Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Biochemical Pharmacology 133 (2017) 63–73

Contents lists available at ScienceDirect

Biochemical Pharmacology
journal homepage: www.elsevier.com/locate/biochempharm

Review

What is an ‘‘ideal” antibiotic? Discovery challenges and path forward


Sheo B. Singh a,⇑, Katherine Young b, Lynn L. Silver c
a
SBS Pharma Consulting, Edison, NJ 08820, USA
b
MRL, Merck & Co., Inc., Kenilworth, NJ 07033, USA
c
LL Silver Consulting, Springfield, NJ 07081, USA

a r t i c l e i n f o a b s t r a c t

Article history: An ideal antibiotic is an antibacterial agent that kills or inhibits the growth of all harmful bacteria in a
Received 28 September 2016 host, regardless of site of infection without affecting beneficial gut microbes (gut flora) or causing undue
Accepted 9 January 2017 toxicity to the host. Sadly, no such antibiotics exist. What exist are many effective Gram-positive antibac-
Available online 10 January 2017
terial agents as well as broad-spectrum agents that provide treatment of certain Gram-negative bacteria
but not holistic treatment of all bacteria. However effectiveness of all antibacterial agents is being rapidly
Keywords: eroded due to resistance. This viewpoint provides an overview of today’s antibiotics, challenges and
Antibiotics
potential path forward of discovery and development of new (ideal) antibiotics.
ESKAPE
Antibacterials
Ó 2017 Elsevier Inc. All rights reserved.
Discovery challenges
Path-forward of discovery

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
2. Brief review of key antibiotic classes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
2.1. b-Lactam antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
2.2. Glycopeptides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
2.3. Lipopeptide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
2.4. Aminoglycosides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
2.5. Tetracyclines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
2.6. Macrolides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
2.7. Oxazolidinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
2.8. Quinolones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
3. Challenges for discovery of an ideal new antibiotic without cross-resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
3.1. Finding new chemical matter for an antibiotic lead . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
3.2. The problem of entry into and retention of antibacterial chemicals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
4. Multi target vs single target . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
5. Broad-spectrum vs narrow spectrum . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
6. New target-lead pair and new lead-old target pair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
7. Potential path forward for a new ideal antibiotic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72

1. Introduction Indeed, it has been estimated that the widespread availability of


antibiotics has added 30 years to human life expectancy in devel-
Antibiotics are essential life-saving drugs that revolutionized oped countries [2]. Since the discovery of penicillin, a number of
medicine, starting with the discovery of penicillin in 1928 [1]. highly effective antibiotics have been discovered and developed
for clinical use in the treatment of bacterial infections [1,3]. Many
of these antibiotics have a broad-spectrum of activity, being effec-
⇑ Corresponding author.
tive in treatment of infections caused by Gram-positive as well as
E-mail addresses: sbs@sbspharma.com, sbsingh101@gmail.com (S.B. Singh).

http://dx.doi.org/10.1016/j.bcp.2017.01.003
0006-2952/Ó 2017 Elsevier Inc. All rights reserved.
64 S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73

Gram-negative bacteria, while others are effective for treatment of (Enterococcus faecium, S. aureus, Klebsiella pneumoniae, Acinetobac-
infections caused by only Gram-positive bacteria. Ever since the ter baumannii, Pseudomonas aeruginosa, and Enterobacter species)
discovery of these drugs the question has been asked, ‘‘What is by the Infectious Disease Society of America (IDSA) that need rapid
an ideal antibiotic?” While specific circumstances may vary, philo- development of new antibiotics [10–12]. No effective agents exist
sophically, an ideal antibiotic is an antibacterial agent that kills or against resistant strains of many of these Gram-negative bacteria,
inhibits the growth of harmful bacteria in a host regardless of site in particular, A. baumannii [12]. The Centers for Disease Control
of infection without affecting beneficial microbes (gut/skin flora) and Prevention (CDC) estimates that over 23,000 people died in
or causing undue toxicity to the host with low potential for resis- United States of America in 2013 due to infections caused by resis-
tance. To accomplish this goal the agent has to demonstrate broad- tant strains of bacteria [6]. A recent comprehensive report suggests
spectrum killing of Gram-positive as well as Gram-negative bacte- that over 700,000 people die every year from drug-resistant infec-
ria; and it has to have exceptional blood/fluid circulation as well as tions world-wide [13]. The low and middle-income countries bear
absorption, distribution, metabolism and excretion (ADME) prop- the brunt of the burden of antibiotic resistance where an estimated
erties allowing a low dose with a large therapeutic window. As 214,000 neonatal sepsis death per year is directly attributable to
almost anything will cause toxicity at a high dose, the therapeutic antibiotic resistant bacteria [2,14]. The greatest burden: when no
window is the only real measure to evaluate risk–benefit ratio. The safe and appropriate drug is available for treatment, an antibiotic
larger the therapeutic window, the better benefit a drug can pro- with low therapeutic index (e.g., polymyxin, colistin) is used to
vide without undue risk to the host. treat certain infections such as multi-drug resistant A. baumannii
No such ideal antibiotic exists or ever existed. Most antibiotics infection as a last resort [12]. New antibiotics with new modes of
in clinical use today do not have a broad-enough spectrum to treat action or a new chemical class with new target interactions and
infections caused by all Gram-positive and Gram-negative bacte- an established mode of action are needed. The question is, why
rial infections [4]. Therefore, most of the antibiotics today are are we where we are after the discovery of penicillin and many
grouped as Gram-positive agents, Gram-negative agents, and a other highly effective antibiotics between 1940 and 1962 [15].
few others with partial coverage of some Gram-positive as well What are the challenges? What have we learned? Is there a poten-
as some Gram-negative bacterial infections With a few exceptions, tial path forward? In this viewpoint we will discuss the challenges
all Gram-negative agents have activity against Gram-positive and present our opinion along with that of others for a potential
pathogens albeit with differing potencies. The selection of antibi- path forward for discovery and development of new (ideal) antibi-
otics for the treatment of bacterial infections is still empirical but otics for treatment of resistant bacterial infections.
is generally based on the physical symptoms presented by patients
and for which a given antibiotic is approved [5]. Such therapy is
started before results of a diagnostic test performed to determine 2. Brief review of key antibiotic classes
which bacterial strains are responsible for the infection are avail-
able [5]. Many of the oral Gram-negative antibiotics also inhibit The true systematic discovery of antibiotics began after the for-
anaerobic pathogens in the gut altering gut flora, and leading to tuitous discovery of penicillin with the discovery of a number of
diseases due to imbalance of gut microbial species. other natural product antibiotics from screening of microbial
In any case, an antibiotic, ideal or not, would not remain an extracts between 1940 and 1962 [1]. This led to the identification
effective antibiotic forever due to the inevitable development of of most of the chemical structures that became antibiotics them-
resistance. Antibiotics, unlike non-infectious disease drugs, have selves or served as chemical scaffolds for the discovery and devel-
a relatively short useful lifespan due to acquired resistance [6]. opment of subsequent generations of antibiotics supporting
So a specific ideal antibiotic will always be transitory and will have today’s antibiotic pipeline, with the exception of two new classes
to be replaced by newer ones when older ones become ineffective. approved after 2000 [15]. The major structural classes of clinical
Drug discovery teams almost always want to develop an ideal antibiotics (Table 1) and overall impact of major classes are briefly
antibiotic but invariably hit a variety of roadblocks, mostly scien- summarized here.
tific, which they cannot overcome, resulting in development of
imperfect but generally safe antibiotic.
Bacterial resistance to antibiotics is inevitable [6,7]. Bacteria 2.1. b-Lactam antibiotics
display various modes of resistance [8]. Resistance mechanisms
can include modification of the target to alter drug-binding, pro- Major b-lactam antibiotics are represented by four classes-
duction of alternate mechanisms to perform the function of the penicillin, cephalosporin, carbapenem, and monobactams (Fig. 1).
target, inactivation of the drug, reduction of drug entry and expres- With the exception of the monobactams, they are composed of a
sion of efflux pumps to eliminate drugs from inside bacterial cells bicyclic-fused ring system with a b-lactam ring being one of those
[8]. In certain cases bacteria are not only resistant to one class of rings. The penicillins contain sulfur in the five membered second
antibiotics but to several classes simultaneously, significantly rings, cephalosporins contain sulfur in the six membered second
limiting treatment options. For example, methicillin-resistant ring, and carbapenems contain a carbon in the five membered sec-
Staphylococcus aureus (MRSA) is not only resistant to penicillins ond ring. Monobactams do not contain a second ring. b-Lactam
but also to cephalosporins and carbapenems [8]. However, MRSA antibiotics inhibit bacterial growth by inhibiting cell wall synthesis
is generally susceptible to several Gram-positive drugs with via binding to a series of enzymes, penicillin-binding proteins
different modes of action (e.g., vancomycin, linezolid, daptomycin). (PBPs) that synthesize and remodel peptidoglycan. They are broad
Nevertheless death rates from invasive MRSA infection remain spectrum and highly effective antibiotics. These classes com-
high [9]. These Gram-positive agents do not work in all cases for manded tremendous efforts on chemical modifications affording
variety of reasons, including failure to reach the site of bacterial over 100 clinical agents in last 75 years [16].
infection, poor tissue penetration, inactivation by body fluids, Sequential and systematic structural diversification of the R
incorrect diagnosis and treatment or some other reason? group in the penicillin class of antibiotics has led to the develop-
What is more alarming currently is the rate of resistance to ment of over 20 clinically useful antibiotics including methicillin,
many drugs occurring in many critical Gram-negative pathogens. amoxicillin and piperacillin (Fig. 1). It is quite remarkable that
This has led to selection of a list of six priority pathogens medicinal chemistry practitioners have synthesized improved
(two Gram-positive and four Gram-negative) termed ESKAPE molecules, often with better spectrum and resistance profiles,
S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73 65

Table 1
Major classes of antibacterial agents used in systemic therapy.

Drug Class Examples Target Pathway inhibited Spectrum


b-Lactams Meropenem, amoxicillin Multiple PBPs Cell wall synthesis Broad spectruma
Glycopeptides Vancomycin Lipid II Cell wall synthesis Gram-positive
Macrolides Erythromycin, azithromycin 50S RNA of ribosome Protein synthesis Gram-positiveb
Oxazolidinones Linezolid 50S RNA of ribosome Protein synthesis Gram-positive
Amphenicols Chloramphenicol 50S RNA of ribosome Protein synthesis Broad Spectrum
Lincosamides Clindamycin 50S RNA of ribosome Protein synthesis Gram-positive
Tetracyclines Doxycycline, tigecycline 30S RNA of ribosome Protein synthesis Broad Spectrum
Aminoglycosides Gentamicin, amikacin 30S RNA of ribosome Protein synthesis Broad Spectrum
Fluoroquinolones Levofloxacin, moxifloxacin Gyrase, Topoisomerase IV DNA synthesis Broad Spectrum
Nitroimidazoles Metronidazole DNA (?) DNA synthesis+ Broad Spectrum
Lipopeptides Daptomycin Membranes Membrane integrity Gram-positive
Polymyxins Colistin Membranes Membrane integrity Gram-negative
a
Broad spectrum indicates clinically useful activity against both Gram-positive and Gram-negative bacteria; some class members have narrower spectra.
b
Some macrolides have useful activity against fastidious Gram-negatives.

NH 2
S

OH N
H R H H
R H N H H
N S H
S N N
O 2 Me
O N R O
N R1 N O
O O O HO 2C N
CO 2H CO 2H CO 2H O SO3H
Penicillins(>20) Cephalosporins (>70) Carbapenems (>7) Monobactam (Aztreonam)
Me
N+
OMe HO H N O
N N H HO
H H
N H HO P N S H H
S O S N N H
MeO O
O N S N NH
N S S N
O O O
CO 2H CO 2H CO 2H

Methicillin Ceftaroline Imipenem


CO 2H
O
NH 2 N O O
H 2N N H HN HO
H H H N H H
N N S NH 2 NMe 2
S S N H
HO NH 2
O O S NH
N N N+ N N
O O O
CO 2H CO 2H CO 2H
Amoxicillin Ceftolozane Meropenem
NEt
O
N
O O
NH N O
H 2N N H HO
H H N H H H
N S NH
S S NH S O
O O N S
N NH NH H N O
N N 2
O O O
CO 2H CO 2H O CO 2H
P iperacillin Ceftobiprole Doripenem
Fig. 1. Chemical structures of selected numbers of b-lactam antibiotics.

one at a time just through modification at a single location of the cephalosporin antibiotics may be clustered into five groups based
molecule [16]. on R and R1 substitution types, spectrum and resistance profiles.
The cephalosporin class (Fig. 1) of b-lactam antibiotics was sub- Ceftaroline, and ceftobiprole (approved in EU) are the latest
ject to much more intensive systematic studies leading to over 70 (2010–2015) broad-spectrum fifth generation cephalosporins
clinical agents often with broader spectrum of activity and superior entered into clinical practice (Fig. 1) [16].
resistance profile. Syntheses of a large number of improved cepha- Thienamycin, the first member of the carbapenem class of
losporin antibiotics were realized only because beneficial chemical b-lactam antibiotics, was discovered in the 1970’s and a derivative
diversification could be performed simultaneously at two struc- (imipenem in combination with the renal dehydropeptidase I
tural sites of the molecule represented by R and R1. The resulting inhibitor, cilastatin) was first approved for clinical use in 1985.
66 S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73

O HN O
H
H HO O N
O OH S H 2N N
N N H
N N N S O B CO 2H
N N HO O
O O N N
CO 2H CO 2H O O SO3H O O SO3H
Cla vu la n ic a c id Tazobactam Avibactam Relebactam Vaborbactam

Fig. 2. Chemical structures of selected b-lactamase inhibitors.

Imipenem (Fig. 1) is one of the most potent and broadest spectrum spectrum b-lactamase inhibitors with inhibition of classes A, C
antibiotics that remain in clinical use as a reserved antibiotic for and some class D b-lactamases. This class of inhibitors is repre-
treatment of infections caused by imipenem susceptible strains sented by avibactam and relebactam (Fig. 2). Avibactam restores
of Gram-negative and Gram-positive bacteria including P. aerugi- most of the Gram-negative activity of ceftazidime, a cephalosporin.
nosa and A. baumannii. Subsequently, additional modifications The combination was approved for human use in 2015 [20]. The
including addition of a b-methyl group at C-2 of the five- imipenem-cilastatin-relebactam combination restores most of
membered core ring along with modifications of R groups allowed the imipenem activity against P. aeruginosa and many other
for development of six additional carbapenem antibiotics (panipe- Gram-negative pathogens harboring resistance due to expression
nem, ertapenem, biapenem, meropenem, doripenem, and tebipe- of class A and C b-lactamases. Imipenem-relebactam combinations
nem) with partially overlapping and/or varying antibacterial along with several other b-lactamase inhibitors of DBO class as
spectrum, properties and safety profile to provide alternatives for well as a boron containing inhibitor (vaborbactam) are in clinical
treatment of serious infections [16]. development [20]. Unfortunately even this strategy has not yet
Aztreonam, the only marketed member of the monobactam been amenable for development of an inhibitor of class B
class (so called because of the lack of a second fused ring) has an metallo-b-lactamase inhibitors to treat NDM-1 or other types of
exclusively Gram-negative spectrum. It is not active against resistance owing to metallo-beta-lactamase-producing pathogens.
Gram-positive and anaerobic bacteria due to its PBP-binding char- Meiji Pharma has published on their MBL-inhibitor ME-1071, but
acteristics. Monobactams are unique among b-lactams in being the spectrum [21] of inhibition does not appear to extend much
non-susceptible to hydrolysis by class B metallo-b-lactamases beyond the IMP family and it is unclear if development is continu-
(MBL). Indeed, resistance to all b-lactams except aztreonam is a ing. Besides inhibiting the metallo-beta-lactamases directly,
handy marker for MBL-producing bacteria. Because many bacteria another approach is to partner a class A/C BLI with an antibiotic
produce b-lactamases of multiple classes and aztreonam is suscep- inherently non-susceptible to hydrolysis such as aztreonam
tible to BLs of classes A and C, it is not a universal solution for the (ATM). Indeed the combination of ATM and avibactam reached
MBL problem [16]. phase I stage of clinical development (clinicaltrials.gov, 27 Septem-
Some of the latest members of the penicillins, cephalosporins ber 2016), though reports on in vitro activity indicated the spec-
and almost all of the carbapenems continue to be used for treat- trum may not be sufficient for MBL-producing P. aeruginosa.
ment of susceptible bacteria. However their utility as standalone Clearly in the absence of a new class of antibiotics to provide
antibiotics is eroding due to increasing resistance because of treatment of ESKAPE pathogens restoring the activity of workhorse
expression of PBP2x in Gram-positive (e.g., PBP2a in MRSA) and b-lactam antibiotics is a sound approach for treating resistant
derepressed or acquired b-lactamases in Gram-negative bacteria Gram-negative infections. However this approach does not fully
[17]. b-Lactamases are enzymes that inactivate b-lactam antibi- restore the activity of critical b-lactam antibiotics because of pres-
otics by hydrolytically opening the b-lactam ring, making them ence of other resistance mechanisms in many Gram-negative
unavailable for binding to PBPs and thus preventing their inhibi- pathogens.
tion of bacterial growth [17,18]. Four classes of b-lactamases (class
A, B, C and D) are expressed in different Gram-negative bacterial 2.2. Glycopeptides
species [17,18].
Fortunately, a strategy employed since the 1980’s leads to inhi- Vancomycin (Fig. 3) is a natural product antibiotic that binds to
bition of these hydrolytic enzymes by even more reactive elec- the terminal dipeptide, D-alanine-D-alanine, of Lipid II, a precursor
trophilic b-lactamase inhibitors, thus sparing b-lactam antibiotics of the peptidoglycan chain of the bacterial cell wall, thus prevent-
from b-lactamases [19]. The first of the b-lactamase inhibitors, ing cell wall synthesis [22]. It is an effective broad-spectrum Gram-
clavulanic acid (Fig. 2), a b-lactam-containing molecule lacking positive agent used to treat infections of methicillin susceptible as
intrinsic antibacterial activity, was discovered and approved in well as methicillin resistant S. aureus and other Gram-positive
combination with amoxicillin for the treatment of infections infections in the hospital [22]. For a while, vancomycin was consid-
caused by certain Gram-negative bacteria producing class A serine ered an antibiotic of last resort and remains critical for treatment
b-lactamases [19]. The combination agent restored most of the of Gram-positive infections including MRSA. However its utility
original amoxicillin activity against Gram-negative bacteria. This is being diminished against Enterococcus faecalis and E. faecium
led to development of the extended spectrum b-lactamase inhibi- infections due to an increasing rate of resistance. Vancomycin is
tor, tazobactam, which in combination with piperacillin restored also effective as an oral formulation for treatment of Clostridium
the original activity of piperacillin against Gram-negative difficile associated diarrhea (CDAD). In 2009, telavancin, a more
pathogens expressing class A b-lactamase enzymes [19]. More lipophilic derivative of vancomycin, was approved for treatment
recently tazobactam has been marketed in combination with the of acute bacterial skin and skin structure infections (AbSSSi)
cephalosporin ceftolozane, which has a modified side-chain at caused by Gram-positive infections including MRSA. It has a long
the 3-position of the cephem nucleus that adds potent anti- half-life in humans of 9 h allowing for once daily dosing. Subse-
pseudomonal activity. After recent discovery efforts failed to pro- quently it was also approved for hospital-acquired pneumonia
duce a new class of antibacterial agents to treat resistant ESKAPE but only as a second line treatment [20,23]. Telavancin retains
pathogens, significant effort was expended on the discovery of activity against vancomycin-resistant bacterial strains due to the
new b-lactamase inhibitors. This effort resulted in the discovery VanB mechanism but not against VanA strains. Recently, dalba-
of the new diazabicyclooctane (DBO) class (Fig. 2) of extended vancin and oritavancin, two newer more potent glycopeptide
S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73 67

Cl
HO NH NH
2
HO
O O
O O O O
O H 2N O
Cl Cl
O O HO O O O

HO OH OH
Cl O Cl O
O O H O O H
H H H H
N N N N N N
O N O N
N N N N
H H H H H H
NH O O NH O O
H 2NOC H 2NOC
HO 2C HO 2C
OH OH
HO Vancomycin HO Oritavancin
O
OH
OH
CO 2H N CO 2H
O H O
O H O
N O O
H 2N N NH
H CO 2H
NH O OH
O HO Cl
O O O
H H
HN HO 2C NH O N N
HN N N NH
H O H H O
H O O H NH O O
HN N N Me 2N Cl
N O HN N NHMe
OH
O H
O O O H O HO O
CONH 2 N O O
NH HO OH
HO O HO
O OH
CO 2H OH
NH 2 O OH

Daptomycin Dalbavancin

Fig. 3. Chemical structures of selected glycopeptides and lipopeptide antibiotics.

H 2N H
H 2N N
HO NH HO
HO NH
O
OH O O
OH
O NH 2
CHO HN NH 2 NH 2 OH
H 2N H 2N H
O NH 2 O N
HN NH 2
OH O HO HO
NM 2 O
OH O O
O OH
HN HN
HO OH OH
S isomicin Plazomicin

Fig. 4. Chemical structures of selected aminoglycoside antibiotics.

antibiotic analogs with improved properties were approved by reg- spectra of dalbavancin and oritavancin are generally similar to van-
ulatory agencies for treatment of Gram-positive infections supple- comycin including its cross-resistance profile except against
menting vancomycin in clinic [20,23]. Dalbavancin and oritavancin vancomycin-resistant strains. Dalbavancin shows in vitro cross-
(Fig. 3) are significantly more potent and have over a 300 h human resistance to the VanA phenotype of E. faecalis and E. faecium
half-life, leading to a significant dosing advantage for treatment of (VRE) but lower levels of cross-resistance the VanB phenotype of
AbSSSi, infusion of just a single dose (1000–1500 mg) compared to VRE [26]. Oritavancin does show some cross-resistance to the VanA
twice daily infusion of vancomycin. Both of these compounds are phenotype VRE (E. faecalis, MIC90 of 0.5 lg/mL and E. faecium,
approved for treatment of vancomycin-sensitive strains of bacteria MIC90 of 0.06 lg/mL) but no cross-resistance to VanB phenotypes
including Enterococcus sp. Dalbavancin is a semisynthetic deriva- (E. faecalis, MIC90 of 0.015 lg/mL and E. faecium, MIC90 of
tive of A40926 factor B, a teicoplanin type natural product, 60.008 lg/mL), due to acquisition of additional binding interac-
whereas oritavancin is a semisynthetic analog of chloroere- tions [27]. While none of the two compounds have been approved
momycin, a vancomycin type glycopeptide. Like vancomycin, dal- for the treatment of infections by VRE strains oritavancin has
bavancin and oritavancin bind to D-ala-D-ala but unlike potential to be used for such treatments.
vancomycin, they gain secondary interaction with the cell mem-
brane [24,25]. Additionally, oritavancin binds to the pentaglycyl 2.3. Lipopeptide
bridging segment of S. aureus peptidoglycan [25]. The lipophilic
structural features of these two agents not only increase half-life Daptomycin (Fig. 3) is a natural lipopeptide, that is potent,
in humans but also provide additional target(s) interactions broad-spectrum and rapidly bactericidal Gram-positive agent that
accounting for improved potency [20,24,25]. The antibacterial is not cross-resistant to any of the other clinically used antibacte-
68 S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73

rial agents due to its novel mode of action [28]. Mechanistically, sisomicin [20] which is less susceptible to many aminoglycoside
daptomycin is inserted into the cell membrane causing depolariza- resistance mechanisms, is currently undergoing phase III develop-
tion and formation of holes, leading to ion leakage and disruption/ ment and has been granted fast track designation for treatment of
rupture of the cell membrane and bacterial death. Daptomycin is a serious and life-threatening carbapenem resistant Enterobacteri-
highly successful intravenous antibiotic for treatment of nosoco- aceae (CRE) infections. (www.achaogen.com, September 4, 2016).
mial Gram-positive infections including MRSA and VRE strains
[28]. 2.5. Tetracyclines

2.4. Aminoglycosides Tetracyclines (Fig. 5) are another old class of broad-spectrum


antibiotics [31]. Ten members of this class of antibiotics have been
Aminoglycosides were discovered in the Golden age of antibi- in clinical practice for more than 60 years. They are inhibitors of
otic discovery-the first member being streptomycin (Fig. 4). More bacterial protein synthesis due to binding to the 16S rRNA of the
than 8 members of this family of antibiotics have been used in clin- 30S ribosomal subunit. Significant class-based resistances due to
ical practice for treatment of both Gram-positive and Gram- expression of tetracycline-specific efflux pumps and ribosome-
negative bacteria due to their broad-spectrum activity [29]. They protection mechanisms have reduced the effectiveness of tetracy-
bind to the 16S rRNA subunit of the 30S ribosome and inhibit bac- clines [31]. However tigecycline (Fig. 5), a glycylcycline, is a
terial protein synthesis [29]. A number of aminoglycosides were broad-spectrum agent that evades most bacterial tetracycline-
used in the clinic in the past but they have fallen out of favor not specific efflux pumps and other tetracycline resistance
only due to resistance development but also due to reversible mechanisms. With the successful launch of tigecycline, several
nephrotoxicity and irreversible ototoxicity [30]. An improved other tetracycline semisynthetic derivatives with broad-spectrum
aminoglycoside, plazomicin (Fig. 4), a semisynthetic derivative of activity are being evaluated in phase III clinical trials [20,30].

HO N N N
H H H H
OH OH
O
H
NH 2 N NH 2
N
OH H OH
OH O OH O O OH O OH O O

Tetracycline Tigecycline

O
N
N N N F N
H H H H H H
OH OH OH
O
H
NH 2 N NH 2 N NH 2
N
OH OH H OH
OH O OH O O OH O OH O O OH O OH O O

S arecycline Omadacycline Eravacycline


Fig. 5. Chemical structures of selected tetracycline antibiotics.

O O
Me
HO N
HO OH HO OMe HO OH
OH NMe 2 OH NMe 2 NMe 2
HO O HO O HO O
O O O O O O

O O O O O O O O O

O OH O OH O OH

Erythromycin Clarithromycin Azithromycin

O N O
N
N N O N N N O N
H 2N OMe
OMe
O O NMe 2
NMe 2
HO HO O
O O O
O O

O O O O
F

Telithromycin S olithromycin
Fig. 6. Chemical structures of selected macrolide antibiotics.
S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73 69

Sarecycline (Fig. 5) is being studied for treatment of acne whereas Gram-positive ABSSSI by once daily dosing for a 6 day duration
the omadacycline (Fig. 5) is being evaluated for AbSSSi and compared to linezolid’s 10 days [20,30]. Tedizolid is more potent
community-acquired bacterial pneumonia (CABP) [20]. After the than linezolid in vitro and shows activity against some linezolid-
development of an efficient convergent total synthesis of tetracy- resistant strains, especially those carrying the horizontally trans-
cline [32,33], it became possible to design compounds that were mitted cfr resistance determinant [38]. Tedizolid is a bactericidal
not easily obtained via semisynthetic routes leading to the synthe- agent in vivo and is not cross-resistant with linezolid-resistant bac-
sis of eravacycline (Fig. 5), the first C-7, C-9-disubstituted fluorocy- terial strains. Tedizolid does not interact with eukaryotic mito-
cline with broad-spectrum antibacterial activity including MDR chondria and has the potential for a better safety profile that
bacteria [20,30]. Bacterial strains harboring tetracycline efflux must be confirmed in clinic [39]. Radezolid (Fig. 7) is another oxa-
pumps are susceptible to eravacycline. It is also active against zolidinone analog undergoing phase II clinical development. Sev-
many multiple drug resistant Gram-negative bacterial strains. eral other oxazolidinones are in earlier stages of clinical
Eravacycline is being studied in phase III trials for treatments of development [20,30].
cIAI and cUTI (www.tetraphase.com, September 4, 2016).
2.8. Quinolones
2.6. Macrolides
Quinolones, exemplified by nalidixic acid (actually a naph-
Macrolides (Fig. 6) have been in clinical use for more than five thyridine) (Fig. 8) are a class of synthetic antibiotic that have been
decades for the oral treatment of community-acquired respiratory used for treatment of bacterial infections for over 35 years [40].
infections [34]. They inhibit bacterial protein synthesis due to Fluoroquinolones were introduced as a second-generation of qui-
binding to the 23S rRNA of the 50S ribosomal subunit [35]. More nolones in 1980’s showing improved potency and antibacterial
than 14 members of the macrolide class have been in clinical use spectrum. The class became a significant contributor to treatment
and three key members of the macrolide family, namely ery- of bacterial infections including major Gram-negative strains such
thromycin, clarithromycin, and azithromycin (Fig. 6), play critical as Pseudomonas aeruginosa. Over 30 quinolones have been
roles in the treatment of respiratory tract infections [34]. However approved for clinical use. The most commonly used broad-
their use is hampered due to development of macrolide resistance spectrum fluoroquinolones are represented by ciprofloxacin and
in S. pneumoniae. Ketolide derivatives overcome this resistance, levofloxacin (Fig. 8) [40]. Quinolones are bactericidal and inhibit
leading to approval of the first ketolide, telithromycin (Fig. 6). bacterial growth by inhibiting the bacterial DNA synthetic
Unfortunately, its use was severely restricted by FDA in a post- enzymes DNA gyrase and topoisomerase IV [40]. Bacteria have
approval decision due to adverse events. Solithromycin (Fig. 6) is developed significant resistant to the quinolones by mutation at
another semisynthetic ketolide that has completed two-phase III one or both of the target binding sites. Newer quinolones having
trials for CABP [20,30]. Both trials met primary end points and have more balanced activity against DNA gyrase and Topoisomerase
been under FDA regulatory review (www.cempra.com, September IV, such as moxifloxacin, are less susceptible to resistance selec-
4, 2016). Recently, as with tetracycline, a highly convergent gen- tion. Ten quinolone antibiotics have been approved out of the total
eral total synthesis of the macrolide class of compounds has been of 32 total antibiotics approved by regulatory agencies between
reported [36] that is currently being exploited from a commercial 200 and 2015 [20]. Three new quinolones are in phase III and
perspective (www.macrolide.com). The newly minted synthetic two in phase II clinical development [20]. The latest approved qui-
route may enable synthesis of diverse analogs of macrolides not nolone antibiotics are nemonoxacin, finafloxacin and ozenoxacin
easily achieved by semisynthesis routes and may allow a renais- (Fig. 8) [20]. In addition to the quinolone class of antibiotics, the
sance of macrolides that overcome resistance. gyrase and topoisomerase IV dual targets bind and are inhibited
by a variety of other chemical classes of small drug-like molecules
2.7. Oxazolidinones including non-quinolone bacterial topoisomerase inhibitors
(NBTIs) and ETX0914 [41]. While none have yet been approved
Oxazolidinones are the newest class of synthetic antibacterial for clinical use, gepotidacin and ETX0914 (Fig. 8) are in phase II
agent represented by linezolid (Fig. 7), the first FDA approved oxa- clinical development for treatment of gonorrhea [20].
zolidinone in 2000 [20,37]; a second member, tedizolid, was
approved in 2014 [20]. Linezolid is a broad-spectrum Gram- 3. Challenges for discovery of an ideal new antibiotic without
positive agent demonstrating efficacy against MRSA infections. It cross-resistance
is an inhibitor of protein synthesis due to binding to the 23S rRNA
of the 50S ribosomal subunit [35]. It is indicated for treatment of 3.1. Finding new chemical matter for an antibiotic lead
serious Gram-positive infections. While linezolid resistance does
occur, it is not (yet) prevalent; however the utility of linezolid is As has been discussed above all antibiotics are natural products
limited to short term treatment (less than 2 weeks) due to reversi- or their semi-synthetic derivatives and synthetic analogs of oxazo-
ble myelosuppression resulting from inhibition of mammalian lidinone and quinolones. The derivatives have been derived by
mitochondrial protein synthesis. Overcoming resistance and incremental improvement of antibiotic properties via step-wise
myelosuppression provided impetus for the discovery and devel- chemical modification of various classes of natural product antibi-
opment of second generation oxazolidinones, leading to approval otics as well as the oxazolidinone and quinolone [1,20,42]. These
of tedizolid (Fig. 7) phosphate in 2014 for treatment of chemical modifications were based on empirical structure activity

O O H O
N
O O N N O N O O
O N N H N N N H
N N N OPO3H 2 N N
H
F F F

Linezolid Tedizolid phosphate Radezolid

Fig. 7. Chemical structures of oxazolidinone antibiotics.


70 S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73

Fig. 8. Chemical structures of selected quinolone and non-quinolone DNA topoisomerase II inhibitor antibiotics.

studies as well as incorporating structure-based design elements, abilities of pharmaceutical companies to pursue antibiotic discov-
wherever structural information was available. Identifying new ery efforts without disproportionate use of resources that could be
antibiotics with improved antibiotic spectrum and resistance prop- applied in other therapeutic areas with readily available biological
erties via further structural changes to the aforementioned struc- target-chemical lead pair starting points for medicinal chemistry.
tural classes is facing significant challenges due to lack of The lack of quality new antibiotic chemical leads (e.g., in compar-
additional beneficially modifiable chemical sites. In addition, ison to historical leads which often possessed desired developable
class-based resistance is inevitable. These issues are of foremost antibacterial spectrum) suitable for starting medicinal chemistry
concern to antibiotic discovery scientists worldwide. Large phar- programs was debated within pharmaceutical companies for many
maceutical companies were very active in making new antibiotics years, fortunately now it is being debated outside of the pharmaceu-
when chemical leads were abundantly available for medicinal tical industry at many public forums [41,43,44]. This is a critical step
chemistry improvement. But there has been a significant decrease for broadcasting the challenges and educating people outside of the
in delivery of new improved antibiotics by chemical modifications antibiotic field, and, most importantly, bringing the issues to the
of the old chemical lead classes and by failure of lead generation attention of decision makers at funding institutions. In the end it is
from screening of chemical libraries (from in-house pharmaceuti- the lack of new quality chemical antibacterial starting point leads that
cal companies as well as publically available chemical libraries). is the biggest roadblock for new antibiotic discovery.
This creates a monumental challenge and seriously hampers

3.2. The problem of entry into and retention of antibacterial chemicals


A = Acve transporters
Gram-posive Gram-negave B = SM efflux pumps
C = OM diffusion pore The cellular machinery of Gram-positive and Gram-negative
D = OM general porin
LPS E = RND efflux
bacteria is surrounded by protective membranes. A single
Pepdoglycan (SM = Single Membrane) cytoplasmic membrane layer in addition to a thick, rigid cell wall
(peptidoglycan) protect Gram-positive bacteria whereas Gram-
Outer
C D Membrane (OM) negative bacteria have a thinner peptidoglycan layer sandwiched
between two membranes, the cytoplasmic membrane (CM) and
Cytoplasmic an outer membrane (OM) (Fig. 9) [45]. These differences in the
A B A B E Membrane protective cellular structure are the root cause of problems in dis-
covery of anti Gram-negative drugs [45]. Compounds inhibiting
Fig. 9. Differences between Gram-positive and Gram-negative permeability barri- intracellular targets must cross these membranes. The OM consists
ers. Gram-positive and Gram-negative envelopes: A = active transporter; B = single
of an asymmetric bilayer that comprises an outer leaflet of
membrane efflux pumps; C = outer membrane facilitated diffusion pore; D = outer
membrane general porin; E = RND efflux pump spanning both outer and cytoplas- lipopolysaccharide (LPS) and an inner leaflet of phospholipids –
mic membranes. All five A–E (transporters, efflux pumps, diffusion pore and porin) together making the OM relatively impervious to both hydropho-
are present in Gram-negative bacterial strains whereas only active transporter (A) bic and hydrophilic compounds (Fig. 9) [45]. To allow passage of
and single membrane efflux pump (B) is present in Gram-positive bacterial strains. required nutrients, it is traversed by water-filled channels called
Many Gram-positive agents show weak or no activity against Gram-negative
bacterial strains because of the outer membrane barrier or the action of efflux
porins, which are selective for hydrophilic, charged compounds
pumps. If outer membrane is breached or efflux inactivated, such compounds gain [46]. On the other hand, the CM is permeable to hydrophobic com-
Gram-negative activity. pounds but allows limited diffusion of hydrophilic compounds. To
S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73 71

enable uptake of required hydrophilic and other nutrients, the CM edge of the causative bacterial strains, they should cover all
contains solute-specific active transport systems such as perme- likely bacterial pathogens for a given site of infection. Traditional
ases. Compounds that can cross the OM may be expelled by efflux culture-based strain identification takes 24–48 h, which presents
pumps whose substrates appear to be small and hydrophobic or treatment challenges for seriously ill patients if empiric treatment
large and zwitterionic [47]. Thus, large molecules are excluded is not rapidly applied. Broad-spectrum antibiotics, particularly oral
by size limitation of porins; lipophilic molecules that could pene- antibiotics, do have disadvantages as they often inhibit growth of
trate the CM may not be allowed entry by the OM and may be sub- anaerobic gut bacteria leading to imbalance in the bacterial popu-
ject to efflux pumps. Small hydrophilic molecules, particularly with lations of gut flora causing disease conditions (e.g., Clostridium dif-
weak positive charge, can cross the OM and enter the periplasm ficile associated diarrhea, CDAD). Use of both a broad-spectrum
and are possibly less subject to efflux pumps, but are unlikely Gram-positive and Gram-negative agent is a second option where
allowed entry by CM. It might be said that the OM, CM and efflux seriously ill patients can be dosed with two antibiotics concomi-
pumps are selective for uptake of solutes that are needed for bac- tantly. A few agents do provide treatment coverage for most
terial survival but tend to exclude chemicals that are foreign to Gram-positive infections including many commonly occurring
bacteria [45]. The question arises: how do any of the existing drug-resistant strains but such an option does not exist for treat-
Gram-negative antibiotics get into the cytoplasm given the orthog- ment of many drug-resistant Gram-negative bacteria [44]. It can
onal sieving properties of the membranes and pumps? While there be argued that narrow spectrum (affecting one or a few similar
are certain exceptions, the existing cytoplasmically targeted species) agents may be easier to discover and should be better/
antibiotics appear to have properties allowing diffusion through safer as they will likely affect only bacterial populations that cause
both the porins of the OM and the phospholipid bilayer of the disease without affecting beneficial bacterial populations. How-
CM. Detailed systematic studies leading to development of guideline ever, the challenges of development of narrow-spectrum or
of principles of permeability (entry) and efflux is critically needed for strain-specific agents should not be minimized, as many targets
successful and efficient development of Gram-negative antibiotics. are common (similar) in most bacteria. While the spectrum of such
To compound this challenge, cell membranes of all Gram- an agent may appear narrow, variable target inhibitory activities
negative pathogens are not identical. In some cases differences due to differences in target binding and/or target access (based
are quite significant. on differences in cell envelope structure), may lead to drastically
different MIC values. Therefore, at a given dose, complete growth
inhibition of the major targeted strain can be achieved while lead-
4. Multi target vs single target
ing to only partial inhibition of less sensitive strains. This situation
may lead to selection of resistance in the partially inhibited popu-
It is inevitable that all antibiotics will develop resistance sooner
lations. This may theoretically create problems - but not always.
or later. Empiric observation of cumulative data on existing antibi-
For example, partial inhibition of Pseudomonas aeruginosa by erta-
otics suggest that antibiotics interacting with more than one bio-
penem has not resulted in an increase in resistance of Pseu-
logical targets (protein, RNA, DNA) have lower propensity for
domonas to other antibiotics including carbapenems [52]. One-
target-based resistance than those targeting a single enzyme target
way this might be potentially circumvented is by identification/
[48]. While the in vivo rate of resistance is more complex it is likely
selection of strain-specific targets and development of individual
that an antibiotic that binds to a single target will rapidly select
antibiotics inhibiting those targets on a strain-by-strain basis.
pre-existing target mutations that arise at a measurable rate per
However, this target-specific approach, if not multi-targeted, has
generation [48]. In fact, selection of target mutations is a technique
the downside of likely leading to rapid resistance selection.
used for mechanism of action determination of antibiotics. Exam-
In any event, strain specific and super narrow antibiotics have
ples of multi-targeting are the multiple penicillin binding proteins
to be coupled with rapid diagnostics for this strategy to be success-
targeted by b-lactams, Gyrase/topoisomerase IV that are targeted
ful. In addition regulatory agencies must make a paradigm shift to
by quinolones, and the rRNA of ribosomes targeted by many pro-
allow strain-based clinical trials and approval vs current disease-
tein synthesis inhibitors that is the product of many rRNA cistrons.
based trials and approval. If successful, the treatment and manage-
The incorporation of structural elements in oritavancin and dal-
ment of monomicrobial bacterial infections will be changed likely
bavancin endowing them with additional binding interactions to
for better. However, treatment of polymicrobial infections will
other targets led to improved potency and spectrum validating
require concurrent administration of a second complementary
another advantage of multi-targeting. In addition, this example
antibiotic or broad-spectrum antibiotic. None of this will help dis-
directly addresses the idea that additional binding sites can be
covery of quality antibiotics until a quality chemical lead-target
crafted in certain molecules, particularly in larger molecules, by
pair is identified. However, once such a quality chemical lead-
structure modification to potentially bring in additional interaction
target pair is identified, this approach may simplify the process
with the same target and/or engagement to another target. A sim-
of lead optimization and may potentially reduce the cost of clinical
ilar argument can be made for the U-shape binding of the natural
development. Unfortunately, this approach will also segment the
product kibdelomycin to the active site as well as many backbone
patient population for individual antibiotics and would likely lead
interactions with Gyrase B and ParE proteins [49,50]. These inter-
to significantly higher cost of treatment unless new pricing strate-
actions likely lower the rate of resistance in S. aureus compared
gies are developed.
to other Gyrase B and ParE inhibitors [51]. Therefore, it is expected
that ideal antibiotics would likely bind to more than one target or
at least will bind to targets with multi-point polar contacts not
6. New target-lead pair and new lead-old target pair
only at the active site but also with the backbone of the target.
While the latter approach needs clinical validation, it is worth con-
Most of the established antibiotic targets are generally privi-
sidering until abundant chemical leads become available.
leged targets affected by specific inhibitor classes with the excep-
tion of the bacterial topoisomerase II target (see below). For
5. Broad-spectrum vs narrow spectrum example, the active sites of penicillin-binding proteins interact
with b-lactam antibiotics, the D-ala-D-ala binding site interacts
Broad-spectrum antibiotics (Table 1) are often ideal for empiri- with glycopeptides, specific ribosomal sites interact with macro-
cal treatment of bacterial infection [5]. In the absence of knowl- lides, tetracycline, and aminoglycosides (Table 1). These molecular
72 S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73

interactions tolerate chemical modifications only on a narrow por- highlighted but also a consortium approach has been advocated
tion of the antibiotic molecule leaving the major portion of the for design and synthesis of new chemical matters with antibiotic
chemical structure untouchable (see examples in Section 2, above). like properties along with other discovery/cell based screening
Peptidoglycan and ribosomes are large biological structures that approaches has been discussed (www.pewtrusts.org).
contain many antibiotic targets; they can be potentially interro- In order to discover ideal antibiotics, whether broad-spectrum or
gated by a variety of chemical matter through individual narrow spectrum, new leads must be discovered. Chemical antibiotic
inhibitor-target pairs that appear to be privileged. Therefore de leads can come from synthetic matter discussed above or from revis-
novo design or natural product based alternate binders/inhibitors iting natural products. Both traditional and genetic based natural
to the clinically validated privileged sites has not been successful. product approaches should be considered as part of the overall goal.
Many other peptidoglycan or protein synthesis inhibitors targeting Many shortcomings of natural product antibacterial discoveries can
other binding sites have been reported but have not been advanced now, with modern methods, be addressed rapidly and efficiently.
to clinical practice. Bacterial topoisomerase II target is the rare Cell based screening approaches are critical for discovery of new
exception. It is composed of four proteins, Gyrase A, B, topoiso- quality chemical matters for further medicinal chemistry optimiza-
merases IV (ParC and ParE). The highly successful quinolone class tion. Multi-targeting is key for a lower rate of resistance, which is
of antibiotics (targeting Gyrase A/ParC) as well as the coumarin an important a criterion for advancing a lead into clinic for develop-
(Gyrase B/ParE) class of natural product antibiotics binds/inhibits ment of an ideal antibiotic regardless of spectrum.
these dual target pairs (Gyrase A/ParC and gyrase B/ParE). Because
of the wide tolerance for binding with biologically functional con-
References
sequences a variety of structural classes have been identified and
optimized to broad-spectrum antibiotics exemplified by the Phase [1] S.B. Singh, J.F. Barrett, Empirical antibacterial drug discovery-Foundation in
II clinical agents gepotidacin and EXT0914 [20]. Many other inhibi- natural products, Biochem. Pharmacol. 71 (2006) 1006–1015.
tors have been reported that interact with these targets at different [2] R. Laxminarayan, P. Matsoso, S. Pant, C. Brower, J.A. Rottingen, K. Klugman,
et al., Access to effective antimicrobials: a worldwide challenge, Lancet 387
sites [41]. (2016) 168–175.
[3] E.D. Brown, G.D. Wright, Antibacterial drug discovery in the resistance era,
7. Potential path forward for a new ideal antibiotic Nature 529 (2016) 336–343.
[4] C.T. Walsh, Antibiotics: Actions, Origin, Resistance, ASM Press, Washington,
DC, 2003.
With identification of challenges, several teams started analyz- [5] J. Klastersky, The changing face of febrile neutropenia-from monotherapy to
ing the chemical structure of antibiotics by computational analysis moulds to mucositis. Why empirical therapy? J. Antimicrob. Chemother. 63
(Suppl 1) (2009) i14–i15.
listed in the Chemical, Manufacturing, and Controls (CMC) data-
[6] T. Frieden, Antibiotic Resistance Threats in the United States, a report
base of FDA approved drugs and compared them with the FDA presented to United States Congress by Centers of Disease Control and
approved non-antibiotic drugs listed in the CMC database. All the Prevention (CDC), 2013. <http://www.cdc.gov>.
analyses indicated clear differences in the physical properties of [7] C. Pillar, D. Sahm, Resistance trends and susceptibility profiles in the US among
prevalent clinical pathogens: lessons from surveillance, in: T.J. Dougherty, M.J.
antibiotics vs other drugs. From these analyses it was also dis- Pucci (Eds.), Antibiotic Discovery and Development, 2, Springer, New York,
cerned that Gram-positive agents fall in a significantly different 2012, pp. 753–792.
property space than Gram-negative agents. Based on the cumula- [8] J.M. Blair, M.A. Webber, A.J. Baylay, D.O. Ogbolu, L.J. Piddock, Molecular
mechanisms of antibiotic resistance, Nat. Rev. Microbiol. 13 (2015) 42–51.
tive analysis the general conclusion is that Gram-positive agents [9] R.M. Klevens, M.A. Morrison, J. Nadle, S. Petit, K. Gershman, S. Ray, et al.,
have larger molecular weight, larger polar surface area (243 vs Invasive methicillin-resistant Staphylococcus aureus infections in the United
70), and lower C log D7.4 ( 0.2) than CMC drugs (1.6) [45]. The data States, J. Am. Med. Assoc. 298 (2007) 1763–1771.
[10] L.B. Rice, Federal funding for the study of antimicrobial resistance in
for Gram-negative drugs were dramatically different from Gram- nosocomial pathogens: no ESKAPE, J. Infect. Dis. 197 (2008) 1079–1081.
positive agents. The Gram-negative agents are generally smaller, [11] H.W. Boucher, G.H. Talbot, J.S. Bradley, J.E. Edwards, D. Gilbert, L.B. Rice, et al.,
more polar and charged with the C log D7.4 for Gram-negative being Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases
Society of America, Clin. Infect. Dis. 48 (2009) 1–12.
2.8 [45,53]. Most compounds in general compound libraries orig- [12] H.W. Boucher, G.H. Talbot, D.K. Benjamin Jr., J. Bradley, R.J. Guidos, R.N. Jones,
inate from other human-health drug discovery programs and et al., 10 x ’20 Progress–development of new drugs active against gram-
mostly consist of simple flat, achiral, and lipophilic structures that negative bacilli: an update from the Infectious Diseases Society of America,
Clin. Infect. Dis. 56 (2013) 1685–1694.
have been loosely designed for oral bioavailability, thus requiring a
[13] K. Kupferschmidt, Long-awaited report outlines how to fight antimicrobial
different set of properties from antibacterials as evidenced by com- resistance—and how to pay for it, Science (2016). Published online 18 May
parison of the CMC database and antibiotics. The differences of 2016.
physical properties of antibiotics in comparison with other drugs [14] R. Laxminarayan, D. Sridhar, M. Blaser, M. Wang, M. Woolhouse, Achieving
global targets for antimicrobial resistance: the UN should promote targets,
underscore the issue of why screening of chemical libraries funding, and governance, Science 353 (2016) 874–875.
(excluding known antibiotic classes) do not produce quality antibi- [15] L.L. Silver, Challenges of antibacterial discovery, Clin. Microbiol. Rev. 24 (2011)
otic cell-active hits. While any molecules with general physical 71–109.
[16] M.G. Page, Beta-lactam antibiotics, in: T.J. Dougherty, M.J. Pucci (Eds.),
properties of antibiotics cannot be assured to have antibiotic activ- Antibiotic Discovery and Development, 1, Springer, New York, 2012, pp. 79–
ity, inclusion of diversified complex structures with many func- 117.
tional groups can provide a reasonable opportunity, but no [17] K. Bush, Evolution of beta-lactamases: past, present, and future, in: T.J.
Dougherty, M.J. Pucci (Eds.), Antibiotic Discovery and Development, 1,
guarantee of success. Therefore, completely new libraries should Springer, New York, 2012, pp. 427–453.
be designed and synthesized keeping in mind the physical proper- [18] R.F. Pratt, Beta-lactamases: Why and How, J. Med. Chem. 59 (2016) 8207–
ties of antibiotics, incorporation of a variety of functional groups 8220.
[19] S.M. Drawz, K.M. Papp-Wallace, R.A. Bonomo, New beta-lactamase inhibitors:
embedded in complex three-dimensional natural product-like chi- a therapeutic renaissance in an MDR world, Antimicrob. Agents Chemother. 58
ral structures, particularly for Gram-negative agents. This is a big (2014) 1835–1846.
task for any single commercial organization to undertake and [20] M.S. Butler, M.A. Blaskovich, M.A. Cooper, Antibiotics in the clinical pipeline at
the end of 2015, J. Antibiot. (Tokyo) 70 (2017) 3–24.
therefore would require a collective approach in precompetitive
[21] D.M. Livermore, S. Mushtaq, A. Morinaka, T. Ida, K. Maebashi, R. Hope, Activity
space. In this context, The Pew Charitable Trusts have issued a of carbapenems with ME1071 (disodium 2,3-diethylmaleate) against
position paper on June 21, 2016 on ‘‘A scientific roadmap for antibi- Enterobacteriaceae and Acinetobacter spp. with carbapenemases, including
otic discovery-A sustained and robust pipeline of new antibacterial NDM enzymes, J. Antimicrob. Chemother. 68 (2013) 153–158.
[22] F.F. Arhin, A. Belley, A. Rafai Far, D. Lehoux, G. Moeck, Glycopeptides and
drugs and therapies is critical to preserve public health” in which Lipoglycopeptides, in: T.J. Dougherty, M.J. Pucci (Eds.), Antibiotic Discovery
not only scientific issues underlying lead generation have been and Development, 1, Springer, New York, 2012, pp. 301–346.
S.B. Singh et al. / Biochemical Pharmacology 133 (2017) 63–73 73

[23] M.S. Butler, K.A. Hansford, M.A. Blaskovich, R. Halai, M.A. Cooper, Glycopeptide tedizolid and linezolid to affect mitochondrial function, Antimicrob. Agents
antibiotics: back to the future, J. Antibiot. (Tokyo) 67 (2014) 631–644. Chemother. 59 (2015) 178–185.
[24] A.Y. Chen, M.J. Zervos, J.A. Vazquez, Dalbavancin: a novel antimicrobial, Int. J. [40] G.A. Jacoby, D.C. Hooper, Review of quinolone family, in: T.J. Dougherty, M.J.
Clin. Pract. 61 (2007) 853–863. Pucci (Eds.), Antibiotic Discovery and Development, 1, Springer, New York,
[25] G.G. Zhanel, F. Schweizer, J.A. Karlowsky, Oritavancin: mechanism of action, 2012, pp. 119–146.
Clin. Infect. Dis. 54 (Suppl 3) (2012) S214–S219. [41] S.B. Singh, Confronting the challenges of discovery of novel antibacterial
[26] M. Billeter, M.J. Zervos, A.Y. Chen, J.R. Dalovisio, C. Kurukularatne, agents, Bioorg. Med. Chem. Lett. 24 (2014) 3683–3689.
Dalbavancin: a novel once-weekly lipoglycopeptide antibiotic, Clin. Infect. [42] S.J. Brickner, D.K. Hutchinson, M.R. Barbachyn, P.R. Manninen, D.A. Ulanowicz,
Dis. 46 (2008) 577–583. S.A. Garmon, et al., Synthesis and antibacterial activity of U-100592 and U-
[27] R.E. Mendes, L.N. Woosley, D.J. Farrell, H.S. Sader, R.N. Jones, Oritavancin 100766, two oxazolidinone antibacterial agents for the potential treatment of
activity against vancomycin-susceptible and vancomycin-resistant multidrug-resistant gram-positive bacterial infections, J. Med. Chem. 39
Enterococci with molecularly characterized glycopeptide resistance genes (1996) 673–679.
recovered from bacteremic patients, 2009–2010, Antimicrob. Agents [43] D.J. Payne, M.N. Gwynn, D.J. Holmes, D.L. Pompliano, Drugs for bad bugs:
Chemother. 56 (2012) 1639–1642. confronting the challenges of antibacterial discovery, Nat. Rev. Drug. Discov. 6
[28] C. Vilhena, A. Bettencourt, Daptomycin: a review of properties, clinical use, (2007) 29–40.
drug delivery and resistance, Mini. Rev. Med. Chem. 12 (2012) 202–209. [44] R. Tommasi, D.G. Brown, G.K. Walkup, J.I. Manchester, A.A. Miller, ESKAPEing
[29] E.S. Armstrong, C.F. Kostrub, R.T. Cass, H.E. Moser, A.W. Serio, G.H. Miller, the labyrinth of antibacterial discovery, Nat. Rev. Drug Discov. 14 (2015) 529–
Aminoglycosides, in: T.J. Dougherty, M.J. Pucci (Eds.), Antibiotic Discovery and 542.
Development, 1, Springer, New York, 2012, pp. 229–269. [45] L.L. Silver, A Gestalt approach to Gram-negative entry, Bioorg. Med. Chem. 16
[30] M.J. Pucci, K. Bush, Investigational antimicrobial agents of 2013, Clin. (2016) 6379–6389.
Microbiol. Rev. 26 (2013) 792–821. [46] M.J. Macielag, Chemical properties of antimicrobials and their uniqueness, in:
[31] P.A. Bradford, C.H. Jones, Tetracyclines, in: T.J. Dougherty, M.J. Pucci (Eds.), T.J. Dougherty, M.J. Pucci (Eds.), Antibiotic Discovery and Development, 2,
Antibiotic Discovery and Development, 1, Springer, New York, 2012, pp. 147– Springer, New York, 2012, pp. 793–820.
179. [47] D.G. Brown, T.L. May-Dracka, M.M. Gagnon, R. Tommasi, Trends and
[32] M.G. Charest, C.D. Lerner, J.D. Brubaker, D.R. Siegel, A.G. Myers, A convergent exceptions of physical properties on antibacterial activity for Gram-positive
enantioselective route to structurally diverse 6-deoxytetracycline antibiotics, and Gram-negative pathogens, J. Med. Chem. 57 (2014) 10144–10161.
Science 308 (2005) 395–398. [48] S.P. East, L.L. Silver, Multitarget ligands in antibacterial research: progress and
[33] C. Sun, Q. Wang, J.D. Brubaker, P.M. Wright, C.D. Lerner, K. Noson, et al., A opportunities, Expert Opin. Drug Discov. 8 (2013) 143–156.
robust platform for the synthesis of new tetracycline antibiotics, J. Am. Chem. [49] J.W. Phillips, M.A. Goetz, S.K. Smith, D.L. Zink, J. Polishook, R. Onishi, et al.,
Soc. 130 (2008) 17913–17927. Discovery of kibdelomycin, a potent new class of bacterial type II
[34] Z.-Q. Xu, M.T. Flavin, D.A. Eiznhamer, Macrolides and Ketolides, in: T.J. topoisomerase inhibitor by chemical-genetic profiling in Staphylococcus
Dougherty, M.J. Pucci (Eds.), Antibiotic Discovery and Development, 1, aureus, Chem. Biol. 18 (2011) 955–965.
Springer, New York, 2012, pp. 181–228. [50] J. Lu, S. Patel, N. Sharma, S.M. Soisson, R. Kishii, M. Takei, et al., Structures of
[35] M.F. Chellat, L. Raguz, R. Riedl, Targeting Antibiotic Resistance, Angew. Chem. kibdelomycin bound to Staphylococcus aureus GyrB and ParE showed a novel
Int. Ed. Engl. 55 (2016) 6600–6626. U-shaped binding mode, ACS Chem. Biol. 9 (2014) 2023–2031.
[36] I.B. Seiple, Z. Zhang, P. Jakubec, A. Langlois-Mercier, P.M. Wright, D.T. Hog, [51] S.B. Singh, Discovery and development of kibdelomycin, a new class of broad-
et al., A platform for the discovery of new macrolide antibiotics, Nature 533 spectrum antibiotics targeting the clinically proven bacterial type II
(2016) 338–345. topoisomerase, Bioorg. Med. Chem. 24 (2016) 6291–6297.
[37] M.R. Barbachyn, Oxazolidinone antibacterial agents, in: T.J. Dougherty, M.J. [52] C.A. Rodriguez-Osorio, C.O. Sanchez-Martinez, J. Araujo-Melendez, E. Criollo, A.
Pucci (Eds.), Antibiotic Discovery and Development, 1, Springer, New York, E. Macias-Hernandez, A. Ponce-de-Leon, et al., Impact of ertapenem on
2012, pp. 271–299. antimicrobial resistance in a sentinel group of Gram-negative bacilli: a 6
[38] J.B. Locke, G.E. Zurenko, K.J. Shaw, K. Bartizal, Tedizolid for the management of year antimicrobial resistance surveillance study, J. Antimicrob. Chemother. 70
human infections: in vitro characteristics, Clin. Infect. Dis. 58 (Suppl 1) (2014) (2015) 914–921.
S35–S42. [53] R. O’Shea, H.E. Moser, Physicochemical properties of antibacterial compounds:
[39] S. Flanagan, E.E. McKee, D. Das, P.M. Tulkens, H. Hosako, J. Fiedler-Kelly, et al., implications for drug discovery, J. Med. Chem. 51 (2008) 2871–2878.
Nonclinical and pharmacokinetic assessments to evaluate the potential of

You might also like