You are on page 1of 8

Biochemical Pharmacology xxx (2017) xxx–xxx

Contents lists available at ScienceDirect

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

Commentary

Repurposing bacterial toxins for intracellular delivery of therapeutic


proteins
Greg L. Beilhartz a, Seiji N. Sugiman-Marangos a, Roman A. Melnyk a,b,⇑
a
Molecular Medicine Program, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada
b
Department of Biochemistry, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada

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

Article history: Despite enormous efforts, achieving efficacious levels of proteins inside mammalian cells remains one of
Received 9 March 2017 the greatest challenges in biologics-based drug discovery and development. The inability of proteins to
Accepted 7 April 2017 readily cross biological membranes precludes access to the wealth of intracellular targets and applica-
Available online xxxx
tions that lie within mammalian cells. Existing methods of delivery commonly suffer from an inability
to target specific cells and tissues, poor endosomal escape, and limited in vivo efficacy. The aim of the pre-
Keywords: sent commentary is to highlight the potential of certain classes of bacterial toxins, which naturally deliver
Bacterial toxins
a large protein into the cytosolic compartment of target cells after binding a host cell-surface receptor
Intracellular protein delivery
Biologics
with high affinity, as robust protein delivery platforms. We review the progress made in recent years
Protein therapeutics toward demonstrating the utility of these systems at delivering a wide variety of protein cargo, with spe-
Diphtheria toxin cial attention paid to three distinct toxin-based platforms. We contend that with recent advances in pro-
tein deimmunization strategies, bacterial toxins are poised to introduce biologics into the inner sanctum
of cells and treat a wealth of heretofore untreatable diseases with a new generation of therapeutics.
Ó 2017 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Existing delivery platforms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Toxin cell entry mechanisms: three distinct strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. Delivery of proteinaceous cargo by AB toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. Practical considerations for delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6. Re-directing toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.1. Cellular tropism. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.2. Tissue tropism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7. Future directions/challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.1. Receptor targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.2. Choice of cargo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.3. Immunogenicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.4. Platform optimization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
8. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

⇑ Corresponding author at: Research Institute, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada.
E-mail address: roman.melnyk@sickkids.ca (R.A. Melnyk).

http://dx.doi.org/10.1016/j.bcp.2017.04.009
0006-2952/Ó 2017 Elsevier Inc. All rights reserved.

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009
2 G.L. Beilhartz et al. / Biochemical Pharmacology xxx (2017) xxx–xxx

1. Introduction deliver their encapsidated cargo to the cytosol by direct membrane


fusion, whereas other non-enveloped viruses have complex endo-
In the past 30 years, since the approval of recombinant human somal escape mechanisms that are not completely understood
insulin, protein-based therapeutics have grown in prominence [17,18]. While VLPs are perfectly suited for vaccine development,
and are now among the fastest-growing class of drugs [1]. In their strong induction of both humoral and cellular immune
2015, 13 new biologics were approved by the FDA, representing responses likely restricts their in vivo use [15].
29% of all approved drugs [2]. In the same year, seven of the top Liposomes and nanoparticles are broadly similar carrier sys-
10 best selling drugs were biologics, led by AbbVie’s Humira, which tems that have been explored for delivery. Liposomes and
brought in $14 billion (USD) in sales [3]. Therapeutic proteins and nanoparticles typically do not have built-in mechanisms for endo-
peptides comprise 85% of the approximately 300 biologics that somal escape, although attempts have been made to decorate the
are currently approved for clinical use [4]. Despite this dramatic surface of nanoparticles or liposomes with CPPs or fusogenic pep-
expansion in large-molecule therapeutics, the full potential of pro- tides/lipids to increase cytosolic delivery [19,20]. While both
teins as drugs is constrained by their inability to cross biological classes can be targeted to specific cells by displaying antibodies
membranes and enter cells. As such, approximately 60% of all or other targeting moieties on their surface, achieving high surface
human proteins – and the entire human genome – remain inacces- densities can be challenging, and even when successful can result
sible to protein-based drugs [5]. Intracellular targets with a clear in faster plasma clearance [21,22]. In vivo toxicity of nanoparticles
therapeutic rationale, including protein-protein interactions, large has been described depending on the nanomaterials involved; lipo-
protein complexes, and important targets that are notoriously dif- somes generally do not show overt toxicity in animal models but
ficult to target with small molecules such as p53 and RAS [6,7], are can be genotoxic even at low doses, as well as being immunogenic
best suited to biologics-based therapies. Moreover, enzymes with to varying degrees based on the specific lipids used [23,24].
tremendous therapeutic potential including genome editing Each of the above protein delivery systems possess one or more
machinery such as CRISPR/Cas9 or TALENs must be delivered as of the ideal attributes, however, no single system has the full com-
nucleic acids or electroporated into cells, complicating their use plement of the features that would be desired in an idealized deliv-
as therapeutics. ery platform. Bacterial toxins, which have evolved the ability to
To solve this issue, several classes of protein carriers have been bind host cell receptors, and subsequently escape from endosomes
explored to circumvent the barrier posed by the plasma mem- to the cytosol of mammalian cells, are replete with features that
brane, however, to date, no single platform has been identified that make them attractive candidates for delivery vectors. Anthrax
is simultaneously efficient, safe, versatile and specific. We contend toxin (B. anthracis), exotoxin A (P. aeruginosa) and diphtheria toxin
that bacterial toxins, which bind specific cells and can efficiently (C. diphtheriae) are three different bacterial toxins that use three
transport a broad spectrum of proteins to the cytosol have great distinct strategies to deliver their catalytic cargo into cells. The
promise as platforms for delivering protein cargo into specific cells modular structure of these toxins, with three exchangeable func-
in vitro and in vivo. Notably, the one class of biologics that enter tional domains, makes them particularly amenable to protein engi-
cells as part of their mechanism is immunotoxins, which use the neering. The major perceived limitation of using toxins in humans
cell-penetrating machinery of bacterial toxins. The field of protein is the immune response, however recent work discussed herein
delivery has greatly matured in the past several years, and novel suggests that this is manageable and no longer a hurdle to
protein engineering techniques combined with improved methods development.
for definitively detecting cargo in the cytosol have set the stage for
bacterial toxins to emerge as a major platform to deliver the next
generation of biologics. 3. Toxin cell entry mechanisms: three distinct strategies

Bacterial exotoxins are secreted proteins that are designed to


2. Existing delivery platforms specifically bind, enter, and damage host tissue. The so-called
‘‘AB class” of toxins are of interest to the field of protein delivery,
Cell-penetrating peptides (CPPs) have been used to deliver pro- as they are autonomous delivery vectors; that is, AB toxins possess
teins of various sizes, as well as diverse nucleic acids and even lipo- all the components necessary to gain access to the cytosol inde-
somes and nanoparticles [8,9]. As a group, CPPs generally show pendent of any other bacterial machinery. Members of this class
varying efficacy based on the CPP used, the cell type to be targeted, all minimally consist of an enzymatic A moiety and a B moiety that
and even buffer conditions [10]. The mechanism by which CPPs both binds a cell-surface receptor and mediates entry into cells.
enter cells has not been fully elucidated, however there is evidence The A and B fragments are typically linked by a furin-cleavable lin-
that CPPs can either directly cross the plasma membrane, or gain ker and a disulfide bond [25]. AB toxins bind to a cell surface recep-
entry via endocytosis and subsequent escape from endosomes tor, which triggers endocytosis, transporting the toxin into the
[11]. One or both of these mechanisms may be used depending early endosome for sorting. From here, some toxins are trafficked
on concentration, temperature, and the choice of counter ion to the endoplasmic reticulum (ER) where they undergo retrograde
[11]. Engineered supercharged proteins such as +36 GFP are highly translocation, while others escape directly from the endosome
positively charged and appear to transport protein and nucleic acid (Fig. 1).
cargo into cells by a mechanism that is similar to CPPs [12]. While Among the toxins that can escape directly from endosomes, two
generally effective in vitro, CPPs suffer from a lack of cell/tissue distinct classes of AB toxins – exemplified by anthrax toxin and
specificity, which contributes to variable efficacy in vivo [13,14]. diphtheria toxin – are observed. In the case of anthrax toxin-like
Virus-like particles (VLPs) are a loosely-defined group of self- platforms, the A and B fragments are independent entities that
assembling structures based on viral proteins. VLPs of diverse associate non-covalently upon oligomerization of the B-
structures can be assembled from various mammalian, plant, fragments, whereas for diphtheria toxin-like platforms, the A and
microbial and insect viruses, and are replication deficient, as they B fragments are encoded on the same polypeptide. The transloca-
lack any genetic material [15]. VLPs are able to encapsulate protein tion pores created by these two types of toxins are also distinct.
cargo fused with viral proteins, or express them on their surface as Whereas the anthrax toxin pore spans the endosomal membrane
is often done in the case of VLP-based vaccines [16]. It has been as a rigid b-barrel, diphtheria toxin creates flexible a-helical pores
demonstrated that certain enveloped VLPs (HSV-1, HIV-1, etc.) across the membrane. Importantly, the properties of these two

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009
G.L. Beilhartz et al. / Biochemical Pharmacology xxx (2017) xxx–xxx 3

types of pores have great influence on the types of cargo that can
be transported by these toxins (vide infra).

4. Delivery of proteinaceous cargo by AB toxins

Anthrax toxin is perhaps the best characterized delivery plat-


form, having been demonstrated to co-deliver over 30 different
molecular cargo fused to the carboxy-terminus of LFN – the
amino-terminal 263-amino acid fragment of the anthrax toxin A-
domain (lethal factor) that recognizes the anthrax toxin pore
[26]. Examples of cargo include peptides, other toxin effector pro-
teins, non-canonical peptides, antibody-like molecules, and
oligonucleotides [27–29]. Not all cargos are delivered equally, as
a comparison of anthrax toxin and Pseudomonas exotoxin A
showed that Pseudomonas exotoxin A could deliver up to three
consecutive designed Ankyrin repeat proteins (DARPins) with
equivalent efficacies, whereas anthrax toxin could only deliver a
single Ankyrin domain (TM = 60 °C) [30]. Due to the rigid nature
of the anthrax toxin pore, and the lack of external factors driving
unfolding and translocation of cargo on the endosomal side, the
two more stably folded cargos (TM > 90 °C) could not be translo-
Fig. 1. Cellular entry mechanisms of bacterial toxins. Diphtheria toxin, anthrax
cated by anthrax. This inability to deliver stably folded cargo rep- toxin and Pseudomonas exotoxin A bind to their specific receptors through their
resents the principal weakness of the anthrax platform, and receptor-binding domains (R), resulting in endocytosis and furin cleavage [83,84].
precludes delivery of tight folders, proteins containing internal In contrast, anthrax toxin is furin-processed on the cell surface, which is required
disulfide bonds, proteins stabilized by bound ligands, and cyclic for oligomerization of the B fragment and subsequent endocytosis [85]. The anthrax
toxin A-domain non-covalently associates with the B fragment heptamer, whereas
peptides [26,31,32]. Pseudomonas exotoxin A does not appear to for diphtheria toxin and Pseudomonas exotoxin A, the A and B fragments remain
have the same limitations as anthrax toxin when it comes to stably linked via a disulfide bridge after furin cleavage. Anthrax and diphtheria toxins are
folded cargo, presumably due to the presence of ER chaperones trafficked to late endosomes, and undergo a conformational change and membrane
that facilitate unfolding and disulfide bond reduction. Likewise, insertion of the transmembrane (T) domain in response to the low endosomal pH
[86–88]. Both toxins then translocate their catalytic A domains through the pore
diphtheria toxin has been demonstrated to efficiently translocate
and into the cytosol [89,90]. The reducing environment of the cytosol allows for the
a wide variety of protein cargos including the highly thermo- release of the disulfide-linked diphtheria toxin A domain [34]. Pseudomonas
and pH-stable fluorophore mCherry [33]. exotoxin A avoids the endolysosomal pathway by being trafficked to the ER via the
trans-Golgi network [91]. In this case, the disulfide linkage is broken by protein
disulfide isomerase (PDI) [92], allowing the A domain to be recognized and
5. Practical considerations for delivery unfolded by ER chaperones [93]. It is thought that the A domain is retrotranslocated
through the sec61 translocon into the cytosol [94].
The quantity of delivered cargo is one of the most important
variables in defining the efficiency of a delivery platform. Each
stage of the delivery process influences the amount of payload that line between solubilizing plasma and endosomal membranes
reaches the cytosol: receptor binding, internalization, furin cleav- [35]. The method described by Verdumen et al. [30] shows promise
age, trafficking, translocation and disulfide reduction and release. due to its versatility, as it can be easily applied to any protein cargo
The biological effect of cargo is fundamentally tied to the amount through the addition of a 15-residue avi-tag. Techniques limited by
reaching the cytosol. Overexpression of prospective cargo in cell the semi-quantitative nature of western blotting can often be
culture is the only positive control available to demonstrate biolog- improved by mass spectrometry if quantification is critical.
ical response, making the interpretation of results ambiguous. Is The need for a reliable quantification method was illustrated
the protein making it to the cytosol in sufficient quantities? How previously with anthrax toxin [28]. Despite the efficient delivery
much protein is needed to elicit the same response as constitutive of several antibody-like cargos as demonstrated by protein synthe-
overexpression? Unfortunately, one of the major impediments in sis inhibition and western blot, the authors noted that the intracel-
the field of protein delivery is lack of standardized methods for lular effects were quite modest, even at the highest concentrations
quantifying cytosolic cargo. Microscopy of fluorescent cargo is a tested. For example, HA4-7c12 is a tandem monobody that binds
strategy that can be used to visualize entry; however, great care with nanomolar affinity to the SH2 domain of the oncoprotein
needs to be taken to account for signal coming from the vast num- Bcr-Abl [36]. While HA4-7c12 was shown to strongly inhibit Bcr-
ber of endosomes that can confound the interpretation of the Abl activity and induce apoptosis when transiently expressed in
amount of cargo reaching the cytosol. While delivery of therapeu- K562 cells, treatment of cells with high doses of LFn-HA4-7c12
tic cargo is normally done in the context of a non-toxic variant of only elicited a moderate effect.
the toxin delivery platform (i.e., the catalytic A-domain is mutated It seems unlikely that increasing binding affinity further would
to be inactive), a commonly used technique, particularly among drastically improve the effect, which raises the questions of how
those that work with bacterial toxins, is the inhibition of protein much monobody is being delivered, and how much is required?
synthesis by the wildtype diphtheria toxin A-domain fused to the While this result demonstrates the proof-of-concept, it raises
cargo protein under investigation. Toxicity as a proximal readout another important aspect of protein delivery in general. Namely,
is a sensitive indicator of cytosolic delivery of cargo due to the is it feasible to stoichiometrically target highly abundant intracel-
extremely potent nature of DT-A, however, it is less useful for lular proteins? The answer to this question remains unclear, but it
quantifying the number of cargo molecules delivered [33]. Meth- would seem a high bar to set for any exogenous protein therapeu-
ods based on cell fractionation using detergents and western blot- tic. Attempting to overtake host gene expression by delivering sto-
ting have been used by many [28,34], however, these methods are ichiometric amounts of exogenous protein would likely require
semi-quantitative and can be technically difficult, given the fine both a very high serum drug concentration, and an extremely

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009
4 G.L. Beilhartz et al. / Biochemical Pharmacology xxx (2017) xxx–xxx

efficient delivery mechanism. Fortunately, enzymes as biologic noted that not all of these moieties will be effective and some opti-
drugs can massively leverage their effects beyond simple target mization may be required (vida infra).
binding, more than compensating for their lower intracellular
doses. Though the development of protein therapeutics has been 6.2. Tissue tropism
so far focused on antibodies and stoichiometric binding of a target
protein, the untapped potential of biologics lies in the delivery of In addition to determining cell type specificity, the targeting
powerful therapeutic enzymes. moiety also determines tissue tropism. This is particularly relevant
AB toxins all evolved highly efficient mechanisms to translocate for biologics developed to treat tissue-specific diseases such as
their cytotoxic cargo, however the cargo enzymes themselves are osteosarcomas or neurological diseases such as Alzheimer’s,
also extremely potent. eEF-2, the target of both diphtheria toxin Parkinson’s or lysosomal storage diseases (LSDs). LSDs result from
and Pseudomonas exotoxin A, is estimated to be present on the a mutation(s) that typically result in a non-functional protein
order of 107 molecules/cell [37], yet it is often claimed that a single involved in the degradation of lysosome contents. These diseases
molecule of diphtheria toxin A-domain is sufficient to kill a cell are commonly treated by enzyme replacement therapy (ERT),
[38]. While the number of molecules that reach the cytosol during where a recombinant version of the missing protein is injected into
an actual intoxication event is undoubtedly far greater, it does patients. Recombinant enzymes enter cells via the mannose-6-
underscore the power of signal amplification inherent to enzy- phosphate receptor (M6PR) and are subsequently trafficked to
matic cargo. For example, assuming the volume of a mammalian lysosomes [52]. Efficacy is limited by M6PR as it is either absent
cell to be 1–10 pL, the intracellular concentration represented or poorly expressed in several important tissue types including
by a single molecule per cell is 10–100 fM [39]. Indeed, the best bone, muscle and the brain vasculature [53]. The absence of
examples of non-native cargo that have been delivered by bacterial M6PR on the blood-brain-barrier (BBB) is particularly problematic
toxins with demonstrable biological effects are enzymes, such as as 75% of LSDs manifest with neurological symptoms [54]. The
bacterial effector domains [28,40], b-lactamase [41], DHFR [32], BBB presents a formidable barrier to delivery of biologics into the
and a-amylase [33]. Regardless of the delivery system used, effort brain, as anything larger than small molecules are unable to diffuse
spent optimizing the properties and potency of enzyme cargo may across.
be equally or even more beneficial than increasing the number of Some receptors have the potential to be exploited for brain pen-
delivered enzyme molecules. etration including receptors for low-density lipoprotein, transfer-
rin and insulin. The use of antibodies or antibody-like molecules
targeting these proteins for receptor-mediated transcytosis is an
6. Re-directing toxins area of active research [55–57]. Remarkably, diphtheria has been
shown to cross the BBB naturally [58–60], via HBEGF-mediated
6.1. Cellular tropism transcytosis of brain endothelial cells. Current methods of deliver-
ing biologics into the brain involve either intrathecal or intracere-
One of the principal advantages of toxin-based platforms is the broventricular injection directly into the brain [61,62]. The ability
specificity gained by targeting a unique cell-surface receptor. of systemically administered protein drugs to cross the BBB and
Moreover, due to their modular structure, both Pseudomonas exo- access the neuronal cytosol would be a marked improvement on
toxin A [30,42] and diphtheria toxin [43,44] can be re-targeted current techniques.
by replacing their receptor binding domains with a ligand (natural
or engineered) to a different receptor. Recent efforts have been
made to similarly re-target the B-moiety of anthrax toxin [30,45], 7. Future directions/challenges
although this is complicated by the fact that removal of the
receptor-binding domain has consequences for proper PA folding. Bacterial toxins are among the best hopes for delivery of thera-
The most notable examples of successful retargeting of toxins peutic proteins to the cytosol of human cells and the treatment of
come from the immunotoxin field. Many have recognized and many diseases and conditions. Their efficacy and versatility allow
exploited the ability of diphtheria toxin to tolerate the replacement for the conjugation of an immense variety of possible cargo. Direct
of its C-terminal receptor-binding domain with cancer-targeted comparison of the delivery efficiencies of anthrax toxin and a CPP
antibodies and ligands. The FDA-approved Ontak (Denileukin difti- revealed that the anthrax-based system was three orders of magni-
tox) is a re-targeted diphtheria toxin variant with the cytokine IL-2 tude more efficient when delivering various antibody-like scaffolds
fused to the C-terminus of the DT1-389 fragment. This re-targets the [28]. Diphtheria toxin has similarly been shown to be superior to
toxic A-fragment to cells expressing the high-affinity IL-2 receptor CPPs at delivering enzymes into cells [33]. There are points to keep
(CD25, CD122 and CD132 trimer), which is often overexpressed on in mind, however, when designing an intracellular therapeutic
leukemia cells. Similarly, DT1-389 fused to epidermal growth factor with bacterial toxins.
(EGF) was developed for the targeting of EGFR-overexpressing
glioblastoma cells, and showed synergistic effects with IL13 fused 7.1. Receptor targeting
to Pseudomonas exotoxin (PE-IL13) [46,47]. Diphtheria toxin fused
with tandem single-chain antibodies against CD3 (Resimmune) has An important consideration for changing the receptor speci-
shown efficacy in patients with cutaneous T-cell lymphoma [48]. ficity for toxins is problems that may arise from receptor ago-
In a further improvement on the same design, a bispecific nism/antagonism. Furthermore, it should not be assumed that
immunotoxin was developed by the fusion of tandem single- the novel receptor will be internalized in response to binding, or
chain antibodies targeting different cell surface receptors, in this that trafficking of the new receptor would be the same as the
case CD19 and CD22 [49]. This construct has shown activity in pre- native one [63]. For example, HER2 (a common target in cancer
clinical models and is entering clinical trials. Pseudomonas exotoxin therapy due to its overexpression in many cancer types) has a
A-based immunotoxins are also in the clinic, including moxetu- low rate of internalization in addition to a rapid recycling to the
momab pasudotox which re-targets Pseudomonas exotoxin A with plasma membrane from early endosomes [64]. This might suggest
an anti-CD22 antibody fragment and LMB-2, which targets CD25- that it would make a poor target for receptor-mediated endocyto-
expressing cells [50,51]. Although diphtheria toxin can accept a sis of a toxin-based therapeutic, yet Affitoxin (an Pseudomonas exo-
wide variety of targeting domains on its C-terminus, it must be toxin A-based immunotoxin) appears to be reasonably potent on

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009
G.L. Beilhartz et al. / Biochemical Pharmacology xxx (2017) xxx–xxx 5

HER2 overexpressing cells, suggesting that rate of internalization is tors. Many applications for protein delivery require multiple, even
not necessarily a key attribute of a ‘‘good” ligand-receptor interac- lifetime injections. Clearly, a host immune response is to be
tion [42]. So, how to best interrogate a novel receptor? To answer avoided. Toxin-based protein carriers are not alone in this, as the
this question, a synthetic antibody library was screened to identify FDA has suggested that drug developers proactively address the
92 HER2-specific single-chain Fvs (scFv) [65]. These were non- issue of immunogenicity and the rise of anti-drug antibodies
covalently associated with a truncated form of Pseudomonas exo- (ADAs) when it comes to biologics [69]. Fortunately, recent
toxin A to form either monovalent (one scFV) or bivalent (tandem advances hold promise that the immunogenicity problem is not
scFvs) immunotoxins. They showed that bivalent arrangements as severe as it once was. In the case of the immunotoxin denileukin
were more potent than monovalent ones for reasons beyond avid- diftitox, although ADA titers increased after treatment, they had no
ity, as the effect varied among the scFvs. Interestingly, they also effect on clinical outcome [70]. The lack of a neutralizing antibody
found that binding affinities and kinetics of the scFvs were not cor- response has been also observed for Pseudomonas exotoxin A-
related with cytotoxicity. Instead, the specific epitope and paratope based immunotoxins, and appears to be a feature of hematological
of the scFvs were deemed to be particularly important, as they malignancies [50]. While this can be beneficial for the treatment of
relate to the pH-dependence of binding and the downstream traf- blood cancers, the repeated use of diphtheria toxin as a protein
ficking of the toxin [65]. This unexpected result could be very use- delivery system for solid-tumor immunotoxins or ERT would
ful when re-targeting toxins for protein delivery. require reducing the immunogenicity of diphtheria toxin itself.
Another way of changing the cell specificity of diphtheria toxin Several examples of successful deimmunization involving the
is by replacing the furin cleavage site with a recognition sequence identification of T and/or B cell epitopes, and subsequent mutage-
for a cell-specific protease. Abi-Habib et al. replaced the furin site nesis to remove them while retaining protein function are now
in a diphtheria toxin-based immunotoxin with a sequence cleav- available [71,72]. Several groups have successfully developed
able by urokinase plasminogen activator (uPA), a technique origi- deimmunized proteins, including diphtheria toxin while retaining
nally used for re-targeting anthrax toxin [66,67]. UPA is a serine catalytic activity [73,74]. Instead of masking the toxin from the
protease that is overexpressed in leukemias and other tumors, immune system, another method is to induce immune tolerance
and the new immunotoxin displayed greater toxicity to cells through induction of regulatory T cells (Tregs) [75]. Recently it
expressing uPA. has been shown that the formation of ADAs can be prevented by
Cell specificity can also be attained by the choice of cytosolic co-administering rapamycin-containing nanoparticles with an
target. The oncogenic bcr-abl fusion protein is a result of a chromo- antigen. This induces tolerogenic dendritic cells, increases Tregs,
somal translocation in chronic myelogenous leukemia, among and reduces B cell activation [76]. This is a promising approach,
others. Anthrax toxin was modified to deliver an LFN-monobody as it promotes immune tolerance of the antigen, rather than
targeting the bcr-abl fusion protein [28]. While the fusion toxin attempting to hide it from the immune system. In a similar vein,
itself is not cell specific, malignant cells will be disproportionally the pre-administration of pentostatin and cyclophosphamide,
sensitive to the monobody cargo. Combining some or all the above which depletes host T and B cells was shown to delay the emer-
techniques may confer exquisite target specificity to these protein gence of neutralizing antibodies against the Pseudomonas exotoxin
delivery vehicles, including the next generation of cancer A-based immunotoxin SS1P, while improving outcomes in a
immunotoxins. mesothelioma trial [77]. While a host immune response to biolog-
ics can reduce their effectiveness, and in severe cases be life-
7.2. Choice of cargo threatening, it need not be an insurmountable hurdle to clinical
success.
Translocation is typically thought to be the rate-limiting step in
toxin delivery. In immunotoxin applications where the cargo is a 7.4. Platform optimization
cytotoxic enzyme, cell specificity outweighs efficiency, as only a
small quantity of cargo is required to kill cells. Other cargo may While bacterial toxins have evolved the ability to target specific
require that more molecules get delivered to elicit the desired cells and escape from endosomes into the cytosol, this ability is
response, as intracellular target proteins vary greatly in abundance uniquely suited to perform its natural function, i.e. to kill cells. This
from many millions of copies, to a dozen or fewer [39]. As such, evolutionary optimization of toxins as dealers of cell death can be
acknowledgement of the limitations of bacterial toxins is required ideal for targeted immunotoxins, however it would appear to be
to determine the most appropriate intracellular targets, cargo, and less suited to other applications. There are several areas where tox-
applications. For example, as mentioned above, anthrax toxin is ins can be improved from a protein delivery standpoint. As dis-
not able to deliver cargo that cannot completely unfold on its cussed previously, replacing the receptor binding moiety, or
own, while Pseudomonas exotoxin A and diphtheria toxin do not developing a bivalent binding domain can decrease off-target
have the same limitation. While Pseudomonas exotoxin A (and pre- effects and improve the therapeutic index. The development of
sumably attached cargo) is unfolded by chaperones and isomerases delivery systems that can deliver multiple copies of an effector in
prior to translocation from the ER, diphtheria toxin appears to be single polypeptide, either by incorporation of release domains or
able to translocate cargo in a folded state [33]. Except for enzymes homo-fusions of cargo enzymes, may increase the effectiveness
involved in redox pathways, cytosolic proteins rarely contain disul- of the drug by delivering multiple cargos per receptor binding
fide bonds [68], so care should be taken with cargo proteins that event. It is also desirable to improve the efficiency of toxin translo-
contain internal disulfide bonds, as the reducing environment of cation, however an incomplete understanding of the mechanism of
the cytosol is not conducive to their proper folding. In all likeli- translocation hinders a rational approach. Nonetheless, there are
hood, the cargo with the greatest chance of achieving a therapeutic several mechanistic steps that are required for translocation that
effect at the lowest doses is likely to be highly active enzymatic lend themselves to optimization, namely furin cleavage, disulfide
cargo that is easily folded and stable in the mammalian cytosol. bond reduction and membrane insertion/disruption.
Furin cleavage is absolutely required for release of the cargo
7.3. Immunogenicity into the cytosol, however in certain instances high receptor density
and low furin expression can make cleavage rate-limiting. While
The development of neutralizing antibodies against the toxin furin cleaves at the consensus RXXR sequence, optimization of
carrier is the biggest hurdle to their development as delivery vec- the surrounding sequence can enhance the efficiency of furin

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009
6 G.L. Beilhartz et al. / Biochemical Pharmacology xxx (2017) xxx–xxx

cleavage [78]. This can improve delivery efficiency, as it has been [4] R.P. Evens, Pharma success in product development-does biotechnology
change the paradigm in product development and attrition, AAPS J. 18 (1)
shown that ‘‘pre-nicking” diphtheria toxin and Pseudomonas exo-
(2016) 281–285.
toxin A with furin results in increased toxicity [79]. Similarly, the [5] M. Uhlen, L. Fagerberg, B.M. Hallstrom, C. Lindskog, P. Oksvold, A. Mardinoglu,
intracellular disulfide bond that links the diphtheria toxin A A. Sivertsson, C. Kampf, E. Sjostedt, A. Asplund, I. Olsson, K. Edlund, E.
domain to the translocation domain has been shown to be exposed Lundberg, S. Navani, C.A. Szigyarto, J. Odeberg, D. Djureinovic, J.O. Takanen, S.
Hober, T. Alm, P.H. Edqvist, H. Berling, H. Tegel, J. Mulder, J. Rockberg, P.
only at low pH, and that reduction of this bond by as yet undiscov- Nilsson, J.M. Schwenk, M. Hamsten, K. von Feilitzen, M. Forsberg, L. Persson, F.
ered mechanisms is the rate-limiting step in diphtheria toxin Johansson, M. Zwahlen, G. von Heijne, J. Nielsen, F. Ponten, Proteomics. Tissue-
intoxication [34]. based map of the human proteome, Science 347 (6220) (2015) 1260419.
[6] A.D. Cox, S.W. Fesik, A.C. Kimmelman, J. Luo, C.J. Der, Drugging the
Despite several studies, the sequence determinants of optimal undruggable RAS: mission possible? Nat. Rev. Drug Discov. 13 (11) (2014)
membrane insertion/translocation are not well understood for 828–851.
many toxins, and perhaps an unbiased approach to this problem [7] K.H. Khoo, C.S. Verma, D.P. Lane, Drugging the p53 pathway: understanding
the route to clinical efficacy, Nat. Rev. Drug Discov. 13 (3) (2014) 217–236.
is warranted. The incorporation of CPPs into the translocating [8] T. Lehto, K. Ezzat, M.J. Wood, S. El Andaloussi, Peptides for nucleic acid
polypeptide, for example, may facilitate toxin translocation, delivery, Adv. Drug Deliv. Rev. 106(Pt A) (2016) 172–182.
despite the differences in mechanisms. The hydrophobicity of the [9] H. Gao, Q. Zhang, Z. Yu, Q. He, Cell-penetrating peptide-based intelligent
liposomal systems for enhanced drug delivery, Curr. Pharm. Biotechnol. 15 (3)
transmembrane helices involved in pore formation varies between (2014) 210–219.
toxins, the significance of which is not yet understood, although [10] C. Bechara, S. Sagan, Cell-penetrating peptides: 20 years later, where do we
the pattern of hydrophobicity is similar across many disparate stand? FEBS Lett. 587 (12) (2013) 1693–1702.
[11] T. Takeuchi, S. Futaki, Current understanding of direct translocation of
types of pore-forming toxins [80].
arginine-rich cell-penetrating peptides and its internalization mechanisms,
Certain small molecules have been shown to enhance endoso- Chem. Pharm. Bull. (Tokyo) 64 (10) (2016) 1431–1437.
mal release [81,82]. Lysosomotropic agents such as chloroquine [12] B.R. McNaughton, J.J. Cronican, D.B. Thompson, D.R. Liu, Mammalian cell
are weak bases that accumulate in endolysosomal compartments penetration, siRNA transfection, and DNA transfection by supercharged
proteins, Proc. Natl. Acad. Sci. U.S.A. 106 (15) (2009) 6111–6116.
and become protonated. At low doses, they inhibit endosome mat- [13] I. Martin, M. Teixido, E. Giralt, Building cell selectivity into CPP-mediated
uration by raising the lumenal pH, but at high concentrations they strategies, Pharmaceuticals (Basel) 3 (5) (2010) 1456–1490.
cause an accumulation of counter ions, followed by osmotic swel- [14] U. Niesner, C. Halin, L. Lozzi, M. Gunthert, P. Neri, H. Wunderli-Allenspach, L.
Zardi, D. Neri, Quantitation of the tumor-targeting properties of antibody
ling and endosome rupture. This has been labeled the ‘‘proton- fragments conjugated to cell-permeating HIV-1 TAT peptides, Bioconjug.
sponge effect”. Other methods that cause endosomal rupture, Chem. 13 (4) (2002) 729–736.
including photochemical disruption, tend to have mechanism- [15] M. Zdanowicz, J. Chroboczek, Virus-like particles as drug delivery vectors, Acta
Biochim. Pol. 63 (3) (2016) 469–473.
based toxicities. Amphipathic peptides derived from viral [16] N. Kushnir, S.J. Streatfield, V. Yusibov, Virus-like particles as a highly efficient
sequences can increase endosomal escape, possibly by endosomal vaccine platform: diversity of targets and production systems and advances in
lysis or vesicle fusion [81]. These techniques are likely most useful clinical development, Vaccine 31 (1) (2012) 58–83.
[17] C.E. Ashley, E.C. Carnes, G.K. Phillips, P.N. Durfee, M.D. Buley, C.A. Lino, D.P.
in an in vitro setting, as co-dosing of these agents with bacterial Padilla, B. Phillips, M.B. Carter, C.L. Willman, C.J. Brinker, C. Caldeira Jdo, B.
toxins or other delivery systems would be complex. Chackerian, W. Wharton, D.S. Peabody, Cell-specific delivery of diverse
cargos by bacteriophage MS2 virus-like particles, ACS Nano 5 (7) (2011)
5729–5745.
8. Conclusions [18] S.J. Kaczmarczyk, K. Sitaraman, H.A. Young, S.H. Hughes, D.K. Chatterjee,
Protein delivery using engineered virus-like particles, Proc. Natl. Acad. Sci. U.S.
A. 108 (41) (2011) 16998–17003.
The rapid emergence of biologics as potent therapeutics has ini- [19] A.M. Koch, F. Reynolds, H.P. Merkle, R. Weissleder, L. Josephson, Transport of
tiated a race to develop platforms to deliver these diverse mole- surface-modified nanoparticles through cell monolayers, ChemBioChem 6 (2)
cules into cells. While antibodies and their analogs are currently (2005) 337–345.
[20] J. Heyes, L. Palmer, K. Bremner, I. MacLachlan, Cationic lipid saturation
enjoying unquestioned dominance in the biologics market, the real
influences intracellular delivery of encapsulated nucleic acids, J. Control.
potential of proteins as drugs is stifled by the cellular membrane of Release 107 (2) (2005) 276–287.
human cells. This barrier eliminates almost two thirds of the [21] T.M. Allen, P.R. Cullis, Liposomal drug delivery systems: from concept to
clinical applications, Adv. Drug Deliv. Rev. 65 (1) (2013) 36–48.
human proteome and the entire genome from therapeutic inter-
[22] P.P. Deshpande, S. Biswas, V.P. Torchilin, Current trends in the use of liposomes
vention by biologics. The use of bacterial toxins for intracellular for tumor targeting, Nanomedicine (Lond) 8 (9) (2013) 1509–1528.
delivery remains an underappreciated approach to gaining entry [23] W.H. De Jong, P.J. Borm, Drug delivery and nanoparticles: applications and
to this target space. While the delivery of proteins into specific hazards, Int. J. Nanomed. 3 (2) (2008) 133–149.
[24] J.B. Szebeni, Adverse immune effects of liposomes: complement activation,
cells and tissues is not a trivial task, the available technology and immunogenicity and immune suppression, in: D. Peer (Ed.), Harnessing
techniques have progressed to the point where engineering and biomaterials for Nanomedicine: Preparation, toxicity and applications, Pan
examining bacterial toxins as effective delivery vehicles is possible. Stanford Publishing, 2009.
[25] K.B. Knudsen, H. Northeved, P.E. Kumar, A. Permin, T. Gjetting, T.L. Andresen, S.
We propose that bacterial toxins represent a promising platform Larsen, K.M. Wegener, J. Lykkesfeldt, K. Jantzen, S. Loft, P. Moller, M.
for delivery of intracellular protein therapeutics that should be Roursgaard, In vivo toxicity of cationic micelles and liposomes,
explored and considered further. Nanomedicine 11 (2) (2015) 467–477.
[26] J.J. Mekalanos, R.J. Collier, W.R. Romig, Enzymic activity of cholera toxin. II.
Relationships to proteolytic processing, disulfide bond reduction, and subunit
Acknowledgements composition, J. Biol. Chem. 254 (13) (1979) 5855–5861.
[27] A.E. Rabideau, B.L. Pentelute, Delivery of non-native cargo into mammalian
cells using anthrax lethal toxin, ACS Chem. Biol. 11 (6) (2016) 1490–1501.
R.A.M. was supported by Canadian Institutes of Health Research [28] P.D. Dyer, T.R. Shepherd, A.S. Gollings, S.A. Shorter, M.A. Gorringe-Pattrick, C.K.
(CIHR) Operating Grant, and a Brain Canada Multi-Investigator Tang, B.N. Cattoz, L. Baillie, P.C. Griffiths, S.C. Richardson, Disarmed anthrax
Research Initiative Grant. toxin delivers antisense oligonucleotides and siRNA with high efficiency and
low toxicity, J. Control Release 220(Pt A) (2015) 316–328.
[29] X. Liao, A.E. Rabideau, B.L. Pentelute, Delivery of antibody mimics into
References mammalian cells via anthrax toxin protective antigen, ChemBioChem 15 (16)
(2014) 2458–2466.
[30] J.C. Milne, S.R. Blanke, P.C. Hanna, R.J. Collier, Protective antigen-binding
[1] B. Leader, Q.J. Baca, D.E. Golan, Protein therapeutics: a summary and
domain of anthrax lethal factor mediates translocation of a heterologous
pharmacological classification, Nat. Rev. Drug Discov. 7 (1) (2008) 21–39.
protein fused to its amino- or carboxy-terminus, Mol. Microbiol. 15 (4) (1995)
[2] FDA, Novel drugs 2015 Summary, in: C.o.D.E.a. Research (Ed.) 2016.
661–666.
[3] PharmaCompass, Top drugs by sales revenue in 2015: Who sold the biggest
[31] W.P. Verdurmen, M. Luginbuhl, A. Honegger, A. Pluckthun, Efficient cell-
blockbuster drugs?, 2016. http://www.pharmacompass.com/radio-compass-
specific uptake of binding proteins into the cytoplasm through engineered
blog/top-drugs-by-sales-revenue-in-2015-who-sold-the-biggest-blockbuster-
modular transport systems, J. Control. Release 200 (2015) 13–22.
drugs. (Accessed February 23, 2017 2017).

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009
G.L. Beilhartz et al. / Biochemical Pharmacology xxx (2017) xxx–xxx 7

[32] I. Zornetta, L. Brandi, B. Janowiak, F. Dal Molin, F. Tonello, R.J. Collier, C. [58] R.J. Boado, Y. Zhang, Y. Wang, W.M. Pardridge, Engineering and expression of a
Montecucco, Imaging the cell entry of the anthrax oedema and lethal toxins chimeric transferrin receptor monoclonal antibody for blood-brain barrier
with fluorescent protein chimeras, Cell. Microbiol. 12 (10) (2010) 1435–1445. delivery in the mouse, Biotechnol. Bioeng. 102 (4) (2009) 1251–1258.
[33] J. Wesche, J.L. Elliott, P.O. Falnes, S. Olsnes, R.J. Collier, Characterization of [59] P.J. Gaillard, A. Brink, A.G. de Boer, Diphtheria toxin receptor-targeted brain
membrane translocation by anthrax protective antigen, Biochemistry 37 (45) drug delivery, International Congress Series 1277(Drug Transport(ers) and the
(1998) 15737–15746. Diseased Brain) (2005) 185–198.
[34] A. Auger, M. Park, F. Nitschke, L.M. Minassian, G.L. Beilhartz, B.A. Minassian, R. [60] J.H. Han, S.A. Kushner, A.P. Yiu, H.L. Hsiang, T. Buch, A. Waisman, B. Bontempi,
A. Melnyk, Efficient delivery of structurally diverse protein cargo into R.L. Neve, P.W. Frankland, S.A. Josselyn, Selective erasure of a fear memory,
mammalian cells by a bacterial toxin, Mol. Pharm. 12 (8) (2015) 2962–2971. Science 323 (5920) (2009) 1492–1496.
[35] E. Papini, R. Rappuoli, M. Murgia, C. Montecucco, Cell penetration of diphtheria [61] C.J. Wrobel, D.C. Wright, R.L. Dedrick, R.J. Youle, Diphtheria toxin effects on
toxin. Reduction of the interchain disulfide bridge is the rate-limiting step of brain-tumor xenografts. Implications for protein-based brain-tumor
translocation in the cytosol, J. Biol. Chem. 268 (3) (1993) 1567–1574. chemotherapy, J. Neurosurg. 72 (6) (1990) 946–950.
[36] J. Niklas, A. Melnyk, Y. Yuan, E. Heinzle, Selective permeabilization for the [62] P. Calias, W.A. Banks, D. Begley, M. Scarpa, P. Dickson, Intrathecal delivery of
high-throughput measurement of compartmented enzyme activities in protein therapeutics to the brain: a critical reassessment, Pharmacol. Ther. 144
mammalian cells, Anal. Biochem. 416 (2) (2011) 218–227. (2) (2014) 114–122.
[37] F. Grebien, O. Hantschel, J. Wojcik, I. Kaupe, B. Kovacic, A.M. Wyrzucki, G.D. [63] J.L. Cohen-Pfeffer, S. Gururangan, T. Lester, D.A. Lim, A.J. Shaywitz, M.
Gish, S. Cerny-Reiterer, A. Koide, H. Beug, T. Pawson, P. Valent, S. Koide, G. Westphal, I. Slavc, Intracerebroventricular delivery as a safe, long-term route
Superti-Furga, Targeting the SH2-kinase interface in Bcr-Abl inhibits of drug administration, Pediatr. Neurol. 67 (2017) 23–35.
leukemogenesis, Cell 147 (2) (2011) 306–319. [64] R. Irannejad, N.G. Tsvetanova, B.T. Lobingier, M. von Zastrow, Effects of
[38] T. Geiger, A. Wehner, C. Schaab, J. Cox, M. Mann, Comparative proteomic endocytosis on receptor-mediated signaling, Curr. Opin. Cell Biol. 35 (2015)
analysis of eleven common cell lines reveals ubiquitous but varying expression 137–143.
of most proteins, Mol. Cell. Proteomics 11 (3) (2012). M111 014050. [65] V. Bertelsen, E. Stang, The mysterious ways of ErbB2/HER2 trafficking,
[39] M. Yamaizumi, E. Mekada, T. Uchida, Y. Okada, One molecule of diphtheria Membranes (Basel) 4 (3) (2014) 424–446.
toxin fragment A introduced into a cell can kill the cell, Cell 15 (1) (1978) 245– [66] S.C. Hou, H.S. Chen, H.W. Lin, W.T. Chao, Y.S. Chen, C.Y. Fu, C.M. Yu, K.F. Huang,
250. A.H. Wang, A.S. Yang, High throughput cytotoxicity screening of anti-HER2
[40] R. Milo, P. Jorgensen, U. Moran, G. Weber, M. Springer, BioNumbers–the immunotoxins conjugated with antibody fragments from phage-displayed
database of key numbers in molecular and cell biology, Nucleic Acids Res. 38 synthetic antibody libraries, Sci. Rep. 6 (2016) 31878.
(2010) D750–D753 (Database issue). [67] R.J. Abi-Habib, S. Liu, T.H. Bugge, S.H. Leppla, A.E. Frankel, A urokinase-
[41] I. Antic, M. Biancucci, K.J. Satchell, Cytotoxicity of the Vibrio vulnificus MARTX activated recombinant diphtheria toxin targeting the granulocyte-macrophage
toxin effector DUF5 is linked to the C2A subdomain, Proteins 82 (10) (2014) colony-stimulating factor receptor is selectively cytotoxic to human acute
2643–2656. myeloid leukemia blasts, Blood 104 (7) (2004) 2143–2148.
[42] H. Hu, S.H. Leppla, Anthrax toxin uptake by primary immune cells as [68] S. Liu, T.H. Bugge, S.H. Leppla, Targeting of tumor cells by cell surface urokinase
determined with a lethal factor-beta-lactamase fusion protein, PLoS ONE 4 plasminogen activator-dependent anthrax toxin, J. Biol. Chem. 276 (21) (2001)
(11) (2009) e7946. 17976–17984.
[43] R. Zielinski, I. Lyakhov, A. Jacobs, O. Chertov, G. Kramer-Marek, N. Francella, A. [69] N.J. Bulleid, Disulfide bond formation in the mammalian endoplasmic
Stephen, R. Fisher, R. Blumenthal, J. Capala, Affitoxin–a novel recombinant, reticulum, Cold Spring Harb. Perspect. Biol. 4 (11) (2012).
HER2-specific, anticancer agent for targeted therapy of HER2-positive tumors, [70] USFAD Administration, Guidance for Industry: Immunogenicity Assessment
J. Immunother. 32 (8) (2009) 817–825. for Therapeutic Protein Products, in: O.o.M.P. Division of Medical Policy
[44] D.P. Williams, K. Parker, P. Bacha, W. Bishai, M. Borowski, F. Genbauffe, T.B. Development, Center for Drug Evaluation and Research (CDER) (Ed.) 2014.
Strom, J.R. Murphy, Diphtheria toxin receptor binding domain substitution [71] F.M. Foss, DAB(389)IL-2 (ONTAK): a novel fusion toxin therapy for lymphoma,
with interleukin-2: genetic construction and properties of a diphtheria toxin- in: Clin. Lymph. 1 (2) (2000) 110–116. discussion 117.
related interleukin-2 fusion protein, Protein Eng. 1 (6) (1987) 493–498. [72] K.E. Griswold, C. Bailey-Kellogg, Design and engineering of deimmunized
[45] L.F. Jean, J.R. Murphy, Diphtheria toxin receptor-binding domain substitution biotherapeutics, Curr. Opin. Struct. Biol. 39 (2016) 79–88.
with interleukin 6: genetic construction and interleukin 6 receptor-specific [73] J.R. Schafer, B.M. Jesdale, J.A. George, N.M. Kouttab, A.S. De Groot, Prediction of
action of a diphtheria toxin-related interleukin 6 fusion protein, Protein Eng. 4 well-conserved HIV-1 ligands using a matrix-based algorithm, EpiMatrix,
(8) (1991) 989–994. Vaccine 16 (19) (1998) 1880–1884.
[46] A. Mechaly, A.J. McCluskey, R.J. Collier, Changing the receptor specificity of [74] A.S. Parker, Y. Choi, K.E. Griswold, C. Bailey-Kellogg, Structure-guided
anthrax toxin, MBio 3 (3) (2012). deimmunization of therapeutic proteins, J. Comput. Biol. 20 (2) (2013) 152–
[47] T.F. Liu, K.A. Cohen, J.G. Ramage, M.C. Willingham, A.M. Thorburn, A.E. Frankel, 165.
A diphtheria toxin-epidermal growth factor fusion protein is cytotoxic to [75] J.U. Schmohl, D. Todhunter, S. Oh, D.A. Vallera, Mutagenic deimmunization of
human glioblastoma multiforme cells, Cancer Res. 63 (8) (2003) 1834–1837. diphtheria toxin for use in biologic drug development, Toxins (Basel) 7 (10)
[48] T.F. Liu, M.C. Willingham, S.B. Tatter, K.A. Cohen, A.C. Lowe, A. Thorburn, A.E. (2015) 4067–4082.
Frankel, Diphtheria toxin-epidermal growth factor fusion protein and [76] A.S. De Groot, L. Moise, J.A. McMurry, E. Wambre, L. Van Overtvelt, P.
Pseudomonas exotoxin-interleukin 13 fusion protein exert synergistic Moingeon, D.W. Scott, W. Martin, Activation of natural regulatory T cells by
toxicity against human glioblastoma multiforme cells, Bioconjug. Chem. 14 IgG Fc-derived peptide ‘‘Tregitopes”, Blood 112 (8) (2008) 3303–3311.
(6) (2003) 1107–1114. [77] T.K. Kishimoto, J.D. Ferrari, R.A. LaMothe, P.N. Kolte, A.P. Griset, C. O’Neil, V.
[49] A.E. Frankel, J.H. Woo, C. Ahn, F.M. Foss, M. Duvic, P.H. Neville, D.M. Neville, Chan, E. Browning, A. Chalishazar, W. Kuhlman, F.N. Fu, N. Viseux, D.H.
Resimmune, an anti-CD3epsilon recombinant immunotoxin, induces durable Altreuter, L. Johnston, R.A. Maldonado, Improving the efficacy and safety of
remissions in patients with cutaneous T-cell lymphoma, Haematologica 100 biologic drugs with tolerogenic nanoparticles, Nat. Nanotechnol. 11 (10)
(6) (2015) 794–800. (2016) 890–899.
[50] D.A. Vallera, H. Chen, A.R. Sicheneder, A. Panoskaltsis-Mortari, E.P. Taras, [78] R. Hassan, A.C. Miller, E. Sharon, A. Thomas, J.C. Reynolds, A. Ling, R.J. Kreitman,
Genetic alteration of a bispecific ligand-directed toxin targeting human CD19 M.M. Miettinen, S.M. Steinberg, D.H. Fowler, I. Pastan, Major cancer
and CD22 receptors resulting in improved efficacy against systemic B cell regressions in mesothelioma after treatment with an anti-mesothelin
malignancy, Leuk. Res. 33 (9) (2009) 1233–1242. immunotoxin and immune suppression, Sci. Transl. Med. 5 (208) (2013)
[51] R.J. Kreitman, I. Pastan, Antibody fusion proteins: anti-CD22 recombinant 208ra147.
immunotoxin moxetumomab pasudotox, Clin. Cancer Res. 17 (20) (2011) [79] S.A. Shiryaev, A.V. Chernov, V.S. Golubkov, E.R. Thomsen, E. Chudin, M.S. Chee,
6398–6405. I.A. Kozlov, A.Y. Strongin, P. Cieplak, High-resolution analysis and functional
[52] R.J. Kreitman, W.H. Wilson, J.D. White, M. Stetler-Stevenson, E.S. Jaffe, S. mapping of cleavage sites and substrate proteins of furin in the human
Giardina, T.A. Waldmann, I. Pastan, Phase I trial of recombinant immunotoxin proteome, PLoS ONE 8 (1) (2013) e54290.
anti-Tac(Fv)-PE38 (LMB-2) in patients with hematologic malignancies, J. Clin. [80] M.F. Chiron, C.M. Fryling, D.J. FitzGerald, Cleavage of pseudomonas exotoxin
Oncol. 18 (8) (2000) 1622–1636. and diphtheria toxin by a furin-like enzyme prepared from beef liver, J. Biol.
[53] W.S. Sly, H.D. Fischer, The phosphomannosyl recognition system for Chem. 269 (27) (1994) 18167–18176.
intracellular and intercellular transport of lysosomal enzymes, J. Cell. [81] Z. Zhang, M. Park, J. Tam, A. Auger, G.L. Beilhartz, D.B. Lacy, R.A. Melnyk,
Biochem. 18 (1) (1982) 67–85. Translocation domain mutations affecting cellular toxicity identify the
[54] R.J. Desnick, E.H. Schuchman, Enzyme replacement therapy for lysosomal Clostridium difficile toxin B pore, Proc. Natl. Acad. Sci. U.S.A. 111 (10) (2014)
diseases: lessons from 20 years of experience and remaining challenges, Annu. 3721–3726.
Rev. Genomics Hum. Genet. 13 (2012) 307–335. [82] A.K. Varkouhi, M. Scholte, G. Storm, H.J. Haisma, Endosomal escape pathways
[55] D.J. Begley, C.C. Pontikis, M. Scarpa, Lysosomal storage diseases and the blood- for delivery of biologicals, J. Control. Release 151 (3) (2011) 220–228.
brain barrier, Curr. Pharm. Des. 14 (16) (2008) 1566–1580. [83] D.S. D’Astolfo, R.J. Pagliero, A. Pras, W.R. Karthaus, H. Clevers, V. Prasad, R.J.
[56] J. Kreuter, D. Shamenkov, V. Petrov, P. Ramge, K. Cychutek, C. Koch-Brandt, R. Lebbink, H. Rehmann, N. Geijsen, Efficient intracellular delivery of native
Alyautdin, Apolipoprotein-mediated transport of nanoparticle-bound drugs proteins, Cell 161 (3) (2015) 674–690.
across the blood-brain barrier, J. Drug Target. 10 (4) (2002) 317–325. [84] J.G. Naglich, J.E. Metherall, D.W. Russell, L. Eidels, Expression cloning of a
[57] M.J. Coloma, H.J. Lee, A. Kurihara, E.M. Landaw, R.J. Boado, S.L. Morrison, W.M. diphtheria toxin receptor: identity with a heparin-binding EGF-like growth
Pardridge, Transport across the primate blood-brain barrier of a genetically factor precursor, Cell 69 (6) (1992) 1051–1061.
engineered chimeric monoclonal antibody to the human insulin receptor, [85] M.Z. Kounnas, R.E. Morris, M.R. Thompson, D.J. FitzGerald, D.K. Strickland, C.B.
Pharm. Res. 17 (3) (2000) 266–274. Saelinger, The alpha 2-macroglobulin receptor/low density lipoprotein

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009
8 G.L. Beilhartz et al. / Biochemical Pharmacology xxx (2017) xxx–xxx

receptor-related protein binds and internalizes Pseudomonas exotoxin A, J. exploits early to late endosome trafficking machinery, Mol. Microbiol. 23 (3)
Biol. Chem. 267 (18) (1992) 12420–12423. (1997) 445–457.
[86] D.B. Lacy, D.J. Wigelsworth, R.A. Melnyk, S.C. Harrison, R.J. Collier, Structure of [91] B.A. Krantz, R.A. Melnyk, S. Zhang, S.J. Juris, D.B. Lacy, Z. Wu, A. Finkelstein, R.J.
heptameric protective antigen bound to an anthrax toxin receptor: a role for Collier, A phenylalanine clamp catalyzes protein translocation through the
receptor in pH-dependent pore formation, Proc. Natl. Acad. Sci. U.S.A. 101 (36) anthrax toxin pore, Science 309 (5735) (2005) 777–781.
(2004) 13147–13151. [92] V.K. Chaudhary, Y. Jinno, D. FitzGerald, I. Pastan, Pseudomonas exotoxin
[87] S. Choe, M.J. Bennett, G. Fujii, P.M. Curmi, K.A. Kantardjieff, R.J. Collier, D. contains a specific sequence at the carboxyl terminus that is required for
Eisenberg, The crystal structure of diphtheria toxin, Nature 357 (6375) (1992) cytotoxicity, Proc. Natl. Acad. Sci. U.S.A. 87 (1) (1990) 308–312.
216–222. [93] B. Tsai, C. Rodighiero, W.I. Lencer, T.A. Rapoport, Protein disulfide isomerase
[88] J. Jiang, B.L. Pentelute, R.J. Collier, Z.H. Zhou, Atomic structure of anthrax acts as a redox-dependent chaperone to unfold cholera toxin, Cell 104 (6)
protective antigen pore elucidates toxin translocation, Nature 521 (7553) (2001) 937–948.
(2015) 545–549. [94] S. Massey, H. Burress, M. Taylor, K.N. Nemec, S. Ray, D.B. Haslam, K. Teter,
[89] T.M. Koehler, R.J. Collier, Anthrax toxin protective antigen: low-pH-induced Structural and functional interactions between the cholera toxin A1 subunit
hydrophobicity and channel formation in liposomes, Mol. Microbiol. 5 (6) and ERdj3/HEDJ, a chaperone of the endoplasmic reticulum, Infect. Immun. 79
(1991) 1501–1506. (11) (2011) 4739–4747.
[90] E. Lemichez, M. Bomsel, G. Devilliers, J. vanderSpek, J.R. Murphy, E.V. Lukianov,
S. Olsnes, P. Boquet, Membrane translocation of diphtheria toxin fragment A

Please cite this article in press as: G.L. Beilhartz et al., Repurposing bacterial toxins for intracellular delivery of therapeutic proteins, Biochem. Pharmacol.
(2017), http://dx.doi.org/10.1016/j.bcp.2017.04.009

You might also like