. Soc. Clin. Oncol., 2002 , Abstract
2154.
[125] Sukumaran, S.; Pittman, K.B.; Patterson, W.K.; Dickson, J.; Yeend, S.,
Townsend, A.; Broadbridge, V.; Price, T.J. A phase I study to determine the
safety, tolerability and maximum tolerated dose of green-lipped mussel
(Perna canaliculus) lipid extract, in patients with advanced prostate and
breast cancer. Ann. Oncol., 2010, 21, 1089-1093.
[126] Jensen, P.R.; Mincer, T.J.; Williams, P.G.; Fenical, W. Marine actinomycete
diversity and natural product discovery. Antonie Van Leeuwenhoek, 2005,
87, 43–48.
[127] Molinski, T.F.; Dalisay, D.S.; Lievens, S.L.; Saludes, J.P. Drug development
from marine natural products. Nat. Rev. Drug Disco v., 2009, 8, 69–85.
[128] Bugni, T.S.; Harper, M.K.; McCulloch, M.W.; Reppart, J.; Ireland, C.M.
Fractionated marine invertebrate extract libraries for drug discovery. Mo le-
cules, 2008, 19, 13, 1372-1383.
3562 Current Medicinal Chemistry, 20 11 Vol. 18, No. 23 Russo et al.
[129] Singh, S.B.; Barrett, J.F. Empirical antibacterial drug discovery – Foundation
in natural products. Biochem. Pha rmacol., 2006, 71, 1006–1015.
[130] Larsson, J.; Gottfries, J.; Muresan, S.; Backlund, A. ChemGPS-NP: Tuned
for navigation in biologically relevant chemical space. J. Nat. Prod., 2007,
70, 789–794.
[131] Henkel, T.; Brunne, R.M.; Muller, H.; Reichel, F. Statistical investigation
into the structural complementarity of natural products and synthetic com-
pounds. Angew. Chem. Int. Ed., 1999 , 38, 643–647.
[132] Verdine, G. The combinatorial chemistry of nature. Nature, 1996, 384, 11–
13.
[133] Tulp, M.; Bohlin, L. Unconventional natural sources for future drug discov-
ery. Drug Discov. Today, 2004, 9, 450–458.
[134] Feher, M.; Schmidt, J.M. Property distributions: Differences between drugs,
natural products, and molecules from combinatorial chemistry. J. Chem. Inf.
Comput. Sci., 2003, 43, 218–227.
[135] Corey, E.J.; Gin, D.Y.; Kania, R.S. Enantioselective Total Synthesis of
Ecteinascidin 743. J. Am. Chem. Soc., 1996, 118, 9202-9203.
[136] http://www.google.it/imgres?imgurl=http://media.diagnosispro.com/drugs/
20100408_9dc35c59-f4f3-43b4-8251-0cf5c06cdc80/gemzar-f001-
v1.jpg&imgrefurl=
[137] http://www.pfeist.net/ALL/arac/sea2arac2.html.
[1138] http://en.diagnosispro.com/drug_information-for/gemzar-gemcitabine-hcl-
for-injection-eli-lilly-and-company-description/3390-241412.
[139] http://www.google.it/imgres?imgurl=http://www.jbl.de/images/gallery/
container/std/39_1657.jpg&imgrefurl.
[140] http://www.google.it/imgres?imgurl=http://4.bp.blogspot.com/_PiEbciaM-
tBE/TBNhH5izl4I/AAAAAAAAAyI/wheROe6yMH4/s320/Eribulin%2Bme
sylate%2B- 01.GIF&imgrefurl.
[141] http://www.google.it/imgres?imgurl=http://www.chemdrug.com/databases/
SYNTHESIS/SYN/28/28719801i.gif&imgrefurl.
[142] http://www.google.it/imgres?imgurl=http://www.chem-station.com/chemist-
db/images/halicondrinB.gif&imgrefurl.
[143] http://www.google.it/imgres?imgurl=http://i293.photobucket.com/albums/
mm53/nexavar/trabectedin/743px-Trabectedin_.png&imgrefurl.
[144] http://www.torinoscienza.it/dossier/nuovi_farmaci_dagli_abissi_4703.
[145] http://www.google.it/imgres?imgurl=http://www.mesa.edu.au/seaweek2011/
images/Aplidium_albicans.jpg&imgrefurl=
[146] http://www.google.it/imgres?imgurl=http://www.translational-
medicine.com/content/figures/1479-5876-4-3-3.gif&imgrefurl=
[147] http://www.chemdrug.com/databases/8_0_doditgutmjvfmnyn.html.
[148] http://www.scbt.kr/datasheet-201449-dolastatin-15.html.
[149] http://www.mayotte-photos-plongee.com/article-lievre-de-mer-dolabella-
auricularia-de-l-ocean-indien-a-mayotte-52390813.html.
[150] http://www.google.it/imgres?imgurl=http://www.aquarium.co.jp/shell/ galle-
ry/nagaubagai.jpg&imgrefurl=http://www.aquarium.co.jp/shell/gallery/
hyouzi.php%3Fnakama%3Dbakagai&usg=__ .
[151] http://www.chemdrug.com/databases/9_0_uqrosqqqigvmckii.html.
[152] http://www.sea-ex.com/fishphotos/mussels,1.html.
Received: March 04, 2011 Revised: June 04, 2011 Accepted: June 05, 2011
... Eribulin mesylate (Halaven ®) for metastatic breast cancer was approved by the FDA in 2010. Researchers from the Eisai Research Institute have assembled a new macrocyclic ketone (Eribulin, E7389) [61]. Halaven ® battle malignancies through reducing tubulin protein, a component of the cytoskeleton necessary to facilitate the development of the tumor and the target of numerous cancer chemotherapy treatments, including taxolin [62]. ...
... Halaven is being consolidated by the new full-scale oncology infrastructure of Eisai. The industry has expanded autonomous marketing in the USA, leveraging its in-house sales team on the roll-out of sales promotion in community oncology clinics and research institutions [61]. ...
Article
Full-text available
The marine environment is an enormous source of marine-derived natural products (MNPs), and future investigation into anticancer drug discovery. Current progress in anticancer drugs offers a rise in isolation and clinical validation of numerous innovative developments and advances in anticancer therapy. However, only a limited number of FDA-approved marine-derived anticancer drugs are available due to several challenges and limitations highlighted here. The use of chitosan in developing marine-derived drugs is promising in the nanotech sector projected for a prolific anticancer drug delivery system (DDS). The cGAS-STING-mediated immune signaling pathway is crucial, which has not been significantly investigated in anticancer therapy and needs further attention. Additionally, a small range of anticancer mediators is currently involved in regulating various JAK/STAT signaling pathways, such as immunity, cell death, and tumor formation. This review addressed critical features associated with MNPs, origin, and development of anticancer drugs. Moreover, recent advances in the nanotech delivery of anticancer drugs and understanding into cancer immunity are detailed for improved human health.
... Several attempts have been made to synthesize the synthetic derivatives of Halichondrin (Fig. 4). [30][31][32] Hemiasterlins It was isolated from Cymbastela, a marine sponge sp. It shows an antimitotic effect at nanomolar concentration. ...
Article
Full-text available
Cancer is a terrifying disease that has become one of the world’s most challenging health concerns, necessitating a proactive approach to its treatment. Nature is a rich source of new chemical entities and a promising avenue for cancer research. Because of their different modes of action on target events in many ways, nature-derived chemicals are considered good possibilities for anticancer drug development. The current review article highlights recent breakthroughs in cancer therapeutics, including natural therapeutic molecules and their mechanisms of action against cancer treatment, as well as their limits, challenges, and prospect. For the review, article data have been collected from previously studied and published research reports, clinical evaluations, case studies, and clinical statistical data generated by scientific research organizations of repute. The chemical structures have been drawn using computational software (chemsketch). The development of promising candidates that can prevent or reduce the proliferation of cancer cells without causing adverse effects from these therapeutics is now underway. Further, nature-derived products are a game-changer because they are simple, safer, environmentally friendly, low-cost, quick, and less toxic than traditional treatment techniques. However, many bioactive chemicals and their analogs have been discovered as possible anticancer treatment options. In the review, the potent therapeutics phytochemicals derived from nature with their tremendous anticancer effects with limitations and prospects have been studied and analyzed based on previously published articles and reports.
... Numerous scientific publications have shown that gastropods are a rich source of bioactive compounds that include steroids, terpenoids, polyketides, FA, and lipopeptides. Many of the drugs found demonstrate anticancer, antibacterial, and antifungal properties [313][314][315][316][317][318][319]. ...
Article
Full-text available
The study of lipopeptides and their related compounds produced by various living organisms from bacteria to marine invertebrates is of fundamental interest for medicinal chemistry, pharmacology, and practical clinical medicine. Using the principles of retrosynthetic analysis of linear and cyclic peptides, the pharmacological activity of unique, unusual, and rare fatty acids (FA) that are part of natural lipopeptides was investigated. To search for new biologically active natural metabolites from natural sources, more than 350 FA incorporated into linear and cyclic peptides isolated from bacteria, cyanobacteria, microalgae, marine invertebrates, fungal endophytes, and microorganisms isolated from sediments are presented. Biological activities have been studied experimentally in various laboratories, as well as data obtained using QSAR (Quantitative Structure-Activity Relationships) algorithms. According to the data obtained, several FA were identified that demonstrated strong antibacterial, antimicrobial, antifungal, or antitumor activity. Along with this, FA have been found that have shown rare properties such as antiviral, antidiabetic, anti-helmintic, anti-inflammatory, anti-psoriatic, anti-ischemic, and anti-infective activities. In addition, FA have been found as potential regulators of lipid metabolism, as well as agents for the treatment of acute neurological disorders, as well as in the treatment of atherosclerosis and multiple sclerosis. For 36 FA, 3D graphs are presented, which demonstrate their predicted and calculated activities.
... As there are several kingdoms of MO: Bacteria, Protozoans, Chromists (including Seaweeds), Fungi, Plants, and Animals including jellyfish, sponges, sea spiders, bryozoans, mussels, sea stars, fish, and whales, they can synthesize several classes of metabolites used to immobilize and capture prey and defend against predators. These molecules enclose the potential to become a lead in AD innovative drug discovery [56][57][58][59][60][61][62][63]. Some of those molecules, the alkaloids, are pharmacologically active secondary metabolites characterized for containing nitrogen in their chemical structures, with at least one nitrogen atom derived directly from an amino acid [64,65]. ...
Article
Full-text available
The incidence of neurodegenerative diseases, such as Alzheimer’s disease (AD), increases continuously demanding the urgent development of anti-Alzheimer’s agents. Marine organisms (MO) have to create their own defenses due to the adverse environment where they live and so synthesize several classes of compounds, such as akaloids, to defend themselves. Therefore, the identification of marine natural products with neuroprotective effects is a necessity. Being that AD is not only a genetic but also an environmental complex disease, a treatment for AD remains to discover. As the major clinical indications (CI) of AD are extracellular plaques formed by β-amyloid (Aβ) protein, intracellular neurofibrillary tangles (NFTs) formed by hyper phosphorylated τ-protein, uncommon inflammatory response and neuron apoptosis and death caused by oxidative stress, alkaloids that may decrease CI, might be used against AD. Most of the alkalolids with those properties are derivatives of the amino acid tryptophan mainly with a planar indole scaffold. Certainly, alkaloids targeting more than one CI, multitarget-directed ligands (MTDL), have the potential to become a lead in AD treatment. Alkaloids to have a maximum of activity against CI, should be planar and contain halogens and amine quaternization.
... As introduced, natural products derive from environmental locations [13,34]. Generally, soil and aquatic environs have been fruitful sources of natural products [10,12,[35][36][37][38][39][40]. Within these environs, commonly identified production hosts include plants and microorganisms with natural products possessing biological functions spanning chemical communication to a weaponized form of competition for resources [41][42][43]. ...
Article
Full-text available
Natural products have had a major impact upon quality of life, with antibiotics as a classic example of having a transformative impact upon human health. In this contribution, we will highlight both historic and emerging methods of natural product bio-manufacturing. Traditional methods of natural product production relied upon native cellular host systems. In this context, pragmatic and effective methodologies were established to enable widespread access to natural products. In reviewing such strategies, we will also highlight the development of heterologous natural product biosynthesis, which relies instead on a surrogate host system theoretically capable of advanced production potential. In comparing native and heterologous systems, we will comment on the base organisms used for natural product biosynthesis and how the properties of such cellular hosts dictate scaled engineering practices to facilitate compound distribution. In concluding the article, we will examine novel efforts in production practices that entirely eliminate the constraints of cellular production hosts. That is, cell free production efforts will be introduced and reviewed for the purpose of complex natural product biosynthesis. Included in this final analysis will be research efforts made on our part to test the cell free biosynthesis of the complex polyketide antibiotic natural product erythromycin.
... Such effects jeopardize further treatment, and eventually lead to advanced stages of malignancy and mortality. Lee Jong-Wook, former Director General of the WHO, has stated that "by the year 2020, cancer could kill more than 10.3 million people per year unless action is taken in both the field of prevention and treatment" (Russo et al. 2011;Schumacher et al. 2011;Indumathy and Dass 2013). In view of this, remarkable advances in cancer research are essential to improving existing therapies and to develop novel cures for cancer. ...
Chapter
There is an increasing need for novel oncotherapeutics to treat cancer and to overcome emerging resistance to current cancer treatment regimen. The marine environment encompasses 70% of the earth's surface and is characterized by unique growth conditions and encloses massive biodiversity. Marine natural products represent validated starting points for drug discovery, since they occupy biologically relevant chemical space. Biodiversity presumably produces chemodiversity as well, providing wider opportunity for discovering unique oncotherapeutics with novel targets and mechanisms of anticancer action. Exploration of marine environment has led to identification of several marine bioactives with promising anticancer activity. Numerous novel bioactive compounds and their synthetic derivatives have displayed in vitro cytotoxic property on neoplastic cell lines and are gaining great attention for further in vivo applications. In this chapter, we are highlighting marine natural products as a new wave of anticancer drugs.
... Terpenes are the most frequently encountered and exhibited a wide range of biological features ranging from antifeedant, anti-inflammatory, antiviral, antifouling, ichthyo-toxic, cytotoxic, to neuroprotective activities (Liang and Guo, 2013). Secondary metabolite compounds have the potency to be used as material anticancer (Russo et al., 2011;Cooper and Yao, 2012;Dobretsov et al., 2016). The study in finding potency bioactive compound as anticancer is important to do due to cancer becomes the highest cause of death in the world. ...
Article
Full-text available
Soft coral is one of the marine organisms that produce secondary metabolites materials and has potency as anticancer. There is much research to find bioactive compounds in the organism, but it still lacks data to find potency of bioactive in soft coral that lives under acidification pressure. The objectives of this study are to determine the characteristic of acidification in Krakatau seas and to assess the potency of cytotoxic activity from soft coral Sarcophyton. Sampling was conducted in three locations, Legon Tuo, Legon Cabe, and Umang-umang Island, to take the sample and measure water quality. Cytotoxic test using MTT method (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolinon) to MCF-7 cancer cell line to measure growth inhibition percentage. Data were analyzed using ANOVA, post hoc analysis, and multivariate analysis. Water quality in Legon Tuo and Legon Cabe have significant different to Umang-umang Island. However, Legon Tuo and Legon Cabe are suitable as acidification site, while Umang-umang Island as the reference site. The characteristic of Krakatau seas, especially in Legon Tuo and Legon Cabe study site, was categorized in low acidification. In the cytotoxic activity, soft coral Sarcophyton from Legon Cabe (acidification site) is more potential than soft coral from Umang-umang Island (reference site).
... N-containing heterocyclic rings generally offer a polarized character which helps in increasing the stability of the molecule-receptor complex by establishing optimum interaction with the receptor and thus, produce anticancer effects [8]. In particular, the structural diversity of biologically active indoles [9] (A-G, Figure 1) and seven-membered rings makes them important structural components in many pharmaceutical agents [8,[10][11][12][13][14][15] Azepines and azepinones have recently gained a considerable amount of attention and interest because of their continuing pharmacological significance. Many natural products and reported scaffolds with anticancer property were found to normally contain tetrahydroazepinone pharmacophore fused with a heterocycle, such as indole, pyrrole or benzofuran, as their main component (H-P, Figure 1) [16][17][18][19][20][21][22][23][24][25][26]. ...
Article
We present here-in the molecular design and chemical synthesis of a novel series of diindoloazepinone derivatives as DNA minor groove binding agents with selective topoisomerase I inhibition. The in vitro cytotoxicity of the synthesized compounds was evaluated against four human cancer cell lines including DU143, HEPG2, RKO and A549 in addition to non-cancerous immortalized human embryonic kidney cells (HEK-293). Compound 11 showed significant cytotoxicity against all the four human cancer cell lines with IC50 values ranging from 4.2 to 6.59 μM. 11 was also found to display 13-fold selective cytotoxicity towards A549 cancerous cells compared to the non-cancerous cell lines (HEK-293). The decatenation, DNA relaxation and intercalation assays revealed that the investigational compounds 10 and 11 act as highly selective inhibitors of Topo-I with DNA minor groove binding ability which was also supported by the results obtained from circular dichroism (CD), UV-visible spectroscopy and viscosity studies. Apoptosis induced by the lead 11 was observed using morphological observations, AO/EB and DAPI staining procedures. Further, dose-dependent increase in the depolarization of mitochondrial membrane was also observed through JC-1 staining. Annexin V-FITC/PI assay confirmed that 11 induced early apoptosis. Additionally, cell cycle analysis indicated that the cells were arrested at sub-G1 phase. Gratifyingly, in silico studies demonstrated promising interactions of 11 with the DNA and Topo I, thus supporting their potential DNA minor groove binding property with relatively selective Topo I inhibition compared to Topo II.
Article
Full-text available
Natural products are substances found in nature that have not been significantly modified by humans [...]
Article
Full-text available
The aquatic ecosystem has proved to be a valuable source of compounds with unusual and special chemical properties, which can be used to enhance the potency and specificity of new drugs by molecular simulation and chemical synthesis. Cancer remains a growing public health threat, and amid recent developments in biomedical research and technology, new anticancer medicines are desperately needed. In both in vitro and in vivo models, marine compounds demonstrated their ability to mediate unique inhibitory actions on a variety of core cellular processes, including apoptosis mechanisms, angiogenesis, proliferation, and invasion, signalling their capacity for use as anticancer drugs. This review focus on the different marine drug structure, possible anticancer mechanism.
Article
The potential of new natural products as an important source for the exploration and development of new drugs and crop protection products is a long way from being exhausted. The statistical analysis of the structures of the natural and synthetically derived compounds has shown conspicuous variations in structural types in the natural products derived from different natural sources, which can be utilized in the search for individual active substances. The occasionally voiced prepossession that natural products have already been sufficiently examined and therefore no more innovations are to be expected can definitely be rejected.
Article
GEMZAR® (gemcitabine·HCl) is a difluorinated analog of deoxy cytidine. It was initially synthesized as a novel anti-viral compound with broad spectrum in vitro activity against both RNA and DNA viruses. However, the compound proved to have a narrow therapeutic index when it was administered daily during the in vivo evaluation of antiviral activity. Using a staggered schedule of administration, GEMZAR is found to be a potent antitumor agent in murine and human xenograft solid tumor models. Studies have shown that gemcitabine diphosphate is a ribonucleotide reductase inhibitor, whereas the triphosphate is a potent and unique terminator of DNA synthesis. In phase I studies a variety of dose schedules were investigated. Based on phase I studies including pharmacokinetic data, phase II studies were initiated. Activity was observed in a variety of solid tumors. The results are specially encouraging for non-small cell lung and pancreatic cancer. GEMZAR is currently undergoing registration review of the Phase III clinical trials for treatment of non-small cell lung and pancreatic cancer.
Article
New antitumor polyether macrolides were successfully isolated from a marine sponge, Halichondria okadai Kadota. One of them, halichondrin B exhibited remarkable in vivo antitumor activity. Physiological properties and structures of these compounds are reported. The structures have been characterized by a long-straight carbon chain, a polyether macrolide, and a novel 2,6,9-trioxatricyclo left bracket 3. 3. 2. 0**3,**7 right bracket decane system which is the first example in natural products.
Article
1. Two new nucleosides have been isolated from the sponge Cryptotethia crypta which had been collected near the coast of Florida. 2. One of the nucleosides has been shown to be a pentofuranosylthymine by its elementary analysis, its hydrolysis to thymine, its formation of a tribenzoate and tri-p-bromobenzoate, and by titration with sodium periodate. The trivial name spongothymidine has been proposed for this nucleoside. 3. Another nucleoside, for which the trivial name spongosine has been proposed, has been shown to be a pentosylmethylaminoöxypurine. The purine has been isolated and converted to a methyldioxypurine. 4. The isolation of thymine and a methylaminooxypurine from the acetone extract of Cryptotethia crypta, collected in the coastal waters of the Bahamas, has been described. 5. It has been shown that the sterol mixture from Cryptotethia crypta consists principally of clionasterol.
Article
A new polyhydroxylated lactone, discodermolide (1), was isolated from a Caribbean marine sponge, Discodermia dissoluta, and its structure was elucidated through a combination of spectroscopic techniques, in particular NMR spectroscopy, and verified by X-ray crystallographic analysis. The carbon skeleton displayed by discodermolide is new; discodermolide is immunosuppressive and cytotoxic