Zhang 2007
Zhang 2007
Zhang 2007
coupled with the relatively straightforward nature of their 500 A [16 ]. In the meantime, the volumes found in over 18
manufacturing. This is probably best illustrated by the 000 binding cavities of protein targets range from
˚3
development of tyrosine kinase inhibitors for the treat-ment 300 to 800 A . Therefore, the average volumes of natural
of cancer [14]. Earlier encouraging data generated with products correlate with the average dimensions of protein
MAbs such as cetuximab and trastuzumab has been cavities (note that protein ligands often do not fill the entire
beneficial to the development of small-molecule inhibi-tors volume of a given protein cavity). Although many natural
such as gefitinib, erlotinib, and lapatinib. products from plants and microorganisms are not meant to
bind to human proteins, many human proteins consist of the
It therefore appears sensible to adopt a more program-matic same building blocks and contain similar structural
approach in discovering new drugs instead of wholly domains to the targets with which natural products have
concentrating on oral bioavailability. That is: coevolved.
to progress initially a parenteral candidate before too Natural products therefore represent privileged chemical
much effort and resources have to be invested for an oral starting points for drug discovery, and yet they have quite
candidate; and distinct structural characteristics from synthetic molecules.
to progress initially a therapeutic antibody wherever For example, when analyzed by either a size-independent
possible (most probably by parenteral administration) ‘chemistry space filter’ or ‘support-vector-machine’
before too much effort and resources have to be invested approach, natural products exhibit better scores of ‘drug-
for a small-molecule candidate. likeness’ than synthetic compounds [17]. Further, natural
products contain on average twice as many oxygen atoms
These may be particularly relevant for ‘first-in-class’ drug and three times fewer nitrogen atoms than synthetic drug
discovery efforts against difficult targets such as proteases molecules [17]. They also contain a slightly higher number
and those involving protein–protein interactions. of hydrogen-bond donors than do synthetic drugs. Natural
products contain approximately four times more chiral
Natural products and drug discovery centers and far fewer aromatic rings, a fact which may
productivity engender upon natural products better selectivity when
A major class of drug molecules that are excluded from the binding to stereo-defined sites.
original analysis which generated the ‘rule-of-five’ are natural
products. This is a serious defect as we know that a large Better exploration of the biologically prevalidated struc-
percentage of marketed drugs are natural products and their tural scaffolds of natural products could increase our
semisynthetic derivatives. A recent analysis of all drugs success rate of finding more active leads. For example, the
approved worldwide between 1981 and 2006 showed that 34% research group of Waldmann, in collaboration with
of all small-molecule drugs are natural products or Novartis, have devised a hierarchical structural
Table 1
US FDA-approved polyketide drugs (up to December 2006)
Synthetic biology is an emerging scientific field which Although the structures of natural products can be highly
seeks to understand and design biological systems and/or complex and diverse, their biosynthesis can be accom-
Figure 1
Polyketide biosynthesis on a modular PKS as exemplified by the biosynthesis of rapamycin. Biosynthesis is initiated by loading of the PKS with 4,5-
dihydroxycyclohexe-1-ene carboxylic acid, with reduction by the loading module ER domain, and subsequent rounds of chain extensions with
malonyl-CoA and methylmalonyl-CoA, in which various levels of reductive processing of the newly formed b-keto groups occur. The modular
organization of the PKS is shown as a series of spherical domains: CoL, CoA-ligase like domain; ER, enoylreductase; ACP, acylcarrier protein; AT,
acyltransferase; DH, dehydratase; KR, b-ketoacylreductase; KS, b-ketoacylsynthase. Biosynthesis is completed by the amidation of the chain with L-
pipecolic acid and macrocylization. The resulting prerapamycin is then converted to rapamycin by the action of several post-PKS acting
monooxygenases and methyltransferase.
Current Opinion in Biotechnology 2007, 18:478–488 www.sciencedirect.com
Drug discovery beyond ‘Rule-of-Five’ Zhang and Wilkinson 483
plished by a remarkably few simple types of reactions. One Island (Rapa Nui). It is a very potent inhibitor of the serine-
example is the biosynthesis of the aliphatic, or reduced, threonine kinase target of rapamycin (TOR) involved in the
polyketides [22], a structurally diverse class of natural phosphatidylinositol 3-kinase (PI3K)/ Akt (protein kinase
products that include drugs such as lovastatin, B) signaling pathway that mediates cell survival and
erythromycin, rifamycin, amphotericin, and rapamycin proliferation [24]. It inhibits TOR by first binding to the
among others (Table 1). The biosynthesis of their poly- immunophilin FK506-binding protein 12 (FKBP-12) and
ketide core structures involves one major type of reac-tion the resulting binary complex then binds to TOR at an
— a decarboxylative Claisen-type condensation in which allosteric site, not competing directly with ATP. It is
various acyl-CoA monomers are selected by a polyketide therefore the most selective kinase inhibitor known till
synthase and added, one by one, to the grow-ing polyketide date.
chain which is tethered covalently to the PKS (Figure 1).
The resulting b-keto esters thus formed can be further The structural complexity of rapamycin has been a major
(optionally) reduced to a hydroxyl group, dehydrated to challenge for analog preparation, and there have been
give an olefin, and finally reduced to a saturated methylene several total syntheses reported over the years [25]. None
unit. These few simple chemical transformations occur with of these, however, is less than 50 steps, nor offers an
specific stereochemical out-comes, and in combination, overall yield >0.5%. It is therefore impractical to use any of
generate tremendous struc-tural diversity. The simplicity of these for lead optimization. As a consequence, the
polyketide biosynthesis is further facilitated by the modular rapamycin analogs that have progressed to market or
organization of the multienzyme PKSs in which highly clinical development are all simple semisynthetic deriva-
similar ‘modules’ — a group of enzymatic domains tives at a single structural point, the secondary hydroxyl
responsible for one ketide extension — act as an assembly functionality at C40 (Figure 2) [24]. The structural sim-
production line, with the structure of the chemical product ilarity of these compounds means they share very similar
‘hardwired’ into the DNA sequence encoding the PKS. biological profiles with little significant improvement, for
Whilst exceptions to the ‘one module for one ketide example, over poor pharmaceutical or pharmacokinetic
extension’ rule do exist [23], in general this simple properties.
template-based process pro-vides a direct and easily
understood relationship between DNA sequence and The biosynthesis of rapamycin is catalyzed by a mixed type
chemical structure, and a paradigm for the rational I PKS/nonribosomal peptide synthetase (NRPS) system
engineering of such systems to generate new natural (Figure 1) [26]. The PKS uses a shikimate-derived 4,5-
product analogs biosynthetically [22]. dihydroxycyclohex-1-enecarboxylic acid starter unit and
carries out a total of 14 successive steps of polyketide chain
As an example, this technology has been successfully used extension. The NRPS then incorporates an L-lysine-derived
to generate novel rapamycin analogs potentially useful as L-pipecolicacid moiety into the chain, followed by cleavage
immunosuppressants, anticancer and anti-inflammatory from the enzyme to produce the key intermediate
drugs. Rapamycin (Figure 2) is a polyketide macrolide prerapamycin [27]. Prerapamycin is further modified by
originally discovered from the culture of Strep-tomyces methyltransferases (RapI, RapM, and RapQ) and cytochrome
hygroscopicus NRRL5491 collected from Easter P450 monooxygenases
Figure 2
Structures of rapamycin and its semisynthetic analogs in clinical use or trials.
www.sciencedirect.com Current Opinion in Biotechnology 2007, 18:478–488
484 Chemical biotechnology
Figure 3
Mutasynthesis of prerapamycin analogs using the genetically engineered strain Streptomyces hygroscopicus MG2-10 and feeding exogenous starter
acid analogs [28].
(RapJ and RapN) to yield the fully processed rapamycin tory potency against TOR kinase activity and exhibits
molecule [28]. potent anticancer activities in various in vitro and in vivo
assays. It is not a substrate of the P-gp efflux pump and has
Interestingly, when a region of DNA including the genes good oral bioavailability. Of particular interest is the
thought to encode those post-PKS acting methyltransfer- effective penetration of the blood–brain barrier by BC210.
ases and monooxygenases (rap KIJMNOQL) were excised, When given intravenously (i.v.) 3 mg/kg in mice, it
the resultant mutant S. hygroscopicus MG2-10 was unable ‘accumulates’ in the brain resulting in the brain to blood
to produce prerapamycin unless fed with exogenous area under curve (AUC) ratio of 1.6, a property which is
pseudo-starter acid 3,4-dihydroxycyclohexane-carboxylic unique among published rapamycin analogs (cf. the ratio
acid or complemented with the gene rapK (Figure 3) [29 ]. for rapamycin is 0.25). This pharmacokinetic property of
This indicated that the product of rapK is involved in starter BC210 makes the compound a highly promising candi-date
acid biosynthesis and/or its regulation. as a potential therapeutic for the treatment of brain tumors
such as glioblastoma multiformes, or neurodegenerative
The production of prerapamycin by feeding exogenous diseases, either alone or in combi-nation with other
starter acid to the MG2-10 mutant, combined with the therapeutic agents [31].
restoration of post-PKS structural changes by expressing all
or selected post-PKS genes (rapI,J,M,N,O,Q), opened up The utility of such genetically engineered biosynthesis
the possibility of the combinatorial biosynthesis of a wide approaches for drug discovery has additional benefit
range of rapamycin analogs [29 ]. For example, it was compared to the more widely used semisynthetic
demonstrated that feeding other carboxylic acids in place of approaches. For example, semisynthetic derivatization
the starter unit acid led to the production of various
often produces analogs with larger molecule sizes than the
rapamycin analogs. One of these analogs, BC210 (Figure
4), which has the metabolically labile C39-meth-oxy group natural product lead, leading to reduced ligand effi-ciency
of rapamycin removed from its structure has shown [32]. Biosynthetic modifications need not suffer from this
drawback. The 39-desmethoxy rapamycin BC210 (Figure
significantly improved metabolic stability (t 1/2 59 min
4) has similar FKBP-12/TOR binding affinity to rapamycin
when incubated with human liver microsomes) and Caco-2
but its molecular weight is slightly less than rapamycin. On
permeability (Papp 29 nm/s) compared to rapamycin (t 1/2 40 the contrary, all semisynthetic derivatives of rapamycin
min with human liver microsomes and P app 2 nm/s) [30]. currently marketed or under clinical development (Figure
BC210 has low nanomolar inhibi- 2) have larger molecular
Figure 4
Design and biosynthetic preparation of BC210: removal of the methoxy moiety at C39 of rapamycin using mutasynthesis provides a compound
with enhanced metabolic stability versus human liver microsomes [29].
weight than rapamycin and equal or less affinity to the 1.2 mM, MW 561) (Figure 5) [34]. A further advantage of
targets FKBP-12/TOR. the nonquinone derivative produced by biosynthetic
engineering over the semisynthetic tanespimycin is that it
This advantage of being able to maintain or increase ligand does not undergo redox cycling and should have signifi-
efficiency by biosynthesis-based lead optimization has also cantly reduced off-target toxicity and consequently an
been observed in several other cases. For example, increased therapeutic window.
optimization of the naturally occurring heat-shock protein-
90 (Hsp90) inhibitor macbecin I by a synthetic biology An increased therapeutic window has also been achieved
approach has generated a nonquinone derivative that has
with a series of borrelidin analogs without increasing their
significantly increased affinity to Hsp90 (K d 0.007 mM) molecular sizes [35]. The biosynthetic derivative BC194
and a reduced molecular weight (MW 519) compared to the exhibited decreased toxicity but increased antiangiogenic
lead molecule (Kd 0.24 mM, MW 559 for macbecin I) activity against human umbi-lical vein endothelial cells
(Figure 5) [33]. On the contrary, semisynthetic (HUVEC) compared to bor-relidin, and its reduced toxicity
optimization of another naturally occurring Hsp90 inhibitor was confirmed in an in vivo study in nude mice. The
geldanamycin has resulted in a 17-ally-lamino derivative, maximum tolerated dose (MTD) of BC194 was >130
tanespimycin, that has similar affinity to Hsp90 (K d 1.3 mg/kg, i.v., whereas the MTD of borrelidin was found to be
mM) but increased molecular weight (MW 586) compared in the range of 5–15 mg/kg, i.v. [35].
to the lead geldanamycin (Kd
Figure 5
Comparison of semisynthetic and synthetic biology-based lead optimization approaches for natural products, exemplified by optimization of
ansamycin-based Hsp90 inhibitors. Binding efficiency indices (BEI) = pK d/MW (kDa). For a small-molecule inhibitor of MW 500 and Kd of 1.0
nM, its BEI = 18 [31].
Conclusions parenteral drug candidate in parallel to an oral drug
Currently, the majority of industrial efforts in drug dis- candidate, in some cases before the availability of an oral
covery, especially in large pharmaceutical companies, are drug candidate and second, to consider the development of
being invested in discovering small molecule, orally bioa- a therapeutic antibody wherever possible in parallel to a
vailable drugs that comply with the ‘rule-of-five’. How- small-molecule drug candidate. These are particularly
ever, only half of all FDA-approved small-molecule drugs relevant for effort against ‘first-in-class’ drug targets and/
are both used orally and compliant with the ‘rule-of-five’ or particularly challenging targets such as proteases and
[3]. In other words, nearly half of all small-molecule drugs those involving protein–protein interactions, where, to
are either not used for oral administration or do not comply discover an orally bioavailable drug candidate can be more
with the ‘rule-of-five’. This incompatibility between huge problematic and slow.
industrial effort and the lower percentage of relevant drugs
in clinical use has almost certainly something to do with In addition, more effort should be invested in natural
economic considerations — oral drugs are more likely to be product research. Novel technologies such as synthetic
‘blockbusters’. Hopefully this is changing with the emer- biology may address many of the challenging issues of
gence of blockbuster biologicals and target-based antic- natural product-based drug discovery, for example struc-
ancer drugs [36], and the drive to develop drugs for niche tural complexity and synthetic feasibility. Synthetic biology
indications [37]. approaches can in practice be simple and straight-forward
in analog preparation. Genetic alterations can be made in
It seems to us that in order to increase drug discovery such a way as to biosynthesize analogs with predesigned
productivity and to address seriously un-met medical needs structural modifications, in very much the same way as
a programmatic approach to bioavailability could be achieved by conventional medicinal chem-istry. Unlike the
adopted. That is first, to consider the development of a total synthesis of complex natural pro-
starter unit biosynthesis. Angew Chem Int Ed Engl 2005, for Cancer Research Annual Meeting Proceedings: 2007 April
44:4757-4760. 14– 18; Philadelphia (PA) [abstract nr 5724]. 2007.
A good example of how genetically engineered biosynthesis can be
used effectively to make analogs of a structurally complex natural 34. Tian ZQ, Liu Y, Zhang D, Wang Z, Dong SD, Carreras CW, Zhou
product lead. Y, Rastelli G, Santi DV, Myles DC: Synthesis and biological
activities of novel 17-aminogeldanamycin derivatives. Bioorg
30. Sheridan RM, Gregory MA, Petkovic H, Gaisser S, Lill RE, Ward Med Chem 2004, 12:5317-5329.
MJ, Kaja AL, Beckman CH, Challis IR, Moss SJ et al.: Brain-
selective mTOR inhibitors as potential therapeutics for 35. Wilkinson B, Gregory MA, Moss SJ, Carletti I, Sheridan RM, Kaja
glioblastoma multiforme. In American Association for Cancer A, Ward M, Olano C, Mendez C, Salas JA et al.: Separation of
Research Annual Meeting Proceedings: 2006 April 1–5; anti-angiogenic and cytotoxic activities of borrelidin by
Washington, DC [abstract nr 5959]. 2006. modification at the C17 side chain. Bioorg Med Chem Lett 2006,
16:5814-5817.
31. Zhang MQ, Sheridan RM: 39-Desmethoxyrapamycin,
compositions and methods of use thereof. US 7,183,289 B2; 36. Reichert JM, Valge-Archer VE: Development trends for
2006. monoclonal antibody cancer therapeutics. Nat Rev Drug Discov
2007, 6:349-356.
32. Abad-Zapatero C, Metz JT: Ligand efficiency indices as
guideposts for drug discovery. Drug Discov Today 2005, 37. Owens J: 2006 drug approvals: finding the niche. Nat Rev Drug
10:464-469. Discov 2007, 6:99-101.
33. Martin CJ, Gaisser S, Vousden WA, Sheehan LJ, Foster T, 38. Wishart DS, Knox C, Guo AC, Shrivastava S, Hassanali M,
Sheridan RM, Nur-E-alam M, Coates N, Moss SJ, Beckmann C Stothard P, Chang Z, Woolsey J: DrugBank: a comprehensive
et al.: Towards ‘best-in-class’ Hsp90 inhibitors: design, resource for in silico drug discovery and exploration. Nucleic
biosynthesis and pre-clinical profiles. In American Association Acids Res 2006, 34:D668-D672.
Current Opinion in Biotechnology 2007, 18:478–488 www.sciencedirect.com