Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Olga Burenkova is active.

Publication


Featured researches published by Olga Burenkova.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Antitumor activity of MLN8054, an orally active small-molecule inhibitor of Aurora A kinase

Mark Manfredi; Jeffrey A. Ecsedy; Kristan Meetze; Suresh K. Balani; Olga Burenkova; Wei Chen; Katherine M. Galvin; Kara M. Hoar; Jessica Huck; Patrick J. LeRoy; Emily T. Ray; Todd B. Sells; Bradley Stringer; Stephen G. Stroud; Tricia J. Vos; Deborah R. Wysong; Mengkun Zhang; Joseph B. Bolen; Christopher F. Claiborne

Increased Aurora A expression occurs in a variety of human cancers and induces chromosomal abnormalities during mitosis associated with tumor initiation and progression. MLN8054 is a selective small-molecule Aurora A kinase inhibitor that has entered Phase I clinical trials for advanced solid tumors. MLN8054 inhibits recombinant Aurora A kinase activity in vitro and is selective for Aurora A over the family member Aurora B in cultured cells. MLN8054 treatment results in G2/M accumulation and spindle defects and inhibits proliferation in multiple cultured human tumor cells lines. Growth of human tumor xenografts in nude mice was dramatically inhibited after oral administration of MLN8054 at well tolerated doses. Moreover, the tumor growth inhibition was sustained after discontinuing MLN8054 treatment. In human tumor xenografts, MLN8054 induced mitotic accumulation and apoptosis, phenotypes consistent with inhibition of Aurora A. MLN8054 is a selective inhibitor of Aurora A kinase that robustly inhibits growth of human tumor xenografts and represents an attractive modality for therapeutic intervention of human cancers.


Cancer Research | 2010

An ErbB3 Antibody, MM-121, Is Active in Cancers with Ligand-Dependent Activation

Birgit Schoeberl; Anthony C. Faber; Danan Li; Mei-Chih Liang; Katherine Crosby; Matthew Onsum; Olga Burenkova; Emily Pace; Zandra E. Walton; Lin Nie; Aaron Fulgham; Youngchul Song; Ulrik Nielsen; Jeffrey A. Engelman; Kwok-Kin Wong

ErbB3 is a critical activator of phosphoinositide 3-kinase (PI3K) signaling in epidermal growth factor receptor (EGFR; ErbB1), ErbB2 [human epidermal growth factor receptor 2 (HER2)], and [hepatocyte growth factor receptor (MET)] addicted cancers, and reactivation of ErbB3 is a prominent method for cancers to become resistant to ErbB inhibitors. In this study, we evaluated the in vivo efficacy of a therapeutic anti-ErbB3 antibody, MM-121. We found that MM-121 effectively blocked ligand-dependent activation of ErbB3 induced by either EGFR, HER2, or MET. Assessment of several cancer cell lines revealed that MM-121 reduced basal ErbB3 phosphorylation most effectively in cancers possessing ligand-dependent activation of ErbB3. In those cancers, MM-121 treatment led to decreased ErbB3 phosphorylation and, in some instances, decreased ErbB3 expression. The efficacy of single-agent MM-121 was also examined in xenograft models. A machine learning algorithm found that MM-121 was most effective against xenografts with evidence of ligand-dependent activation of ErbB3. We subsequently investigated whether MM-121 treatment could abrogate resistance to anti-EGFR therapies by preventing reactivation of ErbB3. We observed that an EGFR mutant lung cancer cell line (HCC827), made resistant to gefitinib by exogenous heregulin, was resensitized by MM-121. In addition, we found that a de novo lung cancer mouse model induced by EGFR T790M-L858R rapidly became resistant to cetuximab. Resistance was associated with an increase in heregulin expression and ErbB3 activation. However, concomitant cetuximab treatment with MM-121 blocked reactivation of ErbB3 and resulted in a sustained and durable response. Thus, these results suggest that targeting ErbB3 with MM-121 can be an effective therapeutic strategy for cancers with ligand-dependent activation of ErbB3.


Molecular Cancer Therapeutics | 2015

Enhanced Targeting of the EGFR Network with MM-151, an Oligoclonal Anti-EGFR Antibody Therapeutic

Jeffrey D. Kearns; Raghida Bukhalid; Mark Sevecka; Gege Tan; Nastaran Gerami-Moayed; Shannon L. Werner; Neeraj Kohli; Olga Burenkova; Callum M. Sloss; Anne M. King; Jonathan Fitzgerald; Ulrik Nielsen; Beni B. Wolf

Although EGFR is a validated therapeutic target across multiple cancer indications, the often modest clinical responses to current anti-EGFR agents suggest the need for improved therapeutics. Here, we demonstrate that signal amplification driven by high-affinity EGFR ligands limits the capacity of monoclonal anti-EGFR antibodies to block pathway signaling and cell proliferation and that these ligands are commonly coexpressed with low-affinity EGFR ligands in epithelial tumors. To develop an improved antibody therapeutic capable of overcoming high-affinity ligand-mediated signal amplification, we used a network biology approach comprised of signaling studies and computational modeling of receptor–antagonist interactions. Model simulations suggested that an oligoclonal antibody combination may overcome signal amplification within the EGFR:ERK pathway driven by all EGFR ligands. Based on this, we designed MM-151, a combination of three fully human IgG1 monoclonal antibodies that can simultaneously engage distinct, nonoverlapping epitopes on EGFR with subnanomolar affinities. In signaling studies, MM-151 antagonized high-affinity EGFR ligands more effectively than cetuximab, leading to an approximately 65-fold greater decrease in signal amplification to ERK. In cell viability studies, MM-151 demonstrated antiproliferative activity against high-affinity EGFR ligands, either singly or in combination, while cetuximab activity was largely abrogated under these conditions. We confirmed this finding both in vitro and in vivo in a cell line model of autocrine high-affinity ligand expression. Together, these preclinical studies provide rationale for the clinical study of MM-151 and suggest that high-affinity EGFR ligand expression may be a predictive response marker that distinguishes MM-151 from other anti-EGFR therapeutics. Mol Cancer Ther; 14(7); 1625–36. ©2015 AACR.


npj Systems Biology and Applications | 2017

Systems biology driving drug development: from design to the clinical testing of the anti-ErbB3 antibody seribantumab (MM-121)

Birgit Schoeberl; Art Kudla; Kristina Masson; Ashish Kalra; Michael D. Curley; Gregory J. Finn; Emily Pace; Brian Harms; Jaeyeon Kim; Jeff Kearns; Aaron Fulgham; Olga Burenkova; Viara P. Grantcharova; Defne Yarar; Violette Paragas; Jonathan Fitzgerald; Marisa Wainszelbaum; Kip A. West; Sara Mathews; Rachel Nering; Bambang Adiwijaya; Gabriela Garcia; Bill Kubasek; Victor Moyo; Akos Czibere; Ulrik Nielsen; Gavin MacBeath

The ErbB family of receptor tyrosine kinases comprises four members: epidermal growth factor receptor (EGFR/ErbB1), human EGFR 2 (HER2/ErbB2), ErbB3/HER3, and ErbB4/HER4. The first two members of this family, EGFR and HER2, have been implicated in tumorigenesis and cancer progression for several decades, and numerous drugs have now been approved that target these two proteins. Less attention, however, has been paid to the role of this family in mediating cancer cell survival and drug tolerance. To better understand the complex signal transduction network triggered by the ErbB receptor family, we built a computational model that quantitatively captures the dynamics of ErbB signaling. Sensitivity analysis identified ErbB3 as the most critical activator of phosphoinositide 3-kinase (PI3K) and Akt signaling, a key pro-survival pathway in cancer cells. Based on this insight, we designed a fully human monoclonal antibody, seribantumab (MM-121), that binds to ErbB3 and blocks signaling induced by the extracellular growth factors heregulin (HRG) and betacellulin (BTC). In this article, we present some of the key preclinical simulations and experimental data that formed the scientific foundation for three Phase 2 clinical trials in metastatic cancer. These trials were designed to determine if patients with advanced malignancies would derive benefit from the addition of seribantumab to standard-of-care drugs in platinum-resistant/refractory ovarian cancer, hormone receptor-positive HER2-negative breast cancer, and EGFR wild-type non-small cell lung cancer (NSCLC). From preclinical studies we learned that basal levels of ErbB3 phosphorylation correlate with response to seribantumab monotherapy in mouse xenograft models. As ErbB3 is rapidly dephosphorylated and hence difficult to measure clinically, we used the computational model to identify a set of five surrogate biomarkers that most directly affect the levels of p-ErbB3: HRG, BTC, EGFR, HER2, and ErbB3. Preclinically, the combined information from these five markers was sufficient to accurately predict which xenograft models would respond to seribantumab, and the single-most accurate predictor was HRG. When tested clinically in ovarian, breast and lung cancer, HRG mRNA expression was found to be both potentially prognostic of insensitivity to standard therapy and potentially predictive of benefit from the addition of seribantumab to standard of care therapy in all three indications. In addition, it was found that seribantumab was most active in cancers with low levels of HER2, consistent with preclinical predictions. Overall, our clinical studies and studies of others suggest that HRG expression defines a drug-tolerant cancer cell phenotype that persists in most solid tumor indications and may contribute to rapid clinical progression. To our knowledge, this is the first example of a drug designed and clinically tested using the principles of Systems Biology.


Cancer Research | 2011

Abstract 655: Combination of MM-111, an ErbB2/ErbB3 bispecific antibody, with endocrine therapies as an effective strategy for treatment of ER+/HER2+ breast cancer

Bo Zhang; Alexandra Huhalov; Olga Burenkova; Sharlene Adams; Stephanie Nguyen; Ryan Overland; Arthur J. Kudla; Ulrik Nielsen; Clet Niyikiza; Charlotte Mcdonagh; Gavin MacBeath

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Approximately 75% of breast cancers are estrogen receptor (ER) positive. Although endocrine therapies such as tamoxifen, fulvestrant, and letrozole have demonstrated significant efficacy in treating ER+ breast cancer patients, intrinsic or acquired resistance has limited their success. Recent studies suggest that crosstalk between ErbB receptor signaling and ER signaling may contribute to resistance to endocrine therapy. Overexpression of human epidermal growth factor receptor 2 (HER2, synonymous with ErbB2) and upregulation of the ErbB3 ligand heregulin are associated with poor prognosis and reduced overall survival. MM-111 is a novel bispecific antibody fusion protein that specifically targets the ErbB2/ErbB3 heterodimer and blocks heregulin binding to ErbB3. MM-111 inhibits ligand-induced ErbB3 phosphorylation, tumor cell cycle progression, and tumor growth when ErbB2 is overexpressed. We hypothesized that combination of endocrine therapies with MM-111 may improve anti-tumor efficacy. In an estrogen-stimulated BT474-M3 ER-positive breast cancer cell three-dimensional spheroid assay, MM-111, when used as a single agent, showed growth inhibitory effects similar to the anti-estrogen drugs tamoxifen and fulvestrant. Combination of MM-111 with anti-estrogen therapy showed superior activity to either drug alone. In the presence of heregulin, MM-111 maintained its growth inhibitory activity, whereas the inhibitory effect of tamoxifen and fulvestrant was diminished. This suggests that activation of ErbB3 confers tumor cell resistance to anti-estrogen therapies. When both estrogen and heregulin were present, the combination of MM-111 and the anti-estrogen drugs demonstrated a significantly greater inhibitory effect than either drug alone. Western blot analysis showed that treatment of BT-474-M3 cells with the combination of MM-111 and the anti-estrogen drugs significantly increased apoptosis markers such as cytochrome C and BAX. Furthermore, an in vivo BT474-M3 xenograft model showed resistance to tamoxifen treatment (5 mg/pellet, 60-day release). In this xenograft model MM-111 sensitized tumor response to tamoxifen and the combination treatment dramatically inhibited tumor growth. In conclusion, the combination of MM-111 and endocrine therapies may provide a potent regimen that overcomes acquired resistance to endocrine therapies in ER+, ErbB2-overexpressing breast cancer patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 655. doi:10.1158/1538-7445.AM2011-655


Molecular Cancer Therapeutics | 2015

Abstract LB-C25: Inhibition of ERBB3 with MM-121, IGF1-R with MM-141 or Met with MM-131 increases the activity of EGFR inhibitor MM-151 in colorectal cancer models expressing multiple resistance ligands

Marisa Wainszelbaum; Jessica Fessler; Johanna Lahdenranta; Olga Burenkova; Nat Gerami-Moayed; Yasmin Hashambhoy-Ramsay; Victoria Rimkunas; Gavin MacBeath

Introduction: The Epidermal Growth Factor Receptor (EGFR) pathway is a key driver of late-stage colorectal cancer (CRC) and EGFR inhibitors, such as cetuximab and panitumumab, prolong survival in a large subset of patients. Not all patients respond to EGFR inhibitor therapy, however, and resistance develops in those who respond. Accumulating evidence suggests that alternative signaling pathways circumvent EGFR blockade. Several studies have shown that tumors with high heregulin, IGF-1 or HGF respond poorly to EGFR-targeted therapies. Further, Phase 2 studies suggest that combined inhibition of EGFR and HGF with panitumumab and rilotumumab significantly improves response rates compared to panitumumab alone. These data support the notion that more than one pathway may be simultaneously active in late-stage CRC and that EGFR-directed therapy may be improved through combination strategies. Merrimack has built a portfolio of biologic agents targeting key oncogenic receptors that block ligand binding and pathway activation, providing a unique opportunity to develop combination therapies in a biomarker-driven fashion. MM-151 targets EGFR; MM-121 (seribantumab) targets ERBB3; MM-141 (istiratumab) targets both IGF-1R and ERBB3; and MM-131 targets Met and EpCAM. Here we investigate responsiveness to different resistance ligands and the ability of drug combinations to block ligand-mediated cell growth more effectively than monotherapy. Experimental procedures: We evaluated the prevalence of resistance ligands in colorectal adenocarcinoma patient samples by RNA in situ hybridization. To assess the effect of ligands and drugs on cell growth we assembled a panel of 29 genetically diverse CRC cell lines. Using a carefully optimized in vitro 3D culture system, we measured cell viability in response to MM-151, MM-121, MM-141, MM-131 or a combination of the drugs using MM-151 as the backbone. The screen was performed in the presence or absence of exogenously added ligands: EGF, heregulin, IGF-1 or HGF. Using these data, we selected representative xenograft models for in vivo analysis of drug combination efficacy. Results: We show that colorectal cancer tumors co-express resistance ligands and that ligand prevalence appears to increase following treatment. Results from our in vitro screen show that CRC cell lines fall into two groups: those that are unresponsive to ligand stimulation and those that are responsive. Interestingly, cell lines that are responsive to one ligand tend to respond to other ligands as well, suggesting that blocking more than one pathway may prove more effective than blocking only a single pathway. In pre-clinical models of cell lines and xenografts, where ligands are co-expressed, combinations of targeted antibodies, using MM-151 as the backbone, have greater activity than monotherapy. Conclusions: These data suggest that subsets of patients with high expression of individual or multiple ligands could benefit from different combinations of targeted therapies. A Phase 1 study evaluating the safety, pharmacology and preliminary activity of the co-administration of MM-151 and MM-121 (seribantumab) in heregulin positive cancer patients is currently enrolling patients with advanced colorectal cancer, non-small cell lung cancer, and head and neck cancer. Citation Format: Marisa J. Wainszelbaum, Jessica Fessler, Johanna Lahdenranta, Olga Burenkova, Nat Gerami-Moayed, Yasmin Hashambhoy-Ramsay, Victoria Rimkunas, Gavin MacBeath. Inhibition of ERBB3 with MM-121, IGF1-R with MM-141 or Met with MM-131 increases the activity of EGFR inhibitor MM-151 in colorectal cancer models expressing multiple resistance ligands. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr LB-C25.


Cancer Research | 2016

Abstract 2148: MM-151 elicits broad and unique inhibition of cells harboring EGFR extracellular domain mutations — results of multiscale experiments with genome-edited cell lines

Hongfang Wang; Shawn P. Carey; Olga Burenkova; Jian Tang; Nastaran Gerami-Moayed; Sabrina Arena; Alberto Bardelli; Rachel Nering; Jeffrey D. Kearns

Mutations in the extracellular domain (ECD) of EGFR have been reported in colorectal (CRC) patients as a mechanism of acquired resistance to the EGFR-directed antibodies cetuximab and panitumumab. Similar to EGFR intracellular domain mutations observed in patients treated with EGFR kinase inhibitors, these EGFR-ECD mutations inhibit the binding of the antibodies while maintaining EGFR signaling activity. MM-151, an investigational agent consisting of three anti-EGFR IgG1 antibodies, has been recently reported to overcome these mutations in over-expression and patient-derived cell line models. Here, we extend these studies to characterize the mechanisms of action that enable MM-151 to provide robust inhibition of EGFR-ECD mutations beyond that achievable by other monoclonal and oligoclonal EGFR antibodies. First, utilizing a cell-free binding assay with recombinant EGFR-ECD mutants, we found that MM-151, but not the Sym004 combination of two antibodies, maintained oligoclonal binding to all mutants. Second, we performed a panel of 2D and 3D in vitro experiments with over-expression and genome-edited (CRISPR-Cas9) cell lines harboring EGFR-ECD mutations to characterize target engagement, cell signaling, proliferation, and the impact of hetero- versus homozygous expression of EGFR wild type or mutant alleles. We observed that the three mechanisms of action for MM-151- antagonism of both low- and high-affinity EGFR ligands, EGFR down-regulation, and immune-effector activity— were preserved in these models. Third, we performed xenograft experiments to evaluate the behavior of MM-151 and found that MM-151 maintained inhibitory activity in an in vivo context. The results of our multiscale experiments identified the impact of combining three antibodies to overcome the high plasticity of the EGFR extracellular domain and thus reveal the potential for clinical evaluation of MM-151 in both EGFR-naive and EGFR-refractory CRC populations. Citation Format: Hongfang Wang, Shawn P. Carey, Olga Burenkova, Jian Tang, Nastaran Gerami-Moayed, Sabrina Arena, Alberto Bardelli, Rachel Nering, Jeffrey D. Kearns. MM-151 elicits broad and unique inhibition of cells harboring EGFR extracellular domain mutations — results of multiscale experiments with genome-edited cell lines. [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 2148.


Cancer Research | 2015

Abstract LB-243: The ErbB3-targeting antibody MM-121 (seribantumab) reverses heregulin-driven resistance to multiple chemotherapies on tumor cell growth

Kristina Masson; Viara P. Grantcharova; Olga Burenkova; Marisa Wainszelbaum; Sergio Iadevaia; Sharlene Adams; Andreas Raue; Akos Czibere; Birgit Schoeberl; Gavin MacBeath

Purpose: Heregulin-mediated activation of the human epidermal growth factor receptor 3 (HER3/ErbB3) is required for the growth and survival of many epithelial cancers. This signaling pathway is also emerging as a mechanism of resistance to targeted agents and chemotherapies. MM-121 (seribantumab) is an investigational human monoclonal anti-ErbB3 antibody that has previously been shown to effectively block ligand-dependent activation of ErbB3 in a range of tumors, and has demonstrated clinical activity in biomarker positive patients in several Phase II trials. The purpose of this study was to examine in three indications of interest, the ability of heregulin to induce resistance to standard chemotherapies and the reversal of this effect by MM-121. Such systematic evaluation of different combinations can serve as a guide for the future clinical development of MM-121. Methods: To assess the effect of heregulin and MM-121 on chemotherapies in cancer cells, we conducted a high throughput proliferation screen in 3D cultures. A panel of 60 cell lines of relevant clinical indications (ovarian, breast and lung cancer) was selected and tested for the sensitivity to respective standard-of-care chemotherapies in the absence or presence of exogenously added heregulin. Using these data, we analyzed the rescuing capacity of heregulin and the MM-121 combination9s sensitivity, and selected representative combinations for in vivo models. Results: We show that in a large panel of cancer cell lines the presence of heregulin can induce resistance to multiple chemotherapies with very different mechanisms of action. The combination of MM-121 with any one of these chemotherapies can reverse the heregulin-meditated rescue and provide an additive treatment effect at therapeutically relevant doses achieved in the clinic. These results were further validated in xenograft mouse models of all three indications, using representative chemotherapies and doses. In addition, biomarker analysis revealed that ErbB3 receptor levels largely determine responsiveness to heregulin and MM-121. Conclusions: MM-121 is an anti-ErbB3 antibody designed to block ligand-mediated signaling, and currently in clinical development. The results presented here demonstrate the role of heregulin in reducing the sensitivity of tumors to standard-of-care chemotherapies, and the effect of ErbB3 pathway inhibition across indications. Citation Format: Kristina Masson, Viara Grantcharova, Olga Burenkova, Marisa Wainszelbaum, Sergio Iadevaia, Sharlene Adams, Andreas Raue, Akos Czibere, Birgit Schoeberl, Gavin MacBeath. The ErbB3-targeting antibody MM-121 (seribantumab) reverses heregulin-driven resistance to multiple chemotherapies on tumor cell growth. [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 LB-243. doi:10.1158/1538-7445.AM2015-LB-243


Cancer Research | 2013

Abstract 5464: In-vitro studies of MM-121/SAR 256212, an anti-ErbB-3 antibody, in combination with erlotinib in EGFR-wild-type NSCLC.

Marisa Wainszelbaum; Mark Sevecka; Olga Burenkova; Gabriela Garcia; William Kubasek; Gavin MacBeath

MM-121/SAR 256212 is a fully human anti ErbB3/HER3 antibody that blocks ligand-induced receptor activation. Formation of EGFR/ErbB3 (ErbB1-3) heterodimers has been implicated as a major driver of tumor growth and survival in non-small cell lung cancer (NSCLC). Although erlotinib remains the standard-of-care treatment for patients with EGFR-wild-type NSCLC who have failed platinum combination chemotherapy, clinical benefit is typically modest. To address this unmet medical need, we investigated the combination of erlotinib with MM-121 in pre-clinical models of NSCLC. We initially assembled a panel of 25 EGFR-WT NSCLC cell lines spanning the most common histological subtypes (adenocarcinoma, squamous and large-cell carcinoma). Clinical studies show that tumors harboring activating Ras mutations less commonly respond to ErbB-directed therapies. To explore the effect of Ras mutations on responsiveness to MM‐121, we also selected our cell lines to include a variety of H-/K-/N-Ras genotypes. Using a carefully optimized in-vitro 3D culture system, we then measured cell viability in response to MM-121, erlotinib or a combination of the drugs in the presence or absence of exogenously added epidermal growth factor (EGF) and/or heregulin-β1 (HRG). Our results indicate that MM-121 inhibits HRG-driven cell proliferation in the studied cell lines. In the five cell lines exhibiting dual-EGF-HRG-driven cell proliferation, the combination of MM-121 with erlotinib demonstrated superior inhibition of cell viability over erlotinib alone.To assess the ability of MM-121 to inhibit tumor cell growth independent of exogenously supplied HRG, we established mouse xenografts from three of the cell lines and treated mice with erlotinib, MM-121, or a combination of the two. All three in-vivo models showed greatly enhanced inhibition of tumor cell growth compared to erlotinib alone in a manner consistent with our in-vitro results. Analyzing our in vitro data we found that mutations in Ras genes (HRAS, KRAS, NRAS) do not preclude response to MM-121 or incremental benefit from the combination of MM-121 with erlotinib over erlotinib alone. Non-adenocarcinoma origin likewise did not preclude response to MM-121, although a weak trend towards diminished activity in the ten non-adenocarcinoma NSCLC cell lines was apparent. Merrimack Pharmaceuticals and Sanofi are co-developing MM-121 and a Phase 1-2 study of MM-121 in combination with erlotinib is currently enrolling patients with EGFR-wild-type NSCLC. Citation Format: Marisa J. Wainszelbaum, Mark S. Sevecka, Olga Burenkova, Gabriela Garcia, William Kubasek, Gavin MacBeath. In-vitro studies of MM-121/SAR 256212, an anti-ErbB-3 antibody, in combination with erlotinib in EGFR-wild-type NSCLC. [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 5464. doi:10.1158/1538-7445.AM2013-5464


Cancer Research | 2010

Abstract 3756: Prediction of xenograft response to MM-121, an anti-ErbB3 inhibitor, using computational modeling and measurements of five biomarkers

Matthew Onsum; Olga Burenkova; Aaron Fulgham; Lin Nie; Ashish Kalra; Dongmei Xiao; Viara P. Grantcharova; Sharlene Adams; Lia Luus; Violette Paragas; Raghida Bukhalid; Sharon Moulis; Lucia Wille; Gabriela Garcia; Victor Moyo; Birgit Schoeberl; Bill Kubasek; Ulrik Nielsen

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC One of the challenges faced by targeted therapeutics currently in the clinic is the relatively small population of patients who derive significant benefit from their use. We report the development of a preclinical classifier which can correctly predict xenograft response to MM-121, an anti-ErbB3 antibody, based on the measurement of a few key biomarkers in cell lysates. Deregulation of the ErbB family receptors is common in many cancers. Using a combination of computational modeling and quantitative experiments we identified ErbB3 as a key mediator of mitogenic signaling downstream of the ErbB receptors. Based on these results, we developed MM-121, a first in class anti-ErbB3 monoclonal antibody that blocks heregulin-induced signaling and inhibits tumor growth in multiple xenograft models of human cancer. Here we present our efforts to derive a predictive biomarker signature that identifies tumors that are responsive to MM-121. Using our computational model of the ErbB signaling pathway we identified the five most critical proteins for predicting activation of phospho-AKT - a key mediator of cell survival and apoptosis. These proteins include MM-121s target, ErbB3, and its ligand, heregulin. We profiled these biomarkers in a large panel of cancer cell lines, and using the measured effect of MM-121 on inhibiting tumor growth in eight xenograft tumor models, we determined a classification rule for predicting xenograft response. We subsequently used this classification rule to correctly predict a priori MM-121 response in 11 xenograft models. These results suggest that our computationally-derived biomarker signature is sufficient for predicting response to MM-121 in xenografts, and could offer significant clinical benefit by helping select patients for MM-121 treatment. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3756.

Collaboration


Dive into the Olga Burenkova's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ulrik Nielsen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabriela Garcia

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kristan Meetze

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Mark Manfredi

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge