Next Article in Journal / Special Issue
Tumor Targeting and Drug Delivery by Anthrax Toxin
Previous Article in Journal
Biofilm-Forming Methicillin-Resistant Staphylococcus aureus Survive in Kupffer Cells and Exhibit High Virulence in Mice
Previous Article in Special Issue
High in Vitro Anti-Tumor Efficacy of Dimeric Rituximab/Saporin-S6 Immunotoxin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Augmenting the Efficacy of Immunotoxins and Other Targeted Protein Toxins by Endosomal Escape Enhancers

1
Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany
2
Institut für Pharmazie, Freie Universität Berlin, 14195 Berlin, Germany
3
Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
*
Author to whom correspondence should be addressed.
Toxins 2016, 8(7), 200; https://doi.org/10.3390/toxins8070200
Submission received: 21 April 2016 / Revised: 14 June 2016 / Accepted: 17 June 2016 / Published: 1 July 2016
(This article belongs to the Collection Immunotoxins 2016)

Abstract

:
The toxic moiety of almost all protein-based targeted toxins must enter the cytosol of the target cell to mediate its fatal effect. Although more than 500 targeted toxins have been investigated in the past decades, no antibody-targeted protein toxin has been approved for tumor therapeutic applications by the authorities to date. Missing efficacy can be attributed in many cases to insufficient endosomal escape and therefore subsequent lysosomal degradation of the endocytosed toxins. To overcome this drawback, many strategies have been described to weaken the membrane integrity of endosomes. This comprises the use of lysosomotropic amines, carboxylic ionophores, calcium channel antagonists, various cell-penetrating peptides of viral, bacterial, plant, animal, human and synthetic origin, other organic molecules and light-induced techniques. Although the efficacy of the targeted toxins was typically augmented in cell culture hundred or thousand fold, in exceptional cases more than million fold, the combination of several substances harbors new problems including additional side effects, loss of target specificity, difficulties to determine the therapeutic window and cell type-dependent variations. This review critically scrutinizes the chances and challenges of endosomal escape enhancers and their potential role in future developments.

Graphical Abstract

1. Introduction

Targeted protein toxins represent one of the high hopes for future drugs in the fight against cancer. They consist of a targeting domain chemically coupled or recombinantly fused to a toxic payload for tumor cell killing [1]. In most of the cases, the targeting moiety is an antibody-based domain including various forms of full regular antibodies, single chain antibodies, bivalent or bispecific diabodies and minibodies [2,3,4,5], and this type of targeted toxins is therefore called immunotoxins. The diversity of these molecules allows the selection of very specific structures for the targeting of tumor cells, however, the protein toxins take their effect inside the cytosol. Therefore, targeting of the toxin to the cell surface is a prerequisite for specificity but not sufficient for efficacy. In many constellations, the toxin remains ineffective since: (1) the antibody is poorly internalized and remains bound to the outside of the cells; (2) is recycled back to the cell surface after internalization; or (3) transported to the lysosomes where it is degraded [6,7,8]. Although these fundamental issues are known for decades, the problems have not been solved yet and no antibody-targeted protein toxin has been approved for tumor therapeutic applications by the authorities to date. It is obvious that a major problem is the endosomal escape within sufficient time. To overcome this drawback, many strategies have been described including approaches to redirect the toxins to endogenous cellular membrane transport complexes of the biosynthetic pathway in the endoplasmic reticulum, disrupting the endosomes, weaken the membrane integrity of endosomal membranes or diving through these membranes with the assistance of cell penetrating peptides [9,10,11,12].
All strategies including physicochemical techniques require molecules that interact more or less directly with membranes and comprise essentially small chemical molecules, secondary metabolites, peptides and proteins. A common feature of all these substances is that they are per se not target-specific and distribute with other kinetics than the targeted toxins. Therefore, a number of requirements can be formulated for endosomal escape enhancers: (1) they must not be toxic for regular cells; (2) they must not mediate the cytosolic uptake of the targeted toxin into off-target cells; (3) their presence at the site of action must by synchronized with the presence of the targeted toxin; (4) they must be biodegradable or excretable; and (5) they must not substantially interfere with metabolic processes of the organism, e.g., interact with hormones. It is easy to recognize that most of these criteria can only be sufficiently tested in in vivo systems, however, most of the published endosomal escape enhancers have only be tested in vitro, which may explain the lack of success of these strategies in clinical applications. The present review describes the different approaches to augment the efficacy of targeted toxins by endosomal escape enhancers including chemical, viral, bacterial and eukaryotic substances. It then discusses the pros and cons of the different techniques with regard to the mentioned requirements and weighs the prospects and risks for future clinical development of the different strategies.

2. Chemical Enhancers

Many different chemical enhancers have been described in the literature to augment the efficacy of targeted toxins. These compounds are generally small chemical molecules with great variability that increase in some way or another the cytotoxicity of targeted toxins. Within the chemical enhancers, the compounds can be classified in several subgroups depending on their chemical nature and molecular mechanism. This includes the subgroups of lysosomotropic amines, carboxylic ionophores, calcium channel antagonists, and other organic compounds. Detailed information is provided for each of the subgroups in the following subsections.

2.1. Lysosomotropic Amines

When cells are treated with targeted toxins, they are commonly internalized into cells by receptor-mediated endocytosis and then undergo their intracellular routing. A substantial amount of targeted toxins are not able to reach their intracellular target and accumulate in the lysosomes where they are degraded. This is one of the main limitations of targeted toxins and results in the decrease of their efficacy [13]. Lysosomotropic amines increase the lysosomal pH and therefore interrupt the degradation of proteins by pH-dependent lysosomal enzymes [14]. Since they have their site of action inside the lysosomes, only targeted toxins that mainly accumulate in these organelles are likely to be enhanced. A list of the lysosomotropic amines that have been reported to facilitate the cytosolic delivery of targeted toxins is presented in Table 1.
Ammonium chloride was firstly used to enhance the cytotoxicity of an anti-melanotransferrin-ricin A chain immunotoxin [15]. The cytotoxic effects were enhanced roughly 100-fold and this encouraged the investigation about combinatorial effects of ammonium chloride with further targeted toxins. The most prominent effects were observed in the case of targeted toxins based on ricin toxin A chain (RTA). As an example, the cytotoxicity of three RTA immunotoxins directed to the cell surface antigens Thy 1.2 (cluster of differentiation 90, CD90), melanotransferrin and CD5 was augmented by 5.7, 42 and 6700 folds, respectively [16]. On the contrary, the cytotoxicity of diphtheria toxin was blocked when it was administered to cells in the presence of ammonium chloride [17], which was not surprising since it has been known that inhibition of the vacuolar-type H(+)-ATPase also blocked diphtheria toxin toxicity [18].
Chloroquine is another lysosomotropic amine that has been extensively studied. The highest enhancing effects were again observed in the case of targeted toxins containing RTA. The immunotoxin T101-RTA was enhanced 2500-fold by chloroquine [16]. Chloroquine was also efficient in enhancing the cytotoxic effects of targeted toxins composed of gelonin [19], pokeweed antiviral protein [20] and saporin [21], although the enhancing effects were moderate.
Other lysosomotropic amines that are able to enhance the efficacy are amantadine [22], methylamine, dimethylamine, trimethylamine [16], lipopolyamines [23], quinacrine (mepacrine) [19] and glycylphenylalanine 2-naphthylamide [24]. All compounds were simultaneously co-administered with the targeted toxins and are believed to act in the same way as proton reservoirs through their amine groups thus preventing the acidification of the lysosomes.

2.2. Carboxylic Ionophores

Carboxylic ionophores have been successful in enhancing the delivery of targeted toxins to the cytosol. Carboxylic ionophores integrate in lipid bilayers and mediate the exchange of monovalent cations across the membranes. An example of this mechanism is the exchange of sodium ions and protons by monensin [35]. When administered to cells, carboxylic ionophores increase the lysosomal pH and this finally results in blocking protein degradation inside the lysosomes [36]. At the same time, the inhibition of lysosomal degradation may augment the cytotoxic effects of targeted toxins as observed for lysosomotropic amines. A list of such carboxylic ionophores is available in Table 2.
Monensin is the most well studied compound within the carboxylic ionophores. The highest enhancement effects were observed in the case of targeted toxins comprising ricin A chain. The cytotoxicity of the immunotoxin T101-RTA was augmented 50,000 folds in the presence of monensin [16]. In another example, an immunotoxin recognizing the neural cell adhesion molecule (SEN36-RTA) was enhanced by 12,000-fold when co-applied with monensin [37]. As a third example, the amount of 3A1-rRTA immunotoxin required to inhibit protein synthesis by 50% was decreased by 1000 folds [38]. Monensin also augmented the cytotoxic effects of a targeted toxin consisting of transferrin and saporin by 1250-fold [39], and more moderately of two gelonin-based targeted toxins directed to the gonadotropin receptor [19] and the disialoganglioside GD2 [32].
On the other hand, the use of monensin in vivo is hindered by poor solubility and a short half-life [40]. In order to overcome these limitations, monensin was conjugated to human serum albumin [41]. Alternatively, monensin was delivered in lipid emulsions [42], encapsulated in liposomes [43] or in poly(lactic-co-glycolic acid) (PLGA) polymer nanoparticles [44]. These strategies that were all conducted in conjunction with ricin A chain-based targeted toxins managed to augment the solubility and to prolong the half-life of the chemical enhancer.
Other carboxylic ionophores that have potentiated the cytotoxic effects of targeted toxins are grisorixin, lasalocid and nigericin. These three compounds enhanced the T101-RTA immunotoxin by 25,000; 33,000 and 6700 folds, respectively [16]. Significant increase in cytotoxicity was observed as well for the combination of nigericin with transferrin-RTA, anti-common acute lymphoblastic leukemia antigen (CALLA, CD10)-RTA [45] and anti-carcinoembryonic antigen (CEA)-RTA [46] targeted toxins.

2.3. Calcium Channel Antagonists

Another group of chemicals that were able to enhance the cytotoxic effect of targeted toxins are the calcium channel antagonists (Table 3). Verapamil was first identified as a cytotoxicity enhancer for two Pseudomonas exotoxin (PE)-based toxins targeted to the epidermal growth factor (EGF) receptor and the transferrin receptor [51]. Verapamil was also reported to enhance targeted toxins based on ricin A chain [52] and gelonin [19]. In order to find compounds with less in vivo toxicity, four verapamil analogs (D792; D595; D528; Sz45) were investigated. These compounds were able to enhance the cytotoxicity of targeted toxins in the range of 2−67 folds [53].
Further calcium channel antagonists that showed cytotoxicity enhancement of targeted toxins are diltiazem and methoxyverapamil, both presenting combinatorial effects with the targeted toxins HB21-PE and EGF-PE [24]. Two indolizines (SR 33287 and SR 33557) potentiated the cytotoxic effects of the anti-CD5 T101-RTA immunotoxin by 620-fold [54]. The highest enhancing effects within this group of compounds were observed in the case of perhexiline that increased the cytotoxicity of two ricin A chain immunotoxins directed against CD5 and HLA-DR class II antigens present in leukemia cells up to 2000 folds [55].
The enhancing ability of calcium channel antagonists is probably associated to the inhibition of lysosomal degradation of targeted toxins and actually does not correlate with the calcium-antagonistic activity [53]. Verapamil delays degradation in lysosomes and this could result in enhanced toxicity, alternatively it may increase the therapeutic potential of targeted toxins by some general effects on membrane permeability [51]. In the cases of perhexiline and indolizines, the delay of protein degradation may occur as a consequence of the fact that they inhibit the acid lysosomal sphingomyelinase [54]. This leads to changes in the membrane lipid composition of intracellular organelles, supposedly modulates the intracellular routing of targeted toxins and facilitates their delivery to the cytosol [55].

2.4. Other Organic Compounds

Other organic compounds that do not fit into the classification defined in the previous subsections have been also reported to enhance the cytotoxicity of targeted toxins. A list of these compounds is shown in Table 4. Retinoic acid enhanced the cytotoxicity of several targeted toxins comprising ricin A chain but failed to deliver diphtheria toxin and Pseudomonas exotoxin to the cytosol [56]. The mechanism of action of retinoic acid may be associated to the modulation of the intracellular trafficking of targeted toxins into the Golgi apparatus.
Further organic compounds have shown significant increase in the cytotoxicity of targeted toxins, however, their mechanism of action is not completely clear and different hypothesis have been formulated for each of the compounds. Brefeldin-A enhanced two ricin A chain immunotoxins and is believed to have its effect on the Golgi apparatus and on the vesicular routing [57]. The combination of Pseudomonas exotoxin-based immunotoxins with cyclosporin A caused remarkable synergistic cytotoxicity in several cancer cell lines, although the combinatorial effect was not linked to the immunosuppressive activity [58]. Similarly, wortmannin enhanced the cytotoxicity of saporin- and gelonin-based targeted toxins, but the effect may occur through an alternative pathway not involving the phosphatidylinositol-3-kinase inhibition [59].
Organic polymers comprise a big variety of compounds that has been investigated mainly for gene delivery [60]. Amongst this group of compounds, there is one, namely polyamidoamine, that has been considered for the enhanced cytosolic delivery of non-targeted toxins [61,62]. Further investigation of polyamidoamine effects in the context of targeted toxins would be of interest.

3. Enhancers of Viral and Bacterial Origin

Viruses have deployed effective intrinsic mechanisms in order to enter the cytosol of their host cells. Viruses are able to fuse directly with cellular membranes resulting in the cytosolic release of the viral particles but they are also internalized via endocytosis [65]. In case they are internalized by endocytosis, they are delivered into early or late endosomes. To exploit the cellular protein machinery, the viruses need to escape from the endosomes, a process that is mediated by specific viral fusion proteins [66] and cell-penetrating peptides (CPPs). Cell penetrating peptides, also termed protein-transduction domains, are widely used as tool to mediate the cytosolic delivery of proteins and other molecules [67,68]. One of the most prominent CPPs is the transactivating factor of the human immunodeficiency virus (HIV) [69]. There are many known different cell penetrating peptides [70]. They are frequently composed of cationic amino acids such as arginine and lysine. CPPs are widely used to augment the endosomal escape of protein therapeutics, e.g., CPPs derived from viruses have been used to deliver different proteins such as green fluorescent protein (GFP), insulin or β-galactosidase into the cytosol of cells [71,72]. As a general delivery platform, a fusion protein of the HIV transactivator of transcription (Tat) and calmodulin was designed in order to drag different calmodulin binding proteins into the cytosol of eukaryotic cells [73]. However a drawback of this approach is the non-specific delivery of protein cargo into off-target cells. Due to their small size of only several amino acids, the sequence of a CPP can be easily integrated into the sequence of targeted, recombinant anti-tumor toxins, which resulted in a targeted CPP. By inserting cleavage sites CPPs can be cleaved off after cytosolic delivery of the protein cargo. One example is the construction of a targeted anti-tumor toxin consisting of human EGF, a CCP from the hepatitis B virus and an endosomal cleavable peptide [74]. The fusion of viral CPPs to ribosome-inactivating proteins improved significantly the efficacy of these toxins against cancer cells [75,76,77]. The products are therefore optimized targeted anti-tumor toxins with fusogenic properties (Table 5).
Bacterial toxins are widely used as toxin components of targeted toxins. The most often used bacterial toxins for the design of targeted toxins are truncated versions of Pseudomonas exotoxin A (PE) [85]. PE comprises three domains, a receptor binding domain (Ia), a membrane translocation domain (II) and the ADP-ribosyl transferase domain (Ib and III) [86]. The truncated domain II mediates the translocation of domain Ia and III into the cytosol via the ER-associated degradation pathway [87]. The specific domains of PE were optimized by evolution in order to facilitate the efficient intracellular delivery of the ADP-ribosyl transferase domain into the cytosol [87,88]. For the design and efficient delivery of targeted toxins, it might be therefore obvious to make use of evolutionarily optimized transduction mechanisms rather than integrating artificial transduction domains into targeted toxins such as CPPs. For instance, it has been shown for diphtheria toxin from Corynebacterium diphtheria that the native diphtheria transduction domain (amino acids 190–390) exhibited the highest cytotoxicity in a human EGF receptor-targeted anti-tumor toxin. Fusions between several viral protein transduction domains and diphtheria toxin A chain were less cytotoxic in target cells expressing the human EGF receptor [89]. It is therefore not surprising that a large number of targeted toxins are based on Pseudomonas exotoxin A and diphtheria toxin [90,91,92,93,94,95,96,97,98,99,100,101]. A fusion protein of interleukin-2 and a truncated diphtheria toxin consisting of 389 amino acids called DAB(389)IL-2 was the first and to date sole targeted protein toxin that has been approved by the U.S. Food and Drug Administration under the name denileukin diftitox (Ontak®) [102]. A ten amino acids long motif in the transmembrane helix 1 of diphtheria toxin that is conserved in anthrax (see next paragraph) and botulinum neurotoxin serotypes mediates the delivery of the toxic domain from the lumen of early endosomes to the cytosol [103]. Using DAB(389)IL-2 it has been demonstrated that introduction of a L221E mutation into a highly conserved residue within this motif results in a nontoxic phenotype that is unable to mediate cytosolic translocation [103]. Thus, this domain acts as an effective endosomal escape enhancer.
A further intriguing approach to augment the endosomal escape process of targeted toxins is the utilization of the protective antigen of Bacillus anthracis. This bacterium produces a lethal toxin (anthrax toxin) that is composed of three domains: the lethal factor (LF), edema factor (EF) and protective antigen (PA). LF is a toxic zinc protease and EF an adenylate cyclase that elevates intracellular cAMP levels. PA mediates the endosomal escape by forming an acid induced pore in the endosomal membrane [104]. PA is activated by cellular furins on the cell surface. Since the PA-mediated delivery of LF and EF is a highly efficient process, attempts have been made to utilize this mechanism for targeted tumor therapies. By replacing the native furin cleavage site by tumor-specific cleavage sites, the PA-mediated endosomal escape process can be utilized for the delivery of ADP-ribosyl transferases [105,106]. The PA-mediated delivery has been also deployed for the target specific delivery of DNases such as the cytolethal distending toxin [107] into tumor cells [108]. However, although the results of these studies are promising, the strategies with modified anthrax toxins are afflicted with non-specific activation by regular cells resulting in elevated side effects.
In addition to the use of specific bacterial protein transduction domains, integral attempts have been made to co-deliver protein based cytolysins into the lysosomes of the cancer cells that have been loaded with targeted toxins. Known cytolysins are streptolysin O from Streptococcus pyogenes [109], perfringolysin O from Chlostridium perfringes [110] and listeriolysin O from Listeria monocytogenes [111]. In general, cytolysins are able to induce pore formation in lipid bilayers resulting in the leakage of membranes. By fusing a targeting ligand (human fibronectin domain) to listeriolysin O, a receptor-specific uptake of this construct into lysosomes was achieved [112]. This was also observed for liposomal listeriolysin O that was co-encapsulated with the type I ribosome-inactivating protein (RIP) gelonin [113]. The co-delivery of enhancers and targeted anti-tumor toxins resulted in an augmented cytosolic release of the anti-tumor toxin and increased efficacy; however, a disadvantage of this strategy is the potential immunogenicity of the enhancer. Although it is advantageous to recruit immune effector cells to the tumor site, it is a drawback when the immune response is directed against the enhancer or toxins themselves, which can, in worst case, result in anaphylactic shock.
Viral and bacterial enhancers for the endosomal escape of targeted toxins revealed great potential for the efficient cytosolic transfer. These enhancers are mostly based on particular protein domains that were optimized by evolution. As targeted toxins are protein-based therapeutics, it is straightforward to utilize viral or bacterial derived enhancer domains in recombinant fusion proteins. This is advantageous since the targeting ligand, the effector molecule (toxin) and the delivery mechanism can be integrated into a single clearly defined therapeutic compound.

4. Enhancers of Eukaryotic Origin

4.1. Proteins and Peptides

The enhancers of eukaryotic origin mainly comprise CPPs and pore forming proteins. Currently, hundreds of CPPs have been found and used in biomedical research. CPPs are derived from various proteins including transactivators of gene transcription, DNA/RNA-binding proteins, antimicrobial peptides, viral particle envelope proteins and plant circular skeletal proteins [12]. CPPs of viral origin have already been described before in this review. The most applied CPP of eukaryotic origin is penetratin, a 16-amino acid long polypeptide corresponding to the third helix of the DNA binding domain (homeodomain) of Antennapedia, a Drosophila transcription factor [114]. The peptide exists in its natural form, in modified forms [115] or as synthetic analogs [116]. It has been widely used for the cytosolic delivery of nucleic acids and proteins but was mainly ignored for targeted toxins [117]. Since the internalization of penetratin is receptor-independent [114] (as typical for all CPPs), it might be difficult to erase this unwanted property in targeted toxins although successfully shown for the PreS2-domain of hepatitis-B virus surface antigen [83,84]. Time will tell us whether CPPs have a future in the delivery of targeted toxins.
A completely different approach is the use of pore forming proteins. Perforins are cytolytic proteins found in the granules of cytotoxic T lymphocytes and natural killer cells that share homology with cholesterol-dependent cytolysins from Gram-positive bacteria [118]. They rather circumvent the endosomal escape instead of enhancing it. Since these proteins form pores, they cannot only be used to bring toxic proteins into the cell but are also toxic by themselves. A recombinant immunotoxin consisting of a fragment containing the N-terminal 34 amino acids of human perforin and the C-terminus of a humanized anti-cytotoxic T-lymphocyte-associated protein (CTLA-4, CD152) scFv antibody selectively depleted activated T cells to prevent transplant rejection [119].
Granzymes are potent apoptosis inducing serine proteases of cytotoxic lymphocytes. Following receptor-mediated endocytosis, perforins mediate the cytosolic uptake of granzymes. A targeted toxin consisting of vascular endothelial growth factor and mutated staphylococcal enterotoxin A was used to attract cytotoxic T lymphocytes, which then secreted perforin and granzyme B (abbreviated as GzmB, Gb or GrB) around the tumor resulting in granzyme B-mediated death of tumor cells supported by perforins [120]. Although reliant on perforins in natural attacks, granzyme B can also effectively kill tumor cells in the absence of perforin when targeted to cell surface receptors [121]. A number of research groups took advantage of this phenomenon (Table 6). Granzyme B genetically fused to H22, a humanized scFv specific for CD64, led to a target cell-specific cytotoxicity with a half-maximal inhibitory concentration (IC50) between 1.7 and 17 nM [122]. A similar cytotoxicity (IC50 between 1.2 and 6.4 nM) was achieved when using granzyme M, however, target cells were killed efficiently even in the presence of the granzyme B inhibitor serpin B9 that is often expressed in solid tumors [123]. Resistance against serpin B9 was also observed for a mutant granzyme B (R201K) [124]. The same mutant was also used with an epidermal growth factor receptor-specific scFv antibody [34]. In another immunotoxin granzyme B was fused to the humanized anti-Her2/neu scFv antibody 4D5 and for further enhancement, a fusogenic peptide (called 26) was introduced [125]. Treatment with nanomolar concentrations of GrB/4D5/26 resulted in specific cytotoxicity, induction of apoptosis, and efficient downregulation of signal pathways. Even tumor cells that are resistant to the tyrosine kinase inhibitor lapatinib or the antibody trastuzumab, and cells resistant to chemotherapeutic agents showed no cross-resistance to the GrB-based fusion proteins [125].
Ricin is a plant ribosome-inactivating protein consisting of a cell-binding B chain and a catalytic A chain. Following endocytosis, ricin is transported in vesicular carriers to the endoplasmic reticulum where the two chains are separated reductively. The A chain embeds in the membrane of the endoplasmic reticulum and then retrotranslocates across this membrane [126]. Although the B chain appears to be not involved in the cytosolic translocation, it has been shown that the presence of the B chain enhances the effect of A chain-based immunotoxins. Highly purified preparations of the two chains were separately coupled to anti-human immunoglobulin antibodies and mixtures of the resulting immunotoxins markedly synergized in vitro in their ability to kill target cells [127]. In contrast, A chain and B chain-containing immunotoxins of irrelevant specificity did not synergize with the specific immunotoxins indicating that the synergy is specific [127]. In vitro killing of target cells by either univalent antibody fragments or divalent full antibodies of rabbit anti-human immunoglobulins coupled to ricin A chain can be specifically potentiated by a piggyback treatment with ricin B chain coupled to goat anti-rabbit immunoglobulins [128]. A bispecific monoclonal antibody recognizing both CEA and RTA was tested for its ability to target the ricin A chain to CEA-expressing tumor cells alone and in combination with the B chain [129]. The antibody induced significant cytotoxicity against target cells using a ricin toxin A chain concentration below that known to be intrinsically cytotoxic. The addition of the B chain then further enhanced the cytotoxicity of the bispecific antibody [129].

4.2. Secondary Metabolites

In addition to proteins and peptides of eukaryotic origin, some secondary plant metabolites show a very promising potential to enhance the endosomal escape of targeted toxins. In particular some glycosylated triterpenoids (saponins) of the oleanane type isolated from Gypsophila paniculata L. (baby’s breath) and Saponaria officinalis L. (common soapwort) have the ability to specifically augment the cytotoxicity of several ribosome-inactivating proteins [131,132,133]. This augmentation is neither due to a permeabilization of the plasma membrane—it occurs at non-permeabilizing concentrations of the glycosylated triterpenoid—nor due to an increase of the rate of endocytic events, but rather due to the mediation of an enhanced endosomal escape [134,135]. Findings hinted at an interaction of the toxins with the saponins at acidic pH as it is found in late endosomes and the lysosome (pH ~5) [136]. Further, the acidic environment is even a prerequisite for the synergistic action since inhibition of vesicle acidification by bafilomycin A or chloroquine restored the survival of the tested cells [137]. It is postulated that the protonation of the glucuronic acid, which is present in this particular group of saponins, and interaction with endosomal cholesterol may play an important role [137,138]. Targeted toxins can become enhanced in their cell killing efficacy by these saponins dependent on cell line and amount of target receptor expression by 3000-fold up to 4,000,000-fold [101,139,140,141], which resulted in a broadening of the therapeutic window in mice between 10-fold and 500-fold. After injection of the targeted toxins, a substantial reduction in the tumor volume occurred and complete remissions were seen in many cases of different tumor models [142,143,144]. The tumor regression across all these studies was in average about 90% and the required dose was only 2% of the dose used for a treatment without saponins. The lower dose resulted in decreased side effects and reduced immunogenicity.
SO1861 from Saponaria officinalis L. and SA1641 from Saponinum album (a saponin composite from Gypsophila spec.) are two of the few saponins that were found to display such tremendous synergism with several type I RIPs, such as saporin [101,133], dianthin [101,145] or agrostin [132]. In contrast, the cytotoxicity of the A chain from the type II RIP ricin was only enhanced 16-fold [101] and the cytotoxicity of the bacterial Pseudomonas exotoxin A remained unaffected by the combinatory treatment [101,137].

5. Synthetic Peptide Enhancers

As described before, there are a number of peptides of viral, bacterial and eukaryotic origin that have been successfully used to enhance the cytotoxicity of targeted toxins. Additionally, peptides have been synthetically produced and evaluated for their potentiating effects.
A main group of synthetic peptides that showed significant increase in protein delivery are polyarginines. Peptides consisting of a different number of sequential arginines increased the cytosolic delivery of proteins such as green fluorescent protein by modulating protein transport in the late endosomes and lysosomes [146].
In the case of targeted toxins, a polyarginine synthetic peptide (Arg9) was fused to an immunotoxin containing a truncated version of Pseudomonas exotoxin (PE35) and directed to the carcinoembryonic antigen (CEA). The Arg9 peptide increased the cytotoxicity of PE35/CEA(Fv)/KDEL markedly [147]. Another synthetic peptide called pJVE doubled the cytotoxic effects of the RIP dianthin when it was delivered to cells as a fusion protein consisting of transferrin as targeting moiety, pJVE as enhancer and dianthin as toxic moiety (Tfn-pJVEDIA). The pJVE peptide interacts with the endosomal membranes and facilitates the release of targeted toxins into the cytosol [80].

6. Physicochemical Techniques

The endosomal entrapment of therapeutics can also be overcome by physicochemical techniques. Such techniques do not rely on the usage of molecules that modulate the intracellular trafficking of toxins by their own. Instead, they are based on the use of physical principles or chemical reactions that directly trigger the intracellular drug delivery and may disrupt the endosomal or lysosomal membranes. Some examples of these techniques are ultrasounds [148], magnetic nanoparticles [149] and plasmonic nanobubbles [150]. Thus far, most of these techniques have only been utilized for the delivery of therapeutics other than proteins such as nucleic acids or small chemotherapeutic drugs. However, a technique called photochemical internalization has been extensively studied for the delivery of targeted toxins.
Photochemical internalization is a strategy based on photodynamic therapy for the cytosolic delivery of protein therapeutics that have been trapped during their intracellular trafficking inside the endosomes and lysosomes [151,152]. The release of targeted toxins is triggered by a light-induced photochemical reaction. When targeted toxins and a photosensitizer are administered to cells, both compounds are internalized and accumulate in the endosomes and lysosomes. Then, cells are exposed to light and reactive oxygen species (mainly singlet oxygen 1O2) are produced by the photosensitizer. The chemical reactions induced by singlet oxygen lead to the disruption of endosomal membranes [153]. Subsequently, targeted toxins are released into the cytosol and they reach their intracellular target. The most common photosensitizers for photochemical internalization are disulfonated compounds based on tetrapyrrole ring systems (porphins) [10,154,155].
This approach has been proven successful in enhancing the cytotoxicity of several targeted toxins (Table 7). They were composed of either gelonin or saporin, and targeted to different cellular receptors, including CD44 [156], CD133 [157,158], chondroitin sulfate proteoglycan 4 [159], EGFR [160], epithelial cell adhesion molecule (EpCAM, EGP-2) [161,162], human epidermal growth factor receptor 2 (HER2) [163] and vascular endothelial growth factor receptor [164]. Photochemical internalization is a minimally invasive strategy for light-controlled endosomal escape of targeted toxins that can be used for many different kinds of cancer cells [10].

7. Discussion

Surgery, radiotherapy and conventional chemotherapy are the most important means in the treatment of cancer. While surgery is limited to accessible tumors and in most cases is not suitable for metastasized cancer, radiotherapy and chemotherapy are accompanied by undesired side effects and adequate efficacy is often missing due to formation of resistances. In the last decades targeted tumor therapies that address more specific properties of tumor cells were developed; they mainly comprise receptor tyrosine kinase inhibitors [169] and antibody-based therapies [2]. The cytotoxic effect of antibodies can be mediated by direct effects of the antibody such as acting as receptor antagonist or by inducing apoptosis, by indirect effects such as complement dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity or modulation of T cell functions, and by specific effects on the tumor environment in particular on vascularization [170]. To augment the effect of antibodies, they were linked to cytotoxic substances, either small molecules or protein-based toxins [171]. The antibodies can be replaced by other targeting ligands such as cytokines or growth factors, which also results in targeted toxins [171]. Conjugates with small toxic molecules are more advanced than immunoconjugates with protein-based toxins, but they still reveal problems with linker chemistry, conjugate homogeneity and overall stability [172,173,174]. Moreover, the toxic substances in antibody small drug conjugates are typically competitive inhibitors, whereas the protein toxins are enzymes. Therefore, only a small number of these toxic proteins must enter the cytosol to mediate cell death [175]. Although protein-based targeted toxins appear to have unbeatable advantages including high target-specific toxicity and recombinant production of homogenous molecules, no antibody-targeted protein toxin for tumor therapeutic applications has been launched to date and only one ligand-targeted fusion protein, denileukin diftitox, was approved by the U.S. Food and Drug Administration [102]. As described in this review article, the low cytosolic uptake of the toxin components is a major concern regarding therapy with targeted toxins. Reasons for this are poor internalization, recycling to the cell surface and lysosomal degradation [6,7,8]. Thus, the advantage of the high efficacy of the protein toxin is undermined by the necessity to apply high doses of the toxin to gain the desired rate of cytosolic translocation. It is notable that denileukin diftitox that uses its evolutionarily developed intrinsic endosomal escaper enhancer did not lead to a breakthrough in the application of targeted toxins even 17 years after approval.
It is therefore obvious that efficacy enhancers are important tools to improve the endosomal escape rate and accordingly decrease the dosage. All the efficacy enhancers presented here (Figure 1), independent of their origin and structure, show substantial enhancer effects, however, none of them is tumor target specific by itself; they all depend on the specificity of the targeted toxin. In the case of off-target effects of the toxin, these unwanted effects might also be enhanced and result in increased side effects. This can be the case due to lack of antibody specificity resulting from the presence of the target receptor on healthy tissue (target dependent toxicity) or due to targeted toxin binding to other cell surface components rather than specifically to its target receptor (target independent toxicity) [176]. Therefore, additional measures are required to prevent enhancement of off-target effects.
Another problem with enhancer substances concerns pharmacokinetic issues. In most cases, the enhancer substance is applied independently from the drug and has its own behavior in liberation, absorption, distribution, metabolization and excretion. Since both compounds, the enhancer and the targeted toxin, must be at the site of interaction at the same time, a careful synchronization of both kinetics is required. Moreover, chemical enhancers, secondary metabolites and photosensitizers are non-specifically taken up by off-target cells. All these facets make it very difficult to determine the mutual dose dependency of both substances, the spatial and temporal distance of both applications, and the therapeutic window.
There are mainly two solutions for the mentioned problems, first, to also target the enhancer molecules to the tumor cells, but independently of the targeted toxin, and second, to combine enhancer and targeted toxin in a single drug molecule. Neither solution can solve both problems discussed before. The first solution avoids off-target uptake of the enhancer and therefore minimizes undesired enhancement of misguided targeted toxins to regular cells but cannot ensure synchronization while the second solution results in synchronization of the kinetics but will still enhance the cytotoxic effect if the antibody binds and is internalized by an off-target cell.
Cell penetrating peptides can be fused to the toxin moiety to make them target cell specific [80,83,84]. The idea is to inactivate the penetrating property until the antibody has been bound to the target receptor and internalized. A possible solution is to place the CPP between targeting moiety and toxin and include an endosomal cleavable peptide that leads to the release of the targeting moiety inside the endosomes, which in turn results in exhibition of the CPP [83,84]. Chemical enhancers and secondary metabolites can be encapsulated into targeted nanocarriers; a pH-sensitive endosomal stimulus can then result in controlled release [177,178]. The nanocarriers can be in addition spiked with the targeted toxins or applied independently, each variant associated with the pros and cons discussed before. Further specificity might be achieved by use or design of toxins that are predominantly activated only in tumor cells as described for apoptin [179,180]. In the case of photochemical internalization, nothing happens even in the case of colocalization of drug and enhancer in off-target cells. Light exposure then results in disruption of the endosomal membrane. Additional specificity is thus being obtained by only illuminating the site of the tumor [151]. This is however achieved at the expense of completeness in the treatment of metastases since unidentified micrometastases cannot be embraced by this system except for unenhanced targeting. This problem might be solved by targeted bioluminescence, however, firefly luciferase bioluminescence does not generate sufficient photons to induce photodynamic toxicity [181].
The use of enhancers leads to a further general problem, the immunogenicity of drugs. In particular, peptide- and protein-based enhancers can contain immunogenic epitopes, and small molecule enhancers, even if not immunogenic by themselves, might boost a possible immune response against the targeted toxin. Strategies to deimmunize enhancers might be applied in the same way as described for targeted toxins [96]. Wherever possible, human protein sequences should be used as exemplified by humanized antibodies or human toxins such as angiogenin or granzymes.

8. Conclusions

Antibody targeted delivery of protein toxins to tumor cells is one of the most promising ideas in the fight against cancer, however, the fact that decades of research have not resulted in any approved antibody-targeted protein toxin for tumor therapy shows that cytosolic protein delivery is a formidable challenge. The main barrier is the endosomal membrane that prevents release of the toxin before degradation or recycling. To overcome this obstacle, a number of strategies were developed that can be summarized as endosomal escape enhancers. The nature of these enhancers is multifarious including chemical substances, cell-penetrating peptides, protein domains, secondary metabolites, and light-sensitive substances. Until now, it is not finally solved how spatio-temporal synchronization of enhancer and drug should be accomplished, and how enhancement of drugs that were misdirected into regular cells should be prevented. Further ideas are definitely required to solve the conflict between linked enhancer/drug delivery and independently targeted enhancers. This might include new controlled delivery techniques and novel tumor activated toxins.

Acknowledgments

The German Research Foundation (DFG) is acknowledged for providing the funding of a postdoctoral research fellowship to Roger Gilabert-Oriol (GI 1135/1-1).

Author Contributions

All authors contributed to the entire review article with particular focus on abstract, introduction, eukaryotic enhancers, discussion and conclusions by H.F., bacterial and viral enhancers by A.W. and all other subsections by R.G.-O.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fuchs, H.; Bachran, C. Design of targeted protein toxins. In Drug Delivery in Oncology–From Basic Research to Cancer Therapy; Kratz, F., Senter, P., Steinhagen, H., Eds.; Wiley-VCH: Weinheim, Germany, 2011; Volume 3, pp. 1443–1487. [Google Scholar]
  2. Alewine, C.; Hassan, R.; Pastan, I. Advances in anticancer immunotoxin therapy. Oncologist 2015, 20, 176–185. [Google Scholar] [CrossRef] [PubMed]
  3. Holliger, P.; Prospero, T.; Winter, G. “Diabodies”: Small bivalent and bispecific antibody fragments. Proc. Natl. Acad. Sci. USA 1993, 90, 6444–6448. [Google Scholar] [CrossRef] [PubMed]
  4. May, C.; Sapra, P.; Gerber, H.P. Advances in bispecific biotherapeutics for the treatment of cancer. Biochem. Pharmacol. 2012, 84, 1105–1112. [Google Scholar] [CrossRef] [PubMed]
  5. Vaughan, C.K.; Sollazzo, M. Of minibody, camel and bacteriophage. Comb. Chem. High Throughput Screen. 2001, 4, 417–430. [Google Scholar] [CrossRef] [PubMed]
  6. Olsnes, S.; Sandvig, K.; Petersen, O.W.; van Deurs, B. Immunotoxins—Entry into cells and mechanisms of action. Immunol. Today 1989, 10, 291–295. [Google Scholar] [PubMed]
  7. Pirker, R.; FitzGerald, D.J.; Hamilton, T.C.; Ozols, R.F.; Laird, W.; Frankel, A.E.; Willingham, M.C.; Pastan, I. Characterization of immunotoxins active against ovarian cancer cell lines. J. Clin. Investig. 1985, 76, 1261–1267. [Google Scholar] [CrossRef] [PubMed]
  8. Ravel, S.; Colombatti, M.; Casellas, P. Internalization and intracellular fate of anti-CD5 monoclonal antibody and anti-CD5 ricin A-chain immunotoxin in human leukemic T cells. Blood 1992, 79, 1511–1517. [Google Scholar] [PubMed]
  9. Fuchs, H.; Bachran, C.; Flavell, D.J. Diving through membranes: molecular cunning to enforce the endosomal escape of antibody-targeted anti-tumor toxins. Antibodies 2013, 2, 209–235. [Google Scholar] [CrossRef]
  10. Selbo, P.K.; Bostad, M.; Olsen, C.E.; Edwards, V.T.; Hogset, A.; Weyergang, A.; Berg, K. Photochemical internalisation, a minimally invasive strategy for light-controlled endosomal escape of cancer stem cell-targeting therapeutics. Photochem. Photobiol. Sci. 2015, 14, 1433–1450. [Google Scholar] [CrossRef] [PubMed]
  11. Wales, R.; Roberts, L.M.; Lord, J.M. Addition of an endoplasmic reticulum retrieval sequence to ricin A chain significantly increases its cytotoxicity to mammalian cells. J. Biol. Chem. 1993, 268, 23986–23990. [Google Scholar] [PubMed]
  12. Zhang, D.; Wang, J.; Xu, D. Cell-penetrating peptides as noninvasive transmembrane vectors for the development of novel multifunctional drug-delivery systems. J. Control. Release 2016, 229, 130–139. [Google Scholar] [CrossRef] [PubMed]
  13. Wu, M. Enhancement of immunotoxin activity using chemical and biological reagents. Br. J. Cancer 1997, 75, 1347–1355. [Google Scholar] [CrossRef] [PubMed]
  14. Seglen, P.O.; Grinde, B.; Solheim, A.E. Inhibition of the lysosomal pathway of protein degradation in isolated rat hepatocytes by ammonia, methylamine, chloroquine and leupeptin. Eur. J. Biochem. 1979, 95, 215–225. [Google Scholar] [CrossRef] [PubMed]
  15. Casellas, P.; Brown, J.P.; Gros, O.; Gros, P.; Hellstrom, I.; Jansen, F.K.; Poncelet, P.; Roncucci, R.; Vidal, H.; Hellstrom, K.E. Human melanoma cells can be killed in vitro by an immunotoxin specific for melanoma-associated antigen p97. Int. J. Cancer 1982, 30, 437–443. [Google Scholar] [CrossRef] [PubMed]
  16. Casellas, P.; Bourrie, B.J.; Gros, P.; Jansen, F.K. Kinetics of cytotoxicity induced by immunotoxins. Enhancement by lysosomotropic amines and carboxylic ionophores. J. Biol. Chem. 1984, 259, 9359–9364. [Google Scholar] [PubMed]
  17. Zhang, Y.; Schulte, W.; Pink, D.; Phipps, K.; Zijlstra, A.; Lewis, J.D.; Waisman, D.M. Sensitivity of cancer cells to truncated diphtheria toxin. PLoS ONE 2010, 5, e10498. [Google Scholar] [CrossRef] [PubMed]
  18. Umata, T.; Moriyama, Y.; Futai, M.; Mekada, E. The cytotoxic action of diphtheria toxin and its degradation in intact Vero cells are inhibited by bafilomycin A1, a specific inhibitor of vacuolar-type H(+)-ATPase. J. Biol. Chem. 1990, 265, 21940–21945. [Google Scholar] [PubMed]
  19. Marcil, J.; Ravindranath, N.; Sairam, M.R. Cytotoxic activity of lutropin-gelonin conjugate in mouse Leydig tumor cells: potentiation of the hormonotoxin activity by different drugs. Mol. Cell. Endocrinol. 1993, 92, 83–90. [Google Scholar] [CrossRef]
  20. Ramakrishnan, S.; Houston, L.L. Inhibition of human acute lymphoblastic leukemia cells by immunotoxins: Potentiation by chloroquine. Science 1984, 223, 58–61. [Google Scholar] [CrossRef] [PubMed]
  21. Lizzi, A.R.; D’Alessandro, A.M.; Zeolla, N.; Brisdelli, F.; D’Andrea, G.; Pitari, G.; Oratore, A.; Bozzi, A.; Ippoliti, R. The effect of AZT and chloroquine on the activities of ricin and a saporin-transferrin chimeric toxin. Biochem. Pharmacol. 2005, 70, 560–569. [Google Scholar] [CrossRef] [PubMed]
  22. Siena, S.; Villa, S.; Bregni, M.; Bonnadonna, G.; Gianni, A.M. Amantadine potentiates T lymphocyte killing by an anti-pan-T cell (CD5) ricin A-chain immunotoxin. Blood 1987, 69, 345–348. [Google Scholar] [PubMed]
  23. Geden, S.E.; Gardner, R.A.; Fabbrini, M.S.; Ohashi, M.; Phanstiel Iv, O.; Teter, K. Lipopolyamine treatment increases the efficacy of intoxication with saporin and an anticancer saporin conjugate. FEBS J. 2007, 274, 4825–4836. [Google Scholar] [CrossRef] [PubMed]
  24. Akiyama, S.; Seth, P.; Pirker, R.; FitzGerald, D.; Gottesman, M.M.; Pastan, I. Potentiation of cytotoxic activity of immunotoxins on cultured human cells. Cancer Res. 1985, 45, 1005–1007. [Google Scholar] [PubMed]
  25. Siena, S.; Lappi, D.A.; Bregni, M.; Formosa, A.; Villa, S.; Soria, M.; Bonadonna, G.; Gianni, A.M. Synthesis and characterization of an antihuman T-lymphocyte saporin immunotoxin (OKT1-SAP) with in vivo stability into nonhuman primates. Blood 1988, 72, 756–765. [Google Scholar] [PubMed]
  26. Siena, S.; Bregni, M.; Formosa, A.; Martineau, D.; Lappi, D.A.; Bonadonna, G.; Gianni, A.M. Evaluation of antihuman T lymphocyte saporin immunotoxins potentially useful in human transplantation. Transplantation 1988, 46, 747–753. [Google Scholar] [CrossRef] [PubMed]
  27. Myers, C.D.; Thorpe, P.E.; Ross, W.C.; Cumber, A.J.; Katz, F.E.; Tax, W.; Greaves, M.F. An immunotoxin with therapeutic potential in T cell leukemia: WT1-ricin A. Blood 1984, 63, 1178–1185. [Google Scholar] [PubMed]
  28. Kronke, M.; Schlick, E.; Waldmann, T.A.; Vitetta, E.S.; Greene, W.C. Selective killing of human T-lymphotropic virus-I infected leukemic T-cells by monoclonal anti-interleukin 2 receptor antibody-ricin A chain conjugates: Potentiation by ammonium chloride and monensin. Cancer Res. 1986, 46, 3295–3298. [Google Scholar] [PubMed]
  29. Faguet, G.B.; Agee, J.F. Four ricin chain A-based immunotoxins directed against the common chronic lymphocytic leukemia antigen: In vitro characterization. Blood 1993, 82, 536–543. [Google Scholar] [PubMed]
  30. Vollmar, A.M.; Banker, D.E.; Mendelsohn, J.; Herschman, H.R. Toxicity of ligand and antibody-directed ricin A-chain conjugates recognizing the epidermal growth factor receptor. J. Cell. Physiol. 1987, 131, 418–425. [Google Scholar] [CrossRef] [PubMed]
  31. Van Oosterhout, Y.V.; Preijers, F.W.; Wessels, H.M.; de Witte, T. Cytotoxicity of CD3-ricin A chain immunotoxins in relation to cellular uptake and degradation kinetics. Cancer Res. 1992, 52, 5921–5925. [Google Scholar] [PubMed]
  32. Mujoo, K.; Reisfeld, R.A.; Cheung, L.; Rosenblum, M.G. A potent and specific immunotoxin for tumor cells expressing disialoganglioside GD2. Cancer Immunol. Immunother. 1991, 34, 198–204. [Google Scholar] [CrossRef] [PubMed]
  33. Uckun, F.M.; Ramakrishnan, S.; Houston, L.L. Immunotoxin-mediated elimination of clonogenic tumor cells in the presence of human bone marrow. J. Immunol. 1985, 134, 2010–2016. [Google Scholar] [PubMed]
  34. Niesen, J.; Hehmann-Titt, G.; Woitok, M.; Fendel, R.; Barth, S.; Fischer, R.; Stein, C. A novel fully-human cytolytic fusion protein based on granzyme B shows in vitro cytotoxicity and ex vivo binding to solid tumors overexpressing the epidermal growth factor receptor. Cancer Lett. 2016, 374, 229–240. [Google Scholar] [CrossRef] [PubMed]
  35. Cox, B.; Firman, P.; Schneider, H. Sodium ion-proton exchange reactions of the carboxylic acid ionophore monensin. J. Am. Chem. Soc. 1985, 107, 4297–4300. [Google Scholar] [CrossRef]
  36. Grinde, B. Effect of carboxylic ionophores on lysosomal protein degradation in rat hepatocytes. Exp. Cell Res. 1983, 149, 27–35. [Google Scholar] [CrossRef]
  37. Derbyshire, E.J.; Stahel, R.A.; Wawrzynczak, E.J. Potentiation of a weakly active ricin A chain immunotoxin recognizing the neural cell adhesion molecule. Clin. Exp. Immunol. 1992, 89, 336–340. [Google Scholar] [CrossRef] [PubMed]
  38. Ramakrishnan, S.; Bjorn, M.J.; Houston, L.L. Recombinant ricin A chain conjugated to monoclonal antibodies: Improved tumor cell inhibition in the presence of lysosomotropic compounds. Cancer Res. 1989, 49, 613–617. [Google Scholar] [PubMed]
  39. Colombatti, M.; Dell’Arciprete, L.; Chignola, R.; Tridente, G. Carrier protein-monensin conjugates: Enhancement of immunotoxin cytotoxicity and potential in tumor treatment. Cancer Res. 1990, 50, 1385–1391. [Google Scholar] [PubMed]
  40. Casellas, P.; Jansen, F.K. Immunotoxin enhancers. Cancer Treat. Res. 1988, 37, 351–369. [Google Scholar] [PubMed]
  41. Hertler, A.A.; Schlossman, D.M.; Borowitz, M.J.; Blythman, H.E.; Casellas, P.; Frankel, A.E. An anti-CD5 immunotoxin for chronic lymphocytic leukemia: Enhancement of cytotoxicity with human serum albumin-monensin. Int. J. Cancer 1989, 43, 215–219. [Google Scholar] [CrossRef] [PubMed]
  42. Griffin, T.; Raso, V. Monensin in lipid emulsion for the potentiation of ricin A chain immunotoxins. Cancer Res. 1991, 51, 4316–4322. [Google Scholar] [PubMed]
  43. Griffin, T.; Rybak, M.E.; Recht, L.; Singh, M.; Salimi, A.; Raso, V. Potentiation of antitumor immunotoxins by liposomal monensin. J. Natl. Cancer Inst. 1993, 85, 292–298. [Google Scholar] [CrossRef] [PubMed]
  44. Ferdous, A.J.; Stembridge, N.Y.; Singh, M. Role of monensin PLGA polymer nanoparticles and liposomes as potentiator of ricin A immunotoxins in vitro. J. Control. Release 1998, 50, 71–78. [Google Scholar] [CrossRef]
  45. Raso, V.; Lawrence, J. Carboxylic ionophores enhance the cytotoxic potency of ligand- and antibody-delivered ricin A chain. J. Exp. Med. 1984, 160, 1234–1240. [Google Scholar] [CrossRef] [PubMed]
  46. Griffin, T.W.; Childs, L.R.; FitzGerald, D.J.; Levin, L.V. Enhancement of the cytotoxic effect of anti-carcinoembryonic antigen immunotoxins by adenovirus and carboxylic ionophores. J. Natl. Cancer Inst. 1987, 79, 679–685. [Google Scholar] [PubMed]
  47. Griffin, T.W.; Pagnini, P.G.; Houston, L.L. Enhancement of the specific cytotoxicity of a breast cancer-associated antigen immunotoxin by the carboxylic ionophore monensin. J. Biol. Response Mod. 1987, 6, 537–545. [Google Scholar] [PubMed]
  48. Roth, J.A.; Ames, R.S.; Fry, K.; Lee, H.M.; Scannon, P.J. Mediation of reduction of spontaneous and experimental pulmonary metastases by ricin A-chain immunotoxin 45-2D9-RTA with potentiation by systemic monensin in mice. Cancer Res. 1988, 48, 3496–3501. [Google Scholar] [PubMed]
