Network


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

Hotspot


Dive into the research topics where Peter Lamb is active.

Publication


Featured researches published by Peter Lamb.


Clinical Cancer Research | 2008

EXEL-7647 Inhibits Mutant Forms of ErbB2 Associated with Lapatinib Resistance and Neoplastic Transformation

Torsten Trowe; Sotiria Boukouvala; Keith Calkins; Richard E. Cutler; Ryan Fong; Roel Funke; Steven Gendreau; Yong D. Kim; Nicole Miller; John R. Woolfrey; Valentina Vysotskaia; Jing Ping Yang; Mary E. Gerritsen; David J. Matthews; Peter Lamb; Timothy S. Heuer

Purpose: Mutations associated with resistance to kinase inhibition are an important mechanism of intrinsic or acquired loss of clinical efficacy for kinase-targeted therapeutics. We report the prospective discovery of ErbB2 mutations that confer resistance to the small-molecule inhibitor lapatinib. Experimental Design: We did in vitro screening using a randomly mutagenized ErbB2 expression library in Ba/F3 cells, which were dependent on ErbB2 activity for survival and growth. Results: Lapatinib resistance screens identified mutations at 16 different ErbB2 amino acid residues, with 12 mutated amino acids mapping to the kinase domain. Mutations conferring the greatest lapatinib resistance cluster in the NH2-terminal kinase lobe and hinge region. Structural computer modeling studies suggest that lapatinib resistance is caused by multiple mechanisms; including direct steric interference and restriction of conformational flexibility (the inactive state required for lapatinib binding is energetically unfavorable). ErbB2 T798I imparts the strongest lapatinib resistance effect and is analogous to the epidermal growth factor receptor T790M, ABL T315I, and cKIT T670I gatekeeper mutations that are associated with clinical drug resistance. ErbB2 mutants associated with lapatinib resistance transformed NIH-3T3 cells, including L755S and T733I mutations known to occur in human breast and gastric carcinomas, supporting a direct mechanism for lapatinib resistance in ErbB2-driven human cancers. The epidermal growth factor receptor/ErbB2/vascular endothelial growth factor receptor inhibitor EXEL-7647 was found to inhibit almost all lapatinib resistance-associated mutations. Furthermore, no ErbB2 mutations were found to be associated with EXEL-7647 resistance and lapatinib sensitivity. Conclusions: Taken together, these data suggest potential target-based mechanisms of resistance to lapatinib and suggest that EXEL-7647 may be able to circumvent these effects.


Leukemia | 2007

The novel tyrosine kinase inhibitor EXEL-0862 induces apoptosis in human FIP1L1-PDGFR-α-expressing cells through caspase-3-mediated cleavage of Mcl-1

Jingxuan Pan; Alfonso Quintás-Cardama; Taghi Manshouri; F. Giles; Peter Lamb; Ayalew Tefferi; J. Cortes; H. Kantarjian; S. Verstovsek

The FIP1-like-1 (FIP1L1)-platelet-derived growth factor receptor-alpha (FIP1L1-PDGFR-α) fusion kinase causes hypereosinophilic syndrome (HES) in a defined subset of patients. Imatinib mesylate is a potent inhibitor of ABL but also of PDGFR-α, and has been associated with durable hematologic responses in patients with HES. However, development of mutations in the tyrosine kinase domain may hamper the activity of tyrosine kinase inhibitors (TKIs), which suggests that novel agents are warranted to prevent or overcome resistance. We evaluated the efficacy of the novel TKI EXEL-0862 in FIP1L1-PDGFR-α-expressing cell lines and in cells from a patient with HES harboring the FIP1L1-PDGFR-α gene. EXEL-0862 inhibited the proliferation of EOL-1 and imatinib-resistant T674I FIP1L1-PDGFR-α-expressing cells and resulted in potent inhibition of the phosphorylation of PDGFR-α and downstream proteins STAT3 and Erk1/2, both in vitro and ex vivo. Moreover, EXEL-0862 induced apoptotic death in EOL-1 cells and imatinib-resistant T674I FIP1L1-PDGFR-α-expressing cells, and resulted in significant downregulation of the antiapoptotic protein Mcl-1 through a caspase-dependent mechanism. Our data establish EXEL-0862 as a solid candidate for the targeted treatment of patients with FIP1L1-PDGFR-α-positive HES.


Molecular Cancer Therapeutics | 2014

Characterization of the Activity of the PI3K/mTOR Inhibitor XL765 (SAR245409) in Tumor Models with Diverse Genetic Alterations Affecting the PI3K Pathway

Peiwen Yu; Laird Ad; Du X; Wu J; Won Ka; Yamaguchi K; Hsu Pp; Fawn Qian; Jaeger Ct; Wentao Zhang; Buhr Ca; Shen P; Abulafia W; Jason Chen; Young J; Plonowski A; Yakes Fm; Felix Chu; Lee M; Bentzien F; Lam St; Dale S; David J. Matthews; Peter Lamb; Paul Foster

Activation of the PI3K (phosphoinositide 3-kinase) pathway is a frequent occurrence in human tumors and is thought to promote growth, survival, and resistance to diverse therapies. Here, we report pharmacologic characterization of the pyridopyrimidinone derivative XL765 (SAR245409), a potent and highly selective pan inhibitor of class I PI3Ks (α, β, γ, and δ) with activity against mTOR. Broad kinase selectivity profiling of >130 protein kinases revealed that XL765 is highly selective for class I PI3Ks and mTOR over other kinases. In cellular assays, XL765 inhibits the formation of PIP3 in the membrane, and inhibits phosphorylation of AKT, p70S6K, and S6 phosphorylation in multiple tumor cell lines with different genetic alterations affecting the PI3K pathway. In a panel of tumor cell lines, XL765 inhibits proliferation with a wide range of potencies, with evidence of an impact of genotype on sensitivity. In mouse xenograft models, oral administration of XL765 results in dose-dependent inhibition of phosphorylation of AKT, p70S6K, and S6 with a duration of action of approximately 24 hours. Repeat dose administration of XL765 results in significant tumor growth inhibition in multiple human xenograft models in nude mice that is associated with antiproliferative, antiangiogenic, and proapoptotic effects. Mol Cancer Ther; 13(5); 1078–91. ©2014 AACR.


Journal of Medicinal Chemistry | 2012

Discovery of a Novel Series of Potent and Orally Bioavailable Phosphoinositide 3-Kinase γ Inhibitors

James W. Leahy; Chris A. Buhr; Henry William Beecroft Johnson; Byung Gyu Kim; Tae-Gon Baik; Jonah Cannoy; Timothy Patrick Forsyth; Joon Won Jeong; Matthew Sangyup Lee; Sunghoon Ma; Kevin Noson; Longcheng Wang; Matthew A. Williams; John M. Nuss; Eric Brooks; Paul A. Foster; Leanne Goon; Nathan Heald; Charles R. Holst; Christopher Jaeger; Scott Lam; Julie Lougheed; Lam Nguyen; Arthur Plonowski; Joanne Song; Thomas J. Stout; Xiang Wu; Michael Yakes; Peiwen Yu; Wentao Zhang

The phosphoinositide 3-kinases (PI3Ks) have been linked to an extraordinarily diversified group of cellular functions making these enzymes compelling targets for the treatment of disease. A large body of evidence has linked PI3Kγ to the modulation of autoimmune and inflammatory processes making it an intriguing target for drug discovery. Our high-throughput screening (HTS) campaign revealed two hits that were nominated for further optimization studies. The in vitro activity of the first HTS hit, designated as the sulfonylpiperazine scaffold, was optimized utilizing structure-based design. However, nonoptimal pharmacokinetic properties precluded this series from further studies. An overlay of the X-ray structures of the sulfonylpiperazine scaffold and the second HTS hit within their complexes with PI3Kγ revealed a high degree of overlap. This feature was utilized to design a series of hybrid analogues including advanced leads such as 31 with desirable potency, selectivity, and oral bioavailability.


Drug Discovery Today | 1998

Biochemical approaches to discovering modulators of the JAK—STAT pathway

Peter Lamb; Peter Tapley; Jonathan Rosen

Regulation of cytokine activity has application in the treatment of numerous diseases, as illustrated by the successful clinical use of recombinant cytokines. The discovery of two key families of signaling proteins — the Janus kinases (JAKs) and the signal transducers and activators of transcription (STATs) — has greatly increased our understanding of cytokine signal transduction. This review describes some of the key interactions between JAKs, STATs and other components of the signal transduction cascade, and the potential for developing high-throughput biochemical screens for the detection of small molecules that target these interactions. Novel compounds of this type have potential as agonists or antagonists of cytokine action.


Molecular Cancer Therapeutics | 2015

The Selective PI3K Inhibitor XL147 (SAR245408) Inhibits Tumor Growth and Survival and Potentiates the Activity of Chemotherapeutic Agents in Preclinical Tumor Models

Paul Foster; Yamaguchi K; Hsu Pp; Fawn Qian; Du X; Wu J; Won Ka; Peiwen Yu; Jaeger Ct; Wentao Zhang; Marlowe Ck; Keast P; Abulafia W; Jason Chen; Young J; Plonowski A; Yakes Fm; Felix Chu; Engell K; Bentzien F; Lam St; Dale S; Yturralde O; David J. Matthews; Peter Lamb; Laird Ad

Dysregulation of PI3K/PTEN pathway components, resulting in hyperactivated PI3K signaling, is frequently observed in various cancers and correlates with tumor growth and survival. Resistance to a variety of anticancer therapies, including receptor tyrosine kinase (RTK) inhibitors and chemotherapeutic agents, has been attributed to the absence or attenuation of downregulating signals along the PI3K/PTEN pathway. Thus, PI3K inhibitors have therapeutic potential as single agents and in combination with other therapies for a variety of cancer indications. XL147 (SAR245408) is a potent and highly selective inhibitor of class I PI3Ks (α, β, γ, and δ). Moreover, broad kinase selectivity profiling of >130 protein kinases revealed that XL147 is highly selective for class I PI3Ks over other kinases. In cellular assays, XL147 inhibits the formation of PIP3 in the membrane, and inhibits phosphorylation of AKT, p70S6K, and S6 in multiple tumor cell lines with diverse genetic alterations affecting the PI3K pathway. In a panel of tumor cell lines, XL147 inhibits proliferation with a wide range of potencies, with evidence of an impact of genotype on sensitivity. In mouse xenograft models, oral administration of XL147 results in dose-dependent inhibition of phosphorylation of AKT, p70S6K, and S6 with a duration of action of at least 24 hours. Repeat-dose administration of XL147 results in significant tumor growth inhibition in multiple human xenograft models in nude mice. Administration of XL147 in combination with chemotherapeutic agents results in antitumor activity in xenograft models that is enhanced over that observed with the corresponding single agents. Mol Cancer Ther; 14(4); 931–40. ©2015 AACR.


Bioorganic & Medicinal Chemistry Letters | 2012

The Design, Synthesis, and Biological Evaluation of Potent Receptor Tyrosine Kinase Inhibitors.

Moon Hwan Kim; Amy Lew Tsuhako; Erick Wang Co; Dana T. Aftab; Frauke Bentzien; Jason Chen; Wei Cheng; Stefan Engst; Levina Goon; Rhett Ronald Klein; Donna T. Le; Morrison B. Mac; Jason Jevious Parks; Fawn Qian; Monica Rodriquez; Thomas J. Stout; Jeffrey H. Till; Kwang-Ai Won; Xiang Wu; F. Michael Yakes; Peiwen Yu; Wentao Zhang; Yeping Zhao; Peter Lamb; John M. Nuss; Wei Xu

Variously substituted indolin-2-ones were synthesized and evaluated for activity against KDR, Flt-1, FGFR-1 and PDGFR. Extension at the 5-position of the oxindole ring with ethyl piperidine (compound 7i) proved to be the most beneficial for attaining both biochemical and cellular potencies. Further optimization of 7i to balance biochemical and cellular potencies with favorable ADME/ PK properties led to the identification of 8h, a compound with a clean CYP profile, acceptable pharmacokinetic and toxicity profiles, and robust efficacy in multiple xenograft tumor models.


Journal of Medicinal Chemistry | 2012

Discovery of a novel class of potent and orally bioavailable sphingosine 1-phosphate receptor 1 antagonists.

Mohamed Abdulkader Ibrahim; Henry William Beecroft Johnson; Joon Won Jeong; Gary L. Lewis; Xian Shi; Robin Tammie Noguchi; Matthew A. Williams; James W. Leahy; John M. Nuss; John Woolfrey; Monica Banica; Frauke Bentzien; Yu-Chien Chou; Anna Gibson; Nathan Heald; Peter Lamb; Larry Mattheakis; David A. Matthews; Aaron Shipway; Xiang Wu; Wentao Zhang; Sihong Zhou; Geetha Shankar

A series of subtype selective sphingosine 1-phosphate receptor 1 (S1P(1)) antagonists are disclosed. Our high-throughput screening campaign revealed hit 1 for which an increase in potency and mouse oral exposure was achieved with minor modifications to the chemical scaffold. In vivo efficacy revealed that at high doses compounds 12 and 15 inhibited tumor growth. Further optimization of our lead series led to the discovery of proline derivatives 37 (XL541) and 38 which had similar efficacy as our first generation analogues at significantly lower doses. Analogue 37 displayed excellent pharmacokinetics and oral exposure in multiple species.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of a new class of glucosylceramide synthase inhibitors.

Elena S. Koltun; Steven Richards; Vicky Chan; Jason Nachtigall; Hongwang Du; Kevin Noson; Adam Antoni Galan; Naing Aay; Art Hanel; Amanda Harrison; Jeff Zhang; Kwang-Ai Won; Danny Tam; Fawn Qian; Tao Wang; Patricia Finn; Kathleen M. Ogilvie; Jon Rosen; Raju Mohan; Christopher J. Larson; Peter Lamb; John M. Nuss; Patrick Kearney

A novel series of potent inhibitors of glucosylceramide synthase are described. The optimization of biochemical and cellular potency as well as ADME properties led to compound 23c. Broad tissue distribution was obtained following oral administration to mice. Thus 23c could be another useful tool compound for studying the effects of GCS inhibition in vitro and in vivo.


Annual Reports in Medicinal Chemistry | 1996

Chapter 27. The Role of JAKs and STATs in Transcriptional Regulation by Cytokines

Peter Lamb; H. Martin Seidel; Robert B. Stein; Jon Rosen

Publisher Summary Cytokines exert their effects on the target cells by binding to specific cell surface receptors, triggering various intracellular events, including rapid changes in gene expression. The binding of a cytokine to its receptor activate the particular members of a family of tyrosine kinases that are known as just another kinases (JAKs). JAKs associate noncovalently with the cytoplasmic domain of the receptor. Activated JAKs then phosphorylate several substrates, including the receptor itself and members of a family of latent cytoplasmic transcription factors known as signal transducers and activators of transcriptions (STATs). Upon phosphorylation of a specific tyrosyl residue, STAT proteins become activated, and form homo- or heterodimeric complexes that translocate to the nucleus and bind to specific DNA sequences termed STAT binding elements (SBEs). The members of the JAK family interact noncovalently with cytoplasmic domains of cytokine receptor subunits. Like the JAKs, STAT proteins also form a complex with the receptor, but only after binding of the cytokine. The STAT1 and STAT2 proteins have been first identified as constituents of a heterotrimeric protein complex termed ISGF3 that bound to specific promoter sequences called IFN α/β-stimulated response elements (ISREs). Recent studies show that STAT1 is phosphorylated on a serine located near the C-terminus of the protein, within a mitogen-activated protein (MAP) kinase consensus sequence that is also present in a similar location in several other STAT proteins. Many cytokines that signal via the JAK/STAT pathway, including IFN α, IFN β IFN γ, IL-2, growth hormone, and the hematopoietic growth factors, Epo, G-CSF, and GMCSF, have proven clinical utility and other cytokines have high potential. The JAK–STAT signal transduction cascade is also amenble to the development of biochemical assays. Thus, the ordered series of steps mediated by JAKs and STATs has presented excellent opportunities for the discovery of novel drugs that modulate cytokine action.

Collaboration


Dive into the Peter Lamb's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alfonso Quintás-Cardama

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge