scholarly journals Early macrophage infiltrates impair pancreatic cancer cell growth by TNF-α secretion

2020 ◽  
Author(s):  
Cansu Tekin ◽  
Hella L Aberson ◽  
Maarten F Bijlsma ◽  
C. Arnold Spek

Abstract Background: Pancreatic ductal adenocarcinoma (PDAC) is a grim disease with high mortality rates. Increased macrophage influx in PDAC is a common hallmark and associated with poor prognosis. Macrophages have high cellular plasticity, which can differentiate into both anti- and pro-tumorigenic properties. Here, we investigated how naïve (M0) macrophages differ from other macrophages in their anti-tumorigenic activities.Methods: In vitro BrdU proliferation and Annexin V cell death analyses were performed on PANC-1 and MIA PaCa-2 PDAC cell lines exposed to conditioned medium of different macrophage subsets. Macrophage secreted factors were measured by transcript analysis and ELISA. Therapeutic antibodies were used to functionally establish the impact of the identified cytokine on PDAC proliferation.Results: Proliferation and cell death assays revealed that only M0 macrophages harbor anti-tumorigenic activities and that M1, M2, and TAMs do not. mRNA analysis and ELISA results suggested TNF-α as a potential candidate to mediate M macrophage induced cell death. To demonstrate the importance of TNF-α in M macrophage-induced cell death, PANC-1 and MIA PaCa-2 cell-lines were exposed to M macrophage conditioned medium in the presence of the TNF-α inhibitor Infliximab, which effectively diminished the anti-tumor activities of M0 macrophages.Conclusion: Newly tumor-infiltrated naive M0 macrophages exert anti-tumorigenic activities via TNF-α secretion. Their subsequent differentiation into either M1, M2, or TAM subsets reduces TNF-α levels, thereby abolishing their cytotoxic activity on PDAC cells. These data suggest that reestablishing TNF-α secretion in differentiated macrophages might yield a therapeutic benefit.

BMC Cancer ◽  
2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Cansu Tekin ◽  
Hella L. Aberson ◽  
Maarten F. Bijlsma ◽  
C. Arnold Spek

Abstract Background Pancreatic ductal adenocarcinoma (PDAC) is a grim disease with high mortality rates. Increased macrophage influx in PDAC is a common hallmark and associated with poor prognosis. Macrophages have high cellular plasticity, which can differentiate into both anti- and pro-tumorigenic properties. Here, we investigated how naïve (M0) macrophages differ from other macrophages in their anti-tumorigenic activities. Methods In vitro BrdU proliferation and Annexin V cell death analyses were performed on PANC-1 and MIA PaCa-2 PDAC cell lines exposed to conditioned medium of different macrophage subsets. Macrophage secreted factors were measured by transcript analysis and ELISA. Therapeutic antibodies were used to functionally establish the impact of the identified cytokine on PDAC proliferation. Results Proliferation and cell death assays revealed that only M0 macrophages harbor anti-tumorigenic activities and that M1, M2, and TAMs do not. mRNA analysis and ELISA results suggested TNF-α as a potential candidate to mediate M0 macrophage induced cell death. To demonstrate the importance of TNF-α in M0 macrophage-induced cell death, PANC-1 and MIA PaCa-2 cell-lines were exposed to M0 macrophage conditioned medium in the presence of the TNF-α inhibitor Infliximab, which effectively diminished the anti-tumor activities of M0 macrophages. Conclusion Newly tumor-infiltrated naive M0 macrophages exert anti-tumorigenic activities via TNF-α secretion. Their subsequent differentiation into either M1, M2, or TAM subsets reduces TNF-α levels, thereby abolishing their cytotoxic activity on PDAC cells. These data suggest that reestablishing TNF-α secretion in differentiated macrophages might yield a therapeutic benefit.


2020 ◽  
Author(s):  
Cansu Tekin ◽  
Hella L Aberson ◽  
Maarten F Bijlsma ◽  
C. Arnold Spek

Abstract Background: Pancreatic ductal adenocarcinoma (PDAC) is a disease with high mortality rates. Increased macrophage influx in PDAC is common and associated with poor prognosis. Macrophages are highly plastic cells that can exhibit both anti- and pro-tumorigenic properties. Here, we investigated how naïve (M0) macrophages differ from other macrophages in their anti-tumorigenic activities.Methods: In vitro BrdU proliferation and Annexin V cell death analyses were performed on PANC-1 and MIA PaCa-2 PDAC cell lines exposed to conditioned medium of different macrophage subsets. Macrophage secreted molecules were measured by transcript analysis and ELISA. Therapeutic antibodies were used to functionally establish the impact of the identified cytokine on PDAC proliferation.Results: Proliferation and cell death assays revealed that only M0 macrophages harbor anti-tumorigenic activities and that M1, M2, and TAMs do not. mRNA analysis and ELISA results suggested TNF-a as a potential candidate to mediate M0 macrophage induced cell death. To demonstrate the importance of TNF-a in M0 macrophage-induced cell death, PANC-1 and MIA PaCa-2 cell-lines were exposed to M0 macrophage conditioned medium in the presence of the TNF-a inhibitor Infliximab, which effectively diminished the anti-tumor activities of M0 macrophages.Conclusion: Newly tumor-infiltrated naive M0 macrophages exert anti-tumorigenic activities via TNF-a secretion. Their subsequent differentiation into either M1, M2, or TAM subsets reduces TNF-a levels, thereby abolishing their cytotoxic activity on PDAC cells. These data suggest that reestablishing TNF-a secretion in differentiated macrophages might yield a therapeutic benefit.


2020 ◽  
Author(s):  
Cansu Tekin ◽  
Hella L Aberson ◽  
Maarten F Bijlsma ◽  
C. Arnold Spek

Abstract Background: Pancreatic ductal adenocarcinoma (PDAC) is a disease with high mortality rates. Increased macrophage influx in PDAC is common and associated with poor prognosis. Macrophages are highly plastic cells that can exhibit both anti- and pro-tumorigenic properties. Here, we investigated how naïve (M0) macrophages differ from other macrophages in their anti-tumorigenic activities.Methods: In vitro BrdU proliferation and Annexin V cell death analyses were performed on PANC-1 and MIA PaCa-2 PDAC cell lines exposed to conditioned medium of known macrophage subsets. Macrophage secreted molecules were measured by transcript analysis and ELISA. Therapeutic antibodies were used to functionally establish the role of the identified cytokine in PDAC proliferation.Results: Proliferation and cell death assays revealed that only M0 macrophages exert anti-tumorigenic activities and that M1, M2, and TAMs do not. mRNA analysis and ELISA results suggested TNF-a as a potential candidate to mediate M0 macrophage induced cell death. To prove or refute the importance of TNF-a in M0 macrophage-induced cell death, PANC-1 and MIA PaCa-2 cell-lines were exposed to M0 macrophage conditioned medium in the presence of the TNF-a inhibitor infliximab, which effectively diminished the anti-tumor activities of M0 macrophages.Conclusion: Freshly infiltrated naive M0 macrophages exert anti-tumorigenic activities via TNF-a secretion. Their subsequent differentiation into either M1, M2, or TAM subsets reduces TNF-a levels, thereby abolishing their cytotoxic activity on PDAC cells. These data suggest that the restoration of TNF-a secretion in differentiated macrophages might yield a therapeutic benefit.


2019 ◽  
Vol 20 (15) ◽  
pp. 1288-1308
Author(s):  
Tahir Maqbool ◽  
Sana J. Awan ◽  
Sabeen Malik ◽  
Faheem Hadi ◽  
Somia Shehzadi ◽  
...  

Background: Natural product with apoptotic activity could serve as a potential new source for anti-cancer medicine. Numerous phytochemicals from plants have shown to exert antineoplastic effects via programmed cell death (apoptosis). Cancer is one of the leading causes of death in prosperous countries. The subject study was intended to evaluate the anticancer properties of Kalonji extracts against cancer cell lines HeLa and HepG2 and normal cell lines BHK and VERO were used as normal controls. Materials & Methods: For the evaluation of anti-proliferative effects, cell viability and cell death in all groups of cells were evaluated via MTT, crystal violet and trypan blue assays. For the evaluation of angiogenesis, Immunocytochemistry and ELISA of VEGF were done. Immunocytochemistry and ELISA of Annexin-V and p53 were performed for the estimation of apoptosis in all groups of cells. Furthermore, LDH assay, antioxidant enzymes activity (GSH, APOX, CAT and SOD) and RT-PCR with proliferative and apoptotic markers along with internal control were also performed. Cancer cells of both cell lines HepG2 and HeLa cells showed reduced viability, angiogenesis and proliferation with increased apoptosis when treated with Kalonji extracts. Whereas anti-oxidative enzymes show enhanced levels in treated cancer cells as compared to untreated ones. Conclusion: It was observed that Kalonji extracts have the ability to induce apoptosis and improve the antioxidant status of HeLa and HepG2 cells. They can also inhibit the proliferation and angiogenesis in both these cancer cell lines.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4459-4459
Author(s):  
Morris Kletzel ◽  
Sarah C. Tallman ◽  
Marie Olszewski ◽  
Wei Huang

Abstract Objective: While busulfan is a commonly used chemotherapeutic agent in the treatment of many hematological diseases, its effectiveness against neuroblastoma is still in question. This study aims to assess the degree of apoptosis and cell death in neuroblastoma cell lines and primary neuroblastoma tumors when exposed to varying doses of busulfan. Materials and Methods: Cultures from established cell lines SKN-SH, SKN-DOX-R, IMR-5, and NGP (n=4), as well as cultures from primary tumors (n=2) were seeded at 106 cells/ml in RPMI640 supplemented with 10% fetal bovine serum (FBS) and transferred to 24-well plates, where cells were exposed to 1ml of busulfan at 0, 0.001, 0.005, 0.01, 0.05, and 0.1mg/ml per well. Cells were incubated at 37°C in a humidified atmosphere of 5% CO2 for 72 hours. Wells were sacrificed after 0, 6, 24, 48 and 72 hours and tested with Annexin V and PI; 10,000 events were measured by flow cytometry. The percentage of apoptotic and dead cells was plotted in a graph and a t-test was performed against the untreated control. Results: After 24 hours, there was a significant decrease in cell viability of each dose when compared to the control untreated cells (p<0.005). 24 Hour % Cell Viability for Varying Doses of Busulfan (mg/ml) Dose 0 Dose 0.001 Dose 0.005 Dose 0.01 Dose 0.05 Dose 0.1 Mean 66.1 44.4 40.3 40.7 37.7 39 SEM 5.56 5.17 5.96 6.17 6.03 5.60 Median 65 33.5 38 39 37 31 Range 39 to 97 14 to 87 4 to 89 6 to 93 4 to 77 5 to 88 The overall mean decrease in cell viability when compared to the control was 25.7%. However, there were only modest differences in effectiveness when comparing the doses, with an average of only 5–7% difference between doses. Further, there was much variability between the different cell lines, some with changes in apoptosis and cell death of over 50%, while other lines showed no changes at all. Limited differences were seen after 6 hours, and after 72 hours any effect of busulfan was masked by cell death due to other factors, as seen through increased cell death in untreated cells. Conclusion: Busulfan induced apoptosis and cell death in vitro in neuroblastoma cell lines at a mean of 76.43% for non-resistant lines, 59.33% for primary tumors and 35% for resistant cell lines (at middle dose 0.01mg/ml). The resistance of certain cell lines confirms the difficulties of treating multi-drug resistant cells in often heterogeneous neuroblastoma tumors. That some cell lines were responsive shows the potential of using busulfan to treat neuroblastoma in the future.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1589-1589
Author(s):  
Dirk Winkler ◽  
Thorsten Zenz ◽  
Daniel Mertens ◽  
Annett Habermann ◽  
Hartmut Döhner ◽  
...  

Abstract The PI3K/AKT pathway acts as a critical regulator of cell survival by stimulating cell proliferation and inhibiting apoptosis and has been implicated in the pathogenesis of lymphoproliferative disorders. Therefore, inhibition of AKT seems to be a highly attractive new approach for the treatment of lymphoma. We treated 9 cell lines with AKT-nhibitor (1, 10, 20 μM) over 24h and 48h respectively: EHEB (B-CLL), GRANTA-519 (MCL), JURKAT (T-ALL) BL-60, NAMALWA and BJAB (all Burkitt’s lymphoma), L363, OPM-2 and RPMI-8226 (all multiple myeloma). To determine the rates and type of AKT-inhibitor induced cell death, FACS analyses for CD19, 7AAD, active caspase-3, cytochrome c were performed. The phosphorylation status of AKT and its downstream proteins GSK3β, p70S6k and S6 was studied by Western blotting after 5–120 minutes. In addition, 11 primary CLL samples with either del 13q (n=3), del 11q (n=2), del 17p (n=3) or a normal karyotype (n=3) were treated with AKT inhibitor (10 μM; 2.5μM; 0.625 μM; 0.156 μM). CLL samples were cultured in both standard medium as well as in HS5-(human stromal cells) conditioned medium to reduce spontaneous apoptosis of CLL in-vitro. 6 out of 11 patients had unmutated VH genes. 8 Patients were untreated, 3 were previously treated. Fludarabine (0.1 μM) was added to AKT-inhibitor in 11 cases to test for synergistic effects. CLL cells were harvested after 48 hours and 5 days to measure cell viability using Celltiter-GLO-Assay. Treatment of cell lines lead to significant rates of AKT-inhibitor induced cell death (table 1), to hyperphosphorylation of AKT and to inhibition of phosphorylation of GSK3β (after 5 min) and S6 (after 20 min) in all cell lines and of p70S6k (after 120 min) in GRANTA, JURKAT, NAMALWA and BJAB. Cell death did not depend on functional p53 gene. Treatment of primary CLL samples with AKT-inhibitor alone was followed by a decrease of cell viability in a time and concentration dependent manner regardless of the medium used (table 2). Only with the lowest concentration and when cultured in HS5-conditioned medium, no further reduction of viable cells was seen between 48h and 5d. Treatment with AKT-inhibitor as a single agent seemed to be at least as effective as treatment with fludarabine. Response was independent of the genetic subgroup, VH mutation status or prior treatment. High risk cases with del 17p responded worse to fludarabine alone when compared to cases without del 17p (i.e. 75% of viable cells after 5d at 10000 μM in cases with del 17p vs. 25% in cases without del 17p). The same fludarabine resistant cases showed good responses to treatment with AKT-inhibitor (9% of viable cells after 5d at 10000 μM in cases with del 17p). A synergistic effect was not achieved by combining AKT-inhibitor and fludarabine. Culture of CLL cells in HS5-conditioned medium resulted in lower rates of spontaneous apoptosis, but also in lower rates of AKT-inhibitor induced cell death. In conclusion, in-vitro treatment with AKT-inhibitor resulted in significant rates of cell death in cell lines and primary CLL cells, even in patients with del 17p or resistance to fludarabine. In cell lines, treatment with AKT-inhibitor was followed by typical features of apoptosis such as activation of caspase-3 and cytochrome c release. In CLL samples, prior treatment did not affect in-vitro response rates. These data underline the involvement of the PI3K/Akt pathway in the pathogenesis of lymphoma and point to an efficacy of the AKT-inhibitor in the treatment of lymphoma, multiple myeloma and CLL in-vivo. Concerning CLL, the AKT-inhibitor seems to be an attractive new treatment option even for cases with high risk cytogenetics. Using HS5-conditioned medium seems to be a well functioning method to reduce spontaneous apoptosis of CLL cells in-vitro. Table 1: rates of cell death, caspase-3 activation and cytochrome c release after treatment of cell lines with AKT inhibitor (1μM, 48h) 7AAD-positive cells active caspase-3 cytochrome c release EHEB 15% − + GRANTA-519 15% + + JURKAT 17% + + BL60 24% + + NAMALWA 25% − (+) BJAB 30% + (+) L363 15% + − OPM-2 41% + + RPMI-8226 32% + (+) Table 2: mean percentage of viable cells after treatment with AKT-Inhibitor (A), fludarabine (F; 0,1μM) and their combination (A + F) measured by Celltiter-GLO-Assay 10000 nM 2500 nM 625 nM 156,25 nM 48h 5d 48h 5d 48h 5d 48h 5d A F A + F A F A + F A A F A+ F A F A+ F A HS5 + (n=8) 94 84 (n=5) 75 (n=5) 45 22 (n=5) 25 (n=5) 88 52 91 84 80 69 22 18 76 85 HS5 − (n=11) 60 79 59 8 39 21 77 27 80 79 76 28 39 34 82 (n=10) 21


Toxins ◽  
2019 ◽  
Vol 11 (9) ◽  
pp. 506 ◽  
Author(s):  
Tony Haykal ◽  
Peter Nasr ◽  
Mohammad H. Hodroj ◽  
Robin I. Taleb ◽  
Rita Sarkis ◽  
...  

Annona cherimola Mill is a large green fruit with black seeds widely known to possess toxic properties due to the presence of Annonaceous acetogenins. The present study investigates the anti-cancer properties of an Annona cherimola Mill ethanolic seed extract on Acute Myeloid Leukemia (AML) cell lines in vitro and elucidates the underlying cellular mechanism. The anti-proliferative effects of the extract on various AML cell lines and normal mesenchymal cells (MSCs) were assessed using WST-1 viability reagent. The pro-apoptotic effect of the extract was evaluated using Annexin V/PI staining and Cell Death ELISA. The underlying mechanism was deciphered by analyzing the expression of various proteins using western blots. Treatment with an A. cherimola seed ethanolic extract promotes a dose- and time-dependent inhibition of the proliferation of various AML cell lines, but not MSCs. Positive Annexin V staining, as well as DNA fragmentation, confirm an increase in apoptotic cell death by upregulating the expression of pro-apoptotic proteins which control both intrinsic and extrinsic pathways of apoptosis. GC/MS analysis revealed the presence of phytosterols, in addition to other bioactive compounds. In conclusion, Annona cherimola Mill seed extract, previously known to possess a potent toxic activity, induces apoptosis in AML cell lines by the activation of both the extrinsic and the intrinsic pathways.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3936-3936
Author(s):  
Yongwei Su ◽  
Xinyu Li ◽  
Holly Edwards ◽  
Lisa Polin ◽  
Juiwanna Kushner ◽  
...  

Abstract Although standard induction therapy initially elicits a promising response in the majority of acute myeloid leukemia (AML) patients, the majority relapse. Leukemia stem cells (LSCs) that survive chemotherapy are believed to be responsible for AML relapse. Therefore, new therapies that eliminate LSCs are desperately needed. ONC201 is a TRAIL inducer and the founding member of the imipridone family. It has been shown to induce apoptosis in LSCs (Ishizawa et al, Science Signaling. 2016; 9:ra17). ONC201 was chemically modified to increase the potency and selectivity against cancer cells, resulting in the new analog ONC213. In this study, we investigated the antileukemic activity and the underlying molecular mechanism of ONC213 in preclinical AML models. ONC213 activity in AML cell lines and primary AML patient samples was first tested in vitro. MTT assay results revealed that ONC213 IC50s ranged from 91.7 nM to 2.4 µM in AML cell lines and primary AML patient samples, which are achievable in vivo based on results from a PK study in mice (a single dose of 50 and 100 mg/kg ONC213 resulted in peak plasma concentrations of 3.7 μM and 8 μM, respectively). Annexin V/propidium iodide staining and flow cytometry analysis results showed variable responses for the AML cell lines tested. After 48 h treatment with 500 nM ONC213, striking induction of cell death in MOLM-13 and MV4-11 cells was detected (at least 72% Annexin V+ cells), while THP-1 and U937 cells showed little to no increase in Annexin V+ cells (6-11%). Similar results were obtained in primary AML patient samples. In contrast to the 48 h treatment of THP-1 and U937 cells, increasing the treatment duration to 120 h resulted in greater than 50% Annexin V+ cells, suggesting that a longer exposure time is necessary in some cell lines. In MV4-11 and MOLM-13 cells, initiation of cell death was detected 8 to 12 h post ONC213 treatment. Colony formation assays revealed that ONC213 treatment significantly reduced colony formation capacity of primary AML patient samples to less than 5% compared to vehicle control, while having no significant effect on normal hematopoietic progenitor cells. A primary AML patient sample was treated with or without ONC213 for 48 h, transplanted into NSG mice, and ten weeks later bone marrow was harvested and human CD45+ cells were measured. ONC213 treatment significantly reduced human AML engraftment compared to vehicle control (0.6% vs. 21.3%; p<0.05), demonstrating that ONC213 kills LSCs in vitro. Next, we examined in vivo efficacy of ONC213 against an AML cell line derived xenograft mouse model. MV4-11 cells were injected into NSGS mice through the tail vein. Three days post-injection, the mice were randomized into vehicle control or 125 mg/kg ONC213 cohorts (5 mice per cohort) and treated daily for 8 days. Modest weight loss was noted but was entirely manageable. ONC213 treatment extended the survival of mice by 88% (median survival 62 vs 33 days). Unlike ONC201, ONC213 treatment of AML cells did not increase the expression of TRAIL. Interestingly, RNAseq results showed that 500 nM ONC213 treatment for 48 h downregulated 33 mRNAs in the oxidative phosphorylation (OXPHOS) pathway, suggesting that ONC213 treatment decreases OXPHOS in AML cells. Thus far, six of the downregulated mRNAs (UQCRQ, SDHA, COX6C, NDUFS5, ATP5D, and NDUFB1) were verified by real-time RT-PCR after both 8 h and 48 h ONC213 treatment. LSCs have been shown to be highly reliant on OXPHOS, while normal hematopoietic stem cells and some bulk AML cells can switch to glycolysis for ATP production during times of OXPHOS inhibition. Thus, ONC213 may kill LSCs through inhibition of OXPHOS. In addition to downregulation of OXPHOS related genes, we found that ONC213 treatment downregulates Mcl-1. Since Mcl-1 mediates resistance to the promising Bcl-2-selective inhibitor ABT-199 (Venetoclax) and inhibition of Bcl-2 impairs OXPHOS, ONC213 would likely synergize with ABT-199 in AML cells. Indeed, combined treatment resulted in striking synergistic induction of apoptosis in both AML cell lines and primary patient samples. Enhanced cell death was detected 8 h post combination treatment in both MOLM-13 and MV4-11 cells. Results from colony formation assays revealed that the combination spares normal hematopoietic progenitor cells. Taken together, ONC213 is active as a single agent and in combination with ABT-199 in AML. Disclosures Allen: Oncoceutics: Employment. Stogniew:Oncoceutics: Employment. Prabhu:Oncoceutics: Employment. Ge:MEI Pharma: Research Funding.


2017 ◽  
Vol 6 (04) ◽  
pp. 5360 ◽  
Author(s):  
Rajkiran Reddy Banala ◽  
Satish Kumar Vemuri ◽  
Gurava Reddy A.V. ◽  
Subbaiah G.P.V.*

Psoriasis is a chronic inflammatory skin disorder characterized by rapid proliferation of keratinocytes and incomplete keratinization. Discovery of safer and more effective anti-psoriatic drugs remains an area of active research at the present time. A431 and B16-F10 cell lines were used as in vitro models. In the present study, we aimed at assessing the Anti-psoriatic activity of aqueous extract of Acalypha indica. We analyzed the efficiency of A. indica leaf extract in inducing cell death and apoptosis in these cell lines. The cell death (Propidium iodide) and apoptosis (Annexin V) was assessed by fluorescence studies and we observed 80% of cell death and 75% of apoptosis in both cell lines. Therefore, this in vitro study suggested that the leaf extract is capable of serving as anti-psoriasis agent or compound.


2021 ◽  
Author(s):  
Maria E Monberg ◽  
Heather Geiger ◽  
Roshan Sharma ◽  
Jaewon J Lee ◽  
Alexander Semaan ◽  
...  

Intratumoral heterogeneity (ITH) is a hallmark of cancer. The advent of single-cell technologies has helped uncover ITH in a high-throughput manner in different cancers across varied contexts. Here we apply single-cell sequencing technologies to reveal striking ITH in assumptively oligoclonal pancreatic ductal adenocarcinoma (PDAC) cell lines. Our findings reveal a high degree of both genomic and transcriptomic heterogeneity in established and globally utilized PDAC cell lines, custodial variation induced by growing apparently identical PDAC cell lines in different laboratories, and profound transcriptomic shifts in transitioning from 2D to 3D spheroid growth models. Our findings also call into question the validity of widely available immortalized, non-transformed pancreatic lines as contemporaneous control lines in experiments. Further, while patient-derived organoid (PDOs) are known to reflect the cognate in vivo biology of the parental tumor, we identify transcriptomic shifts during ex vivo passage that might hamper their predictive abilities over time. The impact of these findings on rigor and reproducibility of experimental data generated using established preclinical PDAC models between and across laboratories is uncertain, but a matter of concern.


Sign in / Sign up

Export Citation Format

Share Document