  49. Derbyshire, E.J.; Wawrzynczak, E.J. An anti-mucin immunotoxin BrE-3-ricin A-chain is potently and selectively toxic to human small-cell lung cancer. Int. J. Cancer 1992, 52, 624–630. [Google Scholar] [CrossRef] [PubMed]
  50. Handa, J.T.; Houston, L.L.; Jaffe, G.J. Monensin enhances the cytotoxic effect of antitransferrin receptor immunotoxin on cultured RPE cells. Curr. Eye Res. 1993, 12, 45–53. [Google Scholar] [CrossRef] [PubMed]
  51. Akiyama, S.; Gottesman, M.M.; Hanover, J.A.; Fitzgerald, D.J.; Willingham, M.C.; Pastan, I. Verapamil enhances the toxicity of conjugates of epidermal growth factor with Pseudomonas exotoxin and antitransferrin receptor with Pseudomonas exotoxin. J. Cell. Physiol. 1984, 120, 271–279. [Google Scholar] [CrossRef] [PubMed]
  52. Pirker, R.; FitzGerald, D.J.; Willingham, M.C.; Pastan, I. Enhancement of the activity of immunotoxins made with either ricin A chain or Pseudomonas exotoxin in human ovarian and epidermoid carcinoma cell lines. Cancer Res. 1988, 48, 3919–3923. [Google Scholar] [PubMed]
  53. Pirker, R.; FitzGerald, D.J.; Raschack, M.; Frank, Z.; Willingham, M.C.; Pastan, I. Enhancement of the activity of immunotoxins by analogues of verapamil. Cancer Res. 1989, 49, 4791–4795. [Google Scholar] [PubMed]
  54. Jaffrezou, J.P.; Levade, T.; Thurneyssen, O.; Chiron, M.; Bordier, C.; Attal, M.; Chatelain, P.; Laurent, G. In vitro and in vivo enhancement of ricin-A chain immunotoxin activity by novel indolizine calcium channel blockers: delayed intracellular degradation linked to lipidosis induction. Cancer Res. 1992, 52, 1352–1359. [Google Scholar] [PubMed]
  55. Jaffrezou, J.P.; Levade, T.; Kuhlein, E.; Thurneyssen, O.; Chiron, M.; Grandjean, H.; Carriere, D.; Laurent, G. Enhancement of ricin A chain immunotoxin activity by perhexiline on established and fresh leukemic cells. Cancer Res. 1990, 50, 5558–5566. [Google Scholar] [PubMed]
  56. Wu, Y.N.; Gadina, M.; Tao-Cheng, J.H.; Youle, R.J. Retinoic acid disrupts the Golgi apparatus and increases the cytosolic routing of specific protein toxins. J. Cell Biol. 1994, 125, 743–753. [Google Scholar] [CrossRef] [PubMed]
  57. Hudson, T.H.; Grillo, F.G. Brefeldin-A enhancement of ricin A-chain immunotoxins and blockade of intact ricin, modeccin, and abrin. J. Biol. Chem. 1991, 266, 18586–18592. [Google Scholar] [PubMed]
  58. Andersson, Y.; Engebraaten, O.; Fodstad, O. Synergistic anti-cancer effects of immunotoxin and cyclosporin in vitro and in vivo. Br. J. Cancer 2009, 101, 1307–1315. [Google Scholar] [CrossRef] [PubMed]
  59. Davol, P.A.; Bizuneh, A.; Frackelton, A.R., Jr. Wortmannin, a phosphoinositide 3-kinase inhibitor, selectively enhances cytotoxicity of receptor-directed-toxin chimeras in vitro and in vivo. Anticancer Res. 1999, 19, 1705–1713. [Google Scholar] [PubMed]
  60. Park, T.G.; Jeong, J.H.; Kim, S.W. Current status of polymeric gene delivery systems. Adv. Drug Deliv. Rev. 2006, 58, 467–486. [Google Scholar] [CrossRef] [PubMed]
  61. Lai, P.S.; Pai, C.L.; Peng, C.L.; Shieh, M.J.; Berg, K.; Lou, P.J. Enhanced cytotoxicity of saporin by polyamidoamine dendrimer conjugation and photochemical internalization. J. Biomed. Mater. Res. Part A 2008, 87, 147–155. [Google Scholar] [CrossRef] [PubMed]
  62. Pattrick, N.G.; Richardson, S.C.; Casolaro, M.; Ferruti, P.; Duncan, R. Poly(amidoamine)-mediated intracytoplasmic delivery of ricin A-chain and gelonin. J. Control. Release 2001, 77, 225–232. [Google Scholar] [CrossRef]
  63. Yefenof, E.; Abboud, G.; Epszteyn, S.; Vitetta, E.S. Treatment of premalignancy: Prevention of lymphoma in radiation leukemia virus-inoculated mice by cyclosporin A and immunotoxin. Proc. Natl. Acad. Sci. USA 1992, 89, 728–732. [Google Scholar] [CrossRef] [PubMed]
  64. Jaffrezou, J.P.; Sikic, B.I.; Laurent, G. Cyclosporin A and cyclosporin SDZ PSC 833 enhance anti-CD5 ricin A-chain immunotoxins in human leukemic T cells. Blood 1994, 83, 482–489. [Google Scholar] [PubMed]
  65. Greber, U.F. Virus and host mechanics support membrane penetration and cell entry. J. Virol. 2016, 90, 3802–3805. [Google Scholar] [CrossRef] [PubMed]
  66. White, J.M.; Whittaker, G.R. Fusion of Enveloped Viruses in Endosomes. Traffic 2016, 17, 593–614. [Google Scholar] [CrossRef] [PubMed]
  67. Copolovici, D.M.; Langel, K.; Eriste, E.; Langel, U. Cell-penetrating peptides: Design, synthesis, and applications. ACS Nano 2014, 8, 1972–1994. [Google Scholar] [CrossRef] [PubMed]
  68. Vives, E.; Schmidt, J.; Pelegrin, A. Cell-penetrating and cell-targeting peptides in drug delivery. Biochim. Biophys. Acta 2008, 1786, 126–138. [Google Scholar] [CrossRef] [PubMed]
  69. Frankel, A.D.; Pabo, C.O. Cellular uptake of the tat protein from human immunodeficiency virus. Cell 1988, 55, 1189–1193. [Google Scholar] [CrossRef]
  70. Reissmann, S. Cell penetration: Scope and limitations by the application of cell-penetrating peptides. J. Pept. Sci. 2014, 20, 760–784. [Google Scholar] [CrossRef] [PubMed]
  71. Foged, C.; Nielsen, H.M. Cell-penetrating peptides for drug delivery across membrane barriers. Expert Opin. Drug Deliv. 2008, 5, 105–117. [Google Scholar] [CrossRef] [PubMed]
  72. Kristensen, M.; Birch, D.; Nielsen, H.M. Applications and Challenges for Use of Cell-Penetrating Peptides as Delivery Vectors for Peptide and Protein Cargos. Int. J. Mol. Sci. 2016, 17, 185. [Google Scholar] [CrossRef] [PubMed]
  73. Salerno, J.C.; Ngwa, V.M.; Nowak, S.J.; Chrestensen, C.A.; Healey, A.N.; McMurry, J.L. Novel cell-penetrating peptide-adaptors effect intracellular delivery and endosomal escape of protein cargos. J. Cell Sci. 2016, 129, 893–897. [Google Scholar] [CrossRef] [PubMed]
  74. Fuchs, H.; Bachran, C.; Li, T.; Heisler, I.; Durkop, H.; Sutherland, M. A cleavable molecular adapter reduces side effects and concomitantly enhances efficacy in tumor treatment by targeted toxins in mice. J. Control. Release 2007, 117, 342–350. [Google Scholar] [CrossRef] [PubMed]
  75. Shin, M.C.; Zhang, J.; Min, K.A.; He, H.; David, A.E.; Huang, Y.; Yang, V.C. PTD-Modified ATTEMPTS for Enhanced Toxin-based Cancer Therapy: An in vivo Proof-of-Concept Study. Pharm. Res. 2015, 32, 2690–2703. [Google Scholar] [CrossRef] [PubMed]
  76. Shin, M.C.; Zhang, J.; Min, K.A.; Lee, K.; Moon, C.; Balthasar, J.P.; Yang, V.C. Combination of antibody targeting and PTD-mediated intracellular toxin delivery for colorectal cancer therapy. J. Control. Release 2014, 194, 197–210. [Google Scholar] [CrossRef] [PubMed]
  77. Shin, M.C.; Zhao, J.; Zhang, J.; Huang, Y.; He, H.; Wang, M.; Min, K.A.; Yang, V.C. Recombinant TAT-gelonin fusion toxin: Synthesis and characterization of heparin/protamine-regulated cell transduction. J. Biomed. Mater. Res. Part A 2015, 103, 409–419. [Google Scholar] [CrossRef] [PubMed]
  78. Fitzgerald, D.J.P.; Padmanabhan, R.; Pastan, I.; Willingham, M.C. Adenovirus-induced release of epidermal growth factor and pseudomonas toxin into the cytosol of KB cells during receptor-mediated endocytosis. Cell 1983, 32, 607–617. [Google Scholar] [CrossRef]
  79. Seth, P.; Fitzgerald, D.; Ginsberg, H.; Willingham, M.; Pastan, I. Evidence that the penton base of adenovirus is involved in potentiation of toxicity of Pseudomonas exotoxin conjugated to epidermal growth factor. Mol. Cell. Biol. 1984, 4, 1528–1533. [Google Scholar] [CrossRef] [PubMed]
  80. Lorenzetti, I.; Meneguzzi, A.; Fracasso, G.; Potrich, C.; Costantini, L.; Chiesa, E.; Legname, G.; Menestrina, G.; Tridente, G.; Colombatti, M. Genetic grafting of membrane-acting peptides to the cytotoxin dianthin augments its ability to de-stabilize lipid bilayers and enhances its cytotoxic potential as the component of transferrin-toxin conjugates. Int. J. Cancer 2000, 86, 582–589. [Google Scholar] [CrossRef]
  81. Chignola, R.; Anselmi, C.; Serra, M.D.; Franceschi, A.; Fracasso, G.; Pasti, M.; Chiesa, E.; Lord, J.M.; Tridente, G.; Colombatti, M. Self-potentiation of Ligand-Toxin Conjugates Containing Ricin A Chain Fused with Viral Structures. J. Biol. Chem. 1995, 270, 23345–23351. [Google Scholar] [CrossRef] [PubMed]
  82. Tolstikov, V.V.; Cole, R.; Fang, H.; Pincus, S.H. Influence of endosome-destabilizing peptides on efficacy of anti-HIV immunotoxins. Bioconjug. Chem. 1997, 8, 38–43. [Google Scholar] [CrossRef] [PubMed]
  83. Heisler, I.; Keller, J.; Tauber, R.; Sutherland, M.; Fuchs, H. A cleavable adapter to reduce nonspecific cytotoxicity of recombinant immunotoxins. Int. J. Cancer 2003, 103, 277–282. [Google Scholar] [CrossRef] [PubMed]
  84. Hetzel, C.; Bachran, C.; Fischer, R.; Fuchs, H.; Barth, S.; Stöcker, M. Small cleavable adapters enhance the specific cytotoxicity of a humanized immunotoxin directed against CD64-positive cells. J. Immunother. 2008, 31, 370–376. [Google Scholar] [CrossRef] [PubMed]
  85. Weldon, J.E.; Skarzynski, M.; Therres, J.A.; Ostovitz, J.R.; Zhou, H.; Kreitman, R.J.; Pastan, I. Designing the furin-cleavable linker in recombinant immunotoxins based on Pseudomonas exotoxin A. Bioconjug. Chem. 2015, 26, 1120–1128. [Google Scholar] [CrossRef] [PubMed]
  86. Hwang, J.; Fitzgerald, D.J.; Adhya, S.; Pastan, I. Functional domains of Pseudomonas exotoxin identified by deletion analysis of the gene expressed in E. coli. Cell 1987, 48, 129–136. [Google Scholar] [CrossRef]
  87. Michalska, M.; Wolf, P. Pseudomonas Exotoxin A: Optimized by evolution for effective killing. Front. Microbiol. 2015, 6, 963. [Google Scholar] [CrossRef] [PubMed]
  88. Wolf, P.; Elsasser-Beile, U. Pseudomonas exotoxin A: From virulence factor to anti-cancer agent. Int. J. Med. Microbiol. IJMM 2009, 299, 161–176. [Google Scholar] [CrossRef] [PubMed]
  89. Bachran, C.; Heisler, I.; Fuchs, H.; Sutherland, M. Influence of protein transduction domains on target-specific chimeric proteins. Biochem. Biophys. Res. Commun. 2005, 337, 602–609. [Google Scholar] [CrossRef] [PubMed]
  90. Bachanova, V.; Frankel, A.E.; Cao, Q.; Lewis, D.; Grzywacz, B.; Verneris, M.R.; Ustun, C.; Lazaryan, A.; McClune, B.; Warlick, E.D.; et al. Phase I study of a bispecific ligand-directed toxin targeting CD22 and CD19 (DT2219) for refractory B-cell malignancies. Clin. Cancer Res. 2015, 21, 1267–1272. [Google Scholar] [CrossRef] [PubMed]
  91. Bachran, C.; Abdelazim, S.; Fattah, R.J.; Liu, S.; Leppla, S.H. Recombinant expression and purification of a tumor-targeted toxin in Bacillus anthracis. Biochem. Biophys. Res. Commun. 2013, 430, 150–155. [Google Scholar] [CrossRef] [PubMed]
  92. Kreitman, R.J.; Arons, E.; Stetler-Stevenson, M.; Fitzgerald, D.J.; Wilson, W.H.; Pastan, I. Recombinant immunotoxins and other therapies for relapsed/refractory hairy cell leukemia. Leuk. Lymphoma 2011, 52 (Suppl. 2), 82–86. [Google Scholar] [CrossRef] [PubMed]
  93. Kreitman, R.J.; Pastan, I. Immunoconjugates in the management of hairy cell leukemia. Best Pract. Res. Clin. Haematol. 2015, 28, 236–245. [Google Scholar] [CrossRef] [PubMed]
  94. Li, Y.M.; Vallera, D.A.; Hall, W.A. Diphtheria toxin-based targeted toxin therapy for brain tumors. J. Neuro Oncol. 2013, 114, 155–164. [Google Scholar] [CrossRef] [PubMed]
  95. Madhumathi, J.; Devilakshmi, S.; Sridevi, S.; Verma, R.S. Immunotoxin therapy for hematologic malignancies: Where are we heading? Drug Discov. Today 2016, 21, 325–332. [Google Scholar] [CrossRef] [PubMed]
  96. Mazor, R.; Onda, M.; Pastan, I. Immunogenicity of therapeutic recombinant immunotoxins. Immunol. Rev. 2016, 270, 152–164. [Google Scholar] [CrossRef] [PubMed]
  97. Meng, J.; Liu, Y.; Gao, S.; Lin, S.; Gu, X.; Pomper, M.G.; Wang, P.C.; Shan, L. A bivalent recombinant immunotoxin with high potency against tumors with EGFR and EGFRvIII expression. Cancer Biol. Ther. 2015, 16, 1764–1774. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, Z.; Pratts, S.G.; Zhang, H.; Spencer, P.J.; Yu, R.; Tonsho, M.; Shah, J.A.; Tanabe, T.; Powell, H.R.; Huang, C.A.; et al. Treg depletion in non-human primates using a novel diphtheria toxin-based anti-human CCR4 immunotoxin. Mol. Oncol. 2015. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, Z.; Wei, M.; Zhang, H.; Chen, H.; Germana, S.; Huang, C.A.; Madsen, J.C.; Sachs, D.H.; Wang, Z. Diphtheria-toxin based anti-human CCR4 immunotoxin for targeting human CCR4+ cells in vivo. Mol. Oncol. 2015, 9, 1458–1470. [Google Scholar] [CrossRef] [PubMed]
  100. Weidle, U.H.; Tiefenthaler, G.; Schiller, C.; Weiss, E.H.; Georges, G.; Brinkmann, U. Prospects of bacterial and plant protein-based immunotoxins for treatment of cancer. Cancer Genom. Proteom. 2014, 11, 25–38. [Google Scholar]
  101. Weng, A.; Thakur, M.; Beceren-Braun, F.; Bachran, D.; Bachran, C.; Riese, S.B.; Jenett-Siems, K.; Gilabert-Oriol, R.; Melzig, M.F.; Fuchs, H. The toxin component of targeted anti-tumor toxins determines their efficacy increase by saponins. Mol. Oncol. 2012, 6, 323–332. [Google Scholar] [CrossRef] [PubMed]
  102. Piascik, P. FDA approves fusion protein for treatment of lymphoma. J. Am. Pharm. Assoc. 1999, 39, 571–572. [Google Scholar] [CrossRef]
  103. Ratts, R.; Trujillo, C.; Bharti, A.; vanderSpek, J.; Harrison, R.; Murphy, J.R. A conserved motif in transmembrane helix 1 of diphtheria toxin mediates catalytic domain delivery to the cytosol. Proc. Natl. Acad. Sci. USA 2005, 102, 15635–15640. [Google Scholar] [CrossRef] [PubMed]
  104. Sun, J.; Jacquez, P. Roles of Anthrax Toxin Receptor 2 in Anthrax Toxin Membrane Insertion and Pore Formation. Toxins 2016, 8, 34. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, S.; Aaronson, H.; Mitola, D.J.; Leppla, S.H.; Bugge, T.H. Potent antitumor activity of a urokinase-activated engineered anthrax toxin. Proc. Natl. Acad. Sci. USA 2003, 100, 657–662. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, S.; Netzel-Arnett, S.; Birkedal-Hansen, H.; Leppla, S.H. Tumor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxin. Cancer Res. 2000, 60, 6061–6067. [Google Scholar] [PubMed]
  107. Wising, C.; Molne, L.; Jonsson, I.M.; Ahlman, K.; Lagergard, T. The cytolethal distending toxin of Haemophilus ducreyi aggravates dermal lesions in a rabbit model of chancroid. Microbes Infect. Inst. Pasteur 2005, 7, 867–874. [Google Scholar] [CrossRef] [PubMed]
  108. Bachran, C.; Hasikova, R.; Leysath, C.E.; Sastalla, I.; Zhang, Y.; Fattah, R.J.; Liu, S.; Leppla, S.H. Cytolethal distending toxin B as a cell-killing component of tumor-targeted anthrax toxin fusion proteins. Cell Death Dis. 2014, 5, e1003. [Google Scholar] [CrossRef] [PubMed]
  109. Mozola, C.C.; Caparon, M.G. Dual modes of membrane binding direct pore formation by Streptolysin O. Mol. Microbiol. 2015, 97, 1036–1050. [Google Scholar] [CrossRef] [PubMed]
  110. Verherstraeten, S.; Goossens, E.; Valgaeren, B.; Pardon, B.; Timbermont, L.; Haesebrouck, F.; Ducatelle, R.; Deprez, P.; Wade, K.R.; Tweten, R.; et al. Perfringolysin O: The Underrated Clostridium perfringens Toxin? Toxins 2015, 7, 1702–1721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Seveau, S. Multifaceted activity of listeriolysin O, the cholesterol-dependent cytolysin of Listeria monocytogenes. Sub Cell. Biochem. 2014, 80, 161–195. [Google Scholar]
  112. Pirie, C.M.; Liu, D.V.; Wittrup, K.D. Targeted cytolysins synergistically potentiate cytoplasmic delivery of gelonin immunotoxin. Mol. Cancer Ther. 2013, 12, 1774–1782. [Google Scholar] [CrossRef] [PubMed]
  113. Provoda, C.J.; Stier, E.M.; Lee, K.D. Tumor cell killing enabled by listeriolysin O-liposome-mediated delivery of the protein toxin gelonin. J. Biol. Chem. 2003, 278, 35102–35108. [Google Scholar] [CrossRef] [PubMed]
  114. Derossi, D.; Calvet, S.; Trembleau, A.; Brunissen, A.; Chassaing, G.; Prochiantz, A. Cell Internalization of the Third Helix of the Antennapedia Homeodomain Is Receptor-independent. J. Biol. Chem. 1996, 271, 18188–18193. [Google Scholar] [CrossRef] [PubMed]
  115. Abes, S.; Turner, J.J.; Ivanova, G.D.; Owen, D.; Williams, D.; Arzumanov, A.; Clair, P.; Gait, M.J.; Lebleu, B. Efficient splicing correction by PNA conjugation to an R6-Penetratin delivery peptide. Nucleic Acids Res. 2007, 35, 4495–4502. [Google Scholar] [CrossRef] [PubMed]
  116. Lundberg, P.; El-Andaloussi, S.; Sütlü, T.; Johansson, H.; Langel, Ü. Delivery of short interfering RNA using endosomolytic cell-penetrating peptides. FASEB J. 2007, 21, 2664–2671. [Google Scholar] [CrossRef] [PubMed]
  117. Dupont, E.; Prochiantz, A.; Joliot, A. Penetratin Story: An Overview. Methods Mol. Biol. 2015, 1324, 29–37. [Google Scholar] [PubMed]
  118. Rosado, C.J.; Buckle, A.M.; Law, R.H.; Butcher, R.E.; Kan, W.T.; Bird, C.H.; Ung, K.; Browne, K.A.; Baran, K.; Bashtannyk-Puhalovich, T.A.; et al. A common fold mediates vertebrate defense and bacterial attack. Science 2007, 317, 1548–1551. [Google Scholar] [CrossRef] [PubMed]
  119. Zeng, L.; Wan, L.; Chen, L.; Li, S.; Lu, Y.; Huang, Q.; Wang, L.; Li, Y.; Cheng, J.; Lu, X. Selective depletion of activated T cells by recombinant immunotoxin containing anti-CTLA-4 single-chain fragment of variable antibody and N-terminal fragment of perforin. Transplant. Proc. 2006, 38, 2151–2153. [Google Scholar] [CrossRef] [PubMed]
  120. Sun, Q.; Jiang, S.; Han, B.; Sun, T.; Li, Z.; Zhao, L.; Gao, Q.; Sun, J. Cytotoxic T lymphocyte-dependent tumor growth inhibition by a vascular endothelial growth factor-superantigen conjugate. Biochem. Biophys. Res. Commun. 2012, 427, 711–717. [Google Scholar] [CrossRef] [PubMed]
  121. Kurschus, F.C.; Kleinschmidt, M.; Fellows, E.; Dornmair, K.; Rudolph, R.; Lilie, H.; Jenne, D.E. Killing of target cells by redirected granzyme B in the absence of perforin. FEBS Lett. 2004, 562, 87–92. [Google Scholar] [CrossRef]
  122. Stahnke, B.; Thepen, T.; Stocker, M.; Rosinke, R.; Jost, E.; Fischer, R.; Tur, M.K.; Barth, S. Granzyme B-H22(scFv), a human immunotoxin targeting CD64 in acute myeloid leukemia of monocytic subtypes. Mol. Cancer Ther. 2008, 7, 2924–2932. [Google Scholar] [CrossRef] [PubMed]
  123. Schiffer, S.; Letzian, S.; Jost, E.; Mladenov, R.; Hristodorov, D.; Huhn, M.; Fischer, R.; Barth, S.; Thepen, T. Granzyme M as a novel effector molecule for human cytolytic fusion proteins: CD64-specific cytotoxicity of Gm-H22(scFv) against leukemic cells. Cancer Lett. 2013, 341, 178–185. [Google Scholar] [CrossRef] [PubMed]
  124. Schiffer, S.; Rosinke, R.; Jost, E.; Hehmann-Titt, G.; Huhn, M.; Melmer, G.; Barth, S.; Thepen, T. Targeted ex vivo reduction of CD64-positive monocytes in chronic myelomonocytic leukemia and acute myelomonocytic leukemia using human granzyme B-based cytolytic fusion proteins. Int. J. Cancer 2014, 135, 1497–1508. [Google Scholar] [CrossRef] [PubMed]
  125. Cao, Y.; Mohamedali, K.A.; Marks, J.W.; Cheung, L.H.; Hittelman, W.N.; Rosenblum, M.G. Construction and characterization of novel, completely human serine protease therapeutics targeting Her2/neu. Mol. Cancer Ther. 2013, 12, 979–991. [Google Scholar] [CrossRef] [PubMed]
  126. Spooner, R.A.; Lord, J.M. Ricin trafficking in cells. Toxins 2015, 7, 49–65. [Google Scholar] [CrossRef] [PubMed]
  127. Vitetta, E.S.; Cushley, W.; Uhr, J.W. Synergy of ricin A chain-containing immunotoxins and ricin B chain-containing immunotoxins in in vitro killing of neoplastic human B cells. Proc. Natl. Acad. Sci. USA 1983, 80, 6332–6335. [Google Scholar] [CrossRef]
  128. Vitetta, E.S.; Fulton, R.J.; Uhr, J.W. Cytotoxicity of a cell-reactive immunotoxin containing ricin A chain is potentiated by an anti-immunotoxin containing ricin B chain. J. Exp. Med. 1984, 160, 341–346. [Google Scholar] [CrossRef] [PubMed]
  129. Embleton, M.J.; Charleston, A.; Robins, R.A.; Pimm, M.V.; Baldwin, R.W. Recombinant ricin toxin A chain cytotoxicity against carcinoembryonic antigen expressing tumour cells mediated by a bispecific monoclonal antibody and its potentiation by ricin toxin B chain. Br. J. Cancer 1991, 63, 670–674. [Google Scholar] [CrossRef] [PubMed]
  130. Schiffer, S.; Hansen, H.P.; Hehmann-Titt, G.; Huhn, M.; Fischer, R.; Barth, S.; Thepen, T. Efficacy of an adapted granzyme B-based anti-CD30 cytolytic fusion protein against PI-9-positive classical Hodgkin lymphoma cells in a murine model. Blood Cancer J. 2013, 3, e106. [Google Scholar] [CrossRef] [PubMed]
  131. Fuchs, H.; Bachran, D.; Panjideh, H.; Schellmann, N.; Weng, A.; Melzig, M.F.; Sutherland, M.; Bachran, C. Saponins as tool for improved targeted tumor therapies. Curr. Drug Targets 2009, 10, 140–151. [Google Scholar] [CrossRef] [PubMed]
  132. Hebestreit, P.; Melzig, M.F. Cytotoxic activity of the seeds from Agrostemma githago var. githago. Planta Medica 2003, 69, 921–925. [Google Scholar] [PubMed]
  133. Melzig, M.F.; Hebestreit, P.; Gaidi, G.; Lacaille-Dubois, M.A. Structure-activity-relationship of saponins to enhance toxic effects of agrostin. Planta Medica 2005, 71, 1088–1090. [Google Scholar] [CrossRef] [PubMed]
  134. Weng, A.; Bachran, C.; Fuchs, H.; Krause, E.; Stephanowitz, H.; Melzig, M.F. Enhancement of saporin cytotoxicity by Gypsophila saponins--more than stimulation of endocytosis. Chem. Biol. Interact. 2009, 181, 424–429. [Google Scholar] [CrossRef] [PubMed]
  135. Weng, A.; Jenett-Siems, K.; Gorick, C.; Melzig, M.F. Enhancement of cytotoxicity of ribosome-inactivating-protein type I by saponinum album is not based on stimulation of phagocytosis. J. Pharm. Pharmacol. 2008, 60, 925–930. [Google Scholar] [CrossRef] [PubMed]
  136. Weng, A.; Thakur, M.; von Mallinckrodt, B.; Beceren-Braun, F.; Gilabert-Oriol, R.; Wiesner, B.; Eichhorst, J.; Bottger, S.; Melzig, M.F.; Fuchs, H. Saponins modulate the intracellular trafficking of protein toxins. J. Control. Release 2012, 164, 74–86. [Google Scholar] [CrossRef] [PubMed]
  137. Bachran, D.; Schneider, S.; Bachran, C.; Weng, A.; Melzig, M.F.; Fuchs, H. The endocytic uptake pathways of targeted toxins are influenced by synergistically acting Gypsophila saponins. Mol. Pharm. 2011, 8, 2262–2272. [Google Scholar] [CrossRef] [PubMed]
  138. Holmes, S.E.; Bachran, C.; Fuchs, H.; Weng, A.; Melzig, M.F.; Flavell, S.U.; Flavell, D.J. Triterpenoid saponin augmention of saporin-based immunotoxin cytotoxicity for human leukaemia and lymphoma cells is partially immunospecific and target molecule dependent. Immunopharmacol. Immunotoxicol. 2015, 37, 42–55. [Google Scholar] [CrossRef] [PubMed]
  139. Bachran, D.; Schneider, S.; Bachran, C.; Urban, R.; Weng, A.; Melzig, M.F.; Hoffmann, C.; Kaufmann, A.M.; Fuchs, H. Epidermal growth factor receptor expression affects the efficacy of the combined application of saponin and a targeted toxin on human cervical carcinoma cells. Int. J. Cancer 2010, 127, 1453–1461. [Google Scholar] [CrossRef] [PubMed]
  140. Gilabert-Oriol, R.; Thakur, M.; von Mallinckrodt, B.; Hug, T.; Wiesner, B.; Eichhorst, J.; Melzig, M.F.; Fuchs, H.; Weng, A. Modified trastuzumab and cetuximab mediate efficient toxin delivery while retaining antibody-dependent cell-mediated cytotoxicity in target cells. Mol. Pharm. 2013, 10, 4347–4357. [Google Scholar] [CrossRef] [PubMed]
  141. Heisler, I.; Sutherland, M.; Bachran, C.; Hebestreit, P.; Schnitger, A.; Melzig, M.F.; Fuchs, H. Combined application of saponin and chimeric toxins drastically enhances the targeted cytotoxicity on tumor cells. J. Control. Release 2005, 106, 123–137. [Google Scholar] [CrossRef] [PubMed]
  142. Bachran, C.; Durkop, H.; Sutherland, M.; Bachran, D.; Muller, C.; Weng, A.; Melzig, M.F.; Fuchs, H. Inhibition of tumor growth by targeted toxins in mice is dramatically improved by saponinum album in a synergistic way. J. Immunother. 2009, 32, 713–725. [Google Scholar] [CrossRef] [PubMed]
  143. Thakur, M.; Mergel, K.; Weng, A.; von Mallinckrodt, B.; Gilabert-Oriol, R.; Durkop, H.; Melzig, M.F.; Fuchs, H. Targeted tumor therapy by epidermal growth factor appended toxin and purified saponin: An evaluation of toxicity and therapeutic potential in syngeneic tumor bearing mice. Mol. Oncol. 2013, 7, 475–483. [Google Scholar] [CrossRef] [PubMed]
  144. Von Mallinckrodt, B.; Thakur, M.; Weng, A.; Gilabert-Oriol, R.; Durkop, H.; Brenner, W.; Lukas, M.; Beindorff, N.; Melzig, M.F.; Fuchs, H. Dianthin-EGF is an effective tumor targeted toxin in combination with saponins in a xenograft model for colon carcinoma. Futur. Oncol. 2014, 10, 2161–2175. [Google Scholar] [CrossRef] [PubMed]
  145. Gilabert-Oriol, R.; Weng, A.; Trautner, A.; Weise, C.; Schmid, D.; Bhargava, C.; Niesler, N.; Wookey, P.J.; Fuchs, H.; Thakur, M. Combinatorial approach to increase efficacy of Cetuximab, Panitumumab and Trastuzumab by dianthin conjugation and co-application of SO1861. Biochem. Pharmacol. 2015, 97, 247–255. [Google Scholar] [CrossRef] [PubMed]
  146. El-Sayed, A.; Futaki, S.; Harashima, H. Delivery of macromolecules using arginine-rich cell-penetrating peptides: Ways to overcome endosomal entrapment. AAPS J. 2009, 11, 13–22. [Google Scholar] [CrossRef] [PubMed]
  147. He, D.; Yang, H.; Lin, Q.; Huang, H. Arg9-peptide facilitates the internalization of an anti-CEA immunotoxin and potentiates its specific cytotoxicity to target cells. Int. J. Biochem. Cell Biol. 2005, 37, 192–205. [Google Scholar] [CrossRef] [PubMed]
  148. Omata, D.; Negishi, Y.; Hagiwara, S.; Yamamura, S.; Endo-Takahashi, Y.; Suzuki, R.; Maruyama, K.; Nomizu, M.; Aramaki, Y. Bubble liposomes and ultrasound promoted endosomal escape of TAT-PEG liposomes as gene delivery carriers. Mol. Pharm. 2011, 8, 2416–2423. [Google Scholar] [CrossRef] [PubMed]
  149. Luo, Z.; Cai, K.; Hu, Y.; Li, J.; Ding, X.; Zhang, B.; Xu, D.; Yang, W.; Liu, P. Redox-responsive molecular nanoreservoirs for controlled intracellular anticancer drug delivery based on magnetic nanoparticles. Adv. Mater. 2012, 24, 431–435. [Google Scholar] [CrossRef] [PubMed]
  150. Lukianova-Hleb, E.Y.; Belyanin, A.; Kashinath, S.; Wu, X.; Lapotko, D.O. Plasmonic nanobubble-enhanced endosomal escape processes for selective and guided intracellular delivery of chemotherapy to drug-resistant cancer cells. Biomaterials 2012, 33, 1821–1826. [Google Scholar] [CrossRef] [PubMed]
  151. Weyergang, A.; Berstad, M.E.; Bull-Hansen, B.; Olsen, C.E.; Selbo, P.K.; Berg, K. Photochemical activation of drugs for the treatment of therapy-resistant cancers. Photochem. Photobiol. Sci. 2015, 14, 1465–1475. [Google Scholar] [CrossRef] [PubMed]
  152. Weyergang, A.; Selbo, P.K.; Berstad, M.E.; Bostad, M.; Berg, K. Photochemical internalization of tumor-targeted protein toxins. Lasers Surg. Med. 2011, 43, 721–733. [Google Scholar] [CrossRef] [PubMed]
  153. Norum, O.J.; Selbo, P.K.; Weyergang, A.; Giercksky, K.E.; Berg, K. Photochemical internalization (PCI) in cancer therapy: From bench towards bedside medicine. J. Photochem. Photobiol. B 2009, 96, 83–92. [Google Scholar] [CrossRef] [PubMed]
  154. Berg, K.; Nordstrand, S.; Selbo, P.K.; Tran, D.T.; Angell-Petersen, E.; Hogset, A. Disulfonated tetraphenyl chlorin (TPCS2a), a novel photosensitizer developed for clinical utilization of photochemical internalization. Photochem. Photobiol. Sci. 2011, 10, 1637–1651. [Google Scholar] [CrossRef] [PubMed]
  155. Selbo, P.K.; Weyergang, A.; Hogset, A.; Norum, O.J.; Berstad, M.B.; Vikdal, M.; Berg, K. Photochemical internalization provides time- and space-controlled endolysosomal escape of therapeutic molecules. J. Control. Release 2010, 148, 2–12. [Google Scholar] [CrossRef] [PubMed]
  156. Bostad, M.; Kausberg, M.; Weyergang, A.; Olsen, C.E.; Berg, K.; Hogset, A.; Selbo, P.K. Light-triggered, efficient cytosolic release of IM7-saporin targeting the putative cancer stem cell marker CD44 by photochemical internalization. Mol. Pharm. 2014, 11, 2764–2776. [Google Scholar] [CrossRef] [PubMed]
  157. Stratford, E.W.; Bostad, M.; Castro, R.; Skarpen, E.; Berg, K.; Hogset, A.; Myklebost, O.; Selbo, P.K. Photochemical internalization of CD133-targeting immunotoxins efficiently depletes sarcoma cells with stem-like properties and reduces tumorigenicity. Biochim. Biophys. Acta 2013, 1830, 4235–4243. [Google Scholar] [CrossRef] [PubMed]
  158. Bostad, M.; Olsen, C.E.; Peng, Q.; Berg, K.; Hogset, A.; Selbo, P.K. Light-controlled endosomal escape of the novel CD133-targeting immunotoxin AC133-saporin by photochemical internalization—A minimally invasive cancer stem cell-targeting strategy. J. Control. Release 2015, 206, 37–48. [Google Scholar] [CrossRef] [PubMed]
  159. Selbo, P.K.; Rosenblum, M.G.; Cheung, L.H.; Zhang, W.; Berg, K. Multi-modality therapeutics with potent anti-tumor effects: Photochemical internalization enhances delivery of the fusion toxin scFvMEL/rGel. PLoS ONE 2009, 4, e6691. [Google Scholar] [CrossRef] [PubMed]
  160. Berstad, M.B.; Cheung, L.H.; Berg, K.; Peng, Q.; Fremstedal, A.S.; Patzke, S.; Rosenblum, M.G.; Weyergang, A. Design of an EGFR-targeting toxin for photochemical delivery: In vitro and in vivo selectivity and efficacy. Oncogene 2015, 34, 5582–5592. [Google Scholar] [CrossRef] [PubMed]
  161. Selbo, P.K.; Sivam, G.; Fodstad, O.; Sandvig, K.; Berg, K. Photochemical internalisation increases the cytotoxic effect of the immunotoxin MOC31-gelonin. Int. J. Cancer 2000, 87, 853–859. [Google Scholar] [CrossRef]
  162. Lund, K.; Bostad, M.; Skarpen, E.; Braunagel, M.; Kiprijanov, S.; Krauss, S.; Duncan, A.; Hogset, A.; Selbo, P.K. The novel EpCAM-targeting monoclonal antibody 3-17I linked to saporin is highly cytotoxic after photochemical internalization in breast, pancreas and colon cancer cell lines. MAbs 2014, 6, 1038–1050. [Google Scholar] [CrossRef] [PubMed]
  163. Bull-Hansen, B.; Cao, Y.; Berg, K.; Skarpen, E.; Rosenblum, M.G.; Weyergang, A. Photochemical activation of the recombinant HER2-targeted fusion toxin MH3-B1/rGel; Impact of HER2 expression on treatment outcome. J. Control. Release 2014, 182, 58–66. [Google Scholar] [CrossRef] [PubMed]
  164. Weyergang, A.; Cheung, L.H.; Rosenblum, M.G.; Mohamedali, K.A.; Peng, Q.; Waltenberger, J.; Berg, K. Photochemical internalization augments tumor vascular cytotoxicity and specificity of VEGF(121)/rGel fusion toxin. J. Control. Release 2014, 180, 1–9. [Google Scholar] [CrossRef] [PubMed]
  165. Bull-Hansen, B.; Berstad, M.B.; Berg, K.; Cao, Y.; Skarpen, E.; Fremstedal, A.S.; Rosenblum, M.G.; Peng, Q.; Weyergang, A. Photochemical activation of MH3-B1/rGel: A HER2-targeted treatment approach for ovarian cancer. Oncotarget 2015, 6, 12436–12451. [Google Scholar] [CrossRef] [PubMed]
  166. Berstad, M.B.; Weyergang, A.; Berg, K. Photochemical internalization (PCI) of HER2-targeted toxins: Synergy is dependent on the treatment sequence. Biochim. Biophys. Acta 2012, 1820, 1849–1858. [Google Scholar] [CrossRef] [PubMed]
  167. Yip, W.L.; Weyergang, A.; Berg, K.; Tonnesen, H.H.; Selbo, P.K. Targeted delivery and enhanced cytotoxicity of cetuximab-saporin by photochemical internalization in EGFR-positive cancer cells. Mol. Pharm. 2007, 4, 241–251. [Google Scholar] [CrossRef] [PubMed]
  168. Weyergang, A.; Selbo, P.K.; Berg, K. Photochemically stimulated drug delivery increases the cytotoxicity and specificity of EGF-saporin. J. Control. Release 2006, 111, 165–173. [Google Scholar] [CrossRef] [PubMed]
  169. Gaumann, A.K.; Kiefer, F.; Alfer, J.; Lang, S.A.; Geissler, E.K.; Breier, G. Receptor tyrosine kinase inhibitors: Are they real tumor killers? Int. J. Cancer 2016, 138, 540–554. [Google Scholar] [CrossRef] [PubMed]
  170. Scott, A.M.; Wolchok, J.D.; Old, L.J. Antibody therapy of cancer. Nat. Rev. Cancer 2012, 12, 278–287. [Google Scholar] [CrossRef] [PubMed]
  171. Fuchs, H.; Bachran, C. Targeted tumor therapies at a glance. Curr. Drug Targets 2009, 10, 89–93. [Google Scholar] [CrossRef] [PubMed]
  172. Diamantis, N.; Banerji, U. Antibody-drug conjugates-an emerging class of cancer treatment. Br. J. Cancer 2016, 114, 362–367. [Google Scholar] [CrossRef] [PubMed]
  173. Polakis, P. Antibody Drug Conjugates for Cancer Therapy. Pharmacol. Rev. 2016, 68, 3–19. [Google Scholar] [CrossRef] [PubMed]
  174. Schumacher, D.; Hackenberger, C.P.; Leonhardt, H.; Helma, J. Current Status: Site-Specific Antibody Drug Conjugates. J. Clin. Immunol. 2016, 36 (Suppl. 1), 100–107. [Google Scholar] [CrossRef] [PubMed]
  175. Yamaizumi, M.; Mekada, E.; Uchida, T.; Okada, Y. One molecule of diphtheria toxin fragment A introduced into a cell can kill the cell. Cell 1978, 15, 245–250. [Google Scholar] [CrossRef]
  176. Shapira, A.; Benhar, I. Toxin-based therapeutic approaches. Toxins 2010, 2, 2519–2583. [Google Scholar] [CrossRef] [PubMed]
  177. Karimi, M.; Eslami, M.; Sahandi-Zangabad, P.; Mirab, F.; Farajisafiloo, N.; Shafaei, Z.; Ghosh, D.; Bozorgomid, M.; Dashkhaneh, F.; Hamblin, M.R. pH-Sensitive stimulus-responsive nanocarriers for targeted delivery of therapeutic agents. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2016. [Google Scholar] [CrossRef] [PubMed]
  178. Taghizadeh, B.; Taranejoo, S.; Monemian, S.A.; Salehi Moghaddam, Z.; Daliri, K.; Derakhshankhah, H.; Derakhshani, Z. Classification of stimuli-responsive polymers as anticancer drug delivery systems. Drug Deliv. 2015, 22, 145–155. [Google Scholar] [CrossRef] [PubMed]
  179. Backendorf, C.; Noteborn, M.H. Apoptin towards safe and efficient anticancer therapies. Adv. Exp. Med. Biol. 2014, 818, 39–59. [Google Scholar] [PubMed]
  180. Los, M.; Panigrahi, S.; Rashedi, I.; Mandal, S.; Stetefeld, J.; Essmann, F.; Schulze-Osthoff, K. Apoptin, a tumor-selective killer. Biochim. Biophys. Acta 2009, 1793, 1335–1342. [Google Scholar] [CrossRef] [PubMed]
  181. Schipper, M.L.; Patel, M.R.; Gambhir, S.S. Evaluation of firefly luciferase bioluminescence mediated photodynamic toxicity in cancer cells. Mol. Imaging Biol. 2006, 8, 218–225. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Endosomal escape enhancers for targeted toxins: (A) as native bacterial toxins, targeted toxins also bind to the target receptor and are internalized; (B) membrane-destabilizing agents and photosensitizers typically enter cells by passive diffusion and accumulate preferably in the endosomal membrane; (C) cell-penetrating peptides (CPPs), native bacterial translocation domains and pore-forming proteins directly mediate the entry of the toxins into the cytosol; (D) membrane-destabilizing agents and photosensitizers rather destroy the membrane integrity resulting in leaky endosomes, which allows translocation of the toxins; and (E) the toxins affect vital metabolic pathways in the cytosol finally resulting in apoptosis of the cell.
Figure 1. Endosomal escape enhancers for targeted toxins: (A) as native bacterial toxins, targeted toxins also bind to the target receptor and are internalized; (B) membrane-destabilizing agents and photosensitizers typically enter cells by passive diffusion and accumulate preferably in the endosomal membrane; (C) cell-penetrating peptides (CPPs), native bacterial translocation domains and pore-forming proteins directly mediate the entry of the toxins into the cytosol; (D) membrane-destabilizing agents and photosensitizers rather destroy the membrane integrity resulting in leaky endosomes, which allows translocation of the toxins; and (E) the toxins affect vital metabolic pathways in the cytosol finally resulting in apoptosis of the cell.
Toxins 08 00200 g001
Table 1. Ammonium chloride and lysosomotropic amines that enhance the cytotoxicity of targeted toxins. All listed substances act by increasing the pH in the lysosomes and thus preventing degradation.
Table 1. Ammonium chloride and lysosomotropic amines that enhance the cytotoxicity of targeted toxins. All listed substances act by increasing the pH in the lysosomes and thus preventing degradation.
EnhancerToxinAntigenic TargetTargeted ToxinEnhancer ConcentrationMax. Enhancement FactorRef.
Amantadinericin A chainCD5T101-RTA1 mM1180[16]
saporinCD5OKT1-SAP1 mM4[25]
CD5T101-SAP1 mM4[25]
CD5SOT1a-SAP1 mM4[26]
Ammonium chloridericin A chainCD5T101-RTA10 mM6700[16]
Thy 1.2 (CD 90)AT15E-RTA10 mM6[16]
melanotransferrin96.5-RTA10 mM42[16]
CD7WT1-RTA6 mM100[27]
CD25anti-TAC-RTA10 mM100[28]
cCLLaCLL2m/RTA10 mM80[29]
EGFREGF-RTA10 mM12[30]
CD3WT32-RTA6 mMSignificant increase[31]
ChloroquinegeloninLH receptorlutropin-gelonin9.6–29 µM15[19]
GD2gelonin-14G2a10 µM10[32]
pokeweed antiviral proteintransferrin receptor5E9-11-PAP10–100 µM65[20]
T cellsT3-3A1-PAP10–100 µM65[20]
CD19B43-PAP40 µMSignificant increase[33]
ricin A chainCD5T101-RTA100 µM2500[16]
saporintransferrin receptorsaporin-transferrin10 µMSignificant increase[21]
granzyme BEGFRGb(R201K)-scFv171150 µM3[34]
Dimethylaminericin A chainCD5T101-RTA10 mM3300[16]
Lipopolyaminessaporinurokinase receptoruPA-saporin5 µg/mL83[23]
Methylaminericin A chainCD5T101-RTA10 mM13,300[16]
QuinacrinegeloninLH receptorlutropin-gelonin2.6–7.6 µM15[19]
Trimethylaminericin A chainCD5T101-RTA10 mM80[16]
β-Glycylphenyl-naphthylamide (GPN)Pseudomonas exotoxintransferrin receptorHB21-PE10–20 µg/mL9[24]
EGFREGF-PE10–20 µg/mL6[24]
Table 2. Carboxylic ionophores that enhance the cytosolic uptake of targeted toxins. The compounds in the list presumably take effect by augmenting the pH in the lysosomes and blocking the lysosomal degradation of targeted toxins.
Table 2. Carboxylic ionophores that enhance the cytosolic uptake of targeted toxins. The compounds in the list presumably take effect by augmenting the pH in the lysosomes and blocking the lysosomal degradation of targeted toxins.
EnhancerToxinAntigenic TargetTargeted ToxinEnhancer ConcentrationMax. Enhancement FactorRef.
Grisorixinricin A chainCD5T101-RTA50 nM25,000[16]
Lasalocidricin A chainCD5T101-RTA1 µM33,000[16]
MonensingeloninLH receptorlutropin-gelonin0.3–2.9 µM15[19]
GD2gelonin-14G2a100 nM10[32]
ricin A chainCD5T101-RTA50 nM50,000[16]
Thy 1.2AT15E-RTA50 nM4[16]
p9796.5-RTA50 nM420[16]
transferrin receptorTfn-RTA10–100 nM30,000[45]
CD10anti-CALLA-RTA10–100 nMSignificant increase[45]
CD25anti-TAC-RTA25 nM400[28]
CEAanti-CEA-RTA0.5–1 µMSignificant increase[46]
p55260F9-rRTA100 nM34[47]
gp7445-2D9-RTA0.5 µMSignificant increase[48]
p55260F9-rRTA10–100 nMSignificant increase[38]
CD73A1-rRTA10–100 nMSignificant increase[38]
transferrin receptorR17-217-rRTA10–100 nMSignificant increase[38]
transferrin receptorTfn-RTA900 nM500[39]
transferrin receptorOKT9-RTA900 nM3300[39]
transferrin receptorOX26-RTA900 nM330[39]
N-CAMSEN36-RTA100 nM12,000[37]
MUC1BrE-3-RTA100 nM100[49]
transferrin receptor454A12MAb-RTA10–100 nM4[50]
saporintransferrin receptorTfn-So6900 nM1250[39]
Nigericinricin A chainCD5T101-RTA10 nM6700[16]
transferrin receptorTfn-RTA10–100 nMSignificant increase[45]
CD10anti-CALLA-RTA10–100 nMSignificant increase[45]
CEAanti-CEA-RTA0.5 µMSignificant increase[46]
Table 3. Calcium channel antagonists enhancing the cytotoxicity of targeted toxins. These substances probably cause their effect by blocking the lysosomal degradation of targeted toxins and modulating their intracellular trafficking.
Table 3. Calcium channel antagonists enhancing the cytotoxicity of targeted toxins. These substances probably cause their effect by blocking the lysosomal degradation of targeted toxins and modulating their intracellular trafficking.
EnhancerToxinAntigenic TargetTargeted ToxinEnhancer ConcentrationMax. Enhancement FactorRef.
DiltiazemPseudomonas exotoxintransferrin receptorHB21-PE10–20 µg/mL6[24]
EGFREGF-PE10–20 µg/mL8[24]
Indolizine (SR 33287; SR33557)ricin A chainCD5T101-RTA5 µM620[54]
Thy 1.2AT15E-RTA5 µM84[54]
Methoxyverapamil (D-600)Pseudomonas exotoxintransferrin receptorHB21-PE5–20 µg/mL12[24]
EGFREGF-PE5–20 µg/mL20[24]
Perhexilinericin A chainCD5T101-RTA1–5 µM2000[55]
HLA-DR class IIHNC-241-RTA1–10 µM100[55]
VerapamilgeloninLH receptorlutropin-gelonin10–41 µM15[19]
Pseudomonas exotoxintransferrin receptorHB21-PE 2.5–20 µg/mL11[24]
EGFREGF-PE10–20 µg/mL40[24]
ricin A chaintransferrin receptor454A12-rRTA20 µg/mL25[52]
p55260F9-rRTA20 µg/mL8[52]
HER2454C11-RTA20 µg/mLSignificant increase[52]
cCLLaCLL2m/RTA20 µg/mL80[29]
Verapamil analogs (D792; D595; D528; Sz45)Pseudomonas exotoxintransferrin receptorHB21-PE20 µM35[53]
ricin A chaintransferrin receptor454A12-rRTA1–20 µg/mL67[53]
p55260F9-rRTA20 µMSignificant increase[53]
Table 4. Other organic compounds that enhance the cytotoxicity of targeted toxins. Listed substances induce their effect on different intracellular vesicular compartments. For some of the compounds, the exact mechanism of action has not been completely elucidated yet.
Table 4. Other organic compounds that enhance the cytotoxicity of targeted toxins. Listed substances induce their effect on different intracellular vesicular compartments. For some of the compounds, the exact mechanism of action has not been completely elucidated yet.
EnhancerToxinAntigenic TargetTargeted ToxinEnhancer ConcentrationMax. Enhancement FactorRef.
Brefeldin-Aricin A chaintransferrin receptor454A12-rRTA0.05–0.5 µg/mLSignificant increase[57]
p55260F9-rRTA0.025–0.05 µg/mLSignificant increase[57]
Cyclosporin APseudomonas exotoxinEGFR425.3PE2 µMSignificant increase[58]
MUC1BM7PE2–4 µM40[58]
EpCAM (EGP-2)MOC31PE2 µMSignificant increase[58]
ricin A chaingp702F10-RTA25 mg/kg (in vivo)100[63]
CD5T101-RTA4 µmol/mL101[64]
Cyclosporin SDZ PSC 833ricin A chainCD5T101-RTA4 µmol/mL105[64]
Retinoic acidricin A chaintransferrin receptor454A12-rRTA10 µM10,000[56]
transferrin receptorTfn-rRTA10 µM1000[56]
B cellsM6-rRTA10 µMSignificant increase[56]
p55260F9-rRTA10 µMSignificant increase[56]
WortmanningeloninbFGFRbFGF-gelonin1–10 µMSignificant increase[59]
saporinbFGFRbFGF-SAP1–10 µMSignificant increase[59]
EGFRHBEGF-SAP1–10 µMSignificant increase[59]
bFGFR11A8-SAP1–10 µMSignificant increase[59]
Table 5. Viruses and virus peptides showing enhancement effects on the cytotoxicity of targeted toxins. It is expected that the mechanism is similar to the natural process of fusogenic peptides of viral origin when viruses enter the cytosol of infected cells.
Table 5. Viruses and virus peptides showing enhancement effects on the cytotoxicity of targeted toxins. It is expected that the mechanism is similar to the natural process of fusogenic peptides of viral origin when viruses enter the cytosol of infected cells.
EnhancerApplicationToxinAntigenic TargetTargeted ToxinEnhancer ConcentrationMax. Enhancement FactorRef.
Adenoviruswhole virusPseudomonas exotoxinEGFRPE-EGF2 × 109 pfu/mL10,000[78]
ricin A chainCEAanti-CEA-RTA3 × 108 pfu/mL33[46]
Penton base protein (adenovirus capsid protein)whole virusPseudomonas exotoxinEGFRPE-EGF9 × 103 viruses/cellSignificant increase[79]
KFT25 (N-terminus of Protein G)viral peptides (fusion proteins)dianthintransferrin receptorTfn-KFT25-DIA≤30,000 ng/mL3.8[80]
ricin A chaintransferrin receptorTfn-KFT25-RTA≤10 pM20[81]
HA23viral peptides (conjugates and free peptides)ricin A chaingp120anti-gp120(HIV)-RTA-HA230–300 µg/ mL5[82]
PreS2-domain of hepatitis-B virus surface antigen (TLM)viral peptides (fusion protein)saporinEGFRsaporin-TLM-EGF≤100 nM1 (in vitro) 2.2 (in vivo)[74,83]
angiogeninCD64anti-CD64-TLM-angiogenin≤100 nM20[84]
Table 6. Toxic proteins of eukaryotic origin that have been used in targeted toxins. The pore forming proteins oligomerize in a Ca2+ dependent manner to form pores on the target cell. They in principle fulfill both functions, the toxic effect and the ability to overcome the membrane barrier, in one molecule. Granzymes are apoptosis inducing serine proteases that are supported by pore forming proteins to enter the cells. An enhancement factor cannot be provided here since efficacy and enhancement are inextricably linked with each other in these systems.
Table 6. Toxic proteins of eukaryotic origin that have been used in targeted toxins. The pore forming proteins oligomerize in a Ca2+ dependent manner to form pores on the target cell. They in principle fulfill both functions, the toxic effect and the ability to overcome the membrane barrier, in one molecule. Granzymes are apoptosis inducing serine proteases that are supported by pore forming proteins to enter the cells. An enhancement factor cannot be provided here since efficacy and enhancement are inextricably linked with each other in these systems.
Enhancer = ToxinAntigenic TargetTargeted ToxinEnhancer = Toxin Concentration (IC50)Ref.
Perforin (N-terminal 34 amino acids)CTLA-4 (CD152)hS83P34200–1000 nM[119]
Perforin / Granzyme BVEGFVEGF-SEA (D227A mutant)released by attracted immune cells[120]
Granzyme BLewis YGzmB-dsFv-B335–140 nM[121]
CD64Gb-H22(scFv)1.7–17 nM[122]
Gb(R201K)-H22(scFv)4–7 nM[124]
CD30Gb(R201K)-Ki4(scFv)1.7 nM[130]
EGFRGb(R201K)-scFv1711133 nM[34]
HER2GrB/4D5242–629 nM[125]
GrB/4D5/2629–93 nM[125]
Granzyme MCD64Gm-H22(scFv)1.2–6.4 nM[123]
Table 7. Targeted toxins enhanced by photochemical internalization. The photochemical reaction of a photosensitizer triggered by light exposure causes the disruption of endosomal membranes. This results in the release of targeted toxins into the cytosol and increases their cytotoxicity.
Table 7. Targeted toxins enhanced by photochemical internalization. The photochemical reaction of a photosensitizer triggered by light exposure causes the disruption of endosomal membranes. This results in the release of targeted toxins into the cytosol and increases their cytotoxicity.
Photosensitizer (Enhancer)ToxinAntigenic TargetTargeted ToxinPhotosensitizer (Enhancer) ConcentrationRef.
AlPcS2ageloninCSPG4scFvMEL/rGel5 µg/mL[159]
TPCS2ageloninEGFRrGel/EGF0.1–0.4 µg/mL[160]
HER2MH3-B1/rGel0.1–0.4 µg/mL[163,165]
VEGFRVEGF121/rGel0.4 µg/mL[164]
saporinCD133CD133/1 (AC133)-saporin0.4–1 µg/mL[157,158]
CD133CD133/2 (293C)-saporin0.2–1 µg/mL[157]
CD44IM7-saporin0.35–1 µg/mL[156]
EpCAM (EGP-2)3–17I-saporin0.35 µg/mL[162]
HER2Trastuzumab-saporin0.2 µg/mL[166]
TPPS2ageloninEpCAM (EGP-2)MOC31-gelonin0.3–1 µg/mL[161]
saporinEGFRCetuximab-saporin0.1–1 µg/mL[167]
EGFREGF-saporin0.1–0.2 µg/mL[168]

Share and Cite

MDPI and ACS Style

Fuchs, H.; Weng, A.; Gilabert-Oriol, R. Augmenting the Efficacy of Immunotoxins and Other Targeted Protein Toxins by Endosomal Escape Enhancers. Toxins 2016, 8, 200. https://doi.org/10.3390/toxins8070200

AMA Style

Fuchs H, Weng A, Gilabert-Oriol R. Augmenting the Efficacy of Immunotoxins and Other Targeted Protein Toxins by Endosomal Escape Enhancers. Toxins. 2016; 8(7):200. https://doi.org/10.3390/toxins8070200

Chicago/Turabian Style

Fuchs, Hendrik, Alexander Weng, and Roger Gilabert-Oriol. 2016. "Augmenting the Efficacy of Immunotoxins and Other Targeted Protein Toxins by Endosomal Escape Enhancers" Toxins 8, no. 7: 200. https://doi.org/10.3390/toxins8070200

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop