Network


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

Hotspot


Dive into the research topics where Katrina Chan is active.

Publication


Featured researches published by Katrina Chan.


Molecular Cancer Therapeutics | 2011

Abstract A171: A potent and selective PI3K inhibitor, INK1117, targets human cancers harboring oncogenic PIK3CA mutations.

Katti Jessen; Linda Kessler; Jeff Kucharski; Xin Guo; Jocelyn Staunton; Matthew R. Janes; Marikka Elia; Urmi Banerjee; Lucy Lan; Shunyou Wang; Josh Stewart; Angelina Luzader; Levan Darjania; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Christian Rommel; Yi Liu

Background: The PI3K pathway is one of the most frequently dysregulated pathways in human cancer. Activating mutations in the PIK3CA gene, encoding the p110 catalytic subunit of PI3K, have been identified as a major mechanism of inducing oncogenic PI3K signaling. The high frequency of PIK3CA mutations suggests that PI3K inhibitors may have therapeutic utility in genetically defined tumor populations. The discovery of isoform-selective PI3K inhibitors has proven to be difficult due to the highly homologous nature of all PI3K isoforms and structurally related kinases. PI3Kα-selective inhibitors may permit more potent inhibition of PI3Kα while minimizing side effects and permitting more alternatives for combination therapy relative to non-selective class I PI3K (pan-PI3K) pathway inhibitors, many of which have entered clinical development. Results: Through structure-guided drug design we have discovered INK1117, a novel, potent and selective PI3K inhibitor with good oral bioavailability. INK1117 potently inhibits PI3K and demonstrates a greater than 100-fold selectivity relative to other class I PI3K family members and mTOR as well as a high degree of selectivity against a large panel of protein kinases. INK1117 blocks proliferation of tumor cell lines bearing PIK3CA mutations, and inhibits cellular phosphorylation and activity of AKT. However, INK1117 shows much less activity in PTEN-deficient tumor cells, which typically display constitutive PI3K pathway activation independent of PI3Kα. Daily, oral administration of INK1117 potently inhibits tumor growth in xenograft models bearing PIK3CA oncogenic mutations and, comparable to in vitro studies, INK1117 was not efficacious in tumor models with PTEN and/or KRAS mutations. In contrast to pan-PI3K inhibitors, INK1117 does not induce significant glucose elevation in glucose unchallenged or challenged rodents while levels of circulating Insulin is mildly elevated. The absence of hyperglycemia but presence of moderate hyperinsulinemia in mice treated with INK1117 is in line with results obtained using genetically engineered PI3K deficient mice. Additionally, INK1117 does not significantly impair B and T cell function in vitro and in vivo. INK1117 in combination with targeted agents such as lapatinib or trastuzumab, displayed enhanced activity both in vitro and in vivo. Our preclinical data show that PI3K isoform-selective kinase inhibitors as a single agent can provide comparable or superior efficacy than panPI3K inhibitors in tumors with PIK3CA mutation with better tolerability. The combination of PI3K inhibitors with other oncogene-targeted drugs revealed further improved anti-tumor efficacy. Conclusion: Selective targeting of PI3K combined with companion diagnostic presents a novel therapeutic strategy for patients with PIK3CA mutated cancer types. INK1117 is a potent and orally efficacious PI3Kα-selective kinase inhibitor currently in phase 1 clinical development. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A171.


Molecular Cancer Therapeutics | 2009

Abstract B148: INK128 is a potent and selective TORC1/2 inhibitor with broad oral antitumor activity

Katayoun Jessen; Shunyou Wang; Linda Kessler; Xin Guo; Jeff Kucharski; Jocelyn Staunton; Lucy Lan; Marikka Elia; Josh Stewart; Jolene Brown; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Christian Rommel; Yi Liu

mTOR Ser/Thr protein kinase operates in two distinct multi‐protein complexes, TORC1 and TORC2, which together regulate growth, metabolism, angiogenesis and survival by integrating nutrient and hormonal environmental signals. The activity of mTOR is frequently up‐regulated in human cancer by constitutive mitogen stimuli or oncogenic mutations upstream of TORC1 and TORC2. Rapamycin provides mechanistic rationale and clinical proof of concept for the therapeutic value of targeting mTOR in human cancer, but it also provided insights into how ATP‐competitive TORC1/2 inhibitors have the potential to demonstrate superior efficacy. Through rational drug design we have discovered INK128, a potent, selective TORC1/2 inhibitor with excellent drug‐like properties. INK128 inhibits mTOR kinase (sub‐nanomolar) in an ATP‐dependent fashion and demonstrates a high degree of selectivity against closely related kinases as well as against a panel of more than 400 kinases. INK128 inhibits both the phosphorylation of S6 and 4EBP1, the downstream substrates of TORC1, and selectively inhibits AKT phosphorylation at Ser473, the downstream substrate of TORC2, in vitro and in vivo. Interestingly, potent inhibition was also observed in cell lines resistant to rapamycin and pan‐PI3K inhibitors. Daily, oral administration of INK128 (alone or in combination) inhibited angiogenesis and tumor growth in multiple xenograft models with predicted dose:exposure PK/PD relationship. We attribute the superior activity of INK128 to the fact that it is a more effective inhibitor of TORC1 relative to rapamycin. We further demonstrated that TORC1/2 inhibitors cause death of murine and human leukemia cells in models of pre‐B acute lymphoblastic leukemia. In vivo, oral daily treatment with TORC1/2 inhibitors delayed leukemia onset and augmented the effects of ABL kinase inhibitors. Unexpectedly, these novel TORC1/2 inhibitors had much weaker effects than rapamycin on proliferation and function of normal lymphocytes. These findings establish that transformed lymphocytes are selectively sensitive to active‐site TORC1/2 inhibitors and further support the development of such compounds for leukemia therapy in addition to solid tumors. In summary, INK128 is a potent, selective, and orally active TORC1/2 dual inhibitor positioned to enter clinical development. TORC1/2 inhibitors are mechanistically distinct from rapamycin and offer a compelling approach to the treatment of cancer by targeting translational control, cell metabolism, growth and angiogenesis. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B148.


Cancer Research | 2012

Abstract 3753: Potent anti-tumor activity of mTOR kinase inhibitor in combination with anti-angiogenic agents in preclinical models of renal cell cancer

Shunyou Wang; Lucy Lan; Mathew Janes; Katti Jessen; Linda Kessler; Jeff Kucharski; Xin Xin Guo; Jocelyn Staunton; Marikka Elia; Urmi Banerjee; Josh Stewart; Levan Darjania; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Christian Rommel; Yi Liu

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, ILnnBackground: Multiple agents that inhibit angiogenesis and tumor cell growth via the VEGF and mTOR (TORC1) pathways have been approved for locally advanced or metastatic renal cell carcinoma (RCC) in recent years. Despite this progress, improving overall survival remains a significant clinical challenge for the treatment of this cancer type. The tumorigenesis of certain types of RCC is dominated by genes that play critical role in hypoxia response, including stimulation of neo-angiogenesis. Anti-angiogenic therapies target the tumor microenvironment but provide little direct impact to the tumor cell, allowing the potential for disease progression despite treatment. Approval of allosteric partial TORC1 inhibitors in RCC stimulated interest in pursuing TORC1/2 inhibitors, that block mTOR signaling more potently and comprehensively, alone and in combination with anti-angiogenic agents in preclinical models of RCC. Results: INK128 is an orally bioavailable, potent and selective ATP site mTOR kinase inhibitor (TORC1/2) that has entered clinical development. INK128 inhibited the phosphorylation of S6 and 4EBP1, downstream substrates of TORC1, and AKT and NRDG1, downstream substrate of TORC2 in RCC cell lines. TORC1/2 pathway inhibition correlated with potent and complete blockade of cell proliferation. In vivo, daily treatment of INK128 as single agent suppressed tumor growth in multiple mouse RCC xenograft models. Pharmacodynamic analysis demonstrated that INK128 inhibits both TORC1 and TORC2 pathways in vivo. INK128 did not alter the tumor-associated microvasculature density despite significant decrease of VEGF supply by lowering the capacity of protein translation of the tumor cells. This prompted us to explore the potential of the combination of INK128 with anti-angiogenic agents to maximize anti-tumor activity by targeting both tumor microenvironment and tumor cells directly. Co-treatment of INK128 with sorafenib resulted in much enhanced activity in RCC tumor models but did not achieve sufficient tolerability. We next employed an alternating dosing strategy of INK128 and sorafenib to improve tolerability. An optimized dose/schedule sequence of both agents caused sustained inhibition of tumor-associated angiogenesis, suppression of mTOR pathway resulting into marked blockade of tumor growth with significant induction of apoptosis and acceptable tolerability. Conclusion: The sequential, yet potent blockade of both mTOR and VEGF pathways obtained by combining INK128 with sorafenib acts synergistically in inducing cell death and tumor regression in preclinical renal cancer models. Adaptive combination of receptor tyrosine kinase-based anti-angiogenic agents with mTOR kinase inhibitors currently in development may offer a safe and effective therapeutic strategy for renal cancer patients who fail to respond to standard of care therapy.nnCitation 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 3753. doi:1538-7445.AM2012-3753


Cancer Research | 2011

Abstract 4501: INK1117: A potent and orally efficacious PI3Kα-selective inhibitor for the treatment of cancer

Katayoun Jessen; Linda Kessler; Jeff Kucharski; Xin Guo; Jocelyn Staunton; Marikka Elia; Matthew R. Janes; Lucy Lan; Shunyou Wang; Josh Stewart; Levan Darjania; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; David A. Fruman; Christian Rommel; Yi Liu

Objective: Discovery, characterization and preclinical evaluation of PI3Kalpha (PI3Kα) selective small molecule kinase inhibitors for the treatment of solid tumors. Background: The PI3K pathway is one of the most frequently dysregulated pathways in human cancer. Activating mutations in the PIK3CA gene, encoding the p110 catalytic subunit of PI3Kα, have been identified as a major mechanism of inducing oncogenic PI3K signaling. The high frequency of PIK3CA mutations suggests that PI3Kα inhibitors may have therapeutic utility in genetically defined tumor populations. The discovery of isoform-selective PI3Kα inhibitors has proven to be difficult due to the highly homologous nature of all PI3K isoforms and structurally related kinases. Multiple non-selective class I PI3K (pan-PI3K) inhibitors have entered clinical development. PI3Kα-selective inhibitors may permit more potent inhibition of PI3Kα while minimizing side effects and permitting more alternatives for combination therapy relative to pan-PI3K pathway inhibitors. Results: Through structure-guided drug design we have discovered INK1117, a novel, potent and selective PI3Kα inhibitor with good oral bioavailability. INK1117 potently inhibits PI3Kα and demonstrates a greater than 100-fold selectivity relative to other class I PI3K family members and mTOR as well as a high degree of selectivity against a large panel of protein kinases. INK1117 blocks proliferation of tumor cell lines bearing PIK3CA mutations, and inhibits cellular phosphorylation and activity of AKT. However, INK1117 shows much less activity in PTEN-deficient tumor cells, which typically display constitutive PI3K pathway activation independent of PI3Kα. INK1117 blocks VEGF signaling and angiogenesis in vitro and in vivo. Daily, oral administration of INK1117 potently inhibits tumor growth in xenograft models bearing PIK3CA oncogenic mutations and, comparable to in vitro studies, INK1117 was not efficacious in tumor models with PTEN and/or KRAS mutations. In contrast to pan-PI3K inhibitors, INK1117 does not appear to impair glucose homeostasis or insulin response in glucose/insulin tolerance tests in rodents. Additionally, INK1117 does not significantly impair B and T cell function in vitro and in vivo. These two factors support the hypothesis that INK1117 may be better tolerated than pan-PI3K inhibitors, particularly when used in combination with other agents. Conclusion: INK1117 is a potent and orally efficacious PI3Kα isoform-selective kinase inhibitor with excellent drug-like properties. Selectively targeting PI3Kα offers a new therapeutic approach for the treatment of cancers with PIK3CA activation and preclinical data suggest that isoform-selective inhibitors may provide equivalent efficacy to pan-PI3K inhibitors in selected tumors with a more favorable safety profile. 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 4501. doi:10.1158/1538-7445.AM2011-4501


Cancer Research | 2012

Abstract 2745: INK128, a TORC1/2 kinase inhibitor, enhances the efficacy of cytotoxic therapies in endometrial tumor models

Linda Kessler; Katti Jessen; Matthew R. Janes; Jocelyn Staunton; Jeff Kucharski; Xin Guo; Marikka Elia; Urmi Banerjee; Lucy Lan; Shunyou Wang; Josh Stewart; Angelina Luzader; Levan Darjania; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Christian Rommel; Yi Liu

Background: The mammalian target of rapamycin (mTOR), which comprises two protein complexes, TORC1 and TORC2, regulates tumor cell growth, metabolism and motility. In endometrial cancer, the mTOR pathway is aberrantly activated by constitutive mitogen stimuli, such as FGFR2 and multiple genetic mutations such as PIK3CA, PIK3R1/2, PTEN and LKB1. The activation of multiple signaling pathways, cross-feedback and redundancy of these pathways led us to explore the combination of mTOR kinase inhibitor with chemotherapeutic agents for achieving maximal efficacy in preclinical mouse models of endometrial cancer. Results: INK128 is a potent and selective TORC1/2 inhibitor with excellent drug-like properties currently advancing in clinical development. INK128 inhibits endometrial tumor cell proliferation in vitro and displays tumor growth inhibition in endometrial tumor models. In most instances, the anti-tumor activity of INK128 is cytostatic compared to the cytotoxic effect of taxol and carboplatin. In endometrial tumor cells of diverse genetic backgrounds, combining INK128 with taxol resulted in a synergistic inhibition of tumor cell proliferation, sustained suppression of PI3K/AKT/mTOR pathway activity, skewed cell cycle and an increase in apoptosis. INK128 decreased the expression of anti-apoptotic protein MCL-1 and therefore mechanistically enhanced taxol-triggered apoptosis. In endometrial mouse tumor models with PTEN and FGFR2 mutations, the combination induced marked tumor regression and significantly delayed tumor re-growth upon discontinuation of the treatment compared to taxol treatment alone. The combination therapy was well-tolerated and the enhanced anti-tumor efficacy correlated with mechanism-based inhibition of several TORC1 and TORC2 pharmacodynamic markers and induction of apoptosis in vivo. Additionally, combining INK128 with taxol or carbotaxol resulted in tumor regression in taxol-resistant endometrial tumor models. The opportunity of optimizing dose and schedule combination regimens as well as biomarkers predicting sensitivity/resistance to INK128/taxol combination will be discussed. Conclusion: The combination of mTOR kinase inhibitors with chemotherapeutic agents is a compelling strategy to improve therapeutic outcome and to overcome chemo-resistance for tumor types with aberrant activation of the PI3K/mTOR pathway such as endometrial cancer. 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 2745. doi:1538-7445.AM2012-2745


Molecular Cancer Therapeutics | 2011

Abstract A172: INK128, an orally active TORC1/2 kinase inhibitor, displays enhanced efficacy when combined with cytotoxic agents.

Linda Kessler; Katti Jessen; Matthew R. Janes; Jocelyn Staunton; Jeff Kucharski; Xin Guo; Marikka Elia; Urmi Banerjee; Lucy Lan; Shunyou Wang; Josh Stewart; Angelina Luzader; Levan Darjania; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Christian Rommel; Yi Liu

Background: The mammalian target of rapamycin (mTOR) comprises two protein complexes, TORC1 and TORC2, which together regulate cell growth, metabolism, angiogenesis, and cell survival. Because TORC1 and TORC2 play a crucial role in several pathways that are frequently dysregulated in human cancer, the TORC1/2 kinase inhibitors provide a promising class of anti-cancer agents. Activation of multiple genomic and signaling pathways, cross-feedback, pathway redundancy and the capacity for compensatory adaptive response in cancer suggest that the optimal therapeutic effect of mTOR inhibitors will be better exploited by the strategy of rational combination therapies. Results: INK128 is a potent and selective TORC1/2 inhibitor with excellent drug-like properties currently in multiple phase 1 studies. Daily, oral administration of INK128 inhibits tumor growth in multiple xenograft models with predicted pharmacokinetic/pharmacodynamic relationship. In most instances the anti-tumor effect of INK128 is cytostatic compared to the cytotoxic effect of chemotherapeutic agents such as Taxol. To enhance the anti-tumor activity of INK128, we investigated the combination of INK128 with cytotoxic agents such as conventional chemo drugs or apoptosis inducing agents. Treatment of tumors with the cytotoxic agents such as Taxol often leads to activation of the PI3K/AKT/mTOR pathway, which limits the anti-tumor activity of Taxol and may eventually lead to drug resistance. Additionally, Taxol resistance mechanisms include the over-expression of members of the Bcl-2 family of proteins such as MCL-1 and BCL-X L , whose expression is regulated by TORC1 and their function possibly by TORC2. Therefore, there is a strong scientific rationale to investigate the combination of a TORC1/2 inhibitor and Taxol. To this end, we have investigated the preclinical efficacy of INK128 in combination with Taxol in vitro and in vivo. Combining INK128 with Taxol resulted in a synergistic inhibition of tumor cell proliferation and the suppression of PI3K/AKT/mTOR signaling pathways in various cancer types. In most instances alterations in cell cycle and an increase in apoptosis was observed. INK128 decreases the expression of anti-apoptotic protein Mcl-1 and therefore resensitizes Taxol-resistant tumor cells to Mcl-1-dependent build up. In various xenograft models, the combination therapies were well-tolerated and displayed enhanced anti-tumor efficacy, inhibited pharmacodynamic markers, and induced apoptosis. Combination of INK128 with a small molecule inhibitor of Bcl-2 (ABT-263) also led to significant increase of apoptosis in tumor cells but not in normal cells in vitro and enhanced anti-tumor activity in mouse tumor models in vivo. Conclusion: In summary, our data show a potential benefit for combining INK128 with cytotoxic agents. INK128 is currently in a Taxol combination phase Ib study in patients with advanced solid malignancies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A172.


Cancer Research | 2011

Abstract 4486: INK128, a novel TORC1/2 inhibitor with potent oral antitumor activity in preclinical models of renal cancer

Shunyou Wang; Katti Jessen; Linda Kessler; Jeff Kucharski; Lucy Lan; Xin Guo; Jocelyn Staunton; Marikka Elia; Josh Stewart; Levan Darjania; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Christian Rommel; Yi Liu

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FLnnObjective: Preclinical characterization and evaluation of INK128 alone and in combination with anti-angiogenic agents in models of renal cell carcinoma.nnBackground: mTOR kinase comprises two distinct multi-protein complexes, TORC1 and TORC2, which together regulate processes critical for cell growth and survival. TORC1 regulates protein translation of cell cycle regulators, angiogenic factors, and factors that control cell motility. TORC2 regulates pathways involved in cell metabolism, survival and motility. Through rational drug design we have identified INK128, a potent and selective small molecule, active-site kinase inhibitor of mTOR with excellent drug-like properties. Activity seen with temsirolimus and everolimus, pharmaceutical derivatives of rapamycin and allosteric partial inhibitors of TORC1, provide clinical proof-of-concept for targeting mTOR for cancer therapy. TORC1 is upstream of HIF1/2 and TORC1 and TORC2 are downstream of the VEGF pathways implicated in the tumorigenesis of metastatic renal cell carcinoma (RCC). We have investigated INK128 in preclinical in vitro and in vivo models of renal cell carcinoma (RCC) and compared its activity to other standard of care agents commonly used to treat this disease.nnResults: INK128 selectively inhibited phosphorylation of S6 and 4EBP1, downstream substrates of TORC1, as well as phosphorylation of AKT downstream of TORC2 in vitro and in vivo. INK128 demonstrated very potent inhibition of tumor cell proliferation and induction of G1 cell cycle arrest in vitro. In mouse tumor models, INK128, rapamycin, avastin and sorafenib displayed anti-tumor efficacy; however, they differ in the mechanisms underlying the anti-tumor activity. INK128 inhibits phosphorylation of AKT, PRAS40, NDRG1, S6 and 4EBP1; rapamycin only inhibits S6 phosphorylation and induces phosphorylation of AKT; sorafenib activates several pathway components that are downstream of TORC1/2. INK128 and rapamycin suppressed tumor growth by directly inhibiting tumor cell proliferation; however neither had much impact on tumor-associated angiogenesis. In contrast, sorafenib and avastin suppressed tumor growth by potently inhibiting tumor angiogenesis. The activity of the combination of INK128 with sorafenib or avastin yielded sustained tumor regression by targeting tumor cells and the microenvironment.nnConclusion: INK128 offers a novel approach for the treatment of renal cell carcinoma by targeting TORC1/2 signaling. Additionally, the mechanism of action of INK128 is different than current therapies and may not display cross resistance with current standard of care agents.nnCitation 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 4486. doi:10.1158/1538-7445.AM2011-4486


Cancer Research | 2010

Abstract 4496: INK128, an orally active TORC1/2 kinase inhibitor, shows broad antitumor activity and enhances efficacy of cytotoxic as well as targeted agents

Linda Kessler; Shunyou Wang; Xin Guo; Jeff Kucharski; Jocelyn Staunton; Lucy Lan; Marikka Elia; Josh Stewart; Jolene Brown; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Katti Jessen; Christian Rommel; Yi Liu

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DCnnmTOR kinase operates via two distinct multi-protein complexes, TORC1 and TORC2, which together regulate growth, metabolism, angiogenesis and survival. Because the PI3K/Akt/mTOR pathway integrates nutrient and hormonal signaling and is frequently dysregulated in human cancer, the mTOR kinase has become an important target for oncology drug development. However, multiple genomic and signaling pathways are often simultaneously activated in cancer, and pathway redundancy and compensatory feedback can blunt the activity of even the most potent anticancer agents. Thus, it is important to study the anti-tumor efficacy of potential anticancer agents, both alone and in combination with cytotoxic and targeted agents, to achieve the optimal therapeutic effect. Through rational drug design we have discovered INK128, a potent and selective TORC1/2 inhibitor with outstanding drug-like properties. INK128 inhibits both the phosphorylation of S6 and 4EBP1, downstream substrates of TORC1, and selectively inhibits AKT phosphorylation at Ser473, the downstream substrate of TORC2, both in vitro and in vivo. Potent inhibition was also observed in cell lines resistant to rapamycin and PanPI3K inhibitors. Daily, oral administration of INK128 inhibited angiogenesis and tumor growth in multiple xenograft models with predicted PK/PD relationship. We have studied INK128 in combination with chemotherapeutic agents as well as with molecular targeted agents both in vitro and in vivo tumor models. In most cases, INK128 demonstrated a synergistically enhanced inhibition of tumor growth and suppression of respective signaling pathways. Additionally, induction of apoptosis was observed only when INK128 was used in combination, suggesting apoptosis as a potential contributor to the observed synergy. A subset of the agents that displayed synergy when combined with INK128 in vitro were evaluated in a number of xenograft models. The combinations were well-tolerated and displayed both enhanced anti-tumor efficacy and enhanced inhibition of key pharmacodynamic markers. In summary, INK128 offers a compelling approach to the treatment of cancer either as a single agent or in combination with other anti-cancer agents.nnCitation 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 4496.


Cancer Research | 2010

Abstract 4488: INK128: An orally active TORC1/2 kinase inhibitor demonstrates potent antitumor activity in preclinical models of renal cell carcinoma

Shunyou Wang; Linda Kessler; Jeff Kucharski; Lucy Lan; Xin Guo; Jocelyn Staunton; Marikka Elia; Josh Stewart; Jolene Brown; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Katti Jessen; Christian Rommel; Yi Liu

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DCnnAnti-angiogenic agents have demonstrated promising activity in patients with renal cell carcinoma (RCC). Patients with clear cell RCC often have mutations or silencing of the von Hippel-Lindau gene, leading to an accumulation of HIF 1 alpha (HIF1α), a key mediator of hypoxia-triggered neo-angiogenesis. The mammalian target of rapamycin (mTOR) is upstream of HIF1a and downstream of the VEGF pathway. mTOR kinase comprises two distinct multi-protein complexes, TORC1 and TORC2, which together regulate growth, metabolism, angiogenesis and survival. Pharmaceutical derivatives of rapamycin, a partial allosteric inhibitor of TORC1, provide clinical proof of concept for targeting mTOR in RCC as well as insights into how ATP-competitive TORC1/2 inhibitors might provide superior efficacy. Through rational drug design we have discovered INK128, a potent, selective TORC1/2 inhibitor with outstanding drug-like properties. We investigated INK128 in preclinical in vitro and in vivo models of RCC. Interestingly, while both INK128 and rapamycin exhibit comparable anti-angiogenic activity in vitro, INK128 correlated with potent and complete blockade of cell proliferation, while rapamycin failed to establish a dose-dependent maximum inhibition of tumor cell proliferation. A comparison of INK128, rapamycin, Nexavar and Avastin demonstrated that all exhibited potent inhibition of tumor growth, via different molecular mechanisms. INK128 inhibits phosphorylation of AKT, S6 and 4EBP1; rapamycin inhibits only S6 phosphorylation and induces AKT phosphorylation; Nexavar and Avastin have little effect on the PI3K/AKT/mTOR pathway. Only INK128 induced autophagy and decreased expression of cyclin D1. INK128 and rapamycin both inhibit expression of HIF-1α and VEGF, which contributes to their anti-angiogenic activity. We conclude that, although Nexavar, Avastin and rapamycin exert their activity primarily through effects on the tumor microenvironment, the anti-tumor activity of INK128 is derived from direct inhibition of tumor cell growth as well as anti-angiogenic activities. In summary, targeting TORC1/2 signaling with INK128 offers a compelling approach to the treatment of RCC.nnCitation 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 4488.


Cancer Research | 2010

Abstract 1668: Pharmacodynamic biomarker development for INK128, a potent and selective inhibitor of TORC1/2 for the treatment of cancer

Katayoun Jessen; Shunyou Wang; Xin Guo; Linda Kessler; Jeff Kucharski; Jocelyn Staunton; Lucy Lan; Marikka Elia; Josh Stewart; Jolene Brown; Liansheng Li; Katrina Chan; Michael Martin; Pingda Ren; Christian Rommel; Yi Liu

Pharmacokinetic / pharmacodynamic (PK/PD) modeling is a valuable strategy to achieve target inhibition as well as predictable and meaningful therapeutic efficacy. The mammalian target of rapamycin (mTOR) comprises two protein complexes, TORC1 and TORC2, which together regulate cell growth, metabolism, angiogenesis, and cell survival. Because TORC1 and TORC2 play a crucial role in several pathways that are frequently dysregulated in human cancer, the mTOR kinase is a compelling target for oncology drug development. Through rational drug design we have identified INK128, a potent and selective small molecule, ATP-competitive, active-site TORC1/2 kinase inhibitor with excellent drug-like properties. Inhibition of phosphorylation of S6 and 4EBP1, downstream markers of TORC1 signaling, was selected for PD analysis in peripheral blood cells (PBCs), skin tissue, and tumor tissue biopsy in mice xenograft tumor models. Time- and dose-dependent inhibition of S6 and 4EBP1 was demonstrated in PBCs by phospho-flow (FACS) analysis. Immunohistochemistry and immunoblot analysis demonstrated a correlation between S6 and 4EBP1 inhibition in tumors or skin tissue and antitumor effect. Additionally, site-selective inhibition of AKT phosphorylation at Ser473, the downstream substrate of TORC2, was also demonstrated in tumors and skin biopsies in mouse xenograft models. Our results demonstrate that daily, oral administration of INK128 selectively inhibits PI3K/AKT/mTOR signaling at the level of TORC1/2, and show that INK128 inhibits growth, and in some cases induces regression, of various tumor xenograft models. Results from these studies display a clear pharmacokinetic and pharmacodynamic relationship. Moreover, the activity of several of these downstream markers can be reproducibly measured in human peripheral blood cells and may permit development of a PK/PD model that might assist to predict PBC and skin tissue PD marker inhibition time-profiles in patients. In summary, INK128 presents a compelling, biomarker-guided approach for the treatment of a variety of cancer by targeting TORC1/2 signaling. 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 1668.

Collaboration


Dive into the Katrina Chan's collaboration.

Top Co-Authors

Avatar

Pingda Ren

University of California

View shared research outputs
Top Co-Authors

Avatar

Yi Liu

Princeton University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeff Kucharski

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Jocelyn Staunton

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Liansheng Li

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Michael Martin

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katti Jessen

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge