Network


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

Hotspot


Dive into the research topics where Andrea D. Basso is active.

Publication


Featured researches published by Andrea D. Basso.


Nature | 2006

BRAF mutation predicts sensitivity to MEK inhibition

David B. Solit; Levi A. Garraway; Christine A. Pratilas; Ayana Sawai; Gad Getz; Andrea D. Basso; Qing Ye; Jose Lobo; Yuhong She; Iman Osman; Todd R. Golub; Judith Sebolt-Leopold; William R. Sellers; Neal Rosen

The kinase pathway comprising RAS, RAF, mitogen-activated protein kinase kinase (MEK) and extracellular signal regulated kinase (ERK) is activated in most human tumours, often through gain-of-function mutations of RAS and RAF family members. Using small-molecule inhibitors of MEK and an integrated genetic and pharmacologic analysis, we find that mutation of BRAF is associated with enhanced and selective sensitivity to MEK inhibition when compared to either ‘wild-type’ cells or cells harbouring a RAS mutation. This MEK dependency was observed in BRAF mutant cells regardless of tissue lineage, and correlated with both downregulation of cyclin D1 protein expression and the induction of G1 arrest. Pharmacological MEK inhibition completely abrogated tumour growth in BRAF mutant xenografts, whereas RAS mutant tumours were only partially inhibited. These data suggest an exquisite dependency on MEK activity in BRAF mutant tumours, and offer a rational therapeutic strategy for this genetically defined tumour subtype.


Journal of Lipid Research | 2006

Thematic review series: Lipid Posttranslational Modifications. Farnesyl transferase inhibitors

Andrea D. Basso; Paul Kirschmeier; W. Robert Bishop

Some proteins undergo posttranslational modification by the addition of an isoprenyl lipid (farnesyl- or geranylgeranyl-isoprenoid) to a cysteine residue proximal to the C terminus. Protein isoprenylation promotes membrane association and contributes to protein-protein interactions. Farnesylated proteins include small GTPases, tyrosine phosphatases, nuclear lamina, cochaperones, and centromere-associated proteins. Prenylation is required for the transforming activity of Ras. Because of the high frequency of Ras mutations in cancer, farnesyl transferase inhibitors (FTIs) were investigated as a means to antagonize Ras function. Evaluation of FTIs led to the finding that both K- and N-Ras are alternatively modified by geranylgeranyl prenyltransferase-1 in FTI-treated cells. Geranylgeranylated forms of Ras retain the ability to associate with the plasma membrane and activate substrates. Despite this, FTIs are effective at inhibiting the growth of human tumor cells in vitro, suggesting that activity is dependent on blocking the farnesylation of other proteins. FTIs also inhibit the in vivo growth of human tumor xenografts and sensitize these models to chemotherapeutics, most notably taxanes. Several FTIs have entered clinical trials for various cancer indications. In some clinical settings, primarily hematologic malignancies, FTIs have displayed evidence of single-agent activity. Clinical studies in progress are exploring the antitumor activity of FTIs as single agents and in combination. This review will summarize the basic biology of FTIs, their antitumor activity in preclinical models, and the current status of clinical studies with these agents.


FEBS Letters | 2011

Structural basis of CX-4945 binding to human protein kinase CK2.

Andrew D. Ferguson; Payal R. Sheth; Andrea D. Basso; Sunil Paliwal; Kimberly Gray; Thierry O. Fischmann; Hung V. Le

Protein kinase CK2 (CK2), a constitutively active serine/threonine kinase, is involved in a variety of roles essential to the maintenance of cellular homeostasis. Elevated levels of CK2 expression results in the dysregulation of key signaling pathways that regulate transcription, and has been implicated in cancer. The adenosine‐5′‐triphosphate‐competitive inhibitor CX‐4945 has been reported to show broad spectrum anti‐proliferative activity in multiple cancer cell lines. Although the enzymatic IC50 of CX‐4945 has been reported, the thermodynamics and structural basis of binding to CK2α remained elusive. Presented here are the crystal structures of human CK2α in complex with CX‐4945 and adenylyl phosphoramidate at 2.7 and 1.3 Å, respectively. Biophysical analysis of CX‐4945 binding is also described. This data provides the structural rationale for the design of more potent inhibitors against this emerging cancer target.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of imidazo[1,2-a]pyrazine-based Aurora kinase inhibitors.

David B. Belanger; Patrick J. Curran; Alan Hruza; Johannes Voigt; Zhaoyang Meng; Amit K. Mandal; M. Arshad Siddiqui; Andrea D. Basso; Kimberly Gray

The synthesis and structure-activity relationships (SAR) of novel, potent imidazo[1,2-a]pyrazine-based Aurora kinase inhibitors are described. The X-ray crystal structure of imidazo[1,2-a]pyrazine Aurora inhibitor 1j is disclosed. Compound 10i was identified as lead compound with a promising overall profile.


ACS Medicinal Chemistry Letters | 2010

Discovery of a Potent, Injectable Inhibitor of Aurora Kinases Based on the Imidazo-[1,2-a]-Pyrazine Core

Tao Yu; Jayaram R. Tagat; Angela Kerekes; Ronald J. Doll; Yonglian Zhang; Yushi Xiao; Sara Esposite; David B. Belanger; Patrick J. Curran; Amit K. Mandal; M. Arshad Siddiqui; Neng-Yang Shih; Andrea D. Basso; Ming Liu; Kimberly Gray; Seema Tevar; Jennifer Jones; Suining Lee; Lianzhu Liang; Samad Ponery; Elizabeth B. Smith; Alan Hruza; Johannes Voigt; Lata Ramanathan; Winifred W. Prosise; Mengwei Hu

The imidazo-[1,2-a]-pyrazine (1) is a dual inhibitor of Aurora kinases A and B with modest cell potency (IC50 = 250 nM) and low solubility (5 μM). Lead optimization guided by the binding mode led to the acyclic amino alcohol 12k (SCH 1473759), which is a picomolar inhibitor of Aurora kinases (TdF K d Aur A = 0.02 nM and Aur B = 0.03 nM) with improved cell potency (phos-HH3 inhibition IC50 = 25 nM) and intrinsic aqueous solubility (11.4 mM). It also demonstrated efficacy and target engagement in human tumor xenograft mouse models.


Biochemistry | 2010

Novel Benzimidazole Inhibitors Bind to a Unique Site in the Kinesin Spindle Protein Motor Domain

Payal R. Sheth; Gerald W. Shipps; Wolfgang Seghezzi; Catherine Smith; Cheng-Chi Chuang; David Paul Sanden; Andrea D. Basso; Lev Vilenchik; Kimberly Gray; D. Allen Annis; Elliott Nickbarg; Yao Ma; Brian R. Lahue; Ronald Herbst; Hung V. Le

Affinity selection-mass spectrometry (AS-MS) screening of kinesin spindle protein (KSP) followed by enzyme inhibition studies and temperature-dependent circular dichroism (TdCD) characterization was utilized to identify a series of benzimidazole compounds. This series also binds in the presence of Ispinesib, a known anticancer KSP inhibitor in phase I/II clinical trials for breast cancer. TdCD and AS-MS analyses support simultaneous binding implying existence of a novel non-Ispinesib binding pocket within KSP. Additional TdCD analyses demonstrate direct binding of these compounds to Ispinesib-resistant mutants (D130V, A133D, and A133D + D130V double mutant), further strengthening the hypothesis that the compounds bind to a distinct binding pocket. Also importantly, binding to this pocket causes uncompetitive inhibition of KSP ATPase activity. The uncompetitive inhibition with respect to ATP is also confirmed by the requirement of nucleotide for binding of the compounds. After preliminary affinity optimization, the benzimidazole series exhibited distinctive antimitotic activity as evidenced by blockade of bipolar spindle formation and appearance of monoasters. Cancer cell growth inhibition was also demonstrated either as a single agent or in combination with Ispinesib. The combination was additive as predicted by the binding studies using TdCD and AS-MS analyses. The available data support the existence of a KSP inhibitory site hitherto unknown in the literature. The data also suggest that targeting this novel site could be a productive strategy for eluding Ispinesib-resistant tumors. Finally, AS-MS and TdCD techniques are general in scope and may enable screening other targets in the presence of known drugs, clinical candidates, or tool compounds that bind to the protein of interest in an effort to identify potency-enhancing small molecules that increase efficacy and impede resistance in combination therapy.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of orally bioavailable imidazo[1,2-a]pyrazine-based Aurora kinase inhibitors.

David B. Belanger; Michael Williams; Patrick J. Curran; Amit K. Mandal; Zhaoyang Meng; Matthew P. Rainka; Tao Yu; Neng-Yang Shih; M. Arshad Siddiqui; Ming Liu; Seema Tevar; Suining Lee; Lianzhu Liang; Kimberly Gray; Bohdan Yaremko; Jennifer Jones; Elizabeth B. Smith; Dan Prelusky; Andrea D. Basso

We report a series of potent imidazo[1,2-a]pyrazine-based Aurora kinase inhibitors. Optimization of the solvent accessible 8-position led to improvements in both oral bioavailability and off-target kinase inhibition. Compound 25 demonstrates anti-tumor activity in an A2780 ovarian tumor xenograft model.


Bioorganic & Medicinal Chemistry Letters | 2011

Bioisosteric approach to the discovery of imidazo[1,2-a]pyrazines as potent Aurora kinase inhibitors.

Zhaoyang Meng; Bheemashankar A. Kulkarni; Angela Kerekes; Amit K. Mandal; Sara Esposite; David B. Belanger; Panduranga Adulla P. Reddy; Andrea D. Basso; Seema Tevar; Kimberly Gray; Jennifer Jones; Elizabeth B. Smith; Ronald J. Doll; M. Arshad Siddiqui

Our continued effort toward the development of the imidazo[1,2-a]pyrazine scaffold as Aurora kinase inhibitors is described. Bioisosteric approach was applied to optimize the 8-position of the core. Several new potent Aurora A/B dual inhibitors, such as 25k and 25l, were identified.


Biochemistry | 2009

Thermodynamics of Nucleotide and Inhibitor Binding to Wild-Type and Ispinesib-Resistant Forms of Human Kinesin Spindle Protein

Payal R. Sheth; Andrea D. Basso; Jose S. Duca; Charles A. Lesburg; Polina Ogas; Kimberly Gray; Lissette Nale; Anthony Mannarino; Andrew Prongay; Hung V. Le

Current antimitotic cancer chemotherapy based on vinca alkaloids and taxanes target tubulin, a protein required not only for mitotic spindle formation but also for the overall structural integrity of terminally differentiated cells. Among many innovations targeting specific mitotic events, inhibition of motor enzymes including KSP (or Eg5) has been validated as a highly productive approach. Many reported KSP inhibitors bind to an induced allosteric site near the site of ATP hydrolysis, and some have been tested in clinical trials with varying degrees of success. This allosteric site was defined in detail by X-ray crystallography of inhibitor complexes, yet complementary information on binding thermodynamics is still lacking. Using two model ATP-uncompetitive inhibitors, monastrol and ispinesib, we report here the results of thermal denaturation and isothermal titration calorimetric studies. These binding studies were conducted with the wild-type KSP motor domain as well as two ispinesib mutants (D130V and A133D) identified to confer resistance to ispinesib treatment. The thermodynamic parameters obtained were placed in the context of the available structural information and corresponding models of the two ispinesib-resistant mutants. The resulting overall information formed a strong basis for future structure-based design of inhibitors of KSP and related motor enzymes.


Molecular Cancer Therapeutics | 2010

SCH 2047069, a Novel Oral Kinesin Spindle Protein Inhibitor, Shows Single-Agent Antitumor Activity and Enhances the Efficacy of Chemotherapeutics

Andrea D. Basso; Ming Liu; Chaoyang Dai; Kimberly Gray; Lissette Nale; Seema Tevar; Suining Lee; Lianzhu Liang; Abdul Ponery; Bohdan Yaremko; Elizabeth M. Smith; Huadong Tang; Payal R. Sheth; M. Arshad Siddiqui; Daniel J. Hicklin; Paul Kirschmeier

Kinesin spindle protein (KSP) is a mitotic kinesin required for the formation of the bipolar mitotic spindle, and inhibition of this motor protein results in mitotic arrest and cell death. KSP inhibitors show preclinical antitumor activity and are currently undergoing testing in clinical trials. These agents have been dosed intravenously using various dosing schedules. We sought to identify a KSP inhibitor that could be delivered orally and thus provide convenience of dosing as well as the ability to achieve more continuous exposure via the use of dose-dense administration. We discovered SCH 2047069, a potent KSP inhibitor with oral bioavailability across species and the ability to cross the blood-brain barrier. The compound induces mitotic arrest characterized by a monaster spindle and is associated with an increase in histone H3 and mitotic protein monoclonal 2 phosphorylation both in vitro and in vivo. SCH 2047069 showed antitumor activity in a variety of preclinical models as a single agent and in combination with paclitaxel, gemcitabine, or vincristine. Mol Cancer Ther; 9(11); 2993–3002. ©2010 AACR.

Collaboration


Dive into the Andrea D. Basso's collaboration.

Researchain Logo
Decentralizing Knowledge