Abstract B28: Targeting the tumor-associated carbohydrate antigen STn with humanized anti-Sialyl-Tn monoclonal antibody-drug conjugates inhibits ovarian cancer tumor growth in vitro and in vivo

Author(s):  
Daniel T. Dransfield ◽  
Jillian M. Prendergast ◽  
David A. Eavarone ◽  
Rawan Nazer ◽  
Linah Al-Alem ◽  
...  
2018 ◽  
Author(s):  
Daniel Dransfield ◽  
Jillian M. Prendergast ◽  
David A. Eavarone ◽  
Rawan Nazer ◽  
Linah Al-Alem ◽  
...  

Cells ◽  
2021 ◽  
Vol 10 (9) ◽  
pp. 2218 ◽  
Author(s):  
Samantha M. Perez ◽  
Julien Dimastromatteo ◽  
Charles N. Landen ◽  
Kimberly A. Kelly

Cancer-specific plectin (CSP) is a pro-tumorigenic protein selectively expressed on the cell surface of major cancers, including ovarian cancer (OC). Despite its assessable localization, abundance, and functional significance, the therapeutic efficacy of targeting CSP remains unexplored. Here, we generated and investigated the anticancer effects of a novel CSP-targeting monoclonal antibody, 1H11, in OC models. Its therapeutic efficacy as a monotherapy and in combination with chemotherapy was evaluated in vitro using two OC cell lines and in vivo by a subcutaneous ovarian cancer model. 1H11 demonstrated rapid internalization and high affinity and specificity for both human and murine CSP. Moreover, 1H11 induced significant and selective cytotoxicity (EC50 = 260 nM), G0/G1 arrest, and decreased OC cell migration. Mechanistically, these results are associated with increased ROS levels and reduced activation of the JAK2-STAT3 pathway. In vivo, 1H11 decreased Ki67 expression, induced 65% tumor growth inhibition, and resulted in 30% tumor necrosis. Moreover, 1H11 increased chemosensitivity to cisplatin resulting in 60% greater tumor growth inhibition compared to cisplatin alone. Taken together, CSP-targeting with 1H11 exhibits potent anticancer activity against ovarian cancer and is deserving of future clinical development.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1653-1653
Author(s):  
Soames F. Boyle ◽  
Jason Kato ◽  
Robert O'Donnell ◽  
Joseph Tuscano

Abstract Abstract 1653 Many non-Hodgkin lymphomas (NHL) and acute lymphoblastic leukemia (ALL) initially respond to therapy, but relapse; at that point further treatment is often limited by chemotherapy-related toxicity. CD22 is a valid target for immunotherapy of NHL and ALL. We have shown that the anti-CD22 ligand blocking monoclonal antibody (mAb), HB22.7, has significant activity. Antibody drug conjugates (ADC) are emerging as the next generation of targeted therapies for lymphoid neoplasms as they may be designed to be effective but with manageable toxicity. We sought to determine if HB22.7 could be used as a platform for CD22-targeted ADCs for the treatment of lymphoid neoplasms. We describe here the effect H22.7-saporin (SAP), a novel ADC composed of HB22.7 and a ribosome inactivating protein on ALL and NHL models. The in vitro cytotoxicity of HB22.7-SAP was assessed using two pre-B cell ALL lines REH and JM1, and NHL cell lines Ramos, Raji, Granta 519, SU-DHL-4, and DOHH-2. HB22.7-SAP had IC50 values of 0.7 and 1.2 ng/mL in ALL and 1.0–8.4 ng/mL in the NHL cell lines. Free HB22.7 plus SAP did not have any cytotoxic effect using the same doses. HB22.7-SAP was not cytotoxic to CD22 negative Jurkat cells. To evaluate HB22.7-SAP's in vivo efficacy against a model of human ALL, NOD/SCID mice were injected with REH leukemia cells. Twenty-four hours later the mice were treated with PBS, free HB22.7 plus SAP, or HB22.7-SAP. Treatment was continued twice weekly for 4 weeks. Mice were euthanized at the onset of hind limb paralysis. The median survival was 52 days as compared to 20 days for the untreated mice (p < 0.0005). Mice treated with HB22.7-SAP had normal blood counts over the study period. In contrast, mice from the PBS or free HB22.7 plus free SAP groups developed leukopenia and thrombocytopenia by day 20, coincident with development of ALL; bone marrow from the two mice that developed hind limb paralysis confirmed ALL. To assess efficacy in a human model of NHL, Raji cells were injected into the flank of previously radiated, female, athymic nude mice. Tumors grew until they reached 100–200 mm3 (day 0). Mice were then exposed to HB22.7-SAP, PBS, or free HB22.7 plus free SAP. Each treatment was administered intraperitoneally, weekly for 3 weeks. To assess significance of starting tumor volume on HB22.7-SAP-mediated tumor growth inhibition, a second model initiated therapy 24 hours after tumor implantation and used the same dose and drug schedule. During the first 14 days, HB22.7-SAP (1 mg/kg) significantly inhibited tumor growth (p < 0.01) on days 5 and 8 as compared to the PBS and free HB22.7 plus free SAP control groups, however, by day 19, the average tumors volume of mice treated with HB22.7-SAP were no different than the controls. In contrast, when HB22.7-SAP was administered 24 hours after subcutaneous injection of Raji cells no mice in the treatment group developed measureable tumors in the 50-day study period. There was no difference in hematologic toxicity between the groups however mice treated with HB22.7-SAP had more therapy-related weight loss. This study demonstrates the promising in vivo and in vitro activity of HB22.7 when used as a platform for CD22-targeted ADCs. The HB22.7-SAP ADC is both specific and cytotoxic to CD22 expressing malignant lymphocytes and that translates into significant efficacy against NHL and ALL. When compared to other anti-CD22 mAb we hypothesize that HB22.7-based ADCs will prove to be potent cytotoxic drugs in-part due to their unique ability to block ligand binding thus conferring an independent lymphocidal property. Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2019 ◽  
Vol 11 (8) ◽  
pp. 1138 ◽  
Author(s):  
Hayashi ◽  
Madokoro ◽  
Yamada ◽  
Nishida ◽  
Morimoto ◽  
...  

Here, we report a novel antibody drug conjugate (ADC) with the humanized anti-CD26 monoclonal antibody YS110 and triptolide (TR-1). YS110 has an inhibitory activity against the CD26-positive tumor growth via the immunological and direct pathway, such as intra-nuclear transportation of CD26 and YS110, and suppressed transcription of RNA polymerase II (Pol II) subunit POLR2A. The ADC conjugated with YS110 and an antitumor compound triptolide (TR-1), which is an inhibitor for TFIIH, one of the general transcription factors for Pol II was developed. YS110 and triptolide were crosslinked by the heterobifunctional linker succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC) and designated Y-TR1. Antitumor efficacy of Y-TR1 against malignant mesothelioma and leukemia cell lines were assessed by the in vitro cell viability assay and in vivo assay using xenografted mouse models. Y-TR1 showed significant cytotoxicity against CD26-positive cell lines but not CD26-negative counterparts in a dose-dependent manner via suppression of mRNA synthesis by impairment of the Pol II activity. The tumors in xenografted mice administered Y-TR1 was smaller than that of the unconjugated YS110 treated mice without severe toxicity. In conclusion, the novel compound Y-TR1 showed antitumor properties against CD26-positive cancer cell lines both in vitro and in vivo without toxicity. The Y-TR1 is a unique antitumor ADC and functions against Pol II.


2020 ◽  
Author(s):  
Juanjuan Shi ◽  
Xijian Xu ◽  
Dan Zhang ◽  
Jiuyan Zhang ◽  
Hui Yang ◽  
...  

Abstract Background: Long non-coding RNA PTPRG antisense RNA 1 (PTPRG-AS1) deregulation has been reported in various human malignancies and identified as an important modulator of cancer development. Few reports have focused on the detailed role of PTPRG-AS1 in epithelial ovarian cancer (EOC) and its underlying mechanism. This study aimed to determine the physiological function of PTPRG-AS1 in EOC. A series of experiments were also performed to identify the mechanisms through which PTPRG-AS1 exerts its function in EOC.Methods: Reverse transcription-quantitative polymerase chain reaction was used to determine PTPRG-AS1 expression in EOC tissues and cell lines. PTPRG-AS1 was silenced in EOC cells and studied with respect to cell proliferation, apoptosis, migration, and invasion in vitro and tumor growth in vivo. The putative miRNAs that target PTPRG-AS1 were predicted using bioinformatics analysis and further confirmed in luciferase reporter and RNA immunoprecipitation assays.Results: Our data verified the upregulation of PTPRG-AS1 in EOC tissues and cell lines. High PTPRG-AS1 expression was associated with shorter overall survival in patients with EOC. Functionally, EOC cell proliferation, migration, invasion in vitro, and tumor growth in vivo were suppressed by PTPRG-AS1 silencing. In contrast, cell apoptosis was promoted by loss of PTPRG-AS1. Regarding the mechanism, PTPRG-AS1 could serve as a competing endogenous RNA in EOC cells by decoying microRNA-545-3p (miR-545-3p), thereby elevating histone deacetylase 4 (HDAC4) expression. Furthermore, rescue experiments revealed that PTPRG-AS1 knockdown-mediated effects on EOC cells were, in part, counteracted by the inhibition of miR-545-3p or restoration of HDAC4.Conclusions: PTPRG-AS1 functioned as an oncogenic lncRNA that aggravated the malignancy of EOC through the miR-545-3p/HDAC4 ceRNA network. Thus, targeting the PTPRG-AS1/miR-545-3p/HDAC4 pathway may be a novel strategy for EOC anticancer therapy.


Cancers ◽  
2018 ◽  
Vol 10 (8) ◽  
pp. 260 ◽  
Author(s):  
Qing Zhang ◽  
Xiaonan Hou ◽  
Bradley Evans ◽  
Jamison VanBlaricom ◽  
Saravut Weroha ◽  
...  

Transforming growth factor beta (TGF-β) signaling has pleiotropic functions regulating cancer initiation, development, and metastasis, and also plays important roles in the interaction between stromal and cancer cells, making the pathway a potential therapeutic target. LY2157299 monohydrate (LY), an inhibitor of TGF-β receptor I (TGFBRI), was examined for its ability to inhibit ovarian cancer (OC) growth both in high-grade serous ovarian cancer (HGSOC) cell lines and xenograft models. Immunohistochemistry, qRT-PCR, and Western blot were performed to study the effect of LY treatment on expression of cancer- and fibroblast-derived genes. Results showed that exposure to TGF-β1 induced phosphorylation of SMAD2 and SMAD3 in all tested OC cell lines, but this induction was suppressed by pretreatment with LY. LY alone inhibited the proliferation, migration, and invasion of HGSOC cells in vitro. TGF-β1-induced fibroblast activation was blocked by LY. LY also delayed tumor growth and suppressed ascites formation in vivo. In addition, independent of tumor inhibition, LY reduces ascites formation in vivo. Using OVCAR8 xenograft specimens we confirmed the inhibitory effect of LY on TGF-β signaling and tumor stromal expression of collagen type XI chain 1 (COL11A1) and versican (VCAN). These observations suggest a role for anti-TGF-β signaling-directed therapy in ovarian cancer.


2020 ◽  
Vol 2020 ◽  
pp. 1-17
Author(s):  
Xiangyu Wang ◽  
Fengmian Wang ◽  
Zhi-Gang Zhang ◽  
Xiao-Mei Yang ◽  
Rong Zhang ◽  
...  

Serine/threonine protein kinase-3 (STK3) is a critical molecule of the Hippo pathway but little is known about its biological functions in the ovarian cancer development. We demonstrated the roles of STK3 in ovarian cancer. Existing databases were used to study the expression profile of STK3. STK3 was significantly downregulated in OC patients, and the low STK3 expression was correlated with a poor prognosis. In vitro cell proliferation, apoptosis, and migration assays, and in vivo subcutaneous xenograft tumor models were used to determine the roles of STK3. The overexpression of STK3 significantly inhibited cell proliferation, apoptosis, and migration of ovarian cancer cells in vitro and tumor growth in vivo. Bisulfite sequencing PCR analysis was performed to validate the methylation of STK3 in ovarian cancer. RNA sequencing and gene set enrichment analysis (GSEA) were used to compare the transcriptome changes in the STK3 overexpression ovarian cancer and control cells. The signaling pathway was analyzed by western blotting. STK3 promoted the migration of CD8+ T-cells by activating nuclear transcription factor κB (NF-κB) signaling. STK3 is a potential predictor of OC. It plays an important role in suppressing tumor growth of ovarian cancer and in chemotaxis of CD8+ T-cells.


2015 ◽  
Vol 12 (6) ◽  
pp. 1872-1879 ◽  
Author(s):  
Penny Bryant ◽  
Martin Pabst ◽  
George Badescu ◽  
Matthew Bird ◽  
William McDowell ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document