scholarly journals ABL001, a Bispecific Antibody Targeting VEGF and DLL4, with Chemotherapy, Synergistically Inhibits Tumor Progression in Xenograft Models

2020 ◽  
Vol 22 (1) ◽  
pp. 241
Author(s):  
Dong-Hoon Yeom ◽  
Yo-Seob Lee ◽  
Ilhwan Ryu ◽  
Sunju Lee ◽  
Byungje Sung ◽  
...  

Delta-like-ligand 4 (DLL4) is a promising target to augment the effects of VEGF inhibitors. A simultaneous blockade of VEGF/VEGFR and DLL4/Notch signaling pathways leads to more potent anti-cancer effects by synergistic anti-angiogenic mechanisms in xenograft models. A bispecific antibody targeting VEGF and DLL4 (ABL001/NOV1501/TR009) demonstrates more potent in vitro and in vivo biological activity compared to VEGF or DLL4 targeting monoclonal antibodies alone and is currently being evaluated in a phase 1 clinical study of heavy chemotherapy or targeted therapy pre-treated cancer patients (ClinicalTrials.gov Identifier: NCT03292783). However, the effects of a combination of ABL001 and chemotherapy on tumor vessels and tumors are not known. Hence, the effects of ABL001, with or without paclitaxel and irinotecan were evaluated in human gastric or colon cancer xenograft models. The combination treatment synergistically inhibited tumor progression compared to each monotherapy. More tumor vessel regression and apoptotic tumor cell induction were observed in tumors treated with the combination therapy, which might be due to tumor vessel normalization. Overall, these findings suggest that the combination therapy of ABL001 with paclitaxel or irinotecan would be a better clinical strategy for the treatment of cancer patients.

Blood ◽  
1995 ◽  
Vol 86 (3) ◽  
pp. 1124-1130 ◽  
Author(s):  
J Michon ◽  
S Moutel ◽  
J Barbet ◽  
JL Romet-Lemonne ◽  
YM Deo ◽  
...  

Abstract Neutrophils isolated from cancer patients treated with granulocyte colony-stimulating factor (G-CSF) express high levels of Fc gamma RI. They exhibited an efficient killing of GD2+ neuroblastoma cells in the presence of an antidisialoganglioside (GD2) mouse monoclonal antibody (MoAb; 7A4, IgG3 kappa). However, this cytotoxicity was totally blocked by human monomeric IgG. In contrast, a bispecific antibody (7A4 bis 22/MDX-260), prepared by chemically linking an F(ab') fragment of 7A4 with an F(ab') fragment of an anti-Fc gamma RI MoAb, 22, which binds outside the Fc binding domain, triggered antibody-dependent cell cytotoxicity, even when neutrophils were preincubated with human monomeric IgG. F(ab')2 22 MoAb abrogated the MDX-260 killing without affecting that of 7A4. The 3G8 MoAb, directed against the Fc gamma RIII binding site, did not inhibit the cytotoxicity induced by either antibody. Thus, these results indicate that G-CSF-activated neutrophils exert their cytotoxic effect against neuroblastoma cells through Fc gamma RI and not Fc gamma RIII, and that the saturation of the high affinity Fc gamma RI by monomeric IgG can be overcome by the use of bispecific antibodies binding epitopes outside the IgG Fc gamma RI binding site. A combined administration of such bispecific antibodies and G-CSF may be, therefore, an efficient therapeutic approach to trigger tumor lysis by cytotoxic neutrophils in vivo.


2017 ◽  
Vol 35 (7_suppl) ◽  
pp. 126-126
Author(s):  
Jaspreet Singh Grewal ◽  
Numan Al-Rayyan ◽  
Jamaal Ritchie ◽  
Paxton Schowe ◽  
Cam Falkner ◽  
...  

126 Background: Myeloid derived suppressor cells (MDSCs) inhibit the expansion of tumor antigen-specific effector CD8+ T cells via different mechanisms including increased expression of arginase, transforming growth factor – β (TGF – β) and indoleamine 2,3-dioxygenase (IDO). Recently, MDSCs were found to over-express hypoxia inducible factor 1 alpha (HIF-1α) which is required for their differentiation. An essential transcriptional target of HIF-1α is 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) which synthesizes fructose 2,6-bisphosphate, an allosteric stimulator of glycolysis and of proliferation via stimulation of cyclin dependent kinase-1 (CDK1). We hypothesized that MDSCs might over-express PFKFB3 which in turn might be required for their function as T cell suppressors. Methods: We used monocytic MDSCs (M-MDSCs) induced by co-culture with A375 melanoma cells, M-MDSCs from metastatic melanoma patients, murine bone marrow MDSCs and splenic M-MDSCs from B16 F10 tumor bearing mice for our studies. T cell suppression assays were performed to analyze M-MDSC suppression and reversal following PFKFB3 blockade. Results: We found that M-MDSCs have increased PFKFB3 expression. We also found that PFK-158 administration in B16 (wild-type) melanoma-bearing mice results in a marked reduction in MDSCs and a simultaneous increase in CD8+ T cell infiltration in the tumors. We analyzed three advanced cancer patients for circulating MDSCs before and after PFK-158 administration as part of a multi-center phase 1 clinical trial. And, we found that the MDSCs were markedly reduced in each patient. In addition, we have generated data for MDSC suppressive activity following in vitro treatment with PFK-158 showing reversal of suppressive activity. Conclusions: Taken together, these data indicate that selective inhibition of PFKFB3 may be a novel approach to target MDSCs and combinations of PFKFB3 inhibitors with immunotherapies may be a rational strategy to promote durable immune-mediated remissions in cancer patients.


2019 ◽  
Vol 20 (3) ◽  
pp. 686 ◽  
Author(s):  
Ewa Kurzejamska ◽  
Mariusz Sacharczuk ◽  
Natalia Landázuri ◽  
Oksana Kovtonyuk ◽  
Marzena Łazarczyk ◽  
...  

Colorectal cancer is the source of one of the most common cancer-related deaths worldwide, where the main cause of patient mortality remains metastasis. The aim of this study was to determine the role of CCL7 (chemokine (C-C motif) ligand 7) in tumor progression and finding whether it could predict survival of colorectal cancer patients. Initially, our study focused on the crosstalk between mesenchymal stem cells (MSCs) and CT26 colon carcinoma cells and resulted in identifying CCL7 as a chemokine upregulated in CT26 colon cancer cells cocultured with MSCs, compared with CT26 in monoculture in vitro. Moreover, we showed that MSCs enhance CT26 tumor cell proliferation and migration. We analyzed the effect of CCL7 overexpression on tumor progression in a murine CT26 model, where cells overexpressing CCL7 accelerated the early phase of tumor growth and caused higher lung metastasis rates compared with control mice. Microarray analysis revealed that tumors overexpressing CCL7 had lower expression of immunoglobulins produced by B lymphocytes. Additionally, using Jh mutant mice, we confirmed that in the CT26 model, CCL7 has an immunoglobulin-, and thereby, B-cell-dependent effect on metastasis formation. Finally, higher expression of CCL7 receptor CCR2 (C-C chemokine receptor type 2) was associated with shorter overall survival of colorectal cancer patients. Altogether, we showed that CCL7 is essentially involved in the progression of colorectal cancer in a CT26 mouse model and that the expression of its receptor CCR2 could be related to a different outcome pattern of patients with colorectal carcinoma.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A757-A757
Author(s):  
Michelle Morrow ◽  
Mustapha Faroudi ◽  
Krishnendu Chakraborty ◽  
Wenjia Liao ◽  
Julia Winnewisser ◽  
...  

BackgroundUpregulation of immune checkpoints, such as LAG-3, plays an important role in promoting resistance to anti-PD-(L)1 therapy. FS118, currently being evaluated in a Phase 1 clinical trial in patients with advanced malignancies, is a tetravalent bispecific antibody targeting LAG-3 and PD-L1 that can overcome immune suppressive signals with greater preclinical activity than a combination of monoclonal antibodies.1 Here, we demonstrate a novel mechanism of action for FS118 in shedding of LAG-3 from the surface of T cells that is not observed with the combination of PD-L1 and LAG-3 antibodies.MethodsHuman ex vivo assays were performed by co-culturing activated CD4+ T cells with iDCs in the presence of Staphylococcal enterotoxin B and FS118, or control reagents. Soluble LAG-3 was measured by ELISA from day 4 to 13. A mouse tumor model used MC38 cells implanted subcutaneously into C57Bl/6 mice. Expression of surface markers was measured on tumor-infiltrating lymphocytes (TILs) from disaggregated tumors and soluble LAG-3 was measured in serum following dosing of mice intraperitoneally with FS118 surrogate or control reagents. Soluble LAG-3 in the serum of patients treated with FS118 was measured by ELISA (Phase 1 trial NCT03440437).ResultsIn an ex vivo T cell assay, FS118 resulted in an increase in the concentration of soluble LAG-3 in the cell culture medium, an effect that was greater than with the combination of the individual bispecific components. Addition of inhibitors of either ADAM10 or ADAM17 to the FS118-treated cells resulted in a decrease in the levels of soluble LAG-3 in the cell culture medium. In MC38 tumor-bearing mice, a mouse surrogate of FS118 decreased the levels of surface LAG-3 expressed by TILs, in contrast to the combination of the bispecific components where an increase in surface LAG-3 was observed. This corresponded with an increase in soluble LAG-3 in the serum following treatment with a mouse surrogate of FS118. Finally, in patients receiving treatment with FS118, a dose dependent increase in soluble LAG-3 was detected in the blood.ConclusionsFS118 mediates LAG-3 shedding from the surface of immune cells via a mechanism that is dependent upon simultaneous binding to both PD-L1 and LAG-3. This shedding was mediated by ADAM10 and ADAM17 metalloproteinases. Removing LAG-3 from the surface of TILs via shedding may be an important mechanism by which FS118 overcomes compensatory upregulation of LAG-3 induced by PD-L1 blockade. Soluble LAG-3 may be an important biomarker for monitoring the pharmacodynamic activity of FS118 in patients.Ethics ApprovalAll animal experiments were conducted under a UK Home Office Project Licence and approved by an Animal Welfare and Ethical Review Board (AWERB) in accordance with the UK Animal (Scientific Procedures) Act 1986 and with EU Directive EU 86/609ReferenceKraman M, Faroudi M, Allen N, Kmiecik K, Gliddon D, Seal C, Koers A, Wydro M, Winnewisser J, Young L, Tuna M, Doody J, Morrow M, Brewis N. FS118, a Bispecific Antibody Targeting LAG-3 and PD-L1, Enhances T-Cell Activation Resulting in Potent Antitumor Activity. Clin Cancer Res 2020;26:3333–3344


Biomedicines ◽  
2021 ◽  
Vol 9 (8) ◽  
pp. 1059
Author(s):  
Jie Chen ◽  
Zhidi Pan ◽  
Lei Han ◽  
Yuexian Zhou ◽  
Huifang Zong ◽  
...  

Lewis Y antigen, a glycan highly expressed on most epithelial cancers, was targeted for cancer treatment but lacked satisfactory results in some intractable and refractory cancers. Thus, it is highly desirable to develop an effective therapy against these cancers, hopefully based on this target. In this work, we constructed a novel T cell-engaging bispecific antibody targeting Lewis Y and CD3 (m3s193 BsAb) with the IgG-[L]-scfv format. In vitro activity of m3s193 BsAb was evaluated by affinity assay to target cells, cytotoxicity assay, cytokines releasing assay, and T cells proliferation and recruiting assays. Anti-tumor activity against gastric cancer was evaluated in vivo by subcutaneous huPBMCs/tumor cells co-grafting model and huPBMCs intravenous injecting model. In vitro, m3s193 BsAb appeared to have a high binding affinity to Lewis Y positive cells and Jurkat cells. The BsAb showed stronger activity than its parent mAb in T cell recruiting, activation, proliferation, cytokine release, and cytotoxicity. In vivo, m3s193 BsAb not only demonstrated higher therapeutic efficacy in the huPBMCs/tumor co-grafting gastric carcinoma model than the parent mAb but also eliminated tumors in the model of intravenous injection with huPBMCs. Strong anti-tumor activity of m3s193 BsAb revealed that Lewis Y could be targeted in T cell-engaging BsAb for gastric cancer therapy.


Sign in / Sign up

Export Citation Format

Share Document