Next Article in Journal
The Role of Galectin-3 in Predicting Congenital Heart Disease Outcome: A Review of the Literature
Next Article in Special Issue
GSK-3 Inhibitor Elraglusib Enhances Tumor-Infiltrating Immune Cell Activation in Tumor Biopsies and Synergizes with Anti-PD-L1 in a Murine Model of Colorectal Cancer
Previous Article in Journal
Transcriptomic, Physiological, and Metabolomic Response of an Alpine Plant, Rhododendron delavayi, to Waterlogging Stress and Post-Waterlogging Recovery
Previous Article in Special Issue
Stability Characterization of the Novel Anti-Cancer HM-10/10 HDL-Mimetic Peptide
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel SPEA Superantigen Peptide Agonists and Peptide Agonist-TGFαL3 Conjugate. In Vitro Study of Their Growth-Inhibitory Effects for Targeted Cancer Immunotherapy

by
Sara S. Bashraheel
1 and
Sayed K. Goda
2,*
1
QU Health, Qatar University (QU), Doha P.O. Box 2713, Qatar
2
College of Science and Technology, University of Derby, Derby DE22 1GB, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10507; https://doi.org/10.3390/ijms241310507
Submission received: 16 May 2023 / Revised: 13 June 2023 / Accepted: 19 June 2023 / Published: 22 June 2023
(This article belongs to the Special Issue Cancer Treatment: New Drugs and Strategies)

Abstract

:
Bacterial superantigens (SAgs) are effective T-cell stimulatory molecules that lead to massive cytokine production. Superantigens crosslink between MHC class II molecules on the Antigen Presenting Cells (APC) and TCR on T-cells. This enables them to activate up to 20% of resting T cells, whilst conventional antigen presentation results in the activation of 0.001–0.0001% of the T cell population. These biological properties of superantigens make them attractive for use in immunotherapy. Previous studies have established the effectiveness of superantigens as therapeutic agents. This, however, was achieved with severe side effects due to the high lethality of the native toxins. Our study aims to produce superantigen-based peptides with minimum or no lethality for safer cancer treatment. In previous work, we designed and synthesized twenty overlapping SPEA-based peptides and successfully mapped regions in SPEA superantigen, causing a vasodilatory response. We screened 20 overlapping SPEA-based peptides designed and synthesized to cover the whole SPEA molecule for T-cell activation and tumor-killing ability. In addition, we designed and synthesized tumor-targeted superantigen-based peptides by fusion of TGFαL3 either from the N′ or C′ terminal of selected SPEA-based peptides with an eight-amino acid flexible linker in between. Our study identified parts of SPEA capable of stimulating human T-cells and producing different cytokines. We also demonstrated that the SPEA-based peptide conjugate binds specifically to cancer cells and can kill this cancer. Peptides induce T-cell activation, and tumor killing might pave the way for safer tumor-targeted superantigens (TTS). We proposed the combination of our new superantigen-based peptide conjugates with other immunotherapy techniques for effective and safer cancer treatment.

Graphical Abstract

1. Introduction

Cancer is a disease of uncontrolled proliferation of cells that used to be healthy and subject to the controlled mechanism. Traditional cancer treatment strategies have been surgery, chemotherapy, and radiotherapy. Due to the severe side effects and resistance to these treatments, we need more tolerant and effective cancer therapies. The focus has been on utilizing the patient’s immune system to find and destroy cancer cells with little or no harm to the healthy ones. Recently several immunotherapy strategies have been developed, such as immune checkpoint inhibitors, Anti-CTLA-4 antibodies, Anti-PD-1 drugs, and Anti-PD-L-1 drugs [1,2,3,4,5,6,7,8,9,10], chimeric antigen receptor T-cells, or CAR-T [11,12,13,14,15,16,17,18,19,20,21].
Superantigens (SAg) are protein toxins known to activate up to 20% of resting T-cells by binding to MHC-II on Antigen Presenting Cells (APC) and Vβ domain of T-cell Receptor from the side and crosslinking both together [22,23]. This feature makes the SAg very attractive for immunotherapy. Superantigens have been heavily studied for cancer immunotherapy as it is effective in T-cell-dependent tumor killing [24,25,26,27,28,29,30]. To minimize cytotoxicity by the massive immune cell activation in the body, SAgs have been fused to anticancer antibody fragments to specifically target the induced T-cell activity towards cancer cells [31,32,33,34]. This approach is known as tumor-targeted superantigens (TTS). Several studies have linked bacterial SAgs to Fab fragment tumor-specific antibodies or ligands targeting a receptor either highly expressed or only expressed on the tumor cells. Ligand-targeted therapeutics (LTTs) have been favored over mAbs due to their lower antigenicity and higher drug penetration level into a solid tumor [35,36].
Epidermal growth factor receptor (EGFR) is a cancer marker in various carcinomas, including breast, head, neck, oesophagal, gastric, pancreatic, colorectal, prostate, bladder, renal, ovarian, and ovarian Non-Small Cell Lung Cancer (NSCLC). The higher EGFR expression level has been associated with the advanced tumor stage [37,38,39,40]. EGFR-directed antibodies have been used in cancer therapy [41]. Moreover, SAgs were fused with the third loop of transforming growth factor α (TGFαL3) to study the possibility of the therapeutic application of TGFαL3-SAg as a novel anti-tumor candidate in EGFR tumor-expressing cells [37,42,43,44].
One side effect of SAgs is Sepsis Shock Syndrome, which results from cytokine accumulation in the body. In vivo studies demonstrate that the severity of toxic shock is linked to the increased systemic level of cytokines, IL-6 in particular [45,46,47,48]. Therefore, it is reasonable to assume that the reduced cytokine level could lead to reduced toxicity of the SAgs. A point mutation D227A in the MHC-II binding site of SEA resulted in reduced toxicity. Tumor-bearing mice treated with Fab-SEA D227A resulted in a significant tumor reduction and less toxicity compared to Fab-SEA protein. The lower toxicity was due to the low binding affinity to MHC-II, resulting in a lower systemic cytokine level [48,49].
The present study proposes a new approach to produce a superantigen-positive lethality-negative molecule for cancer immunotherapy. We synthesized SPEA superantigen-based peptides and screened for peptide agonists of SPEA. We then show that the isolated peptide(s) could interact with T cell receptors and behave as superantigens but with much less activity and production of cytokines. Therefore, these peptides could enhance the patient’s immune system without causing the cytokine storm, usually caused by the full superantigens.
In this study, the identified peptide agonists are then conjugated with cancer-targeted sequence loop GFαL3. Therefore, the produced novel tumor-targeted SAg-based peptides conjugates could accomplish tolerable cancer immunotherapy.

2. Results

2.1. T-Cell Activity Assay

SPEA-based peptides were tested for activity by assessing their ability to activate T-cells. PBMCs treated with SPEA-based peptides were stained with fluorescently labelled anti-CD3 and anti-CD25 antibodies and analyzed using a flow Cytometer for CD3+ CD25+ cells. SPEA was used as a positive control, and DMSO and H2O were used as a negative control. Figure 1 shows that peptides SP2, SP3, and SP15 significantly affected T-cells. Up to 30% T-cell activation was achieved in response to SP2, whereas up to 25% T-cell activation was achieved by SP3 and 15% by SP15. All remaining peptides had no significant T-cell activation effect.

2.2. Production of Cytokines

In Figure 2A and Figure 3A, the concentration of IL-6 produced in response to SPEA, SP2, SP3, and SP15 is around 31,000, 5900, 2500, and 3000 pg/mL, respectively, whereas the fusion peptides TGFSP2, SP2TGF, TGFSP3, SP3TGF, TGFSP15, and SP15TGF (in all peptide conjugates, TGF = TGFαL3 resulted in 3831 pg/mL, 6365 pg/mL, 5624 pg/mL, 5269 pg/mL, 8387 pg/mL, and 779 pg/mL, respectively. The concentration of IL-10 was reduced from 320 pg/mL in response to treatment with SPEA to around 27 pg/mL in response to SP2 and SP3, and 48 pg/mL in response to SP15 (Figure 2B). The level of IL-10 remained at 48 pg/mL for TGFSP2, but elevated to 65 pg/mL in response to SP2TGF, TGFSP3, and SP3TGF, and 241 pg/mL in response to TGFSP15 (Figure 3B). Furthermore, the concentration of IL-1β was 550 pg/mL in response to SPEA. At the same time, it decreased to around 100 pg/mL in response to SP2 and SP3 (Figure 2D). IL-1β concentration produced in response to TGFSP2, SP2TGF, TGFSP3, SP3TGF, and TGFSP15 was 154 pg/mL, 94 pg/mL, 126 pg/mL, 152 pg/mL, and 282 pg/mL, respectively (Figure 3D). The mixed culture incubated with SP2, SP3, or SP15 did not produce TNFa (Figure 2C). The fusion peptides had a low but significant concentration of TNFa produced, around 87 pg/mL, 45 pg/mL, 41 pg/mL, 24 pg/mL, and 46 pg/mL in response to TGFSP2, SP2TGF, TGFSP3, SP3TGF, and TGFSP15, respectively (Figure 3C). SP15TGF was unable to stimulate significant cytokine production (Figure 3C).

2.3. Binding Assessment

The binding of tumor-targeted superantigen-based peptides to EGFR on MDA-MB-468 cells was assessed by ArrayScan™ XTI. SP3TGF and TGFSP15 had the highest percentage of binding to EGFR receptor, up to 45%, whereas SP15TGF had up to 35% binding, and TGFSP2, SP2TGF, and TGFSP3 had 15–25% binding to EGFR receptors (Figure 4).

2.4. MTT Assay

Compared to the Media control, the SPEA-treated PBMCs showed a higher absorbance than the control due to a proliferation effect of the whole superantigen, while SP2TGF and SP3TGF had no toxic effect on the PBMC viability. On the other hand, TGFSP2 and TGFSP3 had decreased cell viability compared to the media (Figure 5).

2.5. Tumor Killing Assay

Tumor cell viability was measured by counting DAPI positive nuclei for each sample as a percentage of TP (Tumor cells with PBMCs) sample. Around 50% tumor killing was detected in response to SP2 and SP3 compared to the corresponding solvent control. At the same time, SP15 had no effect, and SPEA showed 70% tumor killing (Figure 6). Tumor cell viability was also assessed in response to tumor-targeted peptides. TGFSP2 shows the highest percentage of tumor cell count reduction with up to 65% tumor killing. Further, 20–40% killing was achieved in response to SP2TGF, TGFSP3, and SP3TGF. Interestingly, TGFSP15 induced a 25% tumor-killing effect (Figure 7).

3. Discussion

The cytotoxicity and pathogenicity of bacterial superantigens, including SPEA, are due to their ability to activate up to 20% of the T-cell repertoire. This strong interaction is the main reason for its mitogenic activity. These biological properties of superantigens make them very attractive for use in immunotherapy. Previous studies have shown the effectiveness of different superantigens as therapeutics for cancer immunotherapy [27,32,50,51,52,53]. These studies used the entire superantigen molecules, leading to many treatment pitfalls. The complete molecules raised high immunogenicity, followed by neutralization by the patient’s immune system, demolishing its therapeutic effect [52]. The large size of the full superantigen molecules would also be an obstacle to the permeation of the biological barrier in the patient’s body. This is in addition to the severe toxicity which hindered their use in clinical trials. We embarked on a campaign to produce modified and safer forms of superantigens and to test their ability to enhance the immune systems for cancer therapy.
One of the damages bacterial superantigens can cause is severe hypotension, possibly leading to the patient’s death. Our previous work [54] identified the regions of SPEA involved in causing vasodilation and possible hypotension. We carried out the work by designing and synthesizing a series of 20 overlapping peptides spanning the entire sequence of SPEA. The vascular response of each peptide was measured, and three active peptides (SP7, SP11, and SP19) were identified [54]. Removing these sequences and the production of modified superantigens lacking these peptides could lead to safer SPEA variants for cancer treatment.
To produce superantigenicity-positive lethality-negative molecules, we extended our work further. We tested the following 20 overlapping peptides, which were synthesized in our previous work [54], which cover the full amino acid sequence of SPEA for T-cell activation and production of cytokines:
SP1(1–40), SP2(11–50), SP3(21–60), SP4(31–70), SP5(41–80), SP6(51–90), SP7(61–100), SP8(71–110), SP9(81–120), SP10(91–130), SP11(101–140), SP12(111–150), SP13(121–160), SP14(131–170), SP15(141–180), SP16(151–190), SP17(161–200), SP18(171–210), SP19(181–220), and SP20(191–222).
We successfully identified the fragments SP2(11–50), SP3(21–60), and SP15(141–180), which can stimulate the proliferation of human T-cells and produce the tumor necrosis IL-1, IL-6, and IL-10 (Figure 1), and show similar profile as noted by the full SPEA superantigen. The level of cytokines, however, produced is lower than the full superantigen. It was reported that the amino acid sequence TNKKMVTAQELD is the sequence on the superantigen that binds to CD28, and is essential for the induction of cytokines. This sequence is not entirely included in the SP2 and SP3 peptides, which might be the reason for their reduced superantigenicity and toxicity [55]. Therefore, the peptides isolated in this study would avoid the super toxicity caused by the complete superantigen molecules, but still enhance the T-cell proliferation for cancer immune therapy.
IL-6 is a cytokine with pleiotropic activities. It plays a vital role in acquiring immune response by stimulating antibody production and effective T-cell development. IL-6 Figures tumor growth by mobilizing anti-tumor T-cell immune response to control tumor growth. IL-6 plays a crucial role in boosting T-cell trafficking to lymph nodes and tumor sites. IL-6 signaling can also reshape the T cell immune response, shifting it from a suppressive to a responsive state that can effectively act against tumors [56,57]. On the other hand, because of its pleiotropic nature, continuous dysregulated production can develop different diseases [56].
Studies have shown that IL-1α inhibits cell proliferation of MCF-7 breast cancer, A375 melanoma, prostate stem cells, and murine primary mammary cells by causing G0–G1 cell cycle arrest [58,59,60]. Another investigation showed that IL-1 increases the recruitment of neutrophils to the tumor, which increases the neutrophil ability of the tumor killing. The study suggests that due to its action on neutrophils, IL-1 has a robust anti-cancer effect [61]. IL-10 is an anti-inflammatory cytokine that plays a crucial role in tumor killing and inhibits the production of IFNg, IL-2, IL-3, and TNFα [62].
Our findings so far show that three of the peptides SPEA agonists have similar profiles of the full superantigen SPEA in activating the T- cells and producing different types of cytokines with an anti-cancer effect such as IL-1, IL-6, and IL-10, as mentioned above (Figure 2 and Figure 3). These peptides, however, might not have the severe toxicity of the full superantigens. The significant advantage of the peptide’s agonists identified in this study, it would be much easier to modify these peptides to obtain shorter or mutated peptides to produce more active and efficient variants with less toxicity to enhance T-cells against cancer.
There are two main strategies for drug delivery in cancer treatment, aiming to maximize the drug to target the tumor and minimize the exposure of the healthy cells. One uses a delivery vehicle such as nanoparticles [63,64]. The second is a covalent chemical binding of the leading drug to a small moiety which binds specifically to cancer receptors, such as antibody-drug conjugates (ADC) [65], antibody-directed enzyme prodrug therapy (ADEPT) [66], and many others [67]. We, in this study, implemented the second strategy to maximize the superantigen peptides in the cancer tissue.
Using the second strategy, we extended our work to target tumors using these peptides. Epidermal growth factor receptor (EGF), the human EGF receptor, plays an essential role during cell growth [68]. Studies reported that EGFR is overexpressed in cancer cells such as colorectal, breast, and non–small cell lung cancers [69,70,71]. Due to its overexpression on cancer cells, several investigations used the third loop (L3) of transforming growth factor-α (TGF-α) to conjugate with protein antibodies [72,73,74,75], or full superantigen SEB to exert the toxic effect of a drug in the vicinity of cancers to achieve targeted cancer therapy [76].
To accomplish specificity and minimize toxicity, the SPEA peptide agonists were conjugated by the highly expressed TGFαL3. This targeted moiety will lead the peptide agonist to bind specifically to the cancerous cells, activate a safe number of T lymphocytes, and direct them only to the tumor. This will achieve the specific killing of the tumor and leave the healthy cells intact. Considering the misfolding the loop may cause to the peptide agonist, we designed and synthesized TGFaL3-Peptide conjugate (N-terminal) and Peptide-TGFaL3 conjugate (C-terminal) (Figure 8). These conjugates comprise the third loop of transforming growth factor-α (TGF-α), a flexible linker and the peptide agonist (Figure 8 and Table 1). The results in Figure 6 and Figure 7 show that TGFαL3-peptides agonists and free peptide agonist induced cancer-killing, while the results in Figure 6 show that SP2TGF and SP3TGF had no toxic effect on the healthy PBMC viability. Peptide conjugates; TGFSP2 and TGFSP3 however, had decreased cell viability compared to the media.
This work paves the way that the TGFaL3-Peptide conjugates produced in this work could be used for targeting the treatment of EGFR-expressing cancer cells (Figure 9), and will build the ground for in vivo investigations of these novel conjugates.
Developing combined therapies or strategies for treating oncological diseases is a promising approach to improving the effectiveness of antitumor treatment [77].
The combination may produce an additive or synergistic effect and reduce each component’s therapeutic dose.
Several studies reported that combination therapies for oncological diseases had been shown that it is more effective and achieved higher overall survival than individual treatment alone in different types of cancer, including breast cancer [78] and lung cancer [79]. Previous investigations have demonstrated robust tumor immunity against murine melanoma obtained by combining DNA vaccination with dual CTLA-4 and PD-1 blockade [80]. Combining the traditional cancer treatment of radiotherapy with immunotherapy can produce synergistic effects because of the specific interaction between the immune system and radiation [81,82]. A recent in vitro study has shown that a combination of telomerase inhibition and NK cell therapy increased breast cancer cell line apoptosis [83].
Our novel superantigen agonists peptides conjugate add one more robust strategy to be used in combination with other recent cancer immune therapy strategies. We propose that our new superantigen-peptide conjugates could be combined with other immunotherapies for effective cancer treatment (Figure 10).

4. Method and Materials

4.1. Reagents, Antibodies, and Kits

SPEA SAg gene with accession number (KY594414) was synthesised by Geneart GmbH (Regensburg, Germany) and previously cloned and expressed in E. coli. SPEA-based peptides were synthesized and supplied by GenScript (Piscataway, NJ, USA). MDA-MB-468 cell line was obtained from ATCC collections, MDA-MB-468 (ATCC® HTB-132™). Fetal bovine serum, L-Glutamine 200 mM (100×), and Dulbecco modified eagle’s medium (DMEM) were purchased from Life Technologies Co., Paisley, UK. Penicillin-streptomycin and Dulbecco’s phosphate buffered saline (DPBS) were purchased from Sigma-Aldrich Chemie GmbH, Taufkirchen, Germany; DAPI (40′,6-diamidino-2-phenylindole) for nucleic acid staining was purchased from Thermo-Fisher Scientific, Waltham, MA, USA. Lonza Trypsin/EDTA (10×) was purchased from SLS life sciences Co., Warwick, UK. Custom ProcartaPlex Multiplex Panel targeting IL-1β, IL-2, IL-6, IFNγ, TNFα, and IL-10 was purchased from Thermo-Fisher Scientific. Human anti-CD3-APC IgG1 and anti-CD25-FITC were purchased from ImmunoTools (Friesoythe, Germany). Anti-EGFR antibody [EP38Y] and Goat Anti-Rabbit IgG H&L (FITC) were from Abcam (Cambridge, UK).

4.2. T-Cell Activation Assay

Peripheral Blood Mononuclear Cells (PBMC) were freshly isolated from human blood using Lymphoprep. Then, 2.5 × 105 cells/well were seeded into a 96-well plate and were treated with 200 ug/mL of peptides, 800 ug/mL endotoxin-free SPEA, or the solvent controls. Cells were harvested after 72 h incubation at 37 °C, 5% CO2, and stained with fluorescently labelled anti-CD3 and anti-CD25 antibodies. T-cell activation analysis was performed using an Accuri C6 flow cytometer (BD Biosciences) by gating for the lymphocyte subset (Figure S1) and measuring the percentage of CD3+ CD25+ cells. The ability of the labelled peptides to activate T-cells was investigated.

4.3. Design of SPEA-Based Tumor-Targeted SAg Peptide

To design tumor-targeted superantigen-based peptides, we fused selected peptides with TGFαL3 either from the N′ or C′ terminal of the SPEA-based peptides (Figure 8). Eight-amino acid flexible linker (GGSGSGGG) was introduced between the peptides and the TGFαL3 (VCHSGYVGARCEHADLL). The designed peptides (Table 1) were chemically synthesized by ProteoGenix (Schiltigheim, France). The peptides were dissolved in Acetonitrile as recommended by ProteoGenix.

4.4. Production of Cytokines

The cell culture supernatant from above was used to measure the level of different cytokine production in response to SPEA-based peptides and peptide-conjugates, SPEA, or controls by Elisa Assay. Custom ProcartaPlex Multiplex Panel Elisa kit targeting IL-1β, IL-2, IL-6, IFNγ, TNFα, and IL-10 was used as instructed in the manual provided with the kit. The Elisa plate was measured using FlexMap 3D instrument (Luminex, Austin, TX, USA).

4.5. Tumor Cell Binding Assay

The binding of tumor-targeted superantigen-based peptides to EGFR on MDA-MB-468 cells was assessed by measuring its ability to block anti-EGFR antibodies from binding to the EGF receptor. To achieve that, 1 × 104 cells were seeded into 96-well plates and incubated overnight at 37 °C, 5% CO2. After removing the media, the cells were fixed with 3.8% formaldehyde solution in PBS for 15 min at RT. The mixture was removed and the cells were washed with PBST and blocked with 3% BSA in PBS for 30 min at RT. The cells were washed again with PBST and incubated with 200 ug/mL of fusion peptides or media control for 2 h at RT. After three washes with PBST, cells were incubated with an Anti-EGFR antibody (Abcam) for 1 h at RT and then washed three times with PBST. Finally, cells were stained with Goat Anti-Rabbit IgG H&L-FITC and DAPI to stain the cell nuclei for 30 min. ArrayScan™ XTI (Thermo Fisher Scientific, Waltham, MA, USA) was used for automated quantitation of DAPI and FITC positives. The binding of peptides to tumor cells was measured by:
% Binding of peptide to tumor cells = (1 − (response of tumor cell treated with superantigen-based tumor-targeted peptides/response of tumor cell treated with anti-EGFR)) × 100

4.6. MTT Assay

The Fusion peptides were evaluated for their toxicity on healthy cells using an MTT assay. PBMCs were isolated and seeded into a 96-well plate (2.5 × 105 cells/well). Cells were incubated with fusion peptides, endotoxin-free SPEA, or the solvent controls for 24 h at 37 °C and under 5% CO2. Cells were then treated with 20 uL of 5 mg/mL MTT solution (Sigma-Aldrich, St. Louis, MO, USA) and incubated for 4 h at 37 °C and under 5% CO2. The precipitate was dissolved with 200 uL of 20% SDS, and the absorbance was measured at 570 nm. The media (untreated cells) sample is considered 100% viable.

4.7. Assessment of Tumor Killing Ability

Human breast carcinoma cell MDA-MB-468 (obtained from the ATCC: ATCC® HTB-132™) were seeded in a 96-well plate (6.4 × 103 cell/well) and incubated at 37 °C in 5% CO2 overnight to allow cell attachment to the well. Cells were treated with 800 ug/mL of endotoxin-free SPEA superantigen, 200 ug/mL SPEA-based peptides, or tumor-targeted superantigen-based peptides. Then, 3.2 × 104 PBMCs were added to wells with tumor cells and SAg derivatives, and incubated for 48 h at 37 °C incubator with 5% CO2. The cell lysate was removed and used for cytokine assessment. Cells were fixed with 3.8% Formaldehyde solution for 10 min and then stained with DAPI. The number of viable tumor cells was measured by ArrayScan™ XTI (Thermo Fisher Scientific, Waltham, MA, USA) where an automated quantitation of DAPI positive nuclei was performed using the target activation module, and the apoptotic nuclei were distinguished and excluded by their collapsed size, higher chromatin intensities, and nuclear fragmentation [84]. The PBMCs were also excluded by size. Cell count is presented as a percentage of tumor + PBMCs (TP) will count.

4.8. Statistical Analyses

Data are presented as mean ± standard error of the mean (S.E.M) of “n” observations. All graphs were constructed using GraphPad Prism 5 software (San Diego, CA, USA). Statistical analysis was performed using Student’s t-test as appropriate. p values < 0.05 were considered statistically significant.

5. Conclusions

Our work produced novel superantigen SPEA-peptide agonists and SPEA-peptide agonist-EGF conjugates for the targeted treatment of EGFR-expressing cancers. We showed that these peptides and their conjugates produced different cytokines and effectively killed cancer cells, while peptides and peptide-conjugates have no effect on the healthy PBMCs. The great advantage of our work is the ability to carry out mutation studies on our isolated peptide agonists to produce more effective superantigen peptides. Further, to fuse our peptides with other cancer-specific moieties to target treating other cancers. This study would pave the way for in vivo investigation on animals and humans.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms241310507/s1.

Author Contributions

Conceptualization, S.K.G.; Data curation, S.S.B. and S.K.G.; Formal analysis, S.S.B.; Investigation, S.S.B.; Methodology, S.S.B.; Supervision, S.K.G.; Validation, S.S.B. and S.K.G.; Writing—original draft, S.S.B.; Writing—review and editing, S.S.B. and S.K.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was approved by the Anti-Doping Lab-Qatar Institutional Review Board, Ethical approval number: E2017000205.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All data are available with the authors and will be provided if requested.

Acknowledgments

The authors would like to thank the anti-doping laboratory-Qatar for the help and support during this work. We also would like to thank QU Health for covering publication fee for this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jeanson, A.; Tomasini, P.; Souquet-Bressand, M.; Brandone, N.; Boucekine, M.; Grangeon, M.; Chaleat, S.; Khobta, N.; Milia, J.; Mhanna, L.; et al. Efficacy of Immune Checkpoint Inhibitors in KRAS-Mutant Non-Small Cell Lung Cancer (NSCLC). J. Thorac. Oncol. 2019, 14, 1095–1101. [Google Scholar] [CrossRef]
  2. Kuske, M.; Rauschenberg, R.; Garzarolli, M.; Meredyth-Stewart, M.; Beissert, S.; Troost, E.G.C.; Glitza, O.I.C.; Meier, F. Melanoma Brain Metastases: Local Therapies, Targeted Therapies, Immune Checkpoint Inhibitors and Their Combinations—Chances and Challenges. Am. J. Clin. Dermatol. 2018, 19, 529–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lehrer, E.J.; Peterson, J.; Brown, P.D.; Sheehan, J.P.; Quiñones-Hinojosa, A.; Zaorsky, N.G.; Trifiletti, D.M. Treatment of brain metastases with stereotactic radiosurgery and immune checkpoint inhibitors: An international meta-analysis of individual patient data. Radiother. Oncol. 2019, 130, 104–112. [Google Scholar] [CrossRef] [PubMed]
  4. Marrone, K.; Ying, W.; Naidoo, J. Immune-Related Adverse Events from Immune Checkpoint Inhibitors. Clin. Pharmacol. Ther. 2016, 100, 242–251. [Google Scholar] [CrossRef] [PubMed]
  5. Mazieres, J.; Drilon, A.; Lusque, A.B.; Mhanna, L.; Cortot, A.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann. Oncol. 2019, 30, 1321–1328. [Google Scholar] [CrossRef]
  6. Mhanna, L.; Guibert, N.; Milia, J.; Mazieres, J. When to Consider Immune Checkpoint Inhibitors in Oncogene-Driven Non-Small Cell Lung Cancer? Curr. Treat. Options Oncol. 2019, 20, 60. [Google Scholar] [CrossRef] [Green Version]
  7. Robin, T.P.; Breeze, R.E.; Smith, D.E.; Rusthoven, C.G.; Lewis, K.D.; Gonzalez, R.; Brill, A.; Saiki, R.; Stuhr, K.; Gaspar, L.E.; et al. Immune checkpoint inhibitors and radiosurgery for newly diagnosed melanoma brain metastases. J. Neuro-Oncol. 2018, 140, 55–62. [Google Scholar] [CrossRef]
  8. Upadhrasta, S.; Elias, H.; Patel, K.; Zheng, L. Managing cardiotoxicity associated with immune checkpoint inhibitors. Chronic Dis. Transl. Med. 2019, 5, 6–14. [Google Scholar] [CrossRef]
  9. Zhang, Y.; Yao, Q.; Pan, Y.; Fang, X.; Xu, H.; Zhao, T.; Zhu, G.; Jiang, T.; Li, S.; Cao, H. Efficacy and Safety of PD-1/PD-L1 Checkpoint Inhibitors versus Anti-PD-1/PD-L1 Combined with Other Therapies for Tumors: A Systematic Review. Cancers 2023, 15, 682. [Google Scholar] [CrossRef]
  10. Zeng, H.; Xu, Q.; Wang, J.; Xu, X.; Luo, J.; Zhang, L.; Luo, C.; Ying, J.; Li, J. The effect of anti-PD-1/PD-L1 antibodies combined with VEGF receptor tyrosine kinase inhibitors versus bevacizumab in unresectable hepatocellular carcinoma. Front. Immunol. 2023, 14, 1073133. [Google Scholar] [CrossRef]
  11. Sjöstrand, M.; Sadelain, M. Driving CARs to new places: Locally produced BCMA CAR T cells to treat multiple myeloma. Haematologica 2023. [Google Scholar] [CrossRef]
  12. Dai, H.-P.; Kong, D.-Q.; Shen, H.-J.; Cui, W.; Wang, Q.; Li, Z.; Yin, J.; Kang, L.-Q.; Yu, L.; Wu, D.-P.; et al. CAR-T cell therapy followed by allogenic hematopoietic stem cell transplantation yielded comparable outcome between Ph like ALL and other high-risk ALL. Biomark. Res. 2023, 11, 19. [Google Scholar] [CrossRef]
  13. Leick, M.B.; Maus, M.V. Multiomics STEP up in correlative analysis of response to CAR T cells. Nat. Rev. Clin. Oncol. 2023, 20, 285–286. [Google Scholar] [CrossRef]
  14. Rini, S.S.; Kambayana, G. Autologous CD19-Targeted Chimeric Antigen Receptor (CAR)T-Cells as the Future of Systemic Lupus Erythematosus Treatment. Curr. Rheumatol. Rev. 2023, 19, 260–269. [Google Scholar] [CrossRef]
  15. Adabi, N.; Pordel, S.; Rezaee, M.A.; Shobeiri, F.S.; Shobeiri, S.S. Application of CAR-T cell technology in autoimmune diseases and human immunodeficiency virus infection treatment. J. Gene Med. 2023, 25, e3484. [Google Scholar] [CrossRef]
  16. Zhang, R.; Liu, Q.; Zhou, S.; He, H.; Zhao, M.; Ma, W. Mesenchymal stem cell suppresses the efficacy of CAR-T toward killing lymphoma cells by modulating the microenvironment through stanniocalcin-1. Elife 2023, 12, e82934. [Google Scholar] [CrossRef]
  17. Zhang, X.; Wu, J.; Qiao, L.; Chen, L.; Chen, C.; Zhang, H.; Luo, R.; Xiao, Y. Case report: Cryoablation as a novel bridging strategy prior to CAR-T cell therapy for B cell malignancies with bulky disease. Front. Oncol. 2023, 13, 1008828. [Google Scholar] [CrossRef]
  18. Nicod, C.; da Rocha, M.N.; Warda, W.; Roussel, X.; Haderbache, R.; Seffar, E.; Trad, R.; Bouquet, L.; Goncalves, M.; Bosdure, L.; et al. CAR-T cells targeting IL-1RAP produced in a closed semiautomatic system are ready for the first phase I clinical investigation in humans. Curr. Res. Transl. Med. 2023, 71, 103385. [Google Scholar] [CrossRef]
  19. Harrer, D.C.; Dörrie, J.; Schaft, N. CARs and Drugs: Pharmacological Ways of Boosting CAR-T-Cell Therapy. Int. J. Mol. Sci. 2023, 24, 2342. [Google Scholar] [CrossRef]
  20. Asokan, S.; Cullin, N.; Stein-Thoeringer, C.K.; Elinav, E. CAR-T Cell Therapy and the Gut Microbiota. Cancers 2023, 15, 794. [Google Scholar] [CrossRef]
  21. Chen, Y.-J.; Abila, B.; Kamel, Y.M. CAR-T: What Is Next? Cancers 2023, 15, 663. [Google Scholar] [CrossRef] [PubMed]
  22. Li, H.; Llera, A.; Malchiodi, E.L.; Mariuzza, R.A. The structural basis of t cell activation by superantigens. Annu. Rev. Immunol. 1999, 17, 435–466. [Google Scholar] [CrossRef] [PubMed]
  23. Mehindate, K.; Thibodeau, J.; Dohlsten, M.; Kalland, T.; Sékaly, R.P.; Mourad, W. Cross-linking of major histocompatibility complex class II molecules by staphylococcal enterotoxin A superantigen is a requirement for inflammatory cytokine gene expression. J. Exp. Med. 1995, 182, 1573–1577. [Google Scholar] [CrossRef] [Green Version]
  24. Terman, D.S.; Serier, A.; Dauwalder, O.; Badiou, C.; Dutour, A.; Thomas, D.; Brun, V.; Bienvenu, J.; Etienne, J.; Vandenesch, F.; et al. Staphylococcal entertotoxins of the enterotoxin Gene cluster (egcSEs) induce nitrous oxide- and cytokine dependent tumor cell apoptosis in a broad panel of human tumor cells. Front. Cell. Infect. Microbiol. 2013, 3, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ejtehadifar, M.; Halabian, R.; Ghazavi, A.; Khansarinejad, B.; Mosayebi, G.; Fooladi, A.A.I. Bone marrow—mesenchymal stem cells impact on the U937 cells in the presence of staphylococcal enterotoxin B (SEB). Clin. Exp. Pharmacol. Physiol. 2018, 45, 849–858. [Google Scholar] [CrossRef]
  26. Hawkins, R.E.; Gore, M.; Shparyk, Y.; Bondar, V.; Gladkov, O.; Ganev, T.; Harza, M.; Polenkov, S.; Bondarenko, I.; Karlov, P.; et al. A Randomized Phase II/III Study of Naptumomab Estafenatox + IFNα versus IFNα in Renal Cell Carcinoma: Final Analysis with Baseline Biomarker Subgroup and Trend Analysis. Clin. Cancer Res. 2016, 22, 3172–3181. [Google Scholar] [CrossRef] [Green Version]
  27. Superantigens, superantigen-like proteins and superantigen derivatives for cancer treatment. Eur. Rev. Med. Pharmacol. 2021, 25, 1622–1630. [CrossRef]
  28. Golob-Urbanc, A.; Rajčević, U.; Strmšek, Z.; Jerala, R. Design of split superantigen fusion proteins for cancer immunotherapy. J. Biol. Chem. 2019, 294, 6294–6305. [Google Scholar] [CrossRef] [Green Version]
  29. Liu, X.; Zeng, L.; Zhao, Z.; Xie, Y.; Wang, S.; Zhang, J.; He, Y.; Zou, Z.; Zhang, J.; Tao, A. Construction, Expression, and Characterization of rSEA-EGF and In Vitro Evaluation of its Antitumor Activity Against Nasopharyngeal Cancer. Technol. Cancer Res. Treat. 2018, 17, 1533033818762910. [Google Scholar] [CrossRef] [Green Version]
  30. Zhang, G.; Xu, M.; Song, Y.; Su, Z.; Zhang, H.; Zhang, C. TNF-α produced by SEC2 mutant (SAM-3)-activated human T cells induces apoptosis of HepG2 cells. Appl. Microbiol. Biotechnol. 2016, 100, 2677–2684. [Google Scholar] [CrossRef]
  31. Eisen, T.; Hedlund, G.; Forsberg, G.; Hawkins, R. Naptumomab Estafenatox: Targeted Immunotherapy with a Novel Immunotoxin. Curr. Oncol. Rep. 2014, 16, 370. [Google Scholar] [CrossRef] [Green Version]
  32. Dohlsten, M.; Abrahmsén, L.; Björk, P.; Lando, P.A.; Hedlund, G.; Forsberg, G.; Brodin, T.; Gascoigne, N.R.; Förberg, C.; Lind, P. Monoclonal antibody-superantigen fusion proteins: Tumor-specific agents for T-cell-based tumor therapy. Proc. Natl. Acad. Sci. USA 1994, 91, 8945–8949. [Google Scholar] [CrossRef] [Green Version]
  33. Dohlsten, M.; Hedlund, G.; Akerblom, E.; Lando, P.A.; Kalland, T. Monoclonal antibody-targeted superantigens: A different class of anti-tumor agents. Proc. Natl. Acad. Sci. USA 1991, 88, 9287–9291. [Google Scholar] [CrossRef] [Green Version]
  34. Dohlsten, M.; Hansson, J.; Ohlsson, L.; Litton, M.; Kalland, T. Antibody-targeted superantigens are potent inducers of tumor-infiltrating T lymphocytes in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9791–9795. [Google Scholar] [CrossRef] [Green Version]
  35. Allen, T.M. Ligand-targeted therapeutics in anticancer therapy. Nat. Rev. Cancer 2002, 2, 750–763. [Google Scholar] [CrossRef]
  36. Wu, H.-C.; Chang, D.-K.; Huang, C.-T. Targeted Therapy for Cancer. J. Cancer Mol. 2006, 2, 57–66. [Google Scholar]
  37. Xu, Q.; Zhang, X.; Yue, J.; Liu, C.; Cao, C.; Zhong, H.; Ma, Q. Human TGFalpha-derived peptide TGFalphaL3 fused with superantigen for immunotherapy of EGFR-expressing tumours. BMC Biotechnol. 2010, 10, 91. [Google Scholar] [CrossRef] [Green Version]
  38. Grupka, N.L.; Lear-Kaul, K.C.; Kleinschmidt-DeMasters, B.K.; Singh, M. Epidermal growth factor receptor status in breast cancer metastases to the central nervous system. Comparison with HER-2/neu status. Arch. Pathol. Lab. Med. 2004, 128, 974–979. [Google Scholar] [CrossRef]
  39. Dua, R.; Zhang, J.; Nhonthachit, P.; Penuel, E.; Petropoulos, C.; Parry, G. EGFR over-expression and activation in high HER2, ER negative breast cancer cell line induces trastuzumab resistance. Breast Cancer Res. Treat. 2010, 122, 685–697. [Google Scholar] [CrossRef]
  40. Aziz, S.A.; Pervez, S.; Khan, S.; Kayani, N.; Rahbar, M.H. Epidermal growth factor receptor (EGFR) as a prognostic marker: An immunohistochemical study on 315 consecutive breast carcinoma patients. J. Pak. Med. Assoc. 2002, 52, 104–110. [Google Scholar]
  41. Agustoni, F.; Suda, K.; Yu, H.; Ren, S.; Rivard, C.J.; Ellison, K.; Caldwell, C.; Rozeboom, L.; Brovsky, K.; Hirsch, F.R. EGFR-directed monoclonal antibodies in combination with chemotherapy for treatment of non-small-cell lung cancer: An updated review of clinical trials and new perspectives in biomarkers analysis. Cancer Treat. Rev. 2018, 72, 15–27. [Google Scholar] [CrossRef] [PubMed]
  42. Imani-Fooladi, A.A.; Yousefi, F.; Mousavi, S.F.; Amani, J. In Silico Design and Analysis of TGFalphaL3-SEB Fusion Protein as "a New Antitumor Agent" Candidate by Ligand-Targeted Superantigens Technique. Iran. J. Cancer Prev. 2014, 7, 152–164. [Google Scholar] [PubMed]
  43. Yousefi, F.; Mousavi, S.F.; Siadat, S.D.; Aslani, M.M.; Amani, J.; Rad, H.S.; Fooladi, A.A.I. Preparation and In Vitro Evaluation of Antitumor Activity of TGFαL3-SEB as a Ligand-Targeted Superantigen. Technol. Cancer Res. Treat. 2016, 15, 215–226. [Google Scholar] [CrossRef] [PubMed]
  44. Yousefi, F.; Siadat, S.D.; Saraji, A.A.; Hesaraki, S.; Aslani, M.M.; Mousavi, S.F.; Fooladi, A.A.I. Tagging staphylococcal enterotoxin B (SEB) with TGFaL3 for breast cancer therapy. Tumor Biol. 2016, 37, 5305–5316. [Google Scholar] [CrossRef] [PubMed]
  45. Miethke, T.; Duschek, K.; Wahl, C.; Heeg, K.; Wagner, H. Pathogenesis of the toxic shock syndrome: T cell mediated lethal shock caused by the superantigen TSST-1. Eur. J. Immunol. 1993, 23, 1494–1500. [Google Scholar] [CrossRef]
  46. Dama, P.; Ledoux, D.; Nys, M.; Vrindts, Y.; Groote, D.D.E.; Franchimont, P.; Lamy, M. Cytokine Serum Level During Severe Sepsis in Human IL-6 as a Marker of Severity. Ann. Surg. 1992, 215, 356–362. [Google Scholar] [CrossRef]
  47. Waage, A.; Brandtzaeg, P.; Halstensen, A.; Kierulf, P.; Espevik, T. The complex pattern of cytokines in serum from patients with meningococcal septic shock. Association between interleukin 6, interleukin 1, and fatal outcome. J. Exp. Med. 1989, 169, 333–338. [Google Scholar] [CrossRef]
  48. Hansson, J.; Ohlsson, L.; Persson, R.; Andersson, G.; Ilback, N.G.; Litton, M.J.; Kalland, T.; Dohlsten, M. Genetically engineered superantigens as tolerable antitumor agents. Proc. Natl. Acad. Sci. USA 1997, 94, 2489–2494. [Google Scholar] [CrossRef] [Green Version]
  49. Abrahmsen, L.; Dohlsten, M.; Segren, S.; Bjork, P.; Jonsson, E.; Kalland, T. Characterization of two distinct MHC class II binding sites in the superantigen staphylococcal enterotoxin A. EMBO J. 1995, 14, 2978–2986. [Google Scholar] [CrossRef]
  50. Dohlsten, M.; Lando, P.A.; Bjork, P.; Abrahmsen, L.; Ohlsson, L.; Lind, P.; Kalland, T. Immunotherapy of human colon cancer by antibody-targeted superantigens. Cancer Immunol. Immunother. CII 1995, 41, 162–168. [Google Scholar] [CrossRef]
  51. Liu, T.; Li, L.; Yin, L.; Yu, H.; Jing, H.; Liu, Y.; Kong, C.; Xu, M. Superantigen staphylococcal enterotoxin C1 inhibits the growth of bladder cancer. Biosci. Biotechnol. Biochem. 2017, 81, 1741–1746. [Google Scholar] [CrossRef] [Green Version]
  52. Shaw, D.M.; Connolly, N.B.; Patel, P.M.; Kilany, S.; Hedlund, G.; Nordle, O.; Forsberg, G.; Zweit, J.; Stern, P.L.; Hawkins, R.E. A phase II study of a 5T4 oncofoetal antigen tumour-targeted superantigen (ABR-214936) therapy in patients with advanced renal cell carcinoma. Br. J. Cancer 2007, 96, 567–574. [Google Scholar] [CrossRef] [Green Version]
  53. Søgaard, M.; Hansson, J.; Litton, M.J.; Ohlsson, L.; Rosendahl, A.; Lando, P.A.; Antonsson, P.; Kalland, T.; Dohlsten, M. Antibody-targeted superantigens in cancer immunotherapy. Immunotechnology 1996, 2, 151–162. [Google Scholar] [CrossRef]
  54. Bashraheel, S.S.; AlQahtani, A.D.; Rashidi, F.B.; Al-Sulaiti, H.; Domling, A.; Orie, N.N.; Goda, S.K. Studies on vascular response to full superantigens and superantigen derived peptides: Possible production of novel superantigen variants with less vasodilation effect for tolerable cancer immunotherapy. Biomed. Pharmacother. 2019, 115, 108905. [Google Scholar] [CrossRef]
  55. Arad, G.; Levy, R.; Nasie, I.; Hillman, D.; Rotfogel, Z.; Barash, U.; Supper, E.; Shpilka, T.; Minis, A.; Kaempfer, R. Binding of Superantigen Toxins into the CD28 Homodimer Interface Is Essential for Induction of Cytokine Genes That Mediate Lethal Shock. PLoS Biol. 2011, 9, e1001149. [Google Scholar] [CrossRef] [Green Version]
  56. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in Inflammation, Immunity, and Disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef]
  57. Fisher, D.T.; Appenheimer, M.M.; Evans, S.S. The two faces of IL-6 in the tumor microenvironment. Semin. Immunol. 2014, 26, 38–47. [Google Scholar] [CrossRef] [Green Version]
  58. Sgagias, M.K.; Kasid, A.; Danforth, D.N. Interleukin-1α and Tumor Necrosis Factor-α (TNFα) Inhibit Growth and Induce TNF Messenger RNA in MCF-7 Human Breast Cancer Cells. Mol. Endocrinol. 1991, 5, 1740–1747. [Google Scholar] [CrossRef] [Green Version]
  59. Maund, S.L.; Barclay, W.W.; Hover, L.D.; Axanova, L.S.; Sui, G.; Hipp, J.D.; Fleet, J.C.; Thorburn, A.; Cramer, S.D. Interleukin-1α Mediates the Antiproliferative Effects of 1,25-Dihydroxyvitamin D3 in Prostate Progenitor/Stem Cells. Cancer Res. 2011, 71, 5276–5286. [Google Scholar] [CrossRef] [Green Version]
  60. Maund, S.L.; Shi, L.; Cramer, S.D. A Role for Interleukin-1 Alpha in the 1,25 Dihydroxyvitamin D3 Response in Mammary Epithelial Cells. PLoS ONE 2013, 8, e81367. [Google Scholar] [CrossRef] [Green Version]
  61. Le, T.T.; Skak, K.; Schroder, K.; Schroder, W.A.; Boyle, G.M.; Pierce, C.J.; Suhrbier, A. IL-1 Contributes to the Anti-Cancer Efficacy of Ingenol Mebutate. PLoS ONE 2016, 11, e0153975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Oft, M. IL-10: Master Switch from Tumor-Promoting Inflammation to Antitumor Immunity. Cancer Immunol. Res. 2014, 2, 194–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Langer, R. Drug delivery and targeting. Nature 1998, 392 (Suppl. S6679), 5–10. [Google Scholar] [PubMed]
  64. Brannon-Peppas, L.; Blanchette, J.O. Nanoparticle and targeted systems for cancer therapy. Adv. Drug Deliv. Rev. 2004, 56, 1649–1659. [Google Scholar] [CrossRef]
  65. Chari, R.V.J.; Miller, M.L.; Widdison, W.C. Antibody-Drug Conjugates: An Emerging Concept in Cancer Therapy. Angew. Chem. Int. Ed. 2014, 53, 3796–3827. [Google Scholar] [CrossRef]
  66. Sharma, S.K.; Bagshawe, K.D. Antibody Directed Enzyme Prodrug Therapy (ADEPT): Trials and tribulations. Adv. Drug Deliv. Rev. 2017, 118, 2–7. [Google Scholar] [CrossRef]
  67. MacKay, J.A.; Chen, M.; McDaniel, J.R.; Liu, W.; Simnick, A.J.; Chilkoti, A. Self-assembling chimeric polypeptide–doxorubicin conjugate nanoparticles that abolish tumours after a single injection. Nat. Mater. 2009, 8, 993–999. [Google Scholar] [CrossRef] [Green Version]
  68. Bazley, L.A.; Gullick, W.J. The epidermal growth factor receptor family. Endocr.-Relat. Cancer 2005, 12, S17–S27. [Google Scholar] [CrossRef]
  69. Arena, S.; Bellosillo, B.; Siravegna, G.; Martínez, A.; Cañadas, I.; Lazzari, L.; Ferruz, N.; Russo, M.; Misale, S.; González, I.; et al. Emergence of Multiple EGFR Extracellular Mutations during Cetuximab Treatment in Colorectal Cancer. Clin. Cancer Res. 2015, 21, 2157–2166. [Google Scholar] [CrossRef] [Green Version]
  70. Park, H.S.; Jang, M.H.; Kim, E.J.; Kim, H.J.; Lee, H.J.; Kim, Y.J.; Kim, J.H.; Kang, E.; Kim, S.-W.; Kim, I.A.; et al. High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. Mod. Pathol. 2014, 27, 1212–1222. [Google Scholar] [CrossRef] [Green Version]
  71. Azuma, K.; Ota, K.; Kawahara, A.; Hattori, S.; Iwama, E.; Harada, T.; Matsumoto, K.; Takayama, K.; Takamori, S.; Kage, M.; et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann. Oncol. 2014, 25, 1935–1940. [Google Scholar] [CrossRef]
  72. Benelli, R.; Costa, D.; Salvini, L.; Tardito, S.; Tosetti, F.; Villa, F.; Zocchi, M.R.; Poggi, A. Targeting of colorectal cancer organoids with zoledronic acid conjugated to the anti-EGFR antibody cetuximab. J. Immunother. Cancer 2022, 10, e005660. [Google Scholar] [CrossRef]
  73. Lu, Y.; Huang, J.; Li, F.; Wang, Y.; Ding, M.; Zhang, J.; Yin, H.; Zhang, R.; Ren, X. EGFR-specific single-chain variable fragment antibody-conjugated Fe3O4/Au nanoparticles as an active MRI contrast agent for NSCLC. Magn. Reson. Mater. Physics Biol. Med. 2021, 34, 581–591. [Google Scholar] [CrossRef]
  74. Wu, S.-C.; Chen, Y.-J.; Wang, H.-C.; Chou, M.-Y.; Chang, T.-Y.; Yuan, S.-S.; Chen, C.-Y.; Hou, M.-F.; Hsu, J.T.-A.; Wang, Y.-M. Bispecific Antibody Conjugated Manganese-Based Magnetic Engineered Iron Oxide for Imaging of HER2/neu- and EGFR-Expressing Tumors. Theranostics 2016, 6, 118–130. [Google Scholar] [CrossRef]
  75. Leung, S.L.; Zha, Z.; Cohn, C.; Dai, Z.; Wu, X. Anti-EGFR antibody conjugated organic–inorganic hybrid lipid nanovesicles selectively target tumor cells. Colloids Surf. B Biointerfaces 2014, 121, 141–149. [Google Scholar] [CrossRef]
  76. Maleki, F.; Sadeghifard, N.; Hosseini, H.M.; Bakhtiyari, S.; Goleij, Z.; Behzadi, E.; Sedighian, H.; Fooladi, A.A.I. Growth-inhibitory effects of TGFαL3-SEB chimeric protein on colon cancer cell line. Biomed. Pharmacother. 2019, 110, 190–196. [Google Scholar] [CrossRef]
  77. Bashraheel, S.S.; Domling, A.; Goda, S.K. Update on targeted cancer therapies, single or in combination, and their fine tuning for precision medicine. Biomed. Pharmacother. 2020, 125, 110009. [Google Scholar] [CrossRef]
  78. Brandão, M.; Caparica, R.; Eiger, D.; de Azambuja, E. Biomarkers of response and resistance to PI3K inhibitors in estrogen receptor-positive breast cancer patients and combination therapies involving PI3K inhibitors. Ann. Oncol. 2019, 30, x27–x42. [Google Scholar] [CrossRef] [Green Version]
  79. Brunetti, O.; Badalamenti, G.; De Summa, S.; Calabrese, A.; Argentiero, A.; Fucci, L.; Longo, V.; Galetta, D.; Perrotti, P.M.S.; Pinto, R.; et al. Molecular Characterization of a Long-Term Survivor Double Metastatic Non-Small Cell Lung Cancer and Pancreatic Ductal Adenocarcinoma Treated with Gefitinib in Combination with Gemcitabine Plus Nab-Paclitaxel and mFOLFOX6 as First and Second Line Therapy. Cancers 2019, 11, 749. [Google Scholar] [CrossRef] [Green Version]
  80. Kos, S.; Lopes, A.; Preat, V.; Cemazar, M.; Tratar, U.L.; Ucakar, B.; Vanvarenberg, K.; Sersa, G.; Vandermeulen, G. Intradermal DNA vaccination combined with dual CTLA-4 and PD-1 blockade provides robust tumor immunity in murine melanoma. PLoS ONE 2019, 14, e0217762. [Google Scholar] [CrossRef] [Green Version]
  81. Jiang, W.; Chan, C.K.; Weissman, I.L.; Kim, B.Y.; Hahn, S.M. Immune Priming of the Tumor Microenvironment by Radiation. Trends Cancer 2016, 2, 638–645. [Google Scholar] [CrossRef] [PubMed]
  82. Frey, B.; Rückert, M.; Deloch, L.; Rühle, P.F.; Derer, A.; Fietkau, R.; Gaipl, U.S. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol. Rev. 2017, 280, 231–248. [Google Scholar] [CrossRef] [PubMed]
  83. Mazloumi, Z.; Rafat, A.; Asl, K.D.; Charoudeh, H.N. A combination of telomerase inhibition and NK cell therapy increased breast cancer cell line apoptosis. Biochem. Biophys. Res. Commun. 2023, 640, 50–55. [Google Scholar] [CrossRef] [PubMed]
  84. Ismail, H.M.; Zamani, S.; Elrayess, M.A.; Kafienah, W.; Younes, H.M. New Three-Dimensional Poly(decanediol-co-tricarballylate) Elastomeric Fibrous Mesh Fabricated by Photoreactive Electrospinning for Cardiac Tissue Engineering Applications. Polymers 2018, 10, 455. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Percentage of CD3+ CD25+ Cells (activated T-cells) in response to treatment with SPEA-agonist peptides. (A) Data of 20 SPEA-based peptides presented as mean percentage of CD3+ CD25+ cells ± standard error of the mean (S.E.M) of 3 independent experiments (n = 3), * p ˂ 0.05, ** p ˂ 0.01, compared to the corresponding solvent control. (B) Representative flow cytometric dot-plots CD3/CD25 staining in which cells were pre-gated for lymphocytes.
Figure 1. Percentage of CD3+ CD25+ Cells (activated T-cells) in response to treatment with SPEA-agonist peptides. (A) Data of 20 SPEA-based peptides presented as mean percentage of CD3+ CD25+ cells ± standard error of the mean (S.E.M) of 3 independent experiments (n = 3), * p ˂ 0.05, ** p ˂ 0.01, compared to the corresponding solvent control. (B) Representative flow cytometric dot-plots CD3/CD25 staining in which cells were pre-gated for lymphocytes.
Ijms 24 10507 g001
Figure 2. Cytokine production in response to SPEA-based peptides in a mixed culture of PBMCs and MDA-MB-468 cells. (A) concentration of IL-6, (B) concentration of IL-10, (C) concentration of TNFα, and (D) concentration of IL-1β. Data presented as a mean concentration in pg/mL ± standard error of the mean (S.E.M) of 3 experiments (n = 3).* p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with the corresponding solvent (H2O for SP2, SP3, and PBS for SPEA).
Figure 2. Cytokine production in response to SPEA-based peptides in a mixed culture of PBMCs and MDA-MB-468 cells. (A) concentration of IL-6, (B) concentration of IL-10, (C) concentration of TNFα, and (D) concentration of IL-1β. Data presented as a mean concentration in pg/mL ± standard error of the mean (S.E.M) of 3 experiments (n = 3).* p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with the corresponding solvent (H2O for SP2, SP3, and PBS for SPEA).
Ijms 24 10507 g002
Figure 3. Cytokine production in response to SPEA-based tumor-targeted peptides in a mixed culture of PBMCs and MDA-MB-468 cells. (A) concentration of IL-6, (B) concentration of IL-10, (C) concentration of TNFα and (D) concentration of IL-1β. Data presented as a mean concentration in pg/mL ± standard error of the mean (S.E.M) of 3 experiments (n = 3). * p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with the corresponding solvent (ACN for Fusion peptides and PBS for SPEA).
Figure 3. Cytokine production in response to SPEA-based tumor-targeted peptides in a mixed culture of PBMCs and MDA-MB-468 cells. (A) concentration of IL-6, (B) concentration of IL-10, (C) concentration of TNFα and (D) concentration of IL-1β. Data presented as a mean concentration in pg/mL ± standard error of the mean (S.E.M) of 3 experiments (n = 3). * p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with the corresponding solvent (ACN for Fusion peptides and PBS for SPEA).
Ijms 24 10507 g003
Figure 4. SPEA-based tumor-targeted peptides are binding to EGFR. Data of SPEA-based peptides presented as mean percentage of tumor cells treated with anti-EGFR antibody ± standard error of the mean (S.E.M) of 3 experiments (n = 3), * p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with full superantigen.
Figure 4. SPEA-based tumor-targeted peptides are binding to EGFR. Data of SPEA-based peptides presented as mean percentage of tumor cells treated with anti-EGFR antibody ± standard error of the mean (S.E.M) of 3 experiments (n = 3), * p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with full superantigen.
Ijms 24 10507 g004
Figure 5. Effect of SPEA-based tumor-targeted peptides on PBMCs viability. Data presented as mean absorbance ± standard error of the mean (S.E.M) of 3 experiments (n = 3). ** p ˂ 0.01, *** p ˂ 0.001, compared to media control).
Figure 5. Effect of SPEA-based tumor-targeted peptides on PBMCs viability. Data presented as mean absorbance ± standard error of the mean (S.E.M) of 3 experiments (n = 3). ** p ˂ 0.01, *** p ˂ 0.001, compared to media control).
Ijms 24 10507 g005
Figure 6. Effect of different SPEA-based peptides on MDA-MB-468 cell viability in a mixed culture with PBMCs. (A) Data presented as the mean of DAPI positive nuclei as a percentage of TP ± standard error of the mean (S.E.M) of 6 independent experiments (n = 6). * p ˂ 0.05, *** p ˂ 0.001, compared to treatment with corresponding solvent. (B) Representative Images of the DAPI-stained nuclei of viable cells after incubation with the different peptides. The blue color is DAPI positive MDA-MB-468 cells, whereas the orange color is for the excluded PBMCs.
Figure 6. Effect of different SPEA-based peptides on MDA-MB-468 cell viability in a mixed culture with PBMCs. (A) Data presented as the mean of DAPI positive nuclei as a percentage of TP ± standard error of the mean (S.E.M) of 6 independent experiments (n = 6). * p ˂ 0.05, *** p ˂ 0.001, compared to treatment with corresponding solvent. (B) Representative Images of the DAPI-stained nuclei of viable cells after incubation with the different peptides. The blue color is DAPI positive MDA-MB-468 cells, whereas the orange color is for the excluded PBMCs.
Ijms 24 10507 g006
Figure 7. Effect of different SPEA-based tumor-targeted peptides on MDA-MB-468 cell viability in a mixed culture with PBMCs. Data presented as the mean of DAPI positive nuclei as a percentage of TP ± standard error of the mean (S.E.M) of 6 independent experiments (n = 6). * p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with the solvent (ACN).
Figure 7. Effect of different SPEA-based tumor-targeted peptides on MDA-MB-468 cell viability in a mixed culture with PBMCs. Data presented as the mean of DAPI positive nuclei as a percentage of TP ± standard error of the mean (S.E.M) of 6 independent experiments (n = 6). * p ˂ 0.05, ** p ˂ 0.01, *** p ˂ 0.001, compared to treatment with the solvent (ACN).
Ijms 24 10507 g007
Figure 8. Synthesis of superantigen agonist peptide EGF conjugated at either N-terminal or C-terminal.
Figure 8. Synthesis of superantigen agonist peptide EGF conjugated at either N-terminal or C-terminal.
Ijms 24 10507 g008
Figure 9. Targeted cancer treatment using superantigen agonist conjugated peptide, modified [77].
Figure 9. Targeted cancer treatment using superantigen agonist conjugated peptide, modified [77].
Ijms 24 10507 g009
Figure 10. Possible combinations of other immunotherapy techniques with our newly superantigen agonist peptide conjugates to overcome drug resistance.
Figure 10. Possible combinations of other immunotherapy techniques with our newly superantigen agonist peptide conjugates to overcome drug resistance.
Ijms 24 10507 g010
Table 1. SPEA-based tumor-targeted SAg peptide Sequence.
Table 1. SPEA-based tumor-targeted SAg peptide Sequence.
SP2TGFHRSSLVKNLQNIYFLYEGDPVTHENVKSVDQLLSHDLIYNGGSGSGGGVCHSGYVGARCEHADLL
TGFSP2VCHSGYVGARCEHADLLGGSGSGGGHRSSLVKNLQNIYFLYEGDPVTHENVKSVDQLLSHDLIYN
SP3TGFNIYFLYEGDPVTHENVKSVDQLLSHDLIYNVSGPNYDKLKGGSGSGGGVCHSGYVGARCEHADLL
TGFSP3VCHSGYVGARCEHADLLGGSGSGGGNIYFLYEGDPVTHENVKSVDQLLSHDLIYNVSGPNYDKLK
SP15TGFVTAQELDYKVRKYLTDNKQLYTNGPSKYETGYIKFIPKNKGGSGSGGGVCHSGYVGARCEHADLL
TGFSP15VCHSGYVGARCEHADLLGGSGSGGGVTAQELDYKVRKYLTDNKQLYTNGPSKYETGYIKFIPKNK
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bashraheel, S.S.; Goda, S.K. Novel SPEA Superantigen Peptide Agonists and Peptide Agonist-TGFαL3 Conjugate. In Vitro Study of Their Growth-Inhibitory Effects for Targeted Cancer Immunotherapy. Int. J. Mol. Sci. 2023, 24, 10507. https://doi.org/10.3390/ijms241310507

AMA Style

Bashraheel SS, Goda SK. Novel SPEA Superantigen Peptide Agonists and Peptide Agonist-TGFαL3 Conjugate. In Vitro Study of Their Growth-Inhibitory Effects for Targeted Cancer Immunotherapy. International Journal of Molecular Sciences. 2023; 24(13):10507. https://doi.org/10.3390/ijms241310507

Chicago/Turabian Style

Bashraheel, Sara S., and Sayed K. Goda. 2023. "Novel SPEA Superantigen Peptide Agonists and Peptide Agonist-TGFαL3 Conjugate. In Vitro Study of Their Growth-Inhibitory Effects for Targeted Cancer Immunotherapy" International Journal of Molecular Sciences 24, no. 13: 10507. https://doi.org/10.3390/ijms241310507

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