Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

New Therapeutic

Developments against Shiga


Toxin-Producing Escherichia coli
ANGELA R. MELTON-CELSA1 and ALISON D. O’BRIEN1
1
Department of Microbiology and Immunology, Uniformed Services
University of the Health Sciences, Bethesda, MD 20814

ABSTRACT Shiga toxin (Stx)-producing Escherichia coli (STEC) bacteria to intimately adhere to and form attaching
is an etiologic agent of bloody diarrhea. A serious sequela of and effacing lesions on intestinal tissue. The O157:H7
disease, the hemolytic uremic syndrome (HUS) may arise in up
strains, which are responsible for the majority of large
to 25% of patients. The development of HUS after STEC infection
is linked to the presence of Stx. STEC strains may produce one or
outbreaks due to STEC infection, are members of the
more Stxs, and the Stxs come in two major immunological EHEC group. All STEC strains make one or more Stxs;
groups, Stx1 and Stx2. A multitude of possible therapeutics these pathogens may produce two immunologically
designed to inhibit the actions of the Stxs have been developed distinct but highly similar Stxs, Stx1 and Stx2. These
over the past 30 years. Such therapeutics are important because toxins are briefly described in the section on therapeutics
antibiotic treatment of STEC infections is contraindicated due to targeted to the Stxs.
an increased potential for development of HUS. The reason for Some individuals infected with STEC manifest a se-
the increased risk of HUS after antibiotic treatment is likely
rious sequela called hemolytic uremic syndrome (HUS),
because certain antibiotics induce expression of the Stxs, which
are generally associated with lysogenic bacteriophages. There a thrombotic microangiopathy defined by the presence
are a few potential therapeutics that either try to kill STEC of hemolytic anemia, thrombocytopenia, and renal fail-
without inducing Stx expression or target gene expression within ure. The initial insult leading to the development of the
STEC. However, the vast majority of the treatments under thrombotic microangiopathy is damage by the Stx(s) to
development are designed to limit Stx receptor generation or to vascular endothelial cells that express the toxin receptor,
prevent toxin binding, trafficking, processing, or activity within globotriaosylceramide (Gb3). The Stx-mediated injury
the cell. The potential therapies described in this review include
to endothelial cells initiates a cascade of events that
some that have only been tested in vitro and several that
demonstrate efficacy in animals. The therapeutics that are
lead to the activation of platelets and the formation of
currently the furthest along in development (completed phase I thrombi in the small vessels of the kidney and sometimes
and II trials) are monoclonal antibodies directed against Stx1
and Stx2. Received: 10 July 2013, Accepted: 21 August 2013,
Published: 12 September 2014
Editors: Vanessa Sperandio, University of Texas Southwestern
Medical Center, Dallas, TX, and Carolyn J. Hovde, University of Idaho,
BACKGROUND Moscow, ID
Shiga toxin (Stx)-producing Escherichia coli (STEC) Citation: Melton-Celsa AR, O’Brien AD. 2014. New therapeutic
developments against Shiga toxin-producing Escherichia coli.
colonizes the intestine and causes hemorrhagic colitis. Microbiol Spectrum 2(5):EHEC-0013-2013. doi:10.1128
STEC encodes a variety of colonization factors, but /microbiolspec.EHEC-0013-2013.
a significant subset of STEC, the enterohemorrhagic Correspondence: Angela R. Melton-Celsa, angela.melton-celsa
.ctr@usuhs.edu
E. coli (EHEC) strains, have the locus of enterocyte
© 2014 American Society for Microbiology. All rights reserved.
effacement (LEE), the products of which allow the

ASMscience.org/MicrobiolSpectrum 1
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
Melton-Celsa and O’Brien

the central nervous system. Prevention of HUS is im- correlated with increased HUS risk (11). In contrast,
portant as HUS can lead to death or long-term conse- many people were treated with fosfomycin during a
quences such as hypertension and renal disease (1–3). large O157:H7 outbreak in Japan without an apparent
Because STEC strains are intestinal bacterial pathogens, increase in HUS, and perhaps even a protective effect
the inclination by physicians is to treat with antibiotics if the antibiotic was given within the first 2 days of illness
to eliminate the organisms from the gut. However, (12). However, the low overall HUS rate in the Sakai
several studies show that treatment of STEC-infected outbreak (13) suggests that the causative strain may
patients with certain antibiotics may lead to an increase (fortunately) have had reduced virulence compared with
in HUS (antibiotic use is discussed further in the next O157 strains from other outbreaks, and, as such, would
section). Therefore, the focus for therapeutics against make that outbreak a poor platform on which to make
STEC and HUS has been to find (i) compounds that act generalized decisions about treatment with antibiotics.
at the level of the bacterium but do not cause an increase During the O104:H4 outbreak in Germany in 2011
in Stx production; (ii) receptor mimics or other mole- caused by an unusual enteroaggregative E. coli (EAEC)
cules that alter trafficking of the toxin or the cellular strain that makes Stx2, antibiotics were used in many
response to the toxin; (iii) antibodies directed against the patients, some of whom were given up to three anti-
Stxs; or (iv) therapies to prevent or treat the HUS disease biotics at various times after disease onset (14). Because
process. The therapies discussed in this article are listed of the vastly different protocols used to treat patients
in Table 1. during the outbreak in Germany, it is difficult to make
generalized conclusions about antibiotic use based on
that outbreak; however, overall there was not defini-
THE DIFFICULTIES WITH ANTIBIOTIC tive evidence of a benefit due to antibiotic treatment.
USE FOR STEC INFECTIONS Although one surprising study asserted ciprofloxacin
In the United States, antibiotics are not recommended treatment reduced the HUS incidence in patients, the
for treatment of STEC infections because of the in- number of treated patients was small (n = 5), and in
creased risk for the development of HUS (4, 5). How- two of those patients HUS did develop (15). Further-
ever, even though antibiotics are contraindicated for more, in several patients from the O104:H4 outbreak
those with STEC infection, a recent study at FoodNet HUS was reported to develop after ciprofloxacin or
sites found that antibiotics are commonly used in those metronidazole treatment (16, 17). We consider the use
with proven O157 infection (6). The issue of the use of of ciprofloxacin to be especially dangerous because of
antibiotics to treat STEC infections is confounded by a the evidence that ciprofloxacin increases Stx expres-
number of factors: (i) some, but not all, antibiotics at sion in the EAEC O104:H4 strain (18) and STEC O157
sublethal doses increase the expression of Stx by in- strains. Finally, one unique aspect of the outbreak in
ducing the lysogenic phage that encodes the toxin genes Germany was the use of azithromycin to reduce shed-
(see diagram in Fig. 1) (7); (ii) treatment of STEC with a ding of O104:H4 in patients who appeared to be long-
lethal dose of antibiotics may cause release of a large term carriers (19).
bolus of toxin at one time as the bacteria die—a result Animal studies also give contradictory information
that would be bad for a patient; (iii) in some STEC about the use of antibiotics for the treatment of STEC
strains, the Stxs are either chromosomally encoded or infections. In two studies fosfomycin reduced coloniza-
are associated with defective bacteriophages, and thus tion and mortality from STEC infection in mice (20, 21),
antibiotic treatment may not alter Stx expression; (iv) but another study did not show a protective effect by
antibiotics may be used only in the most ill patients, a that antibiotic (22). In a mouse model in which the mice
fact that may skew the apparent risk for HUS develop- are starved for protein calories, the use of trimethoprim-
ment; and (v) published studies on the risk of HUS as- sulfamethoxazole was detrimental when given between
sociated with antibiotic treatment used different days 3 and 5 post infection, whereas the same antibiotic
treatments administered at different stages in the disease as well as ampicillin and fosfomycin were protective
process. In addition, although several studies demon- when given anywhere from days 1 to 5 after infection
strate an increased risk for HUS after treatment with (23). Nevertheless, we should note that the use of am-
antibiotics (8, 9), others do not find an increased risk picillin to treat an infection with an organism (Shigella
(10). Conversely, several studies indicate that the use of dysenteriae or the Stx2+ EAEC O104:H4, respectively)
β-lactam antibiotics, particularly in the first 2 to 3 days resistant to that antibiotic appears to be detrimental to
of illness, and perhaps associated with age <13 years, is humans and mice (24, 25).

2 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
New Therapeutic Developments against Shiga Toxin-Producing Escherichia coli

Overall, the current data do not support the use Similar to LED209, treatment of STEC infection with
of antibiotics for the treatment of STEC infection in the divalent cation zinc reduces adherence to a HeLa
humans, and the majority of the evidence indicates cell monolayer, though through a different mechanism,
that antibiotic treatment is potentially detrimental. The perhaps due to decreased expression of E. coli-secreted
empirical use of antibiotics could be dangerous, since proteins EspA and EspB (29). In addition to a reduction
some of these treatments could increase the risk of in adherence to cells, zinc treatment also appears to
HUS. Furthermore, since there is no clear demonstra- decrease expression of the stx genes. The potential
ble benefit to the use of antibiotics in STEC-infected therapeutic effect of zinc was tested in a rabbit ileal loop
patients, and because antibiotic use itself may pose model, in which rabbit enteropathogenic E. coli strains
possible risk to the patient (allergy or other risks), we transduced with a bacteriophage that encodes stx1 or
believe antibiotics should not be used to treat STEC stx2 were inoculated into the loop in the presence or
infections. absence of zinc. The presence of 1 mM of zinc reduced
the amount of toxin found in the loops, decreased ad-
herence, and lessened histological damage. The next
COMPOUNDS DIRECTED TOWARD STEC avenue to explore for zinc is to determine if the cation
could prevent disease or mortality in an oral infection
Pyocin
model.
A novel strategy to kill O157 strains based on a modified
pyocin (a bactericidal protein, similar to bacteriophage
tail fibers, made by Pseudomonas aeruginosa that usu- THERAPEUTICS THAT INTERFERE
ally targets other P. aeruginosa cells) that consists of WITH TOXIN BINDING, UPTAKE,
the R2 tail fiber fused to a phage spike protein specific TRAFFICKING, OR FUNCTION
for O157 was found to kill O157 strains without in- Stx1 and Stx2 are the key STEC virulence factors that
creasing expression of Stx (26). Subsequent tests dem- lead to the development of HUS. Therefore, therapeutics
onstrated that a slightly modified version of the pyocin, that impede toxin binding, uptake, trafficking, or func-
AvR2-V10.3, given 3 h after infection and once daily tion are strong contenders for treatment of STEC
for the next 2 days to infant rabbits infected with infections. Stx1 and Stx2 share about 56% homology at
O157:H7 strain EDL933 reduced colonization and pre- the amino acid level but are immunologically distinct.
vented diarrhea at the highest dose of pyocin admin- Structurally, the toxins consist of five identical B
istered (27). When the modified pyocin was given to subunits and a single A subunit. The B pentamer binds to
EDL933-infected infant rabbits that exhibited diarrhea, the cellular receptor, Gb3. The A subunit contains the
the amount of loose stool decreased relative to the enzymatic function of the toxin and removes an adenine
control animals given buffer alone and the numbers of residue from the 28S rRNA, an action that destroys
EDL933 organisms decreased in the intestines and ribosome function. After binding to Gb3, the toxin re-
stools. One problem with the pyocin approach is that it ceptor complex is taken up by clathrin-dependent and
is specific for serogroup, so other pyocins would have to -independent mechanisms. The toxin then traffics in a
be developed for non-O157 STEC strains. retrograde direction from the endosome to the Golgi
apparatus to the endoplasmic reticulum (ER). The A
A Small Molecule and a Divalent Cation subunit of the toxin can be nicked within the Golgi body
Another alternative strategy to antibiotics directed to- such that the enzymatic function (within A1) is separated
ward the bacterium uses a small molecule, LED209, by a disulfide bond from the A2 peptide that links A1 to
which inhibits the activity of the QseC sensor that is the B pentamer. The nicked toxin traffics next to the ER
involved in regulating some of the proteins involved in where the disulfide bond between A1 and A2 is reduced.
EHEC adherence and, indirectly, Stx2 expression. Al- The A1 subunit then enters the cytoplasm and targets the
though LED209 inhibited the formation of attaching ribosome. The damage to the ribosome halts protein
and effacing lesions by EHEC on HeLA cells, the com- synthesis and can lead to a ribotoxic stress response and
pound was not effective in reducing colonization or apoptosis (see review in reference 30). A model of Stx
disease in infant rabbits (28). The reason for LED209 trafficking is shown in Fig. 2.
failing to protect in the rabbits may be that the con- Stx1 and Stx2 are the major toxin types produced by
centration of the compound was not high enough at the STEC that cause human disease. There are subtypes of
site of EHEC colonization in the gut. both Stx1 and Stx2, however, and toxin nomenclature

ASMscience.org/MicrobiolSpectrum 3
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
4

Melton-Celsa and O’Brien


TABLE 1 Therapies directed against STEC, the Stx receptor, Stx function, or the cellular response to Stx

Target (step in Fig. 2 at


which therapeutic acts) Therapeutic Function Model used to test function or efficacy Reference(s)
STEC or STEC pathogenesis Pyocin (AvR2-V10.3) Kills O157 In vitro growth; infant rabbits 26, 27
LED209 Reduces attaching and effacing lesions on HeLa cells; infant rabbits 28
HeLa cells but did not reduce colonization
or disease in rabbits
Zinc Reduces stx transcription; HeLa cells; rabbit ileal loops (using rabbit 29
reduces adherence enteropathogenic E. coli transduced with
phage carrying stx1 or stx2)
Receptor (Gb3) C-9 Receptor generation; glucosylceramide HRTEC; Stx2-intoxicated rats 36, 37
synthesis inhibitor synthase
ASMscience.org/MicrobiolSpectrum

Receptor analogs (B) SYNSORB Pk Receptor analog Phase I and II trials 39, 41
STARFISH Receptor analog Vero cells; Stx1-intoxicated mice 42, 43
(does not protect Stx2-intoxicated mice)
Daisy Receptor analog Vero cells; Stx1- and Stx2-intoxicated mice; 43
B2F1-infected, streptomycin-treated mice
SUPER TWIG (1)6 Receptor analog Vero cells; Stx1- and Stx2-intoxicated mice; 44
O157-infected, protein-calorie-deficient mice
Gb3 polymers Receptor analog Mice 45
TVP (also known as Ac-PPP-tet) Receptor analog, Vero cells; O157-infected, 46, 49, 50
binds Stx2 but not Stx1 protein-calorie-deficient mice,
rabbit ileal loops; baboon
MMA-tet Receptor analog Vero cells; O157-infected, 51
protein-calorie-deficient mice
Probiotic that displays Receptor analog Vero cells; streptomycin-treated mice 47, 52–54
Galα1-4Galβ1-4Glc- infected with an Stx2 or Stx2dact producer
HuSAP Binds Stx2 but not Stx1 Stx1- or Stx2-intoxicated mice 57–59
(only protects Stx2-intoxicated animals)

Downloaded from www.asmscience.org by


IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
ASMscience.org/MicrobiolSpectrum
Antitoxin antibodies Polyclonal anti-Stx2 Neutralize Stx2 Gnotobiotic pigs 62
(B, U, T, or E) cαStx1 (human/mouse chimeric Block Stx1 binding (B subunit) Vero cells; Stx1-intoxicated mice; 64, 68, 70
version of 13C4) phase I and II trials
cαStx2 (anti-A subunit, Alters intracellular trafficking and Vero cells; streptomycin-treated mice; 63, 68–70
human/mouse chimeric inhibits enzymatic function phase I and II trials
of 11E10)
Anti-Stx1 (5-5B) Neutralize Stx1 (B subunit) Ramos cells 71
Urtoxazumab (also called Neutralize Stx2 (B subunit) Human renal 73–75
TMA-15; humanized adenocarcinoma cells; Stx2-intoxicated mice;
version of VTm1.1) B2F1-infected streptomycin-treated mice,
phase I trial
Anti-Stx1 monoclonals Neutralize Stx1 (most are anti-B subunit) HeLa cells; Stx1-intoxicated mice 76
Anti-Stx2 (5C12) Neutralize Stx2 (A subunit) HeLa cells; Stx2-intoxicated mice; 77, 78
B2F1-infected streptomycin-treated mice;
O157-infected gnotobiotic piglets
Toxin transport (U or T) Exo2 Blocks transport at the level of the early Vero cells 82, 83
endosome/trans-Golgi interface
Retro-2cycl Toxin transport HeLa cells 85, 86

New Therapeutic Developments against Shiga Toxin-Producing Escherichia coli


Chloroquine Toxin transport HEp-2 cells 87
Small molecules Toxin transport HeLa cells 88
Eeyarestatin 1 Intracellular trafficking HeLa cells 90
Nitrobenzyl-thioinosine Stx1 trafficking Human renal cortical epithelial cells 91
(retention in early endosomes)
Manganese Protects HeLa cells and BALB/c mice from HeLa cells; Stx1-intoxicated BALB/c mice 92, 93
Stx1-mediated lethality; blocks StxB1 (does not protect Stx2-intoxicated mice)
trafficking; does not protect against Stx2
Toxin processing (N) Furin inhibitor Cleavage of A subunit to A1 and A2 HEp-2 cells 94
Toxin function (E) Small molecule Enzymatic activity Cell-free reporter assay 89
Cellular or host response Imatinib Ribotoxic stress response inhibitor HCT-8 cells; infant rabbit 98
to toxin (ribotoxic stress Ouabain Apoptosis inhibition Rat proximal tubule cells (RPTC) 99
response or apoptosis)
Complement factor C5 Eculizumab Anticomplement factor C5 STEC-infected patients 101, 105, 106
5

Downloaded from www.asmscience.org by


IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
Melton-Celsa and O’Brien

FIGURE 1 Induction of the stx-encoding phage by antibiotics such as ciprofloxacin.


Subinhibitory concentrations of some antibiotics induce the lysogenic stx phage to
enter the lytic cycle, and as consequence, stx-bacteriophage are made and released.
Furthermore, 10- to 100-fold more toxin is made and released from the cell. doi:10.1128
/microbiolspec.EHEC-0013-2013.f1

was recently updated so that the prototype Stxs are now was observed (37). Treated rats also showed lower rises
designated Stx1a and Stx2a. The less specific Stx1 and in serum creatinine and urea and reduced renal tubular
Stx2 designations are used when the toxin subtype is injury.
unknown (31). We use the more general designations for
the toxins elsewhere in this review for simplicity. Besides Receptor Analogs
the prototypic Stx2a, two Stx2 subtypes are associated The first drug tested in humans intended for the treat-
with HUS, Stx2c and Stx2d (32, 33). Stx2c and Stx2d ment of HUS was SYNSORB Pk, a silicon dioxide com-
have two amino acid differences in the B subunit as pound that contains the trisaccharide component of
compared to Stx2a, and those changes are responsible Gb3 (38, 39). The theory behind the use of SYNSORB
for reduced cytotoxicity on Vero cells, though Stx2d is Pk is that the compound would bind up free Stx(s)
just as toxic to mice as Stx2a (34). Stx2d has two amino within the intestines of infected patients and prevent
acid differences from Stx2a and Stx2c in the A subunit, that toxin from binding to the functional receptor so
and those two changes contribute to the capacity of the that the toxin could not act either locally or systemically.
toxin to exhibit increased toxicity when treated with SYNSORB Pk was shown to neutralize both Stx1 and
elastase from intestinal mucus (35), a phenotype indi- Stx2 on human renal adenocarcinoma cells (40). Al-
cated with the designation Stx2dact. though SYNSORB Pk was tolerated well in the phase I
trial (41), the double-blind placebo-controlled trial sug-
Receptor Synthesis Inhibitor: C-9 gested no difference between treated and placebo groups
One step at which to stop Stx is at the level of the toxin (39). The reason for the lack of efficacy by SYNSORB Pk
receptor. Various laboratories have tried to protect cells in the latter study may be that the patients were enrolled
or animals from Stxs with compounds that prevent after HUS diagnosis, whereas the best time to neutralize
Gb3 synthesis or that bind to the toxin to prevent toxin- toxin to prevent HUS is most likely before development
receptor interaction. For example, a molecule called of this serious sequela.
C-9 was used in human renal tubular epithelial cells A number of other Stx receptor analogs in addition to
(HRTEC) to prevent the conversion of ceramide to glu- SYNSORB Pk have been developed, such as STARFISH
cosylceramide, an early step in Gb3 synthesis. HRTEC (42), Daisy (43), SUPER TWIG (1)6 (44), Gb3 polymers
pretreated with C-9 for 24 h exhibited reduced levels (45), and Ac-PPPtet (46), as well as a probiotic that
of Gb3 and decreased sensitivity to Stx2 (36). In rats displays an Stx binder on its surface (47). STARFISH is a
treated with C-9 for 2 days before intoxication and for 4 five-“armed” molecule with two receptor mimics at the
days post intoxication, about 50% protection from in- end of each arm; the compound appears to bind two B
jection with bacterial supernatants that contained Stx2 pentamers at the same time and neutralizes both Stx1

6 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
New Therapeutic Developments against Shiga Toxin-Producing Escherichia coli

FIGURE 2 Cellular trafficking of Stx and points in the pathway where therapeutics func-
tion. Therapeutics can interfere with Stx action at several points as it traffics into and
through the cell. The steps in toxin trafficking are briefly diagramed above and outlined
here. The toxin first binds (B) to the receptor Gb3. The toxin/Gb3 complex is taken up (U)
by both clathrin-dependent and -independent mechanisms, and then traffics (T) from the
early endosome to the late endosome to the trans-Golgi apparatus. Within the Golgi the
toxin A subunit is nicked (N), but the toxin remains intact due to a disulfide bond between
the A1 and A2 subunits. The nicked toxin continues to traffic (T) along the retrograde
pathway to the endoplasmic reticulum. The disulfide bond in the A subunit is reduced (R)
within the endoplasmic reticulum and the A1 subunit enters the cytoplasm where it exerts
its enzymatic (E) attack and depurinates the ribosome. The action of the toxin within the
cell can lead to a ribotoxic stress response (RSR) and apoptosis (APOP). doi:10.1128
/microbiolspec.EHEC-0013-2013.f2

and Stx2 in vitro. However, STARFISH has a higher potential therapeutics in an animal model system. To
avidity for Stx1 than Stx2 and was only able to protect find a receptor analog that could be given orally, the
mice injected with Stx1 but not Stx2 (43). Because of the Nishikawa group developed Gb3 polymers that bind to
lack of protective efficacy by STARFISH in the Stx2- the Stxs with higher affinity than SUPER TWIG (1)6,
injection model, a similar but modified divalent trisac- and those polymers, when given by gavage twice daily in
charide inhibitor called Daisy was synthesized (43). the protein-calorie-deficient mouse model on days 3 to 5
Daisy protects Vero cells and mice from both Stx1 and after infection, protect mice from death (45). The strong
Stx2, and in a streptomycin-treated mouse model, neutralization effect observed with SUPER TWIG and
prevented death due to infection with O91:H21 STEC the Gb3 polymers is due to the fact that the B pentamer
strain B2F1 in 50% of the animals (43). The SUPER of the Stxs contains multiple Gb3-binding sites, so re-
TWIG (1)6 identified by Nishikawa’s group carries six ceptor mimics that display multiple copies of the Gb3
trisaccharides and blocks the binding of both Stx1 and trisaccharide bind the toxin tightly. A similar approach
Stx2 to Vero cells, protects mice from Stx2 when ad- as described above was used by another group to display
ministered together with the toxin, and prevents death of the receptor sugar moiety on chitosan, and they similarly
O157:H7-infected mice in a protein-calorie-deficient found that the analog neutralizes in vitro and in vivo
animal model when given intravenously after infection (48).
(44). However, one point to note is that SUPER TWIG According to Nishikawa, however, a drawback to the
(1)12, developed in the latter study, neutralized as well multiple trisaccharide display approach is the complex-
as SUPER TWIG (1)6 in vitro but did not protect in vivo, ity of synthesis (46, 49); therefore, a tetravalent peptide
a finding that demonstrates the importance of testing library was screened to find molecules that bind the Stx2

ASMscience.org/MicrobiolSpectrum 7
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
Melton-Celsa and O’Brien

pentamer (49). In that latter screen, tetravalent peptides Stx2d on Vero cells. In addition, streptomycin-treated
were identified that form a complex with Stx2 and neu- mice are protected from infection with either an Stx2-
tralize the toxin on Vero cells but do not prevent the or Stx2d-producer when fed the Galα1-4Galβ1-4Glc-
toxin/peptide moiety from binding to the cell. Rather, probiotic twice daily. What was not clear in that
the peptide-bound Stx2 was unable to reach the ER. study was whether the mice would continue to do well
Furthermore, O157-infected protein-calorie-deficient mice once the probiotic was discontinued. However, the pro-
gavaged after infection with the peptides were protected biotic with a receptor-analog approach might be pos-
from death; the protection appeared to be dose depen- sible as long as the infecting strain did not persist for
dent and treatment had to be started by day 3 post long-term colonization. Additionally, it may be neces-
infection. Further testing of the optimized tetrapeptide, sary to monitor shedding of the infecting strain over
Ac-PPP-tet (later renamed TVP), demonstrated that time. CWG308:pJCP-Gb3 was found to be effective
the compound inhibits Stx2-mediated fluid accumu- even when killed prior to administration; however,
lation in rabbit ileal loops (46). However, the tetra- the formaldehyde-treated probiotic had to be adminis-
valent peptide only protects when administered orally tered three times daily for complete protection (53).
and not intravenously in the protein-calorie-deficient To develop a probiotic that might be able to be used in
mouse model, so TVP efficacy was tested in the baboon humans, a K-12 strain that expressed the same Galα1-
model of intoxication (50). Baboons given Stx2 and TVP 4Galβ1-4Glc- epitope on its LPS was created (54). In
simultaneously were protected from death, renal injury, that strain, additional mutations were incorporated so
and thrombocytopenia, but not anemia. TVP also res- that no antibiotic resistance markers were needed for
cued 75% of animals that received the drug 24 h post plasmid maintenance. The K-12 probiotic that displays
intoxication and a supplemental dose on days 2, 3, and Galα1-4Galβ1-4Glc- neutralizes both in vitro and in
4. Untreated but intoxicated baboons died by day 6. The vivo (54).
authors of that study suggest that the advantage of TVP
compared to other Stx binders is that the compound is Human Serum Amyloid Component P (HuSAP)
cell-permeable. However, it is unclear how TVP would Reports that there is an Stx2-neutralizing component in
rescue intoxicated cells since the proposed mechanism normal human serum that was not immunoglobulin
for action is for the Stx2-TVP complex to traffic differ- were published in 1993 (55, 56). Later, HuSAP was
ently within the cell than Stx2 alone, and indeed, in identified as the protein from plasma that can neutral-
vitro, cells are only protected if the TVP is added to ize Stx2, but not Stx1 (57). HuSAP administered intra-
cells at the same time as Stx2. In a recent study, the venously protects BALB/c mice given Stx2 1 h later (58).
Nishikawa group identified another tetravalent peptide, Another group showed similar results, though they ad-
MMA-tet, that inhibits both Stx1 and Stx2 on Vero cells ministered the HuSAP twice daily intraperitoneally and
when it is added at the same time as the toxins, and when the Stx2 by the subcutaneous route (59). This group
administered orally, protects protein-calorie-deficient then challenged transgenic C57Bl/6 mice that express
mice infected with STEC strain N-9 (51). The mecha- HuSAP with two 50% lethal doses of Stx2. The HuSAP-
nism whereby MMA-tet protects cells and animals is transgenic mice survived nearly twice as long as the
unclear: MMA-tet forms a complex with the Stx1 B control mice in that study. The reason that the HuSAP-
subunit but does not appear to alter binding or traf- injected BALB/c mice survived longer than the HuSAP-
ficking of the MMA-tet–StxB1 through the cell, at least transgenic mice was hypothesized to be because the
as far as the ER. However, MMA-tet pretreatment of circulating levels of HuSAP were higher in the BALB/c
Vero cells did prevent protein synthesis inhibition by mice injected with HuSAP than in the transgenic ani-
Stx1, a finding that may indicate that MMA-tet some- mals. If, however, the HuSAP-transgenic mice were in-
how prevents the toxin A1 subunit from reaching the jected with a combination of LPS and Stx2, the animals
cytoplasm from the ER. were not protected, even though the LPS did not prevent
Finally, a novel modification of the receptor analog HuSAP-Stx2 interaction or the neutralization by HuSAP
approach to bind up Stx specifically within the intes- of Stx2 for Vero cells in vitro (60). Exactly how HuSAP
tine was designed by Paton’s group: the core trisaccha- binds Stx2 is not clear; Marcato et al. reported that the
ride from Gb3, Galα1-4Galβ1-4Glc-, was displayed on interaction requires both toxin A and B subunits and
the lipopolysaccharide (LPS) structure of a commensal cannot be competed with Daisy (61). The latter finding
E. coli to create a probiotic (52). The constructed strain, suggests that HuSAP does not bind within the Gb3-
CWG308:pJCP-Gb3, neutralizes Stx1, Stx2, Stx2c, and binding sites on the B pentamer.

8 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
New Therapeutic Developments against Shiga Toxin-Producing Escherichia coli

ANTIBODIES TO THE Stxs group also evaluated isotype variants and Fab and F(ab′)2
Because the Stxs are the primary factors responsible for fragments of 5C12 in in vitro and in vivo models of ef-
the development of HUS, neutralization of the toxins ficacy and found that all of the isotype variants demon-
is a critical therapeutic approach for STEC infections. strate in vitro and in vivo neutralization but the Fab and
Polyclonal antiserum against Stx2 protects gnotobiotic F(ab′)2 were only efficacious in the in vitro assay (81).
piglets from infection by O157:H7 strain 86-24 (62). One point of note from the latter study is that the IgG4
Furthermore, monoclonal antibodies to the toxins are variant of 5C12 was the least protective of the isotype
protective in animal models, and two groups have taken variants in vitro, but was as protective in vivo as the best
such therapeutics into phase II safety trials, as detailed in vitro neutralizer (the IgG3 variant). These latter results
below. indicate once again that in vitro and in vivo neutraliza-
Monoclonal antibodies specific for the Stx1 B subunit tion data do not always correlate.
(13C4) and the Stx2 A subunit (11E10) were developed
in the O’Brien laboratory in the mid to late 1980s (63,
64). Those antibodies neutralize the toxins on Vero cells Nonantibody Inhibitors of Toxin Trafficking
(65, 66), and the Stx2 monoclonal antibody is protective or the Cellular Response to Stx
in mice (67). The epitopes for both antibodies have been Brefeldin A (BFA), a fungal toxin that disrupts the Golgi
mapped (65, 66); of note, although 11E10 neutralizes apparatus but also causes tabulation of early endo-
Stx2 in a cell-free protein synthesis assay, the antibody somes, is a known inhibitor of Stx transport; in fact,
also alters the intracellular localization of the toxin such the protective effect of BFA against the Stxs helped
that the toxin-antibody complex does not reach the cy- define the pathway by which Stx is transported through
toplasm (65). Both 11E10 and 13C4 were transformed the cell. Because BFA is a global inhibitor of protein
into human/mouse chimeras by genetic techniques. The transport within the cell, it is not included in Table 1.
“humanized” versions of anti-Stx1 and anti-Stx2 neu- Another Golgi disruptor, Exo2, a small molecule that
tralize the toxins on Vero cells and in either a mouse does not inhibit cholera toxin trafficking, prevents Stx
intoxication model (Stx1) or the streptomycin-treated from reaching the Golgi apparatus (82). Unfortunately,
mouse model of infection with strain B2F1 (68). The cell disruptors such as BFA and Exo2 are toxic to cells.
humanized versions of the antibodies (cαStx1 for 13C4 Another drawback to molecules such as BFA and Exo2
and cαStx2 for 11E10) were evaluated in phase I (69, 70) is that treated cells recover over time, such that incu-
and II trials. The preliminary results of the phase II trial bation of the cells with the inhibitor and the toxin for
indicate that the antibodies were safe and well tolerated longer periods reduces the effectiveness of the thera-
in sick children infected with STEC. peutic. Exo2 was recently derivatized to generate an
Takeda’s group developed monoclonal antibodies inhibitor with lower toxicity that still interferes with Stx
that neutralize Stx1 or Stx2 by preventing receptor trafficking (83). Another small molecule, Retro-2, blocks
binding (71, 72). The anti-Stx2 monoclonal was later Stx from reaching the trans-Golgi network (84, 85) and
humanized and named TMA-15. TMA-15 demonstrates is effective against ricin, a toxin that traffics in the same
Vero cell neutralization and protective efficacy in ani- manner as the Stxs. Recently, a derivative of Retro-2,
mal models (73, 74). Antibody TMA-15 was renamed Retro-2cycl, was identified. Retro-2cycl is 100-fold more
urtoxazumab and evaluated in safety trials in healthy active on HeLa cells than Retro-2 (86). However, the
adults and STEC-infected children (75). Urtoxazumab drawback to Retro-2cycl and derivatives is that the cells
was well tolerated in STEC-infected children, but no needed to be pretreated with the drug to observe pro-
efficacy data are yet published. tection from the toxin (84, 86). Another compound
Finally, Tzipori’s group generated fully humanized known to protect cells from Stx, chloroquine, was also
neutralizing monoclonal anti-Stx1 and -Stx2 antibodies recently shown to permit the toxin to reach the ER (87).
in transgenic mice (76, 77). The anti-Stx2 monoclonal Since chloroquine does not inhibit the enzymatic activity
antibody, 5C12, was developed further, and shown to of Stx nor degrade the toxin in vitro, the drug likely acts
protect piglets and streptomycin-treated mice from by inhibiting the toxin from exiting the ER.
STEC strains that produce Stx2 or Stx2dact, respectively Further small-molecule screens have identified other
(77, 78). Similarly to 11E10, antibody 5C12 neutralizes compounds that inhibit Stx and ricin transport within
Stx2 by altering its intracellular trafficking pattern and Vero cells (88) or the enzymatic activity of both toxins
also has the capacity to prevent protein synthesis inhi- (which have the same mode of action) (89). Another
bition by Stx2 in a cell-free assay (79, 80). The Tzipori small molecule, eeyarestatin I, which interferes with

ASMscience.org/MicrobiolSpectrum 9
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
Melton-Celsa and O’Brien

intracellular trafficking among cellular compartments, the evaluation of therapeutics. However, such a model
was shown to cause a lag in protein synthesis inhibi- would be expensive, and determining effective thera-
tion in HeLa cells by Stx (90). Finally, nitrobenzylthio- peutic doses and the timing for those doses might prove
inosine was shown in 2002 to inhibit the trafficking challenging.
of Stx1, trapping the toxin within the early endosome A recent paper suggests that an inhibitor of apoptosis,
(91); however, no additional information on the use ouabain, protects rat renal proximal tubules from Stx-
of this compound to protect cells or animals has been mediated cell death (99). Kidneys from ouabain-treated
published. mice that were given about four 50% lethal doses of
A recent report suggests that manganese inhibits in- Stx2 showed reduced podocyte depletion as compared
tracellular trafficking of the B subunit of Stx (92), a to phosphate-buffered saline–treated mice. However, no
finding that may or may not extend to the holotoxin. data were reported on whether the ouabain could pro-
However, manganese treatment increased the viability tect the mice from Stx2-mediated lethality.
of HeLa cells incubated with Stx1 in that study and
protected BALB/c mice injected with 500 ng of Stx1
and injected with daily doses of manganese (at least TREATMENT ONCE HUS IS DIAGNOSED
10 mg/kg daily injections were required). Whether such What do you do for patients who already have HUS? We
levels of manganese would be reasonably achievable in leave discussion of specific clinical interventions such as
a patient is not clear. In addition, manganese fails to dialysis and apheresis for medical experts. In terms of a
protect HeLa cells from Stx2 intoxication (93), a finding therapeutic that may interfere in the etiology of HUS,
that makes the potential use of manganese less likely as Lapeyraque and colleagues reported in 2011 the use of
Stx2 is more commonly associated with HUS develop- eculizumab (an antibody directed against complement
ment than Stx1. protein C5 used to treat atypical HUS, a disease that
may arise due to complement dysregulation) in three
children with HUS due to STEC infection (100). Al-
A COMPOUND THAT INTERFERES though the sick children exhibited improvements in
WITH TOXIN PROCESSING platelet counts and reductions in lactate dehydrogenase,
Because the A subunit must be cleaved by a furin- or plasma exchange and hemodialysis were used concur-
trypsin-like protease so that the A1 or enzymatically rently. In addition, no children in the small study re-
active moiety of the toxin may be separated from the ceived just antibody infusion alone. Eculizumab was
holotoxin, furin inhibitors were tested in a cell-based also used to treat many patients with HUS during the
assay for the capacity to protect HEp-2 cells from Stx O104:H4 outbreak in Germany in 2011. Although there
(94). Although one of the inhibitors showed moderate was no evidence of efficacy for eculizumab in treating
inhibition activity against Stx, the inhibition was over- HUS from the outbreak in Germany, the cohorts are
come at higher concentrations of Stx. Furthermore, since difficult to compare because the patients received other
the toxin can be cleaved by enzymes in intestinal mucus concurrent treatments, such as plasma exchange, anti-
(35), as well as by intracellular proteases (95), and be- biotic therapy, immunoglobulin G immunoadsorption,
cause Stx mutated at the trypsin-sensitive site can still be and dialysis (14, 101–105). In addition, many of the
cleaved (96, 97), we suspect that it will be difficult to HUS patients given eculizumab were quite ill and had
protect animals from Stx with protease inhibitors. neurological complications. Therefore, a randomized
controlled trial is necessary to answer the question
Therapies That Interfere with about the efficacy of eculizumab for the treatment of Stx-
the Cellular Response to Stx associated HUS.
Inhibitors of mitogen-activated protein kinase pathways
were used to assess potential protective efficacy against
the Stx2-mediated ribotoxic stress response in HCT- CONCLUSION
8 cells or for oral gavage of Stx2 into infant rabbits (98). Since the first outbreak of STEC infection in the United
The authors observed modest protective capacity by States in 1982, much research has focused on ways to
imatinib in HCT-8 cells and against some aspects of Stx2 neutralize the action of the Stxs. At this time, only
intoxication in the animals, specifically heterophil infil- SYNSORB Pk, urtoxazumab, and the Shiga monoclonal
tration into the intestine. That study indicates that the antibodies cαStx1 and cαStx2 have been tested in phase I
infant rabbit model of Stx2 gavage may prove useful for and II trials intended to treat or prevent HUS. Efforts to

10 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
New Therapeutic Developments against Shiga Toxin-Producing Escherichia coli

prevent STEC infection, such as elimination from the Haller H, Herget-Rosenthal S, Hertenstein B, Hofmann C, Lang M,
food supply and proper food handling, are important as Kielstein JT, Klostermeier UC, Knobloch J, Kuehbacher M, Kunzendorf
U, Lehnert H, Manns MP, Menne TF, Meyer TN, Michael C, Munte T,
well as we try to stop the development of the potentially Neumann-Grutzeck C, Nuernberger J, Pavenstaedt H, Ramazan L,
deadly HUS. Lastly, consideration should be given to the Renders L, Repenthin J, Ries W, Rohr A, LRump LC, Samuelsson O, Sayk
development of a vaccine against the Stxs, minimally for F, Schmidt BM, Schnatter S, Schocklmann H, Schreiber S, von Seydewitz
CU, Steinhoff J, Stracke S, Suerbaum S, van de Loo A, Vischedyk M,
those in research or clinical labs who are exposed to Weissenborn K, Wellhoner P, Wiesner M, Zeissig S, Buning J, Schiffer M,
STEC. Kuehbacher T. 2012. Validation of treatment strategies for entero-
haemorrhagic Escherichia coli O104:H4 induced haemolytic uraemic
ACKNOWLEDGMENT syndrome: case-control study. BMJ 345:e4565.
We declare no conflicts of interest with regard to the manuscript. 15. Geerdes-Fenge HF, Lobermann M, Nurnberg M, Fritzsche C,
Koball S, Henschel J, Hohn R, Schober HC, Mitzner S, Podbielski A,
Reisinger EC. 2013. Ciprofloxacin reduces the risk of hemolytic uremic
REFERENCES
syndrome in patients with Escherichia coli O104:H4-associated diarrhea.
1. Rosales A, Hofer J, Zimmerhackl LB, Jungraithmayr TC, Riedl M,
Infection 41:669–673.
Giner T, Strasak A, Orth-Holler D, Wurzner R, Karch H. 2012. Need for
long-term follow-up in enterohemorrhagic Escherichia coli-associated 16. Binks S, Regan K, Richenberg J, Chevassut T. 2012. Microbes without
hemolytic uremic syndrome due to late-emerging sequelae. Clin Infect Dis frontiers: severe haemolytic-uraemic syndrome due to E coli O104:H4.
54:1413–1421. BMJ Case Rep 2012.
2. Palermo MS, Exeni RA, Fernandez GC. 2009. Hemolytic uremic syn- 17. Colic E, Dieperink H, Titlestad K, Tepel M. 2011. Management of an
drome: pathogenesis and update of interventions. Expert Rev Anti Infect acute outbreak of diarrhoea-associated haemolytic uraemic syndrome
Ther 7:697–707. with early plasma exchange in adults from southern Denmark: an ob-
servational study. Lancet 378:1089–1093.
3. Spinale JM, Ruebner RL, Copelovitch L, Kaplan BS. 2013. Long-term
outcomes of Shiga toxin hemolytic uremic syndrome. Pediatr Nephrol 18. Bielaszewska M, Idelevich EA, Zhang W, Bauwens A, Schaumburg F,
28:2097–2105. Mellmann A, Peters G, Karch H. 2012. Effects of antibiotics on Shiga
toxin 2 production and bacteriophage induction by epidemic Escherichia
4. Thielman NM, Guerrant RL. 2004. Clinical practice. Acute infectious
coli O104:H4 strain. Antimicrob Agents Chemother 56:3277–3282.
diarrhea. N Engl J Med 350:38–47.
19. Nitschke M, Sayk F, Hartel C, Roseland RT, Hauswaldt S, Steinhoff J,
5. Molbak K, Mead PS, Griffin PM. 2002. Antimicrobial therapy in
Fellermann K, Derad I, Wellhoner P, Buning J, Tiemer B, Katalinic A,
patients with Escherichia coli O157:H7 infection. JAMA 288:1014–1016.
Rupp J, Lehnert H, Solbach W, Knobloch JK. 2012. Association between
6. Nelson JM, Griffin PM, Jones TF, Smith KE, Scallan E. 2011. Anti- azithromycin therapy and duration of bacterial shedding among patients
microbial and antimotility agent use in persons with Shiga toxin- with Shiga toxin-producing enteroaggregative Escherichia coli O104:H4.
producing Escherichia coli O157 infection in FoodNet sites. Clin Infect JAMA 307:1046–1052.
Dis 52:1130–1132.
20. Isogai E, Isogai H, Hayashi S, Kubota T, Kimura K, Fujii N, Ohtani
7. Karch H, Goroncy-Bermes P, Opferkuch W, Kroll H-P, O’Brien A. T, Sato K. 2000. Effect of antibiotics, levofloxacin and fosfomycin, on a
1985. Subinhibitory concentrations of antibiotics modulate amount of mouse model with Escherichia coli O157 infection. Microbiol Immunol
Shiga-like toxin produced by Escherichia coli, p 239–245. In Adam D, 44:89–95.
Hahn H, Opferkuch W (ed), The Influence of Antibiotics on the Host-
21. Zhang X, McDaniel AD, Wolf LE, Keusch GT, Waldor MK,
Parasite Relationship II. SpringerVerlag, Berlin, Germany.
Acheson DW. 2000. Quinolone antibiotics induce Shiga toxin-encoding
8. Wong CS, Jelacic S, Habeeb RL, Watkins SL, Tarr PI. 2000. The risk of bacteriophages, toxin production, and death in mice. J Infect Dis 181:
the hemolytic-uremic syndrome after antibiotic treatment of Escherichia 664–670.
coli O157:H7 infections. N Engl J Med 342:1930–1936. 22. Yoshimura K, Fujii J, Taniguchi H, nd S. Yoshida S. 1999. Chemo-
9. Slutsker L, Ries AA, Malone K, Wells JG, Greene KD, Griffin PM. therapy for enterohemorrhagic Escherichia coli O157:H infection in a
1998. A nationwide case-control study of Escherichia coli O157:H7 in- mouse model. FEMS Immunol Med Microbiol 26:101–108.
fection in the United States. J Infect Dis 177:962–966. 23. Kurioka T, Yunou Y, Harada H, Kita E. 1999. Efficacy of antibiotic
10. Panos GZ, Betsi GI, Falagas ME. 2006. Systematic review: are therapy for infection with Shiga-like toxin-producing Escherichia coli
antibiotics detrimental or beneficial for the treatment of patients with O157:H7 in mice with protein-calorie malnutrition. Eur J Clin Microbiol
Escherichia coli O157:H7 infection? Aliment Pharmacol Ther 24:731–742. Infect Dis 18:561–571.
11. Smith KE, PWilke PR, Reiter PL, Hedican EB, Bender JB, Hedberg 24. Zangari T, Melton-Celsa AR, Panda A, Boisen N, Smith MA, Taratov
CW. 2012. Antibiotic treatment of Escherichia coli O157 infection and I, De Tolla LJ, Nataro JP, O’Brien AD. 2013. Virulence of the Shiga toxin
the risk of hemolytic uremic syndrome, Minnesota. Pediatr Infect Dis J type 2-expressing Escherichia coli O104:H4 German outbreak isolate in
31:37–41. two animal models. Infect Immun 81:1562–1574.
12. Ikeda K, Ida O, Kimot K, Takatorige T, Nakanishi N, Tatar K. 1999. 25. Al-Qarawi S, Fontaine RE, Al-Qahtani MS. 1995. An outbreak of
Effect of early fosfomycin treatment on prevention of hemolytic uremic hemolytic uremic syndrome associated with antibiotic treatment of hos-
syndrome accompanying Escherichia coli O157:H7 infection. Clin pital inpatients for dysentery. Emerg Infect Dis 1:138–140.
Nephrol 52:357–362. 26. Scholl D, Cooley M, Williams SR, Gebhart D, Martin D, Bates A,
13. Fukushima H, Hashizume T, Morita Y, Tanaka J, Azuma K, Mandrell R. 2009. An engineered R-type pyocin is a highly specific and
Mizumoto Y, Kaneno M, Matsuura M, Konma K, Kitani T. 1999. sensitive bactericidal agent for the food-borne pathogen Escherichia coli
Clinical experiences in Sakai City Hospital during the massive outbreak of O157:H7. Antimicrob Agents Chemother 53:3074–3080.
enterohemorrhagic Escherichia coli O157 infections in Sakai City, 1996. 27. Ritchie JM, Greenwich JL, Davis BM, Bronson RT, Gebhart D,
Pediatr Int 41:213–217. Williams SR, Martin D, Scholl D, Waldor MK. 2011. An Escherichia coli
14. Menne J, Nitschke M, Stingele R, Abu-Tair M, Beneke J, Bramstedt J, O157-specific engineered pyocin prevents and ameliorates infection by
Bremer JP, Brunkhorst R, Busch V, Dengler R, Deuschl G, Fellermann K, E. coli O157:H7 in an animal model of diarrheal disease. Antimicrob
Fickenscher H, Gerigk C, Goettsche A, Greeve J, Hafer C, Hagenmuller F, Agents Chemother 55:5469–5474.

ASMscience.org/MicrobiolSpectrum 11
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
Melton-Celsa and O’Brien

28. Rasko DA, Moreira CG, Li de R, Reading NC, Ritchie JM, Waldor 45. Watanabe M, Matsuoka K, Kita E, Igai K, Higashi N, Miyagawa A,
MK, Williams N, Taussig R, Wei S, Roth M, Hughes DT, Huntley JF, Watanabe T, Yanoshita R, Samejima Y, Terunuma D, Natori Y,
Fina MW, Falck JR, Sperandio V. 2008. Targeting QseC signaling and Nishikawa K. 2004. Oral therapeutic agents with highly clustered
virulence for antibiotic development. Science 321:1078–1080. globotriose for treatment of Shiga toxigenic Escherichia coli infections.
29. Crane JK, Byrd IW, Boedeker EC. 2011. Virulence inhibition by zinc J Infect Dis 189:360–368.
in Shiga-toxigenic Escherichia coli. Infect Immun 79:1696–1705. 46. Watanabe-Takahashi M, Sato T, Dohi T, Noguchi N, Kano F,
30. Tesh VL. 2012. The induction of apoptosis by Shiga toxins and ricin. Murata M, Hamabata T, Natori Y, Nishikawa K. 2010. An orally ap-
Curr Top Microbiol Immunol 357:137–178. plicable Shiga toxin neutralizer functions in the intestine to inhibit the
31. Scheut F, Teel LD, Beutin L, Pierard D, Buvens G, Karch H, Mellmann intracellular transport of the toxin. Infect Immun 78:177–183.
A, Caprioli A, Tozzoli R, Morabito S, Strockbine NA, Melton-Celsa AR, 47. Paton AW, Morona R, Paton JC. 2001. Neutralization of Shiga toxins
Sanchez M, Persson S, O’Brien AD. 2012. Multicenter evaluation of a Stx1, Stx2c, and Stx2e by recombinant bacteria expressing mimics of
sequence-based protocol for subtyping Shiga toxins and standardizing Stx globotriose and globotetraose. Infect Immun 69:1967–1970.
nomenclature. J Clin Microbiol 50:2951–2963. 48. Li X, Wu P, Cheng S, Lv X. 2012. Synthesis and assessment of
32. Bielaszewska M, Friedrich AW, Aldick T, Schurk-Bulgrin R, Karch H. globotriose-chitosan conjugate, a novel inhibitor of Shiga toxins produced
2006. Shiga toxin activatable by intestinal mucus in Escherichia coli iso- by Escherichia coli. J Med Chem 55:2702–2710.
lated from humans: predictor for a severe clinical outcome. Clin Infect Dis 49. Nishikawa K, Watanabe M, Kita E, Igai K, Omata K, Yaffe MB,
43:1160–1167. Natori Y. 2006. A multivalent peptide library approach identifies a novel
33. Friedrich AW, Bielaszewska M, Zhang WL, Pulz M, Kuczius T, Shiga toxin inhibitor that induces aberrant cellular transport of the toxin.
Ammon A, Karch H. 2002. Escherichia coli harboring Shiga toxin 2 gene FASEB J 20:2597–2599.
variants: frequency and association with clinical symptoms. J Infect Dis 50. Stearns-Kurosawa DJ, Collins V, Freeman S, Debord D, Nishikawa K,
185:74–84. Oh SY, Leibowitz CS, Kurosawa S. 2011. Rescue from lethal Shiga toxin
34. Lindgren SW, Samuel JE, Schmitt CK, O’Brien AD. 1994. The specific 2-induced renal failure with a cell-permeable peptide. Pediatr Nephrol
activities of Shiga-like toxin type II (SLT-II) and SLT-II-related toxins of 26:2031–2039.
enterohemorrhagic Escherichia coli differ when measured by Vero cell 51. Tsutsuki K, Watanabe-Takahashi M, Takenaka Y, Kita E, Nishikawa
cytotoxicity but not by mouse lethality. Infect Immun 62:623–631. K. 2013. Identification of a peptide-based neutralizer that potently inhibits
35. Melton-Celsa AR, Darnell SC, O’Brien AD. 1996. Activation of Shiga- both Shiga toxins 1 and 2 by targeting specific receptor-binding regions.
like toxins by mouse and human intestinal mucus correlates with virulence Infect Immun 81:2133–2138.
of enterohemorrhagic Escherichia coli O91:H21 isolates in orally infected, 52. Paton AW, Morona R, Paton JC. 2000. A new biological agent for
streptomycin-treated mice. Infect Immun 64:1569–1576. treatment of Shiga toxigenic Escherichia coli infections and dysentery in
36. Silberstein C, Copeland DP, Chiang W-L, Repetto HA, Ibarra C. humans. Nat Med 6:265–270.
2008. A glucosylceramide synthase inhibitor prevents the cytotoxic effects 53. Paton JC, Rogers TJ, Morona R, Paton AW. 2001. Oral adminis-
of Shiga toxin-2 on human renal tubular epithelial cells. J Epithel Biol tration of formaldehyde-killed recombinant bacteria expressing a mimic of
Pharmacol 1:71–75. the Shiga toxin receptor protects mice from fatal challenge with Shiga-
37. Silberstein C, Lucero MS, Zotta E, Copeland DP, Lingyun L, Repetto toxigenic Escherichia coli. Infect Immun 69:1389–-1393.
HA, Ibarra C. 2011. A glucosylceramide synthase inhibitor protects rats 54. Pinyon RA, Paton JC, Paton AW, JBotten JA, Morona R. 2004.
against the cytotoxic effects of Shiga toxin 2. Pediatr Res 69:39–394. Refinement of a therapeutic Shiga toxin-binding probiotic for human
38. Armstrong GD, Fodor E, Vanmaele R. 1991. Investigation of Shiga- trials. J Infect Dis 189:1547–1555.
like toxin binding to chemically synthesized oligosaccharide sequences. 55. Bitzan M, Klemt M, Steffens R, Muller-Wiefel DE. 1993. Differences
J Infect Dis 164:1160–1167. in verotoxin neutralizing activity of therapeutic immunoglobulins and sera
39. Trachtman H, Cnaan A, Christen E, Gibbs K, Zhao S, Acheson DW, from healthy controls. Infection 21:140–145.
Weiss R, Kaskel FJ, Spitzer A, Hirschman GH. 2003. Effect of an oral 56. Bitzan M, Ludwig K, Klemt M, Konig H, Buren J, Muller-Wiefel DE.
Shiga toxin-binding agent on diarrhea-associated hemolytic uremic syn- 1993. The role of Escherichia coli O157 infections in the classical
drome in children: a randomized controlled trial. JAMA 290:1337–1344. (enteropathic) haemolytic uraemic syndrome: results of a Central Euro-
40. Takeda T, Yoshino K, Adachi E, Sato Y, Yamagata K. 1999. In vitro pean, multicentre study. Epidemiol Infect 110:183–196.
assessment of a chemically synthesized Shiga toxin receptor analog at- 57. Kimura T, Tani S, Matsumoto Yi Y, Takeda T. 2001. Serum amyloid
tached to chromosorb P (Synsorb Pk) as a specific absorbing agent of Shiga P component is the Shiga toxin 2-neutralizing factor in human blood.
toxin 1 and 2. Microbiol Immunol 43:331–337. J Biol Chem 276:41576–41579.
41. Armstrong GD, Rowe PC, Goodyer P, Orrbine E, Klassen TP, 58. Kimura T, Tani S, Motoki M, Matsumoto Y. 2003. Role of Shiga toxin
Wells G, MacKenzie A, Lior H, Blanchard C, Auclair F, et al. 1995. A 2 (Stx2)-binding protein, human serum amyloid P component (HuSAP), in
phase I study of chemically synthesized verotoxin (Shiga-like toxin) Pk- Shiga toxin-producing Escherichia coli infections: assumption from in vitro
trisaccharide receptors attached to chromosorb for preventing hemolytic- and in vivo study using HuSAP and anti-Stx2 humanized monoclonal an-
uremic syndrome. J Infect Dis 171:1042–1045. tibody TMA-15. Biochem Biophys Res Commun 305:1057–1060.
42. Kitov PI, Sadowska JM, Mulvey G, Armstrong GD, Ling H, Pannu 59. Armstrong GD, Mulvey GL, Marcato P, Griener TP, Kahan MC,
NS, Read RJ, Bundle DR. 2000. Shiga-like toxins are neutralized by tai- Tennent GA, Sabin CA, Chart H, Pepys MB. 2006. Human serum amy-
lored multivalent carbohydrate ligands. Nature 403:66–672. loid P component protects against Escherichia coli O157:H7 Shiga toxin 2
43. Mulve GL, Marcato P, Kitov PI, Sadowska J, Bundle DR, Armstrong in vivo: therapeutic implications for hemolytic-uremic syndrome. J Infect
GD. 2003. Assessment in mice of the therapeutic potential of tailored, Dis 193:1120–1124.
multivalent Shiga toxin carbohydrate ligands. J Infect Dis 187:640–649. 60. Griener TP, Strecker JG, Humphries RM, Mulvey GL, Fuentealba C,
44. Nishikawa K, Matsuoka K, Kita E, Okabe N, Mizuguchi M, Hino K, Hancock RE, Armstrong GD. 2011. Lipopolysaccharide renders trans-
Miyazawa S, Yamasaki C, Aoki J, Takashima S, Yamakawa Y, Nishijima genic mice expressing human serum amyloid P component sensitive to
M, Terunuma D, Kuzuhara H, Natori Y. 2002. A therapeutic agent Shiga toxin 2. PLoS One 6:e21457.
with oriented carbohydrates for treatment of infections by Shiga toxin- 61. Marcato P, Vander Helm K, Mulvey GL, Armstrong GD. 2003. Serum
producing Escherichia coli O157:H7. Proc Natl Acad Sci USA 99:7669– amyloid P component binding to Shiga toxin 2 requires both a subunit and
7674. B pentamer. Infect Immun 71:607–6078.

12 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
New Therapeutic Developments against Shiga Toxin-Producing Escherichia coli

62. Donohue-Rolfe A, Kondova I, Mukherjee J, Chios K, Hutto D, protective human antibodies against Shiga toxin 1. Infect Immun 70:
Tzipori S. 1999. Antibody-based protection of gnotobiotic piglets infected 5896–5899.
with Escherichia coli O157:H7 against systemic complications associated 77. Mukherjee J, Chios K, Fishwild D, Hudson D, O’Donnell S, Rich SM,
with Shiga toxin 2. Infect Immun 67:3645–3648. Donohue-Rolfe A, Tzipori S. 2002. Human Stx2-specific monoclonal
63. Perera LP, Marques LR, O’Brien AD. 1988. Isolation and character- antibodies prevent systemic complications of Escherichia coli O157:H7
ization of monoclonal antibodies to Shiga-like toxin II of entero- infection. Infect Immun 70:612–619.
hemorrhagic Escherichia coli and use of the monoclonal antibodies in a 78. Sheoran AS, Chapman S, Singh P, Donohue-Rolfe A, Tzipori S. 2003.
colony enzyme-linked immunosorbent assay. J Clin Microbiol 26:2127– Stx2-specific human monoclonal antibodies protect mice against lethal
2131. infection with Escherichia coli expressing Stx2 variants. Infect Immun
64. Strockbine NA, Marques LR, Holmes RK, O’Brien AD. 1985. 71:3125–3130.
Characterization of monoclonal antibodies against Shiga-like toxin from 79. Krautz-Peterson G, Chapman-Bonofiglio S, Boisvert K, Feng H,
Escherichia coli. Infect Immun 50:69–700. Herman IM, Tzipori S, Sheoran AS. 2008. Intracellular neutralization of
65. Smith MJ, Melton-Celsa AR, Sinclair JF, Carvalho HM, Robinson Shiga toxin 2 by an a subunit-specific human monoclonal antibody. Infect
CM, O’Brien AD. 2009. Monoclonal antibody 11E10, which neutralizes Immun 76:1931–1939.
shiga toxin type 2 (Stx2), recognizes three regions on the Stx2 A subunit, 80. Jeong KI, Chapman-Bonofiglio S, Singh P, Lee J, Tzipori S, Sheoran
blocks the enzymatic action of the toxin in vitro, and alters the overall AS. 2010. In vitro and in vivo protective efficacies of antibodies that
cellular distribution of the toxin. Infect Immun 77:2730–2740. neutralize the RNA N-glycosidase activity of Shiga toxin 2. BMC
66. Smith MJ, Carvalho HM, Melton-Celsa AR, O’Brien AD. 2006. The Immunol 11:16.
13C4 monoclonal antibody that neutralizes Shiga toxin type 1 (Stx1) 81. Akiyoshi DE, Sheoran AS, Rich CM, Richard L, Chapman-Bonofiglio
recognizes three regions on the Stx1 B subunit and prevents Stx1 from S, Tzipori S. 2010. Evaluation of Fab and F(ab′)2 fragments and isotype
binding to its eukaryotic receptor globotriaosylceramide. Infect Immun variants of a recombinant human monoclonal antibody against Shiga
74:6992–6998. toxin 2. Infect Immun 78:1376-1382.
67. Sauter KA, Melton-Celsa AR, Larkin K, Troxell ML, O’Brien AD, 82. Spooner RA, Watson P, Smith DC, Boal F, Amessou M, Johannes L,
Magun BE. 2008. Mouse model of hemolytic-uremic syndrome caused by Clarkson GJ, Lord JM, Stephens DJ, Roberts LM. 2008. The secretion
endotoxin-free Shiga toxin 2 (Stx2) and protection from lethal outcome by inhibitor Exo2 perturbs trafficking of Shiga toxin between endosomes and
anti-Stx2 antibody. Infect Immun 76:4469–4478. the trans-Golgi network. Biochem J 414:471–484.
68. Edwards AC, Melton-Celsa AR, Arbuthnott K, Stinson JR, Schmitt 83. Guetzoyan LJ, Spooner RA, Boal F, Stephens DJ, Lord JM, Roberts
CK, Wong HC, O’Brien AD. 1998. Vero cell neutralization and mouse LM, Clarkson GJ. 2010. Fine tuning Exo2, a small molecule inhibitor of
protective efficacy of humanized monoclonal antibodies against Esche- secretion and retrograde trafficking pathways in mammalian cells. Mol
richia coli toxins Stx1 and Stx2, p 388–392. In Kaper JB, O’Brien AD (ed), Biosyst 6:2030–2038.
Escherichia coli O157:H7 and Other Shiga Toxin-Producing E. coli 84. Stechmann B, Bai SK, Gobbo E, Lopez R, Merer G, Pinchard S,
Strains. ASM Press, Washington, DC. Panigai L, Tenza D, Raposo G, Beaumelle B, Sauvaire D, Gillet D,
69. Dowling TC, Chavaillaz PA, Young DG, Melton-Celsa A, O’Brien A, Johannes L, Barbier J. 2010. Inhibition of retrograde transport protects
Thuning-Roberson C, Edelman R, Tacket CO. 2005. Phase 1 safety and mice from lethal ricin challenge. Cell 141:231–242.
pharmacokinetic study of chimeric murine-human monoclonal antibody c 85. Park JG, Kahn JN, Tumer NE, Pang YP. 2012. Chemical structure of
alpha Stx2 administered intravenously to healthy adult volunteers. Anti- Retro-2, a compound that protects cells against ribosome-inactivating
microb Agents Chemother 49:1808–1812. proteins. Sci Rep 2:631.
70. Bitzan M, Poole R, Mehran M, Sicard E, Brockus C, Thuning- 86. Noel R, Gupta N, Pons V, Goudet A, Garcia-Castillo MD, Michau A,
Roberson C, Riviere M. 2009. Safety and pharmacokinetics of chimeric Martinez J, Buisson DA, Johannes L, Gillet D, Barbier J, Cintrat JC. 2013.
anti-Shiga toxin 1 and anti-Shiga toxin 2 monoclonal antibodies in healthy N-methyl dihydroquinazolinones derivatives of Retro-2 with enhanced
volunteers. Antimicrob Agents Chemother 53:3081–3087. efficacy against Shiga toxin. J Med Chem 56:3404–3413.
71. Nakao H, Kataoka C, Kiyokawa N, Fujimoto J, Yamasaki S, Takeda 87. Dyve Lingelem AB, Bergan J, Sandvig K. 2012. Inhibitors of
T. 2002. Monoclonal antibody to Shiga toxin 1, which blocks receptor intravesicular acidification protect against Shiga toxin in a pH-indepen-
binding and neutralizes cytotoxicity. Microbiol Immunol 46:777–780. dent manner. Traffic 13:443–454.
72. Nakao H, Kiyokawa N, Fujimoto J, Yamasaki S, Takeda T. 1999. 88. Saenz JB, Doggett TA, Haslam DB. 2007. Identification and charac-
Monoclonal antibody to Shiga toxin 2 which blocks receptor binding and terization of small molecules that inhibit intracellular toxin transport.
neutralizes cytotoxicity. Infect Immun 67:5717–5722. Infect Immun 75:4552–4561.
73. Kimura T, Co MS, Vasquez M, Wei S, Xu H, Tani S, Sakai Y, 89. Wahome PG, Bai Y, Neal LM, Robertus JD, Mantis NJ. 2010.
Kawamura T, Matsumoto Y, Nakao H, Takeda T. 2002. Development of Identification of small-molecule inhibitors of ricin and Shiga toxin using a
humanized monoclonal antibody TMA-15 which neutralizes Shiga toxin cell-based high-throughput screen. Toxicon 56:313–323.
2. Hybrid Hybridomics 21:161–168. 90. Aletrari MO, McKibbin C, Williams H, Pawar V, Pietroni P, Lord
74. Yamagami S, Motoki M, Kimura T, Izumi H, Takeda T, Katsuura Y, JM, Flitsch SL, Whitehead R, Swanton E, High S, Spooner RA. 2011.
Matsumoto Y. 2001. Efficacy of postinfection treatment with anti-Shiga Eeyarestatin 1 interferes with both retrograde and anterograde intracel-
toxin (Stx) 2 humanized monoclonal antibody TMA-15 in mice lethally lular trafficking pathways. PLoS One 6:e22713.
challenged with Stx-producing Escherichia coli. J Infect Dis 184:738–742. 91. Sekino T, Kiyokawa N, Taguchi T, Ohmi K, Nakajima H, Suzuki T,
75. Lopez EL, Contrini MM, Glatstein E, Gonzalez Ayala S, Santoro R, Furukawa S, Nakao H, Takeda T, Fujimoto J. 2002. Inhibition of Shiga
Allende D, Ezcurra G, Teplitz E, Koyama T, Matsumoto Y, Sato H, Sakai toxin cytotoxicity in human renal cortical epithelial cells by nitrobenzyl-
K, Hoshide S, Komoriya K, Morita T, Harning R, Brookman S. 2010. thioinosine. J Infect Dis 185:785–796.
Safety and pharmacokinetics of urtoxazumab, a humanized monoclonal 92. Mukhopadhyay S, Linstedt AD. 2012. Manganese blocks intracellular
antibody, against Shiga-like toxin 2 in healthy adults and in pediatric trafficking of Shiga toxin and protects against Shiga toxicosis. Science
patients infected with Shiga-like toxin-producing Escherichia coli. Anti- 335:332–335.
microb Agents Chemother 54:239–243. 93. Mukhopadhyay S, Redler B, Linstedt AD. 2013. Shiga toxin-binding
76. Mukherjee J, Chios K, Fishwild D, Hudson D, O’Donnell S, Rich SM, site for host cell receptor GPP130 reveals unexpected divergence in toxin-
Donohue-Rolfe A, Tzipori S. 2002. Production and characterization of trafficking mechanisms. Mol Biol Cell 24:2311–2318.

ASMscience.org/MicrobiolSpectrum 13
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16
Melton-Celsa and O’Brien

94. Becker GL, Lu Y, Hardes K, Strehlow B, Levesque C, Lindberg I, Escherichia coli O104:H4-associated haemolytic uraemic syndrome: a
Sandvig K, Bakowsky U, Day R, Garten W, Steinmetzer T. 2012. Highly prospective trial. Lancet 378:1166–1173.
potent inhibitors of proprotein convertase furin as potential drugs for 102. Ullrich S, Bremer P, Neumann-Grutzeck C, Otto H, Ruther C, von
treatment of infectious diseases. J Biol Chem 287:21992–22003. Seydewitz CU, Meyer GP, Ahmadi-Simab K, Rother J, Hogan B, Schwenk
95. Garred O, van Deurs B, Sandvig K. 1995. Furin-induced cleavage and W, Fischbach R, Caselitz J, Puttfarcken J, Huggett S, Tiedeken P, Pober J,
activation of Shiga toxin. J Biol Chem 270:10817–10821. Kirkiles-Smith NC, Hagenmuller F. 2013. Symptoms and clinical course
96. Garred O, Dubinina E, Holm PK, Olsnes S, van Deurs B, Kozlov JV, of EHEC O104 infection in hospitalized patients: a prospective single
Sandvig K. 1995. Role of processing and intracellular transport for opti- center study. PLoS One 8:e55278.
mal toxicity of Shiga toxin and toxin mutants. Exp Cell Res 218:39–49. 103. Loos S, Ahlenstiel T, Kranz B, Staude H, Pape L, Hartel C, Vester U,
97. Burgess BJ, Roberts LM. 1993. Proteolytic cleavage at arginine Buchtala L, Benz K, Hoppe B, Beringer O, Krause M, Muller D, Pohl M,
residues within the hydrophilic disulphide loop of the Escherichia coli Lemke J, Hillebrand G, Kreuzer M, Konig J, Wigger M, Konrad M,
Shiga-like toxin I A subunit is not essential for cytotoxicity. Mol Microbiol Haffner D, Oh J, Kemper MJ. 2012. An outbreak of Shiga toxin-pro-
10:171–179. ducing Escherichia coli O104:H4 hemolytic uremic syndrome in
98. Stone SM, Thorpe CM, Ahluwalia A, Rogers AB, Obata F, Vozenilek Germany: presentation and short-term outcome in children. Clin Infect
A, Kolling GLKan AV, Magun BE, Jandhyala DM. 2012. Shiga toxin 2- Dis 55:753–759.
induced intestinal pathology in infant rabbits is A-subunit dependent and 104. Hauswaldt S, Nitschke M, Sayk F, Solbach W, Knobloch JK. 2013.
responsive to the tyrosine kinase and potential ZAK inhibitor imatinib. Lessons learned from uutbreaks of Shiga toxin producing Escherichia coli.
Front Cell Infect Microbiol 2:135. Curr Infect Dis Rep 15:9.
99. Burlaka I, Liu XL, Rebetz J, Arvidsson I, Yang L, Brismar H, Karpman 105. Kielstein JT, Beutel G, Fleig S, Steinhoff J, Meyer TN, Hafer C,
D, Aperia A. 2013. Ouabain protects against Shiga toxin-triggered apo- Kuhlmann U, Bramstedt J, Panzer U, Vischedyk M, Busch V, Ries W,
ptosis by reversing the imbalance between Bax and Bcl-xL. J Am Soc Mitzner S, Mees S, Stracke S, Nurnberger J, Gerke P, Wiesner M, Sucke B,
Nephrol 24:1413–1423. Abu-Tair M, Kribben A, Klause N, Schindler R, Merkel F, Schnatter S,
100. Lapeyraque AL, Malina M, Fremeaux-Bacchi V, Boppel T, Dorresteijn EM, Samuelsson O, Brunkhorst R. 2012. Best supportive care
Kirschfink M, Oualha M, Proulx F, Clermont MJ, Le Deist F, Niaudet P, and therapeutic plasma exchange with or without eculizumab in Shiga-
Schaefer F. 2011. Eculizumab in severe Shiga-toxin-associated HUS. N toxin-producing E. coli O104:H4 induced haemolytic-uraemic syndrome:
Engl J Med 364:2561–2563. an analysis of the German STEC-HUS registry. Nephrol Dial Transplant
101. Greinache A, Friesecke S, Abel P, Dressel A, Stracke S, Fiene M, Ernst 27:3807–3815.
F, Selleng K, Weissenborn K, Schmidt BM, Schiffer M, Felix SB, Lerch 106. Trachtman H, Austin C, Lewinski M, Stahl RA. 2012. Renal and
MM, Kielstein JT, Mayerle J. 2011. Treatment of severe neurological neurological involvement in typical Shiga toxin-associated HUS. Nat Rev
deficits with IgG depletion through immunoadsorption in patients with Nephrol 8:658–669.

14 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 202.86.180.130
On: Thu, 19 Sep 2019 02:14:16

You might also like