Network


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

Hotspot


Dive into the research topics where Jonathan A. Meulbroek is active.

Publication


Featured researches published by Jonathan A. Meulbroek.


Clinical Cancer Research | 2007

ABT-888, an Orally Active Poly(ADP-Ribose) Polymerase Inhibitor that Potentiates DNA-Damaging Agents in Preclinical Tumor Models

Cherrie K. Donawho; Yan Luo; Yanping Luo; Thomas D. Penning; Joy Bauch; Jennifer J. Bouska; Velitchka Bontcheva-Diaz; Bryan F. Cox; Theodore L. DeWeese; Larry E. Dillehay; Debra Ferguson; Nayereh S. Ghoreishi-Haack; David R. Grimm; Ran Guan; Edward K. Han; Rhonda R. Holley-Shanks; Boris Hristov; Kenneth B. Idler; Ken Jarvis; Eric F. Johnson; Lawrence Kleinberg; Vered Klinghofer; Loren M. Lasko; Xuesong Liu; Kennan C. Marsh; Thomas McGonigal; Jonathan A. Meulbroek; Amanda M. Olson; Joann P. Palma; Luis E. Rodriguez

Purpose: To evaluate the preclinical pharmacokinetics and antitumor efficacy of a novel orally bioavailable poly(ADP-ribose) polymerase (PARP) inhibitor, ABT-888. Experimental Design:In vitro potency was determined in a PARP-1 and PARP-2 enzyme assay. In vivo efficacy was evaluated in syngeneic and xenograft models in combination with temozolomide, platinums, cyclophosphamide, and ionizing radiation. Results: ABT-888 is a potent inhibitor of both PARP-1 and PARP-2 with Kis of 5.2 and 2.9 nmol/L, respectively. The compound has good oral bioavailability and crosses the blood-brain barrier. ABT-888 strongly potentiated temozolomide in the B16F10 s.c. murine melanoma model. PARP inhibition dramatically increased the efficacy of temozolomide at ABT-888 doses as low as 3.1 mg/kg/d and a maximal efficacy achieved at 25 mg/kg/d. In the 9L orthotopic rat glioma model, temozolomide alone exhibited minimal efficacy, whereas ABT-888, when combined with temozolomide, significantly slowed tumor progression. In the MX-1 breast xenograft model (BRCA1 deletion and BRCA2 mutation), ABT-888 potentiated cisplatin, carboplatin, and cyclophosphamide, causing regression of established tumors, whereas with comparable doses of cytotoxic agents alone, only modest tumor inhibition was exhibited. Finally, ABT-888 potentiated radiation (2 Gy/d × 10) in an HCT-116 colon carcinoma model. In each model, ABT-888 did not display single-agent activity. Conclusions: ABT-888 is a potent inhibitor of PARP, has good oral bioavailability, can cross the blood-brain barrier, and potentiates temozolomide, platinums, cyclophosphamide, and radiation in syngeneic and xenograft tumor models. This broad spectrum of chemopotentiation and radiopotentiation makes this compound an attractive candidate for clinical evaluation.


Molecular Cancer Therapeutics | 2016

ABT-414, an Antibody Drug Conjugate Targeting a Tumor-Selective EGFR Epitope

Andrew C. Phillips; Erwin R. Boghaert; Kedar S. Vaidya; Michael J. Mitten; Suzanne M. Norvell; Hugh D. Falls; Peter J. DeVries; Dong Cheng; Jonathan A. Meulbroek; Fritz G. Buchanan; Laura M. McKay; Neal Goodwin; Edward B. Reilly

Targeting tumor-overexpressed EGFR with an antibody–drug conjugate (ADC) is an attractive therapeutic strategy; however, normal tissue expression represents a significant toxicity risk. The anti-EGFR antibody ABT-806 targets a unique tumor-specific epitope and exhibits minimal reactivity to EGFR in normal tissue, suggesting its suitability for the development of an ADC. We describe the binding properties and preclinical activity of ABT-414, an ABT-806 monomethyl auristatin F conjugate. In vitro, ABT-414 selectively kills tumor cells overexpressing wild-type or mutant forms of EGFR. ABT-414 inhibits the growth of xenograft tumors with high EGFR expression and causes complete regressions and cures in the most sensitive models. Tumor growth inhibition is also observed in tumor models with EGFR mutations, including activating mutations and those with the exon 2–7 deletion [EGFR variant III (EGFRvIII)], commonly found in glioblastoma multiforme. ABT-414 exhibits potent cytotoxicity against glioblastoma multiforme patient-derived xenograft models expressing either wild-type EGFR or EGFRvIII, with sustained regressions and cures observed at clinically relevant doses. ABT-414 also combines with standard-of-care treatment of radiation and temozolomide, providing significant therapeutic benefit in a glioblastoma multiforme xenograft model. On the basis of these results, ABT-414 has advanced to phase I/II clinical trials, and objective responses have been observed in patients with both amplified wild-type and EGFRvIII-expressing tumors. Mol Cancer Ther; 15(4); 661–9. ©2016 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Correlation of tumor growth suppression and methionine aminopetidase-2 activity blockade using an orally active inhibitor

Jieyi Wang; Lora A. Tucker; Jason Stavropoulos; Qian Zhang; Yi-Chun Wang; Gail Bukofzer; Amanda Niquette; Jonathan A. Meulbroek; David M. Barnes; Jianwei Shen; Jennifer J. Bouska; Cherrie K. Donawho; George S. Sheppard; Randy L. Bell

This laboratory and others have shown that agents that inhibit the in vitro catalytic activity of methionine aminopeptidase-2 (MetAP2) are effective in blocking angiogenesis and tumor growth in preclinical models. However, these prototype MetAP2 inhibitors are clearly not optimized for therapeutic use in the clinic. We have discovered an orally active class of MetAP2 inhibitors, the anthranilic acid sulfonamides exemplified by A-800141, which is highly specific for MetAP2. This orally bioavailable inhibitor exhibits an antiangiogenesis effect and a broad anticancer activity in a variety of tumor xenografts including B cell lymphoma, neuroblastoma, and prostate and colon carcinomas, either as a single agent or in combination with cytotoxic agents. We also have developed a biomarker assay to evaluate in vivo MetAP2 inhibition in circulating mononuclear cells and in tumors. This biomarker assay is based on the N-terminal methionine status of the MetAP2-specific substrate GAPDH in these cells. In cell cultures in vitro, the sulfonamide MetAP2 inhibitor A-800141 caused the formation of GAPDH variants with an unprocessed N-terminal methionine. A-800141 blocked tumor growth and MetAP2 activity in a similar dose–response in mouse models, demonstrating the antitumor effects seen for A-800141 are causally connected to MetAP2 inhibition in vivo. The sulfonamide MetAP2 inhibitor and GAPDH biomarker in circulating leukocytes may be used for the development of a cancer treatment.


BMC Cancer | 2009

Reversal of oncogene transformation and suppression of tumor growth by the novel IGF1R kinase inhibitor A-928605

William N. Pappano; Paul M Jung; Jonathan A. Meulbroek; Yi-Chun Wang; Robert D. Hubbard; Qian Zhang; Meagan M Grudzien; Niru B. Soni; Eric F. Johnson; George S. Sheppard; Cherrie K. Donawho; Fritz G. Buchanan; Steven K. Davidsen; Randy L. Bell; Jieyi Wang

BackgroundThe insulin-like growth factor (IGF) axis is an important signaling pathway in the growth and survival of many cell and tissue types. This pathway has also been implicated in many aspects of cancer progression from tumorigenesis to metastasis. The multiple roles of IGF signaling in cancer suggest that inhibition of the pathway might yield clinically effective therapeutics.MethodsWe describe A-928605, a novel pyrazolo [3,4-d]pyrimidine small molecule inhibitor of the receptor tyrosine kinases (IGF1R and IR) responsible for IGF signal transduction. This compound was first tested for its activity and selectivity via conventional in vitro kinome profiling and cellular IGF1R autophosphorylation. Additionally, cellular selectivity and efficacy of A-928605 were analyzed in an IGF1R oncogene-addicted cell line by proliferation, signaling and microarray studies. Finally, in vivo efficacy of A-928605 was assessed in the oncogene-addicted cell line and in a neuroblastoma model as a single agent as well as in combination with clinically approved therapeutics targeting EGFR in models of pancreatic and non-small cell lung cancers.ResultsA-928605 is a selective IGF1R inhibitor that is able to abrogate activation of the pathway both in vitro and in vivo. This novel compound dosed as a single agent is able to produce significant growth inhibition of neuroblastoma xenografts in vivo. A-928605 is also able to provide additive effects when used in combination with clinically approved agents directed against EGFR in non-small cell lung and human pancreatic tumor models.ConclusionThese results suggest that a selective IGF1R inhibitor such as A-928605 may provide a useful clinical therapeutic for IGF pathway affected tumors and warrants further investigation.


Bioorganic & Medicinal Chemistry Letters | 2003

In vivo characterization of A-192411: a novel fungicidal lipopeptide (II)

Jonathan A. Meulbroek; Angela M. Nilius; Qun Li; Weibo Wang; Lisa A. Hasvold; Beth Steiner; Daniel A. Dickman; H. Ding; David J. Frost; Robert C. Goldman; Paul A. Lartey; Jacob J. Plattner; Youssef L. Bennani

The ability of the novel antifungal cyclic hexalipopetide A-192411 to treat fungal infections in rodents is presented. Efficacy was demonstrated against Candida albicans as both prolonged survival of systemically infected mice and clearance of viable yeasts from kidneys. The efficacy of A-192411, administered intraperitoneally, was equivalent to amphotercin B at a 4-fold lower dose by weight in the systemic candidiasis models in mice, while the efficacy of A-192441 administered intravenously was equivalent to amphotercin B by weight in the Candida pyelonephritis model in rats. A-192411 also slightly prolonged the survival of immunocompromised mice infected systemically with Aspergillus fumigatus.


Journal of Medicinal Chemistry | 1996

Synthesis and structure-activity relationships of 2-pyridones : A novel series of potent DNA gyrase inhibitors as antibacterial agents

Qun Li; Daniel T. W. Chu; Akiyo Claiborne; Curt S. Cooper; Cheuk Man Lee; Kathleen Raye; Kristine B. Berst; Pamela Donner; Weibo Wang; Lisa A. Hasvold; Anthony K. L. Fung; Zhenkun Ma; Michael D. Tufano; Robert K. Flamm; Linus L. Shen; John L. Baranowski; Angela M. Nilius; Jeff Alder; Jonathan A. Meulbroek; Kennan Marsh; DeAnne Crowell; Yu-hua Hui; Louis Seif; Laura Melcher; Rodger F. Henry; Steven Spanton; Ramin Faghih; Larry L. Klein; S. Ken Tanaka; Jacob J. Plattner


Journal of Medicinal Chemistry | 1999

Synthesis and Antimicrobial Activity of 4H-4-Oxoquinolizine Derivatives: Consequences of Structural Modification at the C-8 Position

Zhenkun Ma; Daniel T. W. Chu; Curt S. Cooper; Qun Li; Anthony K. L. Fung; Sanyi Wang; Linus L. Shen; Robert K. Flamm; Angela M. Nilius; Jeffery D. Alder; Jonathan A. Meulbroek; Yat Sun Or


Journal of Antimicrobial Chemotherapy | 1996

Efficacy of ABT-719, a 2-pyridone antimicrobial, against enterococci, Escherichia coli, and Pseudomonas aeruginosa in experimental murine pyelonephritis

Jonathan A. Meulbroek; Anatol Oleksijew; S. Ken Tanaka; Jeffrey Alder


Archive | 2012

Binding proteins specific for egfr expressed on tumor and uses thereof

Terrance Grant Johns; Andrew Mark Scott; Gerd Ritter; Achim Jungbluth; Elizabeth Stockert; V. P. Collins; Webster Cavenee; Huei-Jen Su Huang; Antony Wilks Burgess; Edouard Collins Nice; Anne Murray; George Mark; Lloyd J. Old; Edward B. Reilly; Andrew C. Phillips; Jonathan A. Meulbroek; Fritz G. Buchanan


Archive | 2016

ANTICUERPOS Y CONJUGADOS DE ANTICUERPO Y FÁRMACO ANTI-EGFR

Edward B. Reilly; Andrew C. Phillips; Chung Hsieh; Lorenzo Benatuil; Fritz G. Buchanan; Jonathan A. Meulbroek; Jennifer Perez

Collaboration


Dive into the Jonathan A. Meulbroek's collaboration.

Top Co-Authors

Avatar

Cherrie K. Donawho

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Mark Scott

Swinburne University of Technology

View shared research outputs
Top Co-Authors

Avatar

Antony Wilks Burgess

Commonwealth Scientific and Industrial Research Organisation

View shared research outputs
Top Co-Authors

Avatar

Edouard Collins Nice

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Terrance Grant Johns

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anne Murray

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Stockert

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge