Cancer wars: natural products strike back Christine Basmadjian1,2, Qian Zhao1,2, Embarek Bentouhami3, Amel Djehal1,3, Canan G. Nebigil4, Roger A…. [600187]
REVIEW ARTICLE
published: 01 May 2014
doi: 10.3389/fchem.2014.00020
Cancer wars: natural products strike back
Christine Basmadjian1,2, Qian Zhao1,2, Embarek Bentouhami3, Amel Djehal1,3, Canan G. Nebigil4,
Roger A. Johnson5,M a r i aS e r o v a2, Armand de Gramont2, Sandrine Faivre2,6, Eric Raymond2,6and
Laurent G. Désaubry1*
1Therapeutic Innovation Laboratory, UMR7200, CNRS/University of Strasbourg, Illkirch, France
2AAREC Filia Research, Clichy, France
3L.C.I.M.N Laboratory, Department of Process Engineering, Faculty of Technology, University Ferhat Abbas, Sétif, Algeria
4Biotechnology and Cell Signaling Laboratory, UMR 7242, CNRS/ University of Strasbourg, Illkirch, France
5Department of Physiology and Biophysics, State University of New Y ork, Stony Brook, NY , USA
6Department of Medical Oncology, Beaujon University Hospital, INSERM U728/AP-HP , Clichy, France
Edited by:
Asier Unciti-Broceta, The University
of Edinburgh, UK
Reviewed by:
Luis Álvarez De Cienfuegos
Rodríguez, University of Granada,
Spain
Francisco Franco-Montalbán,
University of Granada, Spain
*Correspondence:
Laurent G. Désaubry, Therapeutic
Innovation Laboratory (UMR 7200),
Faculté de Pharmacie, 7 4 Route du
Rhin, 67 401 Illkirch, France
e-mail: [anonimizat] products have historically been a mainstay source of anticancer drugs, but in
the 90’s they fell out of favor in pharmaceutical companies with the emergence of
targeted therapies, which rely on antibodies or small synthetic molecules identified by
high throughput screening. Although targeted therapies greatly improved the treatment
of a few cancers, the benefit has remained disappointing for many solid tumors, which
revitalized the interest in natural products. With the approval of rapamycin in 2007 , 12 novel
natural product derivatives have been brought to market. The present review describes the
discovery and development of these new anticancer drugs and highlights the peculiarities
of natural product and new trends in this exciting field of drug discovery.
Keywords: natural products, cancer, drug discovery, pharmacognosy, molecular targets, privileged structures
INTRODUCTION
Recent analyses of tooth plaques showed that ∼50,000 years ago
Neanderthals already used medicinal plants to treat their ailments
(Hardy et al., 2012 ). Currently, more than half of humanity does
not have access to modern medicine and relies on traditional
treatments ( Cordell and Colvard, 2012 ). A recent analysis of the
strategies used in the discovery of new medicines showed that
36% of the first-in-class small-molecules approved by U.S. Food
and Drug Administration (FDA) between 1999 and 2008 were
natural products or natural products derivatives ( Swinney and
Anthony, 2011 ).
Natural products are small-molecule secondary metabolites
that contribute to organism survival. These substances display
considerable structural diversity and “privileged scaffolds,” i.e.,
molecular architectures that are tailored to protein binding, as
first coined by Evans in the late 1980s ( Evans et al., 1988 ). Indeed
natural products have evolved to bind biological targets and elicit
biological effects as chemical weapons or to convey information
from one organism to another. Steroid derivatives are often not
considered as natural products because their design is not based
on a research in pharmacognosy, however we subjectively decided
to include them here due to their importance in drug discovery.
The synthesis of aspirin by Charles Gerhard at Strasbourg
faculty of pharmacy in 1853 paved the road for the medici-
nal chemistry of natural products ( Gerhardt, 1853 ). In 1964,
actinomycin became the first natural product approved for an
indication in oncology. Other natural products based medicines
such as anthracyclines, vinca alkaloids, epipodophyllotoxin lig-
nans, camptothecin derivatives, and taxoids that were launched
before 1997, are still an essential part of the armament for treating
cancers.From 1997 to 2007 no new natural product was approved
for the treatment of cancer ( Bailly, 2009 ). With the imminent
achievement of the genome project, the head of a pharmaceuti-
cal company declared that natural products were outdated. Their
development was greatly reduced and many big pharmaceutical
companies closed their departments of natural product chem-
istry ( Bailly, 2009 ). The future was targeted therapies, which
uses fully synthetic molecules or antibodies to target specific
proteins in tumor growth and progression. In some forms of
leukemia, gastrointestinal, prostate or breast cancers, targeted
therapies greatly delayed tumor progression, and/or improved the
life expectancy of the patients. Some tumors with specific onco-
genic addictions (for example fusion proteins leading to ALK
expression in lung cancer or Bcr-Abl in chronic myeloid leukemia,
KIT expression or mutations in GIST or EGFR mutation in lung
cancer, HER2 amplification in breast cancer or MET overex-
pression in liver tumors) greatly benefited from targeted agents.
However, the vast majority of common tumors were found to
be not dependent of a single “targetable” oncogenic activation.
For instance altogether ALK activations and EGFR mutations
account for less than 10% of lung adenocarcinoma and while
those targeted agents are more efficient than chemotherapy in
oncogenic tumors, antitumor effects are limited to few months.
Importantly, most tumors were shown to activate multiple signal-
ing pathway redundancies and adaptive mechanisms that either
render tumors primarily resistant to targeted drugs or facili-
tate acquired resistance to cell signaling inhibition after only few
months of treatments. As a result, the expected progression-free
survival benefit from targeted therapy is often less than 6-months.
For those later forming complex but rather frequent tumors,
chemotherapy alone remains the cornerstone of treatment with
www.frontiersin.org May 2014 | Volume 2 | Article 20 |1
Basmadjian et al. Anticancer natural products
some limited add-on benefits by use of monoclonal antibodies in
a limited proportion of patients. Combinations of several targetedagents have also been proposed to counteract potential adaptivemechanisms although one should notice that combining targetedagent together was more often associated with unacceptable tox-icity than great clinical synergy. Then there is the additionalinfluence of cost-to-benefit concerns. The financial cost of suchtargeted therapies, to patients or health insurance entities, canbe considered enormous, e.g., thousands to tens of thousands ofeuros per day of extended life. However, the net financial benefitto pharmaceutical companies of those agents that are given onlyfor few months (or years) in only a small proportion of patientsin niche indications may lead to restricted investment by pharma-ceutical industries; blockbuster indications usually provide higherrevenues.
These drawbacks are at the origin of the re-emergence of
natural products in oncology. Since 2007, with the approval ofrapamycin and derivatives of it, 12 natural product derivativeshave been approved for the treatment of cancers ( Ta b l e 1 ).
Recently Stuart Schreiber, Paul Clemons and coworkers at the
Broad Institute in Boston performed a bioinformatics analysis ofnatural product targets and demonstrated that natural productsstatically tend to target proteins with a high number of protein–protein interactions that are particularly essential to an organism(Dan ˇcík et al., 2010 ). This observation is consistent with the com-
mon role played by natural products as chemical weapons againstpredators or competitors.
Henkel et al. at Bayer AG in Germany offered a statistical
analysis of the structural differences between natural productsand fully synthetic drugs ( Henkel et al., 1999 ). Compared with
fully synthetic drugs, natural product tend to have more chi-ral centers, more oxygen atoms, less nitrogen atoms, and morevaried ring systems. Complementary analyses of structural fea-tures of natural products have been reviewed ( Lee and Schneider,
2001; Ortholand and Ganesan, 2004; Ganesan, 2008; Grabowskiet al., 2008 ). A consequence of this structural complexity is that
natural products tend to be more selective toward their tar-gets than fully synthetic drugs, and consequently rarely displayoff-target—induced iatrogenicity.
Moreover, complex natural products tend to act through only
one class of molecular target, even though there are some excep-tions. Indeed, taxanes are known to target β-tubulin and interfere
with microtubule dynamics; however they also bind to Bcl-2 toblock its anti-apoptotic activity. Both β-tubulin and Bcl-2 interact
with the orphan nuclear receptor Nur77 (NGFI-B, TR3, NR4A1).Ferlini et al. showed that in fact taxanes mimic the domainof Nur77 involved in the interaction with β-tubulin and Bcl-
2(Ferlini et al., 2009 ). Another example concerns flavaglines,
an emerging family of natural compounds found in medicinalplants of South-East Asia, which display potent anticancer effectsthrough their direct effects on the scaffold proteins prohibitinsand the initiation factor of translation eIF4a ( Basmadjian et al.,
2013; Thuaud et al., 2013 ).
Modifying the structure of a drug may change the nature
of its molecular target. A striking example concerns the notso rational development of the anticancer medicines etoposideand teniposide ( Figure 1 ). Considering that cardiac glycosidesdisplay enhanced pharmacological properties compared to the
cognate aglycone, Sandoz scientists hypothesized that conjugat-ing podophyllotoxin to a glucose moiety could improve theactivity of this cytotoxic agent that binds tubulin and inhibitsassembly of the mitotic spindle. Fortunately, this glycoconju-gate named etoposide displayed a promising anticancer activitywith reduced adverse effects compared with podophyllotoxin.Surprisingly, etoposide did not affect tubulin polymerization butinhibited another very important target in oncology: DNA topoi-somerase II. This story illustrates well the importance in drugdiscovery of serendipity, which was likened to “looking for aneedle in a haystack and discovering the farmer’s daughter” byProfessor Pierre Potier, inventor of the anticancer drug taxotere(Zard, 2012 ).
Another non-rational issue regarding the SAR of derivatives of
natural compounds concerns the relationship between the chem-ical structure of a drug and its therapeutic indication. Indeed,transforming the structure of a drug may modify the nature ofthe targeted cancer. This is well established for vinca alkaloids for
instance ( Ta b l e 2 ). If we could understand the influence of the
molecular structure of a drug with its optimal therapeutic indi-cation, then we might be able to adapt known medicines to treatcancers that are reluctant to current therapies.
In spite of the major achievements in systems biology and
translational medicine over the last decade, there is still, at best,a presumptive relationship between the efficacy of a drug inpreclinical assays and the likelihood of its value in clinic.
RAPALOGUES: TEMSIROLIMUS®AND EVEROLIMUS®
In 1975, researchers at Ayerst Laboratories (Canada) reported theisolation of rapamycin as a secondary metabolite from a strain ofStreptomyces hygroscopicus based on its antifungal activity ( Sehgal
et al., 1975; Vezina et al., 1975 ). Its name comes from Rapa
Nui (Easter Island) where its producer strain had been collectedfrom a soil sample. Its richly adorned macrocyclic structure wasfully elucidated a few years later ( Swindells et al., 1978; Findlay
and Radics, 1980; McAlpine et al., 1991 ). Rapamycin did not
attract so much attention until the discovery in 1987 of the struc-turally related immunosuppressant FK506 ( Kino et al., 1987a,b ).
Rapamycin was eventually developed without further structuralmodifications as the oral immunosuppressant drug sirolimus. Itwas approved for prevention of rejection in organ transplantationin 1999 ( Calne et al., 1989; Kahan et al., 1991; Watson et al., 1999;
Calne, 2003 ).
Determining the mode of action of rapamycin unraveled one
of the most important signaling pathways in cell biology, whichillustrates another important asp ect of the pharmacology of nat-
ural products. Indeed a common caveat of developing an originalnatural product toward clinical application is the requirement toidentify its molecular target and understand its mode of action(Krysiak and Breinbauer, 2012 ). However, when the target is
identified, it may lead to major breakthroughs in cell biology(Pucheault, 2008 ). Gratefully, current technologies render this
task increasingly easier ( Ares et al., 2013 ).
In 1991, Michael Hall et al. identified the molecular target
of rapamycin in a gene complementation assay in yeast andnamed it TOR for “Target Of Rapamycin” ( Hietman et al., 1991 ).
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry May 2014 | Volume 2 | Article 20 |2
Basmadjian et al. Anticancer natural products
T able 1 | Novel anticancer medicines based on natural products.
Name (trade name), structure Y ear of approval, company Therapeutic indication, mode of action
Temsirolimus (Torisel®): R=R1
Everolimus (Afinitor®), R=R22007 , Wyeth Treatment of renal cell carcinoma (RCC), inhibition of mTOR
2009, Novartis Treatment of advanced kidney cancer, inhibition of mTOR
Ixabepilone (Ixempra®) 2007 , Bristol-Myers Squibb Treatment of aggressive metastatic or locally advanced breast cancer no
longer responding to currently available chemotherapies, stabilization ofmicrotubules
Vinflunine (Javlor®) 2009, Pierre Fabre Treatment of bladder cancer, inhibition of tubulin polymerization
Romidepsin (Istodax®) 2009, Celgene Treatment of cutaneous T -cell lymphoma (CTCL), inhibition of the
isoforms 1 and 2 of histone deacetylases
Trabectedin =ecteinascidin 7 43 (Y ondelis®) 2009, Zeltia and Johnson
and JohnsonTreatment of advanced soft tissue sarcoma and ovarian cancer, induction
of DNA damage
(Continued)
www.frontiersin.org May 2014 | Volume 2 | Article 20 |3
Basmadjian et al. Anticancer natural products
T able 1 | Continued
Name (trade name), structure Y ear of approval, company Therapeutic indication, mode of action
Cabazitaxel (Jevtana®) 2010, Sanofi-Aventis Treatment of hormone-refractory metastatic prostate cancer,
microtubule stabilization
Abiraterone acetate (Zytiga®) 2011, Janssen Treatment of castration-resistant prostate cancer, inhibition of 17
α-hydroxylase/C17 , 20 lyase (CYP17A1)
Eribulin mesylate (Halaven®) 2011, Eisai Co. Treatment of metastatic breast cancer, inhibition of microtubule
dynamics
Homoharringtonine, Omacetaxinemepesuccinate (Synribo
®)2012, Teva Chronic myelogenous leukemia (CML), inhibition of protein synthesis
Carfilzomib (Kyprolis®) 2012, Onyx Treatment of multiple myeloma, inhibition of proteasome
Ingenol mebutate (Picato®) 2012, LEO Pharma Actinic keratosis, activation of PKC δ
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry May 2014 | Volume 2 | Article 20 |4
Basmadjian et al. Anticancer natural products
Three years later, Stuart Shreiber et al. identified its mammalian
homolog referred to today as the kinase mTOR (mammalianTOR) ( Brown et al., 1994 ). The mode of action of rapamycin is
unique: it binds to two proteins at the same time, mTOR andthe immunophilin FKBP-12, to form a ternary complex devoidof any kinase activity. mTOR plays a central role integratingsignals from growth factors, nutrients, stress, and hormones toregulate metabolism, proliferation, cell growth, and apoptosis.However, the exact mechanisms of action of rapamycin deriva-tives, called rapalogues, remain only moderately understood.Some recent evidence indicates that rapalogues may primarilydisplay their anticancer effects through an inhibition of angio-genesis in patients ( Faivre and Raymond, 2008 ). This hypothesis
would explain why rapologues are particularly effective in hyper-vascularized tumors.
Currently, two rapalogues, temsirolimus, and everolimus, have
been developed for the treatment of renal, breast, and pancreascancers, astrocytoma, and mantle cell lymphoma. These drugs
FIGURE 1 | Structures of podophyllotoxin, etoposide, and teniposide.differ in their formulation, application, and dosing schemes,thereby yielding varying bioavailabilities. They are all prepared bysemi-synthesis.
IXABEPILONE (IXEMPRA®)
Drugs that target microtubules, such as taxoids and vinca alka-
loids, continue to represent an important class of chemotherapeu-tic agents ( Jordan and Wilson, 2004 ). Over the last two decades
other classes of naturally occurring nontaxoid compounds, theepothilones ( Gerth et al., 1996; Höfle et al., 1996 ), discoder-
molides ( Gunasekera et al., 1990 ), eleutherobins ( Lindel et al.,
1997 ), and laulimalides ( Mooberry et al., 1999 ) that stabilize
microtubule assemblies similarly to taxol, have been identified(Figure 2 ). Based upon extensive structure-activity data, a com-
mon pharmacophore for these different classes of compounds hasbeen proposed ( Ojima et al., 1999 ).
Not only is epothilone B more cytotoxic than taxol, but
it is also much less sensitive toward the development ofmultidrug-resistance, a major concern in the clinic ( Horwitz,
1994; Bollag et al., 1995; Kirikae et al., 1996 ). This impres-
sive pharmacological profile coupled with the challenge of itstotal synthesis has attracted the attention of some of the mostwell-known organic chemists in the world, including SamuelDanishefsky ( Balog et al., 1996; Su et al., 1997 ), followed by
Nicolaou ( Nicolaou et al., 1997; Y ang et al., 1997 ), Schinzer
(Schinzer et al., 1997 ), Mulzer ( Mulzer et al., 2000 )a n dC a r r e i r a
(Bode and Carreira, 2001 ).
Early investigations indicated that natural epothilones dis-
play poor metabolic stability and unfavorable pharmacokineticproperties ( Lee et al., 2000 ). Several synthetic and semi-synthetic
analogs were then examined and evaluated in preclinical stud-ies. Eventually, isosteric replacement of the lactone by a lac-tam afforded ixabepilone (also known as azaepothilone B) ( Lee
et al., 2008 ). Not only is this drug not susceptible to hydroly-
sis by esterases, conferring metabolic stability, but it also displaysimproved water solubility, which greatly alleviate galenic prob-lems associated with hypersensitivity reaction in patients.
T able 2 | Structures and therapeutic indications of vinca alkaloids.
Name n QR1R2R3R4R5Therapeutic indication
Vinblastine 2 OH H Et OAc Me OMe Lymphomas, germ cell tumors, breast, head and neck cancer and testicular
cancers
Vinorelbine 1 Q =R1=∅(alkene) Et OAc Me OMe Osteosarcoma, breast, and non-small cell lung cancers
Vincristine 2 OH H Et OAc CHO OMe Acute lymphoblastic leukemia, rhabdomyosarcoma, neuroblastoma,
lymphomas, and nephroblastoma
Vindesine 2 OH H Et OH Me NH 2 Melanoma, lung, breast and uterine cancers, leukemia and lymphoma
Vinflunine 1 H H CF 2Me OAc Me OMe Bladder cancer
www.frontiersin.org May 2014 | Volume 2 | Article 20 |5
Basmadjian et al. Anticancer natural products
FIGURE 2 | Chemical structures of representative natural compounds that stabilize microtubule assemblies.
In 2007, the FDA (but not its European equivalent, European
Medicines Agency or EMA) approved ixabepilone for the treat-ment of aggressive metastatic or locally advanced breast cancerno longer responding to currently available chemotherapies.
VINFLUNINE (JAVLOR®)
Vinca alkaloids were the first chemotherapeutic agents that tar-
get microtubules. The first member of this class, vinblastine, wasisolated in 1958 ( Noble et al., 1959 ). Latter, some derivatives, vin-
cristine, vinorelbine, and finally avelbine, were developed to treathematological and solid malignancies in both adult and pediatricpatients ( Ta b l e 1 ).Vinca alkaloids block the polymerization of
tubulin molecules into microtubules to prevent the formation ofthe mitotic spindle.In the course of their study on the reactivity of functionalized
molecules in superacid media, Jacquesy and collaborators foundthat the treatment of vinorelbine with a combination of HF andSbF
5gave a difluoro analog, later called vinflunine ( Scheme 1 )
(Fahy et al., 1997 ). Importantly, this new compound displayed
an enhanced bioavailability compared to other vinca alka-
loids. Indeed, its terminal half-life was calculated to be about40 h and the terminal half-life for its active metabolite (4- O-
deacetylvinflunine) was reported to be 4–6 days in several phase Itrials ( Bennouna et al., 2003, 2005; Johnson et al., 2006 ).
Resistance to vinflunine develops more slowly than with other
vinca alkaloids. In addition, vinflunine in vitro neurotoxicity is
lower than that of vincristine or vinorelbine ( Etiévant et al., 1998,
2001; Estève et al., 2006 ). Further development by Pierre Fabre
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry May 2014 | Volume 2 | Article 20 |6
Basmadjian et al. Anticancer natural products
SCHEME 1 | Synthesis of vinflunine from vinorelbine ( Fahy et al., 1997 ).
and Bristol Myers Squibb laboratories ended with the approval of
vinflunine for the treatment of bladder cancer by the EuropeanMedicines Agency (EMA) in 2009.
ROMIDEPSIN (ISTODAX®)
The cyclic depsi-pentapeptide romidepsin, also called FR901228,FK228, or NSC 630176, was isolated and identified by Ueda andcolleagues at Fujisawa Pharmaceutical in Japan through a screen-ing program of fermentation products able to revert the trans-formed morphology of a Ha-ras NIH3T3 cells to normal ( Ueda
et al., 1994 ). Indeed Ha-ras is an oncogene involved in tumorigen-
esis and consequently represents an important target in oncology.Importantly, romidespsin displayed potent antitumor activitiesagainst A549 and MCF-7 tumors in xenografted mice. Theseresults attracted the attention of NCI scientists who continuedto explore its anticancer properties under a Cooperative Researchand Development Agreement with Fujisawa Corporation (nowAstellas).
When romidepsin was discovered, histone deacetylases
(HDAC) were emerging as important targets for the treatmentof cancer ( Thaler and Mercurio, 2014 ). Screening of micro-
bial metabolites for their effects on transcription showed thatromidepsin behaves similarly to trichostatin A, a known HDACinhibitor ( Nakajima et al., 1998 ). Romidepsin acts as a prodrug,
which is reduced in cells to its active form by glutathione, yield-ing a monocyclic dithiol that preferentially inhibits the isoformsHDAC1 and HDAC2 ( Furumai et al., 2002 ).
In 2002, when it became established that romidepsin
holds a promising therapeutic potential, Fujisawa Corporationbegan clinical trials. Romidepsin was licensed to GloucesterPharmaceuticals in 2004 (latter acquired by Celgene Co)and was approved by the FDA in 2009 for the treat-
ment of cutaneous T-cell lymphoma. The preclinical and clin-ical development has been described in an excellent review(Vandermolen et al., 2011 ).
ECTEINASCIDIN 743 =TRABECTEDIN (YONDELIS®)
In 1969, unidentified alkaloids from the Caribbean tunicate
Ecteinascidia turbinate were shown to display some anticancer
activities, but the structure of these complex alkaloids could notbe determined because of their natural scarcity and the limita-tion of analytical chemistry at that time ( Sigel et al., 1970 ). In
1990, Rinehart et al. from the University of Illinois at Urbana-Champaign elucidated the structure and reported the cytotoxi-city of these tetrahydroisoquinoline alkaloids, the ecteinascidins(Rinehart et al., 1990 ). These compounds displayed greater in
vitro and in vivo antitumor activity than those reported for
the structurally related microbial metabolites saframycins andsafracins.
Ecteinascidin 743, also called trabectedin and ET-743, was
then selected for preclinical development based on its exceptionalin vitro cytotoxicity. Pommier et al. at NCI demonstrated that this
drug binds in the minor groove of DNA and alkylates the exo-cyclic amino group at position 2 of guanine in GC-rich regions(Scheme 2 )(Pommier et al., 1996 ).
Ecteinascidin 743 was shown to block the DNA excision repair
system ( Takebayashi et al., 2001; Zewail-Foote et al., 2001 ), to
cross-link DNA with topoisomerase I ( Martinez et al., 1999;
Takebayashi et al., 1999; Zewail-Foote and Hurley, 1999 ), and
also to inhibit the binding of DNA to some transcription fac-tors ( Bonfanti et al., 1999; Jin et al., 2000; Minuzzo et al.,
2000 ). However, the cascade of events that links DNA damage
www.frontiersin.org May 2014 | Volume 2 | Article 20 |7
Basmadjian et al. Anticancer natural products
SCHEME 2 | DNA Alkylation by ecteinascidin 743.
to the resulting antitumor activity is far from being understood
(D’Incalci and Galmarini, 2010 ).
When ecteinascidin 743 was licensed to PharmaMar; this com-
pany launched a very challenging program of aquaculture toproduce sufficient quantities of tunicate biomass to feed clinicaltrials program. After several years of intensive development, thecumulative total biomass reached some 250 metric tons. However,isolation of ecteinascidin 743 required complex and costly stepsof purification with final yields less than 1 g/ton ( Cuevas and
Francesch, 2009 ). Several total syntheses have been reported, but
they cannot be translated into industrial production ( Corey et al.,
1996; Endo et al., 2002; Chen et al., 2006; Zheng et al., 2006;Fishlock and Williams, 2008; Imai et al., 2012; Kawagishi et al.,2013 ). Eventually, this supply problem was solved by use of a
complex semi-synthesis from cyanosafracin B, which is avail-able in kilogram quantities by fermentation ( Cuevas et al., 2000;
Menchaca et al., 2003 ).
Preclinical studies did not reveal that soft tissue sarcoma is
more sensitive to ecteinascidin 743 than other solid tumors.This response was unveiled first during phase I clinical trialsand confirmed in phase II ( Taamma et al., 2001; Villalona-
Calero et al., 2002; D’Incalci and Jimeno, 2003 ). This drug was
approved under the name of Y ondelis in 2007 in the EuropeanUnion for the treatment of patients with advanced soft tis-sue sarcoma. This compound was the first anticancer medicineof marine origin to be approved. It was followed by eribu-lin (vide infra), validating the concept that marine naturalproducts should be considered important contenders in drugdiscovery.
CABAZITAXEL (JEVTANA®)
The taxane anticancer drug cabazitaxel is a semi-synthetic deriva-tive of the natural taxoid 10-deacetylbaccatin III. It was approvedin 2010 by the FDA, in combination with prednisone, forthe treatment of patients with hormone-refractory metastaticprostate cancer who had already been administered a treatmentcontaining the taxane docetaxel ( Galsky et al., 2010 ). In 2013,
Vrignaud et al. showed that in vitro , cabazitaxel stabilized micro-
tubules as effectively as docetaxel but was also 10 times morepotent than docetaxel in chemotherapy-resistant tumor cells.They also noted that cabazitaxel was active in docetaxal-resistanttumors ( Vrignaud et al., 2013 ). In addition, cabazitaxel pene-
trates the blood-brain barrier. Cabazitaxel was approved 20 yearsafter taxol, illustrating that there is still room to improve wellestablished anticancer medicines.
ABIRATERONE ACETATE (ZYTIGA®)
Abiraterone acetate is an oral inhibitor of androgen synthesis usedsince 2011 for the treatment of castration-resistant prostate can-cer. Previous treatments of prostatic cancers prevented androgenproduction by the testes, but not by the adrenals. Abirateroneacetate is rapidly hydrolyzed in vivo to abiraterone, which is a
selective, irreversible inhibitor of cytochrome P450 17 α(CYP17),
an enzyme that catalyzes the conversion of pregnenolone andprogesterone into DHEA or androstenedione, two precursors oftestosterone. This drug was originally designed and synthesizedby Jarman et al. at the Institute of Cancer Research in Sutton (UK)based on the hypothesis that the nitrogen lone pair of a pyridylmoiety linked to the steroid skeleton would coordinate with theiron atom of the heme cofactor in the active site of CYP17 ( Potter
et al., 1995; Jarman et al., 1998 ).
The inhibition of CYP17 by abiraterone acetate blocks andro-
gen biosynthesis and significantly improves the therapy ofcastration-resistant prostate cancer, which remains a challenge totreat ( Rehman and Rosenberg, 2012 ). Unfortunately, this CYP17
inhibition also decreases glucocorticoid and increases mineralo-corticoid production, which results in the main source of adverseeffects.
Since the invention of abiraterone, different steroids bear-
ing a heteroaromatic substituent on the D ring continued to bedeveloped as CYP17 inhibitors. Among those, galeterone (TOK-001 or VN/124-1) recently entered clinical trials for the treat-ment of chemotherapy-naive, castration-resistant prostate cancer(Figure 3 )(Vasaitis and Njar, 2010 ). Interestingly, this drug not
only inhibits CYP17, but is also an androgen receptor antagonist(Handratta et al., 2005 ).
ERIBULIN MESYLATE (HALAVEN®)
In1985 ,Uemura et al. isolated and identified norhalichon-
drin A from the marine sponge Halichondria okadai based
on its potent in vitro toxicity ( Uemura et al., 1985 ). Related
polyether macrolides, including halichondrin B ( Hirata and
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry May 2014 | Volume 2 | Article 20 |8
Basmadjian et al. Anticancer natural products
FIGURE 3 | Structure of galeterone.
Uemura, 1986 ) were identified in the following years ( Qi and
Ma, 2011 ). T ests with NCI’s 60-cell line screen suggested that
halichondrin B affects tubulin polymerization. Further studiesshowed that this drug displays subtle differences in mechanism ofaction from those of other known antimitotics targeting tubulin.Although halichondrin B displayed promising activity, its preclin-ical investigation has been hampered by its scarcity from naturalsources.
Due to its complexity, the total synthesis of halichondrin
B was considered as an attractive objective by Kishi et al. atHarvard University. This team achieved this formidable chal-lenge in 1992 ( Aicher et al., 1992 ). Further collaborative studies
from this group and Eisai Co. ultimately led to the develop-ment of the simplified and pharmaceutically improved analogeribulin ( Jackson et al., 2009 ). Although it is less complex than
natural halichondrins, eribulin contains 19 stereogenic centers,two exocyclic olefins, seven polyoxygenated pyrans and tetrahy-drofurans, a 22-membered macrolactone ring, and a 36 carbonbackbone. With its 35 steps, eribulin synthesis extended thelimit of feasibility for industrial production. Indeed, eribulin isthe single most complex molecule synthesized at an industrialscale and represents an awe-inspiring testimony to the currentpower of organic synthesis. Eribulin was approved by FDA in2010 to treat patients with metastatic breast cancer who havereceived at least two prior chemotherapy regimens for late-stagedisease.
HOMOHARRINGTONINE =OMACETAXINE MEPESUCCINATE
(CEFLATONIN®)
T oxic seeds of the conifer Cephalotaxus harringtonia K. Koch
varharringtonia belong to the traditional Chinese pharmacopeia.
In observance with Mao Tse-tung’s judgment that Chinesemedicine and pharmacology represent a national treasure thatneeds to be valorized, Chinese investigators established that thetotal alkaloids from C ephalotaxus fortunei Hook.f possesses anti-
tumor activity in preliminary clinical trials ( Group, 1976 ). In the
same period, National Cancer Institute (NCI) scientists foundthat Cephalotaxus harringtonia seed extracts displayed signifi-
cant in vivo activity against L-1210 and P-388 leukemia tumors
in mice. Powell et al. from the U.S. Department of Agricultureisolated and identified the structure of cytotoxic Cephalotaxus
alkaloids: harringtonine, isoharringtonine, homoharringtonine,a n dd e o x y h a r r i n g t o n i n e( Powell et al., 1970 )(Figure 4 ). These
compounds are esters of cephalotaxine, an inactive alkaloid firstisolated by Paudler et al. in 1963 at Ohio University ( Paudler et al.,
1963 ). Homoharringtonine was found to be the most effective inFIGURE 4 | Structures of cytotoxic Cephalotaxus alkaloids.
prolonging survival of P388 leukemic mice ( Powell et al., 1972 ).
Clinical trials performed in China demonstrated the efficacy ofthis agent against acute myeloid leukemia (AML), myelodys-plastic syndrome (MDS), acute promyelocytic leukemia (APL),polycythemia vera, and central nervous system (CNS) leukemia(Kantarjian et al., 2013 ).
Homoharringtonine inhibits protein synthesis ( Huang, 1975 ).
More specifically, it blocks the aminoacyl-tRNA binding to freeribosomes and monosomes, but not to polyribosomes ( Fresno
et al., 1977 ). Tang et al. demonstrated that decreased expression
of the antiapoptotic factor myeloid cell leukemia-1 (Mcl-1) is akey event in this antileukemic mechanism of action ( Tang et al.,
2006 ).
In 1998, a T exan biotech company developed the semisynthetic
form of homoharringtonine, designated “omacetaxine mepesuc-cinate” (Synribo®), and provided a reliable source supply forclinical investigations by ChemGenex and the M.D. AndersonCancer Center ( Robin et al., 1999 ).
This preparation of homoharringtonine [Omacetaxine mepe-
succinate (Synribo®)] has been granted orphan drug status inEurope and the U.S. to treat chronic myelogenous leukemia(CML). It was approved by the US FDA in October 2012 forthe treatment of adult patients with CML after failure of twoor more tyrosine kinase inhibitors (for a review on its clini-cal development, see Kantarjian et al., 2013 ). It is interesting
to note that these approvals occurred more than 40 years afterthe initial discovery of this compound. Even though omacetax-ine has a narrow indication in the U.S. and Europe, it hasbeen part of standard acute myeloid leukemia (AML) ther-apy in China, which begs for extending its use for additionalindications.
CARFILZOMIB (KYPROLIS®)
In 1992, Bristol-Myers Squibb scientists from T okyo reported thestructure of epoxomicin, a microbial tetrapeptide appended withan electrophilic epoxy ketone group. This compound displayedpotent in vivo antitumor activity against murine B16 melanoma
tumors. However, because the mechanism of action could notbe established, its investigation was abandoned, thereby lead-ing to the publication of the initial discovery. Eventually, BMSclosed the research center in T okyo. It was a common practice
www.frontiersin.org May 2014 | Volume 2 | Article 20 |9
Basmadjian et al. Anticancer natural products
during that period for big pharmaceutical companies to close
their departments of natural product chemistry.
In 1999, the potent anticancer activity of epoxomicin attracted
the attention of Craig Crews at Y ale University, who designedthe first synthesis of epoxomicin. In the course of this endeavor,he established the absolute configuration of the epoxide stere-ocenter and synthesized also a biotinylated probe, which wasused to identify the proteasome as the molecular target of epox-omicin. The proteasome is a multiprotein complex that degradesunneeded or damaged proteins by proteolysis. Importantly,epoxomicin does not display any cross-inhibition with pro-teases, which is a major problem encountered with other anti-cancer proteasome inhibitors, such as bortezomib (Velcade®).The source of this selectivity was elucidated by a crystallo-graphic approach ( Groll et al., 2000 ). The crystal structure
of the proteasome bound to epoxomicin revealed the for-mation of a morpholino ring between the amino terminalthreonine of the proteasome and the electrophilic moiety ofepoxomicin, probably through the mechanism displayed inScheme 3 .
The specificity of epoxomicin toward proteasome prompted
Crews to associate with Caltech professor Raymond Deshaies toestablish a start-up company, Proteolix, dedicated to the develop-ment of a clinical candidate. During this process, they identifiedYU-101, which had better inhibitory activity than bortezomib(Figure 5 ). Addition of a morpholine moiety to YU-101 improved
its solubility, thereby creating carfilzomib, which rapidly enteredPhase I and II clinical trials. Importantly, the peripheral neu-ropathy that was observed with bortezomib did not occur withcarfilzomib. In 2009, Onyx Pharmaceuticals acquired Proteolixand this compound was approved for the treatment of multiplemyeloma in 2012.
INGENOL MEBUTATE (PICATO®)
Phorbol diesters, such as 12- O-tetradecanoylphorbol-13-acetate
(TPA), rank among the most potent tumor promoters iden-tified so far ( Figure 6 )(Nishizuka, 1984 ). These compounds
induce tumor formation by activating protein kinase C (PKC).Interestingly, a natural compound extracted from Euphorbia
peplus plants, Ingenol mebutate, also activates PKC but with a dif-
ferent pharmacological profile. Indeed, this compound inducesthe death of precancerous skin lesions induced by sunlight, calledactinic keratosis.
The sap of Euphorbia peplus (known commonly as petty
spurge) is commonly used as an alternative therapy for skindiseases in Australia ( Weedon and Chick, 1976 ). In 1998, its effi-
cacy was established for the self-treatment of skin cancers andactinic keratosis ( Green and Beardmore, 1988 ).
Ingenol mebutate was first identified in 1980 by Evans et al.
from the National Research Center in Cairo (Egypt) ( Sayed et al.,
1980 ). These authors demonstrated also that this compound
is cytotoxic to cancer cells. For more than 20 years, ingenolmebutate remained poorly investigated, until 2004, when thelab of Peter Blumberg at NCI showed that it activates PKC iso-forms, but with a different pharmacological profile than thatof TPA. Importantly, the activation of protein kinase C delta(PKC δ) was shown to promote the production and release of
inflammatory cytokines contributing to the elimination of actinickeratosis.
At the same time, Eric Raymond in Clichy (France) showed
that ingenol mebutate-induced activation of PKC δand reduced
e x p r e s s i o no fP K C αlead to an activation of Ras/Raf/MAPK, an
inhibition of the phosphatidylinositol 3-kinase/AKT signalingpathways, and ultimately to apoptosis of cancer cells ( Benhadji
et al., 2008; Serova et al., 2008; Ghoul et al., 2009 ).
After few years of preclinical investigations, ingenol mebu-
tate entered clinical trials ( Siller et al., 2009 ) and was eventually
approved by FDA and EMA in 2012 for the topical treatment ofactinic keratosis. This compound is produced by extraction fromthe petty spurge plant in low yield (1 g of pure compound/800 kgof plant). T o improve the production of this molecule, JakobFelding of LEO pharma associated with Phil Baran from ScrippsInstitute to develop an elegant synthesis of ingenol in only 14steps from inexpensive ( +)-3-carene ( Jørgensen et al., 2013 ). This
synthesis has been rapidly scaled-up to kilogram levels ( Ritter,
2013 ).
CONJUGATION OF NATURAL PRODUCTS TO ANTIBODIES OR
FOLIC ACID TO TARGET TUMORS
At the end of 19th century, Paul Ehrlich already considered
the conjugation of a toxin to a compound that selectively tar-gets a disease-causing organism to generate a “magic bullet”(“magische kugel ”) that would destroy the origin of the disease
without toxicity to healthy tissues in the body ( Ehrlich, 1897 ).
About 60 years later, Mathé et al. conjugated anti-tumor anti-bodies to the folic acid antagonist, methotrexate ( Loc et al.,
1958 ). Although the experiments in mice were encouraging,
this approach did not attract interest in the scientific commu-nity and returned to limbo for two decades, until 1975, whenGhose et al. demonstrated the efficacy of an anticancer alkylating
SCHEME 3 | Proposed mechanism of alkylation of the proteasome by epoxomicin.
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry M a y2 0 1 4|V o l u m e2|A r t i c l e2 0 |10
Basmadjian et al. Anticancer natural products
FIGURE 5 | Structures of epoxomicin, YU-101, carfilzomib.
FIGURE 6 | Structures of 12- O-tetradecanoylphorbol-13-acetate and ingenol mebutate.
agent, chloranbucil, conjugated to an antibody against a mouse
lymphoma ( Ghose et al., 1975 ). The advent of monoclonal anti-
bodies the same year definitely boosted this field of research(Kohler and Milstein, 1975 ). Since then, almost every cyto-
toxic agent has been conjugated to antibodies following variousstrategies.
After two decades of endeavor, low cytotoxicity, and lack of
specificity of antibodies for their targeted antigens, conjugateinstability, immunogenicity, and heterogeneous product charac-teristics were identified as important sources of failure in theclinic ( Scott et al., 2012; Ho and Chien, 2014 ). However, a sig-
nificant step forward was made with the use of extremely highlytoxic agents such as calicheamicin, maytansine, or auristatin(Figure 7 ). These drugs are so toxic that they cannot be used by
themselves without a targeting agent.
In 2000, four decades after Mathé’s pioneering work and one
century after Ehrlich’s dream, Wyeth received approval to com-mercialize Gemtuzumab ozogamicin (Mylotarg®) which resultsfrom the conjugation of a monoclonal antibody targeting CD33with a calicheamicin derivative. This drug was used for 10years against acute myelogenous leukemia, before being with-drawn in 2010, when it was demonstrated that it does notprovide any significant benefit over conventional cancer ther-apies. In 2011 and 2013, two other immunoconjugates weremarketed: brentuximab vedotin (Adcetris®) and trastuzumabemtansine (Kadcyla®). The first one targets the protein CD30,which is expressed in classical Hodgkin lymphoma and sys-temic anaplastic large cell lymphoma. This antibody is conjugated
to a fully synthetic analog of the antimitotic agent dolastatin
(Figure 7 ).Trastuzumab emtansine results of the conjugation of a mono-
clonal antibody targeting the receptor HER2 (a receptor tyrosine-kinase erbB-2), which is overexpressed mainly in some forms ofbreast and gastric cancers to the highly cytotoxic natural prod-uct maytansine. The development of this class of agents requiresa careful optimization of the monoclonal antibody, the cytotoxicpayload, and the chemical linker ( Ducry, 2012 ). The successful
introduction of immunoconjugates has validated this approachto treat cancers, and currently as many as 415 antibody–drugconjugates are under clinical evaluation.
In addition to antibodies, alternative tumor-selective ligands
have been conjugated to anticancer drugs. Based on observa-tions that cells internalize vitamins, such as folate, by receptor-mediated endocytosis, Leamon, and Low from Purdue Universitydemonstrated in 1991, that macromolecules conjugated to folicacid could be delivered into living cells ( Leamon and Low,
1991 ). Following this seminal observation, hundreds of publi-
cations have improved upon this approach, which is currentlybeing examined in clinical trials. The efficacy of this tech-nology lies on the overexpression of the folate receptor intumors, while it is quasi-absent in normal tissues. Very impor-tantly also, folic acid retains a high affinity to its receptorwhen it is conjugated via its γ-carboxyl ( Vlahov and Leamon,
2012 ).
Early attempts were limited by the release properties of the
conjugates. After two decades of intensive research, some guidingr u l e sw e r ei d e n t i fi e dt ol e a dc o m p o u n d st o w a r dc l i n i c s :
1. anticancer agents must display a high cytotoxicity (similar to
immunoconjugates);
www.frontiersin.org M a y2 0 1 4|V o l u m e2|A r t i c l e2 0 |11
Basmadjian et al. Anticancer natural products
FIGURE 7 | Structures of marketed immunoconjugates.
2. enhanced hydrophilicity, to prevent passive diffusion into nor-
mal tissue;
3. an efficient cleavable linker system that releases the anticancer
drug at a reliable rate once inside the targeted cell;
4. a low molecular weight, to optimize the penetration into solid
tumor tissue with concomitant rapid systemic clearance.
Following these guidelines, five folic acid conjugates have reached
clinical trials, including the most advanced one, vintafolide(EC145), which is currently in a phase 3 trial in women with
cisplatin -resistant ovarian cancer.
In vintafolide, the highly cytotoxic vinblastine is con-
nected to the folate moiety trough a self-immolative linkerand a peptidic spacer ( Figure 8 ). T o provide the desired
hydrophilicity to the final drug-conjugate and prevent unspe-cific internalization, acidic, and basic amino acids such as
aspartic acid and arginine were introduced in the peptide-based
unit.
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry May 2014 | Volume 2 | Article 20 |12
Basmadjian et al. Anticancer natural products
FIGURE 8 | Structure of vintafolide and mechanism of release of the payload in the endosome.
The self-destructive linker system is based on a 1,2-elimination
mechanism by reduction of the disulfide bond between the cys-teine of the spacer and the linker, which occurs in the endosomethrough a not fully understood mechanism ( Figure 8 )(Y ang et al.,
2006 ).
TRADITIONAL HERBAL REMEDIES
In addition to purified molecules, traditional herbal remedies are
slowly emerging in modern Western medicine ( Basu, 2004 ). An
injectible form of an extract of the Chinese medicinal plant Semen
coicis called Kanglaite® (Kang-Lai-T e) has been used in China as
a lipid emulsion since the end of the 90’s for the treatment ofnon-small cell lung, liver, stomach, and breast cancers. It has beenmarketed also in the Russian Federation since 2003 and is thefirst traditional Chinese herbal remedy to enter into clinical tri-als in the US. As with many other traditional Chinese medicines,Kanglaite activity probably results from the combined actions ofmultiple pharmacologically active ingredients that have not beenyet identified ( Xu, 2011 ). Over the last decade, other botanical
drugs have entered clinical trials in the West to treat cancers orother ailments.
NANOPARTICLE DELIVERY OF ANTICANCER DRUGS
Tumor growth requires angiogenesis, i.e., the formation of newblood vessels. In contrast to normal angiogenesis, newly formedvessels in tumors display many structural and functional defects,which permit the leakage of macromolecules. This feature isreferred to as the “enhanced permeability and retention (EPR)effect.” Recent advances in the application of nanotechnology tomedicine enabled the approval of five nanoparticle chemother-apeutics for cancer ( Wang et al., 2012 ). Four liposomal for-
mulations have been approved for clinical use in oncology:pegylated liposomal doxorubicin (DOXIL®, Caelyx®), nonpegy-lated liposomal doxorubicin (Myocet®), and liposomal cytarabine(DepoCyte®) ( Hofheinz et al., 2005 ). Nab-paclitaxel (Abraxane®)
is an albumin bound approved for the treatment of breast can-cer and is undergoing clinical trials for other clinical indications.And finally, Genexol-PM is a polymeric micelle formulation ofpaclitaxel composed of block copolymers of PEG and poly-(D,L-
lactic acid) ( Kim et al., 2004 ).
Although nanomedicine is a new discipline, its translation
into clinics has been rapid. A novel generation of nanoparticlechemotherapeutics is under development and expected to greatlyimprove cancer treatments. These new formulations may alsooffer novel opportunities for established anticancer drugs ( Wang
et al., 2012 ).
MISSED OPPORTUNITIES AND HOW TO RESCUE THEM
In 2010, Bristol-Myers Squibb stopped the phase III clinical trial
of Tanespimycin, an inhibitor of heat shock protein 90, for thetreatment of multiple myeloma, probably because of the expi-ration of the patent in 2014. In addition to drug developmentsthat were terminated because of the shortness of patent life, thereare many interesting drugs that did not reach clinical trial or thatfailed in clinical trial because the conceptual tools to correctly per-form these assays were not available at that time. Indeed, “thereare no bad anticancer agents, only bad clinical trial designs” asstated by Von Hoff (1998) .
Flavaglines, such as rocaglamide, represent a striking example
of natural products that are enjoying reinvigorated investigationafter their original discovery by King et al. from the NationalDefense Medical Center of Taiwan ( King et al., 1985 ). The recent
identification of their molecular targets, the scaffold proteins pro-hibitins and the initiation factor of translation eIF4A, coupledwith a description in Science about the origin of their selective
cytotoxicity in cancer cells should promote further investiga-tions to unveil their therapeutic usefulness ( Basmadjian et al.,
2013; Santagata et al., 2013 ). However, clinical trials with these
compounds are unlikely unless some structurally original andpatentable analogs are identified. Indeed, clinical trials of non-patentable compounds are still scarce ( Roin, 2009; Cvek, 2012 ).
For instance, a non-profit company, the Institute for OneWorldHealth, developed in 2007 paromomycin, which is not patentable,as an effective treatment for visceral leishmaniasis. This wasaccomplished with financial support from the Bill and MelindaGates Foundation, the Special Program for Research and Training
www.frontiersin.org M a y2 0 1 4|V o l u m e2|A r t i c l e2 0 |13
Basmadjian et al. Anticancer natural products
SCHEME 4 | Combination of biotechnology and organic synthesis for the synthesis of ansamitocin derivatives ( T aft et al., 2008 ).
in Tropical Diseases of the United Nations Development Program,
the World Bank, and the World Health Organization ( Sundar
et al., 2007 ). GlobalCures is another example of a non-profit
medical research organization, which aims to develop novel andcost-effective treatments for cancers ( Cvek, 2012 ). State agen-
cies, such as the National Center for Advancing Translational
Sciences are also deeply involved in the development of non-
profitable drugs. Only a radical change in public or internationalpolicy could support the further development of clinically usefulcompounds that are currently fated to be traded as generics.
BIOTECHNOLOGY-BASED GENERATION OF NOVEL NATURAL
PRODUCTS
Since the seminal synthesis of aspirin by Gerhardt (1853) ,a l l
the natural product derivatives were prepared by total synthesisor semi-synthesis. Alternate approaches are currently emergingbased on the progress in the deciphering of biosynthetic pathwaysand advances in biotechnologies. Currently, only a tiny fractionof microbes can be cultured with conventional approaches, yetuncultivated microorganisms represent an attractive source ofnovel natural products. It is now possible to isolate large frag-ments of microbial DNA directly from environmental samplesand to express them in an easily cultured microorganism. Thisapproach provides access to secondary metabolites that were orig-inally produced by inaccessible microorganisms. Additionally,the manipulation of these biosynthetic pathways can lead tonovel natural product derivatives. Metabolic engineering and syn-thetic biology are poised to revolutionize conventional chemicaland pharmaceutical manufacturing in the coming decade ( Ya d av
et al., 2012 ). Recently, methods and concepts of organic synthe-
sis have begun to be integrated to synthetic biology to generatenovel natural product derivatives. Such approaches that mergebiotechnology with organic synthesis are rapidly blooming andare expected to efficiently generate novel natural product analogsi nt h en e a rf u t u r e( Goss et al., 2012; Kirschning and Hahn, 2012 ).
A representative example of such an approach has been the useof an Actinosynnema pretiosum mutant that accepts 3-amino-4-
bromobenzoic acid as a substrate to prepare pharmacologicallyactive ansamitocin derivatives, which can then be transformed byclassical organic reactions ( Scheme 4 ;Taft et al., 2008 ).
CONCLUSION
The success of glivec and herceptin in the 90’s announced
the obsolescence of natural products in therapeutics. A decadelater, many cancer patients continue to die and pharmaceutical
companies have reconsidered their position on the potential ofnatural products in oncology. Indeed, for too many solid tumorsof advanced grades, the only therapeutic options remain exclu-sively palliative. There is therefore an urgent need to developoriginal medicines.
Some of newly developed agents induce a strong cytotox-
icity targeting conventional targets, DNA (for trabectedin) ormicrotubules (for ixabepilone, vinflunine, or eribulin), whileother target specific biochemical events such as steroid biosyn-thesis (abiraterone acetate), histone remodeling (for romidepsin),protein translation (homoharringtonine), or degradation (carfil-zomib). The case of rapamycin derivatives is atypical. These drugsare not cytotoxic, but can be considered as targeted therapy agentsdue to their inhibition of mTOR signaling.
In contrast with targeted therapeutics, which are designed for
a specific type of cancer, the development of natural productsis often more erratic and heavily relies on the skill of pharma-cologists to unravel their mechanism of action and clinicians toidentify the optimal indication in the clinic.
Over the last 15 years, natural products have been rehabil-
itated by pharmaceutical companies, even though some com-plementary approaches, such as molecular modeling based drugdesign are gaining in momentum. This latter methodology, whichwas pioneered by 2013 Nobel laureates, has successfully led toinnovative medicines. When it is possible to predict the 3Dstructure of proteins, then it will probably overshadow othermethods for identifying drug candidates. Until then, naturalproducts should continue to play a major role in drug dis-covery, especially in the treatment of cancers and infectiousdiseases.
ACKNOWLEDGMENTS
We are grateful to the “Association pour la Recherche sur leCancer” (ARC, grant numbers 3940 and SFI20111204054) forgenerous financial support. We also thank AAREC Filia Researchfor fellowships to Christine Basmadjian and Qian Zhao.
REFERENCES
Aicher, T. D., Buszek, K. R., Fang, F. G., Forsyth, C. J., Jung, S. H., Kishi, Y., et al.
(1992). T otal synthesis of halichondrin B and norhalichondrin B. J. Am. Chem.
Soc. 114, 3162–3164. doi: 10.1021/ja00034a086
Ares, J., Durán-Peña, M. J., Hernández-Galán, R., and Collado, I. (2013). Chemical
genetics strategies for identification of molecular targets. Phytochem. Rev. 12,
895–914. doi: 10.1007/s11101-013-9312-6
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry May 2014 | Volume 2 | Article 20 |14
Basmadjian et al. Anticancer natural products
Bailly, C. (2009). Ready for a comeback of natural products in oncology. Biochem.
Pharmacol. 77, 1447–1457. doi: 10.1016/j.bcp.2008.12.013
Balog, A., Meng, D., Kamenecka, T., Bertinato, P ., Su, D.-S., Sorensen, E. J., et al.
(1996). T otalsynthese von (-)-Epothilon A. Angew. Chem. 108, 2976–2978. doi:
10.1002/ange.19961082318
Basmadjian, C., Thuaud, F., Ribeiro, N., and Désaubry, L. (2013). Flavaglines:
potent anticancer drugs that target prohibitins and the helicase eIF4A. Future
Med. Chem. 5, 2185–2197. doi: 10.4155/fmc.13.177
Basu, P . (2004). Trading on traditional medicines. Nat. Biotechnol. 22, 263–265. doi:
10.1038/nbt0304-263
Benhadji, K. A., Serova, M., Ghoul, A., Cvitkovic, E., Le T ourneau, C., Ogbourne,
S. M., et al. (2008). Antiproliferative activity of PEP005, a novel ingenolangelate that modulates PKC functions, alone and in combination with cyto-
toxic agents in human colon cancer cells. Br. J. Cancer 99, 1808–1815. doi:
10.1038/sj.bjc.6604642
Bennouna, J., Campone, M., Delord, J. P ., and Pinel, M.-C. (2005). Vinflunine: a
novel antitubulin agent in solid malignancies. Expert Opin. Investig. Drugs 14,
1259–1267. doi: 10.1517/13543784.14.10.1259
Bennouna, J., Fumoleau, P ., Armand, J.-P ., Raymond, E., Campone, M., Delgado,
F.-M., et al. (2003). Phase I and pharmacokinetic study of the new
vinca alkaloid vinflunine administered as a 10-min infusion every 3 weeks
in patients with advanced solid tumours. Ann. Oncol. 14, 630–637. doi:
10.1093/annonc/mdg174
Bode, J. W., and Carreira, E. M. (2001). Stereoselective syntheses of epothilones
A and B via directed nitrile oxide cycloaddition1. J. Am. Chem. Soc. 123,
3611–3612. doi: 10.1021/ja0155635
Bollag, D. M., McQueney, P . A., Zhu, J., Hensens, O., Koupal, L., Liesch, J., et al.
(1995). Epothilones, a new class of microtubule-stabilizing agents with a taxol-
like mechanism of action. Cancer Res. 55, 2325–2333.
Bonfanti, M., La Valle, E., Fernandez Sousa Faro, J. M., Faircloth, G., Caretti,
G., Mantovani, R., et al. (1999). Effect of ecteinascidin-743 on the interaction
between DNA binding proteins and DNA. Anticancer Drug Des. 14, 179–186.
Brown, E. J., Albers, M. W., Tae Bum, S., Ichikawa, K., Keith, C. T., Lane, W. S., et al.
(1994). A mammalian protein targeted by G1-arresting rapamycin-receptor
complex. Nature 369, 756–758. doi: 10.1038/369756a0
Calne, R. Y. (2003). The development of immunosuppression: the rapamycin
milestone. Transplant. Proc. 35, S15–S17. doi: 10.1016/S0041-1345(03)00209-4
Calne, R. Y., Lim, S., Samaan, A., Collier, D. S. J., Pollard, S. G., White, D. J. G., et al.
(1989). Rapamycin for immunosuppression in organ allografting. The Lancet
334, 227. doi: 10.1016/S0140-6736(89)90417-0
Chen, J., Chen, X., Bois-Choussy, M. L., and Zhu, J. (2006). T otal synthesis of
ecteinascidin 743. J. Am. Chem. Soc. 128, 87–89. doi: 10.1021/ja0571794
Cordell, G. A., and Colvard, M. D. (2012). Natural products and tradi-
tional medicine: turning on a paradigm. J. Nat. Prod. 75, 514–525. doi:
10.1021/np200803m
Corey, E. J., Gin, D. Y., and Kania, R. S. (1996). Enantioselective total synthesis of
ecteinascidin 743. J. Am. Chem. Soc. 118, 9202–9203. doi: 10.1021/ja962480t
Cuevas, C., and Francesch, A. (2009). Development of Y ondelis[registered sign]
(trabectedin, ET-743). A semisynthetic process solves the supply problem. Nat.
Prod. Rep. 26, 322–337. doi: 10.1039/b808331m
Cuevas, C., Pérez, M., Martin, M. J., Chicharro, J. L., Fernandez-Rivas, C., Flores,
M., et al. (2000). Synthesis of ecteinascidin ET-743 and phthalascidin Pt-650
from cyanosafracin B. Org. Lett. 2, 2545–2548. doi: 10.1021/ol0062502
Cvek, B. (2012). Nonprofit drugs as the salvation of the world’s healthcare sys-
tems: the case of antabuse (disulfiram). Drug Discov. Today 17, 409–412. doi:
10.1016/j.drudis.2011.12.010
D’Incalci, M., and Jimeno, J. (2003). Preclinical and clinical results with the nat-
ural marine product ET-743. Expert Opin. Investig. Drugs 12, 1843–1853. doi:
10.1517/13543784.12.11.1843
D’Incalci, M., and Galmarini, C. M. (2010). A review of trabectedin (ET-
743): a unique mechanism of action. Mol. Cancer Ther. 9, 2157–2163. doi:
10.1158/1535-7163.mct-10-0263
Dan ˇcík, V ., Seiler, K. P ., Y oung, D. W., Schreiber, S. L., and Clemons, P . A. (2010).
Distinct biological network properties between the targets of natural products
and disease genes. J. Am. Chem. Soc. 132, 9259–9261. doi: 10.1021/ja102798t
Ducry, L. (2012). Challenges in the development and manufacturing of antibody-
drug conjugates. Methods Mol. Biol. 899, 489–497. doi: 10.1007/978-1-61779-
921-1_29
Ehrlich, P . (1897). Zur kenntnis der antitoxinwirkung. Fortschr. Med. 15, 41–43.Endo, A., Y anagisawa, A., Abe, M., T ohma, S., Kan, T., and Fukuyama, T. (2002).
T otal synthesis of ecteinascidin 743. J. Am. Chem. Soc. 124, 6552–6554. doi:
10.1021/ja026216d
Estève, M.-A., Carré, M., Bourgarel-Rey, V ., Kruczynski, A., Raspaglio, G., Ferlini,
C., et al. (2006). Bcl-2 down-regulation and tubulin subtype composition are
involved in resistance of ovarian cancer cells to vinflunine. Mol. Cancer Ther. 5,
2824–2833. doi: 10.1158/1535-7163.mct-06-0277
Etiévant, C., Barret, J.-M., Kruczynski, A., Perrin, D., and Hill, B. (1998).
Vinflunine (20/prime,20/prime-difluoro- 3/prime,4/prime-dihydrovinorelbine), a novel Vinca alka-
loid, which participates in P-glycoprotein (Pgp)-mediated multidrug resistance
in vivo and in vitro .Invest. New Drugs 16, 3–17. doi: 10.1023/A:10060228
11895
Etiévant, C., Kruczynski, A., Barret, J.-M., Tait, A., Kavallaris, M., and Hill, B.
(2001). Markedly diminished drug resistance-inducing properties of vinflunine
(20/prime,20/prime-difluoro-3/prime,4/prime-dihydrovinorelbine) relative to vinorelbine, identified in
murine and human tumour cells in vivo and in vitro .Cancer Chemother.
Pharmacol. 48, 62–70. doi: 10.1007/s002800100275
Evans, B. E., Rittle, K. E., Bock, M. G., Dipardo, R. M., Freidinger, R. M., Whitter, W.
L., et al. (1988). Methods for drug discovery: development of potent, selective,
orally effective cholecystokinin antagonists. J. Med. Chem. 31, 2235–2246. doi:
10.1021/jm00120a002
Fahy, J., Duflos, A., Ribet, J.-P ., Jacquesy, J.-C., Berrier, C., Jouannetaud, M.-P .,
et al. (1997). Vinca alkaloids in superacidic media: a method for creating a
new family of antitumor derivatives. J. Am. Chem. Soc. 119, 8576–8577. doi:
10.1021/ja971864w
Faivre, S., and Raymond, E. (2008). Mechanism of action of rapalogues: the
antiangiogenic hypothesis. Expert Opin. Investig. Drugs 17, 1619–1621. doi:
10.1517/13543784.17.11.1619
Ferlini, C., Cicchillitti, L., Raspaglio, G., Bartollino, S., Cimitan, S., Bertucci, C.,
et al. (2009). Paclitaxel directly binds to Bcl-2 and functionally mimics activityof Nur77. Cancer Res. 69, 6906–6914. doi: 10.1158/0008-5472
Findlay, J. A., and Radics, L. (1980). On the chemistry and high field nuclear mag-
netic resonance spectroscopy of rapamycin. Can. J. Chem. 58, 579–590. doi:
10.1139/v80-090
Fishlock, D., and Williams, R. M. (2008). Synthetic studies on Et-743. Assembly of
the pentacyclic core and a formal total synthesis .J .O r g .C h e m . 73, 9594–9600.
doi: 10.1021/jo801159k
Fresno, M., Jiménez, A., and Vázquez, D. (1977). Inhibition of translation
in eukaryotic systems by harringtonine. Eur. J. Biochem. 72, 323–330. doi:
10.1111/j.1432-1033.1977.tb11256.x
Furumai, R., Matsuyama, A., Kobashi, N., Lee, K. H., Nishiyama, M., Nakajima,
H., et al. (2002). FK228 (depsipeptide) as a natural prodrug that inhibits class I
histone deacetylases. Cancer Res. 62, 4916–4921
Galsky, M. D., Dritselis, A., Kirkpatrick, P ., and Oh, W. K. (2010). Cabazitaxel. Nat.
Rev. Drug Discov. 9, 677–678. doi: 10.1038/nrd3254
Ganesan, A. (2008). The impact of natural products upon modern drug discovery.
Curr. Opin. Chem. Biol. 12, 306–317. doi: 10.1016/j.cbpa.2008.03.016
Gerhardt, C. (1853). Untersuchungen über die wasserfreien organischen Säuren.
Justus Liebigs Ann. Chem. 87, 57–84. doi: 10.1002/jlac.18530870107
Gerth, K., Bedorf, N., Hofle, G., Irschik, H., and Reichenbach, H. (1996).
Epothilons A and B: antifungal and cytotoxic compounds from Sorangium cellu-
losum (Myxobacteria). Production, physico-chemical and biological properties .
J. Antibiot. (Tokyo) 49, 560–563. doi: 10.7164/antibiotics.49.560
Ghose, T., Guclu, A., and Tai, J. (1975). Suppression of an AKR Lymphoma
by antibody and chlorambucil. J. Natl. Cancer Inst. 55, 1353–1357. doi:
10.1093/jnci/55.6.1353
Ghoul, A. D., Serova, M., Astorgues-Xerri, L., Bieche, I., Bousquet, G., Varna, M.,
et al. (2009). Epithelial-to-mesenchymal transition and resistance to ingenol 3-angelate, a novel protein kinase C modulator, in colon cancer cells. Cancer Res.
69, 4260–4269. doi: 10.1158/0008-5472.can-08-2837
Goss, R. J. M., Shankar, S., and Fayad, A. A. (2012). The generation of “unNatu-
ral” products: synthetic biology meets synthetic chemistry. Nat. Prod. Rep. 29,
870–889. doi: 10.1039/C2NP00001F
Grabowski, K., Baringhaus, K.-H., and Schneider, G. (2008). Scaffold diversity of
natural products: inspiration for combinatorial library design. Nat. Prod. Rep.
25, 892–904. doi: 10.1039/B715668P
Green, A. C., and Beardmore, G. L. (1988). Home treatment of skin cancer
and solar keratoses. Australas. J. Dermatol. 29, 127–130. doi: 10.1111/j.1440-
0960.1988.tb00383.x
www.frontiersin.org M a y2 0 1 4|V o l u m e2|A r t i c l e2 0 |15
Basmadjian et al. Anticancer natural products
Groll, M., Kim, K. B., Kairies, N., Huber, R., and Crews, C. M. (2000). Crystal struc-
ture of the 20 s proteasome:TMC-95A complex: a non-covalent proteasome
inhibitor. J. Am. Chem. Soc. 122, 1237–1238. doi: 10.1021/ja993588m
Group, C. R. C. (1976). Cephalotaxine esters in the treatment of acute leukemia. A
preliminary clinical assessment. Chin. Med. J. (Engl). 2, 263–272.
Gunasekera, S. P ., Gunasekera, M., Longley, R. E., and Schulte, G. K.
(1990). Discodermolide: a new bioactive polyhydroxylated lactone from the
marine sponge Discodermia dissoluta .J. Org. Chem. 55, 4912–4915. doi:
10.1021/jo00303a029
Handratta, V . D., Vasaitis, T. S., Njar, V . C. O., Gediya, L. K., Kataria, R., Chopra, P .,
et al. (2005). Novel C-17-heteroaryl steroidal CYP17 inhibitors/antiandrogens:
synthesis, in vitro biological activity, pharmacokinetics, and antitumor activ-
ity in the LAPC4 human prostate cancer xenograft model. J. Med. Chem. 48,
2972–2984. doi: 10.1021/jm040202w
Hardy, K., Buckley, S., Collins, M. J., Estalrrich, A., Brothwell, D., Copeland, L.,
et al. (2012). Neanderthal medics? Evidence for food, cooking, and medici-
nal plants entrapped in dental calculus. Naturwissenschaften 99, 617–626. doi:
10.1007/s00114-012-0942-0
Henkel, T., Brunne, R. M., Müller, H., and Reichel, F. (1999). Statistical investi-
gation into the structural complementarity of natural products and synthetic
compounds. Angew. Chem. Int. Ed. Engl. 38, 643–647. doi: 10.1002/(SICI)1521-
3773(19990301)38:5 <643::AID-ANIE643 >3.0.CO;2-G
Hietman, J., Movva, N. R., and Hall, M. N. (1991). Targets for cell cycle arrest
by the immunosuppressant rapamycin in yeast. Science 253, 905–909. doi:
10.1126/science.1715094
Hirata, Y., and Uemura, D. (1986). Halichondrins – antitumor polyether
macrolides from a marine sponge. Pure Appl. Chem. 58, 701–710. doi:
10.1351/pac198658050701
Ho, R. J. Y., and Chien, J. (2014). Trends in translational medicine and drug
targeting and delivery: new insights on an old concept—targeted drug deliv-ery with antibody–drug conjugates for cancers. J. Pharm. Sci. 103, 71–77. doi:
10.1002/jps.23761
Hofheinz, R. D., Gnad-Vogt, S. U., Beyer, U., and Hochhaus, A. (2005).
Liposomal encapsulated anti-cancer drugs. Anticancer Drugs 16, 691–707. doi:
10.1097/01.cad.0000167902.53039.5a
Höfle, G., Bedorf, N., Steinmetz, H., Schomburg, D., Gerth, K., and Reichenbach,
H. (1996). Epothilone A and B – novel 16-membered macrolides with cyto-
toxic activity: isolation, crystal structure, and conformation in solution. Angew.
Chem. Int. Ed. Engl. 35, 1567–1569. doi: 10.1002/anie.199615671
Horwitz, S. B. (1994). Taxol (paclitaxel): mechanisms of action. Ann. Oncol.
5(Suppl. 6), S3–S6.
Huang, M. T. (1975). Harringtonine, an inhibitor of initiation of protein biosyn-
thesis. Mol. Pharmacol. 11, 511–519.
Imai, T., Nakata, H., Y okoshima, S., and Fukuyama, T. (2012). Synthetic stud-
ies toward ecteinascidin 743 (Trabectedin). Synthesis 44, 2743–2753. doi:
10.1055/s-0032-1316579
Jackson, K. L., Henderson, J. A., and Phillips, A. J. (2009). The halichondrins and
E7389. Chem. Rev. 109, 3044–3079. doi: 10.1021/cr900016w
Jarman, M., Barrie, S. E., and Llera, J. M. (1998). The 16,17-double bond is needed
for irreversible inhibition of human cytochrome P45017 αby abiraterone (17-
(3-Pyridyl)androsta-5,16-dien-3 β-ol) and related steroidal inhibitors. J. Med.
Chem. 41, 5375–5381. doi: 10.1021/jm981017j
Jin, S., Gorfajn, B., Faircloth, G., and Scotto, K. W. (2000). Ecteinascidin 743, a
transcription-targeted chemotherapeutic that inhibits MDR1 activation. Proc.
Natl. Acad. Sci. U.S.A. 97, 6775–6779. doi: 10.1073/pnas.97.12.6775
Johnson, P ., Geldart, T., Fumoleau, P ., Pinel, M.-C., Nguyen, L., and Judson,
I. (2006). Phase I study of Vinflunine administered as a 10-minute infu-
sion on days 1 and 8 every 3 weeks. Invest. New Drugs 24, 223–231. doi:
10.1007/s10637-005-3902-0
Jordan, M. A., and Wilson, L. (2004). Microtubules as a target for anticancer drugs.
Nat. Rev. Cancer 4, 253–265. doi: 10.1038/nrc1317
Jørgensen, L., McKerrall, S. J., Kuttruff, C. A., Ungeheuer, F., Felding, J., and Baran,
P . S. (2013). 14-step synthesis of ( +)-ingenol from ( +)-3-carene. Science 341,
878–882. doi: 10.1126/science.1241606
Kahan, B. D., Chang, J. Y., and Sehgal, S. N. (1991). Preclinical evaluation of a new
potent immunosuppressive agent, rapamycin. Transplantation 52, 185–191. doi:
10.1097/00007890-199108000-00001
Kantarjian, H. M., O’Brien, S., and Cortes, J. (2013).
Homoharringtonine/Omacetaxine mepesuccinate: the long and windingroad to food and drug administration approval. Clin. Lymphoma Myeloma
Leuk. 13, 530–533. doi: 10.1016/j.clml.2013.03.017
Kawagishi, F., T oma, T., Inui, T., Y okoshima, S., and Fukuyama, T. (2013). T otal
synthesis of ecteinascidin 743. J. Am. Chem. Soc. 135, 13684–13687. doi:
10.1021/ja408034x
Kim, T. Y., Kim, D. W., Chung, J. Y., Shin, S. G., Kim, S. C., Heo, D. S., et al. (2004).
Phase I and pharmacokinetic study of Genexol-PM, a Cremophor-free, poly-
meric micelle-formulated paclitaxel, in patients with advanced malignancies.
Clin. Cancer Res. 10, 3708–3716. doi: 10.1158/1078-0432.CCR-03-0655
King, M. L., Chiang, C. C., Ling, H. C., Ochiai, M., Fujita, E., and Mcphail, A.
T. (1985). X-Ray crystal structure of rocaglamide, a novel antileulemic 1H-
cyclopenta[b]benzofuran from Aglaia elliptifolia. J. Chem. Soc. Chem. Commun.
1982, 1150–1151. doi: 10.1039/C39820001150
Kino, T., Hatanaka, H., Hashimoto, M., Nishiyama, M., Goto, T., Okuhara, M., et al.
(1987a). FK-506, a novel immunosuppressant isolated from a Streptomyces.
I. Fermentation, isolation, and physico-chemical and biological characteristics.
J. Antibiot. (Tokyo) 40, 1249–1255.
Kino, T., Hatanaka, H., Miyata, S., Inamura, N., Nishiyama, M., Y ajima, T., et al.
(1987b). FK-506, a novel immunosuppressant isolated from a Streptomyces. II.
Immunosuppressive effect of FK-506 in vitro .J. Antibiot. (Tokyo) 40, 1256–1265.
Kirikae, T., Ojima, I., Kirikae, F., Ma, Z., Kuduk, S. D., Slater, J. C., et al. (1996).
Structural requirements of taxoids for nitric oxide and tumor necrosis fac-tor production by murine macrophages. Biochem. Biophys. Res. Commun. 227,
227–235. doi: 10.1006/bbrc.1996.1494
Kirschning, A., and Hahn, F. (2012). Merging chemical synthesis and biosyn-
thesis: a new chapter in the total synthesis of natural products and nat-
ural product libraries. Angew. Chem. Int. Ed. Engl. 51, 4012–4022. doi:
10.1002/anie.201107386
Kohler, G., and Milstein, C. (1975). Continuous cultures of fused cells secreting
antibody of predefined specificity. Nature 256, 495–497. doi: 10.1038/256495a0
Krysiak, J., and Breinbauer, R. (2012). Activity-based protein profiling for
natural product target discovery. Top. Curr. Chem. 324, 43–84. doi:
10.1007/128_2011_289
Leamon, C. P ., and Low, P . S. (1991). Delivery of macromolecules into living cells:
a method that exploits folate receptor endocytosis. Proc. Natl. Acad. Sci. U.S.A.
88, 5572–5576. doi: 10.1073/pnas.88.13.5572
Lee, F. F., Borzilleri, R., Fairchild, C., Kamath, A., Smykla, R., Kramer, R., et al.
(2008). Preclinical discovery of ixabepilone, a highly active antineoplastic
agent. Cancer Chemother. Pharmacol. 63, 157–166. doi: 10.1007/s00280-008-
0724-8
Lee, F. Y. F., Vite, G., Borzilleri, R. M., Arico, M. A., Clark, J. L., Fager, K. L., et al.
(2000). Preclinical pharmacology of the epothilone B analog BMS-247550—
an epothilone analog possessing potent activity against paclitaxel sensitive andresistant human tumors. Clin. Cancer Res. 6:4580s.
Lee, M.-L., and Schneider, G. (2001). Scaffold architecture and pharmacophoric
properties of natural products and trade drugs:? application in the design of
natural product-based combinatorial libraries. J. Comb. Chem. 3, 284–289. doi:
10.1021/cc000097l
Lindel, T., Jensen, P . R., Fenical, W., Long, B. H., Casazza, A. M., Carboni, J., et al.
(1997). Eleutherobin, a new cytotoxin that mimics paclitaxel (Taxol) by sta-
bilizing microtubules. J. Am. Chem. Soc. 119, 8744–8745. doi: 10.1021/ja971
7828
Loc, T. B., Mathé, G., and Bernard, J. (1958). Effet sur la leucémie 1210 de la
souris d’une combinaison par diazotation d’A-méthopterine ct de γ-globulins
de hamsters porteurs de cette leucémic par hétérogreffe. C. R. Acad. Sci. Paris
246, 1626–1628.
Martinez, E. J., Owa, T., Schreiber, S. L., and Corey, E. J. (1999). Phthalascidin,
a synthetic antitumor agent with potency and mode of action compara-ble to ecteinascidin 743. Proc. Natl. Acad. Sci. U.S.A. 96, 3496–3501. doi:
10.1073/pnas.96.7.3496
McAlpine, J. B., Swanson, S. J., Jackson, M., and Whittern, D. N. (1991).
Revised NMR assignments for rapamycin. J. Antibiot. (Tokyo) 44, 688–690. doi:
10.7164/antibiotics.44.688
Menchaca, R., Martinez, V ., Rodriguez, A., Rodriguez, N., Flores, M., Gallego, P .,
et al. (2003). Synthesis of natural ecteinascidins (ET-729, ET-745, ET-759B, ET-
736, ET-637, ET-594) from cyanosafracin B. J. Org. Chem. 68, 8859–8866. doi:
10.1021/jo034547i
Minuzzo, M., Marchini, S., Broggini, M., Faircloth, G., D’Incalci, M., and
Mantovani, R. (2000). Interference of transcriptional activation by the
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry May 2014 | Volume 2 | Article 20 |16
Basmadjian et al. Anticancer natural products
antineoplastic drug ecteinascidin-743. Proc. Natl. Acad. Sci. U.S.A. 97,
6780–6784. doi: 10.1073/pnas.97.12.6780
Mooberry, S. L., Tien, G., Hernandez, A. H., Plubrukarn, A., and Davidson, B.
S. (1999). Laulimalide and isolaulimalide, new paclitaxel-like microtubule-stabilizing agents. Cancer Res. 59, 653–660.
Mulzer, J., Mantoulidis, A., and Öhler, E. (2000). T otal syntheses of epothilones B
and D. J. Org. Chem. 65, 7456–7467. doi: 10.1021/jo0007480
Nakajima, H., Kim, Y. B., T erano, H., Y oshida, M., and Horinouchi, S. (1998).
FR901228, a potent antitumor antibiotic, is a novel histone deacetylaseinhibitor. Exp. Cell Res. 241, 126–133. doi: 10.1006/excr.1998.4027
Nicolaou, K. C., Ninkovic, S., Sarabia, F., Vourloumis, D., He, Y., Vallberg, H., et al.
(1997). T otal syntheses of epothilones A and B via a macrolactonization-basedstrategy. J. Am. Chem. Soc. 119, 7974–7991. doi: 10.1021/ja971110h
Nishizuka, Y. (1984). The role of protein kinase C in cell surface signal transduction
and tumour promotion. Nature 308, 693–698. doi: 10.1038/308693a0
Noble, R. L., Beer, C. T., and Cutts, J. H. (1959). Further biological activities of vin-
caleukoblastine – an alkaloid isolated from Vinca rosea (L.). Biochem. Pharmacol.
1, 347–348. doi: 10.1016/0006-2952(59)90123-6
Ojima, I., Chakravarty, S., Inoue, T., Lin, S., He, L., Horwitz, S. B., et al. (1999). A
common pharmacophore for cytotoxic natural products that stabilize micro-
tubules. Proc. Natl. Acad. Sci. U.S.A. 96, 4256–4261. doi: 10.1073/pnas.96.
8.4256
Ortholand, J.-Y., and Ganesan, A. (2004). Natural products and combinato-
rial chemistry: back to the future. Curr. Opin. Chem. Biol. 8, 271–280. doi:
10.1016/j.cbpa.2004.04.011
Paudler, W. W., Kerley, G. I., and McKay, J. (1963). The alkaloids of Cephalotaxus
drupacea and Cephalotaxus fortunei .J. Org. Chem. 28, 2194–2197. doi:
10.1021/jo01044a010
Pommier, Y., Kohlhagen, G., Bailly, C., Waring, M., Mazumder, A., and Kohn, K. W.
(1996). DNA sequence- and structure-selective alkylation of guanine N2 in theDNA minor groove by ecteinascidin 743, a potent antitumor compound from
the caribbean tunicate Ecteinascidia turbinata .Biochemistry 35, 13303–13309.
doi: 10.1021/bi960306b
Potter, G. A., Barrie, S. E., Jarman, M., and Rowlands, M. G. (1995). Novel
steroidal inhibitors of human cytochrome P45017.alpha.-hydroxylase-C17,20-lyase): potential agents for the treatment of prostatic cancer. J. Med. Chem. 38,
2463–2471. doi: 10.1021/jm00013a022
Powell, R. G., Weisleder, D., and Smith, C. R. (1972). Antitumor alkaloids from
Cephalotaxus harringtonia :s t r u c t u r ea n da c t i v i t y . J. Pharm. Sci. 61, 1227–1230.
doi: 10.1002/jps.2600610812
Powell, R. G., Weisleder, D., Smith C. R. Jr., and Rohwedder, W. K.
(1970). Structures of harringtonine, isoharringtonine, and homohar-
ringtonine. Tetrahedron Lett. 11, 815–818. doi: 10.1016/S0040-4039(01)
97839-6
Pucheault, M. (2008). Natural products: chemical instruments to apprehend bio-
logical symphony. Org. Biomol. Chem. 6, 424–432. doi: 10.1039/b713022h
Qi, Y., and Ma, S. (2011). The medicinal potential of promising marine
macrolides with anticancer activity. ChemMedChem 6, 399–409. doi:
10.1002/cmdc.201000534
Rehman, Y., and Rosenberg, J. E. (2012). Abiraterone acetate: oral androgen biosyn-
thesis inhibitor for treatment of castration-resistant prostate cancer. Drug Des.
Devel. Ther. 6, 13–18. doi: 10.2147/DDDT.S15850
Rinehart, K. L., Holt, T. G., Fregeau, N. L., Stroh, J. G., Keifer, P . A., Sun, F., et al.
(1990). Ecteinascidins 729, 743, 745, 759A, 759B, and 770: potent antitumor
agents from the Caribbean tunicate Ecteinascidia turbinata .J. Org. Chem. 55,
4512–4515. doi: 10.1021/jo00302a007
Ritter, S. K. (2013). 2013’s notable advances. Chem. Eng. News 91, 16–19.
Robin, J.-P ., Dhal, R., Dujardin, G., Girodier, L., Mevellec, L., and Poutot, S. (1999).
The first semi-synthesis of enantiopure homoharringtonine via anhydroho-
moharringtonine from a preformed chiral acyl moiety. Tetrahedron Lett. 40,
2931–2934. doi: 10.1016/S0040-4039(99)00327-5
Roin, B. N. (2009). Unpatentable drugs and the standards of patentability. Tex. Law
Rev. 87, 503–570.
Santagata, S., Mendillo, M. L., Tang, Y.-C., Subramanian, A., Perley, C. C., Roche, S.
P ., et al. (2013). Tight coordination of protein translation and HSF1 activation
supports the anabolic malignant state. Science 341, 1238303-1–1238303-10. doi:
10.1126/science.1238303
Sayed, M. D., Riszk, A., Hammouda, F. M., El-Missiry, M. M., Williamson, E. M.,
and Evans, F. J. (1980). Constituents of Egyptian Euphorbiaceae. IX. Irritant andcytotoxic ingenane esters from Euphorbia paralias L.Experientia 36, 1206–1207.
doi: 10.1007/BF01976131
Schinzer, D., Limberg, A., Bauer, A., Böhm, O. M., and Cordes, M. (1997). T otal
synthesis of (-)-epothilone A. Angew. Chem. Int. Ed. Engl. 36, 523–524. doi:
10.1002/anie.199705231
Scott, A. M., Wolchok, J. D., and Old, L. J. (2012). Antibody therapy of cancer. Nat.
Rev. Cancer 12, 278–287. doi: 10.1038/nrc3236
Sehgal, S. N., Baker, H., and Vezina, C. (1975). Rapamycin (AY 22,989), a new anti-
fungal antibiotic. II. Fermentation, isolation and characterization . J. Antibiot.
(Tokyo) 28, 727–732.
Serova, M., Ghoul, A. D., Benhadji, K. A., Faivre, S., Le T ourneau, C., Cvitkovic,
E., et al. (2008). Effects of protein kinase C modulation by PEP005, a novelingenol angelate, on mitogen-activated protein kinase and phosphatidylinos-
itol 3-kinase signaling in cancer cells. Mol. Cancer Ther. 7, 915–922. doi:
10.1158/1535-7163.mct-07-2060
Sigel, M. M., Wellham, L. L., Lichter, W., Dudeck, L. E., Gargus, J. L., and Lucas, A.
H. (1970). “Anticellular and antitumor activity of extracts from tropical marineinvertebrates,” in Food-Drugs from the Sea Proceedings 1969 ,e dH .W .Y o u n g k e n
Jr. (Washington, DC: Marine T echnology Society), 281–294.
Siller, G., Gebauer, K., Welburn, P ., Katsamas, J., and Ogbourne, S. M. (2009).
PEP005 (ingenol mebutate) gel, a novel agent for the treatment of actinic
keratosis: results of a randomized, double-blind, vehicle-controlled, multicen-tre, phase IIa study. Australas. J. Dermatol. 50, 16–22. doi: 10.1111/j.1440-
0960.2008.00497.x
Su, D.-S., Meng, D., Bertinato, P ., Balog, A., Sorensen, E. J., Danishefsky, S. J., et al.
(1997). T otal synthesis of (–)-epothilone B: an extension of the suzuki coupling
method and insights into structure–activity relationships of the epothilones.
Angew. Chem. Int. Ed. Engl. 36, 757–759. doi: 10.1002/anie.199707571
Sundar, S., Jha, T. K., Thakur, C. P ., Sinha, P . K., and Bhattacharya, S. K. (2007).
Injectable paromomycin for visceral leishmaniasis in India. N. Engl. J. Med. 356,
2571–2581. doi: 10.1056/NEJMoa066536
Swindells, D. C. N., White, P . S., and Findlay, J. A. (1978). The X-ray crys-
tal structure of rapamycin, C51H79NO13. Can. J. Chem. 56, 2491–2492. doi:
10.1139/v78-407
Swinney, D. C., and Anthony, J. (2011). How were new medicines discovered? Nat.
Rev. Drug Discov. 10, 507–519. doi: 10.1038/nrd3480
Taamma, A., Misset, J. L., Riofrio, M., Guzman, C., Brain, E., Lopez Lazaro, L.,
et al. (2001). Phase I and pharmacokinetic study of ecteinascidin-743, a new
marine compound, administered as a 24-hour continuous infusion in patients
with solid tumors. J. Clin. Oncol. 19, 1256–1265.
Taft, F., Brünjes, M., Floss, H. G., Czempinski, N., Grond, S., Sasse, F., et al. (2008).
Highly active ansamitocin derivatives: mutasynthesis using an AHBA-blocked
mutant. Chembiochem 9, 1057–1060. doi: 10.1002/cbic.200700742
Takebayashi, Y., Pourquier, P ., Y oshida, A., Kohlhagen, G., and Pommier, Y. (1999).
Poisoning of human DNA topoisomerase I by ecteinascidin 743, an anticancer
drug that selectively alkylates DNA in the minor groove. Proc. Natl. Acad. Sci.
U.S.A. 96, 7196–7201. doi: 10.1073/pnas.96.13.7196
Takebayashi, Y., Pourquier, P ., Zimonjic, D. B., Nakayama, K., Emmert, S., Ueda, T.,
et al. (2001). Antiproliferative activity of ecteinascidin 743 is dependent upon
transcription-coupled nucleotide-excision repair. Nat. Med. 7, 961–966. doi:
10.1038/91008
Tang, R., Faussat, A.-M., Majdak, P ., Marzac, C., Dubrulle, S., Marjanovic, Z., et al.
(2006). Semisynthetic homoharringtonine induces apoptosis via inhibition ofprotein synthesis and triggers rapid myeloid cell leukemia-1 down-regulation
in myeloid leukemia cells. Mol. Cancer Ther. 5, 723–731. doi: 10.1158/1535-
7163.mct-05-0164
Thaler, F., and Mercurio, C. (2014). T owards selective inhibition of histone deacety-
lase isoforms: what has been achieved, where we are and what will be next.ChemMedChem 9, 523–536. doi: 10.1002/cmdc.201300413
Thuaud, F., Ribeiro, N., Nebigil, C. G., and Désaubry, L. (2013). Prohibitin ligands
in cell death and survival: mode of action and therapeutic potential. Chem. Biol.
20, 316–331. doi: 10.1016/j.chembiol.2013.02.006
Ueda, H., Nakajima, H., Hori, Y., Goto, T., and Okuhara, M. (1994).
Action of FR901228, a novel antitumor bicyclic depsipeptide produced by
Chromobacterium violaceum no. 968, on Ha-ras transformed NIH3T3 cells.
Biosci. Biotechnol. Biochem. 58, 1579–1583.
Uemura, D., Takahashi, K., Y amamoto, T., Katayama, C., Tanaka, J., Okumura,
Y., et al. (1985). Norhalichondrin A: an antitumor polyether macrolide from
a marine sponge. J. Am. Chem. Soc. 107, 4796–4798. doi: 10.1021/ja00302a042
www.frontiersin.org May 2014 | Volume 2 | Article 20 |17
Basmadjian et al. Anticancer natural products
Vandermolen, K. M., McCulloch, W., Pearce, C. J., and Oberlies, N. H. (2011).
Romidepsin (Istodax, NSC 630176, FR901228, FK228, depsipeptide): a natural
product recently approved for cutaneous T-cell lymphoma. J. Antibiot. (Tokyo)
64, 525–531. doi: 10.1038/ja.2011.35
Vasaitis, T. S., and Njar, V . C. O. (2010). Novel, potent anti-androgens of therapeutic
potential: recent advances and promising developments. Future Med. Chem. 2,
667–680. doi: 10.4155/fmc.10.14
Vezina, C., Kudelski, A., and Sehgal, S. N. (1975). Rapamycin (AY 22,989), a new
antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation
of the active principle. J. Antibiot. (Tokyo) 28, 721–726.
Villalona-Calero, M. A., Eckhardt, S. G., Weiss, G., Hidalgo, M., Beijnen, J. H., Van
Kesteren, C., et al. (2002). A phase I and pharmacokinetic study of ecteinascidin-
743 on a daily x 5 schedule in patients with solid malignancies. Clin. Cancer Res.
8, 75–85.
Vlahov, I. R., and Leamon, C. P . (2012). Engineering folate-drug conjugates to
target cancer: from chemistry to clinic. Bioconjug. Chem. 23, 1357–1369. doi:
10.1021/bc2005522
Von Hoff, D. D. (1998). There are no bad anticancer agents, only bad clinical trial
designs–twenty-first Richard and Hinda Rosenthal Foundation Award Lecture.
Clin. Cancer Res. 4, 1079–1086.
Vrignaud, P ., Sémiond, D., Lejeune, P ., Bouchard, H., Calvet, L., Combeau, C.,
et al. (2013). Preclinical antitumor activity of cabazitaxel, a semisynthetic tax-
ane active in taxane-resistant tumors. Clin. Cancer Res. 19, 2973–2983. doi:
10.1158/1078-0432.ccr-12-3146
Wang, A. Z., Langer, R., and Farokhzad, O. C. (2012). Nanoparticle delivery of
cancer drugs. Annu. Rev. Med. 63, 185–198. doi: 10.1146/annurev-med-040210-
162544
Watson, C. J. E., Friend, P . J., Jamieson, N. V ., Frick, T. W., Alexander, G.,
Gimson, A. E., et al. (1999). Sirolimus: a potent new immunosuppressant
for liver transplantation. Transplantation 67, 505–509. doi: 10.1097/00007890-
199902270-00002
Weedon, D., and Chick, J. (1976). Home treatment of basal cell carcinoma. Med. J.
Aust. 1, 928.
Xu, Z. (2011). Modernization: one step at a time. Nature 480, S90–S92. doi:
10.1038/480S90a
Y adav, V . G., De Mey, M., Giaw Lim, C., Kumaran Ajikumar, P ., and
Stephanopoulos, G. (2012). The future of metabolic engineering and syn-
thetic biology: towards a systematic practice. Metab. Eng. 14, 233–241. doi:
10.1016/j.ymben.2012.02.001Y ang, J., Chen, H., Vlahov, I. R., Cheng, J.-X., and Low, P . S. (2006). Evaluation of
disulfide reduction during receptor-mediated endocytosis by using FRET imag-
ing. Proc. Natl. Acad. Sci. U.S.A. 103, 13872–13877. doi: 10.1073/pnas.06014
55103
Y ang, Z., He, Y., Vourloumis, D., Vallberg, H., and Nicolaou, K. C. (1997). T otal
synthesis of epothilone A: the olefin metathesis approach. Angew. Chem. Int.
Ed. Engl. 36, 166–168. doi: 10.1002/anie.199701661
Zard, S. Z. (2012). Some aspects of the chemistry of nitro compounds. Helv. Chim.
Acta 95, 1730–1757. doi: 10.1002/hlca.201200324
Zewail-Foote, M., and Hurley, L. H. (1999). Ecteinascidin 743: a minor groove alky-
lator that bends DNA toward the major groove. J. Med. Chem. 42, 2493–2497.
doi: 10.1021/jm990241l
Zewail-Foote, M., Li, V .-S., Kohn, H., Bearss, D., Guzman, M., and Hurley, L.
H. (2001). The inefficiency of incisions of ecteinascidin 743–DNA adducts
by the UvrABC nuclease and the unique structural feature of the DNA
adducts can be used to explain the repair-dependent toxicities of this
antitumor agent. Chem. Biol. 8, 1033–1049. doi: 10.1016/S1074-5521(01)
00071-0
Zheng, S., Chan, C., Furuuchi, T., Wright, B. J. D., Zhou, B., Guo, J., et al. (2006).
Stereospecific formal total synthesis of ecteinascidin 743. Angew. Chem. Int. Ed.
Engl. 45, 1754–1759. doi: 10.1002/anie.200503983
Conflict of Interest Statement: The authors declare that the research was con-
ducted in the absence of any commercial or financial relationships that could be
construed as a potential conflict of interest.
Received: 06 March 2014; accepted: 04 April 2014; published online: 01 May 2014.
Citation: Basmadjian C, Zhao Q, Bentouhami E, Djehal A, Nebigil CG, Johnson RA,
Serova M, de Gramont A, Faivre S, Raymond E and Désaubry LG (2014) Cancer wars:
natural products strike back. Front. Chem. 2:20. doi: 10.3389/fchem.2014.00020
This article was submitted to Medicinal and Pharmaceutical Chemistry, a section of
the journal Frontiers in Chemistry.
Copyright © 2014 Basmadjian, Zhao, Bentouhami, Djehal, Nebigil, Johnson, Serova,
de Gramont, Faivre, Raymond and Désaubry. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The use, dis-
tribution or reproduction in other forums is permitted, provided the original author(s)
or licensor are credited and that the original publication in this journal is cited, in
accordance with accepted academic practice. No use, distribution or reproduction is
permitted which does not comply with these terms.
Frontiers in Chemistry | Medicinal and Pharmaceutical Chemistry M a y2 0 1 4|V o l u m e2|A r t i c l e2 0 |18
Copyright Notice
© Licențiada.org respectă drepturile de proprietate intelectuală și așteaptă ca toți utilizatorii să facă același lucru. Dacă consideri că un conținut de pe site încalcă drepturile tale de autor, te rugăm să trimiți o notificare DMCA.
Acest articol: Cancer wars: natural products strike back Christine Basmadjian1,2, Qian Zhao1,2, Embarek Bentouhami3, Amel Djehal1,3, Canan G. Nebigil4, Roger A…. [600187] (ID: 600187)
Dacă considerați că acest conținut vă încalcă drepturile de autor, vă rugăm să depuneți o cerere pe pagina noastră Copyright Takedown.
