Network


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

Hotspot


Dive into the research topics where Judy Lucas is active.

Publication


Featured researches published by Judy Lucas.


Cancer Research | 2009

Biochemical, Cellular, and In vivo Activity of Novel ATP-Competitive and Selective Inhibitors of the Mammalian Target of Rapamycin

Ker Yu; Lourdes Toral-Barza; Celine Shi; Weiguo Zhang; Judy Lucas; Boris Shor; Jamie Kim; Jeroen C. Verheijen; Kevin J. Curran; David Malwitz; Derek Cecil Cole; John W. Ellingboe; Semiramis Ayral-Kaloustian; Tarek S. Mansour; James Joseph Gibbons; Robert T. Abraham; Pawel Wojciech Nowak; Arie Zask

The mammalian target of rapamycin (mTOR) is centrally involved in cell growth, metabolism, and angiogenesis. While showing clinical efficacy in a subset of tumors, rapamycin and rapalogs are specific and allosteric inhibitors of mTOR complex 1 (mTORC1), but they do not directly inhibit mTOR complex 2 (mTORC2), an emerging player in cancer. Here, we report chemical structure and biological characterization of three pyrazolopyrimidine ATP-competitive mTOR inhibitors, WAY-600, WYE-687, and WYE-354 (IC(50), 5-9 nmol/L), with significant selectivity over phosphatidylinositol 3-kinase (PI3K) isofoms (>100-fold). Unlike the rapalogs, these inhibitors acutely blocked substrate phosphorylation by mTORC1 and mTORC2 in vitro and in cells in response to growth factor, amino acids, and hyperactive PI3K/AKT. Unlike the inhibitors of PI3K or dual-pan PI3K/mTOR, cellular inhibition of P-S6K1(T389) and P-AKT(S473) by the pyrazolopyrimidines occurred at significantly lower inhibitor concentrations than those of P-AKT(T308) (PI3K-PDK1 readout), showing mTOR selectivity in cellular setting. mTOR kinase inhibitors reduced AKT downstream function and inhibited proliferation of diverse cancer cell lines. These effects correlated with a strong G(1) cell cycle arrest in both the rapamycin-sensitive and rapamycin-resistant cells, selective induction of apoptosis, repression of global protein synthesis, and down-regulation of angiogenic factors. When injected into tumor-bearing mice, WYE-354 inhibited mTORC1 and mTORC2 and displayed robust antitumor activity in PTEN-null tumors. Together, our results highlight mechanistic differentiation between rapalogs and mTOR kinase inhibitors in targeting cancer cell growth and survival and provide support for clinical development of mTOR kinase inhibitors as new cancer therapy.


Science Signaling | 2010

Ammonia Derived from Glutaminolysis Is a Diffusible Regulator of Autophagy

Christina H. Eng; Ker Yu; Judy Lucas; Eileen White; Robert T. Abraham

A volatile metabolic by-product can promote cell survival through the induction of autophagy. From Metabolism to Autophagy Autophagy is a catabolic process in which macromolecules and organelles are degraded to reclaim nutrients during starvation or to eliminate damaged, potentially toxic cellular components. Dysregulation of autophagy has been implicated in the pathogenesis of various diseases, including cancer and Parkinson’s disease. Noting that culture medium conditioned by actively proliferating cells increased autophagy in secondary cell cultures, Eng et al. identified ammonia, a volatile by-product of a metabolic process known as glutaminolysis, as a diffusible stimulator of autophagy. Glutaminolysis can be higher in tumor cells than in normal cells, and in mice implanted with tumor xenografts derived from human cancer cell lines, ammonia concentrations in tumor-associated fluids were higher than those found in the general circulation of the murine host and were similar to those required for autophagy stimulation. Moreover, ammonia-induced autophagy protected cells from cell death triggered by exposure to tumor necrosis factor–α. Together, these results suggest that treatments that alter intracellular glutamine concentrations or target glutamine metabolism may be possible strategies in cancer therapy. Autophagy is a tightly regulated catabolic process that plays key roles in normal cellular homeostasis and survival during periods of extracellular nutrient limitation and stress. The environmental signals that regulate autophagic activity are only partially understood. Here, we report a direct link between glutamine (Gln) metabolism and autophagic activity in both transformed and nontransformed human cells. Cells cultured for more than 2 days in Gln-containing medium showed increases in autophagy that were not attributable to nutrient depletion or to inhibition of mammalian target of rapamycin. Conditioned medium from these cells contained a volatile factor that triggered autophagy in secondary cell cultures. We identified this factor as ammonia derived from the deamination of Gln by glutaminolysis. Gln-dependent ammonia production supported basal autophagy and protected cells from tumor necrosis factor–α (TNF-α)–induced cell death. Thus, Gln metabolism not only fuels cell growth but also generates an autocrine- and paracrine-acting regulator of autophagic flux in proliferating cells.


Cancer Research | 2010

Beyond Rapalog Therapy: Preclinical Pharmacology and Antitumor Activity of WYE-125132, an ATP-Competitive and Specific Inhibitor of mTORC1 and mTORC2

Ker Yu; Celine Shi; Lourdes Toral-Barza; Judy Lucas; Boris Shor; Jae Eun Kim; Weiguo Zhang; Robert Mahoney; Christine Gaydos; LuAnna Tardio; Sung Kyoo Kim; Roger Conant; Kevin J. Curran; Joshua Kaplan; Jeroen C. Verheijen; Semiramis Ayral-Kaloustian; Tarek S. Mansour; Robert T. Abraham; Arie Zask; James Joseph Gibbons

The mammalian target of rapamycin (mTOR) is a major component of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway that is dysregulated in 50% of all human malignancies. Rapamycin and its analogues (rapalogs) partially inhibit mTOR through allosteric binding to mTOR complex 1 (mTORC1) but not mTOR complex 2 (mTORC2), an emerging player in cancer. Here, we report WYE-125132 (WYE-132), a highly potent, ATP-competitive, and specific mTOR kinase inhibitor (IC(50): 0.19 +/- 0.07 nmol/L; >5,000-fold selective versus PI3Ks). WYE-132 inhibited mTORC1 and mTORC2 in diverse cancer models in vitro and in vivo. Importantly, consistent with genetic ablation of mTORC2, WYE-132 targeted P-AKT(S473) and AKT function without significantly reducing the steady-state level of the PI3K/PDK1 activity biomarker P-AKT(T308), highlighting a prominent and direct regulation of AKT by mTORC2 in cancer cells. Compared with the rapalog temsirolimus/CCI-779, WYE-132 elicited a substantially stronger inhibition of cancer cell growth and survival, protein synthesis, cell size, bioenergetic metabolism, and adaptation to hypoxia. Oral administration of WYE-132 to tumor-bearing mice showed potent single-agent antitumor activity against MDA361 breast, U87MG glioma, A549 and H1975 lung, as well as A498 and 786-O renal tumors. An optimal dose of WYE-132 achieved a substantial regression of MDA361 and A549 large tumors and caused complete regression of A498 large tumors when coadministered with bevacizumab. Our results further validate mTOR as a critical driver for tumor growth, establish WYE-132 as a potent and profound anticancer agent, and provide a strong rationale for clinical development of specific mTOR kinase inhibitors as new cancer therapy.


Cancer Research | 2005

SKI-606, a Src/Abl Inhibitor with In vivo Activity in Colon Tumor Xenograft Models

Jennifer M. Golas; Judy Lucas; Carlo Etienne; Jonathan Golas; Carolyn Discafani; Latha Sridharan; Erwin R. Boghaert; Kim Arndt; Fei Ye; Diane H. Boschelli; Fangbiao Li; Craig Titsch; Christine Huselton; Inder Chaudhary; Frank Boschelli

Src up-regulation is a common event in human cancers. In colorectal cancer, increased Src levels are an indicator of poor prognosis, and progression to metastatic disease is associated with substantial increases in Src activity. Therefore, we examined the activity of SKI-606, a potent inhibitor of Src and Abl kinases, against colon tumor lines in vitro and in s.c. tumor xenograft models. SKI-606 inhibited Src autophosphorylation with an IC(50) of approximately 0.25 micromol/L in HT29 cells. Phosphorylation of Tyr(925) of focal adhesion kinase, a Src substrate, was reduced by similar concentrations of inhibitor. Antiproliferative activity on plastic did not correlate with Src inhibition in either HT29 or Colo205 cells (IC(50)s, 1.5 and 2.5 micromol/L, respectively), although submicromolar concentrations of SKI-606 inhibited HT29 cell colony formation in soft agar. SKI-606 also caused loosely aggregated Colo205 spheroids to condense into compact spheroids. On oral administration to nude mice at the lowest efficacious dose, peak plasma concentrations of approximately 3 micromol/L, an oral bioavailability of 18%, and a t(1/2) of 8.6 hours were observed. SKI-606 was orally active in s.c. colon tumor xenograft models and caused substantial reductions in Src autophosphorylation on Tyr(418) in HT29 and Colo205 tumors. SKI-606 inhibited HT29 tumor growth on once daily administration, whereas twice daily administration was necessary to inhibit Colo205, HCT116, and DLD1 tumor growth. These results support development of SKI-606 as a therapeutic agent for treatment of colorectal cancer.


Journal of Medicinal Chemistry | 2010

Bis(morpholino-1,3,5-triazine) derivatives: potent adenosine 5'-triphosphate competitive phosphatidylinositol-3-kinase/mammalian target of rapamycin inhibitors: discovery of compound 26 (PKI-587), a highly efficacious dual inhibitor.

Aranapakam Mudumbai Venkatesan; Christoph Martin Dehnhardt; Efren Delos Santos; Zecheng Chen; Osvaldo Dos Santos; Semiramis Ayral-Kaloustian; Gulnaz Khafizova; Natasja Brooijmans; Robert Mallon; Irwin Hollander; Larry Feldberg; Judy Lucas; Ker Yu; James Joseph Gibbons; Robert T. Abraham; Inder Chaudhary; Tarek S. Mansour

The PI3K/Akt signaling pathway is a key pathway in cell proliferation, growth, survival, protein synthesis, and glucose metabolism. It has been recognized recently that inhibiting this pathway might provide a viable therapy for cancer. A series of bis(morpholino-1,3,5-triazine) derivatives were prepared and optimized to provide the highly efficacious PI3K/mTOR inhibitor 1-(4-{[4-(dimethylamino)piperidin-1-yl]carbonyl}phenyl)-3-[4-(4,6-dimorpholin-4-yl-1,3,5-triazin-2-yl)phenyl]urea 26 (PKI-587). Compound 26 has shown excellent activity in vitro and in vivo, with antitumor efficacy in both subcutaneous and orthotopic xenograft tumor models when administered intravenously. The structure-activity relationships and the in vitro and in vivo activity of analogues in this series are described.


Clinical Cancer Research | 2011

Antitumor Efficacy of PKI-587, a Highly Potent Dual PI3K/mTOR Kinase Inhibitor

Robert Mallon; Larry Feldberg; Judy Lucas; Inder Chaudhary; Christoph Martin Dehnhardt; Efren Delos Santos; Zecheng Chen; Osvaldo Dos Santos; Semiramis Ayral-Kaloustian; Aranapakam Mudumbai Venkatesan; Irwin Hollander

Purpose: The aim of this study was to show preclinical efficacy and clinical development potential of PKI-587, a dual phosphoinositide 3-kinase (PI3K)/mTOR inhibitor. Experimental Design: In vitro class 1 PI3K enzyme and human tumor cell growth inhibition assays and in vivo five tumor xenograft models were used to show efficacy. Results: In vitro, PKI-587 potently inhibited class I PI3Ks (IC50 vs. PI3K-α = 0.4 nmol/L), PI3K-α mutants, and mTOR. PKI-587 inhibited growth of 50 diverse human tumor cell lines at IC50 values of less than 100 nmol/L. PKI-587 suppressed phosphorylation of PI3K/mTOR effectors (e.g., Akt), and induced apoptosis in human tumor cell lines with elevated PI3K/mTOR signaling. MDA-MB-361 [breast; HER2+, PIK3CA mutant (E545K)] was particularly sensitive to this effect, with cleaved PARP, an apoptosis marker, induced by 30 nmol/L PKI-587 at 4 hours. In vivo, PKI-587 inhibited tumor growth in breast (MDA-MB-361, BT474), colon (HCT116), lung (H1975), and glioma (U87MG) xenograft models. In MDA-MB-361 tumors, PKI-587 (25 mg/kg, single dose i.v.) suppressed Akt phosphorylation [at threonine(T)308 and serine(S)473] for up to 36 hours, with cleaved PARP (cPARP) evident up to 18 hours. PKI-587 at 25 mg/kg (once weekly) shrank large (∼1,000 mm3) MDA-MB-361 tumors and suppressed tumor regrowth. Tumor regression correlated with suppression of phosphorylated Akt in the MDA-MB-361 model. PKI-587 also caused regression in other tumor models, and efficacy was enhanced when given in combination with PD0325901 (MEK 1/2 inhibitor), irinotecan (topoisomerase I inhibitor), or HKI-272 (neratinib, HER2 inhibitor). Conclusion: Significant antitumor efficacy and a favorable pharmacokinetic/safety profile justified phase 1 clinical evaluation of PKI-587. Clin Cancer Res; 17(10); 3193–203. ©2011 AACR.


Journal of Medicinal Chemistry | 2009

ATP-Competitive Inhibitors of the Mammalian Target of Rapamycin: Design and Synthesis of Highly Potent and Selective Pyrazolopyrimidines.

Arie Zask; Jeroen C. Verheijen; Kevin J. Curran; Joshua Kaplan; David J. Richard; Pawel Wojciech Nowak; David Malwitz; Natasja Brooijmans; Joel Bard; Kristine Svenson; Judy Lucas; Lourdes Toral-Barza; Weiguo Zhang; Irwin Hollander; James Joseph Gibbons; Robert T. Abraham; Semiramis Ayral-Kaloustian; Tarek S. Mansour; Ker Yu

The mammalian target of rapamycin (mTOR), a central regulator of growth, survival, and metabolism, is a validated target for cancer therapy. Rapamycin and its analogues, allosteric inhibitors of mTOR, only partially inhibit one mTOR protein complex. ATP-competitive, global inhibitors of mTOR that have the potential for enhanced anticancer efficacy are described. Structural features leading to potency and selectivity were identified and refined leading to compounds with in vivo efficacy in tumor xenograft models.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Macroautophagy is dispensable for growth of KRAS mutant tumors and chloroquine efficacy

Christina H. Eng; Zuncai Wang; Diane Tkach; Lourdes Toral-Barza; Savuth Ugwonali; Shanming Liu; Stephanie Fitzgerald; Elizabeth George; Elizabeth Frias; Nadire R. Cochran; Rowena De Jesus; Gregory McAllister; Gregory R. Hoffman; Kevin Bray; Luanna Lemon; Judy Lucas; Valeria R. Fantin; Robert T. Abraham; Leon O. Murphy; Beat Nyfeler

Significance Kirsten rat sarcoma (KRAS) mutant tumors are believed to depend on autophagy for growth and survival. This study details the unexpected finding that autophagy-related 7, an enzyme essential for macroautophagy, can be deleted in several KRAS-driven cancer lines without affecting growth in vitro or in vivo. These data indicate that KRAS mutation status does not predict cell-autonomous addiction to autophagy. Furthermore, this report addresses a long-standing question regarding the mechanism of chloroquine, a lysosomotropic agent often used to interrogate effects of autophagy inhibition. Although chloroquine is antiproliferative and synergizes with targeted anticancer drugs, these effects are independent of macroautophagy. Future studies are needed to identify appropriate genetic stratification parameters to predict efficacy toward chloroquine and to characterize such agents further as anticancer combination partners. Macroautophagy is a key stress-response pathway that can suppress or promote tumorigenesis depending on the cellular context. Notably, Kirsten rat sarcoma (KRAS)-driven tumors have been reported to rely on macroautophagy for growth and survival, suggesting a potential therapeutic approach of using autophagy inhibitors based on genetic stratification. In this study, we evaluated whether KRAS mutation status can predict the efficacy to macroautophagy inhibition. By profiling 47 cell lines with pharmacological and genetic loss-of-function tools, we were unable to confirm that KRAS-driven tumor lines require macroautophagy for growth. Deletion of autophagy-related 7 (ATG7) by genome editing completely blocked macroautophagy in several tumor lines with oncogenic mutations in KRAS but did not inhibit cell proliferation in vitro or tumorigenesis in vivo. Furthermore, ATG7 knockout did not sensitize cells to irradiation or to several anticancer agents tested. Interestingly, ATG7-deficient and -proficient cells were equally sensitive to the antiproliferative effect of chloroquine, a lysosomotropic agent often used as a pharmacological tool to evaluate the response to macroautophagy inhibition. Moreover, both cell types manifested synergistic growth inhibition when treated with chloroquine plus the tyrosine kinase inhibitors erlotinib or sunitinib, suggesting that the antiproliferative effects of chloroquine are independent of its suppressive actions on autophagy.


Molecular Cancer Therapeutics | 2013

Long-term Tumor Regression Induced by an Antibody–Drug Conjugate That Targets 5T4, an Oncofetal Antigen Expressed on Tumor-Initiating Cells

Puja Sapra; Marc Damelin; John F. DiJoseph; Kimberly Marquette; Kenneth G. Geles; Jonathan Golas; Maureen Dougher; Bitha Narayanan; Andreas Giannakou; Kiran Khandke; Russell Dushin; Elana Ernstoff; Judy Lucas; Mauricio Leal; George Hu; Christopher J. O'Donnell; Lioudmila Tchistiakova; Robert T. Abraham; Hans-Peter Gerber

Antibody–drug conjugates (ADC) represent a promising therapeutic modality for the clinical management of cancer. We sought to develop a novel ADC that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells (TIC), which comprise the most aggressive cell population in the tumor. We optimized an anti-5T4 ADC (A1mcMMAF) by sulfydryl-based conjugation of the humanized A1 antibody to the tubulin inhibitor monomethylauristatin F (MMAF) via a maleimidocaproyl linker. A1mcMMAF exhibited potent in vivo antitumor activity in a variety of tumor models and induced long-term regressions for up to 100 days after the last dose. Strikingly, animals showed pathologic complete response in each model with doses as low as 3 mg antibody/kg dosed every 4 days. In a non–small cell lung cancer patient-derived xenograft model, in which 5T4 is preferentially expressed on the less differentiated tumor cells, A1mcMMAF treatment resulted in sustained tumor regressions and reduced TIC frequency. These results highlight the potential of ADCs that target the most aggressive cell populations within tumors, such as TICs. In exploratory safety studies, A1mcMMAF exhibited no overt toxicities when administered to cynomolgus monkeys at doses up to 10 mg antibody/kg/cycle × 2 and displayed a half-life of 5 days. The preclinical efficacy and safety data established a promising therapeutic index that supports clinical testing of A1mcMMAF. Mol Cancer Ther; 12(1); 38–47. ©2012 AACR.


Biochemical Pharmacology | 2014

Advances in patient-derived tumor xenografts: From target identification to predicting clinical response rates in oncology

Edward Rosfjord; Judy Lucas; Gang Li; Hans-Peter Gerber

Most oncology compounds entering clinical development have passed stringent preclinical pharmacology evaluation criteria. However, only a small fraction of experimental agents induce meaningful antitumor activities in the clinic. Low predictability of conventional preclinical pharmacology models is frequently cited as a main reason for the unusually high clinical attrition rates of therapeutic compounds in oncology. Therefore, improvement in the predictive values of preclinical efficacy models for clinical outcome holds great promise to reduce the clinical attrition rates of experimental compounds. Recent reports suggest that pharmacology studies conducted with patient derived xenograft (PDX) tumors are more predictive for clinical outcome compared to conventional, cell line derived xenograft (CDX) models, in particular when therapeutic compounds were tested at clinically relevant doses (CRDs). Moreover, the study of the most malignant cell types within tumors, the tumor initiating cells (TICs), relies on the availability of preclinical models that mimic the lineage hierarchy of cells within tumors. PDX models were shown to more closely recapitulate the heterogeneity of patient tumors and maintain the molecular, genetic, and histological complexity of human tumors during early stages of sequential passaging in mice, rendering them ideal tools to study the responses of TICs, tumor- and stromal cells to therapeutic intervention. In this commentary, we review the progress made in the development of PDX models in key areas of oncology research, including target identification and validation, tumor indication search and the development of a biomarker hypothesis that can be tested in the clinic to identify patients that will benefit most from therapeutic intervention.

Collaboration


Dive into the Judy Lucas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge