Network


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

Hotspot


Dive into the research topics where James J. Starling is active.

Publication


Featured researches published by James J. Starling.


Journal of Biological Chemistry | 2012

Reactivation of Mitogen-activated Protein Kinase (MAPK) Pathway by FGF Receptor 3 (FGFR3)/Ras Mediates Resistance to Vemurafenib in Human B-RAF V600E Mutant Melanoma

Vipin Yadav; Xiaoyi Zhang; Jiangang Liu; Shawn T. Estrem; Shuyu Li; Xueqian Gong; Sean Buchanan; James Robert Henry; James J. Starling; Sheng-Bin Peng

Background: B-RAF V600E melanomas rapidly develop resistance to B-RAF inhibitors in the clinic. Results: FGFR3/Ras signaling is elevated and induces resistance to vemurafenib in vemurafenib-resistant cells. Conclusion: FGFR3/Ras confers resistance to B-RAF inhibition via MAPK pathway reactivation. Significance: A novel mechanism of resistance to B-RAF inhibitors is described and potential therapeutic strategies are suggested. Oncogenic B-RAF V600E mutation is found in 50% of melanomas and drives MEK/ERK pathway and cancer progression. Recently, a selective B-RAF inhibitor, vemurafenib (PLX4032), received clinical approval for treatment of melanoma with B-RAF V600E mutation. However, patients on vemurafenib eventually develop resistance to the drug and demonstrate tumor progression within an average of 7 months. Recent reports indicated that multiple complex and context-dependent mechanisms may confer resistance to B-RAF inhibition. In the study described herein, we generated B-RAF V600E melanoma cell lines of acquired-resistance to vemurafenib, and investigated the underlying mechanism(s) of resistance. Biochemical analysis revealed that MEK/ERK reactivation through Ras is the key resistance mechanism in these cells. Further analysis of total gene expression by microarray confirmed a significant increase of Ras and RTK gene signatures in the vemurafenib-resistant cells. Mechanistically, we found that the enhanced activation of fibroblast growth factor receptor 3 (FGFR3) is linked to Ras and MAPK activation, therefore conferring vemurafenib resistance. Pharmacological or genetic inhibition of the FGFR3/Ras axis restored the sensitivity of vemurafenib-resistant cells to vemurafenib. Additionally, activation of FGFR3 sufficiently reactivated Ras/MAPK signaling and conferred resistance to vemurafenib in the parental B-RAF V600E melanoma cells. Finally, we demonstrated that vemurafenib-resistant cells maintain their addiction to the MAPK pathway, and inhibition of MEK or pan-RAF activities is an effective therapeutic strategy to overcome acquired-resistance to vemurafenib. Together, we describe a novel FGFR3/Ras mediated mechanism for acquired-resistance to B-RAF inhibition. Our results have implications for the development of new therapeutic strategies to improve the outcome of patients with B-RAF V600E melanoma.


International Journal of Cancer | 2003

Modulation of P-glycoprotein but not MRP1- or BCRP-mediated drug resistance by LY335979

Robert L. Shepard; Jin Cao; James J. Starling; Anne H. Dantzig

Our study examines the ability of LY335979 (Zosuquidar trihydrochloride) to modulate 3 distinct ABC transporters that are mechanisms of drug resistance: P‐glycoprotein (Pgp, ABCB1), multidrug resistance associated protein (MRP1, ABCC2) and breast cancer resistance protein (BCRP, ABCG2). Pgp‐mediated resistance can be modulated by coadministration with the highly potent, selective inhibitor, LY335979. Modulation of resistance by mitoxantrone and vinorelbine, 2 drugs used to treat certain solid tumors, was examined in a 3‐day cytotoxicity assay using a panel of HL60 leukemia cell lines or MCF‐7 breast cancer transfectants. LY335979, at 0.5 μM, substantially reversed mitoxantrone resistance and fully reversed vinorelbine resistance of Pgp‐expressing HL60/Vinc cells. However, LY335979 did not modulate drug resistance in the MRP1‐expressing HL60/ADR or drug‐sensitive parental HL60 cells. To ascertain if LY335979 modulates BCRP‐mediated drug resistance, the sensitivity of 26‐fold mitoxantrone resistant, BCRP‐transfected MCF‐7 cells was evaluated. Addition of 5 μM LY335979, a concentration ∼100‐fold higher than the affinity of Pgp, had little to no effect on the BCRP transfectant. [125I]Iodomycin photolabeled Pgp in CEM/VLB100 membranes and was inhibited by 5 μM LY335979 and GF120918. No photolabeling of MRP or BCRP occurred in H69AR or MCF‐7/BCRP membranes, respectively. These results further demonstrate that LY335979 is highly specific for Pgp and does not modulate MRP1‐ or BCRP‐mediated resistance and can be used in combination with mitoxantrone and vinorelbine in tumor cells.


Journal of Medicinal Chemistry | 2009

Synthesis, Crystallization, and Biological Evaluation of an Orally Active Prodrug of Gemcitabine

David M. Bender; Jingqi Bao; Anne H. Dantzig; William D. Diseroad; Kevin L. Law; Nicholas A. Magnus; Jeffrey A. Peterson; Everett J. Perkins; Yangwei J. Pu; Susan M. Reutzel-Edens; David Michael Remick; James J. Starling; Gregory A. Stephenson; Radhe K. Vaid; Deyi Zhang; James R. McCarthy

The design, synthesis, and biological characterization of an orally active prodrug (3) of gemcitabine are described. Additionally, the identification of a novel co-crystal solid form of the compound is presented. Valproate amide 3 is orally bioavailable and releases gemcitabine into the systemic circulation after passing through the intestinal mucosa. The compound has entered clinical trials and is being evaluated as a potential new anticancer agent.


Cancer Cell | 2015

Inhibition of RAF Isoforms and Active Dimers by LY3009120 Leads to Anti-tumor Activities in RAS or BRAF Mutant Cancers

Sheng-Bin Peng; James Robert Henry; Michael Kaufman; Wei-Ping Lu; Bryan D. Smith; Subha Vogeti; Thomas J. Rutkoski; Scott C. Wise; Lawrence Chun; Youyan Zhang; Robert D. Van Horn; Tinggui Yin; Xiaoyi Zhang; Vipin Yadav; Shih-Hsun Chen; Xueqian Gong; Xiwen Ma; Yue Webster; Sean Buchanan; Igor Mochalkin; Lysiane Huber; Lisa Kays; Gregory P. Donoho; Jennie L. Walgren; Denis J. McCann; Phenil J. Patel; Ilaria Conti; Gregory D. Plowman; James J. Starling; Daniel L. Flynn

LY3009120 is a pan-RAF and RAF dimer inhibitor that inhibits all RAF isoforms and occupies both protomers in RAF dimers. Biochemical and cellular analyses revealed that LY3009120 inhibits ARAF, BRAF, and CRAF isoforms with similar affinity, while vemurafenib or dabrafenib have little or modest CRAF activity compared to their BRAF activities. LY3009120 induces BRAF-CRAF dimerization but inhibits the phosphorylation of downstream MEK and ERK, suggesting that it effectively inhibits the kinase activity of BRAF-CRAF heterodimers. Further analyses demonstrated that LY3009120 also inhibits various forms of RAF dimers including BRAF or CRAF homodimers. Due to these unique properties, LY3009120 demonstrates minimal paradoxical activation, inhibits MEK1/2 phosphorylation, and exhibits anti-tumor activities across multiple models carrying KRAS, NRAS, or BRAF mutation.


Bioorganic & Medicinal Chemistry Letters | 2002

Tricyclic isoxazoles are novel inhibitors of the multidrug resistance protein (MRP1)

Bryan H. Norman; Joseph Michael Gruber; Sean Patrick Hollinshead; Joseph W. Wilson; James J. Starling; Kevin L. Law; Tracy D. Self; Linda B. Tabas; Daniel C. Williams; Donald C. Paul; Margaret M. Wagner; Anne H. Dantzig

Tricyclic isoxazoles were identified from a screen as a novel class of selective multidrug resistance protein (MRP1) inhibitors. From a screen lead, SAR efforts resulted in the preparation of LY 402913 (9h), which inhibits MRP1 and reverses drug resistance to MRP1 substrates, such as doxorubicin, in HeLa-T5 cells (EC(50)=0.90 microM), while showing no inherent cytotoxicity. Additionally, LY 402913 inhibits ATP-dependent, MRP1-mediated LTC(4) uptake into membrane vesicles prepared from the MRP1-overexpressing HeLa-T5 cells (EC(50)=1.8 microM). LY 402913 also shows selectivity ( approximately 22-fold) against the related transporter, P-glycoprotein, in HL60/Adr and HL60/Vinc cells. Finally, when dosed in combination with the oncolytic MRP1 substrate vincristine, LY 402913 delays the growth of MRP1-overexpressing tumors in vivo.


Molecular Cancer Therapeutics | 2013

The CDK4/6 Inhibitor LY2835219 Overcomes Vemurafenib Resistance Resulting from MAPK Reactivation and Cyclin D1 Upregulation

Vipin Yadav; Teresa F. Burke; Lysiane Huber; Robert D. Van Horn; Youyan Zhang; Sean Buchanan; Edward M. Chan; James J. Starling; Richard P. Beckmann; Sheng-Bin Peng

B-RAF selective inhibitors, including vemurafenib, were recently developed as effective therapies for melanoma patients with B-RAF V600E mutation. However, most patients treated with vemurafenib eventually develop resistance largely due to reactivation of MAPK signaling. Inhibitors of MAPK signaling, including MEK1/2 inhibitor trametinib, failed to show significant clinical benefit in patients with acquired resistance to vemurafenib. Here, we describe that cell lines with acquired resistance to vemurafenib show reactivation of MAPK signaling and upregulation of cyclin D1 and are sensitive to inhibition of LY2835219, a selective inhibitor of cyclin-dependent kinase (CDK) 4/6. LY2835219 was demonstrated to inhibit growth of melanoma A375 tumor xenografts and delay tumor recurrence in combination with vemurafenib. Furthermore, we developed an in vivo vemurafenib-resistant model by continuous administration of vemurafenib in A375 xenografts. Consistently, we found that MAPK is reactivated and cyclin D1 is elevated in vemurafenib-resistant tumors, as well as in the resistant cell lines derived from these tumors. Importantly, LY2835219 exhibited tumor growth regression in a vemurafenib-resistant model. Mechanistic analysis revealed that LY2835219 induced apoptotic cell death in a concentration-dependent manner in vemurafenib-resistant cells whereas it primarily mediated cell-cycle G1 arrest in the parental cells. Similarly, RNAi-mediated knockdown of cyclin D1 induced significantly higher rate of apoptosis in the resistant cells than in parental cells, suggesting that elevated cyclin D1 activity is important for the survival of vemurafenib-resistant cells. Altogether, we propose that targeting cyclin D1–CDK4/6 signaling by LY2835219 is an effective strategy to overcome MAPK-mediated resistance to B-RAF inhibitors in B-RAF V600E melanoma. Mol Cancer Ther; 13(10); 2253–63. ©2014 AACR.


Bioorganic & Medicinal Chemistry Letters | 1991

A novel targeted delivery system utilizing a cephalosporin-oncolytic prodrug activated by an antibody β-lactamase conjugate for the treatment of cancer

Timothy Alan Shepherd; Louis Nickolaus Jungheim; Damon L. Meyer; James J. Starling

Abstract Cephalosporins substituted at C-3′ with oncolytic agents such as vinca alkaloids have been developed as prodrugs for the treatment of solid tumors. Conversion of prodrug to drug is mediated by an immunoconjugate consisting of a β-lactamase enzyme covalently attached to a monoclonal antibody Fab′ fragment which is pre-localized at the tumor.


British Journal of Haematology | 2008

p38 mitogen‐activated protein kinase inhibitor LY2228820 enhances bortezomib‐induced cytotoxicity and inhibits osteoclastogenesis in multiple myeloma; therapeutic implications

Kenji Ishitsuka; Teru Hideshima; Paola Neri; Sonia Vallet; Norihiko Shiraishi; Yutaka Okawa; Zhenxin Shen; Noopur Raje; Tanyel Kiziltepe; Enrique M. Ocio; Dharminder Chauhan; Pierfrancesco Tassone; Nikhil C. Munshi; Robert M. Campbell; Alfonso De Dios; Chuan Shih; James J. Starling; Kazuo Tamura; Kenneth C. Anderson

The interaction between multiple myeloma (MM) cells and the bone marrow (BM) microenvironment induces proliferation and survival of MM cells, as well as osteoclastogenesis. This study investigated the therapeutic potential of novel p38 mitogen‐activated protein kinase (p38MAPK) inhibitor LY2228820 (LY) in MM. Although cytotoxicity against MM cell lines was modest, LY significantly enhanced the toxicity of bortezomib by down‐regulating bortezomib‐induced heat shock protein 27 phosphorylation. LY inhibited interleukin‐6 secretion from long term cultured‐BM stromal cells and BM mononuclear cells (BMMNCs) derived from MM patients in remission. LY also inhibited macrophage inflammatory protein‐1α secretion from patient MM cells and BMMNCs as well as normal CD14 positive osteoclast precursor cells. Moreover, LY significantly inhibited in vitro osteoclastogenesis from CD14 positive cells induced by macrophage‐colony stimulating factor and soluble receptor activator of nuclear factor‐κB ligand. Finally, LY also inhibited in vivo osteoclatogenesis in a severe combined immunodeficiency mouse model of human MM. These results suggest that LY represents a promising novel targeted approach to improve MM patient outcome both by enhancing the effect of bortezomib and by reducing osteoskeletal events.


Molecular Cancer Therapeutics | 2014

Characterization of LY2228820 Dimesylate, a Potent and Selective Inhibitor of p38 MAPK with Antitumor Activity

Robert M. Campbell; Bryan D. Anderson; Nathan A. Brooks; Harold B. Brooks; Edward M. Chan; Alfonso De Dios; Raymond Gilmour; Jeremy R. Graff; Enrique Jambrina; Mary M. Mader; Denis J. McCann; Songqing Na; Stephen Parsons; Susan E. Pratt; Chuan Shih; Louis Stancato; James J. Starling; Courtney M. Tate; Juan A. Velasco; Yong Wang; Xiang S. Ye

p38α mitogen-activated protein kinase (MAPK) is activated in cancer cells in response to environmental factors, oncogenic stress, radiation, and chemotherapy. p38α MAPK phosphorylates a number of substrates, including MAPKAP-K2 (MK2), and regulates the production of cytokines in the tumor microenvironment, such as TNF-α, interleukin-1β (IL-1β), IL-6, and CXCL8 (IL-8). p38α MAPK is highly expressed in human cancers and may play a role in tumor growth, invasion, metastasis, and drug resistance. LY2228820 dimesylate (hereafter LY2228820), a trisubstituted imidazole derivative, is a potent and selective, ATP-competitive inhibitor of the α- and β-isoforms of p38 MAPK in vitro (IC50 = 5.3 and 3.2 nmol/L, respectively). In cell-based assays, LY2228820 potently and selectively inhibited phosphorylation of MK2 (Thr334) in anisomycin-stimulated HeLa cells (at 9.8 nmol/L by Western blot analysis) and anisomycin-induced mouse RAW264.7 macrophages (IC50 = 35.3 nmol/L) with no changes in phosphorylation of p38α MAPK, JNK, ERK1/2, c-Jun, ATF2, or c-Myc ≤ 10 μmol/L. LY2228820 also reduced TNF-α secretion by lipopolysaccharide/IFN-γ–stimulated macrophages (IC50 = 6.3 nmol/L). In mice transplanted with B16-F10 melanoma, tumor phospho-MK2 (p-MK2) was inhibited by LY2228820 in a dose-dependent manner [threshold effective dose (TED)70 = 11.2 mg/kg]. Significant target inhibition (>40% reduction in p-MK2) was maintained for 4 to 8 hours following a single 10 mg/kg oral dose. LY2228820 produced significant tumor growth delay in multiple in vivo cancer models (melanoma, non–small cell lung cancer, ovarian, glioma, myeloma, breast). In summary, LY2228820 is a p38 MAPK inhibitor, which has been optimized for potency, selectivity, drug-like properties (such as oral bioavailability), and efficacy in animal models of human cancer. Mol Cancer Ther; 13(2); 364–74. ©2013 AACR.


International Journal of Cancer | 2002

A very early induction of major vault protein accompanied by increased drug resistance in U-937 cells

Yi Hu; Andrew G. Stephen; Jin Cao; Lee R. Tanzer; Christopher A. Slapak; Steadman D. Harrison; Viswanath Devanarayan; Anne H. Dantzig; James J. Starling; Leonard H. Rome; Robert E. Moore

U‐937 human leukemia cells were selected for resistance to doxorubicin in the presence or absence of a specific drug modulator that inhibits the activity of P‐glycoprotein (Pgp), encoded by the multidrug‐resistance gene (MDR1). Parental cells expressed low basal levels of the multidrug‐resistance‐associated gene (MRP1) and major vault protein (MVP) mRNAs and no MDR1 mRNA. Two doxorubicin‐resistant cell lines were selected. Both drug‐resistant cell lines upregulated the MVP mRNA level 1.5‐fold within 1 cell passage. The MVP mRNA level continued to increase over time as the doxorubicin selection pressure was increased. MVP protein levels generally paralleled the mRNA levels. The 2 high molecular weight vault protein mRNAs were always expressed at constitutive levels. Fully formed vault particles consisting of the MVP, the 2 high molecular weight proteins and the vault RNA assembled and accumulated to increased levels in drug‐selected cells. MVP induction is therefore the rate‐limiting step for vault particle formation in U‐937 cells. By passage 25 and thereafter, the selected cells were resistant to doxorubicin, etoposide, mitoxantrone and 5‐fluorouracil by a pathway that was independent of MDR1, MRP1, MRP2 and breast cancer resistance protein. In summary, U‐937 doxorubicin‐selected cells are programmed to rapidly upregulate MVP mRNA levels, to accumulate vault particles and to become multidrug resistant.

Collaboration


Dive into the James J. Starling's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge