Network


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

Hotspot


Dive into the research topics where Allison L. Zulli is active.

Publication


Featured researches published by Allison L. Zulli.


Molecular Cancer Therapeutics | 2007

The selective poly(ADP-ribose) polymerase-1(2) inhibitor, CEP-8983, increases the sensitivity of chemoresistant tumor cells to temozolomide and irinotecan but does not potentiate myelotoxicity

Sheila J. Miknyoczki; Hong Chang; Jennifer Grobelny; Sonya Pritchard; Candace S. Worrell; Natalie McGann; Mark A. Ator; Jean Husten; James Deibold; Robert L. Hudkins; Allison L. Zulli; Ralph E. Parchment; Bruce Ruggeri

The effect of the potent and selective poly(ADP-ribose) (PAR) polymerase-1 [and PAR polymerase-2] inhibitor CEP-8983 on the ability to sensitize chemoresistant glioblastoma (RG2), rhabdomyosarcoma (RH18), neuroblastoma (NB1691), and colon carcinoma (HT29) tumor cells to temozolomide- and camptothecin-induced cytotoxicity, DNA damage, and G2-M arrest and on the potentiation of chemotherapy-induced myelotoxicity was evaluated using in vitro assays. In addition, the effect of the prodrug CEP-9722 in combination with temozolomide and/or irinotecan on PAR accumulation and tumor growth was also determined using glioblastoma and/or colon carcinoma xenografts relative to chemotherapy alone. CEP-8983 sensitized carcinoma cells to the growth-inhibitory effects of temozolomide and/or SN38 increased the fraction of and/or lengthened duration of time tumor cells accumulated in chemotherapy-induced G2-M arrest and sensitized tumor cells to chemotherapy-induced DNA damage and apoptosis. A granulocyte-macrophage colony-forming unit colony formation assay showed that coincubation of CEP-8983 with temozolomide or topotecan did not potentiate chemotherapy-associated myelotoxicity. CEP-9722 (136 mg/kg) administered with temozolomide (68 mg/kg for 5 days) or irinotecan (10 mg/kg for 5 days) inhibited significantly the growth of RG2 tumors (60%) or HT29 tumors (80%) compared with temozolomide or irinotecan monotherapy, respectively. In addition, CEP-9722 showed “stand alone” antitumor efficacy in these preclinical xenografts. In vivo biochemical efficacy studies showed that CEP-9722 attenuated PAR accumulation in glioma xenografts in a dose- and time-related manner. These data indicate that CEP-8983 and its prodrug are effective chemosensitizing agents when administered in combination with select chemotherapeutic agents against chemoresistant tumors. [Mol Cancer Ther 2007;6(8):2290–302]


Journal of Medicinal Chemistry | 2012

A Selective, Orally Bioavailable 1,2,4-Triazolo[1,5-A]Pyridine-Based Inhibitor of Janus Kinase 2 for Use in Anticancer Therapy: Discovery of Cep-33779.

Benjamin J. Dugan; Diane E. Gingrich; Eugen F. Mesaros; Karen L. Milkiewicz; Matthew A. Curry; Allison L. Zulli; Pawel Dobrzanski; Cynthia Serdikoff; Mahfuza Jan; Thelma S. Angeles; Mark S. Albom; Jennifer L. Mason; Lisa D. Aimone; Sheryl L. Meyer; Zeqi Huang; Kevin J. Wells-Knecht; Mark A. Ator; Bruce Ruggeri; Bruce D. Dorsey

Members of the JAK family of nonreceptor tyrosine kinases play a critical role in the growth and progression of many cancers and in inflammatory diseases. JAK2 has emerged as a leading therapeutic target for oncology, providing a rationale for the development of a selective JAK2 inhibitor. A program to optimize selective JAK2 inhibitors to combat cancer while reducing the risk of immune suppression associated with JAK3 inhibition was undertaken. The structure-activity relationships and biological evaluation of a novel series of compounds based on a 1,2,4-triazolo[1,5-a]pyridine scaffold are reported. Para substitution on the aryl at the C8 position of the core was optimum for JAK2 potency (17). Substitution at the C2 nitrogen position was required for cell potency (21). Interestingly, meta substitution of C2-NH-aryl moiety provided exceptional selectivity for JAK2 over JAK3 (23). These efforts led to the discovery of CEP-33779 (29), a novel, selective, and orally bioavailable inhibitor of JAK2.


Journal of Medicinal Chemistry | 2012

Synthesis and Biological Profile of the pan-Vascular Endothelial Growth Factor Receptor/Tyrosine Kinase with Immunoglobulin and Epidermal Growth Factor-Like Homology Domains 2 (VEGF-R/TIE-2) Inhibitor 11-(2-Methylpropyl)-12,13-dihydro-2-methyl-8-(pyrimidin-2-ylamino)-4H-indazolo[5,4-a]pyrrolo[3,4-c]carbazol-4-one (CEP-11981): A Novel Oncology Therapeutic Agent

Robert L. Hudkins; Nadine C. Becknell; Allison L. Zulli; Ted L. Underiner; Thelma S. Angeles; Lisa D. Aimone; Mark S. Albom; Hong Chang; Sheila J. Miknyoczki; Kathryn Hunter; Susan Jones-Bolin; Hugh Zhao; Edward R. Bacon; John P. Mallamo; Mark A. Ator; Bruce Ruggeri

A substantial body of evidence supports the utility of antiangiogenesis inhibitors as a strategy to block or attenuate tumor-induced angiogenesis and inhibition of primary and metastatic tumor growth in a variety of solid and hematopoietic tumors. Given the requirement of tumors for different cytokine and growth factors at distinct stages of their growth and dissemination, optimal antiangiogenic therapy necessitates inhibition of multiple, complementary, and nonredundant angiogenic targets. 11-(2-Methylpropyl)-12,13-dihydro-2-methyl-8-(pyrimidin-2-ylamino)-4H-indazolo[5,4-a]pyrrolo[3,4-c]carbazol-4-one (11b, CEP-11981) is a potent orally active inhibitor of multiple targets (TIE-2, VEGF-R1, 2, and 3, and FGF-R1) having essential and nonredundant roles in tumor angiogenesis and vascular maintenance. Outlined in this article are the design strategy, synthesis, and biochemical and pharmacological profile for 11b, which completed Phase I clinical assessing safety and pharmacokinetics allowing for the initiation of proof of concept studies.


Bioorganic & Medicinal Chemistry Letters | 2011

Identification of pyridazin-3-one derivatives as potent, selective histamine H3 receptor inverse agonists with robust wake activity

Robert L. Hudkins; Lisa D. Aimone; Thomas R. Bailey; Robert J. Bendesky; Reddeppa reddy Dandu; Derek Dunn; John A. Gruner; Kurt A. Josef; Yin-Guo Lin; Jacquelyn A. Lyons; Val R. Marcy; Joanne R. Mathiasen; Babu G. Sundar; Ming Tao; Allison L. Zulli; Rita Raddatz; Edward R. Bacon

H(3)R structure-activity relationships on a novel class of pyridazin-3-one H(3)R antagonists/inverse agonists are disclosed. Modifications of the pyridazinone core, central phenyl ring and linker led to the identification of molecules with excellent target potency, selectivity and pharmacokinetic properties. Compounds 13 and 21 displayed potent functional H(3)R antagonism in vivo in the rat dipsogenia model and demonstrated robust wake activity in the rat EEG/EMG model.


Bioorganic & Medicinal Chemistry Letters | 2008

Design and synthesis of dihydroindazolo[5,4-a]pyrrolo[3,4-c]carbazole oximes as potent dual inhibitors of TIE-2 and VEGF-R2 receptor tyrosine kinases.

Reddeppareddy Dandu; Allison L. Zulli; Edward R. Bacon; Ted L. Underiner; Candy Robinson; Hong Chang; Sheila Miknyoczki; Jennifer Grobelny; Bruce Ruggeri; Shi Yang; Mark S. Albom; Thelma S. Angeles; Lisa D. Aimone; Robert L. Hudkins

Fused dihydroindazolopyrrolocarbazole oximes have been identified as low nanomolar, potent dual TIE-2 and VEGF-R2 receptor tyrosine kinase inhibitors with excellent cellular potency. Development of the structure-activity relationships (SAR) led to identification of compounds 35 and 40 as potent, selective dual TIE-2/VEGF-R2 inhibitors with favorable pharmacokinetic properties. Compound 35 was orally active in tumor models with no observed toxicity.


Bioorganic & Medicinal Chemistry Letters | 2010

8-THP-DHI analogs as potent Type I dual TIE-2/VEGF-R2 receptor tyrosine kinase inhibitors

Robert L. Hudkins; Allison L. Zulli; Ted L. Underiner; Thelma S. Angeles; Lisa D. Aimone; Sheryl L. Meyer; Daniel Pauletti; Hong Chang; Elena V. Fedorov; Steven C. Almo; Alexander A. Fedorov; Bruce Ruggeri

A novel series of 8-(2-tetrahydropyranyl)-12,13-dihydroindazolo[5,4-a]pyrrolo[3,4-c]carbazoles (THP-DHI) was synthesized and evaluated as dual TIE-2 and VEGF-R2 receptor tyrosine kinase inhibitors. Development of the structure-activity relationships (SAR) with the support of X-ray crystallography led to identification of 7f and 7g as potent, selective dual TIE-2/VEGF-R2 inhibitors with excellent cellular potency and acceptable pharmacokinetic properties. Compounds 7f and 7g were orally active in tumor models with no observed toxicity.


Bioorganic & Medicinal Chemistry Letters | 2012

4-Phenoxypiperidine pyridazin-3-one histamine H3 receptor inverse agonists demonstrating potent and robust wake promoting activity

Robert L. Hudkins; Allison L. Zulli; Reddeppa reddy Dandu; Ming Tao; Kurt A. Josef; Lisa D. Aimone; R. Curtis Haltiwanger; Zeqi Huang; Jacquelyn A. Lyons; Joanne R. Mathiasen; Rita Raddatz; John A. Gruner

Structure-activity relationships for a series of phenoxypiperidine pyridazin-3-one H(3)R antagonists/inverse agonists are disclosed. The search for compounds with improved hERG and DAT selectivity without the formation of in vivo active metabolites identified 6-[4-(1-cyclobutyl-piperidin-4-yloxy)-phenyl]-4,4-dimethyl-4,5-dihydro-2H-pyridazin-3-one 17b. Compound 17b met discovery flow criteria, demonstrated potent H(3)R functional antagonism in vivo in the rat dipsogenia model and potent wake activity in the rat EEG/EMG model at doses as low as 0.1 mg/kg ip.


Bioorganic & Medicinal Chemistry Letters | 2012

4,5-Dihydropyridazin-3-one derivatives as histamine H3 receptor inverse agonists

Robert L. Hudkins; Lisa D. Aimone; Reddeppa reddy Dandu; Derek Dunn; John A. Gruner; Zeqi Huang; Kurt A. Josef; Jacquelyn A. Lyons; Joanne R. Mathiasen; Ming Tao; Allison L. Zulli; Rita Raddatz

H(3)R structure-activity relationships for a new class of 4,5-dihydropyridazin-3-one H(3)R antagonists/inverse agonists are disclosed. Modification of the 4,5-dihydropyridazinone moiety to block in vivo metabolism identified 4,4-dimethyl-6-{4-[3-((R)-2-methyl-pyrrolidin-1-yl)-propoxy]-phenyl}-4,5-dihydro-2H-pyridazin-3-one 22 as a lead candidate demonstrating potent in vivo functional H(3)R antagonism in the rat dipsogenia model and robust wake promoting activity in the rat EEG/EMG model.


Bioorganic & Medicinal Chemistry Letters | 2008

TIE-2/VEGF-R2 SAR and in vitro activity of C3-acyl dihydroindazolo[5,4-a]pyrrolo[3,4-c]carbazole analogs.

Ted L. Underiner; Bruce Ruggeri; Lisa D. Aimone; Mark S. Albom; Thelma S. Angeles; Hong Chang; Robert L. Hudkins; Kathryn Hunter; Kurt A. Josef; Candy Robinson; Linda Weinberg; Shi Yang; Allison L. Zulli

Orally bioavailable, dual inhibitors of TIE-2/VEGF-R2 were identified by elaborating the C3/N13 SAR around a fused pyrrolodihydroindazolocarbazole scaffold. Analogs bearing a C3-thiophencarbonyl group were evaluated in enzymatic and cellular biochemical assays; two orally bioavailable analogs were further profiled in functional assays and found to inhibit microvessel growth in rat aortic explant cultures and inhibit Ang-1-stimulated chemotaxis of HUVECs.


Bioorganic & Medicinal Chemistry Letters | 2012

Substituted phenoxypropyl-(R)-2-methylpyrrolidine aminomethyl ketones as histamine-3 receptor inverse agonists.

Allison L. Zulli; Lisa D. Aimone; Joanne R. Mathiasen; John A. Gruner; Rita Raddatz; Edward R. Bacon; Robert L. Hudkins

Optimization of a series of aminomethyl ketone diamine H(3)R antagonists to reduce the brain exposure by lowering the pKa, led to molecules with improved pharmacokinetic properties. Compounds 9, 19, and 25 had high affinity for human H(3)R and demonstrated in vivo H(3)R functional activity in the rat dipsogenia model. Compound 9 displayed modest wake-promoting activity in the rat EEG/EMG model.

Collaboration


Dive into the Allison L. Zulli's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kurt A. Josef

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge