Combined Treatment with the Mek Inhibitor PD0325901 and Arsenic Trioxide Has Potent Antitumor Activity in Vivo against Human Multiple Myeloma Xenograft Model.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1712-1712
Author(s):  
Paolo Lunghi ◽  
Laura Mazzera ◽  
Guerino Lombardi ◽  
Micaela Ricca ◽  
Attilio Corradi ◽  
...  

Abstract Despite recent advances in therapy, Multiple myeloma (MM) remains incurable because of the high resistance to apoptosis and both intrinsic and acquired drug resistance. Therefore, new therapeutic strategies are needed to improve patient outcome. We recently demonstrated that blockade of the MEK/ERK signaling module, using the small-molecule inhibitors PD184352 or PD0325901 (PD), strikingly enhances arsenic trioxide (ATO)-induced cytotoxicity in MM cells through a multiple modulation of apoptotic regulatory proteins, including p53 family proteins, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors, several Bcl-2 family proteins and caspases, that depend on the functionality of the p53 pathway (Blood prebublished on line june 26, 2008). Furthermore, we also demonstrated that PD plus ATO treatment induces early tumor (volume approximately 200 mm3) regression, prolongs survival and is well tolerated in vivo in a human plasmacytoma xenograft model. The aim of this study was to investigate whether the combined treatment with PD and ATO is effective in animals with more advanced tumors; thus we used a murine model in which MM RPMI 8226 cells were injected subcutaneously into NOD-SCID mice and when the tumors reached approximately 1000mm3, mice were randomized (n=6/group) to receive vehicle or PD0325901 at 10 mg/kg administered by oral gavage or ATO (3.75 mg/kg) injected intraperitoneally or PD/ATO on a 5-days-a-week schedule for 3 consecutive weeks. Treatment of RPMI 8226 MM-advanced tumor-bearing mice with PD0325901 (10mg/kg) significantly reduced MM-tumor growth as compared to control (P <.01 Tukey-Kramer test), ATO (3.75 mg/kg) had minimal effect on the growth of tumors, which increased as in control mice. Importantly, when PD (10mg/kg) was combined with ATO (3.75mg/Kg), there was a significant reduction in tumor size and growth rate relative to untreated or PD treated mice (P<.001 for PD/ATO versus control, and P<.01 for PD/ATO versus PD Tukey-Kramer test). The combination of PD and ATO (3.75mg/Kg) significantly prolonged survival compared with treatment with either drug alone and was well tolerated in vivo because no differences in body weight and general appearance was noted in mice during the treatment. We next investigated the in vivo effects of the drug combination on proliferation and apoptosis; whole tumor-cell tissues and tumor lysates from mice treated for five days (n=2/group) were subjected to immunohistochemical staining and immunoblotting to assess in vivo phosphorylation of ERK, the proliferative antigen, Ki-67, and cleaved caspase-3. Tumor tissues from PD0325901 (10mg/kg) treatments resulted in profound p-ERK inhibition compared with tumor tissues from vehicle control or ATO-treated animals. In agreement with these data, a significant decrement in the number of Ki-67 positive plasma cells was noted in tumor sections from PD-treated mice relative to tumors from mice receiving either vehicle control or ATO (3.75mg/Kg) treatment alone thereby confirming the tumors growth retardation observed in PD-treated mice. Either PD (10mg/kg) or ATO (3.75mg/Kg) alone did not increase caspase activation compared with tumors from control cohorts. However, the combination PD/ATO dramatically activated caspase-3 in advanced tumors. Notably, consistent with our previous in vitro study demonstrating the involvement of the Bim pathway in MM PD/ATO-induced apoptosis, immunoblotting of MM tumors form PD plus ATO-treated mice showed an elevated ratio of proapoptotic Bim to antiapoptotic Mcl-1 compared with treatment with either drug alone. Collectively, our previous and present findings suggest that combining PD with ATO induces both cytostatic and cytotoxic responses in vivo, resulting in regression of early or advanced tumors, prolongs survival in vivo, and is well tolerated in vivo. In conclusion, our preclinical in vivo studies provide the framework for testing PD0325901 and ATO combination therapy in clinical trials aimed to improve patient outcome in MM.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1846-1846
Author(s):  
Paolo Lunghi ◽  
Laura Mazzera ◽  
Guerino Lombardi ◽  
Micaela Ricca ◽  
Manuela Abeltino ◽  
...  

Abstract Abstract 1846 Although novel drugs such as Bortezomib and Thalidomide have extended the overall survival of multiple myeloma patients they often do not achieve lasting cures, providing an impetus to search for novel therapeutic modalities. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL, also known as Apo2L) is a member of the TNF family of death receptor ligands and has significant potential for use in cancer therapy because of its potent ability to selectively kill cancer cells while sparing normal cells. Preclinical studies have demonstrated that combinations with cytotoxic chemotherapy or other targeted agents may enhance the antitumor activity of TRAIL through cross-talk between the intrinsic and extrinsic pathways. We here demonstrate that targeting Aurora A and B kinases with selective pan-Aurora kinases inhibitors (AKIs) (VX-680, Vertex/Merck; or PHA-680632, Nerviano Medical Sciences, Pfizer) strikingly enhances TRAIL-induced cytotoxicity in TRAIL-sensitive and -resistant human myeloma cell lines (HMCLs) via potentiation of both the death receptor-mediated and mitochondrial apoptotic pathways. In particular, we found that co-treatment with pan-AKIs elevated (DR4+DR5)/(DcR1+DcR2) ratio, caspase-8 activation, Bid fragmentation, mitochondrial depolarization and caspase-9 activation of TRAIL-treated HMCLs. Furthermore we found that the combination of pan-AKIs with TRAIL enhanced the levels of the pro-apoptotic protein Bak and reduced the basal and/or TRAIL-induced expression of the anti-apoptotic Bfl-1/A1 and Mcl-1 at the mitochondrial level; notably, loss of either Mcl-1 or Bfl-1/A1 expression greatly increased TRAIL sensitivity in both sensitive and resistant HMCLs analyzed. To assess the in vivo efficacy of combining TRAIL and Pan-AKIs, we tested these compounds using mouse tumor human plasmacytoma xenograft model in which the drug-resistant RPMI 8226/R5 cells (1.0×107 cells per mouse) were injected subcutaneously into NOD-SCID mice. When the tumors reached approximately 250 mm3 mice bearing RPMI 8226/R5 tumors were randomized (n=12/group) to receive vehicle or MK-0457 or PHA-680632 at 50mg/kg or TRAIL (300μg/per mouse) or MK-0457/TRAIL or PHA-680632/TRAIL. Both Pan-AKIs and TRAIL were administered by intraperitoneal injection. Mice were treated with daily doses of Pan-Aurora inhibitors for 9 days, and two doses of TRAIL. Treatment of RPMI 8226/R5 MM-tumor-bearing mice with MK-0457 or PHA-680632 significantly reduced MM-tumor growth as compared to control (P <.01, Tukey-Kramer test), TRAIL had minimal effect on the growth of tumors, which increased as in control mice. Importantly, when Pan-Aurora inhibitors were combined with TRAIL, there was a significant (P<.001 Dunnet test) reduction in tumor growth relative to either treatment alone. Furthermore, the combination pan-AKIs/TRAIL significantly (P<.001; Kaplan-Meier method and compared using the log-rank test, followed by a Bonferroni correction for multiple comparisons) prolonged survival compared with treatment with either drug alone and was well tolerated in vivo. We next investigated the in vivo effects of the drug combination on proliferation and apoptosis; whole tumor-cell tissues and tumor lysates from mice treated for six days (n=3/group) were subjected to immunohistochemical staining and immunoblotting to assess in vivo phosphorylation histone H3 on Ser10 (phospho-H3), a direct downstream target of the Aurora Kinases, cleaved caspase-3 and PARP fragmentation. Tumor tissues from pan-AKIs treatments resulted in profound phospho-Histone H3 inhibition compared with tumor tissues from vehicle control or TRAIL-treated animals thereby confirming the tumors growth retardation observed in pan-AKIs-treated mice. Either pan-AKIs or TRAIL alone slightly increased caspase-3 cleavage/activation and PARP fragmentation compared with tumors from control cohorts. However, the combination pan-AKIs/TRAIL dramatically increased caspase-3 cleavage/activation, PARP degradation in tumors. In conclusion, our preclinical in vitro and in vivo studies provide the framework for testing pan-AKIs and TRAIL combination therapy in clinical trials aimed to improve patient outcome in MM. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii45-iii46
Author(s):  
W Kim

Abstract BACKGROUND Glioblastoma (GBM) is the most common and aggressive human primary brain malignancy. The key properties of GBM, stemness and invasiveness, are known to be associated with a highly unfavorable prognosis. Notably, the process of epithelial-mesenchymal transition (EMT) is closely related to the progression of GBM. On the basis of reports that 2′-hydroxycinnamaldehyde (HCA) and its derivative, 2′-benzoyloxycinnamaldehyde (BCA), suppresses EMT in several human cancer cells, we sought to evaluate the therapeutic efficacy of HCA and BCA, alone and in combination with temozolomide (TMZ), on GBM tumorspheres (TSs). MATERIAL AND METHODS Two human GBM TSs were treated with HCA, BCA, or TMZ. Therapeutic effects were evaluated by measuring ATP levels, neurosphere formation, 3D-invasion in collagen matrix, and viability. Protein expression profiles after drug treatment were evaluated by western blotting. In vivo anticancer efficacy of drugs was examined in a mouse orthotopic xenograft model. RESULTS Combined treatment of GBM TSs with HCA or BCA and TMZ significantly reduced cell viability, stemness, and invasiveness. Expression levels of stemness-, invasiveness-, and mesenchymal transition-associated markers, Zeb1, N-cadherin, and β-catenin, were also substantially decreased by the combined treatment. The combined treatment also reduced tumor growth in a mouse orthotopic xenograft model. CONCLUSION Our findings suggest that HCA and BCA, combined with TMZ, are potential therapeutic agents in the treatment of GBM.


2018 ◽  
Vol 38 (5) ◽  
Author(s):  
Ruosi Yao ◽  
Xiaoyang Sun ◽  
Yu Xie ◽  
Xiaoshen Sun ◽  
Yao Yao ◽  
...  

Increasing evidence shows that c-Myc oncoprotein is tightly associated with multiple myeloma (MM) progression. Herein, we identified compound 7594-0035, which is a novel inhibitor that specifically targets c-Myc. It was identified from the ChemDiv compound database by molecular docking-based, high-throughput virtual screening. Compound 7594-0035 inhibited MM cell proliferation in vitro, induced cell cycle G2-phase arrest, and triggered MM cell death by disturbing the stability of c-Myc protein. Additionally, we also found that compound 7594-0035 overcame bortezomib (BTZ) drug resistance and increased the killing effect on MM cells in combination with BTZ. The severe combined immune deficiency (SCID) mouse xenograft model revealed that compound 7594-0035 partially decreased the primary tumor growth of Roswell Park Memorial Institute (RPMI)-8226 cells in vivo. The novel small molecular compound 7594-0035 described in the present study that targets c-Myc protein is likely to be a promising therapeutic agent for relapsed/refractory MM.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3022-3022
Author(s):  
Jennifer S. Carew ◽  
Claudia M. Espitia ◽  
Weiguo Zhao ◽  
Valeria Visconte ◽  
Kevin R. Kelly ◽  
...  

Abstract Multiple myeloma (MM) is an incurable plasma cell malignancy and represents the second most common adult hematologic malignancy in the United States. MM is relatively asymptomatic during its early stages and as a result, the majority of patients have advanced disease at diagnosis. Innovations in the treatment of MM, including the development of proteasome inhibitors such as bortezomib (Velcade) have improved clinical outcomes. However, many patients fail to respond to these agents or relapse after initial response highlighting the need for novel therapeutic strategies. Constitutive activation of the MYC oncogene is a frequent pathogenic event in MM that drives disease progression. Aberrant MYC transcriptional activity can increase the levels of a number of factors that are associated with disease progression and drug resistance making it an appealing therapeutic target. Recent studies have demonstrated that inhibition of bromodomain and extra terminal (BET) protein family members including BRD4 decreases the expression of c-MYC and other key oncogenic factors. Here, we demonstrate that shRNA-mediated knockdown of BRD4 or treatment with the BET antagonist JQ1 decreased the expression of c-MYC, BCL-xL, and BCL-2, induced p21 levels, diminished cell viability, and triggered apoptosis in MM cell lines. Comprehensive gene expression profiling of the pharmacodynamic effects of JQ1 revealed that the histone deacetylase HDAC6 was also highly significantly elevated in all MM cell lines and primary patient specimens treated with this agent. Several earlier studies demonstrated that aberrant HDAC6 expression/activity contributes to malignant progression and resistance to a number of classes of anticancer agents including proteasome inhibitors. Based on the roles of HDAC6 in malignant pathogenesis, we hypothesized that its induction may reduce the anti-myeloma activity of JQ1. To test this hypothesis, we utilized both genetic and pharmacological approaches to impair HDAC6 function [shRNA-mediated knockdown of HDAC6, the pan-HDAC inhibitor vorinostat, and the HDAC6-selective inhibitor ACY-1215 (rocilinostat)] and evaluated the consequential impact on the anti-MM effects of JQ1. Notably, antagonzing HDAC6 activity synergistically enhanced the activity of JQ1 in a panel of MM cell lines. These effects were also observed in primary CD138+ cells obtained from patients with MM in a manner that was not affected by prior treatment history. The increased efficacy of these therapeutic combinations was associated with further reductions in c-MYC, BCL-2, and BCL-xL along with significant increases in apoptosis induction as evidenced by enhanced caspase-3 cleavage and DNA fragmentation. Importantly, administration of ACY-1215 was very well tolerated (less than 5% mean transient reduction in body weight) and significantly augmented the in vivo anti-myeloma activity of JQ1 in the RPMI-8226 MM xenograft model as disease burden in combination treated animals was substantially lower than those that received either monotherapy. Immunohistochemical analyses demonstrated that the combination of JQ1 and ACY-1215 led to significantly lower MM cell proliferation (PCNA), increased apoptosis (active caspase-3), and diminished expression of c-MYC and BCL-2. These data suggest for the first time that induction of HDAC6 may represent a key mechanism that promotes drug resistance and limits the efficacy of bromodomain inhibitor therapy. Taken together, our findings demonstrate that abrogation of HDAC6 activity with ACY-1215 or vorinostat is a novel approach to augment the efficacy of bromodomain inhibitors in MM that warrants further investigation. Disclosures Carew: Boehringer Ingelheim: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3005-3005
Author(s):  
Bjoern Jacobi ◽  
Lea Stroeher ◽  
Nadine Leuchtner ◽  
Hakim Echchannaoui ◽  
Alexander Desuki ◽  
...  

Abstract Introduction Starvation of tumor cells from the amino acid arginine has recently gained particular interest because of the downregulation of the rate-limiting enzyme argininosuccinate synthethase 1 (ASS1) in various cancer entities. ASS1-deficient cells cannot resynthesize arginine from citrulline and are therefore considered arginine auxotrophic. The arginine depleting enzyme arginine deiminase (ADI-PEG20, Polaris Pharmaceuticals) is currently tested in phase I-III clinical trials for different arginine auxotrophic cancers. The natural arginine analogue canavanine can compete with arginine for arginyl-tRNA-binding sites and consequently be incorporated into nascent proteins instead of arginine. Canavanine could therefore potentially further disturb intracellular protein homeostasis, especially under arginine deprivation. The sensitivity of myeloma cells towards arginine depletion strategies has not been analyzed so far. Methods Human myeloma cell lines and CD138-sorted primary human myeloma cells from patient bone marrow were screened for ASS1 expression by western blotting (WB). The cells were cultured in arginine free medium and assessed for proliferation and metabolic activity (CFSE/MTT assays), apoptosis (caspase-3 cleavage) and cell death (annexinV/propidium iodide). Canavanine was supplied in both arginine-sufficient and -deficient conditions. The level of intracellular protein stress was determined by WB and/or flow cytometry analysis for ubiquitinated proteins, phosphorylated eukaryotic initiation factor 2α (peIF2α) and the spliced isoform of the X-Box binding protein 1 (Xbp1s). Repetitive ADI-PEG20 ± canavanine application i.p. were tested in vivo in an U266 myeloma xenograft model in NOD/SCID/IL2Rcg-/- (NSG) mice. Arginine and canavanine levels in plasma were determined by HPLC. Tumor growth was measured, mice were assessed for survival, weight and side effects. Tumor tissues were analyzed for caspase-3 cleavage and Ki67 expression by immunohistochemistry. Results 5 of 6 myeloma cell lines were negative for ASS1. Also, ASS1 was either not or only weakly expressed in the majority of primary CD138+ myeloma patient samples. Arginine starvation induced an arrest of cell proliferation and/or metabolic activity of primary myeloma cells and myeloma cell lines after 18-24 h. Addition of citrulline could only rescue ASS1 positive myeloma cells due to the intracellular resynthesis of arginine. Arginine starvation alone led to delayed induction of apoptosis (e.g. 35% cell death of NCI-H929 cells after 72 h of treatment). Addition of 100 mM canavanine strongly increased cell death specifically in the context of arginine deficiency (e.g. cell death in NCI-H929 cells: 87% after 24 h, 100 % after 48h) while it was non-toxic and had no effect on cell viability under physiological arginine conditions. Co-application of canavanine induced ubiquitination of cellular proteins and led to the prolongation of a fatal unfolded protein response (UPR) as measured by markedly elevated Xbp1s levels. Prolonged UPR ultimately led to the induction of apoptosis as reflected by annexin V binding and caspase-3 cleavage. In an U266 myeloma NSG xenograft model, systemic arginine depletion by ADI-PEG20 suppressed tumor growth in vivo and significantly prolonged median survival of mice when compared with the control group (22±3 vs. 15±3 days). Canavanine treatment alone had no influence on viability (13±0 days). However, the combination of ADI-PEG20 and canavanine demonstrated the longest median survival (27±7 days). Histological examination of explanted tumors showed the highest rates of caspase-3 cleavage in the ADI-PEG20/canavanine group. Conclusion Myeloma cells are mostly arginine auxotrophic and can be selectively targeted by arginine starvation. Combination of arginine depletion with the arginine analogue canavanine leads to a highly efficient and specific tumor cell eradication and should be further optimized in multiple myeloma preclinical models. Disclosures Bomalaski: Polaris Pharmaceuticals Inc.: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees.


Author(s):  
Cinzia Lanzi ◽  
Enrica Favini ◽  
Laura Dal Bo ◽  
Monica Tortoreto ◽  
Noemi Arrighetti ◽  
...  

Abstract Background Synovial sarcoma (SS) is an aggressive soft tissue tumor with limited therapeutic options in advanced stage. SS18-SSX fusion oncogenes, which are the hallmarks of SS, cause epigenetic rewiring involving histone deacetylases (HDACs). Promising preclinical studies supporting HDAC targeting for SS treatment were not reflected in clinical trials with HDAC inhibitor (HDACi) monotherapies. We investigated pathways implicated in SS cell response to HDACi to identify vulnerabilities exploitable in combination treatments and improve the therapeutic efficacy of HDACi-based regimens. Methods Antiproliferative and proapoptotic effects of the HDACi SAHA and FK228 were examined in SS cell lines in parallel with biochemical and molecular analyses to bring out cytoprotective pathways. Treatments combining HDACi with drugs targeting HDACi-activated prosurvival pathways were tested in functional assays in vitro and in a SS orthotopic xenograft model. Molecular mechanisms underlying synergisms were investigated in SS cells through pharmacological and gene silencing approaches and validated by qRT-PCR and Western blotting. Results SS cell response to HDACi was consistently characterized by activation of a cytoprotective and auto-sustaining axis involving ERKs, EGR1, and the β-endoglycosidase heparanase, a well recognized pleiotropic player in tumorigenesis and disease progression. HDAC inhibition was shown to upregulate heparanase by inducing expression of the positive regulator EGR1 and by hampering negative regulation by p53 through its acetylation. Interception of HDACi-induced ERK-EGR1-heparanase pathway by cell co-treatment with a MEK inhibitor (trametinib) or a heparanase inhibitor (SST0001/roneparstat) enhanced antiproliferative and pro-apoptotic effects. HDAC and heparanase inhibitors had opposite effects on histone acetylation and nuclear heparanase levels. The combination of SAHA with SST0001 prevented the upregulation of ERK-EGR1-heparanase induced by the HDACi and promoted caspase-dependent cell death. In vivo, the combined treatment with SAHA and SST0001 potentiated the antitumor efficacy against the CME-1 orthotopic SS model as compared to single agent administration. Conclusions The present study provides preclinical rationale and mechanistic insights into drug combinatory strategies based on the use of ERK pathway and heparanase inhibitors to improve the efficacy of HDACi-based antitumor therapies in SS. The involvement of classes of agents already clinically available, or under clinical evaluation, indicates the transferability potential of the proposed approaches.


2021 ◽  
Vol 12 ◽  
Author(s):  
Huibo Dai ◽  
Bangyun Ma ◽  
Xingbin Dai ◽  
Jie Pang ◽  
Jingyu Wang ◽  
...  

Shengma Biejia decoction (SMBJD), a traditional Chinese formula recorded in the Golden Chamber, has been widely used for the treatment of malignant tumors. However, its underlying molecular targets and mechanisms are still unclear. This study showed that SMBJD inhibited tumor growth and stimulated hemogram recovery significantly in a multiple myeloma xenograft model. Western blot and immunohistochemistry assays of tumor tissues showed that SMBJD reduced the ratio of autophagy-related proteins LC3-II/LC3-I, while P62 and apoptosis-related proteins cleaved caspase-3/caspase-3 and Bax/Bcl-2 were upregulated. In vitro experiments demonstrated the time-dependent and dose-dependent cytotoxicity of SMBJD on multiple myeloma cell lines H929 and U266 through MTT assays. The LC3-II/LC3-I ratio and number of GFP-LC3 puncta showed that SMBJD inhibited the autophagy process of H929 and U266 cells. Moreover, both SMBJD and 3-methyladenine (3-MA) caused a decrease in LC3-II/LC3-I, and SMBJD could not reverse the upregulation of LC3-II/LC3-I caused by bafilomycin A1 (Baf-A1). Furthermore, the results of annexin V-FITC and propidium iodide double staining demonstrated that SMBJD treatment induced the apoptosis of H929 and U266 cells. These data prove that SMBJD inhibits autophagy and promotes apoptosis in H929 and U266 cells. The results also show that rapamycin could reduce the rate of SMBJD-induced apoptosis in H929 and U266 cells, at a concentration which had no effect on apoptosis but activated autophagy. In addition, analysis of the mechanism indicated that levels of phosphorylated ERK and phosphorylated mTOR were increased by treatment with SMBJD in vivo and in vitro. These results indicate that SMBJD, an old and effective herbal compound, could inhibit the viability of H929 and U266 cells and induce autophagy-mediated apoptosis through the ERK/mTOR pathway. Thus, it represents a potential therapy strategy for multiple myeloma.


2014 ◽  
Vol 32 (3_suppl) ◽  
pp. 221-221 ◽  
Author(s):  
Amit Deorukhkar ◽  
Nga Diep ◽  
Dev Chatterjee ◽  
Parmeswaran Diagaradjane ◽  
John S. Bomalaski ◽  
...  

221 Background: The benefits of chemoradiation therapy in patients with locally advanced pancreatic cancer (LAPC) are limited due to the inherent radioresistance of pancreatic cancer (PC) and high systemic toxicity of current radiosensitizers (e.g., gemcitabine). Hence, the search for newer radiosensitizers with unique anticancer properties continues. Single amino acid arginine starvation is a new promising therapeutic approach for solid tumors (e.g., PC), that are auxotrophic for non-essential amino acids. Arginine degrading enzyme, arginine deiminase (ADI), deprives cells of arginine and thereby exerts its anti-proliferative effects, especially in cancer cells deficient in enzyme argininosuccinate synthase (ASS1). Here we evaluate the effects of ADI-polyethylene glycol formulation (ADI-PEG20) as a radiosensitizer in PC. Methods: The toxicity of ADI-PEG20 in vitro was evaluated using XTT. Effect of ADI-PEG20 as radiosensitizer was determined by clonogenic cell survival. For in vivo, mice with PC tumor xenografts (Panc1), randomized into four groups, were treated with vehicle (PBS), ADI-PEG20 (5 IU/mouse; twice weekly), radiation (IR; 2 Gy × 5 times), and ADI-PEG20 with IR. Tumors were measured following treatment and the tumor re-growth delay time for each group was calculated. Immunohistochemical analysis of Ki-67 and VEGF was done on tumor tissues (paraffin sections) by routine immunofluorescence. Results: ADI-PEG20 selectively sensitized ASS1 deficient PC cells to IR at low, non-toxic concentrations (0.04 and 0.08 μg/mL for 72 h; DER at 10% SF for Panc1 was 1.39 and 1.52; for Miapaca-2, 1.09 and 1.25 respectively), but not ASS1 positive cells (L3.6pl). In vivo, ADI-PEG20 profoundly sensitized PC cells to IR. IR treatment alone delayed the tumor doubling time (7.6 ± 1.7 days compared to the non-treated controls); however, combining ADI-PEG20 with IR delayed the tumor growth by an additional 10 ± 1.3 days (p<0.05). Immunohistochemical analysis of tumor tissues suggested that ADI-PEG20 with IR down-regulates the expression of Ki-67 and VEGF. Conclusions: ADI-PEG20 potently radiosensitizes PC cells in vitro and in vivo. The detailed molecular mechanism of this radiosensitization warrants further investigations.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2601-2601
Author(s):  
Jie Lin ◽  
Yongji Wu ◽  
Shujie Wang ◽  
Dajun Yang ◽  
Yongqiang Zhao

Abstract Multiple myeloma remains an incurable malignancy and overall survival has not been improved despite responses to conventional and high-dose chemotherapy. Over-expression of both Bcl-2 and Mcl-1 is frequent in multiple myeloma which renders myeloma cells resistant to apoptosis by chemotherapy, and overexpression is associated with relapse and poorer survival. Inhibition of the anti-apoptotic function of Bcl-2 family member proteins such as Bcl-2 and Mcl-1 represents an attractive new strategy for developing anticancer drugs. Apogossypolone (ApoG2) is a novel derivative of the naturally occurring polyphenolic compound gossypol which has aldehyde moieties removed and further modification to make it pharmaceutically more stable. More particularly, ApoG2 is a potent inhibitor of Mcl-1 and Bcl-2, with Ki value of 25 nM for Mcl-1, 35 nM for Bcl-2, respectively. In this study, trypan blue dye exclusion, Hoechst 33258 staining, DNA ladder formation and annexin-V-PI flow cytometric analysis were used to determine the cellular activities of ApoG2 on cell growth inhibition, cell viability, cell cycle and apoptosis. Cleavage of caspase-3 and caspase-9 was analyzed by colorimetirc assay. Xenograft model of Wus1 cells (from Peking Union Medical College Hospital) in nude mice were used to determine the antitumor activity of compounds. We found that ApoG2 resulted in a dose and time-dependent inhibition of multiple myeloma cell proliferation, with IC50 value to both U266 and Wus1 cells at 0.1 to 0.2 uM at 48 hours after treatment. ApoG2 effectively induced apoptosis of multiple myeloma cells as evidenced by typical morphological changes, transmission electron microscopy, DNA ladder formation and increase in the percentage of cells in subdiploid peak. Colorimetric assays further showed activation of both caspase-3 and caspase-9. In a parallel direct comparison study, ApoG2 was more potent than the parental compound gossypol in both growth inhibition and induction of apoptosis. Of interest, cell cycle analysis of both U266 and Wus1 cells treated with ApoG2 produced a slightly G2 arrest, increasing from 9.7% to 19.6% in U266 cells, and from 9.8% to 31.7% in Wus1 cells, respectively. This was different from gossypol which induced mainly G1 arrest. Preliminary in vivo antitumor activity of ApoG2 was examined in xenograft model of Wus1 cells in nude mice, and growth inhibition (T/C) of 32.7% and 33.4% was obtained at 60 mg/kg, and 40 mg/kg, respectively. In addition, there was no body weight loss for both treated groups in comparison with the vehicle treated mice. Our results demonstrated that a potent pan inhibitor of Bcl-2 and Mcl-1 ApoG2 had significant effect of antiproliferation and induction of apoptosis on multiple myeloma cells in vitro and in vivo. ApoG2 may represent a promising new anticancer agent with a novel molecular mechanism and warrant further investigation as a single agent or in combination for human multiple myeloma with Bcl-2/Mcl-1 overexpression.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3707-3707
Author(s):  
Edward Allan R. Sison ◽  
Daniel Magoon ◽  
Li Li ◽  
Barbara Romagnoli ◽  
Garry Douglas ◽  
...  

Abstract Background: Interaction between surface receptor CXCR4 (s-CXCR4) and chemokine SDF-1 (CXCL12) is critical in signaling between leukemic blasts and the bone marrow (BM) microenvironment. We previously demonstrated: 1) chemotherapy-induced upregulation of s-CXCR4 in acute myeloid leukemia (AML) and ALL enhances stromal protection from chemotherapy-induced apoptosis; 2) the FDA-approved CXCR4 inhibitor plerixafor reverses stromal protection and chemotherapy resistance both in vitro in stromal co-cultures of pre-B cell ALL cell lines and in vivo in xenografts of primary samples of infant MLL-rearranged (MLL-R) ALL; 3) the novel Protein Epitope Mimetic POL5551, a selective and potent antagonist of CXCR4, blocks the SDF-1-binding site of CXCR4, inhibits SDF-1-induced chemotaxis, and reverses stromal-mediated protection from chemotherapy in vitro in pre-B and T ALL cell lines. Here, we further characterize the effects of POL5551 (POL) on surface adhesion molecule expression in ALL, and its in vivo effects in a xenograft model of HR pediatric ALL. In Vitro Methods/Results: We have previously shown that POL inhibits 12G5 antibody binding to s-CXCR4 in ALL cell lines, suggesting an overlapping of the two binding sites. We sought to verify these results using primary samples of pediatric ALL. We treated 3 pre B and 3 T cell ALL primary samples with a dose range of POL and measured s-CXCR4 by FACS at multiple time points. POL inhibition of 12G5 binding was potent (average IC50 at 2 hours pre B 8.3 nM, T 1.4 nM), rapid (<1 hour) and sustained (>24 hours). Further, POL was significantly more potent at inhibiting 12G5 binding than plerixafor (average IC50 at 2 hours pre B 18.4 nM, T 8.4 nM). To further characterize POL’s effects in ALL, we treated 2 pre B and 2 T cell ALL cell lines with POL or vehicle control and then treated with SDF-1α or vehicle control. Treatment with POL inhibited SDF-1α-induced phosphorylation of ERK1/2 in a dose-dependent manner. In parallel, we measured POL-induced compensatory upregulation of the alternative surface adhesion molecules CXCR7 and VLA-4 (CD49d), and found that POL led to increased CXCR7 expression at early time points that began to decrease after 24 hours. We did not find a consistent effect of POL on CD49d surface expression. Xenograft Methods/Results: Infant MLL-R ALL primary samples (n=4) were transplanted into sublethally irradiated NSG mice. After 2 weeks, mice were treated on days 1-3 of 2 consecutive weeks with 1) vehicle control (C), 2) POL (5 mg/kg SC), 3) AraC (200 mg/kg IP), or 4) POL followed by AraC 4 hours later (POL+AraC). One week after treatment, cells were harvested from BM, spleen, and peripheral blood (PB). Leukemic blasts were defined as human CD19+ and CD45+. Overall leukemic burden (average % blasts in BM+spleen+PB) did not differ between mice treated with either C (56.2%) or POL (49.5%, p=0.12). However, treatment with AraC (36.7%, p=3E-07) or POL+AraC (26.3%, p=4E-15) significantly decreased total leukemic burden compared to C. Notably, POL+AraC significantly decreased total leukemic burden compared to AraC alone (p=0.001), demonstrating that POL increased overall sensitivity to AraC. When analyzed by organ-specific leukemic burden, POL+AraC resulted in decreased leukemic burden compared to AraC alone in BM (42.8 vs. 49.8%, p=0.27), spleen (16.9 vs. 30.8%, p=0.002), and PB (19.3 vs. 29.6%, p=0.008). Interestingly, AraC and POL+AraC led to significantly increased CXCR7 expression (blasts from BM p<0.01, spleen p<0.04, and PB p<0.03 vs. C) as well as a trend toward increased CD49d expression compared to C. Conclusions: POL5551 is a potent antagonist of s-CXCR4 in pediatric ALL cell lines and primary samples. Surface expression of adhesion molecules in ALL blasts is dynamic and affected by anti-leukemic therapy. Importantly, POL5551 enhances sensitivity to AraC in a xenograft model of infant MLL-R ALL, a HR subtype of pediatric ALL. Our findings support the continued development of BM microenvironment-targeted agents as a therapeutic strategy for pediatric ALL. Disclosures Romagnoli: Polyphor Ltd.: Employment. Douglas:Polyphor Ltd.: Employment. Tuffin:Polyphor Ltd.: Employment. Zimmermann:Polyphor Ltd.: Employment.


Sign in / Sign up

Export Citation Format

Share Document