Network


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

Hotspot


Dive into the research topics where Robert L. Hudkins is active.

Publication


Featured researches published by Robert L. Hudkins.


Bioorganic & Medicinal Chemistry Letters | 2002

Mixed lineage kinase activity of indolocarbazole analogues

Chikara Murakata; Masami Kaneko; George Gessner; Thelma S. Angeles; Mark A. Ator; Teresa M. O'Kane; Beth Ann McKenna; Beth Ann Thomas; Joanne R. Mathiasen; Michael S. Saporito; Donna Bozyczko-Coyne; Robert L. Hudkins

The MLK1-3 activity for a series of analogues of the indolocarbazole K-252a is reported. Addition of 3,9-bis-alkylthiomethyl groups to K-252a results in potent and selective MLK inhibitors. The in vitro and in vivo survival promoting activity of bis-isopropylthiomethyl-K-252a (16, CEP-11004/KT-8138) is reported.


Progress in Medicinal Chemistry | 2002

Discovery of CEP-1347/KT-7515, an inhibitor of the JNK/SAPK pathway for the treatment of neurodegenerative diseases.

Michael S. Saporito; Robert L. Hudkins; Anna C. Maroney

Apoptosis has been proposed as a mechanism of cell death in Alzheimers, Huntingtons and Parkinsons diseases and the occurrence of apoptosis in these disorders suggests a common mechanism. Events such as oxidative stress, calcium toxicity, mitochondria defects, excitatory toxicity, and deficiency of survival factors are all postulated to play varying roles in the pathogenesis of the diseases. However, the transcription factor c-jun may play a role in the pathology and cell death processes that occur in Alzheimers disease. Parkinsons disease (PD) is also a progressive disorder involving the specific degeneration and death of dopamine neurons in the nigrostriatal pathway. In Parkinsons disease, dopaminergic neurons in the substantia nigra are hypothesized to undergo cell death by apoptotic processes. The commonality of biochemical events and pathways leading to cell death in these diseases continues to be an area under intense investigation. The current therapy for PD and AD remains targeting replacement of lost transmitter, but the ultimate objective in neurodegenerative therapy is the functional restoration and/or cessation of progression of neuronal loss. This chapter will describe a novel approach for the treatment of neurodegenerative diseases through the development of kinase inhibitors that block the active cell death process at an early transcriptional independent step in the stress activated kinase cascade. In particular, preclinical data will be presented on the c-Jun Amino Kinase pathway inhibitor, CEP-1347/KT-7515, with respect to its properties that make it a desirable clinical candidate for treatment of various neurodegenerative diseases.


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]


Current Topics in Medicinal Chemistry | 2010

Histamine H3 Antagonists for Treatment of Cognitive Deficits in CNS Diseases

Rita Raddatz; Ming Tao; Robert L. Hudkins

The H(3) histamine receptor is expressed in many brain regions, including those involved in sleep/wake regulation and cognitive functions. Inhibition of the H(3) receptor leads to increased release of multiple neurotransmitters in these regions, making this receptor an ideal target for the potential enhancement of arousal and/or cognitive processes. The high interest level by several pharmaceutical companies in H(3) receptors as potential drug targets has produced rapid advancement in novel compound series with different properties, providing a variety of preclinical tools as well as advancing several candidates into clinical trials. Multiple chemotypes have demonstrated efficacy in preclinical models covering a range of cognitive deficits, suggesting the potential value of H(3) antagonists as cognition enhancers in a variety of disease states. These studies have revealed the actions of this class of compounds at the cellular, neuronal systems and behavioral levels. Recently, compounds with improved selectivity, pharmacokinetics and preclinical safety profiles have advanced into clinical trials for a number of potential indications. The results of these clinical trials are eagerly awaited and will increase our understanding of the properties of H(3) receptor antagonists that will provide therapeutic value.


Current Drug Targets - Cns & Neurological Disorders | 2002

Targeting the JNK pathway for therapeutic benefit in CNS disease.

Donna Bozyczko-Coyne; Michael S. Saporito; Robert L. Hudkins

The c-Jun N-terminal Kinase (JNK) pathway leading to c-Jun phosphorylation plays a causal role in apoptosis of isolated primary embryonic neurons and of multiple neuronal cell lines following a wide variety of stimuli. Activation of this pathway may also contribute to the neuronal atrophy and death that is associated with neurodegenerative pathological conditions including Alzheimers, Parkinsons, Huntingtons Diseases and stroke. Here, the data that providelinks between the activation of the JNK pathway and its potential to play an operative role in CNS disease are reviewed. Also included is the progress on development of inhibitors targeting the JNK pathway for therapeutic benefit.


Journal of Medicinal Chemistry | 2011

Discovery and Characterization of 6-{4-[3-(R)-2-Methylpyrrolidin-1-yl)propoxy]phenyl}-2H-pyridazin-3-one (CEP-26401, Irdabisant): A Potent, Selective Histamine H3 Receptor Inverse Agonist

Robert L. Hudkins; Rita Raddatz; Ming Tao; Joanne R. Mathiasen; Lisa D. Aimone; Nadine C. Becknell; Catherine Prouty; Lars Jacob Stray Knutsen; Mehran Yazdanian; Gilbert Moachon; Mark A. Ator; John P. Mallamo; Michael J. Marino; Edward R. Bacon; Michael T. Williams

Optimization of a novel series of pyridazin-3-one histamine H(3) receptor (H(3)R) antagonists/inverse agonists identified 6-{4-[3-(R)-2-methylpyrrolidin-1-yl)propoxy]phenyl}-2H-pyridazin-3-one (8a, CEP-26401; irdabisant) as a lead candidate for potential use in the treatment of attentional and cognitive disorders. 8a had high affinity for both human (K(i) = 2.0 nM) and rat (K(i) = 7.2 nM) H(3)Rs with greater than 1000-fold selectivity over the hH(1)R, hH(2)R, and hH(4)R histamine receptor subtypes and against an in vitro panel of 418 G-protein-coupled receptors, ion channels, transporters, and enzymes. 8a demonstrated ideal pharmaceutical properties for a CNS drug in regard to water solubility, permeability and lipophilicity and had low binding to human plasma proteins. It weakly inhibited recombinant cytochrome P450 isoforms and human ether-a-go-go-related gene. 8a metabolism was minimal in rat, mouse, dog, and human liver microsomes, and it had good interspecies pharmacokinetic properties. 8a dose-dependently inhibited H(3)R agonist-induced dipsogenia in the rat (ED(50) = 0.06 mg/kg po). On the basis of its pharmacological, pharmaceutical, and safety profiles, 8a was selected for preclinical development. The clinical portions of the single and multiple ascending dose studies assessing safety and pharmacokinetics have been completed allowing for the initiation of a phase IIa for proof of concept.


Journal of Pharmacology and Experimental Therapeutics | 2012

CEP-26401 (Irdabisant), a Potent and Selective Histamine H3 Receptor Antagonist/Inverse Agonist with Cognition-Enhancing and Wake-Promoting Activities

Rita Raddatz; Robert L. Hudkins; Joanne R. Mathiasen; John A. Gruner; Dorothy G. Flood; Lisa D. Aimone; Siyuan Le; Hervé Schaffhauser; Emir Duzic; Maciej Gasior; Donna Bozyczko-Coyne; Michael J. Marino; Mark A. Ator; Edward R. Bacon; John P. Mallamo; Michael T. Williams

CEP-26401 [irdabisant; 6-{4-[3-((R)-2-methyl-pyrrolidin-1-yl)-propoxy]-phenyl}-2H-pyridazin-3-one HCl] is a novel, potent histamine H3 receptor (H3R) antagonist/inverse agonist with drug-like properties. High affinity of CEP-26401 for H3R was demonstrated in radioligand binding displacement assays in rat brain membranes (Ki = 2.7 ± 0.3 nM) and recombinant rat and human H3R-expressing systems (Ki = 7.2 ± 0.4 and 2.0 ± 1.0 nM, respectively). CEP-26401 displayed potent antagonist and inverse agonist activities in [35S]guanosine 5′-O-(γ-thio)triphosphate binding assays. After oral dosing of CEP-26401, occupancy of H3R was estimated by the inhibition of ex vivo binding in rat cortical slices (OCC50 = 0.1 ± 0.003 mg/kg), and antagonism of the H3R agonist R-α-methylhistamine- induced drinking response in the rat dipsogenia model was demonstrated in a similar dose range (ED50 = 0.06 mg/kg). CEP-26401 improved performance in the rat social recognition model of short-term memory at doses of 0.01 to 0.1 mg/kg p.o. and was wake-promoting at 3 to 30 mg/kg p.o. In DBA/2NCrl mice, CEP-26401 at 10 and 30 mg/kg i.p. increased prepulse inhibition (PPI), whereas the antipsychotic risperidone was effective at 0.3 and 1 mg/kg i.p. Coadministration of CEP-26401 and risperidone at subefficacious doses (3 and 0.1 mg/kg i.p., respectively) increased PPI. These results demonstrate potent behavioral effects of CEP-26401 in rodent models and suggest that this novel H3R antagonist may have therapeutic utility in the treatment of cognitive and attentional disorders. CEP-26401 may also have therapeutic utility in treating schizophrenia or as adjunctive therapy to approved antipsychotics.


Annual Reports in Medicinal Chemistry | 2007

Chapter 4 Recent Advances in Drug Discovery of Histamine H3 Antagonists

Robert L. Hudkins; Rita Raddatz

Publisher Summary H 3 antagonists can function to increase the release of various neurotransmitters, including histamine, acetylcholine, norepinephrine, serotonin and dopamine. They have potential utility in addressing a variety of CNS disorders, including deficits in wakefulness and attention, attentiondeficit hyperactivity disorder (ADHD), various dementias, schizophrenia and obesity. The search for H 3 antagonists with drug-like properties has been almost exclusively focused on amine-based compounds. These tertiary amine scaffolds exhibit an extremely broad diversity of structural classes and pharmacophores, and demonstrate an inherent tolerance of the H 3 R to accommodate considerable functional substitutions with large lipophilic aryl groups, polar hydrogen bond donor and acceptor groups or additional basic amines, accounting for the variety and often simplicity of H 3 scaffolds. Moreover, drug-like tertiary amines with reduced side effect liabilities have been identified and advanced into clinical evaluation. Many excellent reviews have been recently published on H 3 chemistry and biology. An important aspect in understanding the H 3 R and its ligands is the high degree of constitutive activity in vitro and in vivo .


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.


Journal of Medicinal Chemistry | 2008

Mixed-lineage kinase 1 and mixed-lineage kinase 3 subtype-selective dihydronaphthyl[3,4-a]pyrrolo[3,4-c]carbazole-5-ones: optimization, mixed-lineage kinase 1 crystallography, and oral in vivo activity in 1-methyl-4-phenyltetrahydropyridine models.

Robert L. Hudkins; James L. Diebold; Ming Tao; Kurt A. Josef; Chung Ho Park; Thelma S. Angeles; Lisa D. Aimone; Jean Husten; Mark A. Ator; Sheryl L. Meyer; Beverly P. Holskin; John T. Durkin; Alexander A. Fedorov; Elena V. Fedorov; Steven C. Almo; Joanne R. Mathiasen; Donna Bozyczko-Coyne; Michael S. Saporito; Richard W. Scott; John P. Mallamo

The optimization of the dihydronaphthyl[3,4-a]pyrrolo[3,4-c]carbazole-5-one R(2) and R(12) positions led to the identification of the first MLK1 and MLK3 subtype-selective inhibitors within the MLK family. Compounds 14 (CEP-5104) and 16 (CEP-6331) displayed good potency for MLK1 and MLK3 inhibition with a greater than 30- to 100-fold selectivity for related family members MLK2 and DLK. Compounds 14 and 16 were orally active in vivo in a mouse MPTP biochemical efficacy model that was comparable to the first-generation pan-MLK inhibitor 1 (CEP-1347). The MLK1 structure-activity relationships were supported by the first-reported X-ray crystal structure of MLK1 bound with 16.

Collaboration


Dive into the Robert L. Hudkins's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kurt A. Josef

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge