The Novel Proteasome Inhibitor MLN9708 Demonstrates Efficacy in a Genetically-Engineered Mouse Model of DeNovo Plasma Cell Malignancy.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3849-3849 ◽  
Author(s):  
Siegfried Janz ◽  
Brian G Van Ness ◽  
Vishala Neppalli ◽  
Ray Liu ◽  
Michael D Pickard ◽  
...  

Abstract Abstract 3849 Poster Board III-785 Introduction The first-in-class proteasome inhibitor VELCADË (bortezomib) is a critical component of chemotherapeutic strategy in treating multiple myeloma (MM), a plasma cell malignancy characterized by monoclonal gammopathy and osteolytic lesions. However, patients eventually develop bortezomib resistance, highlighting the continuous challenge to further improve myeloma therapy. We have recently identified a novel proteasome inhibitor MLN9708 that shows greater antitumor activity than bortezomib in a number of preclinical xenograft models. Here we describe antitumor activity of MLN9708 in a genetically engineered mouse model of human MM. In this model, neoplastic plasma cell development is driven by deregulated expression of Myc (c-myc, myelocytomatosis oncogene) and Bcl-x (official gene name: Bcl2l1; encodes the anti-apoptotic Bcl-XL oncoprotein). This is the first preclinical in vivo study of proteasome inhibitors employing a transgenic mouse model of human MM in which plasma cell neoplasms develop de novo. Materials Upon exposure to aqueous solutions or plasma, MLN9708 immediately hydrolyzes to MLN2238, the biologically active form. MLN2238 was used for all preclinical studies described below. Methods We have previously demonstrated that intercrossing C57BL/6 mice carrying the iMycCa transgene (insertion of Myc in the immunoglobulin heavy-chain locus, Igh) with FVB/N mice carrying the 3'KE-Bcl-XL transgene (enforced expression of Bcl-x driven by the immunoglobulin light-chain 3' k enhancer and Vk21 promoter) produces double transgenic F1 hybrid iMycCa/Bcl-XL mice that develop plasma cell malignances with short onset (135 days on average) and full penetrance (100% tumor incidence; J. Clin. Invest. 113:1763-1773, 2004). These double transgenic iMycCa/Bcl-XL mice develop hypergammaglobulinemia and tissue plasmacytosis by 6-8 weeks of age and rapidly succumb to malignant plasma cell tumors by less than 200 days of age. Preliminary studies based on 11 tumor-bearing iMycCa/Bcl-XL mice showed this form of plasma cell malignancy recapitulates important features of human MM, including serum paraproteins, infiltration of bone marrow with malignant plasma cells, osteolytic lesions and myeloma-like global gene expression profiles (Cancer Res. 67:4069-4078, 2007). Here we used the iMycCa/Bcl-XL mouse model of human MM to assess the antitumor activity of bortezomib and MLN9708 in a preclinical setting. Results Untreated double transgenic iMycCa/Bcl-XL mice (n=30) invariably developed plasma cell tumors with short onset (median tumor-free survival = 112 days) and showed marked elevations in plasma immunoglobulin IgM, IgG1, IgG2a and IgG2b levels. To assess antitumor activity of bortezomib and MLN2238, 9-week-old iMycCa/Bcl-XL mice were treated intravenously (IV) twice per week (BIW) with bortezomib (1.2 mg/kg) or MLN2238 (18 mg/kg) for 6 consecutive weeks. These doses represent the maximum tolerated dose (MTD) for each drug as previously determined in normal C57BL/6 x FVB/N F1 hybrid mice. Treatment of iMycCa/Bcl-XL mice with bortezomib (n=30) significantly prolonged tumor-free survival compared to untreated controls (median tumor-fee survival = 139 days; hazard ratio = 0.116; 95% CI = 0.057 to 0.235; p<0.0001). Importantly, treatment of iMycCa/Bcl-XL mice with MLN2238 (n=30) also significantly prolonged tumor-free survival relative to untreated controls (median tumor-free survival = 148 days; hazard ratio = 0.144; 95% CI = 0.071 to 0.289; p<0.0001). Immunoglobulin levels, overall tumor burden, osteolytic lesions (mCT) and biochemical parameters of myeloma bone disease after treatment with bortezomib and MLN2238 will be presented. Conclusion The novel proteasome inhibitor MLN9708 demonstrates striking antitumor activity in a genetically engineered mouse model of human MM, significantly prolonging tumor-free survival of double transgenic iMycCa/Bcl-XL mice. MLN9708 is currently in human clinical development for both hematological and solid tumor indications. Disclosures: Janz: Millennium – The Takeda Oncology Company: Research Funding. Van Ness:Millennium: Research Funding; Scientific Advisory Board, International Myeloma Foundation: Membership on an entity's Board of Directors or advisory committees. Liu:Milllennium: Employment, Equity Ownership. Pickard:Milllennium: Employment. Terkelsen:Milllennium: Employment. Bradley:Milllennium: Employment, Equity Ownership, Research Funding. Kupperman:Milllennium: Employment. Manfredi:Milllennium: Employment. Lee:Milllennium: Employment, Equity Ownership.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3835-3835 ◽  
Author(s):  
Michael Fitzgerald ◽  
Yueying Cao ◽  
Bret Bannerman ◽  
Zhi Li ◽  
Olga Tayber ◽  
...  

Abstract Abstract 3835 Poster Board III-771 Introduction The first generation proteasome inhibitor VELCADE® (bortezomib) is indicated for the treatment of patients with multiple myeloma (MM), a form of plasma cell malignancy (PCM). MLN9708 is our novel proteasome inhibitor that selectively and reversibly binds to, and potently inhibits the b5 site of the 20s proteasome in preclinical studies. We have recently demonstrated that MLN9708 significantly prolongs tumor-free survival of double transgenic iMycCa/Bcl-XL mice, a genetically-engineered mouse model of de novo PCM. Here we describe the in vivo evaluation of cell lines derived from double transgenic iMycCa/Bcl-XL mice and the antitumor activity of MLN9708 in a disseminated mouse model of iMycCa/Bcl-XL PCM. Materials MLN9708 immediately hydrolyzes to MLN2238, the biologically active form, upon exposure to aqueous solutions or plasma. MLN2238 was used for all preclinical studies described below. Double transgenic iMycCa/Bcl-XL mice develop de novo PCM, in which neoplastic plasma cell development is driven by the targeted expression of the oncoprotein Myc and anti-apoptotic Bcl-XL (J. Clin. Invest. 113:1763-1773, 2004). DP54 and DP42 are plasma cell tumor cell lines isolated from the bone marrow and lymph nodes, respectively, of syngeneic mice previously inoculated with iMycCa/Bcl-XL tumors (Cancer Res. 67:4069-4078, 2007). In vitro, DP54 and DP42 cells express both the Myc and Bcl-XL transgenes, various plasma cell and B-cell markers including CD38, CD138 and B220, and have gene expression profiles very similar to human MM. Methods Cell viability studies were performed to determine the antiproliferative effects of MLN2238 in DP54 and DP42 cells in vitro. To evaluate DP54 and DP42 cells in vivo, these cells were aseptically inoculated into the tail vein of NOD-SCID mice. Progressions of the resultant PCM were monitored and tumor burdens were evaluated by magnetic resonance imaging (MRI), ex vivo mCT imaging, and histopathology. Mouse plasma samples were collected at the end of the studies and levels of immunoglobulin were assessed. To establish a preclinical disseminated mouse model of iMycCa/Bcl-XL PCM, freshly dissociated DP54-Luc cells (constitutively expressing firefly luciferase under a mouse Ig-k promoter) were aseptically inoculated into the tail vein of NOD-SCID mice. Once tumor growth has been established, mice were randomized into treatment groups and then treated with vehicle, bortezomib (at 0.7mg/kg intravenously [IV] twice weekly [BIW]) or MLN2238 (at 11 mg/kg IV BIW) for 3 consecutive weeks. Tumor burden was measured by bioluminescent imaging. Results In vitro, both DP54 and DP42 cells were sensitive to MLN2238 treatment (LD50 values of 14 and 25 nM, respectively). In vivo, NOD-SCID mice rapidly succumbed to PCM after being inoculated with DP54 and DP42 cells (25 and 14 days post-inoculation, respectively), where the disease was accompanied by marked elevation of plasma immunoglobulins. MRI scans revealed the presence of multiple lesions and several abnormalities were found including: cranial deformation, bowel distortion, splenomegaly and renal edema. Tumor infiltrates, ranging from minor to extensive, were identified in multiple organ compartments (brain<kidney<liver<lymph nodes<spleen<bone marrow) by histopathological analysis. Ex vivo mCT imaging has also revealed signs of bone erosion in the cranial sagittal sutures. Dissemination of DP54-Luc cells after tail vein inoculations was detected by in vivo bioluminescent and confirmed by ex vivo imaging where luminescent tumor nodules were identified in the spleen, kidneys, liver, intestine, lymph nodes, spinal bone and cranium. To assess the antitumor activity of MLN2238, an efficacy study was performed using the DP54-Luc disseminated model. Tumor burden (bioluminescence), skeletal malformation (mCT) and overall survival after treatment with bortezomib and MLN2238 will be presented. Conclusion The DP54-Luc disseminated mouse model of double transgenic iMycCa/Bcl-XL PCM recapitulated several key features of human MM and provided real-time assessment of novel MM therapy preclinically. MLN9708 is currently in human clinical development for both hematological and solid tumor indications. Disclosures: Cao: Milllennium: Employment, Equity Ownership. Bannerman:Milllennium: Employment. Li:Milllennium: Employment. Bradley:Milllennium: Employment, Equity Ownership, Research Funding. Silverman:Milllennium: Employment. Janz:Milllennium: Research Funding. Van Ness:Milllennium: Research Funding. Kupperman:Milllennium: Employment. Manfredi:Milllennium: Employment. Lee:Milllennium: Employment, Equity Ownership.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2709-2709
Author(s):  
Erik Kupperman ◽  
Edmund Lee ◽  
Yueying Cao ◽  
Bret Bannerman ◽  
Michael Fitzgerald ◽  
...  

Abstract Abstract 2709 Poster Board II-685 Introduction: The ubiquitin-proteasome system processes the majority of cellular proteins and is the principal manner by which cells regulate protein homeostasis. The successful development of bortezomib for multiple myeloma and previously treated mantle cell lymphoma has validated the proteasome as a therapeutic target for hematological malignancies. MLN9708 was identified in screens for a proteasome inhibitor with greater antitumor activity than bortezomib in preclinical xenograft models. MLN9708 immediately hydrolyzes to MLN2238, the biologically active form, upon exposure to aqueous solutions or plasma. MLN2238 was used for all preclinical studies described below. Results: MLN2238 inhibited the 20S proteasome b5 site (IC50 of 3.4 nM), an engineered proteasome substrate (4x ubiquitin-luciferase reporter) and blocked TNFa-induced activation of the NFkB pathway. Cell viability studies confirmed that MLN2238 has potent activity against both myeloma and lymphoma cell lines. The proteasome dissociation half life of MLN2238 was determined to be approximately 6-fold faster than bortezomib, consistent with data generated from Proteasome-Glo wash out experiments where proteasome activity recovered more quickly in MLN2238-treated cells compared to bortezomib. In immunocompromised mice MLN2238 achieved exposures that resulted in significant blood and tumor proteasome inhibition and had increased plasma and tumor exposure compared to bortezomib (when dosed at MTD). In Sprague-Dawley rats, MLN2238 had improved plasma exposure, lower plasma clearance, higher blood Vdss, better oral F% and higher plasma protein binding than bortezomib. In WSU-DLCL2 xenografts, a model of diffuse large B cell lymphoma, greater antitumor activity was seen in mice treated with IV or SC doses of MLN2238 compared to bortezomib. For example, MLN2238 dosed SC QD at its MTD resulted in a T/C of 0.29 compared to bortezomib dosed SC QD at its MTD which resulted in a T/C of 0.79. Pharmacodynamic (PD) responses (in mice) were assessed by evaluating tumor 20S b5 site-specific activity and expression levels of GADD34, an unfolded protein response (UPR) pathway gene shown to be upregulated in response to proteasome inhibition. Consistent with the efficacy difference between MLN2238 and bortezomib in WSU-DLCL2 xenografts, MLN2238 demonstrated an improved PD response compared to bortezomib, showing higher levels of tumor proteasome inhibition and pathway marker elevation. MLN2238 and bortezomib were then evaluated for their ability to reduce tumor burden and improve overall survival in a disseminated model of lymphoma. Tumor burden was tracked over time via bioluminescent scans in NOD-SCID mice inoculated with OCI-Ly7-luciferase cells. The strongest antitumor response was seen in mice treated with MLN2238 SC QD at its MTD, and this dosing regimen also significantly prolonged overall survival compared to vehicle treated controls (median survival was 54 vs. 33 days). Weaker antitumor responses were seen following treatment with bortezomib (either at its SC QD MTD or weekly IV MTD) and these dose regimens did not significantly prolong survival in this study. Conclusions: MLN2238 is a potent, reversible and orally bioavailable proteasome inhibitor with improved pharmacokinetics, pharmacodynamics and antitumor activity in preclinical xenograft models compared to bortezomib. MLN9708 is currently in clinical development for a variety of oncology indications. Disclosures: Kupperman: Milllennium: Employment. Lee:Milllennium: Employment, Equity Ownership. Cao:Milllennium: Employment, Equity Ownership. Bannerman:Milllennium: Employment. Fitzgerald:Millennium Pharmaceuticals: Employment. Berger:Millennium Pharmaceuticals: Employment. Yu:Millennium Pharmaceuticals: Employment. Zhang:Millennium Pharmaceuticals: Employment. Hales:Millennium Pharmaceuticals: Employment. Bruzzese:Millennium Pharmaceuticals: Employment. Liu:Millennium Pharmaceuticals: Employment. Blank:Millennium Pharmaceuticals: Employment. Garcia:Millennium Pharmaceuticals: Employment. Tsu:Millennium Pharmaceuticals: Employment. Dick:Millennium Pharmaceuticals: Employment. Fleming:Millennium Pharmaceuticals: Employment. Yu:Millennium Pharmaceuticals: Employment. Manfredi:Millennium Pharmaceuticals: Employment. Rolfe:Millennium Pharmaceuticals: Employment. Bolen:Millennium Pharmaceuticals: Employment.


PLoS ONE ◽  
2015 ◽  
Vol 10 (10) ◽  
pp. e0140253 ◽  
Author(s):  
Jamie D. Weyandt ◽  
Benjamin L. Lampson ◽  
Sherry Tang ◽  
Matthew Mastrodomenico ◽  
Diana M. Cardona ◽  
...  

2004 ◽  
Vol 25 ◽  
pp. S242 ◽  
Author(s):  
Adam J. Simon ◽  
Lin Chen ◽  
Eric A. Price ◽  
Min Xu ◽  
Adam Lucka ◽  
...  

2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii300-iii300
Author(s):  
Chen Shen ◽  
David Picketts ◽  
Oren Becher

Abstract Diffuse Intrinsic Potine Glioma (DIPG) is a rare pediatric brain tumor for which no cure or efficacious therapies exist. Previous discoveries have revealed that, DIPG harbors distinct genetic alterations, when compared with adult high-grade glioma (HGG) or even with non-DIPG pediatric HGGs. ATRX alteration is found in 9% of clinical cases of DIPG, and significantly overlaps with H3.3K27M mutation and p53 loss, the two most common genetic changes in DIPG, found in 80% and 77% clinical cases, respectively. Here we developed genetically engineered mouse model of brainstem glioma using the RCAS-Tv-a system by targeting PDGF-B overexpression, p53 loss, H3.3K27M mutation and ATRX loss-of function to Nestin-expression brainstem progenitor cells of the neonatal mouse. Specifically, we used Nestin-Tv-a; p53 floxed; ATRX heterozygous female and Nestin-Tv-a; p53 floxed; ATRX floxed male breeders, generated offsprings with ATRX loss of function (n=18), ATRX heterozygous females (n=6), and ATRX WT (n=10). Median survial of the three groups are 65 days, 88 days and 51 days, respectively. Also, ATRX null mice is lower in tumor incidence (44.4%), compared with ATRX WT (80%). We evaluated the pathological features of DIPG with or without ATRX alteration, RNA-seq is performed to identify differentially expressed genes between ATRX WT and loss-of-function. In conclution, this study generated the first genetically modified mouse model studying ATRX loss-of-function in DIPG, and suggested that ATRX loss-of-function in DIPG may slow down tumorigenesis and decrease tumor incidence.


2020 ◽  
Vol 1 (3) ◽  
pp. 100165
Author(s):  
Fernando M. Nunez ◽  
Jessica C. Gauss ◽  
Flor M. Mendez ◽  
Santiago Haase ◽  
Pedro R. Lowenstein ◽  
...  

2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi121-vi121
Author(s):  
Daniel Zamler ◽  
Er-Yen Yen ◽  
Takashi Shingu ◽  
Jiangong Ren ◽  
Cynthia Kassab ◽  
...  

Abstract The introduction of immunotherapies has been paradigm shifting for cancers that were previously a death sentence. However, preclinical/clinical studies on glioblastoma (GBM) have generated mixed outcomes in patients, likely due to its great heterogeneity of immune microenvironment, particularly the myeloid cell populations. Primary patient studies have been limited by a difficulty in performing longitudinal studies, uncontrolled environmental conditions, and genetic variability. There is also, unfortunately, a paucity of mouse models that effectively re-capitulate the immune microenvironment of the human disease. To address these difficulties, we have established the Qk/p53/Pten (QPP) triple knockout mouse model established in our lab. The QPP model uses a cre-lox system to induce Qk deletion on a Pten−/−; p53−/− background which helps NSCs maintain their stemness outside the SVZ in Nes-CreERT2;QkiL/L PtenL/L p53L/L mice, which develops glioblastoma with survival of ~105 days. We have preliminarily assessed the QPP tumors as a faithful model to study the immune response to GBM and found them to recapitulate human GBM with respect to differential response to checkpoint blockade therapy and myeloid and T-cells histopathologically, particularly regarding upregulation of Arginase-1 (Arg1). Arg1 is the canonical marker for tumor-associated macrophages (TAMs), which is a major population of myeloid cells that greatly infiltrate in human GBM, sometimes making up more than ~30% of all GBM cells. Given TAMs’ prevalence in the tumor microenvironment and their upregulation of Arg1 in both human GBM and our QPP model, we are testing whether manipulation of Arg1 will impact TAM function and influence GBM growth. We are also evaluating arginine metabolism in TAMs effect on T cell function in GBM. Lastly, we have developed a genetically engineered mouse model to study the role of Arg1 knockout in a GBM context in-vivo. Our studies suggest that Arg1 plays an important role in GBM immune interaction.


Sign in / Sign up

Export Citation Format

Share Document