Network


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

Hotspot


Dive into the research topics where Josef Vagner is active.

Publication


Featured researches published by Josef Vagner.


Current Opinion in Chemical Biology | 2008

Peptidomimetics, a synthetic tool of drug discovery

Josef Vagner; Hongchang Qu; Victor J. Hruby

The demand for modified peptides with improved stability profiles and pharmacokinetic properties is driving extensive research effort in this field. Many structural modifications of peptides guided by rational design and molecular modeling have been established to develop novel synthetic approaches. Recent advances in the synthesis of conformationally restricted building blocks and peptide bond isosteres are discussed.


Expert Opinion on Therapeutic Targets | 2004

Hitting multiple targets with multimeric ligands.

Heather L. Handl; Josef Vagner; Haiyong Han; Eugene A. Mash; Victor J. Hruby; Robert J. Gillies

Multimeric ligands consist of multiple monomeric ligands attached to a single backbone molecule, creating a multimer that can bind to multiple receptors or targets simultaneously. Numerous examples of multimeric binding exist within nature. Due to the multiple and simultaneous binding events, multimeric ligands bind with an increased affinity compared to their corresponding monomers. Multimeric ligands may provide opportunities in the field of drug discovery by providing enhanced selectivity and affinity of binding interactions, thus providing molecular-based targeted therapies. However, gaps in our knowledge currently exist regarding the quantitative measures for important design characteristics, such as flexibility, length and orientation of the inter-ligand linkers, receptor density and ligand sequence. In this review, multimeric ligand binding in two separate phases is examined. The prerecruitment phase describes the binding of one ligand of a multimer to its corresponding receptor, an event similar to monomeric ligand binding. This results in transient increases in the local concentration of the other ligands, leading to apparent cooperativity. The postrecruitment phase only occurs once all receptors have been aligned and bound by their corresponding ligand. This phase is analogous to DNA–DNA interactions in that the stability of the complex is derived from physical orientation. Multiple factors influence the kinetics and thermodynamics of multimeric binding, and these are discussed.


Angewandte Chemie | 2008

Heterobivalent Ligands Crosslink Multiple Cell-Surface Receptors: The Human Melanocortin-4 and δ-Opioid Receptors†

Josef Vagner; Liping Xu; Heather L. Handl; Jatinder S. Josan; David L. Morse; Eugene A. Mash; Robert J. Gillies; Victor J. Hruby

Cell-surface receptor mediated signaling is mechanistically complex. Hetero- and homo-multimerization of receptors appears to occur naturally and is a significant regulatory component of signal transduction.[1] Additionally, exogenous entities, such as cells and viruses, can interact with multiple heterologous receptors and induce clustering.[2] Such multivalent interactions are characterized by enhanced affinities (avidities) relative to monovalent binding and enhanced specificities with heteromultivalent interactions. For example, polymers containing α-MSH (α-melanocyte stimulating hormone) ligands bind with higher affinity to melanoma cells compared to monovalent α-MSH ligands.[3–5] Recapitulation of these natural phenomena using synthetic multivalent agents has been proposed for many years.[3–9] Although homomultivalent agents are known, there is little precedent for synthetic heteromultivalent targeting of cell-surface receptors. Herein we detail the synthesis and bioevaluation of heterobivalent ligands (htBVLs) targeted to two heterologous cell-surface receptors.


Clinical Cancer Research | 2012

Noninvasive Detection of Breast Cancer Lymph Node Metastasis Using Carbonic Anhydrases IX and XII Targeted Imaging Probes

Narges K. Tafreshi; Marilyn M. Bui; Kellsey Bishop; Mark C. Lloyd; Steven A. Enkemann; Alexis S. Lopez; Dominique Abrahams; Bradford W. Carter; Josef Vagner; Stephen R. Gobmyer; Robert J. Gillies; David L. Morse

Purpose: To develop targeted molecular imaging probes for the noninvasive detection of breast cancer lymph node metastasis. Experimental Design: Six cell surface or secreted markers were identified by expression profiling and from the literature as being highly expressed in breast cancer lymph node metastases. Two of these markers were cell surface carbonic anhydrase isozymes (CAIX and/or CAXII) and were validated for protein expression by immunohistochemistry of patient tissue samples on a breast cancer tissue microarray containing 47 normal breast tissue samples, 42 ductal carcinoma in situ, 43 invasive ductal carcinomas without metastasis, 46 invasive ductal carcinomas with metastasis, and 49 lymph node macrometastases of breast carcinoma. Targeted probes were developed by conjugation of CAIX- and CAXII-specific monoclonal antibodies to a near-infrared fluorescent dye. Results: Together, these two markers were expressed in 100% of the lymph node metastases surveyed. Selectivity of the imaging probes were confirmed by intravenous injection into nude mice-bearing mammary fat pad tumors of marker-expressing cells and nonexpressing cells or by preinjection of unlabeled antibody. Imaging of lymph node metastases showed that peritumorally injected probes detected nodes harboring metastatic tumor cells. As few as 1,000 cells were detected, as determined by implanting, under ultrasound guidance, a range in number of CAIX- and CAXII-expressing cells into the axillary lymph nodes. Conclusion: These imaging probes have potential for noninvasive staging of breast cancer in the clinic and elimination of unneeded surgery, which is costly and associated with morbidities. Clin Cancer Res; 18(1); 207–19. ©2011 AACR.


Molecular Imaging | 2008

Gold nanorods targeted to delta opioid receptor: plasmon-resonant contrast and photothermal agents.

Kvar C. Black; Nathaniel D. Kirkpatrick; Timothy S. Troutman; Liping Xu; Josef Vagner; Robert J. Gillies; Jennifer K. Barton; Urs Utzinger; Marek Romanowski

Molecularly targeted gold nanorods were investigated for applications in both diagnostic imaging and disease treatment with cellular resolution. The nanorods were tested in two genetically engineered cell lines derived from the human colon carcinoma HCT-116, a model for studying ligand-receptor interactions. One of these lines was modified to express delta opioid receptor (δOR) and green fluorescent protein, whereas the other was receptor free and expressed a red fluorescent protein, to serve as the control. Deltorphin, a high-affinity ligand for δOR, was stably attached to the gold nanorods through a thiol-terminated linker. In a mixed population of cells, we demonstrated selective imaging and destruction of receptor-expressing cells while sparing those cells that did not express the receptor. The molecularly targeted nanorods can be used as an in vitro ligand-binding and cytotoxic treatment assay platform and could potentially be applied in vivo for diagnostic and therapeutic purposes with endoscopic technology.


Molecular Cancer Therapeutics | 2009

Enhanced targeting with heterobivalent ligands

Liping Xu; Josef Vagner; Jatinder S. Josan; Ronald M. Lynch; David L. Morse; Brenda Baggett; Haiyong Han; Eugene A. Mash; Victor J. Hruby; Robert J. Gillies

A novel approach to specifically target tumor cells for detection and treatment is the proposed use of heteromultivalent ligands, which are designed to interact with, and noncovalently crosslink, multiple different cell surface receptors. Although enhanced binding has been shown for synthetic homomultivalent ligands, proof of cross-linking requires the use of ligands with two or more different binding moieties. As proof-of-concept, we have examined the binding of synthetic heterobivalent ligands to cell lines that were engineered to coexpress two different G-protein-coupled human receptors, i.e., the human melanocortin 4 receptor (MC4R) expressed in combination with either the human δ-opioid receptor (δOR) or the human cholecystokinin-2 receptor (CCK2R). Expression levels of these receptors were characterized by time-resolved fluorescence saturation binding assays using Europium-labeled ligands; Eu-DPLCE, Eu-NDP-α-MSH, and Eu-CCK8 for the δOR, MC4R, and CCK2R, respectively. Heterobivalent ligands were synthesized to contain a MC4R agonist connected via chemical linkers to either a δOR or a CCK2R agonist. In both cell systems, the heterobivalent constructs bound with much higher affinity to cells expressing both receptors, compared with cells with single receptors or to cells where one of the receptors was competitively blocked. These results indicate that synthetic heterobivalent ligands can noncovalently crosslink two unrelated cell surface receptors, making feasible the targeting of receptor combinations. The in vitro cell models described herein will lead to the development of multivalent ligands for target combinations identified in human cancers. [Mol Cancer Ther 2009;8(8):2356–65]


Journal of Medicinal Chemistry | 2008

A Structure–Activity Relationship Study and Combinatorial Synthetic Approach of C-Terminal Modified Bifunctional Peptides That Are δ/μ Opioid Receptor Agonists and Neurokinin 1 Receptor Antagonists

Takashi Yamamoto; Padma Nair; Josef Vagner; Tally M. Largent-Milnes; Peg Davis; Shou Wu Ma; Edita Navratilova; Sharif Moye; Suneeta Tumati; Josephine Lai; Henry I. Yamamura; Todd W. Vanderah; Frank Porreca; Victor J. Hruby

A series of bifunctional peptides with opioid agonist and substance P antagonist bioactivities were designed with the concept of overlapping pharmacophores. In this concept, the bifunctional peptides were expected to interact with each receptor separately in the spinal dorsal horn where both the opioid receptors and the NK1 receptors were found to be expressed, to show an enhanced analgesic effect, no opioid-induced tolerance, and to provide better compliance than coadministration of two drugs. Compounds were synthesized using a two-step combinatorial method for C-terminal modification. In the method, the protected C-terminal-free carboxyl peptide, Boc-Tyr( tBu)- d-Ala-Gly Phe-Pro-Leu-Trp(Boc)-OH, was synthesized as a shared intermediate using Fmoc solid phase chemistry on a 2-chlorotrityl resin. This intermediate was esterified or amidated in solution phase. The structure-activity relationships (SAR) showed that the C-terminus acted as not only a critical pharmacophore for the substance P antagonist activities, but as an address region for the opioid agonist pharmacophore that is structurally distant from the C-terminal. Among the peptides, H-Tyr- d -Ala-Gly-Phe-Pro-Leu-Trp-NH-Bzl ( 3) demonstrated high binding affinities at both delta and mu receptors ( K i = 10 and 0.65 nM, respectively) with efficient agonist functional activity in the mouse isolated vas deferens (MVD) and guinea pig isolated ileum (GPI) assays (IC 50 = 50 and 13 nM, respectively). Compound 3 also showed a good antagonist activity in the GPI assay with substance P stimulation ( K e = 26 nM) and good affinity for the hNK1 receptor ( K i = 14 nM). Consequently, compound 3 is expected to be a promising and novel type of analgesic with bifunctional activities.


Tetrahedron Letters | 2000

A solid phase traceless synthesis of quinoxalinones

Viktor Krchňák; Lajos Szabo; Josef Vagner

A solid-phase traceless synthesis of quinoxalinones in three combinatorial steps is reported. An aldehyde functionalized polystyrene resin was reductively alkylated by amino acid methyl esters, and then the resin bound secondary amines were reacted with o-fluoronitrobenzenes. The resulting o-nitroanilines were reduced by tin chloride to the dianilines, which spontaneously cyclized. The amide nitrogen of the dihydroquinoxalinones obtained was alkylated by alkyl halides. After cleavage from the resin, the dihydroquinoxalinones were air oxidized to quinoxalinones.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Heterobivalent ligands target cell-surface receptor combinations in vivo

Liping Xu; Jatinder S. Josan; Josef Vagner; Michael R. Caplan; Victor J. Hruby; Eugene A. Mash; Ronald M. Lynch; David L. Morse; Robert J. Gillies

A challenge in tumor targeting is to deliver payloads to cancers while sparing normal tissues. A limited number of antibodies appear to meet this challenge as therapeutics themselves or as drug-antibody conjugates. However, antibodies suffer from their large size, which can lead to unfavorable pharmacokinetics for some therapeutic payloads, and that they are targeted against only a single epitope, which can reduce their selectivity and specificity. Here, we propose an alternative targeting approach based on patterns of cell surface proteins to rationally develop small, synthetic heteromultivalent ligands (htMVLs) that target multiple receptors simultaneously. To gain insight into the multivalent ligand strategy in vivo, we have generated synthetic htMVLs that contain melanocortin (MSH) and cholecystokinin (CCK) pharmacophores that are connected via a fluorescent labeled, rationally designed synthetic linker. These ligands were tested in an experimental animal model containing tumors that expressed only one (control) or both (target) MSH and CCK receptors. After systemic injection of the htMVL in tumor-bearing mice, label was highly retained in tumors that expressed both, compared with one, target receptors. Selectivity was quantified by using ex vivo measurement of Europium-labeled htMVL, which had up to 12-fold higher specificity for dual compared with single receptor expressing cells. This proof-of-principle study provides in vivo evidence that small, rationally designed bivalent htMVLs can be used to selectively target cells that express both, compared with single complimentary cell surface targets. These data open the possibility that specific combinations of targets on tumors can be identified and selectively targeted using htMVLs.


Journal of Medicinal Chemistry | 2011

Development of melanoma-targeted polymer micelles by conjugation of a melanocortin 1 receptor (MC1R) specific ligand.

Natalie M. Barkey; Narges K. Tafreshi; Jatinder S. Josan; Channa R. De Silva; Kevin N. Sill; Victor J. Hruby; Robert J. Gillies; David L. Morse; Josef Vagner

The incidence of malignant melanoma is rising faster than that of any other cancer in the United States. Because of its high expression on the surface of melanomas, MC1R has been investigated as a target for selective imaging and therapeutic agents against melanoma. Eight ligands were screened against cell lines engineered to overexpress MC1R, MC4R, or MC5R. Of these, compound 1 (4-phenylbutyryl-His-dPhe-Arg-Trp-NH(2)) exhibited high (0.2 nM) binding affinity for MC1R and low (high nanomolar) affinities for MC4R and MC5R. Functionalization of the ligand at the C-terminus with an alkyne for use in Cu-catalyzed click chemistry was shown not to affect the binding affinity. Finally, formation of the targeted polymer, as well as the targeted micelle formulation, also resulted in constructs with low nanomolar binding affinity.

Collaboration


Dive into the Josef Vagner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert J. Gillies

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

David L. Morse

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Liping Xu

University of Arizona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Theodore J. Price

University of Texas at Dallas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge