Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an immunosuppressed, apoptosis-resistant phenotype. TLY012 is pegylated recombinant Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL), an orphan drug for chronic pancreatitis and systemic sclerosis. Innate immune TRAIL signaling suppresses cancer. We hypothesized that the combination of immune checkpoint-blocking anti-PD-1 antibody and TLY012 would have synergistic anti-tumor efficacy in immune-competent PDAC-bearing mice. PDAC tumor-bearing C57Bl/6 mice treated with 10 mg/kg anti-mouse PD-1 antibody twice weekly and 10 mg/kg TLY012 three times weekly had reduced tumor growth and tumor volume at 70 days compared to either drug alone (all P < 0.005). B-cell activating factor (BAFF), which promotes PDAC tumors, decreased to 44% of control mice with dual treatment at 7 days and remained decreased at 3 months. Long-term dual treatment showed the highest plasma levels of proinflammatory cytokines interferon-gamma (average 5.6 times control level, P=0.046), CCL5 (average 14.1 times control level, P=0.048), and interleukin-3 (IL-3, average 71.1 times control level, P=0.0053). Flow cytometry showed trends toward decreased circulating regulatory T cells, increased NK cells, and a higher proportion of CD8+ T cells within tumors in the dual treatment group. In summary, the combination of anti-PD-1 and TLY012 prevented the growth of PDAC in an immunocompetent mouse model while increasing tumor-infiltrating CD8+ T cells, decreasing circulating T-regulatory cells and altering plasma cytokine expression of CCL5, interferon-gamma, and IL-3 to promote proinflammatory, antitumor effects. Combining TLY012 and anti-mouse PD-1 modifies immune cell and cytokine levels to induce a more proinflammatory immune environment that contributes to decreased PDAC tumor growth.
Keywords: TLY012, TRAIL, BAFF, CCL5, IFN-gamma, IL3, inflammation, cancer
Introduction
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly disease with little progress in long-term survival over the last several decades [1-5]. Much work has been done to characterize molecular alterations and drivers in pancreatic cancer including among different ethnic groups, early onset pancreatic cancer, and treatment effects on the genomic landscape [6-13]. Some of the key driver alterations include mutations in KRAS, p53, p16, SMAD4, KDM6A, ARID1A, and BRCA2, signatures of DNA damage and repair deficiency, mismatch repair deficiency as well as amplified genes such as ERBB2, MET, PIK3CA among others [6,7,11,12]. While much progress has been made with immunotherapy across tumor types, and much understanding of the biology of pancreatic cancer, there has been little impactful progress on advanced PDAC in part due to an immunosuppressed, apoptosis-resistant phenotype [14-23].
The TNF-Related Apoptosis-Inducing Ligand (TRAIL) is part of the host immune system that suppresses transformed cells, virally infected cells as well as cancer and its metastases [24-35]. Nearly 3 decades ago, the discovery of pro-apoptotic TRAIL Death Receptor DR5 as a direct transcriptional target of the p53 tumor suppressor gene directly linked the innate immune system to the host response to suppress tumorigenesis [36-42]. Deletion of TRAIL receptor DR5 in mice led to apoptotic resistance in vivo as well as tumorigenesis [43,44]. It was later recognized that the TRAIL gene is also a p53 target gene and subsequent work identified TRAIL-inducing compound #10 (TIC10; also known as ONC201) [45,46]. We previously reported that the combination of TRAIL or TRAIL receptor DR5 agonist antibodies and ONC201 has particularly potent anti-tumor effects in vivo across cancer types including pancreatic cancer [47-50]. We have also previously reported a TRAIL-inducing micro-RNA that we have not, as of the date of publication of this manuscript, translated to in vivo studies or to the treatment of patients [51].
The TRAIL pathway influences both inflammation and tumorigenesis as well as fibrosis, all relevant to PDAC and its potential novel therapeutics [44,52-55]. TLY012 is pegylated recombinant Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL) [55,56].
In the present studies, we explore the hypothesis that TLY012’s modulation of the tumor microenvironment has the potential for synergistic effects when combined with immune checkpoint blockade. We demonstrate potent anti-tumor effects of TLY012 combined with anti-mouse PD-1 and show immune alterations including in B- and T-cell immunity as well as tumor-promoting cytokines. Our results prompt further preclinical and clinical studies evaluating the novel treatment combination of TLY012 and anti-PD1.
Materials and methods
Cell culture
Cell lines used for this study were acquired from the American Type Culture Collection (ATCC), unless otherwise indicated. All pancreatic cancer cells were grown in Dulbecco’s Modified Eagle Medium (DMEM) with 10% Fetal Bovine Serum (FBS). Human Foreskin Fibroblasts (HFF) were grown in DMEM with 15% FBS. rhTRAIL was generated in-house using a protocol previously developed by our lab and detailed in Kim et al. TLY012 was provided by Theraly Fibrosis, Inc. TLY012 was diluted in sterile phosphate buffered saline (PBS) for in vitro experiments.
Cell viability assay
To assess cell viability, cells were plated overnight in 96-well plates at a density of 1.0 × 104 cells/well. All cells were plated in triplicates. After 72 hours, the media in the wells was replaced with either fresh media (controls) or with media containing various doses of rhTRAIL or TLY012. After 4 hours of incubation, cells were treated with Cell-Titer Glo (Promega) and imaged to assess cell viability. Synergy and combination indices were determined using Compusyn, which uses the Chou-Talalay method for determining synergy.
Cytokine profiling
Murine plasma samples were collected at 7 days and 3 months post-treatment with vehicle control, anti-PD-1, TLY012, or the combination of anti-PD-1 + TLY012. Samples were analyzed using an R&D systems Murine Premixed Multi-Analyte Kit (R&D Systems, Inc., Minneapolis, MN, USA) and a Luminex 200 Instrument (LX200-XPON-RUO, Luminex Corporation, Austin, TX, USA) according to the manufacturer’s instructions. Samples were analyzed in duplicate and average sample values are reported in pg/mL.
Flow cytometric analysis
Murine spleens and tumors were collected 3 months post-treatment. Flow cytometry viability staining was conducted by suspending murine spleen and tumor single cell suspensions in Zombie Violet solution (BioLegend, San Diego, CA, USA) according to manufacturer instructions for 30 minutes at room temperature. Staining for membrane surface proteins was conducted using conjugated primary antibodies for 1 hour on ice, according to manufacturer instructions. The following antibodies were used for the described T cell profiling experiments: CD45 Monoclonal Antibody (30-F11), eVolve™ 605, eBioscience™, Catalog # 83-0451-42; BD Pharmingen™ APC-Cy™7 Rat Anti-Mouse CD3 Molecular Complex, Clone 17A2 (RUO), Catalog # 560590; CD4 Monoclonal Antibody (RM4-5), PE-Cyanine7, eBioscience™, Catalog # 25-0042-82; BD Pharmingen™ PE Rat Anti-Mouse CD8a, Clone 53-6.7 (RUO), Catalog # 553032; Invitrogen CD69 Monoclonal Antibody (H1.2F3), FITC, eBioscience™, Catalog # 11-0691-81; Invitrogen FOXP3 Monoclonal Antibody (FJK-16s), APC, eBioscience™, Catalog # 17-5773-82. The following antibodies were used for the described NK cell profiling experiments: CD45 Monoclonal Antibody (30-F11), eVolve™ 605, eBioscience™, Catalog # 83-0451-42; BD Pharmingen™ PE Rat Anti-Mouse CD3 Molecular Complex, Clone 17A2 (RUO), Catalog # 555275; Invitrogen NK1.1 Monoclonal Antibody (PK136), APC, eBioscience™, Catalog # 17-5941-82; APC/Cyanine7 anti-mouse/human CD11b Antibody, Clone M1/70, Catalog # 101226; Invitrogen CD27 Monoclonal Antibody (LG.7F9), FITC, eBioscience™, Catalog # 11-0271-82; Invitrogen KLRG1 Monoclonal Antibody (2F1), PE-Cyanine7, eBioscience™, Catalog # 25-5893-82. Cells were fixed using IC Fixation Buffer (eBioscience™, San Diego, CA, USA) for 30 minutes according to manufacturer instructions. Cells were resuspended in Flow Cytometry Staining Buffer (R&D Systems, Minneapolis, MN, USA) and were analyzed using a BD Biosciences LSR II and FlowJo version 10.1 (FlowJo, Ashland, OR, USA).
Natural killer cell immunophenotyping
The NK cell flow cytometry panel included the following directly-conjugated primary antibodies: CD45 Monoclonal Antibody (30-F11), eVolve™ 605, eBioscience™, Catalog # 83-0451-42; BD Pharmingen™ PE Rat Anti-Mouse CD3 Molecular Complex, Clone 17A2 (RUO), Catalog # 555275; Invitrogen NK1.1 Monoclonal Antibody (PK136), APC, eBioscience™, Catalog # 17-5941-82; APC/Cyanine7 anti-mouse/human CD11b Antibody, Clone M1/70, Catalog # 101226; Invitrogen CD27 Monoclonal Antibody (LG.7F9), FITC, eBioscience™, Catalog # 11-0271-82; Invitrogen KLRG1 Monoclonal Antibody (2F1), PE-Cyanine7, eBioscience™, Catalog # 25-5893-82. Gating strategies are as follows: NK cell: live/CD45/CD3-/NK1.1+; Mature NK cell: live/CD45/CD3-/NK1.1+/KRLG1+; Activated NK cell: live/CD45/CD3-/NK1.1+/CD11b+; NK cell subset 1: live/CD45/CD3-/NK1.1+/CD11blowCD27low; NK cell subset 2: live/CD45/CD3-/NK1.1+/CD11blowCD27high; NK cell subset 3: live/CD45/CD3-/NK1.1+/CD11bhighCD27high; NK cell subset 4: live/CD45/CD3-/NK1.1+/CD11bhighCD27low.
T cell immunophenotyping
The T cell flow cytometry panel included the following directly-conjugated primary antibodies: CD45 Monoclonal Antibody (30-F11), eVolve™ 605, eBioscience™, Catalog # 83-0451-42; BD Pharmingen™ APC-Cy™7 Rat Anti-Mouse CD3 Molecular Complex, Clone 17A2 (RUO), Catalog # 560590; CD4 Monoclonal Antibody (RM4-5), PE-Cyanine7, eBioscience™, Catalog # 25-0042-82; BD Pharmingen™ PE Rat Anti-Mouse CD8a, Clone 53-6.7 (RUO), Catalog # 553032; Invitrogen CD69 Monoclonal Antibody (H1.2F3), FITC, eBioscience™, Catalog # 11-0691-81; Invitrogen FOXP3 Monoclonal Antibody (FJK-16s), APC, eBioscience™, Catalog # 17-5773-82. Gating strategies are as follows: CD4+ T cell: live/CD45+/CD3+/CD4+/FOXP3-; CD8+ T cell: live/CD45+/CD3+/CD8+; Treg: live/CD45+/CD3+/CD4+/FOXP3+; Activated CD8+ T cell: live/CD45+/CD3+/CD8+/CD69+.
In vivo tumor xenograft studies
All in vivo studies conducted for this manuscript were approved by the Brown University IACUC. For in vivo tumor xenograft studies, we used female C57Bl/6 mice. Mice were aged 5-8 weeks at the time of tumor inoculation. Cells were mixed in a 50:50 Matrigel (Corning): PBS solution and mixed at various dilutions. The total inoculation volume was 200 µL, irrespective of the tumor model or the number of cells inoculated. The vehicle is a solution of 20% Cremophor EL (Sigma-Aldrich), 70% PBS, and 10% DMSO. rhTRAIL was administered through intravenous tail vein injections. TLY012 was administered via intraperitoneal injections.
Tumor volume calculations
Measurements were taken using Vernier calipers. The equation we employed for calculating tumor volume is: Volume = (Width2 * Length)/2. Treatment was initiated once the tumors reached an optimal volume between 100-150 mm3.
Statistical analysis
The statistical significance of differences between pairs was determined using unpaired Student’s t-tests. The statistical significance between groups was determined using a One-way ANOVA followed by a post-hoc Tukey’s multiple comparisons test. The minimal level of significance was P < 0.05. Following symbols * and ** represent, P < 0.05 and P < 0.01, respectively.
Results
Potent anti-tumor effects in PDAC-implanted syngeneic C57Bl/6 mice treated with anti-mouse PD-1 and TLY012
TLY012, a novel TRAIL formulation, activates the host innate immune system through the TRAIL pathway and targets fibrosis within the tumor microenvironment. These properties including removal of an immune suppressive microenvironment are well suited for synergy with other agents such as immune checkpoint therapy that may have improved penetration and efficacy in the tumor microenvironment.
We set up a mouse model treatment scheme (Figure 1A) to test the hypothesis that combination of immune checkpoint-blocking anti-PD-1 antibody and TLY012 would have synergistic anti-tumor efficacy in immune-competent PDAC-bearing mice.
Figure 1.
Syngeneic mice with PDAC treated with anti-mouse PD-1 and TLY012 have delayed tumor growth and reduced tumor volume. A. In vivo PDAC mouse model and treatment schema for anti-tumor efficacy, biomarker, and immune analysis. The number of mice used for short-term as well as long-term treatments is as indicated for each cohort. B. Syngeneic mice with PDAC treated with both anti-mouse PD-1 and TLY012 resulted in slower tumor growth and reduced tumor volume at 70 days compared to either drug alone (all P < 0.005). Tumor weights are shown in the graph in the lower right for the respective treatment groups. C. Images of available tumors at the end of the experiment are shown. D. Weights of mice in different treatment groups over the course of the experiment.
We used immune competent C57/Bl6 mice as the host for our experiments where 50,000 KPC-Luc PDAC cells were implanted subcutaneously. As shown in Figure 1A, we included cohorts of mice for short-term analysis including analysis of tumor-infiltrating lymphocytes (TIL) as well as systemic immune analysis, cytokine analysis, immunohistochemical biomarker analysis and toxicity analysis. Cohort sizes are indicated in Figure 1A. In addition, there were mice allocated to a long-term cohort for similar analyses. Treatments included “adjuvant control” (6 mice for short-term and 9 mice for long-term), TLY012 at 10 mg/kg 3 times per week by intraperitoneal (IP) injection (4 mice for short-term and 6 mice for long term), anti-mouse PD1 at 10 mg/kg 2 times per week by IP injection (4 mice for short-term and 6 mice for long term), and the combination of TLY012 plus anti-PD1 therapy (4 mice for short-term and 6 mice for long term).
The result of the long-term in vivo study demonstrates potent anti-tumor effects in PDAC-implanted syngeneic C57Bl/6 mice treated with anti-mouse PD-1 and TLY012 (Figure 1B). KPC-Luc tumors grew over 20 days at which point treatments were initiated as indicated in Figure 1A. Short-term analysis of immune cells (TILs and systemically), cytokines and immunohistochemistry as well as toxicity analysis were performed at 7 days after treatment initiation for the different cohorts of mice. Long-term analysis was performed as indicated after continued treatments of the long-term treatment cohorts as indicated in Figure 1A. The results demonstrate statistically significant reduction in tumor volumes and tumor weights in the monotherapy cohorts (TLY012 at 10 mg/kg 3 times per week IP or anti-mouse PD1 at 10 mg/kg 2 times per week by IP injection) and potent suppression of tumors with combination therapy with TLY012 plus anti-PD1 therapy over the course of the experiment as shown in Figure 1B.
Available tumors at the end of the experiment were photographed (Figure 1C) and correspond to the results shown in Figure 1B. The mice tolerated the treatments well with no reduction in mouse weights (Figure 1D).
Decreased circulating regulatory T-cells, increased NK-cells, and a higher proportion of CD8+ T cells within tumors after dual treatment with anti-mouse PD-1 and TLY012
We performed flow cytometric analyses of spleen and tumor T-cells and NK-cells to examine the effects of individual anti-mouse PD-1 or TLY012, as well as the dual anti-mouse PD-1 and TLY012 treatment group in treated syngeneic C57Bl/6 KPC-Luc PDAC tumor-bearing mice (Figure 2).
Figure 2.
Decreased circulating regulatory T-cells, increased NK-cells, and a higher proportion of CD8+ T cells within tumors after dual treatment with anti-mouse PD-1 and TLY012. A. Flow cytometric immune analysis of spleen T cells following 3 month treatment with TLY012, anti-PD1 or the combination of TLY012 and anti-PD1 in a syngeneic pancreatic cancer mouse model (KPC-Luc). B. Flow cytometric immune analysis of tumor T following treatment with TLY012, anti-PD1 or the combination of TLY012 and anti-PD1 in a syngeneic pancreatic cancer mouse model (KPC-Luc). C. Flow cytometric immune analysis of spleen NK cells following treatment with TLY012, anti-PD1 or the combination of TLY012 and anti-PD1 in a syngeneic pancreatic cancer mouse model (KPC-Luc). D. Flow cytometric immune analysis of tumor NK cells following treatment with TLY012, anti-PD1 or the combination of TLY012 and anti-PD1 in a syngeneic pancreatic cancer mouse model (KPC-Luc).
We observed a reduction in splenic T-regulatory as cytotoxic CD8+ T-cells (Figure 2A) along with a relative enrichment in analyzed tumors of cytotoxic CD8+ T-cells in combination anti-mouse PD-1 and TLY012-treated tumors (Figure 2B) in the syngeneic KPC PDAC model. We observed enrichment of immature NK cells in the spleen of combo treated mice (Figure 2C) and similarly the tumors of dual anti-mouse PD-1 and TLY012 treated PDAC tumor-bearing C57Bl/6 mice showed enrichment with immature NK cells (Figure 2D). Thus, the combination of TLY012 plus anti-PD1 enriches treated tumors with cytotoxic CD8+ T-cells as well as immature NK cells. These findings are consistent with the observed synergistic anti-tumor efficacy of the combination therapy in vivo.
B-cell activating factor (BAFF), which correlates with tumor progression in PDAC, was reduced in treatment groups at 7 days and 3 months
B-cell activating factor (BAFF) correlates with tumor progression in PDAC [57]. Plasma BAFF concentrations were reduced in anti-PD1-treated and dual TLY012 plus anti-PD1-treated mice at 7 days and was reduced in single and dual-treated groups after three months (Figure 3). Reduced BAFF levels are consistent with the observed anti-PDAC effects of treatment with TLY012 plus anti-PD1.
Figure 3.
Reduced tumor-promoting B-cell activating factor (BAFF) levels in anti-PD1 treated and dual TLY012 plus anti-PD1 treated mice at 7 days and in single and dual treated groups at three months. Day 7 murine plasma cytokine levels are in the left panel and the levels at 3 months are shown in the right panel for the different treatment groups as indicated. The numbers shown for plasma BAFF levels on the X-axes are in pg/mL.
Long-term plasma cytokine analysis following treatment with TLY012, anti-PD1 or the combination of TLY012 and anti-PD1 in syngeneic KPC PDAC mouse model
We assessed plasma cytokine levels at 3 months in the TLY012, anti-PD1 or combination treatment groups versus control mice (Figure 4). A few proinflammatory cytokines including interferon-gamma and CCL5 were elevated while VEGFR2 trended towards reduction in the long-term dual treatment group (Figures 4, 5). These results are consistent with the observed anti-PDAC effects of treatment with TLY012 plus anti-PD1.
Figure 4.
Long-term cytokine levels in KPC PDAC tumor-bearing C57Bl/6 mice for control, TLY012, anti-PD1, or dual therapy with TLY012 plus anti-PD1 as indicated. Three-month murine plasma cytokine levels are shown for the different treatment groups as indicated. Cytokine concentrations were normalized to the control group. Fold change is over control is represented by circle diameter.
Figure 5.
Alterations in selected cytokines at 3 months in KPC PDAC tumor-bearing C57Bl/6 mice for control, TLY012, anti-PD1, or dual therapy with TLY012 plus anti-PD1 as indicated. A. Plasma IFN-gamma levels at 3 months in pg/mL for control, TLY012, anti-PD1, or dual therapy with TLY012 plus anti-PD1 as indicated. B. CCL5 levels at 3 months in pg/mL for control, TLY012, anti-PD1, or dual therapy with TLY012 plus anti-PD1 as indicated. C. VEGFR2 levels at 3 months in pg/mL for control, TLY012, anti-PD1, or dual therapy with TLY012 plus anti-PD1 as indicated.
Discussion
We report potent anti-tumor effects following long-term treatment for 3 months with the combination of innate immune anti-fibrotic TLY012 and immune checkpoint blocker anti-mouse PD-1 in a KPC PDAC syngeneic C57Bl/6 mouse model. The treatments were associated with changes in immune cell populations in spleens and tumors and plasma cytokine levels to induce a more proinflammatory immune environment that correlates to decreased PDAC tumor growth.
A limitation of our study is that the tumors were implanted subcutaneously and not orthotopically which could influence the results due to differences in the local microenvironment. Clearly, this would need to be further addressed in future studies.
Another limitation is that while we performed flow cytometric analysis of tumor tissue and spleens, as well as plasma cytokine profiling, we did not carry out immunohistochemical staining of tumors at early or late time points to provide further evidence of altered biomarkers and proposed mechanisms. We also did not test the requirement of NK- or T-cell alterations or cytokine alterations with regard to the observed anti-PDAC effects in vivo. We also did not evaluate the anti-fibrotic effects of TLY012 as monotherapy or in combination with anti-PD1 in vivo.
It is noteworthy that IFNγ and CCL5 were increased in the combined treatment group (TLY012 + anti-PD1). However, how these changes may directly contribute to decreased tumor growth remains an open question for future research. The observed changes in T cells and NK cell populations mentioned above have not been tested as far as their requirement for tumor growth suppression. In addition future studies will need to unravel the importance of Tregs alterations such as their decrease in the spleen, but not in the tumor tissues.
Despite the limitations, our results provide a novel preclinical combination therapy of innate immune TLY012 and immune checkpoint blocker anti-mouse PD-1 that has potent efficacy in a KPC PDAC syngeneic C57Bl/6 mouse model and without evidence of toxicity. The results support the further exploration and clinical testing of TLY012 alone and in combination with anti-PD1 therapy in pancreatic cancer as well as other malignancies.
Acknowledgements
W.S.E-D. is an American Cancer Society Research Professor and is supported by the Mencoff Family University Professorship at Brown University. This work was supported by a grant from D&D Pharmatech to W.S.E-D. This work was presented in part at the 2022 meeting of the American Association for Cancer Research and the 2022 meeting of the Society of Surgical Oncology.
Disclosure of conflict of interest
W.S.E-D. is the Scientific Founder of Oncoceutics, Inc., a subsidiary of Chimerix, and Founder of p53-Therapeutics, Inc., and SMURF-Therapeutics, Inc. Dr. El-Deiry has disclosed his relationships and potential conflict of interest to his academic institution/employer and is fully compliant with NIH and institutional policy that is managing this potential conflict of interest. The disclosed relationships by W.S.E-D. are not directly relevant to the present manuscript as none of the treatments used are owned or licensed by the entities he founded. S.L. is co-Founder, CEO and Chairman of D&D Pharmatech.
References
- 1.Halbrook CJ, Lyssiotis CA, Pasca di Magliano M, Maitra A. Pancreatic cancer: advances and challenges. Cell. 2023;186:1729–1754. doi: 10.1016/j.cell.2023.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hu ZI, O’Reilly EM. Therapeutic developments in pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2024;21:7–24. doi: 10.1038/s41575-023-00840-w. [DOI] [PubMed] [Google Scholar]
- 3.Hernández-Blanquisett A, Quintero-Carreño V, Martínez-Ávila MC, Porto M, Manzur-Barbur MC, Buendía E. Metastatic pancreatic cancer: where are we? Oncol Rev. 2024;17:11364. doi: 10.3389/or.2023.11364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kolbeinsson HM, Chandana S, Wright GP, Chung M. Pancreatic cancer: a review of current treatment and novel therapies. J Invest Surg. 2023;36:2129884. doi: 10.1080/08941939.2022.2129884. [DOI] [PubMed] [Google Scholar]
- 5.Park W, Chawla A, O’Reilly EM. Pancreatic cancer: a review. JAMA. 2021;326:851–862. doi: 10.1001/jama.2021.13027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, Quinn MC, Robertson AJ, Fadlullah MZ, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Wani S, Wilson PJ, Markham E, Cloonan N, Anderson MJ, Fink JL, Holmes O, Kazakoff SH, Leonard C, Newell F, Poudel B, Song S, Taylor D, Waddell N, Wood S, Xu Q, Wu J, Pinese M, Cowley MJ, Lee HC, Jones MD, Nagrial AM, Humphris J, Chantrill LA, Chin V, Steinmann AM, Mawson A, Humphrey ES, Colvin EK, Chou A, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Pettitt JA, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Jamieson NB, Graham JS, Niclou SP, Bjerkvig R, Grützmann R, Aust D, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Falconi M, Zamboni G, Tortora G, Tempero MA Australian Pancreatic Cancer Genome Initiative. Gill AJ, Eshleman JR, Pilarsky C, Scarpa A, Musgrove EA, Pearson JV, Biankin AV, Grimmond SM. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495–501. doi: 10.1038/nature14169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Pantaleo A, Forte G, Fasano C, Lepore Signorile M, Sanese P, De Marco K, Di Nicola E, Latrofa M, Grossi V, Disciglio V, Simone C. Understanding the genetic landscape of pancreatic ductal adenocarcinoma to support personalized medicine: a systematic review. Cancers (Basel) 2023;16:56. doi: 10.3390/cancers16010056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ben-Aharon I, Elkabets M, Pelossof R, Yu KH, Iacubuzio-Donahue CA, Leach SD, Lowery MA, Goodman KA, O’Reilly EM. Genomic landscape of pancreatic adenocarcinoma in younger versus older patients: does age matter? Clin Cancer Res. 2019;25:2185–2193. doi: 10.1158/1078-0432.CCR-18-3042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhang X, Mao T, Zhang B, Xu H, Cui J, Jiao F, Chen D, Wang Y, Hu J, Xia Q, Ge W, Li S, Yue M, Ma J, Yao J, Wang Y, Wang Y, Shentu D, Zhang X, Chen S, Bai Y, Wang Y, Zhang X, Liu Q, Sun Y, Fu D, Liu Y, Xiong L, Wang L. Characterization of the genomic landscape in large-scale Chinese patients with pancreatic cancer. EBioMedicine. 2022;77:103897. doi: 10.1016/j.ebiom.2022.103897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Xie IY, Gallinger S. The genomic landscape of recurrent pancreatic cancer is modified by treatment. Nat Rev Gastroenterol Hepatol. 2020;17:389–390. doi: 10.1038/s41575-020-0321-9. [DOI] [PubMed] [Google Scholar]
- 11.Poruk KE, Moran A, Doctor V, Elvin JA, Radhi S, Loaiza-Bonilla A, Schink JC, Markman M. Mutational landscape of pancreatic adenocarcinoma identified by prospective clinical sequencing in a nationwide cancer network. J. Clin. Oncol. 2022;40:4137–4137. [Google Scholar]
- 12.Bazzichetto C, Luchini C, Conciatori F, Vaccaro V, Di Cello I, Mattiolo P, Falcone I, Ferretti G, Scarpa A, Cognetti F, Milella M. Morphologic and molecular landscape of pancreatic cancer variants as the basis of new therapeutic strategies for precision oncology. Int J Mol Sci. 2020;21:8841. doi: 10.3390/ijms21228841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sivapalan L, Kocher HM, Ross-Adams H, Chelala C. The molecular landscape of pancreatic ductal adenocarcinoma. Pancreatology. 2022;22:925–936. doi: 10.1016/j.pan.2022.07.010. [DOI] [PubMed] [Google Scholar]
- 14.Morrison AH, Byrne KT, Vonderheide RH. Immunotherapy and prevention of pancreatic cancer. Trends Cancer. 2018;4:418–428. doi: 10.1016/j.trecan.2018.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Dauer P, Nomura A, Saluja A, Banerjee S. Microenvironment in determining chemo-resistance in pancreatic cancer: neighborhood matters. Pancreatology. 2017;17:7–12. doi: 10.1016/j.pan.2016.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Timmer FEF, Geboers B, Nieuwenhuizen S, Dijkstra M, Schouten EAC, Puijk RS, de Vries JJJ, van den Tol MP, Bruynzeel AME, Streppel MM, Wilmink JW, van der Vliet HJ, Meijerink MR, Scheffer HJ, de Gruijl TD. Pancreatic cancer and immunotherapy: a clinical overview. Cancers (Basel) 2021;13:4138. doi: 10.3390/cancers13164138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Laface C, Memeo R, Maselli FM, Santoro AN, Iaia ML, Ambrogio F, Laterza M, Cazzato G, Guarini C, De Santis P, Perrone M, Fedele P. Immunotherapy and pancreatic cancer: a lost challenge? Life (Basel) 2023;13:1482. doi: 10.3390/life13071482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ullman NA, Burchard PR, Dunne RF, Linehan DC. Immunologic strategies in pancreatic cancer: making cold tumors hot. J. Clin. Oncol. 2022;40:2789–2805. doi: 10.1200/JCO.21.02616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hosein AN, Dougan SK, Aguirre AJ, Maitra A. Translational advances in pancreatic ductal adenocarcinoma therapy. Nat Cancer. 2022;3:272–286. doi: 10.1038/s43018-022-00349-2. [DOI] [PubMed] [Google Scholar]
- 20.Yeo D, Giardina C, Saxena P, Rasko JEJ. The next wave of cellular immunotherapies in pancreatic cancer. Mol Ther Oncolytics. 2022;24:561–576. doi: 10.1016/j.omto.2022.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mucciolo G, Roux C, Scagliotti A, Brugiapaglia S, Novelli F, Cappello P. The dark side of immunotherapy: pancreatic cancer. Cancer Drug Resist. 2020;3:491–520. doi: 10.20517/cdr.2020.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bear AS, Vonderheide RH, O’Hara MH. Challenges and opportunities for pancreatic cancer immunotherapy. Cancer Cell. 2020;38:788–802. doi: 10.1016/j.ccell.2020.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Francescone R, Barbosa Vendramini-Costa D, Franco-Barraza J, Wagner J, Muir A, Lau AN, Gabitova L, Pazina T, Gupta S, Luong T, Rollins D, Malik R, Thapa RJ, Restifo D, Zhou Y, Cai KQ, Hensley HH, Tan Y, Kruger WD, Devarajan K, Balachandran S, Klein-Szanto AJ, Wang H, El-Deiry WS, Vander Heiden MG, Peri S, Campbell KS, Astsaturov I, Cukierman E. Netrin G1 promotes pancreatic tumorigenesis through cancer-associated fibroblast-driven nutritional support and immunosuppression. Cancer Discov. 2021;11:446–479. doi: 10.1158/2159-8290.CD-20-0775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17:395–417. doi: 10.1038/s41571-020-0341-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Montinaro A, Walczak H. Harnessing TRAIL-induced cell death for cancer therapy: a long walk with thrilling discoveries. Cell Death Differ. 2023;30:237–249. doi: 10.1038/s41418-022-01059-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Thapa B, Kc R, Uludağ H. TRAIL therapy and prospective developments for cancer treatment. J Control Release. 2020;326:335–349. doi: 10.1016/j.jconrel.2020.07.013. [DOI] [PubMed] [Google Scholar]
- 27.Snajdauf M, Havlova K, Vachtenheim J Jr, Ozaniak A, Lischke R, Bartunkova J, Smrz D, Strizova Z. The TRAIL in the treatment of human cancer: an update on clinical trials. Front Mol Biosci. 2021;8:628332. doi: 10.3389/fmolb.2021.628332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Almasan A, Ashkenazi A. Apo2L/TRAIL: apoptosis signaling, biology, and potential for cancer therapy. Cytokine Growth Factor Rev. 2003;14:337–348. doi: 10.1016/s1359-6101(03)00029-7. [DOI] [PubMed] [Google Scholar]
- 29.Stuckey DW, Shah K. TRAIL on trial: preclinical advances in cancer therapy. Trends Mol Med. 2013;19:685–694. doi: 10.1016/j.molmed.2013.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lemke J, von Karstedt S, Zinngrebe J, Walczak H. Getting TRAIL back on track for cancer therapy. Cell Death Differ. 2014;21:1350–1364. doi: 10.1038/cdd.2014.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Singh D, Tewari M, Singh S, Narayan G. Revisiting the role of TRAIL/TRAIL-R in cancer biology and therapy. Future Oncol. 2021;17:581–596. doi: 10.2217/fon-2020-0727. [DOI] [PubMed] [Google Scholar]
- 32.Ashkenazi A, Herbst RS. To kill a tumor cell: the potential of proapoptotic receptor agonists. J Clin Invest. 2008;118:1979–1990. doi: 10.1172/JCI34359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Takeda K, Hayakawa Y, Smyth MJ, Kayagaki N, Yamaguchi N, Kakuta S, Iwakura Y, Yagita H, Okumura K. Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nat Med. 2001;7:94–100. doi: 10.1038/83416. [DOI] [PubMed] [Google Scholar]
- 34.Oh YT, Sun SY. Regulation of cancer metastasis by TRAIL/death receptor signaling. Biomolecules. 2021;11:499. doi: 10.3390/biom11040499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sato K, Hida S, Takayanagi H, Yokochi T, Kayagaki N, Takeda K, Yagita H, Okumura K, Tanaka N, Taniguchi T, Ogasawara K. Antiviral response by natural killer cells through TRAIL gene induction by IFN-alpha/beta. Eur J Immunol. 2001;31:3138–3146. doi: 10.1002/1521-4141(200111)31:11<3138::aid-immu3138>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
- 36.Wu GS, Burns TF, McDonald ER 3rd, Jiang W, Meng R, Krantz ID, Kao G, Gan DD, Zhou JY, Muschel R, Hamilton SR, Spinner NB, Markowitz S, Wu G, El-Deiry WS. KILLER/DR5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet. 1997;17:141–143. doi: 10.1038/ng1097-141. [DOI] [PubMed] [Google Scholar]
- 37.Wu GS, Kim K, El-Deiry WS. KILLER/DR5, a novel DNA-damage inducible death receptor gene, links the p53-tumor suppressor to caspase activation and apoptotic death. Adv Exp Med Biol. 2000;465:143–151. doi: 10.1007/0-306-46817-4_13. [DOI] [PubMed] [Google Scholar]
- 38.Wu GS, Burns TF, Zhan Y, Alnemri ES, El-Deiry WS. Molecular cloning and functional analysis of the mouse homologue of the KILLER/DR5 tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor. Cancer Res. 1999;59:2770–2775. [PubMed] [Google Scholar]
- 39.Takimoto R, El-Deiry WS. Wild-type p53 transactivates the KILLER/DR5 gene through an intronic sequence-specific DNA-binding site. Oncogene. 2000;19:1735–1743. doi: 10.1038/sj.onc.1203489. [DOI] [PubMed] [Google Scholar]
- 40.Wang S, El-Deiry WS. Requirement of p53 targets in chemosensitization of colonic carcinoma to death ligand therapy. Proc Natl Acad Sci U S A. 2003;100:15095–15100. doi: 10.1073/pnas.2435285100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang S, El-Deiry WS. Inducible silencing of KILLER/DR5 in vivo promotes bioluminescent colon tumor xenograft growth and confers resistance to chemotherapeutic agent 5-fluorouracil. Cancer Res. 2004;64:6666–6672. doi: 10.1158/0008-5472.CAN-04-1734. [DOI] [PubMed] [Google Scholar]
- 42.Kim K, Fisher MJ, Xu SQ, El-Deiry WS. Molecular determinants of response to TRAIL in killing of normal and cancer cells. Clin Cancer Res. 2000;6:335–346. [PubMed] [Google Scholar]
- 43.Finnberg N, Gruber JJ, Fei P, Rudolph D, Bric A, Kim SH, Burns TF, Ajuha H, Page R, Wu GS, Chen Y, McKenna WG, Bernhard E, Lowe S, Mak T, El-Deiry WS. DR5 knockout mice are compromised in radiation-induced apoptosis. Mol Cell Biol. 2005;25:2000–2013. doi: 10.1128/MCB.25.5.2000-2013.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Finnberg N, Klein-Szanto AJ, El-Deiry WS. TRAIL-R deficiency in mice promotes susceptibility to chronic inflammation and tumorigenesis. J Clin Invest. 2008;118:111–123. doi: 10.1172/JCI29900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kuribayashi K, Krigsfeld G, Wang W, Xu J, Mayes PA, Dicker DT, Wu GS, El-Deiry WS. TNFSF10 (TRAIL), a p53 target gene that mediates p53-dependent cell death. Cancer Biol Ther. 2008;7:2034–2038. doi: 10.4161/cbt.7.12.7460. [DOI] [PubMed] [Google Scholar]
- 46.Allen JE, Krigsfeld G, Mayes PA, Patel L, Dicker DT, Patel AS, Dolloff NG, Messaris E, Scata KA, Wang W, Zhou JY, Wu GS, El-Deiry WS. Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects. Sci Transl Med. 2013;5:171ra17. doi: 10.1126/scitranslmed.3004828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ralff MD, Jhaveri A, Ray JE, Zhou L, Lev A, Campbell KS, Dicker DT, Ross EA, El-Deiry WS. TRAIL receptor agonists convert the response of breast cancer cells to ONC201 from anti-proliferative to apoptotic. Oncotarget. 2020;11:3753–3769. doi: 10.18632/oncotarget.27773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ray JE, Ralff MD, Jhaveri A, Zhou L, Dicker DT, Ross EA, El-Deiry WS. Antitumorigenic effect of combination treatment with ONC201 and TRAIL in endometrial cancer in vitro and in vivo. Cancer Biol Ther. 2021;22:554–563. doi: 10.1080/15384047.2021.1977067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jhaveri AV, Zhou L, Ralff MD, Lee YS, Navaraj A, Carneiro BA, Safran H, Prabhu VV, Ross EA, Lee S, El-Deiry WS. Combination of ONC201 and TLY012 induces selective, synergistic apoptosis in vitro and significantly delays PDAC xenograft growth in vivo. Cancer Biol Ther. 2021;22:607–618. doi: 10.1080/15384047.2021.1976567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Parker CS, Zhou L, Prabhu VV, Lee S, Miner TJ, Ross EA, El-Deiry WS. ONC201/TIC10 plus TLY012 anti-cancer effects via apoptosis inhibitor downregulation, stimulation of integrated stress response and death receptor DR5 in gastric adenocarcinoma. Am J Cancer Res. 2023;13:6290–6312. [PMC free article] [PubMed] [Google Scholar]
- 51.Lulla AR, Zhou Y, Ralff MD, Lev A, Dicker DT, El-Deiry WS. miR-3132 upregulates surface TRAIL to induce apoptotic cell death in cancer cells. Am J Cancer Res. 2022;12:315–326. [PMC free article] [PubMed] [Google Scholar]
- 52.Guicciardi ME, Gores GJ. Paving the TRAIL to anti-fibrotic therapy. Hepatology. 2016;64:29–31. doi: 10.1002/hep.28520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lu B, Peng L, Luo S, Zhou J, Xu N, Dong C, Yan Z, Li H, Li Q. Long circulation of PEG-TRAIL improves anti-hepatic fibrosis effect of TRAIL via targeting activated hepatic stellate cells. Front Mater. 2021;8:682844. [Google Scholar]
- 54.Habiel DM, Moreira AP, Ismailoglu UB, Dunleavy MP, Cavassani KA, van Rooijen N, Coelho AL, Hogaboam CM. TRAIL-dependent resolution of pulmonary fibrosis. Mediators Inflamm. 2018;2018:7934362. doi: 10.1155/2018/7934362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Oh Y, Park O, Swierczewska M, Hamilton JP, Park JS, Kim TH, Lim SM, Eom H, Jo DG, Lee CE, Kechrid R, Mastorakos P, Zhang C, Hahn SK, Jeon OC, Byun Y, Kim K, Hanes J, Lee KC, Pomper MG, Gao B, Lee S. Systemic PEGylated TRAIL treatment ameliorates liver cirrhosis in rats by eliminating activated hepatic stellate cells. Hepatology. 2016;64:209–223. doi: 10.1002/hep.28432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chae SY, Kim TH, Park K, Jin CH, Son S, Lee S, Youn YS, Kim K, Jo DG, Kwon IC, Chen X, Lee KC. Improved antitumor activity and tumor targeting of NH(2)-terminal-specific PEGylated tumor necrosis factor-related apoptosis-inducing ligand. Mol Cancer Ther. 2010;9:1719–1729. doi: 10.1158/1535-7163.MCT-09-1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Koizumi M, Hiasa Y, Kumagi T, Yamanishi H, Azemoto N, Kobata T, Matsuura B, Abe M, Onji M. Increased B cell-activating factor promotes tumor invasion and metastasis in human pancreatic cancer. PLoS One. 2013;8:e71367. doi: 10.1371/journal.pone.0071367. [DOI] [PMC free article] [PubMed] [Google Scholar]