scholarly journals A First-In-Class, Humanized Antibody Targeting Alternatively Spliced Tissue Factor: Preclinical Evaluation in an Orthotopic Model of Pancreatic Ductal Adenocarcinoma

2021 ◽  
Vol 11 ◽  
Author(s):  
Clayton S. Lewis ◽  
Aniruddha Karve ◽  
Kateryna Matiash ◽  
Timothy Stone ◽  
Jingxing Li ◽  
...  

In 2021, pancreatic ductal adenocarcinoma (PDAC) is the 3rd leading cause of cancer deaths in the United States. This is largely due to a lack of symptoms and limited treatment options, which extend survival by only a few weeks. There is thus an urgent need to develop new therapies effective against PDAC. Previously, we have shown that the growth of PDAC cells is suppressed when they are co-implanted with RabMab1, a rabbit monoclonal antibody specific for human alternatively spliced tissue factor (asTF). Here, we report on humanization of RabMab1, evaluation of its binding characteristics, and assessment of its in vivo properties. hRabMab1 binds asTF with a KD in the picomolar range; suppresses the migration of high-grade Pt45.P1 cells in Boyden chamber assays; has a long half-life in circulation (~ 5 weeks); and significantly slows the growth of pre-formed orthotopic Pt45.P1 tumors in athymic nude mice when administered intravenously. Immunohistochemical analysis of tumor tissue demonstrates the suppression of i) PDAC cell proliferation, ii) macrophage infiltration, and iii) neovascularization, whereas RNAseq analysis of tumor tissue reveals the suppression of pathways that promote cell division and focal adhesion. This is the first proof-of-concept study whereby a novel biologic targeting asTF has been investigated as a systemically administered single agent, with encouraging results. Given that hRabMab1 has a favorable PK profile and is able to suppress the growth of human PDAC cells in vivo, it comprises a promising candidate for further clinical development.

2021 ◽  
Author(s):  
Katrin J Ciecielski ◽  
Antonio Mulero-Sanchez ◽  
Alexandra Berninger ◽  
Laura Ruiz Canas ◽  
Astrid Bosma ◽  
...  

Mutant KRAS is present in over 90% of pancreatic as well as 30-40% of lung and colorectal cancers and is one of the most common oncogenic drivers. Despite decades of research and the recent emergence of isoform-specific KRASG12C-inhibitors, most mutant KRAS isoforms, including the ones frequently associated with pancreatic ductal adenocarcinoma (PDAC), cannot be targeted directly. Moreover, targeting single RAS downstream effectors induces adaptive mechanisms leading to tumor recurrence or resistance. We report here on the combined inhibition of SHP2, a non-receptor tyrosine phosphatase upstream of KRAS, and ERK, a serine/threonine kinase and a key molecule downstream of KRAS in PDAC. This combination shows synergistic anticancer activity in vitro, superior disruption of the MAPK pathway, and significantly increased apoptosis induction compared to single-agent treatments. In vivo, we demonstrate good tolerability and efficacy of the combination. Concurrent inhibition of SHP2 and ERK induces significant tumor regression in multiple PDAC mouse models. Finally, we show evidence that 18F-FDG PET scans can be used to detect and predict early drug responses in animal models. Based on these compelling results, we will investigate this drug combination in a clinical trial (SHERPA, SHP2 and ERK inhibition in pancreatic cancer, NCT04916236), enrolling patients with KRAS-mutant PDAC.


Pancreatology ◽  
2013 ◽  
Vol 13 (3) ◽  
pp. S19
Author(s):  
Rainer Heuchel ◽  
Marco Gerling ◽  
Ying Zhao ◽  
Jithin Jose ◽  
Jessica Norberg ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1486-1486
Author(s):  
Dusten Unruh ◽  
Farah Sagin ◽  
Mariette Adam ◽  
Patrick Van Dreden ◽  
Barry J Woodhams ◽  
...  

Abstract Tissue Factor (TF) present in blood cells and plasma is referred to as blood-borne or circulating TF. TF has been implicated in the pathogenesis of several chronic disease states, most notably cardiovascular disease/thrombosis, diabetes, and cancer. Full-length TF is an integral membrane protein while alternatively spliced TF can be secreted in a free form and features a unique C-terminal domain enabling its selective detection in bio-specimens. Recently, asTF was shown to circulate in the blood of metastatic breast cancer patients at concentrations exceeding 1 ng/mL (Kocaturk et al, PNAS 2013), and it promoted tumor growth and spread in an orthotopic model of pancreatic ductal adenocarcinoma (PDAC, Unruh et al, Int J Cancer, 2014). asTF protein acts as a cell agonist driving angiogenesis, cancer cell proliferation, and monocyte recruitment via integrin binding. It is not known whether circulating asTF may contribute to or serve as a biomarker in patients suffering from cardiovascular disease, diabetes, and/or solid cancers including PDAC. We evaluated circulating asTF in healthy subjects and individuals with ongoing acute coronary syndrome (ACS); diabetes mellitus (DM); ongoing ACS+DM; and PDAC. Samples of platelet poor plasma from 204 subjects were obtained from University of Cincinnati Cancer Institute’s Tumor Bank and Diagnostica Stago collections, blood specimens drawn from emergency room visitors at four medical centers in the US, and George King Bio-Medical, Inc. ACS was defined by positive troponin levels; DM was self-identified. Blood was drawn into tubes containing heparin (ASC, DM, ACS+DM), acid citrate dextrose (PDAC), or sodium citrate (healthy subjects), centrifuged at 3000 rpm for 15 min at 4°C, and stored at -80°C until use. Blinded asTF ELISA was performed on plasma samples as per the prototype-tailored procedure (Diagnostica Stago). Samples with asTF concentrations ≥0.2 ng/mL were deemed positive. asTF concentrations are presented as mean±SD. Kruskal-Wallis one-way analysis of variance was used to compare differences in concentration levels between the cohorts; Chi-Square and/or Fisher’s exact test were used to compare proportions. asTF protein was detectable in the plasma of 3/19 (15.8%) subjects in the healthy cohort (CORE Set 50, George King Bio-Medical); 7/38 (18.4%) in the no ACS/no DM cohort (emergency room visitors’ control group); 2/40 (5%) in the DM cohort; 5/39 (12.8%) in the ACS cohort; 4/25 (16.0%) in the ACS/DM cohort; and 20/43 (46.5%) in the PDAC cohort; the proportion of PDAC patients positive for asTF was significantly higher compared to that in all other cohorts (p<0.01, Chi-Square test). The mean asTF concentrations in the cohorts were as follows: PDAC, 0.403±0.912 ng/mL; healthy subjects, 0.169±0.596 ng/mL; emergency room visitors’ control group, 0.159±0.357 ng/mL; ACS, 0.0925±0.258 ng/mL; DM, 0.0423±0.19 ng/mL; ACS+DM, 0.208±0.642 ng/mL; the differences between mean asTF levels in the cohorts did not reach significance. Next, we evaluated asTF’s potential as a biomarker to help detect a more aggressive PDAC phenotype. Among the 43 patients with PDAC, 36 were initially deemed resectable and 7 unresectable due to the presence of metastatic disease as determined by diagnostic screening; following exploratory laparoscopic surgery, 11 out of 36 patients initially deemed resectable were deemed unresectable due to the presence of metastatic disease. When the entire PDAC cohort was split into bona fide resectable (25) and unresectable (18) sub-cohorts, positivity for asTF was significantly more prevalent in the unresectable sub-cohort irrespective of the results of initial evaluation and/or pre-operative CA19-9 levels (asTF ≥0.2 ng/mL: 13 unresectable and 7 resectable patients; asTF<0.2 ng/mL: 5 unresectable and 18 resectable patients, p=0.0059, Fisher’s exact test). We here report that asTF at levels ≥0.2 ng/mL occurs more frequently in the plasma of patients with PDAC compared to healthy subjects and/or individuals with ACS, DM, and ACS/DM. Further, PDAC patients whose plasma asTF levels were equal to or exceeded 0.2 ng/mL had a significantly lower chance to qualify for tumor resection, irrespective of initial pre-surgical diagnostic evaluation. asTF may thus comprise a novel marker of aggressive PDAC phenotype with potential utility in patient stratification, warranting prospective evaluation of larger PDAC patient cohorts. Disclosures No relevant conflicts of interest to declare.


Oncotarget ◽  
2016 ◽  
Vol 7 (18) ◽  
pp. 25264-25275 ◽  
Author(s):  
Dusten Unruh ◽  
Betül Ünlü ◽  
Clayton S. Lewis ◽  
Xiaoyang Qi ◽  
Zhengtao Chu ◽  
...  

Cancers ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 3816
Author(s):  
Loryn Holokai ◽  
Jayati Chakrabarti ◽  
Joanne Lundy ◽  
Daniel Croagh ◽  
Pritha Adhikary ◽  
...  

Purpose: Pancreatic ductal adenocarcinoma (PDAC) has the lowest five-year survival rate of all cancers in the United States. Programmed death 1 receptor (PD-1)-programmed death ligand 1 (PD-L1) immune checkpoint inhibition has been unsuccessful in clinical trials. Myeloid-derived suppressor cells (MDSCs) are known to block anti-tumor CD8+ T cell immune responses in various cancers including pancreas. This has led us to our objective that was to develop a clinically relevant in vitro organoid model to specifically target mechanisms that deplete MDSCs as a therapeutic strategy for PDAC. Method: Murine and human pancreatic ductal adenocarcinoma (PDAC) autologous organoid/immune cell co-cultures were used to test whether PDAC can be effectively treated with combinatorial therapy involving PD-1 inhibition and MDSC depletion. Results: Murine in vivo orthotopic and in vitro organoid/immune cell co-culture models demonstrated that polymorphonuclear (PMN)-MDSCs promoted tumor growth and suppressed cytotoxic T lymphocyte (CTL) proliferation, leading to diminished efficacy of checkpoint inhibition. Mouse- and human-derived organoid/immune cell co-cultures revealed that PD-L1-expressing organoids were unresponsive to nivolumab in vitro in the presence of PMN-MDSCs. Depletion of arginase 1-expressing PMN-MDSCs within these co-cultures rendered the organoids susceptible to anti-PD-1/PD-L1-induced cancer cell death. Conclusions: Here we use mouse- and human-derived autologous pancreatic cancer organoid/immune cell co-cultures to demonstrate that elevated infiltration of polymorphonuclear (PMN)-MDSCs within the PDAC tumor microenvironment inhibit T cell effector function, regardless of PD-1/PD-L1 inhibition. We present a pre-clinical model that may predict the efficacy of targeted therapies to improve the outcome of patients with this aggressive and otherwise unpredictable malignancy.


2013 ◽  
Vol 134 (1) ◽  
pp. 9-20 ◽  
Author(s):  
Dusten Unruh ◽  
Kevin Turner ◽  
Ramprasad Srinivasan ◽  
Begüm Kocatürk ◽  
Xiaoyang Qi ◽  
...  

2011 ◽  
Vol 17 (22) ◽  
pp. 7015-7023 ◽  
Author(s):  
David Z. Chang ◽  
Ying Ma ◽  
Baoan Ji ◽  
Huamin Wang ◽  
Defeng Deng ◽  
...  

Radiology ◽  
2015 ◽  
Vol 277 (3) ◽  
pp. 644-661 ◽  
Author(s):  
Paul F. Laeseke ◽  
Ru Chen ◽  
R. Brooke Jeffrey ◽  
Teresa A. Brentnall ◽  
Jürgen K. Willmann

BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jung Hyun Jo ◽  
Sun A Kim ◽  
Jeong Hoon Lee ◽  
Yu Rang Park ◽  
Chanyang Kim ◽  
...  

Abstract Background Cancer stem cells (CSCs) are implicated in carcinogenesis, cancer progression, and recurrence. Several biomarkers have been described for pancreatic ductal adenocarcinoma (PDAC) CSCs; however, their function and mechanism remain unclear. Method In this study, secretome analysis was performed in pancreatic CSC-enriched spheres and control adherent cells for biomarker discovery. Glutaredoxin3 (GLRX3), a novel candidate upregulated in spheres, was evaluated for its function and clinical implication. Results PDAC CSC populations, cell lines, patient tissues, and blood samples demonstrated GLRX3 overexpression. In contrast, GLRX3 silencing decreased the in vitro proliferation, migration, clonogenicity, and sphere formation of cells. GLRX3 knockdown also reduced tumor formation and growth in vivo. GLRX3 was found to regulate Met/PI3K/AKT signaling and stemness-related molecules. ELISA results indicated GLRX3 overexpression in the serum of patients with PDAC compared to that in healthy controls. The sensitivity and specificity of GLRX3 for PDAC diagnosis were 80.0 and 100%, respectively. When GLRX3 and CA19–9 were combined, sensitivity was significantly increased to 98.3% compared to that with GLRX3 or CA19–9 alone. High GLRX3 expression was also associated with poor disease-free survival in patients receiving curative surgery. Conclusion Overall, these results indicate GLRX3 as a novel diagnostic marker and therapeutic target for PDAC targeting CSCs.


Sign in / Sign up

Export Citation Format

Share Document