33 research outputs found

    Abstract 2458: Targeting membrane fluidity as a therapeutic strategy in cancer using BPM 31510

    No full text
    Abstract Cancer cells membranes are relatively more fluid compared to healthy cells. Higher fluidity in cancer cells closely relate to their invasive potential, proliferation, and metastatic ability. Pharmacological modulation of membrane fluidity as a novel therapeutic strategy for potential treatment of cancer is investigated in this study. BPM 31510, a proprietary CoQ10 based liposomal formulation currently in clinical trials affects cell membrane fluidity to influence cancer cell survival. To study the effect of BPM 31510 on biophysical parameters of membrane structure in cancer cells, the CoQ10 concentration in the liposomes was systematically increased and the membrane rigidity (Fluorescence Anisotropy) as function of temperature was measured. A progressive (significant, p&amp;lt;0.05) increase in rigidity of liposomal membranes was observed with increase in CoQ10 concentration to a maxima, after which any further increase in CoQ10 concentration resulted in decreases in membrane rigidity. Interestingly, the local maxima in rigidity matched with the concentration of CoQ10 in BPM 31510 formulation. A spectrum of cell lines including cancer of breast, pancreas, liver, prostate, ovarian cell lines and corresponding healthy non-disease control cell lines including fibroblast, smooth muscles, primary prostate were treated with BPM 31510. For all cell lines, there was a temporal increase in cell membrane rigidity between 0-8 hours following treatment with BPM 31510. The magnitude of increase in membrane rigidity was higher for cells with higher initial membrane fluidity for cancer cells compared to healthy cells. Further, magnitude and rate of this reorganization of cell lipid and protein profile, bioenergetics and metabolic state as a maladaptive response of cells to BPM 31510 correlated with the magnitude of change in cell membrane rigidity. Finally, decrease in cell viability was higher for cells with higher membrane fluidity compared to cells with rigid membrane after 48 hours of treatment of BPM 31510. In in vivo preclinical animal model of cancer, there was a strong correlation between BPM 31510 in-plane rigidity measurements to observed bio-distribution and efficacy. This work provides profound insight into CoQ10 effect on cell membrane dynamics and mechanism of action of BPM 31510. The study provides compelling evidence in support of targeting membrane fluidity characteristics in cancer cells as a novel modality with therapeutic potential in the treatment of cancer. Citation Format: Sumit Garg, Sirisha Dhavala, Katerina Krumova, Vivek K. Vishnudas, Joaquin J. Jimenez, Michael Kiebish, Rangaprasad Sarangarajan, Niven R. Narain. Targeting membrane fluidity as a therapeutic strategy in cancer using BPM 31510. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2458. doi:10.1158/1538-7445.AM2015-2458</jats:p

    Abstract 4934: Employing theBerg Interrogative Biology® systems platform to elucidate cellular signaling changes that contribute to the pathophysiology of hepatocellular carcinoma (HCC)

    No full text
    Abstract Hepatocellular Carcinoma (HCC) is one of the leading causes of cancer-related death worldwide, ranked as the third most fatal cancer after lung and stomach carcinomas. The diverse etiology, high morbidity/mortality, lack of diagnostic markers for early diagnosis and the highly variable clinical course of HCC have hindered advances in diagnosis and treatment. After years of studying the HCC, the understanding of molecular mechanism operational in HCC remains incomplete. The genomic, transcriptomic and comparative proteomic profiles have yielded some important insights for HCC research. However, many studies focused on single aspect of the cellular changes associated with HCC, hindering the full understanding of biological systems in their true complexity and dynamics. We describe a novel approach combining the power of cell biology, integrated proteomics platforms and an informatics platform that generates causal protein networks. In order to delineate the role of post-translation modification and enzymes that partake in such mechanisms, we incorporated activity based proteomics employing kinase enrichment probes and phosphoproteome mapping of total proteins in HCC cellular models. Multikinase inhibitor Sorafenib, the first line chemotherapeuitc agent for the advanced HCC patients, was used to probe the global kinase activity and protein phosphorylation changes associated with this treatment. Comparative proteomics, phosphoproteome and kinase activity data was integrated into the AI based REFS™ informatics platform. Causal networks of protein interaction specifically from a functional stand point namely kinase activity and potential targets that kinases can phosphorylate were generated. In addition, using cellular functional read out, proteins/ kinases that modulate phosphorylation of targets and mechanistically drive pathophysiological cellular behavior were determined. The approach outlined here enables global characterization of cellular responses, insights into mechanisms of chemo sensitivity and potential targets/biomarkers for clinical management of HCC. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4934. doi:1538-7445.AM2012-4934</jats:p

    Abstract 208: BPM 31510-induced alteration in Complex II activity is functionally linked to cell death activation pathway in a preclinical model of triple-negative breast cancer

    No full text
    Abstract Although only 15-20% of total breast cancer diagnoses are of the triple-negative breast cancer (TNBC) subtype, they account for a significant portion of the mortality rate due to their more aggressive phenotype and a high risk of reoccurrence. Metabolic rewiring supports breast cancer progression and metastasis, particularly in ER-negative and triple-negative (TNBC) breast tumors. Thus, we examined the effects of BPM 31510, a metabolic-modulating agent in clinical trials for solid tumors, in in vitro and in vivo ER-negative and TNBC models. BPM 31510 EC50/EC&amp;gt;90 values were determined for a panel of the breast cancer cell lines and compared to non-tumorigenic MCF12A cells in vitro, and the MDA-MB231 and SkBr-3, TNBC and ER-negative models respectively, were found to be the most sensitive to BPM 31510. Treatment with BPM 31510 (EC50 and EC&amp;gt;90 doses) resulted in a time- and dose-dependent decrease the viable cell population (PI- and Annexin V-negative) and a concomitant increase in cells in early and late apoptosis (PI-negative and PI-positive Annexin V-positive cells, respectively), suggesting that BPM 31510 activates regulated cell death pathways. Consistent with the in vitro data, MDA-MB231 tumor-bearing mice had smaller tumors after 30 days of treatment with BPM 31510 and increased cleaved caspase 3 staining in resected tumors. In vitro, BPM 31510-dependent breast cancer cell death was preceded by mitochondrial membrane potential depolarization (TMRE flow cytometry) and alterations in mitochondrial respiration characterized by a consistent, dose-dependent decrease in succinate (Complex II)-fueled respiration with more varied responses to BPM 31510 in cells provided the Complex I substrates (pyruvate or palmitoyl carnitine). To investigate the role of Complex II in BPM 31510-mediated cell death, pharmacological inhibitors of the dicarboxylate site (malonate) and Qp site (atpenin A5) of Complex II were used in combination with BPM 31510 to assess the resultant effects on cell death in MDA-MB231 cells. Co-treatment with malonate significantly attenuated BPM 31510-mediated cell death while atpenin A5 did not affect BPM 31510-induced cell death, indicating succinate oxidation at the dicarboxylate site of Complex II is required, in part, for induction of cell death by BPM 31510. Together, these data demonstrate BPM 31510 has a potent anti-cancer activity in preclinical breast cancer models and define a functional link between Complex II activity and the mechanism of action for BPM 31510. Citation Format: Tulin Dadali, Anne R. Diers, Arleide Lee, Ezer Benaim, Joaquin J. Jimenez, Stephane Gesta, Vivek K. Vishnudas, Rangaprasad Sarangarajan, Niven R. Narain. BPM 31510-induced alteration in Complex II activity is functionally linked to cell death activation pathway in a preclinical model of triple-negative breast cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 208.</jats:p

    Integrated Metabolomics Assessment of Human Dried Blood Spots and Urine Strips

    No full text
    (1) Background: Interest in the application of metabolomics toward clinical diagnostics development and population health monitoring has grown significantly in recent years. In spite of several advances in analytical and computational tools, obtaining a sufficient number of samples from patients remains an obstacle. The dried blood spot (DBS) and dried urine strip (DUS) methodologies are a minimally invasive sample collection method allowing for the relative simplicity of sample collection and minimal cost. (2) Methods: In the current report, we compared results of targeted metabolomics analyses of four types of human blood sample collection methods (with and without DBS) and two types of urine sample collection (DUS and urine) across several parameters including the metabolite coverage of each matrix and the sample stability for DBS/DUS using commercially available Whatman 903TM paper. The DBS/DUS metabolomics protocols were further applied to examine the temporal metabolite level fluctuations within hours and days of sample collection. (3) Results: Several hundred polar metabolites were monitored using DBS/DUS. Temporal analysis of the polar metabolites at various times of the day and across days identified several species that fluctuate as a function of day and time. In addition, a subset of metabolites were identified to be significantly altered across hours within a day and within successive days of the week. (4) Conclusion: A comprehensive DBS/DUS metabolomics protocol was developed for human blood and urine analyses. The described methodology demonstrates the potential for enabling patients to contribute to the expanding bioanalytical demands of precision medicine and population health studies

    Abstract 1736: Mitochondrial priming of apoptotic machinery in pancreatic cancer by BPM 31510 to enhance efficacy of chemotherapy

    No full text
    Abstract Recent evidence suggests that the cytotoxic effects of chemotherapy, specifically in cancer cells is a consequence of “pre-treatment priming” event wherein exposure to the chemotherapeutic agents pushes the cancer cells beyond an apoptotic threshold to effectuate cell death. (Letai, A et. al, Science. 2011 Nov 25;334 (6059) BPM 31510 is a ubidecarenone containing proprietary formulation that effectuates a metabolic switch from glycolysis towards enhanced mitochondrial oxidative phosphorylation resulting in the recapitulation of apoptosis in cancer. The current study was aimed at investigating whether pretreatment with BPM 31510 results in mitochondrial priming, thereby augmenting the cytotoxic effect of standard of care chemotherapeutic agents. The human derived pancreatic cancer cell line Mia-Paca-2 was subjected to either (a) pretreatment with BPM 31510 followed by co-treatment with gemcitabine or (b) co-treatment with BPM 31510 and gemcitabine. BPM 31510 exposure resulted in decreased cell proliferation rates that were significantly higher than gemcitabine alone in Mia-Paca-2 cell line. Pre-treatment with BPM 31510 augmented the cytotoxic potential of gemcitabine, a phenomenon that was also observed in the co-treatment regimen. In vivo pancreatic cancer model using Mia-Paca-2 cell lines were developed to further investigate the priming effect of BPM 31510. Results demonstrate that 1-week and 2-week pretreatment with intravenous BPM 31510 followed by gemcitabine results in improved survival in the pancreatic cancer model compared to the co-treatment regimen. The data demonstrates that addition of BPM 31510 synergizes the cytotoxic effect of gemcitabine in pancreatic cancer. In addition, pretreatment with BPM 31510 followed by gemcitabine treatment is associated with improved survival. Taken together, the data suggest that BPM 31510 may be a viable mitochondrial priming agent to sensitize cancer cells to the cytotoxic effects of gemcitabine in pancreatic cancer. Citation Format: Niven R. Narain, Tony E. Walshe, Arleide Lee, Rakib Ouro-Djoba, Lucia Mauro, Adel Yunis, Vivek K. Vishnudas, Rangaprasad Sarangarjan, Joaquin J. Jimenez. Mitochondrial priming of apoptotic machinery in pancreatic cancer by BPM 31510 to enhance efficacy of chemotherapy. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1736. doi:10.1158/1538-7445.AM2013-1736</jats:p
    corecore