7.14 Fungi and Fungal Disease
7.14 Fungi and Fungal Disease
7.14 Fungi and Fungal Disease
419
420 Fungi and Fungal Disease
Figure 1 Micro- and macroscopic images of pathogenic fungi. (a) Electron micrograph of Candida albicans in yeast form and
(b) mixed yeast–mycelium form from a urine sample (per acid stained). (c) Methenamine silver staining of clinical samples
highlights the hyphal growth of the filamentous pathogen Aspergillus fumigatus invading lung tissue. The spore-forming (conidial
head) structure of A. fumigatus is also highlighted (d) within invaded lung tissue, and (e) in more detail as an electron micrograph
following laboratory culture. Extensive invasion can lead to macroscopic emergence of Aspergillus biomass, as shown by
(f) visible growth on lung tissue. Colony presentation of (g) Aspergillus and (h) C. albicans on agar plates. (a–d; f–h: reproduced
by permission of Dr David Ellis, School of Molecular & Biomedical Science, University of Adelaide and the Kaminski digital image
library; e: reproduced by permission of Dr Jean-Paul Latge of the Pasteur Institute.)
Table 2 Subcutaneous mycoses. The causative pathogen is emboldened for the clinical case highlighted
Reproduced by permission of Dr David Ellis, School of Molecular & Biomedical Science, University of Adelaide.
agent and its metabolic products. The causative pathogen is usually a filamentous dermatophyte (Table 3), from which
Trychophyton (usually T. mentagrophytes or T. rubrum), Epidermophyton, and Microsporium species are by far the most
common pathogens, although superficial candidiasis is not uncommon.7 These pathogens infect various areas of the
body, from which the infection description is derived (e.g., Tinea capitis (scalp and/or hair), Tinea corporis (a dermatophyte
Fungi and Fungal Disease 423
Table 3 Cutaneous mycoses. The causative pathogen is emboldened for the clinical case highlighted
Reproduced by permission of Dr David Ellis, School of Molecular & Biomedical Science, University of Adelaide.
infection of the trunk, legs, and arms), T. pedis (feet), T. mannum (hands), T. unguium (nails)). The vast majority of
infections are mild, and readily diagnosed by microscopy and culture of samples, following symptoms of localized
irritation, lesions, and inflammation. These infections respond well to topical therapies such as azole and allylamine
drugs (in particular, terbinafine).8 In cases where the infection is showing signs of deeper and wider invasion into
peripheral tissues, oral systemic therapy is required, usually by itraconazole or terbinafine.9,10 Of these infections,
T. unguium or onychomycosis is the least well addressed in terms of therapy.8 Although generally a mild infection due to
dermatophytes and also Candida species, it can spread to case discomfort and localized disfigurement. Effective therapy
is restricted to long-term (3–6 month) oral terbinafine or itraconazole. Topical agents have very limited efficacy,8,10 but
would be preferred to limit systemic exposure of the oral agents, where liver tests are often required to monitor possible
side effects.
Reproduced by permission of Dr David Ellis, School of Molecular & Biomedical Science, University of Adelaide.
Figure 2 Tissue invasion by Candida albicans in kidney: (a) colony formation on tissue surface highlighted and (b) an
apsergilloma (fungal ball) caused by Aspergillus fumigatus (excised from lung).
new systemic antifungal agents. It is this area of antifungal drug discovery that is of particular challenge to the
medicinal chemist. Diagnosis of fungal infections and treatment are described in this chapter in more detail, together
with emerging targets and chemistry starting points for the discovery of new antifungal drugs.
resistant to conventional antifungal therapy, and may cause severe morbidity and mortality in immunocompromised
hosts. Emerging pathogens are increasingly reported as causing invasive mycoses refractory to amphotericin B therapy,
although the new agents caspofungin (Cancidas) and voriconazole in particular have been shown to have impressive
potency against these species in vitro and in the clinic.25–30 Pfaller and Diekema provide an excellent review of new and
emerging fungal infections together with changes in epidemiology and treatment modalities.25
* esophageal candidiasis (frequently associated with AIDS and severe immunosuppression following treatment for
leukemia or solid tumors);
* gastrointestinal candidiasis;
* pulmonary candidiasis;
* peritonitis (usually from colonization of indwelling catheters used for peritoneal dialysis or gastrointestinal
perforation);
* urinary tract and renal candidiasis (pyelonephritis);
* meningitis;
* hepatic and hepatosplenic candidiasis;
* endocarditis, myocarditis, and pericarditis;
* candidemia (Candida septicemia) and disseminated candidiasis.
The complications associated with Candida infections is compounded by the emergence of non-albicans species as
both a colonizer and a pathogen. More than 17 different species of Candida have been identified as etiologic agents
of bloodstream infections, with approximately 95% of these being caused by four species: C. albicans, C. glabrata,
C. parapsilosis, and C. tropicalis.25 The greater efficacy of fluconazole as a prophylactic in transplant patients has indeed
led to a shift toward the less susceptible C. glabrata species as the predominant pathogen in such immunocompromised
patients, as well as more emerging opportunistic pathogens.25 The inherent resistance of a few non-albicans species to
commonly used systemic antifungals such as fluconazole and amphotericin B can pose a therapeutic challenge in the
management of candidaemia. Encouragingly, newer drugs, including broad-spectrum triazoles such as voriconazole, and
candins such as caspofungin, are emerging as therapeutic alternatives for non-albicans infection treatment, as discussed
elsewhere (see 7.15 Major Antifungal Drugs).
Fungi and Fungal Disease 427
7.14.2.2 Aspergillus
Aspergillosis is a spectrum of diseases of humans (and animals) caused by members of the genus Aspergillus. Aspergillus
fumigatus is overwhelmingly the predominant pathogenic species. The type of disease and severity depends upon the
physiologic state of the host and the species of Aspergillus involved. The etiological agents are ubiquitous, and include
A. fumigatus, A. flavus, A. niger, A. nidulans, and A. terreus.
Numerous virulence factors have been associated with aspergillosis, including phospholipases and proteases to
facilitate adhesion and invasion,38,42 and the biosynthesis of factors that enable evasion of limited host defenses in the
immunosuppressed patient.41 As with Candida infections, the clinical manifestations resulting from Aspergillus virulence
are varied, although pulmonary infection and aspergilloma caused by the saprophytic colonization of preformed cavities
(Figure 2b) is the most common. Acute invasive pulmonary aspergillosis (usually a result of prolonged neutropenia,
especially in leukemia patients or in bone marrow transplant recipients) generates symptoms similar to acute bacterial
pneumonia. Dissemination can lead to tissue-specific infections. Abscesses may occur in the brain (cerebral asper-
gillosis), kidney (renal aspergillosis), heart (endocarditis, myocarditis), bone (osteomyelitis), and gastrointestinal tract.
Ocular lesions (mycotic keratitis, endophthalmitis, and orbital aspergilloma) manifest themselves as erythematous
papules or macules with progressive central necrosis.
diagnosis is often the favored option, even when the administered agent has severe side effects, such as those
associated with amphotericin B. However, in recent years, the choice of antifungal drugs for systemic diseases has
increased with the more widespread use of fluconazole for C. albicans infections (in particular maintenance
therapy), to voriconazole (particularly for Aspergillus infections, and broad-spectrum use), and caspofungin (which is a
better-tolerated infused agent compared with amphotericin).58 A comprehensive general review of the diagnosis (and
management) of fungal infections has been published.3
Much has been achieved recently in the improvement of laboratory diagnosis of mycoses, such as advances in blood
culture systems, and the development of new biochemical assays, antigen detection assays, and molecular methodo-
logies.59 More standardized susceptibility testing guidelines provide for better therapeutic interventions. In an era of
economic cutbacks in healthcare, future challenges include the development of cost-effective and technically
simplified systems, which will provide for the early detection and identification of common and emerging fungal
pathogens. The conventional diagnostic methods such as culture are often slow and lack sensitivity and specificity,
resulting in additional alternative diagnostic assays. Among the most promising new techniques are the detection of
fungal DNA, metabolites (see following sections), and serology. Fungal DNA can be detected with high sensitivity and
specificity when performed with specimens from sterile sites such as blood.60,61 Polymerase chain reaction (PCR)
assays can be used to detect a broad range of fungal pathogens, and combined with species identification, although a
lack of standardization has restricted its use. The sero-diagnosis of invasive fungal infections has become an important
tool in the management of invasive fungal infections. Both serology and PCR can be used to monitor the response to
antifungal therapy.60–62 These newer approaches are generally used to help confirm the diagnosis of systemic infection;
however, their impact on mortality may be greatest when they are used to screen high-risk patients.61 While the
mainstay of infectious disease diagnosis remains in microbiological culture with sensitivity follow-up, significant steps
in providing rapid diagnosis in commercialized and standardized assays have been achieved.
particular mannan-based metabolites derived from the cell wall of Candida species.70,71 The Platelia Candida sandwich
ELISA represents a sensitive serological method, utilizing a monoclonal antibody that recognizes the sequences of
linked oligomannoses present in a large number of mannoproteins extracted from different Candida species. An issue
of concern regarding the Platelia Aspergillus antigen/antibody assay is the variable reactivity of different Candida
species.72
Butenafine Topical
Naftifine Topical
Clotrimazole Topical
Econazole Topical
Miconazole Topical
Oxiconazole Topical
Sulconazole Topical
Terconazole Topical
Tioconazole Topical
Micafungin Intravenous
Anidulafungin Intravenous
Pimaricin Ophthalmic
Haloprogin Topical
Tolnaftate Topical
Undecylenate Topical
Fungi and Fungal Disease 431
Primary metabolism
Acetyl-CoA
Squalene
Squalene epoxide
Lanosterol
(nonfunctional sterol)
HO
Fungi Human
Amphotericin
Cytochrome
P450
demethylase
HO
HO
Ergosterol (functional Azoles Cholesterol (functional
sterol in fungi) sterol in humans)
Figure 3 Sterol biosynthesis pathway and target sites for antifungal drugs.
creams and pessaries, and oral fluconazole is effective for vaginal thrush.3 In light of these infections being in general
well addressed, and thereby the need for the involvement of the medicinal chemist to discover new agents being
reduced, the greater challenge of finding and prescribing antifungal drugs for the treatment of systemic infections by
Candida, Aspergillus, and emerging pathogens will be focused on here.
overall safety of newer antifungal drugs. Comprehensive reviews of treatment modalities and clinical pharmacology of
currently available antifungal drugs have been published, and are very useful reference points.82–85
The treatment of systemic fungal infections has been an evolutionary process as newer agents have become available
and comparative trials for efficacy and safety have proceeded. Generally, amphotericin led the way as a broad-spectrum
intravenous agent, which has efficacy but very poor tolerance. Azole antifungals represent the next major advance and
have added to the antifungal arsenal. Early azoles such as ketoconazole showed significant side effects, and were rapidly
supplanted by fluconazole as a highly safe systemic antifungal, with good efficacy against the predominant pathogen
C. albicans. Further progress was made with broader-spectrum azole itraconazole, although this met with limitations due
to reduced tolerance and unpredictable pharmacokinetics relative to fluconazole. In more recent years the introduction
of a new generation of azoles and glucan synthase-inhibiting candin antifungals have substantially broadened
therapeutic options for clinicians. The profile of the leading antifungals amphotericin and fluconazole and new agents
are reviewed in more detail below.
7.14.5.3.1 Amphotericin
Amphotericin was first isolated from Streptomyces nodosus in 1955. It is an amphoteric compound composed of a
hydrophilic polyhydroxyl chain along one side and a lipophilic polyene hydrocarbon chain on the other (see 7.15 Major
Antifungal Drugs). This parenternal drug was the gold standard antifungal for neutropenic patients with invasive
candidiasis for many years. The three lipid formulations are liposomal amphotericin B (AmBisome), amphotericin B
lipid complex (Abelcet), and amphotericin B colloidal dispersion (Amphocil). These formulations are more expensive
than conventional amphotericin B, but generally have the advantage of less infusional and renal toxicity, and provide the
opportunity to administer higher doses of amphotericin B.86 Animal studies suggest similar efficacy between liposomal
amphotericin B and amphotericin B lipid complex, as do retrospective comparisons of patient outcomes.82
Amphotericin and polyenes in general operate by inhibiting sterol function in the fungal cell membrane, to exert an
antifungal effect (Figure 3; for a review, see Polak87). Amphotericin achieves functional ergosterol blockade by directly
binding this functional sterol, to induce dysfunctional membrane properties, including leakage of intracellular
potassium, magnesium, sugars, and metabolites, and then cellular death. The mechanism of action is the same for all
the preparations, and is due to the intrinsic antifungal activity of amphotericin B. Target binding is only moderately
selective with respect to cholesterol binding, in particular at cholesterol-rich sites such as the kidney tubules, which
underpins the mechanistic toxicity of amphotericin B.15,88 In addition to the poor tolerance, the pharmacokinetic
profile is poorly defined, and administration is limited to intravenous infusion. Amphotericin B is not bioavailable
following oral administration.87,88 Generally speaking, amphotericin B has a very broad range of activity, and is active
against most pathogenic fungi. Notable exceptions include the emerging pathogens Trichosporon beigleii and Fusarium
species.89,90 It is testament to the rapid progression and high mortality and morbidity of systemic fungal infections that
such a poorly tolerated drug as amphotericin has been the gold standard for antifungal therapy for so many years.
7.14.5.3.2 Fluconazole
Fluconazole is an extremely safe and effective drug for the treatment and prevention of C. albicans infections
(see 7.15 Major Antifungal Drugs). Fluconazole also has the advantages of oral and intravenous administration, and has
highly predictable pharmacokinetics.91,92 However, like all azoles, it is fungistatic and not fungicidal against C. albicans,
and has a very limited spectrum with no efficacy against molds. Furthermore, some Candida species are intrinsically
resistant to fluconazole. Almost all C. krusei are resistant, and approximately 50% of C. glabrata isolates are resistant or
have intermediate dose-dependent susceptibility to fluconazole.3,25 Until recently, oral fluconazole has been the drug of
choice for controlling oropharyngeal candidiasis in AIDS patients, and for prophylactic use (predominantly transplant
and cancer patients).93,94 The success of azoles and polyenes in addressing a high medical need has been significant,
despite their mutually exclusive limitations. Both drug classes operate by inhibiting sterol function in the fungal cell
membrane, to exert an antifungal effect (see Figure 3).87 However, azoles indirectly target ergosterol function via
fungal-specific inhibition of 14-sterol demethylase, to block the synthesis of functional ergosterol from nonfunctional
lanosterol. No significant effect on human/mammalian cholesterol biosynthesis is achieved in vivo as a result of this
activity, which underpins the mechanistic safety of azoles and fluconazole in particular.95,96
infections. Voriconazole is an extended-spectrum triazole that is fungicidal for many filamentous fungi, including
Aspergillus, Scedosporium, Fusarium, and Paecilomyces, and is active against all species of Candida.97,98 Clinical trials have
shown that treatment with voriconazole cleared Candida from the blood as quickly as amphotericin B, with a lower
incidence of treatment-related adverse events. Subsequently, it was approved for the first-line treatment of invasive
aspergillosis and as salvage therapy for fungal infections caused by the pathogens Scedosporium apiospermum and Fusarium
species. More recently, voriconazole was approved for use in treating esophageal candidiasis.98 This is a reflection of its
impressive activity against filamentous fungi, in particular Aspergillus, where it exerts fungicidal activity, as seen in a
variety of in vivo and in vitro models.99,100 Voriconazole is given either by the oral or the intravenous route. Unlike
fluconazole, voriconazole is not renally cleared in light of its higher lipophilicity. Clearance is cytochrome P450-
mediated. The effects of voriconazole on cytochrome P450-mediated metabolism means that clinicians must be aware
of drug–drug interactions. The clinical pharmacokinetics of voriconazole have been described in detail by Purkins
et al.101 Voriconazole represents the most advanced and widely used third-generation azole antifungal, with
posaconazole and ravuconazole (see 7.15 Major Antifungal Drugs) also showing promising broad-spectrum activity.
The most obvious rational starting point for a drug discovery program would be to target the cell wall of fungal
pathogens, in light of its essentiality for pathogen viability, and total absence in host cells, despite the commonality of
their eukaryotic origin. This approach has been aggressively targeted by drug researchers, as seen by the massive success
demonstrated with the penicillin and cephalosporin series of antibacterial antibiotics.102 Despite the complex nature of
fungal cell walls (a highly structured complex of mannan, b-glucan, and n-acetyl glucosamine (chitin)-based polymers103)
and their biosynthesis, highlighting a host of target areas for antifungal drug discovery, it is arguably disappointing that
relatively little success has been made (with one exception) in terms of novel drug series. Numerous attempts and
screens to find inhibitors of chitin synthases have been undertaken, and inhibitors such as the natural products
nikkomycin and poloxin have been discovered.104 However, these have not led to clinical candidates, and other more
amenable inhibitors to a drug discovery program have not been forthcoming. Similarly, the pradamicins, which bind to
mannans in the cell wall, have proven to be of limited value as an antifungal drug lead.102 These are far from being
antifungal drugs due to inherent limitations in potency and unfavorable physicochemical properties that limit their
systemic bioavailability. Despite the substantial effort of medicinal chemists and biotransformation scientists to improve
the pradamicins, little of therapeutic use has emerged from these starting points. One notable exception
in the search for new drugs that target the cell wall of fungi is the discovery, development, and successful com-
mercialization of the echinocandin and pneumocandin drugs (reviewed by Denning,105 and discussed in detail in
7.15 Major Antifungal Drugs). These agents inhibit the glucan synthase enzyme responsible for the b-glucan polymer, a
major constituent of the cell wall. Clinical trials have highlighted amphotericin-like efficacy, in empirical-based therapy,
with superior tolerance.86 The most successful of these to date is caspofungin (Cancidas), which is a semisynthetic
analog of a pneumocandin lead.106 Caspofungin has subsequently progressed through clinical development, and met
with considerable success for the empirical therapy of fungal infections in febrile neutropenic patients. Approval was
based on results from the largest prospective antifungal empirical therapy trial published to date in neutropenic patients,
where caspofungin was as effective as amphotericin (Ambisome) but with fewer side effects.86 This represents a
breakthrough in supplanting the gold standard amphotericin B with a safer agent for many deep-seated life-threatening
infections. Caspofungin is also active against aspergillosis,107 and has been used successfully in combination with
voriconazole for this life-threatening mycoses.108 Caspofungin is currently the most widely used glucan synthase
inhibitor for the treatment of serious fungal infections. Micafungin (Mycamine), has a very similar profile, and has been
approved for prophylaxis against Candida infection as well as esophageal candidasis. Other agents in this class are also
showing great promise. Clinical studies with anidulafungin (Eraxis) have been encouraging,109 where recent Phase III
studies showed it to be superior to fluconazole against invasive candidiasis, yet with a similar safety profile.
However, a limitation of caspofungin, and the candin class of antifungal drugs in general, is the lack of an oral route of
administration. Efforts by medicinal chemists and drug discoverers are ongoing to find new small molecule templates for
the inhibition of glucan synthase (e.g., see Onishi110), although this has currently met with limited success.
7.14.6 Antifungal Drug Discovery and Development: Future Directions and New
Modes of Action
7.14.6.1 Antifungal Compounds in Clinical Development
7.14.6.1.1 Isoleucyl-tRNA synthetase inhibitors (ITRS)
ITRS is the target of icofungipen (formerly PLD-118, see Figure 4), which is a highly novel clinical development
compound for the treatment of Candida infections.111,112 Icofungipen is a synthetic derivative of the naturally occurring
434 Fungi and Fungal Disease
Isofungipen Cispentacin
b-amino acid cispentacin (derived from Bacillus cereus and Streptomyces setonii) that blocks isoleucyl-tRNA synthetase,
resulting in the inhibition of protein synthesis and growth of fungal cells. Icofungipen, like fluconazole, has the
advantage of renal clearance to drive highly predictive pharmacokinetics and avoid drug–drug interactions as a result
of hepatic cytochrome P450-based clearance. Like fluconazole, it is also a small, highly soluble and orally bioavailable
molecule. Although active against a number of Candida species, including fluconazole-resistant strains, it has only
moderate antifungal potency in vitro (minimum inhibitory concentration (MIC) ¼ 8–64 mg mL 1 with no fungicidal
activity). However, this compound showed promising efficacy in models of candidiasis (C. albicans), which was
similar to that seen with amphotericin B.112,113 Icofungipen showed a clear pharmacokinetic–pharmacodynamic
relationship, with no overt safety alerts, as shown by a well-tolerated profile against numerous toxicity markers. In light
of promising preclinical data, icofungipen has progressed to Phase II for oropharyngeal comparison with fluconazole,
where efficacy was seen, albeit less than that demonstrated for fluconazole, and with a more frequent dosing regimen.114
Unlike most natural-product antifungal leads, the small molecular size and simple structure of icofungipen make it
an attractive starting point for the medicinal chemist, since added substituents to increase potency may not
compromise the physicochemical properties, and thus hamper the favorable oral bioavailability and phamacokinetics.115
Serine
Palmitoyl-CoA
Ketodihydrosphingosine
Dihydrosphingosine
Fungi Humans
Phytosphingosine Sphingosine
Fatty acid
Ceramide Ceramide
AureobasidinA
Inositol-P-ceramide Sphingomyelin
Mannosyl-inositol-P-ceramide Gangliosides
Mannosyl-(inositol)2-ceramide Cerebrosides
O
N N
O N
O
O O
O O N
N O
O
N N
O N
O
Figure 6 Aureobasidin A.
436 Fungi and Fungal Disease
antimicrobial agents. This approach has also led to the discovery of new antifungal targets of great potential. An
excellent review of natural-product antifungal leads, clinical candidates, and details of their mode of action has been
done by Vicente et al.117
mCoA
NMT/mCoA binary Peptide substrate (e.g., ARF)
complex
Target for benzothiazole
inhibitors
NMT-Myr-peptide
Myristoylated peptide CoA
Figure 7 The catalytic cycle of NMT and the site of inhibition for benzothiazole and peptide-based inhibitors.
Fungi and Fungal Disease 437
Compound Properties
CP-123,457
UK-370,485
UK-362,091
Figure 8 Three-dimensional image of benzothiazole occupation of the Candida albicans NMT active site cleft (peptide-binding
site) as derived from x-ray diffraction of NMT-inhibitor co-crystals.
was found to have potent antifungal activity against Candida (all species) and Aspergillus, with a high therapeutic index
relative to human cell line cytotoxicity. A genetic approach utilizing the yeast Saccharomyces cerevisiae was used to
identify the target of antifungal compounds. Three lines of evidence showed that UK-118,005 inhibited cell growth by
targeting RNA Pol III in yeast. First, a dominant mutation in the g domain of Rpo31p, the largest subunit of RNA Pol
III, conferred resistance to the compound. Second, UK-118,005 rapidly inhibited tRNA synthesis in wild-type cells but
438 Fungi and Fungal Disease
Tyr 225
Phe 115 IIe 111 WAT 762 Glu 109
Leu 415
WAT 887
WAT 898
Tyr 354
Phe 240
Figure 9 Two-dimensional representation of benzothiazole (UK-370,485) interactions with active site amino acid residues in
the Candida albicans NMT.
Asp − Cryptococcus
Aspergillus
Candida resistant species
Human
Ser − Cryptococcus
Aspergillus
Candida resistant species
Human
Figure 10 Interaction of key amino acid residues of Candida albicans NMT with a benzothiazole highlighting inhibitor. Van der
Waals interaction of the benzothiazole moiety with the hydrophobic Ile111 and Phe339 residues of C. albicans NMT. The
equivalent polar residues (incompatible with respect to forming interactions with the benzothiazole) from other species are
highlighted.
not in UK-118,005-resistant mutants. Third, in biochemical assays, UK-118,005 inhibited tRNA gene transcription
in vitro by the wild-type but not the mutant Pol III enzyme. Further examination showed UK-118,005 to inhibit
RNA Pol III transcription systems derived from C. albicans and human cells. The identification of these inhibi-
tors demonstrates that RNA Pol III can be targeted by small synthetic molecules, to exert a potent antifungal
Fungi and Fungal Disease 439
effect. UK-118,005 showed broad-spectrum antifungal activity (MIC ¼ 3.1–12.5 mg mL–1 against Candida species and
A. fumigatus (Table 8). Unfortunately, this compound showed little selectivity for the fungal/human enzyme in
controlled kinetic assays, indicating the need for an extensive medicinal chemistry campaign to generate a fungal
selective inhibitor.
−
ATP + HCO3 ADP + Pi
Biotin carboxylase
BCCP BCCP-COO−
Acetyl-CoA Malonyl-CoA
Figure 11 Acetyl-CoA carboxylase (ACCase) catalysis of malonyl-CoA formation from acetyl-CoA. Catalysis is a multistep
process, taking place at different catalytic sites of a single multifunctional protein. The reaction proceeds via the ATP-dependent
carboxylation of a biotin group at the biotin carboxylase domain of ACCase, with the transfer of this (by a biotin carboxyl carrier
peptide – BCCP) to the carboxyl transferase domain of ACCase.
OMe
OH
O O
O OH
OH
OMe
Figure 12 Soraphen A.
440 Fungi and Fungal Disease
H3C CH3
R O
O OH O
HO O HO O
CH3 CH3
Figure 13 Antifungal sordarins. (a) Core template, (b) parent sordarin (R group), and (c) GR1305402 (R group).
fungi, and therefore the great degree of similarity between the fungal and mammalian protein synthesis machineries.
Two soluble elongation factors show some fungal specificity: EF3, a factor that is required by fungal ribosomes only, and
EF2, which has been demonstrated to possess at least one functional distinction from its mammalian counterpart. The
sordarins are the most important family of antifungal agents acting at the protein synthesis level. Compounds in this
class inhibit in vitro translation in C. albicans, C. tropicalis, C. kefyr, and C. neoformans, to varying degrees.117 The lack of
activity of the sordarins against C. krusei, C. glabrata, and C. parapsilosis, in comparison with their extremely high levels of
potency against C. albicans, suggests that these compounds have a highly specific binding site, which may also be the
basis for the greater selectivity of these compounds in inhibiting fungal, but not the mammalian, protein synthesis. The
most advanced inhibitors of fungal protein biosynthesis are analogs of the natural product lead sordarin lead, GR135402
(see Figure 13).146,147 Its spectrum of activity includes C. albicans, C. tropicalis, and C. neoformans, where impressive
antifungal activity has been observed in vitro (MICo1 mg mL 1 in many cases), but not in other Candida or Aspergillus
species, which severely restricts its potential as a quality lead. Efficacy has been seen in animal models,147 although at
high dose and predominantly via nonoral routes, reflecting the potential for rapid clearance and limited oral
bioavailability.
References
1. San-Blas, G.; Calderone, R., Eds. Pathogenic Fungi. Structural Biology and Taxonomy; Caister Academic Press: Norwich, UK, 2004; Vol. 1.
2. Brandt, M. E.; Warnock, D. W. J. Chemother. 2003, 15, 36–47.
3. Warnock, D.; Richardson, M. Fungal Infection Diagnosis and Management, 3rd ed.; Blackwell: Oxford, 2003.
4. Dutriaux, C.; Saint-Cyr, I.; Desbois, N.; Cales-Quist, D.; Diedhou, A.; Boisseau-Garsaud, A. M. Ann. Dermatol. Venereol. 2005, 132, 259–262.
5. McGinnis, M. R.; Nordoff, N.; Li, R. K.; Pasarell, L.; Warnock, D. W. Med. Mycol. 2001, 39, 369–371.
6. Al-Abdely, H. M. Curr. Opin. Infect. Dis. 2004, 17, 527–532.
7. Allelo, A.; Hay, R. J. Medical Mycology. In Topley & Wilson’s Microbiology and Infectious Infections, 9th ed.; Edward Arnold: London, 1997; Vol. 4.
8. Gupta, A. K.; Cooper, E. A.; Ryder, J. E.; Nicol, K. A.; Chow, M.; Chaudhry, M. M. Am. J. Clin. Dermatol. 2004, 5, 225–237.
9. Baran, R.; Gupta, A. K.; Pierard, G. E. Expert Opin. Pharmacother. 2005, 6, 609–624.
10. Gupta, A. K.; Ryder, J. E.; Lynch, L. E.; Tavakkol, A. J. Drugs Dermatol. 2005, 4, 302–308.
11. Prabhu, R. M.; Bonnell, M.; Currier, B. L.; Orenstein, R. Clin. Infect. Dis. 2004, 39, e74–e77.
12. Cortez, K. J.; Walsh, T. J.; Bennett, J. E. Clin. Infect. Dis. 2003, 36, 1619–1622.
13. Proia, L. A.; Tenorio, A. R. Antimicrob. Agents Chemother. 2004, 48, 2341.
14. Groll, A. H.; Shah, P. M.; Mentzel, C.; Schneider, M.; Just-Nuebling, G.; Huebner, K. J. Infect. 1996, 33, 23–32.
15. Bodey, G. Candidiasis. Pathogenesis, Diagnosis, and Treatment, 2nd ed.; Raven Press: New York, 1993.
16. Cunha, B. A. Crit. Care Clin. 1998, 14, 263–282.
17. Selvarangan, R.; Bui, U.; Limaye, A. P.; Cookson, B. T. J. Clin. Microbiol. 2003, 41, 5660–5664.
18. Pfaller, M. A. Clin. Infect. Dis. 1996, 22, S89–S94.
19. Hajjeh, R. A.; Sofair, A. N.; Harrison, L. H.; Lyon, G. M.; Arthington-Skaggs, B. A.; Mirza, S. A.; Phelan, M.; Morgan, J.; Lee-Yang, W.; Ciblak,
M. A. et al. J. Clin. Microbiol. 2004, 42, 1519–1527.
20. Hobson, R. P. J. Hosp. Infect. 2003, 55, 159–168.
21. Fleming, R. V.; Walsh, T. J.; Anaissie, E. J. Infect. Dis. Clin. North Am. 2002, 16, 915–933.
22. Walsh, T. J.; Groll, A.; Hiemenz, J.; Fleming, R.; Roilides, E.; Anaissie, E. Clin. Microbiol. Infect. 2004, 10, 48–66.
23. Groll, A. H.; Walsh, T. J. Clin. Microbiol. Infect. 2001, 7, 8–24.
24. Baddley, J. W.; Moser, S. A. Clin. Lab. Med. 2004, 24, 721–735.
25. Pfaller, M. A.; Diekema, D. J. J. Clin. Microbiol. 2004, 42, 4419–4431.
Fungi and Fungal Disease 441
26. Garbino, J.; Ondrusova, A.; Baglivo, E.; Lew, D.; Bouchuiguir-Wafa, K.; Rohner, P. Scand. J. Infect. Dis. 2002, 34, 701–703.
27. Mattei, D.; Mordini, N.; Lo Nigro, C.; Gallamini, A.; Osenda, M.; Pugno, F.; Viscoli, C. Mycoses 2003, 46, 511–514.
28. Lewis, R. E.; Wiederhold, N. P.; Klepser, M. E. Antimicrob. Agents Chemother. 2005, 49, 945–951.
29. Schaenman, J. M.; Digiulio, D. B.; Mirels, L. F.; McClenny, N. M.; Berry, G. J.; Fothergill, A. W.; Rinaldi, M. G.; Montoya, J. G. J. Clin. Microbiol.
2005, 43, 973–977.
30. Husain, S.; Munoz, P.; Forrest, G.; Alexander, B. D.; Somani, J.; Brennan, K.; Wagener, M. M.; Singh, N. Clin. Infect. Dis. 2005, 40, 89–99.
31. McNeil, M. M.; Nash, S. L.; Hajjeh, R. A.; Phelan, M. A.; Conn, L. A.; Plikaytis, B. D.; Warnock, D. W. Clin. Infect. Dis. 2001, 33, 641–647.
32. Rees, J. R.; Pinner, R. W.; Hajjeh, R. A.; Brandt, M. E.; Reingold, A. L. Clin. Infect. Dis. 1998, 27, 1138–1147.
33. Ghannoum, M. A.; Abu-Elteen, K. H. Mycoses 1990, 33, 265–282.
34. Yang, Y. L. J. Microbiol. Immunol. Infect. 2003, 36, 223–228.
35. Rinaldi, M. G. Biology and Pathogenicity of Candida Species. In Candidiasis; Bodey, G., Ed.; Raven Press: New York, 1993, pp 1–20.
36. Bignell, E.; Rogers, T.; Haynes, K. Host Recognition by Fungal Pathogens. In Pathogenic Fungi: Host Interactions and Emerging Strategies for Control;
San-Blas, G., Calderone, R. A, Eds.; Caister Academic Press: Norwich, 2004; Chapter 1, pp 3–28.
37. Munro, C. A.; Bates, S.; Buurman, E. T.; Hughes, H. B.; Maccallum, D. M.; Bertram, G.; Atrih, A.; Ferguson, M. A.; Bain, J. M.; Brand, A.
et al. J. Biol. Chem. 2005, 280, 1051–1060.
37a. Mycology Online. http://www.mycology.adelaide.edu.au (accessed April 2006).
38. Shen, D. K.; Noodeh, A. D.; Kazemi, A.; Grillot, R.; Robson, G.; Brugere, J. F. FEMS Microbiol. Lett. 2004, 239, 87–93.
39. Fotedar, R.; Al-Hedaithy, S. S. Mycoses 2005, 48, 62–67.
40. Dolan, J. W.; Bell, A. C.; Hube, B.; Schaller, M.; Warner, T. F.; Balish, E. Med. Mycol. 2004, 42, 439–447.
41. Langfelder, K.; Streibel, M.; Jahn, B.; Haase, G.; Brakhage, A. A. Fungal Genet. Biol. 2003, 38, 143–158.
42. Kogan, T. V.; Jadoun, J.; Mittelman, L.; Hirschberg, K.; Osherov, N. J. Infect. Dis. 2004, 189, 1965–1973.
43. Varvitarian, S.; Smith, C. B. Pathogenesis, Host Resistance and Predisposing Factors. In Candidiasis. Pathogenesis, Diagnosis, and Treatment, 2nd ed.;
Bodey, G., Ed.; Raven Press: New York, 1993, pp 59–84.
44. Ashman, R. B.; Farah, C. S.; Wanasaengsakul, S.; Hu, Y.; Pang, G.; Clancy, R. L. Immunol. Cell Biol. 2004, 82, 196–204.
45. Speth, C.; Rambach, G.; Lass-Florl, C.; Dierich, M. P.; Wurzner, R. Mycoses 2004, 47, 93–103.
46. Polonelli, L.; Casadevall, A.; Han, Y.; Bernardis, F.; Kirkland, T. N.; Matthews, R. C.; Adriani, D.; Boccanera, M.; Burnie, J. P.; Cassone, A.
et al. Med. Mycol. 2000, 38, 281–292.
47. Clemons, K. V.; Calich, V. L.; Burger, E.; Filler, S. G.; Grazziutti, M.; Murphy, J.; Roilides, E.; Campa, A.; Dias, M. R.; Edwards, J. E., Jr., et al.
Med. Mycol. 2000, 38, 99–111.
48. Morelli, R.; Rosenberg, L. T. J. Immunol. 1971, 107, 476–480.
49. Romani, L. Curr. Opin. Microbiol. 1999, 2, 363–367.
50. Romani, L. J. Leukoc. Biol. 2000, 68, 175–179.
51. Kalfa, V. C.; Roberts, R. L.; Stiehm, E. R. Ann. Allergy Asthma Immunol. 2003, 90, 259–264.
52. Mencacci, A.; Cenci, E.; Bacci, A.; Montagnoli, C.; Bistoni, F.; Romani, L. Curr. Pharm. Biotechnol. 2000, 1, 235–251.
53. Standiford, T. J. Curr. Opin. Pulm. Med. 1997, 3, 81–88.
54. Traynor, T. R.; Huffnagle, G. B. Med. Mycol. 2001, 39, 41–50.
55. Boktour, M. R.; Kontoyiannis, D. P.; Hanna, H. A.; Hachem, R. Y.; Girgawy, E.; Bodey, G. P.; Raad, I. I. Cancer 2004, 101, 1860–1865.
56. Mansour, M. K.; Levitz, S. M. Curr. Opin. Microbiol. 2002, 5, 359–365.
57. Bellocchio, S.; Montagnoli, C.; Bozza, S.; Gaziano, R.; Rossi, G.; Mambula, S. S.; Vecchi, A.; Mantovani, A.; Levitz, S. M.; Romani, L.
J. Immunol. 2004, 172, 3059–3069.
58. Groll, A. H.; Walsh, T. J. Expert Opin. Investig. Drugs 2001, 10, 1545–1558.
59. O’Shaughnessy, E. M.; Shea, Y. M.; Witebsky, F. G. Infect. Dis. Clinics N. Am. 2003, 17, 135–158.
60. Verweij, P. E.; Meis, J. Transplant Infect. Dis. 2000, 2, 80–87.
61. McLintock, L. A.; Jones, B. L. Br. J. Haematol. 2004, 126, 289–297.
62. Kappe, R.; Rimek, D. Progr. Drug Res. 2003, 2, 39–57.
63. Pazos, C.; Ponton, J.; Del Palacio, A. J. Clin. Microbiol. 2005, 43, 299–305.
64. Odabasi, Z.; Mattiuzzi, G.; Estey, E.; Kantarjian, H.; Saeki, F.; Ridge, R. J.; Ketchum, P. A.; Finkelman, M. A.; Rex, J. H.; Ostrosky-Zeichner, L.
Clin. Infect. Dis. 2004, 39, 199–205.
65. Viscoli, C.; Machetti, M.; Gazzola, P.; De Maria, A.; Paola, D.; Van Lint, M. T.; Gualandi, F.; Truini, M.; Bacigalupo, A. J. Clin. Microbiol. 2002, 40,
1496–1499.
66. Sanguinetti, M.; Posteraro, B.; Pagano, L.; Pagliari, G.; Fianchi, L.; Mele, L.; La Sorda, M.; Franco, A.; Fadda, G. J. Clin. Microbiol. 2003, 41,
3922–3925.
67. Maertens, J.; Glasmacher, A.; Selleslag, D.; Ngai, A.; Ryan, D.; Layton, M.; Taylor, A.; Sable, C.; Kartsonis, N. Clin. Infect. Dis. 2005, 41, 9–14.
68. Maertens, J.; Verhaegen, J.; Lagrou, K.; Van Eldere, J.; Boogaerts, M. Blood 2001, 97, 1604–1610.
69. Boutboul, F.; Alberti, C.; Leblanc, T.; Sulahian, A.; Gluckman, E.; Derouin, F.; Ribaud, P. Clin. Infect. Dis. 2002, 34, 939–943.
70. Weiner, M. H.; Yount, W. J. J. Clin. Invest. 1976, 58, 1045–1053.
71. Lehmann, P. F.; Reiss, E. Mycopathologia 1980, 70, 89–93.
72. Yera, H.; Sendid, B.; Francois, N.; Camus, D.; Poulain, D. Eur. J. Clin. Microbiol. Infect. Dis. 2001, 20, 864–870.
73. Morace, G.; Pagano, L.; Sanguinetti, M.; Posteraro, B.; Mele, L.; Equitani, F.; D’Amore, G.; Leone, G.; Fadda, G. J. Clin. Microbiol. 1999, 37,
1871–1875.
74. Wahyuningsih, R.; Freisleben, H. J.; Sonntag, H. G.; Schnitzler, P. J. Clin. Microbiol. 2000, 38, 3016–3021.
75. Chen, S. C.; Halliday, C. L.; Meyer, W. Med. Mycol. 2002, 40, 333–357.
76. Kawazu, M.; Kanda, Y.; Goyama, S.; Takeshita, M.; Nannya, Y.; Niino, M.; Komeno, Y.; Nakamoto, T.; Kurokawa, M.; Tsujino, S. et al. Am. J.
Hematol. 2003, 72, 27–30.
77. Jordanides, N. E.; Allan, E. K.; McLintock, L. A.; Copland, M.; Devaney, M.; Stewart, K.; Parker, A. N.; Johnson, P. R.; Holyoake, T. L.; Jones, B.
L. Bone Marrow Transplant. 2005, 35, 389–395.
78. Loeffler, J.; Dorn, C.; Hebart, H.; Cox, P.; Magga, S.; Einsele, H. Diag. Microbiol. Infect. Dis. 2003, 45, 217–220.
79. Huang, D. B.; Ostrosky-Zeichner, L.; Wu, J. J.; Pang, K. R.; Tyring, S. K. Dermatol. Ther. 2004, 17, 517–522.
80. Petranyi, G.; Meingassner, J. G.; Mieth, H. Antimicrob. Agents Chemother. 1987, 31, 1558–1561.
81. Favre, B.; Ryder, N. S. Arch. Biochem. Biophys. 1997, 340, 265–269.
442 Fungi and Fungal Disease
82. Slavin, M. A.; Szer, J.; Grigg, A. P.; Roberts, A. W.; Seymour, J. F.; Sasadeusz, J.; Thursky, K.; Chen, S. C.; Morrissey, C. O.; Heath, C. H.
et al. Intern. Med. J. 2004, 34, 192–200.
83. Pappas, P. G.; Rex, J. H.; Sobel, J. D.; Filler, S. G.; Dismukes, W. E.; Walsh, T. J.; Edwards, J. E. Clin. Infect. Dis. 2004, 38, 161–189.
84. Rex, J. H.; Walsh, T. J.; Sobel, J. D.; Filler, S. G.; Pappas, P. G.; Dismukes, W. E.; Edwards, J. E. Clin. Infect. Dis. 2000, 30, 662–678.
85. Stevens, D. A.; Kan, V. L.; Judson, M. A.; Morrison, V. A.; Dummer, S.; Denning, D. W.; Bennett, J. E.; Walsh, T. J.; Patterson, T. F.; Pankey, G. A.
Clin. Infect. Dis. 2000, 30, 696–709.
86. Walsh, T. J.; Teppler, H.; Donowitz, G. R.; Maertens, J. A.; Baden, L. R.; Dmoszynska, A.; Cornely, O. A.; Bourque, M. R.; Lupinacci, R. J.;
Sable, C. A. et al. N. Engl. J. Med. 2004, 351, 1391–1402.
87. Polak, A. Prog. Drug Res. 2003, 59–190.
88. Zager, R. A. Am. J. Kidney Dis. 2000, 36, 238–249.
89. Walsh, T. J.; Melcher, G. P.; Rinaldi, M. G.; Lecciones, J.; McGough, D. A.; Kelly, P.; Lee, J.; Callender, D.; Rubin, M.; Pizzo, P. A. J. Clin.
Microbiol. 1990, 28, 1616–1622.
90. Arikan, S.; Lozano-Chiu, M.; Paetznick, V.; Nangia, S.; Rex, J. H. J. Clin. Microbiol. 1999, 37, 3946–3951.
91. Jezequel, S. G. J. Pharm. Pharmacol. 1994, 46, 196–199.
92. Cha, R.; Sobel, J. D. Expert Rev. Anti Infect. Ther. 2004, 2, 357–366.
93. Perfect, J. R. Oncology (Huntingt.) 2004, 18, 15–23.
94. Perfect, J. R. Oncology (Williston Park) 2004, 18, 5–14.
95. Vanden Bossche, H.; Marichal, P.; Gorrens, J.; Coene, M. C. Br. J. Clin. Pract. Suppl. 1990, 71, 41–46.
96. Richardson, K.; Cooper, K.; Marriott, M. S.; Tarbit, M. H.; Troke, P. F.; Whittle, P. J. Rev. Infect. Dis. 1990, 12, S267–S271.
97. Donnelly, J. P.; De Pauw, B. E. Clin. Microbiol. Infect. 2004, 10, 107–117.
98. Herbrecht, R. Expert Rev. Anti Infect. Ther. 2004, 2, 485–497.
99. Martin, M. V.; Yates, J.; Hitchcock, C. A. Antimicrob. Agents Chemother. 1997, 41, 13–16.
100. Gallagher, J. C.; Dodds Ashley, E. S.; Drew, R. H.; Perfect, J. R. Expert Opin. Pharmacother. 2003, 4, 147–164.
101. Purkins, L.; Wood, N.; Greenhalgh, K.; Allen, M. J.; Oliver, S. D. Br. J. Clin. Pharmacol. 2003, 56, 10–16.
102. Debono, M.; Gordee, R. S. Annu. Rev. Microbiol. 1994, 48, 471–497.
103. Rafael Sentandreu, M. V. E.; Valentı́n, E.; Ruiz Herrera, J. The Structure and Composition of the Fungal Cell Wall. In Pathogenic Fungi: Structural
Biology and Taxonomy; San-Blas, G., Calderone, R. A., Eds.; Caister Academic Press: Norwich, 2004; Chapter 1, pp 3–40.
104. Munro, C.; Gow, N. Chitin Biosynthesis as a Target for Antifungals. In Antifungal Agents. Discovery and Mode of Action; G. Dixon, L.; Copping, D.,
Eds.; Hollowman Bios Scientific Publishers: Oxford, 1995, pp 161–171.
105. Denning, D. W. Lancet 2003, 362, 1142–1151.
106. Abruzzo, G. K.; Flattery, A. M.; Gill, C. J.; Kong, L.; Smith, J. G.; Pikounis, V. B.; Balkovec, J. M.; Bouffard, A. F.; Dropinski, J. F.; Rosen, H.
et al. Antimicrob. Agents Chemother. 1997, 41, 2333–2338.
107. Maertens, J.; Raad, I.; Petrikkos, G.; Boogaerts, M.; Selleslag, D.; Petersen, F. B.; Sable, C. A.; Kartsonis, N. A.; Ngai, A.; Taylor, A. et al. Clin.
Infect. Dis. 2004, 39, 1563–1571.
108. Schuster, F.; Moelter, C.; Schmid, I.; Graubner, U. B.; Kammer, B.; Belohradsky, B. H.; Fuhrer, M. Pediatr. Blood Cancer 2005, 44, 682–685.
109. Murdoch, D.; Plosker, G. L. Drugs 2004, 64, 2249–2258, 2259–2260.
110. Onishi, J.; Meinz, M.; Thompson, J.; Curotto, J.; Dreikorn, S.; Rosenbach, M.; Douglas, C.; Abruzzo, G.; Flattery, A.; Kong, L. et al. Antimicrob.
Agents Chemother. 2000, 44, 368–377.
111. Parnham, M. J.; Bogaards, J. J.; Schrander, F.; Schut, M. W.; Oreskovic, K.; Mildner, B. Biopharm. Drug Dispos. 2005, 26, 27–33.
112. Petraitiene, R.; Petraitis, V.; Kelaher, A. M.; Sarafandi, A. A.; Mickiene, D.; Groll, A. H.; Sein, T.; Bacher, J.; Walsh, T. J. Antimicrob. Agents
Chemother. 2005, 49, 2084–2092.
113. Petraitis, V.; Petraitiene, R.; Kelaher, A. M.; Sarafandi, A. A.; Sein, T.; Mickiene, D.; Bacher, J.; Groll, A. H.; Walsh, T. J. Antimicrob. Agents
Chemother. 2004, 48, 3959–3967.
114. Brockmeyer N, R. M.; Oreskovic, K. Phase II study of icofungipen (PLD-118) in the treatment of oropharyngeal candidiasis (OPC) in HIV-positive patients,
44th Interscience Conference of Antimicrobial Agents, 2004.
115. Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Adv. Drug Deliv. Rev. 2001, 46, 3–26.
116. Sugimoto, Y.; Sakoh, H.; Yamada, K. Curr. Drug Targets Infect. Disord. 2004, 4, 311–322.
117. Vicente, M. F.; Basilio, A.; Cabello, A.; Pelaez, F. Clin. Microbiol. Infect. 2003, 9, 15–32.
118. Zhong, W.; Jeffries, M. W.; Georgopapadakou, N. H. Antimicrob. Agents Chemother. 2000, 44, 651–653.
119. Nagiec, M. M.; Nagiec, E. E.; Baltisberger, J. A.; Wells, G. B.; Lester, R. L.; Dickson, R. C. J. Biol. Chem. 1997, 272, 9809–9817.
120. Ikai, K.; Takesako, K.; Shiomi, K.; Moriguchi, M.; Umeda, Y.; Yamamoto, J.; Kato, I.; Naganawa, H. J. Antibiot. (Tokyo) 1991, 44, 925–933.
121. Takesako, K.; Kuroda, H.; Inoue, T.; Haruna, F.; Yoshikawa, Y.; Kato, I.; Uchida, K.; Hiratani, T.; Yamaguchi, H. J. Antibiot. (Tokyo) 1993, 46,
1414–1420.
122. Endo, M.; Takesako, K.; Kato, I.; Yamaguchi, H. Antimicrob. Agents Chemother. 1997, 41, 672–676.
123. Hashida-Okado, T.; Ogawa, A.; Endo, M.; Yasumoto, R.; Takesako, K.; Kato, I. Mol. Gen. Genet. 1996, 251, 236–244.
124. Kuroda, M.; Hashida-Okado, T.; Yasumoto, R.; Gomi, K.; Kato, I.; Takesako, K. Mol. Gen. Genet. 1999, 261, 290–296.
125. Heidler, S. A.; Radding, J. A. Antimicrob. Agents Chemother. 1995, 39, 2765–2769.
126. Kurome, T.; Inoue, T.; Takesako, K.; Kato, I. J. Antibiot. (Tokyo) 1998, 51, 359–367.
127. Lodge, J. K.; Johnson, R. L.; Weinberg, R. A.; Gordon, J. I. J. Biol. Chem. 1994, 269, 2996–3009.
128. Weinberg, R. A.; McWherter, C. A.; Freeman, S. K.; Wood, D. C.; Gordon, J. I.; Lee, S. C. Mol. Microbiol. 1995, 16, 241–250.
129. Devadas, B.; Freeman, S. K.; Zupec, M. E.; Lu, H. F.; Nagarajan, S. R.; Kishore, N. S.; Lodge, J. K.; Kuneman, D. W.; McWherter, C. A.;
Vinjamoori, D. V. et al. J. Med. Chem. 1997, 40, 2609–2625.
130. Sikorski, J. A.; Devadas, B.; Zupec, M. E.; Freeman, S. K.; Brown, D. L.; Lu, H. F.; Nagarajan, S.; Mehta, P. P.; Wade, A. C.; Kishore, N. S.
et al. Biopolymers 1997, 43, 43–71.
131. Nagarajan, S. R.; Devadas, B.; Zupec, M. E.; Freeman, S. K.; Brown, D. L.; Lu, H. F.; Mehta, P. P.; Kishore, N. S.; McWherter, C. A.; Getman, D.
P. et al. J. Med. Chem. 1997, 40, 1422–1438.
132. Lodge, J. K.; Jackson-Machelski, E.; Devadas, B.; Zupec, M. E.; Getman, D. P.; Kishore, N.; Freeman, S. K.; McWherter, C. A.; Sikorski, J. A.;
Gordon, J. I. Microbiology 1997, 143, 357–366.
133. Dorr, P. 3D structure analysis coupled with high throughput screening. Biochemical Society and SGM: Abstract 0950. 148th Ordinary Meeting,
March 26–30, 2001.
Fungi and Fungal Disease 443
134. Weston, S. A.; Camble, R.; Colls, J.; Rosenbrock, G.; Taylor, I.; Egerton, M.; Tucker, A. D.; Tunnicliffe, A.; Mistry, A.; Mancia, F. et al. Nat. Struct.
Biol. 1998, 5, 213–221.
135. Sogabe, S.; Masubuchi, M.; Sakata, K.; Fukami, T. A.; Morikami, K.; Shiratori, Y.; Ebiike, H.; Kawasaki, K.; Aoki, Y.; Shimma, N. et al. Chem. Biol.
2002, 9, 1119–1128.
136. Futterer, K.; Murray, C. L.; Bhatnagar, R. S.; Gokel, G. W.; Gordon, J. I.; Waksman, G. Acta Crystallogr. D Biol. Crystallogr. 2001, 57, 393–400.
137. Ebara, S.; Naito, H.; Nakazawa, K.; Ishii, F.; Nakamura, M. Biol. Pharm. Bull. 2005, 28, 591–595.
138. Ebiike, H.; Masubuchi, M.; Liu, P.; Kawasaki, K.; Morikami, K.; Sogabe, S.; Hayase, M.; Fujii, T.; Sakata, K.; Shindoh, H. et al. Bioorg. Med.
Chem. Lett. 2002, 12, 607–610.
139. Wu, L.; Pan, J.; Thoroddsen, V.; Wysong, D. R.; Blackman, R. K.; Bulawa, C. E.; Gould, A. E.; Ocain, T. D.; Dick, L. R.; Errada, P. et al. Eukaryot.
Cell 2003, 2, 256–264.
140. Schneiter, R.; Hitomi, M.; Ivessa, A. S.; Fasch, E. V.; Kohlwein, S. D.; Tartakoff, A. M. Mol. Cell Biol. 1996, 16, 7161–7172.
141. Pridzun L, S. F.; Reichenbach, H. Inhibition of Fungal ACCase: A Novel Target Discovered with the Myxobacterial Compound Soraphen. In
Antifungal Agents – Discovery and Mode of Action; Dixon, G. K., Copping, L. G., Holloman, D. W., Eds.; Bios Publ. SCI: Oxford, 1995, pp 99–108.
142. Harwood, J. L. Biochim. Biophys. Acta 1996, 1301, 7–56.
143. Al-Feel, W.; DeMar, J. C.; Wakil, S. J. Proc. Natl. Acad. Sci. USA 2003, 100, 3095–3100.
144. Hasslacher, M.; Ivessa, A. S.; Paltauf, F.; Kohlwein, S. D. J. Biol. Chem. 1993, 268, 10946–10952.
145. Evenson, K. J.; Gronwald, J. W.; Wyse, D. L. Plant Physiol. 1994, 105, 671–680.
146. Kinsman, O. S.; Chalk, P. A.; Jackson, H. C.; Middleton, R. F.; Shuttleworth, A.; Rudd, B. A.; Jones, C. A.; Noble, H. M.; Wildman, H. G.;
Dawson, M. J. et al. J. Antibiot. (Tokyo) 1998, 51, 41–49.
147. Dominguez, J. M.; Kelly, V. A.; Kinsman, O. S.; Marriott, M. S.; Gomez de las Heras, F.; Martin, J. J. Antimicrob. Agents Chemother. 1998, 42,
2274–2278.
148. San-Blas, G.; Calderone, R. Eds. Pathogenic Fungi. Host Interactions and Emerging for Control; Caister Academc Press: Norwich, 2004, Vol. 2.
149. Doctor Fungus. http://www.doctorfungus.org (accessed April 2006).
150. The Aspergillus Website. http://www.aspergillus.man.ac.uk (accessed April 2006).
Biography
Patrick Dorr gained his BSc in microbiology from Sheffield University, UK, in 1987, and PhD from Kent University,
UK, in 1990, following research in bacterial metabolism in Professor Chris Knowles’ laboratory. A subsequent move to
industry resulted in 5 years at Rhone-Poulenc, researching into new targets for pesticide discovery. In 1995, he moved
to Pfizer GRD in Sandwich, as a team leader in antifungals discovery, undertaking research in many aspects of drug
discovery, from new target validation and lead seeking to efficacy studies in infection models. He is currently leading
the CCR5 and HIV entry preclinical teams for the discovery of new antiviral drugs.
& 2007 Elsevier Ltd. All Rights Reserved Comprehensive Medicinal Chemistry II
No part of this publication may be reproduced, stored in any retrieval system or transmitted ISBN (set): 0-08-044513-6
in any form by any means electronic, electrostatic, magnetic tape, mechanical, photocopying,
recording or otherwise, without permission in writing from the publishers ISBN (Volume 7) 0-08-044520-9; pp. 419–443