5 results on '"Sonal Gera"'
Search Results
2. Data from Allosteric SHP2 Inhibitor, IACS-13909, Overcomes EGFR-Dependent and EGFR-Independent Resistance Mechanisms toward Osimertinib
- Author
-
Philip Jones, Nancy E. Kohl, Timothy P. Heffernan, Joseph R. Marszalek, Giulio F. Draetta, Andy M. Zuniga, Simon S. Yu, Christopher C. Williams, Erika Suzuki, Nakia D. Spencer, Sahil Seth, Vandhana Ramamoorthy, Michael Peoples, Robert A. Mullinax, Meredith A. Miller, Timothy McAfoos, Pijus K. Mandal, Xiaoyan Ma, Anastasia M. Lopez, Chiu-Yi Liu, Jeffrey J. Kovacs, Zhijun Kang, Yongying Jiang, Justin K. Huang, Virginia Giuliani, Sonal Gera, Guang Gao, Ningping Feng, Qing Chang, Christopher L. Carroll, Caroline C. Carrillo, Jason P. Burke, Christopher A. Bristow, Benjamin J. Bivona, Maria Emilia Di Francesco, Jason B. Cross, Connor A. Parker, Sarah Johnson, Qi Wu, Angela L. Harris, Faika Mseeh, Paul Leonard, Barbara Czako, Brooke A. Meyers, and Yuting Sun
- Abstract
Src homology 2 domain-containing phosphatase (SHP2) is a phosphatase that mediates signaling downstream of multiple receptor tyrosine kinases (RTK) and is required for full activation of the MAPK pathway. SHP2 inhibition has demonstrated tumor growth inhibition in RTK-activated cancers in preclinical studies. The long-term effectiveness of tyrosine kinase inhibitors such as the EGFR inhibitor (EGFRi), osimertinib, in non–small cell lung cancer (NSCLC) is limited by acquired resistance. Multiple clinically identified mechanisms underlie resistance to osimertinib, including mutations in EGFR that preclude drug binding as well as EGFR-independent activation of the MAPK pathway through alternate RTK (RTK-bypass). It has also been noted that frequently a tumor from a single patient harbors more than one resistance mechanism, and the plasticity between multiple resistance mechanisms could restrict the effectiveness of therapies targeting a single node of the oncogenic signaling network. Here, we report the discovery of IACS-13909, a specific and potent allosteric inhibitor of SHP2, that suppresses signaling through the MAPK pathway. IACS-13909 potently impeded proliferation of tumors harboring a broad spectrum of activated RTKs as the oncogenic driver. In EGFR-mutant osimertinib-resistant NSCLC models with EGFR-dependent and EGFR-independent resistance mechanisms, IACS-13909, administered as a single agent or in combination with osimertinib, potently suppressed tumor cell proliferation in vitro and caused tumor regression in vivo. Together, our findings provide preclinical evidence for using a SHP2 inhibitor as a therapeutic strategy in acquired EGFRi-resistant NSCLC.Significance:These findings highlight the discovery of IACS-13909 as a potent, selective inhibitor of SHP2 with drug-like properties, and targeting SHP2 may serve as a therapeutic strategy to overcome tumor resistance to osimertinib.
- Published
- 2023
- Full Text
- View/download PDF
3. Abstract 2338: The GLS1 inhibitor IPN60090 enhances antitumor immune response through metabolic reprogramming of T cells and impacts on the tumor microenvironment
- Author
-
Timothy P. Heffernan, Michael J. Soth, Mikhila Mahendra, Nakia D. Spencer, Timothy A. Yap, Sonal Gera, Jennifer R. Molina, Erika Suzuki, Angela L. Harris, Jeffrey J. Kovacs, Giulio Draetta, Ningping Feng, Kang Le, Christopher A. Bristow, and Philip Jones
- Subjects
Glutamine ,Cancer Research ,Tumor microenvironment ,Stromal cell ,Immune system ,Oncology ,Cancer research ,Context (language use) ,Pembrolizumab ,Biology ,CD8 ,Blockade - Abstract
Therapeutic agents targeting metabolism in the tumor microenvironment have been of increasing interest in recent years, however, the complexities of the interplay between tumor, stromal and immune cell interactions add complexity to these therapeutic approaches. We have previously disclosed the development of the GLS1 inhibitor, IPN60090, which is currently progressing through Phase 1 studies (NCT03894540) in multiple indication-specific biomarker-positive patient populations. In order to fully appreciate the opportunity to enhance IPN60090 activity in patients, we explored the impact of GLS1 inhibition on the activity of the immune system. Glutamine metabolism has been shown to play important and varied roles within the immune compartment including, but not limited to, roles in T-cell activation, T-cell effector functions and progression of exhaustion phenotypes. During T-cell activation, glutamine is utilized to drive activated T-cells towards a glycolytic phenotype. Eventually, activated T-cells exhaust and shift away from glycolysis towards fatty acid oxidation. Interestingly, it has been reported that the interaction of PD1 with PD-L1 blocks glutamine import and decreases glycolysis. Given these data, we hypothesized that inhibition of GLS1 with IPN60090 might enhance checkpoint blockade by increasing levels of glutamine in the tumor microenvironment, thus enhancing anti-tumor immune responses. In ex vivo culture systems, we show that IPN60090-mediated GLS1 inhibition increases the glycolytic activity of CD4+ and CD8+ T-cells, suggesting that GLS1 inhibition allows cells to maintain an energetically favorable phenotype. Furthermore, we show that IPN60090 enhances checkpoint blockade through cooperation with αPD1 therapy in two syngeneic mouse models which do not harbor predictive biomarkers of response to IPN60090 and which are refractory to checkpoint blockade. The observed synergy is due in part to IPN60090-dependent depletion of regulatory T-cells (Treg) and a concurrent increase in the CD8+ T-cell to Treg ratio in the tumor microenvironment. These data suggest that IPN60090 may show clinical benefit by enhancing immune response in the context of checkpoint blockade and have served as the justification for phase 1b trials in combination with Pembrolizumab which will enroll in 2021. Citation Format: Erika Suzuki, Jennifer Molina, Nakia D. Spencer, Christopher A. Bristow, Angela L. Harris, Ningping Feng, Mikhila Mahendra, Sonal Gera, Michael J. Soth, Kang Le, Timothy A. Yap, Giulio Draetta, Philip Jones, Timothy P. Heffernan, Jeffrey J. Kovacs. The GLS1 inhibitor IPN60090 enhances antitumor immune response through metabolic reprogramming of T cells and impacts on the tumor microenvironment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2338.
- Published
- 2021
- Full Text
- View/download PDF
4. Abstract 1655: Discovery and development of IACS-010759, a novel inhibitor of Complex I currently in phase I studies to exploit oxidative phosphorylation dependency in acute myeloid leukemia and solid tumors
- Author
-
Maria Emilia Di Francesco, Joseph R. Marszalek, Timothy McAfoos, Christopher L. Carroll, Zhijun Kang, Gang Liu, Jay P. Theroff, Jennifer P. Bardenhager, Madhavi L. Bandi, Jennifer R. Molina, Sonal Gera, Marina Protopopova, Yuting Sun, Mary K. Geck Do, Ningping Feng, Jason P. Gay, Florian Muller, Marina Konopleva, Funda Meric-Bernstam, Carlo Toniatti, Timothy P. Heffernan, Giulio F. Draetta, and Philip Jones
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Melanoma ,Myeloid leukemia ,Cancer ,Context (language use) ,Mitochondrion ,medicine.disease ,Lymphoma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Mechanism of action ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,medicine.symptom ,business ,Triple-negative breast cancer - Abstract
Over the past few years we and others reported that specific populations of tumor cells including AML, subsets of lymphoma, glioblastoma, triple negative breast cancer (TNBC), melanoma and pancreatic ductal adenocarcinoma (PDAC) are highly dependent upon oxidative phosphorylation (OXPHOS) to meet their energy and biomass needs. Inhibition of OXPHOS in the context of these dependent tumor populations represents therefore an exciting therapeutic opportunity. Through an extensive medicinal chemistry campaign we discovered IACS-010759, a potent, selective small molecule inhibitor of complex I of the mitochondria electron transport chain that possesses excellent pharmacokinetic (PK) and pharmacologic properties, making it suitable for clinical development. We advanced IACS-010759 through IND studies and have recently initiated Phase I studies in patients with relapsed/refractory acute myeloid leukemia (AML) and advanced solid tumors and lymphomas (NCT02882321 and NCT03291938). In this presentation we will describe the identification of a novel series of Complex I inhibitors and their optimization into the clinical candidate compound IACS-010759. Several challenges were successfully overcome, including the optimization of the pharmacokinetic profile and the identification of inhibitors with minimal activity shift across preclinical species, thus enabling a thorough evaluation of the efficacy and toxicology profile. Aspects of the extensive translational research conducted to elucidate the mechanism of action of IACS-010759 and to position it into the clinic will be discussed, including the compelling pharmacological response observed in multiple PDX models of primary AML, and PDX xenograft models of lymphoma, TNBC, glioblastoma, melanoma and PDAC. The observed response was associated with robust pharmacodynamic read-out as assessed by modulation of oxygen consumption rate (OCR), aspartate and specific transcriptional changes. The presentation will also cover the preclinical development activities which resulted in IACS-010759 advancing into on-going phase 1 evaluation in AML and solid tumors. Citation Format: Maria Emilia Di Francesco, Joseph R. Marszalek, Timothy McAfoos, Christopher L. Carroll, Zhijun Kang, Gang Liu, Jay P. Theroff, Jennifer P. Bardenhager, Madhavi L. Bandi, Jennifer R. Molina, Sonal Gera, Marina Protopopova, Yuting Sun, Mary K. Geck Do, Ningping Feng, Jason P. Gay, Florian Muller, Marina Konopleva, Funda Meric-Bernstam, Carlo Toniatti, Timothy P. Heffernan, Giulio F. Draetta, Philip Jones. Discovery and development of IACS-010759, a novel inhibitor of Complex I currently in phase I studies to exploit oxidative phosphorylation dependency in acute myeloid leukemia and solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1655.
- Published
- 2018
- Full Text
- View/download PDF
5. Abstract 335: Title: IACS-010759 is a novel clinical candidate that targets AML cells by inducing a metabolic catastrophe through inhibition of oxidative phosphorylation
- Author
-
Jennifer R. Molina, Marina Protopopova, Madhavi Bandi, Jennifer Bardenhagen, Christopher Bristow, Christopher Carroll, Edward Chang, Ningping Feng, Jason Gay, Mary Geck Do, Jennifer Greer, Sha Huang, Yongying Jiang, Marina Konopleva, Polina Matre, Jing Han, Zhijun Kang, Gang Liu, Timothy McAfoos, Pietro Morlacchi, Melinda Smith, Sonal Gera, Jay Theroff, Quanyun Xu, Juliana Velez, Carlo Toniatti, Timothy Heffernan, Giulio Draetta, M. Emilia Di Francesco, Philip Jones, and Joseph R. Marszalek
- Subjects
Cancer Research ,Cell growth ,Melanoma ,Oxidative phosphorylation ,Biology ,Bioinformatics ,medicine.disease ,Citric acid cycle ,Quinone binding ,Oncology ,Cell culture ,Apoptosis ,Cancer research ,medicine ,Glycolysis - Abstract
Tumor cells depend on both glycolysis and oxidative phosphorylation (OXPHOS) for energy and biomass production leading to robust cell proliferation. Recent data has demonstrated a dependence of various tumor types on mitochondrial OXPHOS, which represents an exciting therapeutic opportunity. Through an extensive medicinal chemistry campaign, IACS-10759 was identified as a potent, selective inhibitor of complex I of the electron transport chain, which is orally bioavailable and has excellent PK and physicochemical properties in preclinical species. Our group and others have demonstrated that a variety of tumor types including: AML, plus subsets of lymphoma, breast, melanoma and PDAC are highly dependent on OXPHOS to meet energy and biomass demands. Treatment of multiple cell lines and patient derived xenograft (PDX) models in multiple cancer types with IACS-10759 led to decreased oxygen consumption rate (OCR). IACS-10759 treatment also led to a robust decrease in cell viability and often an increase in apoptosis with EC50 values between 1 nM - 50 nM across multiple lines. Through a series of mechanistic studies we established that IACS-10759 blocks complex I of the electron transport at the quinone binding site. In an orthotopic xenograft model of primary AML cells derived from a patient who was refractory to standard of care and salvage therapies, 42 days of IACS-10759 treatment with 3 and 10 mg/kg orally using a 5 on/2 off schedule extended the median survival by greater than 2-fold. Efficacy was paralleled by robust modulation of OCR, aspartate, and p-AMPK levels. Additionally, tumor growth inhibition or regression was also observed in cell line and PDX xenograft models of lymphoma, triple negative breast, melanoma and PDAC treated with IACS-10759, indicating that subsets of several non-AML indications are also dependent on OXPHOS. Mechanistically, extensive metabolic profiling and flux analysis revealed that the response to IACS-10759 was associated with induction of a metabolic imbalance that negatively impacted energy homeostasis, amino acid biosynthesis, and NTP production due to reduced conversion of NADH to NAD+ by complex I, decreased ATP production, TCA cycle flux and nucleotide biosynthesis. As a result of the robust response in multiple cell lines, primary patient samples, and efficacy in PDX models, IACS-10759 has been advanced through IND enabling studies. GLP safety and toxicology have been completed, and we expect to file an IND at the end of 1Q2016 and initiate a Phase I clinical trial in AML during the second quarter of 2016. Citation Format: Jennifer R. Molina, Marina Protopopova, Madhavi Bandi, Jennifer Bardenhagen, Christopher Bristow, Christopher Carroll, Edward Chang, Ningping Feng, Jason Gay, Mary Geck Do, Jennifer Greer, Sha Huang, Yongying Jiang, Marina Konopleva, Polina Matre, Jing Han, Zhijun Kang, Gang Liu, Timothy McAfoos, Pietro Morlacchi, Melinda Smith, Sonal Gera, Jay Theroff, Quanyun Xu, Juliana Velez, Carlo Toniatti, Timothy Heffernan, Giulio Draetta, M. Emilia Di Francesco, Philip Jones, Joseph R. Marszalek. Title: IACS-010759 is a novel clinical candidate that targets AML cells by inducing a metabolic catastrophe through inhibition of oxidative phosphorylation. [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 335.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.