Network


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

Hotspot


Dive into the research topics where Gary L. Olson is active.

Publication


Featured researches published by Gary L. Olson.


Oncotarget | 2016

Discovery and clinical introduction of first-in-class imipridone ONC201

Joshua E. Allen; C. Leah B. Kline; Varun Vijay Prabhu; Jessica Wagner; Jo Ishizawa; Neel Madhukar; Avital Lev; Marie Baumeister; Lanlan Zhou; Amriti R. Lulla; Martin Stogniew; Lee Schalop; Cyril H. Benes; Howard L. Kaufman; Richard S. Pottorf; B. Rao Nallaganchu; Gary L. Olson; Fahd Al-Mulla; Madeleine Duvic; Gen Sheng Wu; David T. Dicker; Mala Kiran Talekar; Bora Lim; Olivier Elemento; Wolfgang Oster; Joseph R. Bertino; Keith T. Flaherty; Michael L. Wang; Gautam Borthakur; Michael Andreeff

ONC201 is the founding member of a novel class of anti-cancer compounds called imipridones that is currently in Phase II clinical trials in multiple advanced cancers. Since the discovery of ONC201 as a p53-independent inducer of TRAIL gene transcription, preclinical studies have determined that ONC201 has anti-proliferative and pro-apoptotic effects against a broad range of tumor cells but not normal cells. The mechanism of action of ONC201 involves engagement of PERK-independent activation of the integrated stress response, leading to tumor upregulation of DR5 and dual Akt/ERK inactivation, and consequent Foxo3a activation leading to upregulation of the death ligand TRAIL. ONC201 is orally active with infrequent dosing in animals models, causes sustained pharmacodynamic effects, and is not genotoxic. The first-in-human clinical trial of ONC201 in advanced aggressive refractory solid tumors confirmed that ONC201 is exceptionally well-tolerated and established the recommended phase II dose of 625 mg administered orally every three weeks defined by drug exposure comparable to efficacious levels in preclinical models. Clinical trials are evaluating the single agent efficacy of ONC201 in multiple solid tumors and hematological malignancies and exploring alternative dosing regimens. In addition, chemical analogs that have shown promise in other oncology indications are in pre-clinical development. In summary, the imipridone family that comprises ONC201 and its chemical analogs represent a new class of anti-cancer therapy with a unique mechanism of action being translated in ongoing clinical trials.


Cell Cycle | 2017

Preclinical evaluation of the imipridone family, analogues of clinical stage anti-cancer small molecule ONC201, reveals potent anti-cancer effects of ONC212

Jessica Wagner; Christina Leah B. Kline; Marie D. Ralff; Avital Lev; Amriti R. Lulla; Lanlan Zhou; Gary L. Olson; Bhaskara Rao Nallaganchu; Cyril H. Benes; Joshua E. Allen; Varun Vijay Prabhu; Martin Stogniew; Wolfgang Oster; Wafik S. El-Deiry

ABSTRACT Anti-cancer small molecule ONC201 upregulates the integrated stress response (ISR) and acts as a dual inactivator of Akt/ERK, leading to TRAIL gene activation. ONC201 is under investigation in multiple clinical trials to treat patients with cancer. Given the unique imipridone core chemical structure of ONC201, we synthesized a series of analogs to identify additional compounds with distinct therapeutic properties. Several imipridones with a broad range of in vitro potencies were identified in an exploration of chemical derivatives. Based on in vitro potency in human cancer cell lines and lack of toxicity to normal human fibroblasts, imipridones ONC206 and ONC212 were prioritized for further study. Both analogs inhibited colony formation, and induced apoptosis and downstream signaling that involves the integrated stress response and Akt/ERK, similar to ONC201. Compared to ONC201, ONC206 demonstrated improved inhibition of cell migration while ONC212 exhibited rapid kinetics of activity. ONC212 was further tested in >1000 human cancer cell lines in vitro and evaluated for safety and anti-tumor efficacy in vivo. ONC212 exhibited broad-spectrum efficacy at nanomolar concentrations across solid tumors and hematological malignancies. Skin cancer emerged as a tumor type with improved efficacy relative to ONC201. Orally administered ONC212 displayed potent anti-tumor effects in vivo, a broad therapeutic window and a favorable PK profile. ONC212 was efficacious in vivo in BRAF V600E melanoma models that are less sensitive to ONC201. Based on these findings, ONC212 warrants further development as a drug candidate. It is clear that therapeutic utility extends beyond ONC201 to include additional imipridones.


Cancer Research | 2017

Abstract 3245: Preclinical evaluation of the imipridone family of small molecules, including analogues of clinical-stage anti-cancer small molecule ONC201, reveals potent anti-cancer effects of ONC212

Jessica Wagner; C. Leah B. Kline; Gary L. Olson; Bhaskara Rao Nallaganchu; Richard S. Pottorf; Varun Vijay Prabhu; Martin Stogniew; Joshua E. Allen; Wafik S. El-Deiry

We previously identified a novel, potent anti-cancer small molecule ONC201, which upregulates the integrated stress response (ISR) through ATF4/CHOP/DR5 and acts as a dual inactivator of Akt and ERK, leading to TRAIL gene activation. After completing a first-in-human phase I clinical trial that revealed exceptional safety, therapeutic pharmacokinetic (PK) profile and tumor engagement, ONC201 is under investigation in several advanced cancer Phase I/II trials. Given the unique imipridone core chemical structure of ONC201, we synthesized a family of analogues in an effort to identify additional chemical family members with distinct therapeutic properties. Based on in vitro potency improvements in human cancer cell lines and therapeutic window approximations with normal human fibroblasts, select analogues were investigated in animals for toxicity, maximum tolerated dose (MTD), and antitumor efficacy. ONC212 is one of the most promising new imipridones that was further evaluated to establish the PK profile, oral bioavailability, and efficacy in tumor types that are less sensitive to ONC201. Compared to ONC201, we noted distinct and more rapid kinetics of activity as well as improved potency in multiple human cancer cell lines in vitro. ONC212 has a broad therapeutic window, an acceptable PK profile, and is orally well-tolerated in mice. With no evidence of toxicity at efficacious doses in both colon and triple negative breast cancer, we have begun further evaluation of antitumor efficacy studies in ONC201-resistant tumor types. Efficacy studies with ONC212 are ongoing in melanoma models that are sensitive to ONC212 but less sensitive to ONC201 in vitro. Preliminary data indicates potent tumor growth reduction by ONC212 in vivo in ONC201-resistant melanoma xenografts. With a wide safety margin, potent antitumor activity in ONC201-insenstive tumors, and drug-like characteristics, ONC212 is being further developed as a drug candidate from the new imipridone class of compounds that complements the spectrum of activity of ONC201. Citation Format: Jessica Wagner, C. Leah Kline, Gary Olson, Bhaskara Nallaganchu, Richard Pottorf, Varun Prabhu, Martin Stogniew, Joshua Allen, Wafik El-Deiry. Preclinical evaluation of the imipridone family of small molecules, including analogues of clinical-stage anti-cancer small molecule ONC201, reveals potent anti-cancer effects of ONC212 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3245. doi:10.1158/1538-7445.AM2017-3245


Cancer Research | 2016

Abstract 349: Structure-activity relationships and mechanistic analysis of analogues of the clinical-stage anti-cancer small molecule ONC201

Jessica Wagner; Gary L. Olson; Nallaganchu Rao Bhaskara; Richard S. Pottorf; Garnett J. Mathew; Cyril H. Benes; Rohinton Tarapore; Martin Stogniew; Lee Schalop; Wolfgang Oster; Josh E. Allen; Wafik S. El-Deiry

TRAIL is an endogenous protein that initiates apoptosis selectively in cancer without toxic side effects, prompting interest in therapeutic modulation. We previously screened for small molecules that could upregulate the endogenous TRAIL gene to trigger apoptosis and restore anti-tumor immunity within tumor cells in a p53-independent manner. We showed that ONC201 (previously referred to as TIC10) is a dual inactivator of Akt and ERK, leading to nuclear translocation of Foxo3a and TRAIL gene activation (Allen et al., Science Translational Medicine, 2013). We recently found that ONC201 causes an early-stage upregulation of the integrated stress response through ATF4/CHOP/DR5 (Kline et al., Science Signaling, in press) and can inhibit cancer stem cell self-renewal (Prabhu et al., Cancer Research, 2015). ONC201 recently completed its first-in-man phase I clinical trial and several other trials in select advanced cancers are ongoing (NCT02250781, NCT02324621, NCT02420795, NCT02392572, NCT02609230, NCT02525692, NCT02038699). Leveraging the unique pharmacophore of ONC201, we synthesized ONC201 analogues in search for compounds with distinct therapeutic properties. After establishing the importance of the pyrido[3,4-e]pyrimidinone core structure of ONC201 in its anti-tumor activity (Wagner et al., Oncotarget, 2014), we performed detailed structure activity relationship (SAR) studies. We evaluated several ONC201 analogues with differing N-substituents around the core structure. Certain ONC201 analogues exhibit more rapid kinetics of activity and lowered IC50 values in some human cancer cell lines in vitro. Interim results of ONC212 sensitivity profiling in >100 genetically annotated cell lines from the Genomic of Drug Sensitivity in Cancer collection have corroborated this improvement in potency. One analogue, ONC212, has demonstrated compelling efficacy against several tumor types in vivo with no evidence of toxicity at therapeutic doses. Furthermore, in vitro mechanism studies have demonstrated overlap between ONC201- and ONC212-mediated signaling in tumor cells that includes activation of the integrated stress response. With a wide safety margin, distinct pharmacokinetics (PK), and robust potency, ONC212 is being developed as the next drug candidate from the new class of compounds defined by the novel pharmacophore of ONC201 in indications that complement the parent compound9s use spectrum. Citation Format: Jessica Wagner, Gary Olson, Nallaganchu Rao Bhaskara, Richard S. Pottorf, Garnett J. Mathew, Cyril H. Benes, Rohinton Tarapore, Martin Stogniew, Lee Schalop, Wolfgang Oster, Josh E. Allen, Wafik S. El-Deiry. Structure-activity relationships and mechanistic analysis of analogues of the clinical-stage anti-cancer small molecule ONC201. [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 349.


Cancer Research | 2016

Abstract 4826: ONC212 exhibits increased cytotoxicity relative to ONC201 in a subset of human pancreatic cancer cell lines

Avital Lev; Jessica Wagner; David T. Dicker; Martin Stogniew; Joshua E. Allen; Lee Schalop; Wolfgang Oster; Gary L. Olson; Wafik S. El-Deiry

Pancreatic cancer is one of the most deadly types of cancer with five-year survival rate of ∼6%. This poor prognosis is attributed to aggressive growth of the tumor cells, metastasis at an early stage and resistance to standard chemotherapy. Therefore, finding new treatments and improving outcomes of pancreatic cancer patients have been high priorities. ONC201 is a small molecule that selectively targets a broad range of tumor types tested. It is currently in phase I/II clinical trials for advanced solid, hematopoietic, and lymphoid tumors. The ONC201 analogue, ONC212, is in preclinical development for certain cancers that are not lead indications for the parent compound ONC201. We examined the efficacy of ONC201 and ONC212 in a panel of human pancreatic cancer cell lines. Among seven cell lines tested, three cell lines were relatively insensitive to ONC201, with IC50 values of 100 iM or greater. However, these cell lines showed greater sensitivity to ONC212 with IC50 values of 31-56 iM. We compared ONC201-resistant PANC-1 cells and ONC201-sensitive HPAF-II cells. Both ONC201 and ONC212 induced apoptosis in HPAF-II cells, as assessed by PARP cleavage, while PARP cleavage was detected only when PANC-1 cells were treated with ONC212 but not ONC201. We are currently evaluating the efficacy of ONC201 and ONC212 in a panel of patient-derived low passage pancreatic cancer cell lines. We found a correlation between high expression levels of RTKs (c-MET, ALK, EGFR, IGFR, HER2) and decreased sensitivity to ONC201 in pancreatic cancer cell lines. We investigated the effect of ONC201 on EGFR and IGFR, which are highly expressed in PANC-1 and found that ONC201 does not inhibit receptor activation by the specific growth factors. We are studying the effects of ONC201 and ONC212 on RTKs and their signal transduction pathways. Lastly, since aggressive drug-resistant pancreatic cancer cells express high levels of RTKs, we are developing a rationale combining ONC201/ONC212 with specific small molecule RTK inhibitors such as crizotinib or lapatinib. We are exploring these combination treatments using cell lines, organoids and PDX models to unravel specific therapy options for therapy-refractory pancreatic cancer cells. Our goal is to provide the preclinical rationale for clinical trials of ONC201 or ONC212 as mono-agents or in combination with other therapies for pancreatic cancer patients. Citation Format: Avital Lev, Jessica Wagner, David T. Dicker, Martin Stogniew, Joshua E. Allen, Lee Schalop, Wolfgang Oster, Gary L. Olson, Wafik S. El-Deiry. ONC212 exhibits increased cytotoxicity relative to ONC201 in a subset of human pancreatic cancer 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 4826.


Cancer Research | 2015

Abstract 4499: Cytotoxicity, biochemical activity, and structural analysis of ONC201 and comparisons to a biologically inactive isomer

Jessica Wagner; Christina Leah B. Kline; Richard S. Pottorf; Bhaskara Rao Nallaganchu; Gary L. Olson; David T. Dicker; Joshua E. Allen; Wafik S. El-Deiry

We previously identified TRAIL-inducing compound 10 (TIC10), also known as NSC-350625, as a small molecule being developed under the name ONC201 that has potent anti-tumor efficacy and a benign safety profile in preclinical cancer models. Further investigation of the preclinical profile of this drug candidate led in early 2014 to the FDA acceptance of the investigational drug application (IND) for oral ONC201 to treat patients with advanced cancer. The initially disclosed chemical structure of ONC201 provided by Stahle et. al in an expired patent was described as an imidazo[1,2-a]pyrido[4,3-d]pyrimidine derivative. Our first report of its anticancer activity by Allen et. al. included mass spectrometry and 1H NMR that indicated both were consistent with the structure depicted by Stahle et. al and the National Cancer Institute. A recent publication reported that the structure of ONC201 differs from the initial description and is in fact an angular [3,4-e] isomer of the previously depicted structure. Here, we report X-ray crystallography and other structural studies of ONC201 produced by Oncoceutics in a dihydrochloride salt form for clinical use that confirm the angular [3,4-e] structure and indicate that the material is not a mixture of the two isomers. Furthermore, we confirm that although fragmentation by mass spectrometry for the isomers is identical, the angular [3,4-e] isomer can be definitively identified as ONC201 by implementing specific various spectroscopy techniques to identify differences between ONC201 and the linear isomer. In accordance with our structural analysis, in vitro activity assays in cancer cells indicate that the previously disclosed anti-cancer activity and mechanism of action are indeed associated exclusively with the [3,4-e] structure and not the [4,3-d] linear isomer. Together these studies confirm the angular [3,4-e] structure of ONC201 as the highly active and pure anti-cancer drug candidate that was utilized in prior preclinical pharmacology studies and is now entering clinical trials in several oncology indications. Citation Format: Jessica Wagner, Christina Leah Kline, Richard S. Pottorf, Bhaskara Rao Nallaganchu, Gary L. Olson, David T. Dicker, Joshua E. Allen, Wafik S. El-Deiry. Cytotoxicity, biochemical activity, and structural analysis of ONC201 and comparisons to a biologically inactive isomer. [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 4499. doi:10.1158/1538-7445.AM2015-4499


Oncotarget | 2014

The angular structure of ONC201, a TRAIL pathway-inducing compound, determines its potent anti-cancer activity

Jessica Wagner; Christina Leah B. Kline; Richard S. Pottorf; Bhaskara Rao Nallaganchu; Gary L. Olson; David T. Dicker; Joshua E. Allen; Wafik S. El-Deiry


Archive | 2010

Arginase inhibitors and methods of use

David W. Christianson; Bruce Edward Tomczuk; Richard S. Pottorf; Andrew Vargha Colasanti; Gary L. Olson


Archive | 2010

PROSTAGLANDIN TRANSPORTER INHIBITORS AND USES THEREOF

Victor L. Schuster; Yuling Chi; Andrew S. Wasmuth; Richard S. Pottorf; Gary L. Olson


Archive | 2011

Arginase inhibitors and methods of use thereof

Bruce Edward Tomczuk; Gary L. Olson; Richard S. Pottorf; Jane Wang; Bhaskara Rao Nallaganchu

Collaboration


Dive into the Gary L. Olson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joshua E. Allen

Penn State Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lee Schalop

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge