Abstract
Bladder cancer is the sixth most common cancer in the United States, and it exhibits an alarming 70% recurrence rate. Thus, the development of more efficient antibladder cancer approaches is a high priority. Accordingly, this work provides the basis for a transformative anticancer strategy that takes advantage of the unique characteristics of the bladder. Unlike mucin-shielded normal bladder cells, cancer cells are exposed to the bladder lumen and overexpress EGFR. Therefore, we used an EGF-conjugated anthrax toxin that after targeting EGFR was internalized and triggered apoptosis in exposed bladder cancer cells. This unique agent presented advantages over other EGF-based technologies and other toxin-derivatives. In contrast to known agents, this EGF-toxin conjugate promoted its own uptake via receptor microclustering even in the presence of Her2 and induced cell death with a LC50 < 1 nM. Furthermore, our data showed that exposures as short as ≈3 min were enough to commit human (T24), mouse (MB49) and canine (primary) bladder cancer cells to apoptosis. Exposure of tumor-free mice and dogs with the agent resulted in no toxicity. In addition, the EGF-toxin was able to eliminate cells from human patient tumor samples. Importantly, the administration of EGF-toxin to dogs with spontaneous bladder cancer, who had failed or were not eligible for other therapies, resulted in ~30% average tumor reduction after one treatment cycle. Because of its in vitro and in vivo high efficiency, fast action (reducing treatment time from hours to minutes) and safety, we propose that this EGF–anthrax toxin conjugate provides the basis for new, transformative approaches against bladder cancer.
Keywords: bladder cancer, anthrax toxin, therapy, EGF, EGFR
Introduction
Bladder cancer poses a significant public health concern with an estimated 81,190 new cases and 17,240 related deaths in 2018 just in the United States.1 Importantly, 70% of newly diagnosed patients will suffer disease recurrence after transurethral resection of a bladder tumor and more than 20% will develop invasive bladder cancer.2 In fact, the majority of patients return frequently for office visits, cystoscopic procedures and intravesical treatments, making bladder cancer one of the most expensive malignancies to treat.3–5
Despite efforts to address these issues with adjuvant therapies involving the intravesical instillation of chemotherapeutic and immunomodulatory agents, high rates of disease recurrence and progression continue to persist. These strategies (e.g., using mitomycin C or Bacillus Calmette–Guerin [BCG]) require at least 2 hr-long treatment per session6,7 and have been linked to local and systemic side effects including urinary symptoms, cystitis, fever and inflammatory response.8–12 This situation is worsened by the current BCG shortage.13,14 Therefore, there is a pressing need for more efficient therapeutic approaches against bladder cancer. Importantly, novel efficient countermeasures against this malignancy should be able to exploit the unique advantages that the bladder architecture presents while overcoming its specific challenges.
Umbrella cells protect the epithelia from contact with urine by forming tight junctions, depositing an apical semicrystalline uroplakin coat and by bearing an insulating glycosaminoglycans (GAG) layer.15–17 However, since malignant bladder cells are less differentiated, they present a poorly assembled GAG layer, virtually no uroplakin deposits and are loosely associated with each other.16,18 These characteristics result in a differential bladder lumen exposure of cancer vs. normal cells,16 thereby presenting a great opportunity for treating tumors while minimizing effects on normal cells. Nevertheless, dilution of the bladder content by urine influx and elimination of therapeutics due to bladder voiding requires the use of targeted anticancer drugs against tumor cells.19
Since bladder cancer cells are known to upregulate epidermal growth factor (EGF) receptor (EGFR) expression,20 we tested the use of an EGF-targeted bacterial toxin designed to bypass the limitations of other EGF/toxin-based approaches as an antibladder cancer agent: Since EGFR-targeting therapeutics require receptor endocytosis to be active, impaired EGFR uptake (e.g., by Her2/neu upregulation or the presence of mutations affecting EGFR internalization) are predicted to negatively impact their efficacy.21–23 Importantly, to bypass such limitations we previously devised an approach relying on receptor microclustering to induce nanoparticle uptake by bladder tumor cells.24 These microclustering effects can also be elicited by multivalent agents such as anthrax toxin that, in contrast to monovalent toxins (e.g., diphtheria toxin25–27), forms oligomers at the cell surface promoting its own internalization.28
Here we present evidence that indicates that an EGF–anthrax toxin fusion efficiently targeted and eliminated human, mouse and canine bladder tumor cells, offering a novel, efficacious and fast strategy (minutes vs. hours with current treatments) against both superficial and invasive bladder cancer. Furthermore, our data indicate that Her2 was overexpressed in dog tumors and nevertheless the toxin was highly effective in vitro and in vivo against canine bladder cancer cells. Indeed, six dogs with terminal, treatment-resistant bladder cancer exhibited consistent tumor mass reduction because of exposure to this EGF-toxin. Importantly, in addition to being very specific for EGFR overexpressing cells, this agent is safer than others27 due to its binary nature.28,29 Although this approach is intended to be administered intravesically (NOT systemically) and at a low nM dosage, if any leakage into the bloodstream/adjacent organs would occur, each agent’s component would be independently diluted, making toxin reassembly virtually impossible.29
In conclusion, we believe that the studies presented in this work provide the basis for the design of an innovative and transformative therapeutic strategy against bladder cancer.
Materials and Methods
Protein expression and purification
EGF-PA′ and lethal factor N-terminus fused to the catalytic domain of diphtheria toxin A (LFN-DTA) were expressed by the BL21 (DE3) E. coli strain (New England Biolabs, Ipswich, MA), under the induction of 1 mM IPTG for 2 hr at 30°C. EGF-PA′ was purified as described previously.30 Recombinant LFN-DTA was expressed as a His6-SUMO fusion using the Champion pet-SUMO expression system (Invitrogen, Carlsbad, CA) and purified as previously described.31
Cell sources, origin, maintenance and transfection
Human T24 (RRID: CVCL_0554) bladder cancer cell line was obtained from ATCC while murine MB49 (RRID: CVCL_7076) was provided by the Ratliff lab. The MB49 low EGFR-expressing (MB49LE) cell line is a spontaneous variant isolated during our study. All cell lines were cultured in Dulbecco’s modified Eagle medium (DMEM; Sigma, St. Louis, MO), 5% Penicillin/Streptomycin (Corning, Corning, NY), 5% l-glutamine (Corning) and 10% fetal bovine serum (Atlas Biologicals, Fort Collins, CO) while maintained at 37°C in the presence of 5% CO2.
All experiments were performed with mycoplasma-free cells and the human T24 cell line was authenticated using STR profiling.
Patient tumor disaggregation.
Samples from freshly resected human or canine bladder tumors were washed three times with phosphate-buffered saline (PBS), transferred onto a 150 mm-dish where they were minced with a sterile razor blade, 10 ml of 5 mg/ml or 5% w/v collagenase type 4 (Worthington Biochemical, Lakewood, NJ) were added and incubated at RT for 45 min. The tissue culture dish was then placed at 37°C for 30 min. The sample was transferred to a 50 ml tube and centrifuged at 1,200 rpm for 5 min. The supernatant was discarded, and the pellet was washed three times with PBS and centrifuged once more. After being resuspended using a 10 ml transfer pipette, cells were then passed through a 70 μm cell strainer (BD Biosciences, San Jose, CA) to achieve a single-cell suspension. Cells were resuspended in fully supplemented DMEM+20% fetal bovine serum (FBS) and plated on fibronectin-coated six-well plates (Biomedical Technologies Inc., Stoughton, MA).
Transfection.
MB49LE cells were seeded in a six-well plate containing glass coverslips and allowed to grow to 70% confluence to then be transfected with an EGFR-green fluorescent protein (GFP) plasmid using the Fugene6 (Promega, Madison, WI) transfection reagent according to the manufacturer’s protocol. Then, 24 hr later, the cells were fixed in 4% formaldehyde, mounted on slides and the transfectants were visualized via epifluorescence microscopy using a GFP filter.
EGF–TMR and EGF-toxin binding and internalization
All experiments with human cells were done in the presence of PBS and 50% human urine, while those using murine and canine cells were performed in PBS supplemented with 50% murine and canine urine, respectively. When needed, cytotoxicity was assessed using standard MTT assays.
Stepwise incubation format.
(A). Fluorescently tagged EGF:
2.0 × 105 cells were seeded in fully supplemented media on coverslips in six-well plates, the cells were serum-starved for 12 hr just before use. Cells were rinsed with ice-cold PBS and incubated with 0.5 μg/ml (70 nM) EGF conjugated with tetra-methyl rhodamine (EGF-TMR; Invitrogen, Carlsbad, CA) in PBS/50% urine at 4°C for 45 min and then washed with ice-cold PBS to remove any unbound ligand. Next, coverslips were transferred to 37°C in prewarmed PBS/50% urine for 0 and 30 min to allow internalization. Coverslips were rinsed in ice-cold PBS and the bound, but noninternalized ligand was removed by washing with ice-cold acidic buffer (0.2 M glycine, 0.15 M NaCl, pH = 3.0) for 45 sec, washed and fixed in 4% formaldehyde in PBS for 10 min at RT. Coverslips were mounted on slides and 15 randomly chosen fields were imaged using a Zeiss Axiovert 200 M epifluorescence microscope.
(B). EGF-toxin:
Approximately 2–3 × 104 cells (to yield MTT assay’s high sensitivity medium linear range) were seeded at least 24 hr prior to experiments, and they were serum starved for 12 hr before treatment. Cells were then washed with ice-cold PBS to remove residual FBS and treated with varying concentrations of EGF-PA′ for 45 min at 4°C. After the incubation period, cells were washed with ice-cold PBS to remove any unbound ligand. LFN-DTA (10 nM) in PBS (at 37°C) was added to cells and incubated for times ranging from 0 to 30 min (controls without LFN-DTA were also performed). After incubation period, cells were washed with ice-cold PBS. Bound but noninternalized ligand was removed by washing with ice-cold 0.2 M glycine pH = 3.0, 0.15 M NaCl for 45 sec. Cells were then washed with ice-cold PBS and placed in fully supplemented DMEM media for times ranging from 48 to 72 hr at which point viability was assessed using MTT assays. Experiments were performed by triplicate and at least three times.
(C). EGF-toxin/EGF competition assay:
As described above, cells were plated in six-well plates and serum starved for at least 12 hr prior to treatment. Cells were incubated with 0.63 nM EGF-PA′ with and without 100-fold higher concentrations of wild-type, unlabeled EGF (Cytoskeleton). After EGF-PA′ binding in either the absence or the presence of wild-type EGF for 45 min at 4°C, the cells were washed in PBS and then incubated with 10 nM LFN-DTA for 30 min at 37°C. After rinsing with PBS, the cells were washed with acidic buffer (pH = 3) to strip off the bound, but noninternalized, EGF-PA′/LFN-DTA complexes as described above. Cells were washed in PBS once more and placed in fully supplemented DMEM. Then, 48 hr later, cell viability was determined using MTT assays.
Simultaneous incubation format.
(A). Fluorescently-tagged EGF:
2.0 × 105 cells were plated and serum starved as indicated above. Cells were then rinsed with PBS and incubated with 0.5 μg/ml (70 nM) EGF-TMR at 37°C in PBS/urine for the desired length of time. Cells were then washed with PBS and fixed in 4% formaldehyde in PBS for 10 min at RT and imaged as described above.
(B). EGF-toxin:
2–3 × 104 cells were seeded, and serum starved as indicated above. Cells were then treated and washed in PBS and treated with a single cocktail comprising of EGF-PA′ + LFN-DTA at varying concentrations for the desired length of time. Cells were then washed in PBS and placed in fully supplemented DMEM for 48–72 hr. Viability was assessed using MTT assays. Dose–response curves were plotted and nonlinear regression was applied using the GraphPad Prism software.
Alternatively, apoptotic, necrotic and healthy cell populations were determined using the Apoptosis, Necrosis & Healthy Cell Quantitation Kit (Biotium, Fremont, CA) according to the manufacture’s protocol. Cells were then visualized by fluorescence microscopy.
In vivo antitumor effect of the EGF-toxin on canine spontaneous bladder cancer
After approval of the Purdue Animal Care and Use Committee, the safety of the EGF toxin was confirmed in four laboratory dogs, and then the EGF toxin was tested in a pilot study of six tumor-bearing dogs. The dogs had naturally occurring histopathologically confirmed urothelial carcinoma expressing EGFR detected by immunohistochemistry. These dogs were ones presented to the Purdue University Veterinary Teaching Hospital for treatment of their bladder cancer and they had either failed other treatments or were not eligible for other bladder cancer treatments. The dog owners were informed of the trial and they elected to enroll their dog, the dog owner provided written informed consent. If the dogs had previously received cyclooxygenase (COX)-inhibitors, and had experienced tumor progression on the COX-inhibitor, but needed the medications to control arthritis pain or other painful conditions, they were allowed to continue to receive the COX-inhibitor while in the pilot study. Since COX-inhibitors have antitumor activity in dogs with bladder cancer, it was important to only allow the drugs if cancer progression had occurred on that medication prior to the pilot study.32 Before and after treatment, the dogs were evaluated with blood work (CBC, serum biochemical profile), urinalysis, thoracic radiography, abdominal ultrasonography and a standardized bladder ultrasound protocol33 to measure the bladder masses.33 The ultrasound exams were standardized for machine, operator, dog position, probe position and angle, degree of bladder distension and image analysis program. When this standardized protocol is followed, the interassay variability was <10% and was used to ascertain tumor size.
The dogs were treated with the EGF toxin for up to 5 days in a week (one cycle). Prior to the intravesical administration of EGF-toxin, dogs were sedated with 0.2 mg/kg butorphenol-IV and 3–5 mg/kg dexdomotor-IV and a urinary catheter was passed transurethrally. The bladder was then emptied of its contents and washed with 50 ml of saline solution for 5 min; this step was repeated twice. The EGF-toxin was then instilled intravesically via the urinary catheter into the bladder at a concentration of 200 nM EGF-PA′/400 nM LFN-DTA (in 25–75 ml total volume depending on the size of the dog) and left for 1 hr. During this time the dog was positioned in right lateral recumbency, then shifted to dorsal recumbency and then shifted to left lateral recumbency, being shifted every 20 min. After the 1 hr-dwell time, the solution was removed from the bladder and the bladder washed with 50–100 ml of PBS. The sedation was then reversed by administering Atipamazole (the same dosage as Dexdomotor-IV), and the dog was allowed to recover.
Statistical analysis
Normally distributed data were represented as the mean ± standard deviation of triplicate measurements. Statistical significance of value differences was assessed by applying the t-test. Some biological data analyzed within this work did not follow a normal distribution (failed normality tests). Given their nature, data distributions were shown as box-plots (constructed with Sigmaplot-14) depicting the median, 25–75 (bottom and top of box, respectively) and 10–90 (whiskers) percentiles. When appropriate, statistical significance was evaluated using the nonparametric Wilcoxon test.
Supplemental Methods can be found in the Supporting Information Materials section.
Data availability
Data is available upon request.
Results
The EGF-PA′/LFN-DTA toxin system
To design efficient antibladder cancer approaches it is necessary to consider the bladder’s unique advantages and challenges as a target for therapy: on the one hand, and in contrast to normal bladder epithelia, cancer cells are exposed to the lumen of the bladder (Fig. 1a). On the other hand, the potential beneficial impact of the instillation of nontargeted therapeutics on tumor cells is limited by agent dilution due to constant urine influx, and elimination caused by periodic voiding of the bladder. Therefore, only high affinity targeted therapeutics are expected to overcome these challenges.
Since it is well-known that bladder cancer cells overexpress EGFR,20 we speculated that an EGF-targeted bacterial toxin would be effective in bladder cancer therapeutics. The EGF-toxin system mechanism of action30,31,34 is depicted in Figure 1b. Briefly, an EGF–anthrax protective antigen mutant fusion-protein (EGF-PA′; where PA′ stands for a mutant PA unable to bind anthrax receptor) recognizes EGFR, is proteolytically activated by a cellular, furin-family protease, and assembles as an heptameric or octameric prepore complex on the plasma membrane and recruits up to three molecules of LFN-DTA (Fig. 1b). In contrast to other EGF-agents,27 endocytosis of this EGF-toxin is triggered by PA′ oligomer-mediated microclustering of EGFR. After internalization, the lower pH of the endosome induces a conformational change in the EGF-PA′ oligomer leading to pore formation and translocation of LFN-DTA molecules into the cytosol, which in turn initiates apoptosis by inactivation of elongation factor-2 (EF2; Fig. 1b).
Binding and internalization of EGF-TMR and EGF-toxin in the presence of instillation buffer and urine
To determine the viability of the proposed strategy, we took advantage of the fact that human (h) EGF can also bind mouse and dog EGFR. Specifically, we tested if under conditions that emulate the environment of the bladder during treatment (instillation buffer in the presence of urine and absence of bovine serum), fluorescently labeled human epidermal growth factor (hEGF) was capable of targeting human (T24), mouse (MB49) bladder cancer cell lines as well as dog bladder cancer cells (isolated from canine spontaneous bladder tumors).
During a typical treatment instillation (i.e., after bladder voiding and rinsing) urine is expected to be present at less than 50% of the total bladder content; however, we exaggerated its contribution in the assays to maximize putative detrimental effects of its components (e.g., urea) on receptor binding.
After 30 min of incubation of cells with EGF-TMR in saline solution + 50% of homologous urine at 37°C, we proceeded to eliminate unbound ligand by rinsing, and to remove the bound but noninternalized fraction using an acidic wash.24 Our results indicate that soluble EGF was able to bind its receptor in the presence of saline and urine, and it was also internalized by the different bladder cancer cells (see Fig. 1c, upper panels for representative images). Accordingly, the EGF-toxin was highly efficient for elimination of human, mouse and dog cancer cells under the same conditions (Fig. 1c, lower panel). It should be noted that the EGF-toxin was highly effective against dog tumor and T24 cells even when these were Her2-positive (Fig. 1d, Supporting Information Fig. S1). This is important as Her2 is known to interfere EGFR internalization21,22 and RNA-seq data from 28 dog bladder tumors (vs. 4 normal urothelial layer) indicated that Her2 was enriched in dog bladder tumors in these animals (Supporting Information Fig. S2).
EGF-toxin action on bladder cancer cells
Next, we utilized a stepwise approach to characterize three different aspects of toxin action on bladder cancer cells: toxin binding to EGFR (Fig. 2), and the time-courses of toxin oligomer assembly/internalization/LFN-DTA translocation (Fig. 3), and toxin-induced cell death (Fig. 4).
Binding: establishment of the dose–response relationship for the EGF-toxin system.
We exposed the cells to a range of EGF-PA′ concentrations at 4°C for 45 min (Binding step, Fig. 2a, left) followed by removal of the unbound fraction, while maintaining constant 37°C incubation time at 30 min (prepore assembly, complex internalization and LFN-DTA translocation). After that, we eliminated bound, but noninternalized ligand by acidic wash (the efficiency of this wash was controlled by monitoring the removal of receptor-bound EGF-TMR in a parallel sample—see Materials and methods). Then the cells were incubated at 37°C in complete media for 48 hr and subjected to MTT cell viability assays or cell imaging. Using this experimental set up with different bladder cancer cells, we determined their corresponding EGF-toxin lethal concentration required to kill 50% of the population (LC50; Fig. 2a, right, Table 1). Specifically, human T24, mouse MB49 and cells isolated from several spontaneous canine tumors showed similar sensitivity to the EGF-toxin with a LC50 ranging from 0.21 to 0.54 nM (Table 1). Importantly, addition of excess of WT EGF competed-off EGF-PA′ binding to EGFR, suppressing the EGF-toxin cytotoxic effect (Fig. 2b). These results clearly indicate that the EGF-toxin activity depends on the specific recognition of EGFR. Further supporting the idea that sensitivity to the EGF-toxin depends on the levels of EGFR expressed by the targeted cell, the MB49LE cell line variant (shown to have low levels of EGFR—Supporting Information Fig. S3A) had a LC50 two orders of magnitude above the one from the MB49 parental cell line (Table 1). Importantly, transfection of MB49LE cells with a construct for the expression of EGFR-GFP from a cytomegalovirus promoter greatly enhanced the cell line sensitivity to the EGF-toxin (Supporting Information Fig. S3B). Real-time quantitative reverse transcription-polymerase chain reaction (QRT-PCR) results indicated that EGFR messenger RNA levels of MB49LE cells were comparable to mouse normal bladder epithelial cells (Supporting Information Fig. S3A).
Table 1.
Bladder cancer cells | LC50 (nM) | LC100 (nM) |
---|---|---|
| ||
Human T24 | 0.31 ± 0.05 | 2.2 ± 0.5 |
Mouse MB49WT | 0.21 ± 0.02 | 1.9 ± 0.2 |
MB49LE1 | 20 ± 6 | 205 ± 29 |
Spontaneous canine tumor cells2 | 0.33 ± 0.04 | 2.6 ± 0.4 |
0.54 ± 0.07 | 3.6 ± 0.6 | |
0.34 ± 0.073 | 2.5 ± 0.73 |
MB49LE: variant expressing very low levels of EGFR.
Cells obtained from disaggregation of tumor biopsies. All samples except one (see note 3) were immortalized.
Primary culture obtained from one of the dogs treated with the EGF-toxin.
In addition, our results indicate that Her2 is expressed in T24, but not in MB49 cells (Supporting Information Fig. S1); however, both cell lines presented similar sensitivity to the EGF-toxin (Table 1).
Toxin oligomer assembly and internalization time-course.
Next, we maintained constant the EGF-PA′ concentration to its LC50 value during binding at 4°C while we varied the duration of the incubation at 37°C (after unbound ligand elimination; Fig. 3). This experimental design was used to determine the time-dependence for the assembly/internalization/translocation phase of the EGF-toxin action (Fig. 3, left). Specifically, we established that using EGF-PA′ LC50, 15–30 min were needed to achieve 50% cell elimination (Fig. 3 right, upper panel).
Importantly, under the same experimental conditions, but using EGF-PA′ at its absolute lethal concentration (LC100), we established that exposure times to the toxin as short as 3 min assured maximal tumor cell killing by the toxin (Fig. 3, right, lower panel). These results suggest that at this dose, the amount of pore assembled and the fraction of LFN-DTA released into the cytosol during this short exposure time is sufficient to assure tumor cell elimination.
Toxin-induced cell death time-course.
After incubating cells with EGF-PA′ LC100 for 45 min at 4°C, unbound toxin was eliminated, and the samples were incubated at 37°C for 30 min (Fig. 4). Next, the bound noninternalized ligand was stripped off using acidic wash, and the toxin effects on the cells were monitored by MTT and apoptosis assays at different times (Fig. 4 and Supporting Information Fig. S4, respectively).
Four hours after removal of the toxin, we detected fluorescent annexin-V binding on treated cells, a phenomenon indicative of membrane alterations (exposure of phosphatidylserine in outer leaflet) compatible with apoptosis (Supporting Information Fig. S4). However, by MTT assays, maximal cell death was observed between 24 and 48 hr after exposure to the toxin (Fig. 4a, right panel). In addition, we also conducted experiments in which confluent T24 cell monolayers were exposed to complete/reconstituted EGF-toxin and cell elimination was monitored periodically and quantitatively by microscopy (Fig. 4b). These experiments, in agreement with the toxin-induced cell death time-courses, also indicated sustained cell loss as a function of time, with maximal cell elimination by 24–48 hr (Fig. 4b).
Incubation of human tumor cells with reconstituted EGF-toxin
Exposure of T24 cells to EGF-PA′/LFN-DTA in saline/urine, at 37°C and without elimination of unbound or noninternalized proteins did not result in any significant change in EGF-toxin sensitivity by the studied cells as compared to the stepwise approach results (Supporting Information Fig. S5A). In addition, using EGF-PA′ at its LC50, we titrated the LFN-DTA concentration needed to obtain the expected 50% cell population elimination. Our results showed that LFN-DTA concentrations below 10 nM were suboptimal for supporting 50% cell death by the EGF-PA′ LC50 (Supporting Information Fig. S5B).
Importantly, primary cultures obtained from freshly resected human bladder tumors were tested for EGF-TMR binding/internalization and cell elimination by the EGF-toxin. Our results showed that similar to other bladder cancer cells, primary tumor cells from eight different patients were able to bind and internalize EGF-TMR in the presence of PBS and urine (Figs. 5a and 5b shows two representative examples). In contrast to other cell sources, we could not verify EGFR overexpression a priori for these samples. In fact, our data indicated that there was some degree of EGFR expression level variation among patients, and even some variability within tumor cells from a single patient (Figs. 5a and 5b). However, our results showed that overall patient cancer cells displayed sensitivity to the EGF-toxin yielding an 85% median elimination rate of tumor cells using 2 nM EGF-PA′/10 nM LFN-DTA concentration (Fig. 5c).
The EGF-toxin displays antitumor activity in vivo in dogs with spontaneous, terminal bladder cancer
As the first step toward in vivo studies, we tested the EGF-toxin for potential adverse effects in tumor-free animals. Specifically, the toxin was instilled into the bladder of control animals: six mice and four dogs. No toxicity was detected in the animals by any analysis made (including daily observation and physical exam, urinalyses, complete blood counts and serum biochemical profiles for over a month—data not shown). Therefore, we proceeded to treat six dogs with bulky (in some cases partially blocking the exit of urine), naturally occurring invasive urothelial carcinomas, resistant to conventional therapies with 10 × LC100 dose of EGF-toxin. The overexpression of EGFR (as compared to controls) was verified in these dogs by immunohistochemistry (Figs. 5d and 5e).
In all cases, dogs treated with the EGF-toxin had some reduction in tumor volume (even when multiple tumors were present) with an average of ~30% decrease in size after a single treatment cycle. In some cases, the tumor size was reduced by 10%/day during the 5 days of treatment (see example in Fig. 5f and Table 2). This response after just one treatment cycle is encouraging because invasive urothelial carcinoma in dogs closely mimics human invasive bladder cancer in behavior and treatment response.32 Importantly, the EGF-toxin treatment was very well tolerated. Only a mild bladder irritation (slight increase in amount of blood in the urine and increase in frequency of urination) was noted by treatment cycle Day 5. These resolved within 2 days after treatment ended.
Table 2.
Dog # | Tumor volume (cm3)1 |
Number of doses2 | Age, sex, breed | Weight (kg) | ||
---|---|---|---|---|---|---|
Before | After | Change (%) | ||||
| ||||||
1 | 36.6 | 22 | 40 | 5 | 12 years, Male, Mixed | 23 |
2 | 101.2 | 81.1 | 20 | 3 | 12 years, Male, Collie | 28 |
3 | 4.9 | 3.2 | 35 | 5 | 9 years, Male, Sheltie | 17 |
4 | 6 | 5.3 | 12 | 5 | 12 years, Female, Mixed | 31 |
5 | 5.7 | 3.9 | 31 | 5 | 11 years, Male, Beagle | 14 |
6 | 3.9 | 2.5 | 36 | 4 | 12 years, Male Beagle | 14 |
Measured by ultrasound (see Materials and methods).
Within a single treatment cycle.
Discussion
The results presented in our study set the basis for the development of a therapeutic strategy against bladder cancer considered transformative due to its (i) high efficiency for targeting bladder cancer cells; (ii) fast action, that is, having the potential of drastically decreasing patient treatment time from hours (current therapies) to minutes; (iii) its ability to eliminate Her2-positive cells; (iv) demonstrated in vivo efficacy and (v) its enhanced safety.
The constant influx of urine into the lumen of the bladder and the necessary voiding of the bladder content are serious challenges to the instillation of nontargeted therapeutics against bladder cancer.19 This makes the devising of targeted approaches a must for efficacious antibladder cancer strategies.
Since EGFR is overexpressed in both superficial and invasive bladder carcinomas,20 this receptor emerges as a suitable target against bladder cancer. However, use of the anti-EGFR monoclonal antibody cetuximab against bladder cancer have yielded disappointing results, failed to prevent disease progression and developed drug resistance/patient toxicity.35–37
Since internalization of EGFR complexes plays an important role in the success of these targeted agents, the presence of EGFR internalization-impairing mutations or upregulation of Her2/neu (known to interfere with EGFR endocytosis21) are among the factors known to impair the efficacy of EGF-based strategies.21–23
Therefore, and based on previous developments from our lab24 we decided to use an agent that while targeting EGFR, would be able to induce its own internalization through a clustering-dependent mechanism. Specifically, we used a chimeric protein resulting from the fusion of EGF (targeting ligand) and the binary anthrax toxin (cytotoxic component and microclustering active element). Very importantly, endocytosis of this agent is triggered by oligomerization of the PA subunit of the anthrax toxin (Fig. 1),28 that is, contrary to other toxins (e.g., diphtheria toxin25–27) anthrax assembles oligomers at the plasma membrane inducing its own uptake.28Therefore, as opposed to other EGF-based technologies, the EGF-toxin is able to bypass Her2s negative effect on EGFR internalization (Figs. 1c and 1d; Table 1 shows similar sensitivity by the Her2+ T24 cells and the Her2− MB49 line) and is predicted to be insensitive to EGFR mutations that inhibit its endocytosis.
This agent not only fills-in an important need for better, more efficient antibladder cancer therapeutics, but we humbly think that it will be transformative for bladder cancer treatment for the following reasons.
-
This is an approach of very high efficacy (with subnanomolar LC50) against bladder tumor cells—Figures 1 and 2, Table 1. It is known that more than 90% of the EGFR in cells display an affinity (as measured by EGF-EGFR dissociation constant, KD) for EGF of ~2 nM.38 As expected, since our data reflects biological activity, the LC50 range value for EGFR-expressing bladder cancer cells (~0.2–0.5 nM; Table 1) is substantially lower than the KD. This is often seen with pharmacologically active ligands39,40 and suggests that is not necessary to bind a substantial number of receptors on the cell to trigger maximal biological activity, this is expected for the EGF-toxin considering the enzymatic nature of LFN-DTA.
Importantly, the EGF-toxin approach is very specific for cells overexpressing EGFR; that is, cancer cells (Fig. 2b, Table 1 and Supporting Information Fig. S3B). Indeed, we found that the EGFR-low level expressing MB49LE cells are two orders of magnitude less sensitive than the MB49 original cell line (Table 1). However, transfection/expression of GFP-EGFR in these cells substantially increased their sensitivity for the EGF-toxin (Supporting Information Fig. S3B).
The EGF-toxin exhibits very fast action (min vs. hr of current treatments). The time-course of toxin action (Fig. 3) indicates that the kinetics of PA-clustering mediated uptake is not substantially different from the rate of other ligand-receptor complexes. Indeed, pulse-chase experiments indicate that 5 min after binding, a substantial fraction of EGF can be found in the early endosomal compartment.41,42 This is very important because it points out to the potential of this agent to drastically reduce patient treatment time from hours to minutes.
The EGF-toxin is capable of eliminating tumor cells even in the presence of Her2. The EGFR-related protein Her2 is a poor prognosis marker, but also an EGFR internalization-interfering factor21; however, our results clearly demonstrate that the EGF-toxin is highly effective (Figs. 1c and 1d, Table 1) independently of Her2 expression. These observations indicate that the microcluster-induced endocytosis mechanism24,28 overrides Her2-mediated inhibition of EGFR internalization. In fact, while uptake of conventional EGF-derived agents27 occurs by a mechanism initiated by EGFR dimerization affected by the formation of EGFR-Her2 dimers, EGF-PA′ triggers a PA-mediated microclustering internalization event.28 Therefore, we also predict that this agent would be able to eliminate cells displaying EGFR-internalization mutants.
-
This approach is efficacious in vivo, even when used in patient dogs refractory to all other available treatments (Figs. 5d–5f, Table 2). In addition to be readily applicable to superficial bladder cancer, we speculate that its high efficacy may turn this strategy into a substitute for bladder resection in some invasive bladder cancer cases.
Cell killing experiments performed on T24 cell monolayers in which we monitored toxin action by direct observation of cell elimination (creation of “holes” in the cell monolayer), suggested that as early as 4–8 hr after treatment, the outer layer of a putative tumor would be breached to an extent that a second toxin application at this time would allow access to the underlying layers of the tumor. Furthermore, the looser tumor cell junctions would allow deeper tumor penetration of this molecular-sized agent as compared to nanoparticulated entities or bacteria (e.g., BCG). We speculate that these observations should be taken into account for the design of future treatment strategies using this agent.
The EGF–toxin is a safe approach. This approach is safer than any other toxin-based approach reported in the literature.27 Among the factors that assure the safety of this approach: (a) the toxin is instilled in the lumen of the bladder (NOT in the bloodstream); therefore, as discussed above, only the exposed tumor cells are accessed by the agent. (b) Even in the event of a toxin leak into circulation, our data indicate that due to their heightened sensitivity (see item #1 above) the dose required for bladder cancer cell elimination is substantially lower than the one required for toxin intoxication (e.g., bladder cancer cell LC100 ~ 2 nM vs. mice killing dose ~ 1 μM43). Even if the entire volume of the mouse bladder instillate (80 μl) would to leak into the bloodstream (~1.5 ml), the resulting toxin concentration would be 10,000 times below the lethal dose. (c) In contrast to one-component agents (e.g., diphtheria toxin27), binary toxins (such as anthrax) are safer as dilution greatly decreases the probability of both components reconstituting on normal cells. In addition, when the potential toxicity of the bladder-instilled agent was tested in control animals (mice and dogs) it yielded negative results; furthermore, administration of the toxin to dogs with spontaneous bladder cancer led to substantial tumor reduction without discernible toxic side-effects.
To summarize, the approach presented in this article can be transformative for the treatment of superficial bladder cancer due to its high efficacy both in vitro and in vivo and its fast action. We also speculate that its high efficiency in vivo could make it a viable substitute for cystectomy in early stages of invasive bladder cancer.
Supplementary Material
What’s new?
Bladder cancer is hard to treat and after surgery has a notoriously high recurrence rate (70%). Here the authors propose a new therapeutic strategy combining epidermal growth factor (EGF) with anthrax toxin. Since bladder cancer cells are exposed to urine and carry high levels of EGF receptor, the toxin after intravesical application was specifically taken up by cancer cells and induced rapid apoptosis, regardless of whether cancer cells expressed another EGF receptor, Her2, or not. This underscores the broad potential of the approach for treatment of bladder tumors in the future.
Acknowledgements
We thank members of the Aguilar, Knapp and Ratliff labs for discussions and critical reading of the manuscript, and to Claudia B. Hanna for excellent technical assistance. This work was supported by grants R21-CA151961 (NIH/NCI) and PILOT from Purdue Center for Cancer Research to RCA.
Abbreviations:
- ADP
adenosine diphosphate
- BCG
Bacillus Calmette–Guerin
- CMV
cytomegalovirus
- DMEM
Dulbecco’s modified Eagle medium
- EF2
elongation factor 2
- EGF
epidermal growth factor
- EGFR
epidermal growth factor receptor
- FBS
fetal bovine serum
- GAG
glycosaminoglycan
- GFP
green fluorescent protein
- hEGF
human epidermal growth factor
- KD
dissociation constant
- LC100
lethal concentration required to kill 100% of the population
- LC50
lethal concentration required to kill 50% of the population
- LFN-DTA
lethal factor N-terminus fused to the catalytic domain of diphtheria toxin A
- MB49LE
MB49 low EGFR-expressing
- mRNA
messenger RNA
- MTT
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- nM
nanomolar
- PA
protective antigen
- PBS
phosphate-buffered saline
- QRTPCR
real-time quantitative reverse transcription-polymerase chain reaction
- RNA-seq
ribonucleic acid-sequencing
- TMR
tetra-methyl rhodamine
Footnotes
Conflict of interest: The authors declare no potential conflicts of interest.
Additional Supporting Information may be found in the online version of this article.
References
- 1.Society AC. Cancer facts and figures 2018. Atlanta, GA: American Cancer Society, 2018. [Google Scholar]
- 2.Chou R, Selph S, Buckley DI, et al. Intravesical therapy for the treatment of nonmuscle invasive bladder cancer: a systematic review and meta-analysis. J Urol 2017;197:1189–99. [DOI] [PubMed] [Google Scholar]
- 3.Mossanen M, Gore JL. The burden of bladder cancer care: direct and indirect costs. Curr Opin Urol 2014;24:487–91. [DOI] [PubMed] [Google Scholar]
- 4.Sievert KD, Amend B, Nagele U, et al. Economic aspects of bladder cancer: what are the benefits and costs? World J Urol 2009;27:295–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Boormans JL, Zwarthoff EC. Limited funds for bladder cancer research and what can we do about it. Bladder Cancer 2016;2:49–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.ACS. Treatment of bladder cancer by stage. Atlanta, GA: American Cancer Society, 2019. [Google Scholar]
- 7.Koga H, Ozono S, Tsushima T, et al. Maintenance intravesical bacillus Calmette-Guerin instillation for ta, T1 cancer and carcinoma in situ of the bladder: randomized controlled trial by the BCG Tokyo Strain Study Group. Int J Urol 2010;17:759–66. [DOI] [PubMed] [Google Scholar]
- 8.Chade DC, Shariat SF, Dalbagni G. Intravesical therapy for urothelial carcinoma of the urinary bladder: a critical review. Int Braz J Urol 2009;35:640–50. discussion 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zlotta AR, Fleshner NE, Jewett MA. The management of BCG failure in non-muscle-invasive bladder cancer: an update. Can Urol Assoc J 2009;3:S199–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hall MC, Chang SS, Dalbagni G, et al. Guideline for the management of nonmuscle invasive bladder cancer (stages ta, T1, and tis): 2007 update. J Urol 2007;178:2314–30. [DOI] [PubMed] [Google Scholar]
- 11.Lamm DL, McGee WR, Hale K. Bladder cancer: current optimal intravesical treatment. Urol Nurs 2005;25:323–6. [PubMed] [Google Scholar]
- 12.Koya MP, Simon MA, Soloway MS. Complications of intravesical therapy for urothelial cancer of the bladder. J Urol 2006;175:2004–10. [DOI] [PubMed] [Google Scholar]
- 13.Bandari J, Maganty A, MacLeod LC, et al. Manufacturing and the market: rationalizing the shortage of bacillus Calmette-Guerin. Eur Urol Focus 2018;4:481–4. [DOI] [PubMed] [Google Scholar]
- 14.Messing EM. The BCG shortage. Bladder Cancer 2017;3:227–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Romih R, Korosec P, de Mello W Jr, et al. Differentiation of epithelial cells in the urinary tract. Cell Tissue Res 2005;320:259–68. [DOI] [PubMed] [Google Scholar]
- 16.DeGraff DJ, Cates JM, Mauney JR, et al. When urothelial differentiation pathways go wrong: implications for bladder cancer development and progression. Urol Oncol 2013;31:802–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fry CH, Meng E, Young JS. The physiological function of lower urinary tract smooth muscle. Auton Neurosci 2010;154:3–13. [DOI] [PubMed] [Google Scholar]
- 18.Hurst RE, Greenwood-Van Meerveld B, Wisniewski AB, et al. Increased bladder permeability in interstitial cystitis/painful bladder syndrome. Transl Androl Urol 2015;4:563–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Douglass L, Schoenberg M. The future of Intravesical drug delivery for non-muscle invasive bladder cancer. Bladder Cancer 2016;2:285–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Messing EM. Clinical implications of the expression of epidermal growth factor receptors in human transitional cell carcinoma. Cancer Res 1990;50:2530–7. [PubMed] [Google Scholar]
- 21.Hendriks BS, Opresko LK, Wiley HS, et al. Coregulation of epidermal growth factor receptor/human epidermal growth factor receptor 2 (HER2) levels and locations: quantitative analysis of HER2 overexpression effects. Cancer Res 2003;63:1130–7. [PubMed] [Google Scholar]
- 22.Yamaoka T, Ohba M, Ohmori T. Molecular-targeted therapies for epidermal growth factor receptor and its resistance mechanisms. Int J Mol Sci 2017;18:E2420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wang Q, Chen X, Wang Z. Dimerization drives EGFR endocytosis through two sets of compatible endocytic codes. J Cell Sci 2015;128:935–50. [DOI] [PubMed] [Google Scholar]
- 24.Coon BG, Crist S, Gonzalez-Bonet AM, et al. Fibronectin attachment protein from bacillus Calmette-Guerin as targeting agent for bladder tumor cells. Int J Cancer 2012;131:591–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ratts R, Zeng H, Berg EA, et al. The cytosolic entry of diphtheria toxin catalytic domain requires a host cell cytosolic translocation factor complex. J Cell Biol 2003;160:1139–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Murphy JR. Mechanism of diphtheria toxin catalytic domain delivery to the eukaryotic cell cytosol and the cellular factors that directly participate in the process. Toxins (Basel) 2011;3:294–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Yang X, Kessler E, Su LJ, et al. Diphtheria toxin-epidermal growth factor fusion protein DAB389EGF for the treatment of bladder cancer. Clin Cancer Res 2013;19:148–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Young JA, Collier RJ. Anthrax toxin: receptor binding, internalization, pore formation, and translocation. Annu Rev Biochem 2007;76:243–65. [DOI] [PubMed] [Google Scholar]
- 29.Zahaf NI, Schmidt G. Bacterial toxins for cancer therapy. Toxins (Basel) 2017;9:E236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mechaly A, McCluskey AJ, Collier RJ. Changing the receptor specificity of anthrax toxin. MBio 2012;3:e00088–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.McCluskey AJ, Olive AJ, Starnbach MN, et al. Targeting HER2-positive cancer cells with receptor-redirected anthrax protective antigen. Mol Oncol 2013;7:440–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Knapp DW, Ramos-Vara JA, Moore GE, et al. Urinary bladder cancer in dogs, a naturally occurring model for cancer biology and drug development. ILAR J 2014;55:100–18. [DOI] [PubMed] [Google Scholar]
- 33.Honkisz SI, Naughton JF, Weng HY, et al. Evaluation of two-dimensional ultrasonography and computed tomography in the mapping and measuring of canine urinary bladder tumors. Vet J 2018;232:23–6. [DOI] [PubMed] [Google Scholar]
- 34.LainE, MartineL, LadanD, et al. Molecular motions as a drug target: mechanistic simulations of anthrax toxin edema factor function led to the discovery of novel allosteric inhibitors. Toxins (Basel ) 2012;4:580–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.DeVita VT Jr. Chu E a history of cancer chemotherapy. Cancer Res 2008;68:8643–53. [DOI] [PubMed] [Google Scholar]
- 36.Philips GK, Halabi S, Sanford BL, et al. A phase II trial of cisplatin (C), gemcitabine (G) and gefitinib for advanced urothelial tract carcinoma: results of cancer and leukemia group B (CALGB) 90102. Ann Oncol 2009;20:1074–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hussain M, Daignault S, Agarwal N, et al. A randomized phase 2 trial of gemcitabine/cisplatin with or without cetuximab in patients with advanced urothelial carcinoma. Cancer 2014;120:2684–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ozcan F, Klein P, Lemmon MA, et al. On the nature of low- and high-affinity EGF receptors on living cells. Proc Natl Acad Sci USA 2006;103:5735–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Macdougall IC. Optimizing the use of erythropoietic agents—pharmacokinetic and pharmacodynamic considerations. Nephrol Dial Transplant 2002;17(Suppl 5):66–70. [DOI] [PubMed] [Google Scholar]
- 40.Johnson M Beta2-adrenoceptors: mechanisms of action of beta2-agonists. Paediatr Respir Rev 2001;2:57–62. [DOI] [PubMed] [Google Scholar]
- 41.Burke P, Schooler K, Wiley HS. Regulation of epidermal growth factor receptor signaling by endocytosis and intracellular trafficking. Mol Biol Cell 2001;12:1897–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tomas A, Futter CE. Eden ER. EGF receptor trafficking: consequences for signaling and cancer. Trends Cell Biol 2014;24:26–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Shoop WL, Xiong Y, Wiltsie J, et al. Anthrax lethal factor inhibition. Proc Natl Acad Sci USA 2005;102:7958–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dhawan D, Paoloni M, Shukradas S, et al. Comparative gene expression analyses identify luminal and basal subtypes of canine invasive Urothelial carcinoma that mimic patterns in human invasive bladder cancer. PLoS One 2015;10:e0136688. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Data is available upon request.