Abstract Human umbilical cord serum is full of molecules that play vital roles in foetal development. This study aimed to explore the effects of RR-171, a novel peptide derived from umbilical cord serum, on pancreatic cancer cells and to elucidate its mechanisms. The anti-pancreatic cancer properties of RR-171 were detected by a cell counting kit-8, colony formation, flow cytometry, LDH release and EdU incorporation assays. RNA sequencing and gene enrichment analysis were applied to identify the differentially expressed genes and enriched pathways. Western blotting analysis was used to detect the expression of proteins. A subcutaneous xenograft model was used to examine the effect of RR-171 on pancreatic cancer cells in vivo. The results demonstrated that RR-171 inhibited the viability, proliferation and colony formation of pancreatic cancer cells in a dose-dependent manner. Gene enrichment analysis revealed that RR-171 inhibits the Wnt signaling pathway. Moreover, RR-171 significantly induced apoptosis and pyroptosis in pancreatic cancer cells in a dose-dependent manner. Z-VAD-FMK partly reversed the proapoptotic effect of RR-171, and VX-765 partly reversed the pro-pyroptotic effect of RR-171. Finally, RR-171 inhibited the growth of pancreatic cancer cells in a subcutaneous xenograft mice model and suppressed the expression of Ki-67 and PCNA in tumors. In conclusion, RR-171 induces apoptosis and pyroptosis through multiple pathways and inhibits pancreatic cancer growth, suggesting that RR-171 might be a potential agent for the treatment of pancreatic cancer. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-025-96465-x. Keywords: Umbilical cord serum, Peptide, RR-171, Apoptosis, Pyroptosis, Pancreatic cancer Subject terms: Cancer, Drug discovery, Medical research, Molecular medicine Introduction Pancreatic cancer is the seventh major cause of cancer-related death and ranks 12th among 36 cancers in terms of morbidity^[36]1,[37]2. The common risk factors for pancreatic cancer consist of smoking, chronic pancreatitis, diabetes, obesity, and other occupational chemical exposures^[38]3,[39]4. Surgery is still the cornerstone of treatment for pancreatic cancer, but comprehensive treatments, including radiotherapy and chemotherapy, also play vital roles in the fight against this disease^[40]5,[41]6. Although, advances have been made in the treatment of pancreatic cancer in recent years. Owing to the lack of prompt screening and diagnostic methods, most patients are already in an advanced stage when they are diagnosed, the success of these treatments is relatively low, and the overall 5-year survival ratio is not more than 10%^[42]7. Moreover, pancreatic cancer is an immunologically “cold” cancer characterized by a lack of infiltration of effector T cells, prominent myeloid inflammation and a low tumor mutational burden, which thwarts immunotherapy^[43]8,[44]9. Thus, novel agents for the treatment of pancreatic cancer are urgently needed. Peptides are tiny molecules with less than fifty amino acids in length and are often derived from natural or synthetic products^[45]10. Peptides can be used as pharmaceuticals for various diseases, including asthma, stroke, diabetes, heart disease and wound healing^[46]11. In terms of anticancer treatment, peptides have also been exploited for various therapeutic and diagnostic strategies^[47]12. For example, the peptides leuprolide and goserelin are clinically used to treat prostate cancer^[48]13. In addition, peptides can be applied as tumor-targeting carriers in peptide-drug conjugates through the combination of cytotoxic drugs with peptide ligands that can target tumor-specific biomarkers or cancer cell-membrane receptors, which can lessen side effects and improve the specificity of cytotoxic drugs^[49]14,[50]15. The peptide-drug conjugate ^177Lu-dotatate has been approved for the treatment of gastroenteropancreatic neuroendocrine tumors^[51]16. The above results suggest that peptides might be promising choices for pancreatic cancer therapy. Human umbilical cord blood, which is characterized by the presence of various stem cells, proteins, cytokines and lipids, plays a vital role in foetal development^[52]17. Up to now, human umbilical cord blood has been widely applied in clinical settings by utilizing hematopoietic stem/progenitor cells, for example, Fanconi anemia, tissue regeneration, and malignant hematological diseases^[53]18. In recent years, umbilical cord blood serum (UCBS) has attracted increasing attention because it contains abundant cytokines and molecules that play important roles in cell proliferation, differentiation and immune regulation^[54]19. However, studies on the role of UCBS components in the oncology field are rare. A previous study reported that UCBS inhibits the viability, proliferation, and migration of human prostate cancer cell lines^[55]20, which represents a novel direction in the oncology therapy field. Thus, anticancer components might be found in UCBS, which might stimulate the discovery of new agents for pancreatic cancer treatment. RR-171, a novel peptide with seventeen amino acids (peptide sequence: RRRRLVAGVLVLLRRRR), was identified and modified in our laboratory from UCBS though proteomics biotechnology method. The biological function segment is the sequence LVAGVLVLL, and the four arginines at both ends are added to increase its solubility. The molecular weight of RR-171 is 2145.68 g/mol, the isoelectric point of RR-171 is 12.4, and net charge is 8^[56]21. The random coil of RR-171 accounts for 35.7%, the antiparallel structure accounts for 33.9%, and beta-turn accounts for 20.1%^[57]21. In our previous study, we reported that RR-171 exhibits anticancer properties^[58]21. However, the specific role of RR-171 in pancreatic cancer has not been investigated. In the current study, we investigated the antitumor effect of RR-171 peptide on human pancreatic cancer cells both in vitro and in vivo and then further explored the underlying mechanisms of this effect in pancreatic cancer cells. Materials and methods Cell culture and reagents The hTERT-immortalized human normal pancreatic epithelial cell line (HPNE) and pancreatic cancer cell lines (Panc-1, Bxpc-3, and Capan-2) were purchased from ATCC. HPNE and Panc-1 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM, Gibco, Carlsbad, USA) containing 10% fetal bovine serum (FBS) (Gibco) and 1% penicillin/streptomycin (Gibco). Bxpc-3 and Capan-2 were cultured in RPMI 1640 medium (Gibco) containing 10% FBS (Gibco) and 1% penicillin/streptomycin (Gibco). These cells were cultured at 37℃ in a 5% CO[2] humidified incubator. Z-VAD-FMK and VX-765 were purchased from Selleck Chemicals (Selleck, Houston, USA). Both reagents were dissolved in DMSO (Sigma Aldrich), and diluted to 100 mM and stored in a − 80℃ freezer. The peptide RR-171 was synthesized by Glbiochem (Shanghai, China). Cell viability assay The cells were seeded in 96-well plates (4500 cells/well). After cell adhesion, the medium was replaced with fresh medium or medium containing DMSO or different concentrations of RR-171 for 24 h. Then, the cells were incubated with medium containing 10% Cell Counting Kit-8 reagent (CCK-8; Beyotime Biotechnology, China) for 2 h at 37℃. The OD value was measured with a microplate reader (BioTek Epoch, USA) at 450 nm. The IC[50] values were counted by nonlinear regression analysis using GraphPad Prism 8 software. Colony formation assay The cells were seeded in 6-well plates (800 cells/well). After the cells adhesion steadily, the medium was replaced with fresh medium or different concentrations of RR-171 for 24 h, after which the medium was replaced with fresh medium for 14 days. These cells were washed with PBS and then fixed with 4% paraformaldehyde (Beyotime Biotechnology, China). After fixation, the cells were washed once with PBS and then stained with crystal violet (Beyotime Biotechnology, China) for 30 min. EdU incorporation assay The cells were seeded in 96-well plates (2500 cells/well). After cell adhesion, fresh medium or different concentrations of RR-171 were added to Bxpc-3 and Capan-2 cells for 48 h. The BeyoClick™ EdU Cell Proliferation Kit with Alexa Fluor 488 (Beyotime Biotechnology, China) was used according to the manufacturer’s protocol. The cells were observed and imaged with a fluorescence microscope after EdU staining. RNA sequencing and gene enrichment analysis Capan-2 cells were incubated with 20 µM RR-171 for 24 h and then lysed in Trizol reagent (Thermo Fisher Scientific, USA). The total RNA quantity and purity were assessed using NEBNext Ultra TM RNA Library Prep Kit (NEB, E7490) and AMPure XP system. After the library was constructed, we performed PE150 sequencing on an Illumina Novaseq 6000 according to the manufacturer’s protocol. We applied the DEGseq algorithm to filter the differentially expressed genes (DEGs) after significance analysis and FDR analysis under the criteria (|log2FC| > 1, P value < 0.05). Gene set enrichment analysis (GSEA) was performed using GSEA software (Broad Institute, Cambridge, MA). The R package ClusterProfiler was used to identify the significant GO (Gene Ontology) categories and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways^[59]22–[60]24. A network was built on the basis of the significant pathways (P value < 0.05). Volcano plots and bubble plots were drawn by the R software on the basis of the differential gene and pathway analyses. Flow cytometry analysis of apoptosis Capan-2 and Bxpc-3 cells were seeded in 6-well (3 × 10^5 cells/well) plates, and then the peptide RR-171 was added to the wells at concentrations of 0, 5, 10, and 20 µM for 24 h. After that, the cells were digested and washed twice with PBS and then resuspended in Annexin V binding Buffer. Next, the cell suspension were stained with 10 µl Propidium Iodide and 5 µl FITC Annexin V (Biolegend, San Diego, CA) or 5 µl APC Annexin V (eBioscience, San Diego, CA) reagents at room temperature (25℃) for 15 min in the dark. Subsequently, 400 µl Annexin V binding Buffer was added to each tube. Finally, the stained samples were detected by using a flow cytometer (MA900, Sony, Japan). The results were analyzed by using FlowJo 10. Scanning electron microscopy Capan-2 cells were seeded in 24-wells plates (5 × 10^4 cells/well). After the cells adhesion, RR-171, or fresh medium for the control group, was added to the wells and incubated for 24 h. Then, the cells were washed with PBS, fixed in PBS containing 2.5% glutaraldehyde for 2 h and washed with PBS for three times for 15 min. Next, the cells were dehydrated with increasing concentrations of ethanol (from 30%, 40%, 50%, 60%, 70%, 80%, 90–100%). The cells were then sprayed with an ion sputter. The cells were subsequently observed via an S-3400 N scanning electron microscope (Hitachi, Tokyo, Japan). Protein analysis by Western blotting Protein extraction was performed using RIPA lysis buffer and phenylmethylsulfonyl fluoride. Western blot analysis of Capan-2 and Bxpc-3 cells treated with different concentrations of RR-171 was performed as previously described^[61]25. After protein extraction, protein quantification was performed via a BCA protein quantification kit (BOSTER, Wuhan, China). The protein samples were separated via SDS-PAGE and transferred to polyvinylidene fluoride membranes. Next, the membranes were blocked with QuickBlock™ Blocking Buffer (Beyotime Biotechnology, China) for 15 min and then incubated with primary antibody at 4 °C overnight. The membranes were subsequently washed with TBST (10 min, 3 times) and incubated with secondary HRP-conjugated antibodies for 2 h at room temperature. The membranes were subsequently washed with TBST (10 min, 3 times) and developed with a chemiluminescence kit (Thermo Fisher Scientific Inc. IL, USA). Finally, the protein signals were detected using the ChemiDoc MP Imaging System (Bio-Rad, Hercules, CA, USA). Antibodies against Bax and Bcl-2 were purchased from BOSTER Biological Technology. Antibodies against Caspase-1, Caspase-3, Caspase-7, Caspase-9, Cleaved-PARP, IL-1β, IL-18, GSDMD, NLRP-3 and HRP-conjugated antibodies were purchased from Cell Signaling Technology. Antibodies against Wnt-1, β-actin, β-catenin and GSK3β were purchased from Proteintech. Lactate dehydrogenase (LDH) release analysis A lactate dehydrogenase release assay kit (c0017, Beyotime, China) was used to detect the percentage of LDH released by the cells. The cells were seeded in 96 well plates (5000–6000 cells/well); after the cells were adhesion, different agent treatments were performed. After treatment, the 96-well plates were centrifugated (400g, 5 min), the supernatant was collected, and the experiment was subsequently performed based on the manufacturer’s instructions. The percentage of LDH release was calculated as follows: LDH release = (LDH sample/LDH maximum) × 100%. Subcutaneous xenograft model The animal experiments were reviewed and approved by the Animal Ethics Committee of Air Force Medical University. The animal study was designed in accordance with the ARRIVE guidelines. Female nude mice (BALB/c nude, 5–6 weeks old) were obtained from GemPharmatech (Jiangsu, China) and housed at the Air Force Medical University Animal Experiment Center. Capan-2 cells (6 × 10^6) were injected subcutaneously into the right flanks of the nude mice. Five days later, the mice were randomly divided into two groups (n = 6). The experimental group was intraperitoneally injected with RR-171 (20 mg/kg) every 3 days for three weeks. The control group was intraperitoneally injected with 100 µl of normal saline solution. Tumor length (L) and width (W) and mouse weight were measured every third day. The tumor volume was calculated as follows: tumor volume (V) = (L×W^2)/2. Finally, at the end of the experiment, the nude mice were euthanized by sodium pentobarbital overdose by intraperitoneal injection, after which the lungs, livers, spleens, kidneys, and hearts were excised, and the tumors were removed and weighed. All methods were carried out in accordance with relevant guidelines and regulations. Hematoxylin-eosin (HE) and immunohistochemical staining Hematoxylin-eosin (HE) staining was performed using a Hematoxylin and eosin staining kit (c0105M, Beyotime, China). The experiment was performed according to the manufacturer’s instructions. The tumor tissues were fixed with 4% paraformaldehyde, embedded in paraffin and sectioned. Immunohistochemical staining was performed as described previously^[62]26. The immunohistochemical staining results were analyzed by Image j 1.53 software. The average density was calculated via the following formula: average density = IOD/area. Statistical analysis All experimental data were statistically analyzed by using GraphPad Prism 8.0 software. The data are expressed as the mean ± standard deviation (SD). An unpaired t test (between two groups) or one-way ANOVA (multiple groups) was used for comparisons. P value less than 0.05 was identified as statistically significant. Results RR-171 inhibits pancreatic cancer cell viability and proliferation At the beginning, we treated Panc-1, Capan-2, Bxpc-3 and HPNE cells with RR-171 and conducted CCK-8 experiments. The results revealed that RR-171 obviously inhibited pancreatic cancer cell lines in a dose-dependent manner. The IC[50] values of RR-171 in Capan-2, Bxpc-3 and Panc-1 cells at 24 h were 24.70 µM, 15.71 µM and 75.68 µM, respectively (Fig. [63]1A). While the inhibitory effect of RR-171 on HPNE cells was attenuated, the IC[50] value was 144.50 µM (Supplementary Fig. [64]1). On the basis of these results, we selected Capan-2 and Bxpc-3 cells for further study. The results of plate cloning experiments indicated that the number of clones formed by Capan-2 and Bxpc-3 cells dramatically decreased with increasing RR-171 concentrations (Fig. [65]1B). The results of the EdU staining assay also showed that RR-171 distinctly suppressed the proliferation of Capan-2 and Bxpc-3 cells in a dose-dependent manner (Fig. [66]1C). These results demonstrated that RR-171 inhibited pancreatic cancer cell viability and proliferation. However, the inhibitory effect of RR-171 on normal pancreatic cells was limited. Fig. 1. [67]Fig. 1 [68]Open in a new tab RR-171 suppresses pancreatic cancer cell viability and proliferation in vitro. (A) Capan-2, Bxpc-3, and Panc-1 cell viability was determined using a CCK-8 analysis after treatment with different concentrations of RR-171. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the 0 µM RR-171 treatment group. (B) Colony formation assay to identify the effect of RR-171 on pancreatic cancer cell proliferation. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the 0 µM RR-171 treatment group. (C) The proliferation of Capan-2 and Bxpc-3 cells was determined using an EdU assay after treatment with increasing concentrations of RR-171. The nuclei were stained with Hoechst (blue), and the proliferating cells were stained with EdU (green) (200×). Differential gene expression and pathway enrichment analysis of pancreatic cancer cells after RR-171 treatment Capan-2 cells were treated with RR-171 for 24 h, after which RNA was extracted for RNA sequencing. Compared with those in the control group, there were 1616 downregulated genes and 556 upregulated genes in the RR-171-treated group (|log 2 fold change(FC)| > 1, false discovery rate (FDR) < 0.05) (Fig. [69]2A). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses and gene set enrichment analysis (GSEA) of the genes in Capan-2 cells after RR-171 treatment were performed. The DEGs were enriched mainly in the negative regulation of the cell population, apoptosis, cell death and other signaling pathways (Fig. [70]2B–D). Through further GO and pathway network analysis, we found that the Wnt signaling and MAPK pathway were focused (Fig. [71]2E). Combined with the GSEA results, Wnt signaling attracted our attention. Thus, we performed western blot analysis for key proteins related to canonical Wnt signaling pathway. The results showed that Wnt-1, GSK3β and β-catenin were decreased in a dose-dependent manner after RR-171 treatment (Fig. [72]2F). These results suggest that RR-171 can inhibit the canonical Wnt signaling pathway in pancreatic cancer cells, which might be one of mechanism by which RR-171 inhibits pancreatic cancer cells. Moreover, the results of the Go and KEGG analyses suggested further exploration of the other effects of RR-171 on pancreatic cancer cells. Fig. 2. [73]Fig. 2 [74]Open in a new tab Differential gene expression and pathway enrichment analysis of pancreatic cancer cells treated with RR-171. (A) Capan-2 cells were treated with RR-171 for 24 h for RNA sequencing, and a volcano plot of differential expression was generated. [Downregulated genes are in blue, upregulated genes are in red, nonregulated genes are in gray (|log 2 fold change (FC)| > 1, false discovery rate (FDR) < 0.05)]. Volcano plots were drawn by the R software via differential gene analysis. (B) GSEA of the differentially expressed genes using GSEA software. (C) GO pathway analysis of differentially expressed genes. (D) KEGG pathway analysis of differentially expressed genes. ClusterProfiler package was applied to identify the significant GO and KEGG categories, and FDR was used to correct the p-values via R software. (E) Pathway network analysis of the KEGG pathways [the network was built on the basis of the relationships among KEGG pathways that are significant terms (P value < 0.05)]. (F) Western blot analysis of cell lysates treated with different concentrations of RR-171 for 24 h. Detection of Wnt signaling pathway-related proteins. Relative expression of protein = protein/β-actin band intensity. RR-171 induces caspase-dependent apoptosis in pancreatic cancer cells After incubation with RR-171, the cells were stained with PI and Annexin-V. Flow cytometry results indicated that RR-171 distinctively increased the percentage of apoptotic Capan-2 and Bxpc-3 cells in a dose-dependent manner (Fig. [75]3A). The western blot results demonstrated that, as the RR-171 concentration increased in Capan-2 and Bxpc-3 cells, the expression of the antiapoptotic protein Bcl-2 decreased, whereas the expression of the proapoptotic protein Bax increased in a dose-dependent manner (Fig. [76]3B).Moreover, we examined apoptosis-associated proteins in the caspase signaling pathway. We found that the levels of caspase-3, caspase-7, and caspase-9 decreased in a dose-dependent manner, whereas the level of cleaved PARP increased (Fig. [77]3C), which suggested that RR-171 activated the apoptosis-associated caspase signaling pathway to induce the apoptosis of pancreatic cancer cells. Next, we treated Capan-2 and Bxpc-3 cells with the pan-caspase inhibitor Z-VAD-FMK alone or in combination with RR-171 for 24 h. Flow cytometry demonstrated that Z-VAD-FMK treatment significantly reduced the ratio of apoptosis in both cell lines (Fig. [78]3D). Taken together, these results suggest that RR-171 induces caspase-dependent apoptosis in pancreatic cancer cells. Fig. 3. [79]Fig. 3 [80]Open in a new tab RR-171 induces apoptosis in pancreatic cancer cells. (A) After Capan-2 and Bxpc-3 cells were treated with RR-171 for 24 h, the ratio of apoptotic cells was detected by flow cytometry using PI/Annexin V-FITC staining. *P < 0.05 and ***P < 0.001compared with the 0 µM RR-171 treatment group. (B,C) Western blotting detection of proteins related to apoptosis in Capan-2 and Bxpc-3 cells treated with 0, 5, 10, and 20 µM RR-171 for 24 h. (D) Capan-2 and Bxpc-3 cells were treated with RR-171 and Z-VAD-FMK or DMSO, and the apoptosis ratio was detected by flow cytometry using PI/Annexin V-APC double staining. **P < 0.01 and ***P < 0.001 compared with the control group. RR-171 induces pyroptosis in pancreatic cancer cells The differential gene expression enrichment analysis results suggest that cell death might play a part in the effect of RR-171 on pancreatic cancer cells. Thus, we explored the morphology of Capan-2 cells treated with RR-171. Scanning electron microscopy (SEM) observed that the connections among cells were reduced with 5 µM RR-171 treatment, balloon-like membrane protrusions formed with10 µM RR-171 treatment, and cell debris was induced with 20 µM RR-171 treatment (Fig. [81]4A). We then observed that pores formed in the cell membrane, which might have caused cell swelling and rupture (Fig. [82]4B). Additionally, LDH release from Capan-2 and Bxpc-3 cells was significantly increased after RR-171 treatment (Fig. [83]4C; P < 0.01). These results suggest that pyroptosis might be induced by RR-171. Thus, we detected pyroptosis-associated proteins. The western blotting results demonstrated that RR-171 treatment led to elevated levels of caspase-1, NLRP-3, GSDMD, IL-1β and IL-18 in a dose-dependent manner in both cell lines (Fig. [84]4E, F). Next, we applied the pan-caspase inhibitor Z-VAD-FMK to explore the effect of RR-171 combined with Z-VAD-FMK. We found that Z-VAD-FMK did not inhibit the increase in LDH release in Capan-2 cells and partly inhibited the increase in LDH release in Bxpc-3 cells (Fig. [85]4D). We then utilized VX-765, a pyroptosis inhibitor, for RR-171 treatment of pancreatic cancer cells. The results showed that VX-765 significantly inhibited the increase in LDH release induced by RR-171 in both cell lines (Fig. [86]4G; P < 0.05). Western blot analysis also showed that VX-765 decreased the elevated protein levels of IL-18, Il-1β, Caspase-1, and NLRP-3 (Fig. [87]4H, I). Fig. 4. [88]Fig. 4 [89]Open in a new tab RR-171 induces pyroptosis in pancreatic cancer cells. (A,B) Capan-2 cells were treated with different concentrations of RR-171, and morphological changes were observed via SEM (red arrows, morphological changes). (C) After Capan-2 and Bxpc-3 cells were treated with RR-171 for 24 h, cytotoxicity was detected by lactate dehydrogenase (LDH) release into the cell culture medium. ***P < 0.001 compared with the control group. (D) Capan-2 and Bxpc-3 cells were treated with Z-VAD-FMK and RR-171 for 24 h. Cytotoxicity was detected by LDH release assay. *P < 0.05; ***P < 0.001; ns means nonsignificant. (E,F) Western blotting detection of proteins of pyroptosis markers in Capan-2 and Bxpc-3 cells treated with 0, 5, 10, 20 µM RR-171 for 24 h. (G) Capan-2 and Bxpc-3 cells were treated with VX-765 and RR-171 for 24 h. Cytotoxicity was detected by LDH release assay. *P < 0.05; **P < 0.01; and ***P < 0.001. (H,I) Western blotting detection of proteins of pyroptosis markers in Capan-2 and Bxpc-3 cells treated with VX-765 alone or in combination with RR-171 for 24 h. RR-171 suppresses tumor growth in Capan-2 cell xenograft mice To identify whether RR-171 has anti-pancreatic cancer tumor properties in vivo, we established Capan-2 cell subcutaneous xenografts in nude mice. The tumor volume was significantly reduced beginning on Day 17 in the xenograft model injected intraperitoneally with RR-171 (Fig. [90]5A–C). At the end of the animal experiment, the tumor weight was also significantly reduced (Fig. [91]5D). There was no significant difference in the body weight of the nude mice between the two groups (Fig. [92]5D). To explore the potential toxicity effects of RR-171 on nude mice, we obtained lung, heart, liver, spleen, and kidney samples from the nude mice at the end of the experiment. HE staining results indicated that RR-171 had no significant effects on the above five parenchymal organs (Supplementary Fig. [93]2). Moreover, the immunohistochemistry results showed that RR-171 significantly decreased the expression levels of Ki-67 and PCNA (Fig. [94]5E). These results demonstrated that RR-171 can inhibit pancreatic cancer cell tumor growth in vivo. Fig. 5. [95]Fig. 5 [96]Open in a new tab RR-171 inhibits tumor growth in pancreatic cancer cell xenograft mice. (A,B) Images of the mice and subcutaneous xenograft tumors formed in the nude mice model. (C) Tumor volumes were measured and counted every three days. **P < 0.01; ***P < 0.001; the arrow indicates the beginning of treatment. (D) Tumor weight and body weight at the end of the experiment. **P < 0.01. (E) Immunohistochemistry results of Ki-67 and PCNA expression in the tumors (200×). *P < 0.05 and **P < 0.01. Discussion Pancreatic cancer remains a troubling illness to treat^[97]27. Surgery continues to be the only chance for curing pancreatic cancer, and it has become a relatively safe treatment in experienced medical centers^[98]28. However, surgery alone is not sufficient because most affected patients present with advanced stage disease and miss the chance for surgery, and high-rate of relapse after curative surgery without addition therapy^[99]29. Thus, comprehensive treatment strategies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, have been developed in recent decades^[100]30. Although the overall 5-year survival of patients with pancreatic cancer has improved over the preceding years, the overall prognosis of patients with pancreatic cancer remains unsatisfactory, and the treatment for patients with pancreatic cancer is still a formidable conundrum^[101]29,[102]31. Identifying potential agents for the treatment of pancreatic cancer has important implications. In this study, we explored the peptide RR-171 as a potential drug for the treatment of pancreatic cancer. Umbilical cord blood has been developed as a therapy for numerous applications, including stem cell transplantation, immunotherapy, and regenerative medicine^[103]18,[104]32. In recent years, umbilical cord blood derived molecules or cells have been explored for the treatment of malignancies^[105]19. For example, Chen et al. reported that the use of umbilical cord blood-derived natural killer cells alone or in combination with Bevacizumab can be a novel treatment for patients with colorectal cancer^[106]33. Raquel et al. found that umbilical cord blood serum can significantly inhibit the viability, proliferation and migration of human prostate cancer cell lines^[107]34, but they did not identify the specific constituents that play anticancer roles. In our previous study, we reported that the peptide RR-171 derived from umbilical cord blood serum has anti-pancreatic cancer properties^[108]21. The peptide RR-171 has 17 amino acids, the core function part of which is the medial nine amino acids, and the terminal eight arginines are added to increase the solubility of the peptide. In present study, RR-171 inhibited the viability and proliferation of pancreatic cancer cell lines. However, normal pancreatic ductal epithelial cells were less sensitive to the cellular cytotoxicity of RR-171. We speculated that, just as normal epithelial cells are less sensitive to chemotherapy drugs, RR-171 might also be less toxic to relatively poorly proliferating cells. We subsequently explored the mechanisms by which RR-171 inhibits pancreatic cancer cell lines. This study revealed that RR-171 can inhibit the canonical Wnt signaling pathway. Increasing evidence indicates that Wnt signaling pathway activation plays a vital role in all aspects of human tumor development and progression, including growth, stemness, angiogenesis, resistance, and immune evasion^[109]35. Wnt signaling pathway-associated inhibitors have also been developed for preclinical and clinical trials in the treatment of malignancies^[110]36. A previous study showed consistent activation of core signaling pathways, including the Wnt signaling pathway, in pancreatic cancer^[111]37. Further studies have also demonstrated that epithelial Wnt signaling is crucial for pancreatic cancer progression^[112]38. In brief, RR-171 might exert a tumor suppressive role in pancreatic cancer through the inhibition of the canonical Wnt signaling pathway. Interestingly, in our previous study, we found that RR-171 can activate the NF-KappaB pathway in hepatocellular carcinoma^[113]21. The reason for this difference might be that the two pathways play different key roles in the two tumors. Moreover, the RNA sequencing results suggested that RR-171 treatment might induce apoptosis in pancreatic cancer cell lines. We revealed that RR-171 can significantly induce apoptosis in both pancreatic cancer cell lines by activating caspase pathway signaling. This result is similar to our previous finding that RR-171 can promote the apoptosis of hepatocellular carcinoma cells. In addition, through SEM scanning, we found that pores formed on the surface of pancreatic cancer cell lines and that LDH release increased after treatment with RR-171. These results suggest that RR-171 might induce pyroptosis in pancreatic cancer cell lines. We examined markers of pyroptosis, including NLRP-3, GSDMD, IL-1β, IL-18 and Caspase-1, which were increased after treatment with RR-171. We then applied the pyroptosis inhibitor VX765, which reversed the increase in LDH release. These results demonstrated that RR-171-induced pyroptosis in pancreatic cancer cells might occur through triggering the Caspase-1/NLRP-3/GSDMD pathway. In our previous study, pores were observed in hepatocellular carcinoma cells after RR-171 treatment^[114]21, while deeply studies have not investigated this phenomenon. In conclusion, RR-171 can induce apoptosis and pyroptosis in pancreatic cancer cells. Next, to identify the anticancer functions of RR-171 in vivo, we established a subcutaneous xenograft model to examine the function of RR-171 in vivo. The results showed that the volume and weight of tumors were significantly suppressed by RR-171 treatment. The immunohistological results revealed that RR-171 inhibited the expression of Ki-67 and PCNA, which was consistent with the tumor volume. In addition, the HE results demonstrated that the parenchymal organ toxicity of RR-171 was not apparent in vivo, which suggests that RR-171 might be a relatively safe agent in nude mice. We further revealed that RR-171 can inhibit the growth of pancreatic cancer in vivo, which is consistent with in vitro results. In our previous study, we demonstrated that RR-171 might interact with α-tubulin and inhibit tubulin polymerization in hepatocellular carcinoma cells^[115]21. Another study showed that anti-tubulin agents can arrest cell division and facilitate one of the apoptosis pathways and that modified tubulin inhibitors have the ability to improve the outcomes of patients with pancreatic cancer through overcoming chemoresistance in these patients^[116]39,[117]40. These results suggest that the specific target of RR-171 in pancreatic cancer cells might also be associated with tubulin polymerization, which needs further study. Eventually, the bioactivity of RR-171 was not high enough, and the IC[50] reached only micromolar concentrations. Modifications might be explored to improve the bioactivity of RR-171 in our future studies. Moreover, the effects of combined treatment with RR-171 and radiotherapy or chemotherapy might be evaluated in our future studies, and modifications must be explored in the future to improve its bioactivity. Overall, this study revealed that a new peptide, RR-171, induces apoptosis and pyroptosis through multiple pathways and inhibits pancreatic cancer growth. RR-171 might exert anti-pancreatic cancer effects by suppressing the Wnt signaling pathway. Our study provides new directions and strategies for the development of new agents for the treatment of pancreatic cancer. Electronic supplementary material Below is the link to the electronic supplementary material. [118]Supplementary Material 1^ (2MB, pdf) Acknowledgements