First Selective KIT D816V Inhibitor for Patients with Systemic Mastocytosis

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3217-3217 ◽  
Author(s):  
Erica K. Evans ◽  
Brian L. Hodous ◽  
Alexandra Gardino ◽  
Julia Zhu ◽  
Adam Shutes ◽  
...  

Abstract Systemic mastocytosis is a disease characterized by the abnormal proliferation and accumulation of mast cells. In aggressive cases, these mast cells accumulate in organs such as bone marrow, liver and spleen and result in compromised organ function with average patient survival only 3 to 5 years after diagnosis. The mast cells of nearly all systemic mastocytosis patients harbor a heterozygous D816V mutation in the activation loop of KIT conferring constitutive, ligand-independent activation of this receptor tyrosine kinase, suggesting this mutation is a driver of disease. While KIT D816V can be targeted by small molecules such as dasatinib and midostaurin, these agents have activity against many human kinases resulting in dose limiting toxicities in the clinic that prevent complete suppression of KIT D816V activity in vivo. In vitro, their potent activity against multiple kinases leads to uncertainties regarding their mechanism of action. Thus far, selective inhibition of the KIT D816V mutation has not been achieved. However starting with a novel chemical library optimized for kinase selectivity, we have identified BLU-285, a small molecule inhibitor targeting KIT exon 17 mutants including the activated KIT D816V kinase. BLU-285 potently disrupts KIT D816V oncogenic signaling as measured by inhibition of both KIT D816V autophosphorylation and phosphorylation of the downstream substrates Akt and Stat3 in the human mast cell leukemia cell line HMC1.2. In vitro, BLU-285 inhibits proliferation and induces apoptosis in the mouse mastocytoma cell line P815. In vivo, BLU-285 is a well-tolerated, orally bioavailable agent that achieves dose dependent tumor growth inhibition in a P815 mouse xenograft model with tumor regression observed at 30 mg/kg once daily dosing. Tumor growth inhibition correlates with inhibition of KIT autophosphorylation; greater than 80% target suppression throughout the 24-hour dosing period is required for effective tumor growth inhibition. Prolonged target suppression is achievable with BLU-285 but not dasatinib, even when dosed at the MTD in mouse. Furthermore, to more closely mimic the nature of systemic mastocytosis, we have developed a disseminated model of disease whereby the in vivo growth of P815-luciferase expressing cells inoculated intravenously can be measured by whole body bioluminescence. Treatment of mice with systemic disease leads to dose dependent inhibition of disease, with a 3-fold increase in survival time when dosed 30 mg/kg QD. In addition, as anticipated by its selectivity profile, BLU-285 is very well tolerated in vivo with no impact on body weight at efficacious doses. Our data demonstrate that selective inhibition of KIT D816V with BLU-285 achieves complete and prolonged inactivation of the disease-driving kinase and suggests that BLU-285 may provide a compelling new therapy for patients with systemic mastocytosis. Disclosures Evans: Blueprint Medicines: Employment, Equity Ownership. Hodous:Blueprint Medicines: Employment, Equity Ownership. Gardino:Blueprint Medicines: Employment, Equity Ownership. Zhu:Blueprint Medicines: Employment, Equity Ownership. Shutes:Blueprint Medicines: Employment, Equity Ownership. Davis:Blueprint Medicines: Employment, Equity Ownership. Kim:Blueprint Medicines: Employment, Equity Ownership. Wilson:Blueprint Medicines: Employment, Equity Ownership. Wilson:Blueprint Medicines: Employment, Equity Ownership. Zhang:Blueprint Medicines: Employment, Equity Ownership. Kohl:Blueprint Medicines: Employment, Equity Ownership. Guzi:Blueprint Medicines: Employment, Equity Ownership. Lengauer:Blueprint Medicines: Employment, Equity Ownership.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1844-1844
Author(s):  
John Richards ◽  
Myriam N Bouchlaka ◽  
Robyn J Puro ◽  
Ben J Capoccia ◽  
Ronald R Hiebsch ◽  
...  

AO-176 is a highly differentiated, humanized anti-CD47 IgG2 antibody that is unique among agents in this class of checkpoint inhibitors. AO-176 works by blocking the "don't eat me" signal, the standard mechanism of anti-CD47 antibodies, but also by directly killing tumor cells. Importantly, AO-176 binds preferentially to tumor cells, compared to normal cells, and binds even more potently to tumors in their acidic microenvironment (low pH). Hematological neoplasms are the fourth most frequently diagnosed cancers in both men and women and account for approximately 10% of all cancers. Here we describe AO-176, a highly differentiated anti-CD47 antibody that potently targets hematologic cancers in vitro and in vivo. As a single agent, AO-176 not only promotes phagocytosis (15-45%, EC50 = 0.33-4.1 µg/ml) of hematologic tumor cell lines (acute myeloid leukemia, non-Hodgkin's lymphoma, multiple myeloma, and T cell leukemia) but also directly targets and kills tumor cells (18-46% Annexin V positivity, EC50 = 0.63-10 µg/ml) in a non-ADCC manner. In combination with agents targeting CD20 (rituximab) or CD38 (daratumumab), AO-176 mediates enhanced phagocytosis of lymphoma and multiple myeloma cell lines, respectively. In vivo, AO-176 mediates potent monotherapy tumor growth inhibition of hematologic tumors including Raji B cell lymphoma and RPMI-8226 multiple myeloma xenograft models in a dose-dependent manner. Concomitant with tumor growth inhibition, immune cell infiltrates were observed with elevated numbers of macrophage and dendritic cells, along with increased pro-inflammatory cytokine levels in AO-176 treated animals. When combined with bortezomib, AO-176 was able to elicit complete tumor regression (100% CR in 10/10 animals treated with either 10 or 25 mg/kg AO-176 + 1 mg/kg bortezomib) with no detectable tumor out to 100 days at study termination. Overall survival was also greatly improved following combination therapy compared to animals treated with bortezomib or AO-176 alone. These data show that AO-176 exhibits promising monotherapy and combination therapy activity, both in vitro and in vivo, against hematologic cancers. These findings also add to the previously reported anti-tumor efficacy exhibited by AO-176 in solid tumor xenografts representing ovarian, gastric and breast cancer. With AO-176's highly differentiated MOA and binding characteristics, it may have the potential to improve upon the safety and efficacy profiles relative to other agents in this class. AO-176 is currently being evaluated in a Phase 1 clinical trial (NCT03834948) for the treatment of patients with select solid tumors. Disclosures Richards: Arch Oncology Inc.: Employment, Equity Ownership, Other: Salary. Bouchlaka:Arch Oncology Inc.: Consultancy, Equity Ownership. Puro:Arch Oncology Inc.: Employment, Equity Ownership. Capoccia:Arch Oncology Inc.: Employment, Equity Ownership. Hiebsch:Arch Oncology Inc.: Employment, Equity Ownership. Donio:Arch Oncology Inc.: Employment, Equity Ownership. Wilson:Arch Oncology Inc.: Employment, Equity Ownership. Chakraborty:Arch Oncology Inc.: Employment, Equity Ownership. Sung:Arch Oncology Inc.: Employment, Equity Ownership. Pereira:Arch Oncology Inc.: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5123-5123 ◽  
Author(s):  
Mark Joseph Axelrod ◽  
Peter Fowles ◽  
Jeff Silverman ◽  
Astrid Clarke ◽  
Jennifer Tang ◽  
...  

Abstract Background Entospletinib (GS-9973) selectively inhibits spleen tyrosine kinase (SYK), a critical signaling component of the BCR pathway that is expressed primarily in cells of hematopoietic lineage including normal and malignant B-lymphocytes. Entospletinib is currently in phase II clinical trials, where it has demonstrated both a high degree of safety as well as efficacy against chronic lymphocytic leukemia (Sharman, J., et al. Blood, 2015) and other B cell malignancies. Despite these successes, new therapeutic options, including combinations with standard of care agents, are needed in order to achieve the goal of curing disease through finite treatment. We show here that the combination of entospletinib and vincristine causes synergistic apoptosis in vitro in a broad panel of cell lines derived from hematological cancers including diffuse large B cell lymphoma (DLBCL), acute lymphocytic leukemia, follicular lymphom), multiple myeloma, and acute myelogenous leukemia. We also evaluated and compared the in vivo efficacy of entospletinib and vincristine as singe agents and in combination in a DLBCL tumor xenograft model using the SU-DHL-10 cell line. Methods In vitro growth inhibition of a panel of malignant hematological cell lines was assessed using CellTiter-Glo™ Assay (Promega) after 72h incubation with entospletinib or vincristine alone or in combination. Synergy was evaluated using the Bliss model of independence (Meletiadis, J., et al., Med Mycol, 2005). In vivo, SU-DHL-10 cells (5 x 106 cells) were implanted subcutaneously in the axilla in male SCID beige mice. All mice were sorted into study groups on Day 16 such that each group's mean tumor volume fell within 10% of the overall mean (197mm3). Dosing was initiated on Day 16 and animals were dosed for 17 days. Plasma concentrations of entospletinib and vincristine were assessed on Day 19, and the entospletinib 75 mg/kg dose was lowered on Day 22 to 50 mg/kg to approximate the human achievable SYK target coverage of EC80. Efficacy and tolerability were evaluated by tumor measurements and body weight monitored three times weekly. Tumor burden data were analyzed by the application of a two-way analysis of variance (ANOVA), with post-hoc analysis. Results In vitro combinations of entospletinib with low concentrations of vincristine resulted in marked inhibition of cell proliferation and induction of apoptosis in a broad panel of 19 tumor cell lines representing major B cell malignancies including DLBCL. The combination of entospletinib with vincristine had a profound inhibitory effect on proliferation in all subtypes of DLBCL. Entospletinib was evaluated at a concentration equivalent to the Cminof the clinical dose and vincristine was used at concentrations (≤ 10 nM) that had little to no significant single agent effect in these cell lines. In vivo in a SU-DHL-10 xenograft model, entospletinib dosed alone at 25 or 75/50 mg/kg significantly inhibited tumor growth, causing 39% and 20% tumor growth inhibition (TGI), respectively, compared to the vehicle-treated control group. Vincristine administered at either 0.15 and 0.5 mg/kg Q7D x 3 also resulted in significant TGI (42% and 85% TGI, respectively). The addition of entospletinib (75/50 mg/kg) to 0.5 mg/kg or 0.15 mg/kg vincristine resulted in a significant increase in TGI from 85% to 96% (p= 0.001) and 42% to 71% (p< 0.0001), respectively. The addition of entospletinib (25 mg/kg) to vincristine did not significantly increase the tumor growth inhibition. While the groups receiving either entospletinib or vincristine as single agents had no complete or partial tumor regression, 50% of the mice receiving the combination of 75/50 mg/kg entospletinib with 0.5 mg/kg vincristine had partial responses, 8% had complete regression and 8% were tumor free at the end of study (Figure 1). Conclusion Entospletinib and vincristine demonstrated efficacy and tolerability both alone and in combination in the SU-DHL-10 DLBCL cell line xenograft model in SCID beige mice. Vincristine combinations with entospletinib showed significantly greater efficacy than vincristine alone. These data support the further clinical development of entospletinib in combination with vincristine for the treatment of DLBCL. a ENTO: PO: Q12H x 2 (Day 16-32) b VCR: IV: Q7D x 3 (Days 18, 25, 32) Figure 1. Tumor Regressions in an Entospletinib/ Vincristine Treated Murine DLBCL Xenograft Figure 1. Tumor Regressions in an Entospletinib/ Vincristine Treated Murine DLBCL Xenograft Disclosures Axelrod: Gilead Sciences: Employment, Equity Ownership. Fowles:Gilead Sciences: Employment, Equity Ownership. Silverman:Gilead Sciences: Employment, Equity Ownership. Clarke:Gilead Sciences: Employment, Equity Ownership. Tang:Gilead Sciences: Employment, Equity Ownership. Rousseau:Gilead Sciences: Employment, Equity Ownership. Webb:Gilead Sciences: Employment, Equity Ownership. Di Paolo:Gilead Sciences: Employment, Equity Ownership.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1592-1592 ◽  
Author(s):  
Jessica J Huck ◽  
Mengkun Zhang ◽  
Marc L Hyer ◽  
Mark G Manfredi

Abstract Aurora A kinase is a serine/threonine protein kinase that is essential for normal transit of cells through mitosis. In many tumor types the Aurora A gene is amplified and/or the protein is over-expressed. The Aurora A small-molecule inhibitor MLN8237 demonstrated robust tumor growth inhibition in xenograft models of solid tumors grown subcutaneously (S.C.) in immunocompromised mice. Here we explored the antitumor activity of MLN8237 in models of diffuse large B-cell lymphoma (DLBCL) both in vitro and in vivo. In vivo three established DLBCL xenograft models (OCI-Ly7, OCI-Ly19, and WSU-DLCL2; all cells expressing luciferase) and a primary DLBCL tumor model PHTX-22-06 were tested using MLN8237 at different doses. Rituximab, an anti-CD20 monoclonal antibody that is active against CD20+ malignant B cells and is a standard of care agent was used for comparison. Using these model systems, tumor cells were injected either I.V. (to evaluate disseminated disease), or S.C. in severe combined immunodeficient mice (SCID). Animals were dosed orally for 21 days with MLN8237 (QD or BID) at various doses, or Rituximab dosed at 10mg/kg IV (once/week) and tumor growth inhibition was monitored using either bioluminescent imaging for the disseminated models or vernier calipers for the S.C. models. Tumor growth inhibition by MLN8237 was dose dependent with 20 mg/kg bid being the most efficacious dose (TGI&gt;100% in both disseminated OCI-Ly19 and WSU models). All animals in the OCI-Ly19 disseminated model 20 mg/kg BID treatment group demonstrated regressions and remained disease free until the end of the study, day 65. In this study the Rituximab treated animals were euthanized on day 31 due to a high level of tumor burden. In the primary tumor model, PHTX-22-06, MLN8237 dosed at 20 mg/kg BID was also the most efficacious with a TGI of 95%. Moreover, tumor growth inhibition was durable as determined by prolonged tumor growth delay (&gt;50 days). Significant efficacy was achieved in all models tested, whether grown as disseminated or subcutaneous models. A noted increase in durability of response was observed with MLN8237 treatment when compared with previous data from solid tumor models. In vitro, MLN8237 treatment increased levels of apoptosis in the OCI-Ly19 cells in comparison to the solid tumor cell line HCT-116 (colon). Greater Annexin V positive cells and greater cleaved PARP and Caspase-3 signals were detected in the MLN8237 treated OCI-Ly19 cells when compared to HCT-116 cells. The demonstration of robust and durable anti-tumor activity in preclinical models treated with MLN8237 provides the basis for its clinical evaluation as a treatment option for DLBCL. MLN8237 is currently in multiple Phase I clinical trials.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1729-1729
Author(s):  
Luis Borges ◽  
Mark A Wallet ◽  
Chiamin-Liao Bullaughey ◽  
Michael F Naso ◽  
Buddha Gurung ◽  
...  

Abstract Induced-pluripotent stem cells (iPSCs) can be differentiated into various somatic cells, including different immune cell types. We have engineered iPSC-derived NK cells with multiple features to generate therapeutic candidates designed to eliminate cancer cells while avoiding recognition by the host immune system. The unlimited replication capacity of iPSCs facilitates the engineering of several genetic modifications without the risk of driving cells to exhaustion as in the case of cell products derived from fully differentiated immune cells. Once all edits are completed, our cells are single-cell cloned and each clone is genetically characterized to select clones without off-target insertions or deletions. Following the genetic characterization, selected clones are differentiated and tested in vitro and in vivo to identify the final clinical candidate. The use of a single-cell iPSC clone enables the generation of a master cell bank producing a highly uniform cell product that can be made available off-the-shelf at any clinical site. CNTY-101 is an iPSC-derived CAR-NK clinical candidate for the treatment of B-cell malignancies. It incorporates six gene edits designed to improve persistence and functionality as well as safety. These modifications include edits to reduce graft rejection due to alloreactivity, the expression of a homeostatic cytokine to improve functionality and persistence, the introduction of a chimeric antigen receptor (CAR) targeting CD19 to mediate tumor cell engagement and killing, as well a safety switch to eliminate the cells, if ever necessary. To prevent rejection by the patient's CD8 T cells, the beta-2-microbulin (ß2M) gene was disrupted with simultaneous insertion of a transgene encoding the HLA-E protein tethered with ß2M and a peptide. HLA-E was introduced to prevent NK cell cytotoxicity against the engineered cells, which lack HLA-I. For resistance to CD4 T cell-mediated allogenic immune rejection, the class II major histocompatibility complex transactivator (CIITA) gene was disrupted with simultaneous insertion of a transgene encoding the extra-cellular and transmembrane domains of EGFR, and the NK cell growth factor IL-15. EGFR provides an elimination tag that can be engaged by clinically approved anti-EGFR antibodies, such as cetuximab. Finally, the CAR transgene targeting the CD19 antigen was inserted into the AAVS1 safe harbor locus. Our data indicates that CNTY-101 iNK cells have strong antitumor activity against lymphoma cell lines both in vitro and in vivo. In vitro, CNTY-101 eliminates lymphoma cell lines through multiple rounds of killing without reaching exhaustion. Clones expressing higher levels of IL-15 tend to have better persistence and functionality, with some clones showing robust cytotoxicity for over fifteen rounds of serial killing. In vivo, the clones that demonstrated better in vitro serial killing tend to mediate the best anti-tumor activity in lymphoma xenograft models. Upon 3 weekly doses, the most active candidate clone demonstrated significant tumor growth inhibition after administration of fresh (91 % tumor growth inhibition) or cryopreserved cells (76 % tumor growth inhibition). The efficacy of the EGFR-safety switch was also investigated both in vitro and in vivo. In vitro, addition of cetuximab to co-cultures of IL-2-activated PBMC and cells mediated antibody-dependent cellular cytotoxicity (ADCC) in a concentration-dependent fashion, with an EC50 of 2 ng/ml. In vivo, there was a 96% reduction in the number of iPSC-derived CAR-NK cells in the lungs and a 95% reduction in the number of CAR-NK cells in the blood of mice that received cetuximab versus PBS-treated mice. In summary, CNTY-101 is a novel, multi-engineered, allogeneic CAR-iNK product candidate for the treatment of B-cell malignancies. It includes multiple immune evasion features to prevent recognition by the patient's immune system and expression of IL-15 to facilitate persistence and functionality. We have initiated GMP manufacturing of CNTY-101 and plan to enter clinical trials in 2022. Disclosures Borges: Century Therapeutics: Current Employment, Current equity holder in publicly-traded company. Wallet: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Bullaughey: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Naso: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Gurung: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Keating: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Carton: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Wheeler: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Campion: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Mendonca: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Jessup: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Beqiri: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Chin: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Millar Quinn: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Morse: Century Therapeutics: Current Employment, Current holder of stock options in a privately-held company.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3915-3915 ◽  
Author(s):  
Frank Herting ◽  
Sabine Bader ◽  
Pablo Umana ◽  
Christian Klein

Abstract Abstract 3915 GA101 is type II, glycoengineered CD20 antibody currently in PhII/III clinical trials. GA101 mediates enhanced direct cell death with a concomitant reduction of CDC; and high ADCC induction due to increased affinity for FcgRIIIa. We have shown that GA101 compared to rituximab mediates superior efficacy in NHL xenograft models including the induction of complete tumor remission. In clinical practice the combination of rituximab with chemotherapy e.g. CHOP, CVP, bendamustine, fludarabine or FC results in a substantial clinical benefit. To assess the potential of GA101 for combination with bendamustine or fludarabine, in vivo combination studies in s.c. Z138 (MCL) xenografts in Scid beige mice were devised. GA101 and rituximab at sub-optimal doses of 1 mg/kg (once weekly) were combined with 3 mg/kg bendamustine (days 19, 20, 21, 22); or with 40 mg/kg fludarabine (days 22, 23, 24) and compared to the corresponding monotherapy arms. GA101 in combination with bendamustine mediated statistically superior efficacy in terms of tumor growth inhibition (TGI) compared to the combination of rituximab and bendamustine: TGI values on day 33 were 29% for rituximab, 42% for rituximab + bendamustine, 47% for GA101 and 72% for GA101 + bendamustine. Treatment with bendamustine did not show significant antitumor activity. Statistical evaluation based on sAUC showed a more than additive and significant effect on tumor growth for the combination of GA101 with bendamustine compared to the corresponding monotherapy arms. GA101 in combination with fludarabine demonstrated statistically superior efficacy in terms of TGI and yielded a significant difference compared to the combination of rituximab and fludarabine or GA101 as monotherpy. TGI values on day 36 were 50% for fludarabine, 60% for rituximab, 85% for rituximab + fludarabine, 86% for GA101 and >100% for GA101 + fludarabine. Furthermore, the superiority of the GA101-fludarabine combination was demonstrated by the observation of 3 tumor-free animals at the end of the study. ABT-263 (navitoclax) is a Bcl-2 family inhibitor that is currently in Phase I/II clinical trials for lymphoid malignancies. To provide evidence that GA101 can be combined with ABT-263 and the experimental Bcl-2 family inhibitor ABT-737, in vivo combination studies in s.c. SU-DHL4 (DLBCL) xenografts in Scid beige mice were devised. 10 mg/kg GA101 and rituximab (once weekly) were combined with 50 mg/kg ABT-737 (i.p., days 19, 22, 24, 26, 29, 31, 33). In a second study, 3 mg/kg GA101 or 10 mg/kg rituximab (once weekly) were combined with 100 mg/kg ABT-263 (orally, once daily). GA101 at a sub-optimal dose of 10 mg/kg demonstrated statistically superior efficacy in combination with ABT-737 in terms of tumor growth inhibition compared to GA101 alone or the combination of 10 mg/kg rituximab and ABT-737. Both combination treatments were statistically significant compared to the corresponding monotherapy arms. TGI values based on means on day 36 were 20% for ABT-737, 45% for rituximab, 92% for rituximab + ABT-737, 96% for GA101 and >100% for GA101 + ABT-737. The superiority of the combination of GA101 and ABT-737 was supported by complete tumor regression in all animals whereas none was observed with the combination of rituximab and ABT-737. GA101 at a sub-optimal dose of 3 mg/kg or rituximab at a dose of 10 mg/kg mediated statistically superior efficacy in terms of tumor growth inhibition in combination with ABT-263 compared to the respective monotherapy arms. TGI values based on means on day 43 were 15% for ABT-263, 89% for rituximab, >100% for rituximab + ABT-263, 80% for GA101 (at the sub-optimal dose) and >100% for GA101 (sub-optimal dose) + ABT-263. Taken together i) GA101 as single agent was at least as efficacious as the combination of rituximab with bendamustine, fludarabine or ABT-737; ii) the combination of GA101 with bendamustine or fludarabine was superior to the respective monotherapy arms and resulted in an enhanced, at least additive effect of the combination; iii) the combination of GA101 with Bcl-2 family inhibitors ABT-737 and ABT-263 was superior to the respective monotherapy arms and resulted in an enhanced effect of the combination including the induction of tumor remission. These data strongly support the further clinical investigation of GA101 in combination with Fludarabine, Bendamustine or Bcl-2 family inhibitors. Disclosures: Herting: Roche: Employment. Bader:Roche: Employment. Umana:Roche: Employment, Equity Ownership. Klein:Roche: Employment, Equity Ownership.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3298-3298 ◽  
Author(s):  
Christian Argueta ◽  
Hua Chang ◽  
Trinayan Kashyap ◽  
Sivan Elloul ◽  
Sharon Friedlander ◽  
...  

Abstract Background: XPO1 (Exportin-1/CRM1) is the non-redundant nuclear exporter of over 200 cargos including the major tumor suppressor proteins. Deregulated nuclear export by changes in XPO1 expression is a common characteristic for a broad range of cancers and may aid in the evasion of anti-neoplastic mechanisms. As a result, inhibition of XPO1 has emerged as a promising area of cancer treatment. The Selective Inhibitor of Nuclear Export (SINE) compounds, selinexor, as well as a second generation, KPT-8602, bind to the XPO1 cargo binding pocket and disrupt XPO1-mediated nuclear export, resulting in cancer specific cell death. Although selinexor has been evaluated in >1,500 patients and has manageable tolerability, KPT-8602 may have improved tolerability and efficacy based on decreased brain penetration in animal models allowing more frequent dosing. Currently, the safety, tolerability and efficacy of KPT-8602 is being evaluated in a phase 1 trial of patients with relapsed/refractory multiple myeloma (MM; NCT02649790). Since selinexor synergizes with a broad array of anti-MM agents in patients, KPT-8602 is an excellent candidate for combination therapies. In this study, we investigated the use and mechanism of combining KPT-8602 with the pan-histone deacetylase (HDAC) inhibitor, panobinostat, in MM cell lines and in a xenograft mouse model of MM. Methods : MM.1S cells were treated with single agent KPT-8602, panobinostat or a combination of both. The effects of KPT-8602 and/or panobinostat on cell viability were examined using standard viability assays after 72 hours of treatment. Total RNA or protein levels were examined after 24 hours using quantitative PCR or immunoblots, respectively. Immune compromised mice were injected subcutaneously with MM.1S cells. Tumors were allowed to grow to ~150 mm3before treatment. The mice were treated with vehicle, sub-therapeutic doses of KPT-8602 (5 mg/kg PO QDx5) or panobinostat (5 mg/kg IP QDx5) alone or in combination. Tumor growth and animal weights was monitored to determine tumor growth inhibition (TGI), tumor regression, and tolerability to treatment. The tumors were then harvested for immunohistochemical (IHC) analysis. Results : The combination of KPT-8602 and panobinostat was highly effective against MM.1S cell viability. A synergistic anti-cancer effect was observed against MM.1S cells grown in culture and in mice. In cells, the MTT IC50of KPT-8602 was shifted from 50 to 23 nM by the addition of sub cytotoxic concentrations of panobinostat. In mice, single agent treatment with KPT-8602 led to 96.5% tumor growth inhibition whereas panobinostat resulted in 69.4% tumor growth inhibition within 22 days. Remarkably, in the combination of KPT-8602 and panobinostat, 3 out of 8 tumors totally disappeared and the overall tumor regression was 95%, (Figure 1). Both drugs, as single agents and in combination were well tolerated and no significant changes in weight were observed. Gene and protein expression studies revealed that although both compounds target independent proteins (e.g. HDACs or XPO1), the combination significantly enhances markers of cell death (cleavage of PARP-1, caspase-3, etc.). Curiously, KPT-8602 enhances the inhibitory effect panobinostat has on deacetylation as evidenced by histone acetylation. Moreover, DNA damage, as indicated by ϒ-H2AX, significantly increases in the presence of both compounds. Conclusion : KPT-8602 and panobinostat are dissimilar drugs with unique mechanisms of action, and individually affect a broad range of cellular processes. Here we show that the combination of these drugs can dramatically increase the already potent anti-cancer properties of these compounds in MM cell lines. In addition, KPT-8602 enhances the inhibitory effect exerted by panobinostat on histone deacetylation, which coincides with an increase induction of DNA damage. It should be noted that both panobinostat and SINE compounds have been shown to downregulate checkpoint and DNA damage response (DDR) proteins (e.g. RAD51 and Chk1). We hypothesize that the combination of KPT-8602 and panobinostat promotes significant chromatin remodeling in the presence of a compromised DDR pathway, which destabilizes genomic integrity in MM cells and leads to a synergistic effect on cell viability. Together, these data provide rational support for the study of KPT-8602 and panobinostat in clinical trials. Figure 1 Figure 1. Disclosures Argueta: Karyopharm Therapeutics: Employment, Equity Ownership. Chang:Karyopharm Therapeutics: Employment, Equity Ownership. Kashyap:Karyopharm Therapeutics: Employment, Equity Ownership. Elloul:Rubius Therapeutics: Employment. Friedlander:Karyopharm Therapeutics: Employment. Lee:Karyopharm Therapeutics: Employment, Equity Ownership. Kauffman:Karyopharm Therapeutics Inc: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Shacham:Karyopharm Therapeutics: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Senapedis:Karyopharm Therapeutics: Employment, Equity Ownership. Baloglu:Karyopharm Therapeutics: Employment, Equity Ownership.


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.


Sign in / Sign up

Export Citation Format

Share Document