Network


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

Hotspot


Dive into the research topics where Alexander Zheleznyak is active.

Publication


Featured researches published by Alexander Zheleznyak.


Molecular Cell | 1999

Ubiquitin-Related Proteins Regulate Interaction of Vimentin Intermediate Filaments with the Plasma Membrane

Ai-Ling Wu; Jun Wang; Alexander Zheleznyak; Eric J. Brown

Integrin-associated protein (IAP, CD47) is a plasma membrane receptor for thrombospondins and signal regulatory proteins (SIRPs) that has an essential role in host defense through its association with integrins. The IAP gene encodes alternatively spliced carboxyterminal cytoplasmic tails that have no previously described function. IAP cytoplasmic tails can bind two related proteins that mediate interaction between IAP and vimentin-containing intermediate filaments, named proteins linking IAP with cytoskeleton (PLICs). Integrins interact with PLICs indirectly, through IAP. Transfection of PLICs induces redistribution of vimentin and cell spreading in IAP-expressing cells. This novel connection between plasma membrane and cytoskeleton is likely to be significant in many adhesion-dependent cell functions.


ACS Nano | 2011

Evaluation of Multivalent, Functional Polymeric Nanoparticles for Imaging Applications

Monica Shokeen; Eric D. Pressly; Aviv Hagooly; Alexander Zheleznyak; Nicholas Ramos; Ashley L. Fiamengo; Michael J. Welch; Craig J. Hawker; Carolyn J. Anderson

A series of multivalent, functional polymer nanoparticles with diagnostic/imaging units and targeting ligands for molecular targeting were synthesized with the loading of the chain-end-functionalized GRGDS peptide targeting sequence (model system based on integrin α(v)β(3)) ranging from 0 to 50%. Accurate structural and functional group control in these systems was achieved through a modular approach involving the use of multiple functionalized macromonomer/monomer units combined with living free radical polymerization. In cellulo results show an increase in uptake in α(v)β(3) integrin-positive U87MG glioblastoma cells with increasing RGD loading and a possible upper limit on the effectiveness of the number of RGD peptides for targeting α(v)β(3) integrin. Significantly, this increased targeting efficiency is coupled with in vivo biodistribution results, which show decreased blood circulation and increased liver uptake with increasing RGD loading. The results demonstrate the importance of controlling ligand loading in order to achieve optimal performance for therapeutic and imaging applications for multivalent nanoparticle-based systems.


The Journal of Nuclear Medicine | 2008

Preparation and Biological Evaluation of 64Cu-CB-TE2A-sst2-ANT, a Somatostatin Antagonist for PET Imaging of Somatostatin Receptor–Positive Tumors

Thaddeus J. Wadas; Martin Eiblmaier; Alexander Zheleznyak; Christopher D. Sherman; Riccardo Ferdani; Kexian Liang; Samuel Achilefu; Carolyn J. Anderson

Recently, the somatostatin receptor subtype 2 (SSTR2) selective antagonist sst2-ANT was determined to have a high affinity for SSTR2. Additionally, 111In-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-sst2-ANT showed high uptake in an SSTR2-transfected, tumor-bearing mouse model and suggested that radiolabeled SSTR2 antagonists may be superior to agonists for imaging SSTR2-positive tumors. This report describes the synthesis and evaluation of 64Cu-CB-4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane-sst2-ANT (64Cu-CB-TE2A-sst2-ANT) as a PET radiopharmaceutical for the in vivo imaging of SSTR2-positive tumors. Methods: Receptor-binding studies were performed to determine the dissociation constant of the radiopharmaceutical 64Cu-CB-TE2A-sst2-ANT using AR42J rat pancreatic tumor cell membranes. The internalization of 64Cu-CB-TE2A-sst2-ANT was compared with that of the 64Cu-labeled agonist 64Cu-CB-TE2A-tyrosine3-octreotate (64Cu-CB-TE2A-Y3-TATE) in AR42J cells. Both radiopharmaceuticals were also compared in vivo through biodistribution studies using healthy rats bearing AR42J tumors, and small-animal PET/CT of 64Cu-CB-TE2A-sst2-ANT was performed. Results: The dissociation constant value for the radiopharmaceutical was determined to be 26 ± 2.4 nM, and the maximum number of binding sites was 23,000 fmol/mg. 64Cu-CB-TE2A-sst2-ANT showed significantly less internalization than did 64Cu-CB-TE2A-Y3-TATE at time points from 15 min to 4 h. Biodistribution studies revealed that the clearance of 64Cu-CB-TE2A-sst2-ANT from the blood was rapid, whereas the clearance of 64Cu-CB-TE2A-sst2-ANT from the liver and kidneys was more modest at all time points. Tumor-to-blood and tumor-to-muscle ratios were determined to be better for 64Cu-CB-TE2A-sst2-ANT than those for 64Cu-CB-TE2A-Y3-TATE at the later time points, although liver and kidney uptake was significantly higher. Small-animal imaging using 64Cu-CB-TE2A-sst2-ANT revealed excellent tumor-to-background contrast at 4 h after injection, and standardized uptake values remained high even after 24 h. Conclusion: The PET radiopharmaceutical 64Cu-CB-TE2A-sst2-ANT is an attractive agent, worthy of future study as a PET radiopharmaceutical for the imaging of somatostatin receptor–positive tumors.


The Journal of Nuclear Medicine | 2011

In Vitro and In Vivo Evaluation of 64Cu-Labeled SarAr-Bombesin Analogs in Gastrin-Releasing Peptide Receptor–Expressing Prostate Cancer

Kimberly A. Lears; Riccardo Ferdani; Kexian Liang; Alexander Zheleznyak; Rebecca Andrews; Christopher D. Sherman; Samuel Achilefu; Carolyn J. Anderson; Buck E. Rogers

Bombesin is a 14–amino-acid amphibian peptide that binds with high affinity to the gastrin-releasing peptide receptor (GRPR), which is overexpressed on a variety of solid tumors. It has been demonstrated that bombesin analogs can be radiolabeled with a variety of radiometals for potential diagnosis and treatment of GRPR-positive tumors. In this regard, several studies have used different chelators conjugated to the 8 C-terminal amino acids of bombesin(7-14) for radiolabeling with 64Cu. These analogs have demonstrated GRPR-specific small-animal PET of tumors but have various advantages and disadvantages. The objective of this study was to conjugate the previously described (1-N-(4-aminobenzyl)-3,6,10,13,16,19-hexaazabicyclo[6.6.6]-eicosane-1,8-diamine) (SarAr) chelator to bombesin(7-14), radiolabel the conjugate with 64Cu, and evaluate in vitro and in vivo. Methods: SarAr was synthesized as previously published and conjugated to bombesin(7-14) by solid-phase peptide synthesis using standard Fmoc chemistry. Succinic acid (SA), 8-aminooctanoic acid (Aoc), and Gly-Ser-Gly (GSG) were used as linkers between SarAr and bombesin(7-14) to yield the resulting SarAr-SA-Aoc-bombesin(7-14) and SarAr-SA-Aoc-GSG-bombesin(7-14) peptides. The unlabeled peptides were evaluated in a competitive binding assay using PC-3 prostate cancer cells and 125I-Tyr4-bombesin to determine the inhibitory concentration of 50%. The peptides were radiolabeled with 64Cu and evaluated for internalization into PC-3 cells in vitro and for in vivo tumor uptake in mice bearing PC-3 xenografts using biodistribution and small-animal PET/CT studies. Results: The competitive binding assay demonstrated that both SarAr-SA-Aoc-bombesin(7-14) and SarAr-SA-Aoc-GSG-bombesin(7-14) bound with high affinity to GRPR with an inhibitory concentration of 50% of 3.5 and 4.5 nM, respectively. Both peptides were radiolabeled with 64Cu at room temperature without further purification and demonstrated similar internalization into PC-3 cells. In vivo, the radiolabeled peptides demonstrated tumor-specific uptake (13.0 and 8.5 percentage injected dose per gram for 64Cu-SarAr-SA-Aoc-bombesin(7-14) and 64Cu-SarAr-SA-Aoc-GSG-bombesin(7-14), respectively, at 1 h) and imaging that was comparable to, or better than, that of the previously reported 64Cu-labeled bombesin analogs. The 64Cu-SarAr-SA-Aoc-GSG-bombesin(7-14) had more rapid blood clearance and lower tumor and normal-tissue uptake than 64Cu-SarAr-SA-Aoc-bombesin(7-14), resulting in similar tumor-to-blood ratios for each analog (15.1 vs. 11.3 for 64Cu-SarAr-SA-Aoc-bombesin(7-14) and 64Cu-SarAr-SA-Aoc-GSG-bombesin(7-14), respectively, at 1 h). Conclusion: These studies demonstrate that 64Cu-SarAr-SA-Aoc-bombesin(7-14) and 64Cu-SarAr-SA-Aoc-GSG-bombesin(7-14) bound with high affinity to GRPR-expressing cells and that these peptides can be used for PET of GRPR-expressing prostate cancer.


The Journal of Nuclear Medicine | 2009

Targeting the αvβ3 Integrin for Small-Animal PET/CT of Osteolytic Bone Metastases

Thaddeus J. Wadas; Hongju Deng; Jennifer E. Sprague; Alexander Zheleznyak; Katherine N. Weilbaecher; Carolyn J. Anderson

This article describes the evaluation of the radiopharmaceutical 64Cu-CB-TE2A-c(RGDyK) (64Cu-RGD) as an imaging agent for osteolytic bone metastases and their associated inflammation by targeting of the αvβ3 integrin on osteoclasts and the proinflammatory cells involved at the bone metastatic site. Methods: The 64Cu-RGD radiotracer was evaluated in the transgenic mouse expressing Tax (Tax+), which spontaneously develops osteolytic tumors throughout the vertebrae and hind limbs, using biodistribution studies and small-animal PET/CT. Histologic analysis was also performed on Tax+ mouse tails, using hematoxylin and eosin and tartrate-resistant acid phosphatase to confirm the presence of osteolytic bone lesions and the presence of osteoclasts, respectively. Additionally, a proof-of-principle study was conducted with a small group of Tax+ animals presenting with osteolytic lesions. These animals were treated with the bisphosphonate zoledronic acid and imaged with 64Cu-RGD to determine whether this radiopharmaceutical was sensitive enough to detect a response to the bisphosphonate therapy. Results: Biodistribution studies using 64Cu-RGD demonstrated that Tax+ mice between the ages of 6 and 12 mo had a greater accumulation of activity in their tail vertebrae than did the wild-type (WT) cohort (P = 0.013). Additionally, Tax+ mice between the ages of 6 and 12 mo had significantly more tracer activity associated with their tail vertebrae than did Tax+ mice older than 12 mo (P = 0.003), suggesting that earlier bone metastases cause an increased recruitment of αvβ3-expressing cells. Small-animal PET/CT with 64Cu-RGD was conducted on Tax+ and WT mice. On the basis of standardized uptake value analysis, Tax+ mice had approximately 2-fold more tail-associated activity than did WT animals (P = 0.0157). Additionally, decreases in uptake were observed in the tails of Tax+ mice after treatment with the osteoclast inhibitor zoledronic acid, and histologic analysis of Tax+ mouse-tail vertebrae revealed the presence of Tax+ tumor cells, osteoclasts, and proinflammatory cells within the bone microenvironment. Conclusion: Together, these data suggest that 64Cu-RGD has the potential to effectively image osteolytic bone metastases and monitor the physiologic changes in the bone metastatic microenvironment after osteoclast-inhibiting bisphosphonate therapy.


Molecular Pharmaceutics | 2014

Evaluation of 89Zr-pertuzumab in Breast Cancer Xenografts

Bernadette V. Marquez; Oluwatayo F. Ikotun; Alexander Zheleznyak; Brian D. Wright; Amrita Hari-Raj; Richard A. Pierce; Suzanne E. Lapi

Pertuzumab is a monoclonal antibody that binds to HER2 and is used in combination with another HER2–specific monoclonal antibody, trastuzumab, for the treatment of HER2+ metastatic breast cancer. Pertuzumab binds to an HER2 binding site distinct from that of trastuzumab, and its affinity is enhanced when trastuzumab is present. We aim to exploit this enhanced affinity of pertuzumab for its HER2 binding epitope and adapt this antibody as a PET imaging agent by radiolabeling with 89Zr to increase the sensitivity of HER2 detection in vivo. Here, we investigate the biodistribution of 89Zr-pertuzumab in HER2–expressing BT-474 and HER2–nonexpressing MDA-MB-231 xenografts to quantitatively assess HER2 expression in vivo. In vitro cell binding studies were performed resulting in retained immunoreactivity and specificity for HER2–expressing cells. In vivo evaluation of 89Zr-pertuzumab was conducted in severely combined immunodeficient mice, subcutaneously inoculated with BT-474 and MDA-MB-231 cells. 89Zr-pertuzumab was systemically administered and imaged at 7 days postinjection (p.i.) followed by terminal biodistribution studies. Higher tumor uptake was observed in BT-474 compared to MDA-MB-231 xenografts with 47.5 ± 32.9 and 9.5 ± 1.7% ID/g, respectively at 7 days p.i (P = 0.0009) and blocking studies with excess unlabeled pertuzumab showed a 5-fold decrease in BT-474 tumor uptake (P = 0.0006), confirming the in vivo specificity of this radiotracer. Importantly, we observed that the tumor accumulation of 89Zr-pertuzumab was increased in the presence of unlabeled trastuzumab, at 173 ± 74.5% ID/g (P = 0.01). Biodistribution studies correlate with PET imaging quantification using max SUV (r = 0.98, P = 0.01). Collectively, these results illustrate that 89Zr-pertuzumab as a PET imaging agent may be beneficial for the quantitative and noninvasive assessment of HER2 expression in vivo especially for patients undergoing trastuzumab therapy.


The Journal of Nuclear Medicine | 2012

Molecular Imaging of Very Late Antigen–4 (α4β1 Integrin) in the Premetastatic Niche

Monica Shokeen; Alexander Zheleznyak; Jessica M. Wilson; Majiong Jiang; Ruiwu Liu; Riccardo Ferdani; Kit S. Lam; Julie K. Schwarz; Carolyn J. Anderson

Despite advances in cancer treatment over the past few decades, metastatic disease remains the primary cause of morbidity and mortality. Recent reports suggest the formation of a “premetastatic niche” before the metastatic cascade, where niche is defined as the microenvironment for tumor cells to be able to engraft and proliferate at secondary sites. Bone marrow–derived (BMD) cells that express vascular endothelial growth factor receptor–1 and very late antigen–4 (VLA-4) have been shown to arrive at sites of metastasis to form a receptive environment for tumor cells. Here we describe experiments toward imaging of VLA-4–positive BMD cells using a high-affinity PET probe, 64Cu-labeled 11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2] hexadecane (CB-TE2A)-LLP2A. Methods: VLA-4–negative MDA-MB-231/firefly luciferase (fluc) human breast tumor cells were injected intraarterially in the left ventricle in nude mice. Tumor metastasis in mice was monitored for 30 d by bioluminescence imaging and small-animal PET/CT. Small-animal PET images were collected 2 h after mice were injected in the tail vein with 64Cu-CB-TE2A-LLP2A (5.6–11.1 MBq [150–300 μCi; specific activity, 400 μCi/μg]). Cellular uptake of 64Cu-CB-TE2A-LLP2A was determined in VLA-4–positive B16F10 mouse melanoma cells and VLA-4–negative MDA-MB-231/fluc human breast cancer tumor cells. Biodistribution experiments in nude mice bearing VLA-4–positive B16F10 subcutaneous tumors in the flank were conducted to validate targeting of VLA-4–positive cells in vivo. Results: Uptake of 64Cu-CB-TE2A-LLP2A was higher in VLA-4–positive human melanoma B16F10 cells than in VLA-4–negative MDA-MB-231 cells (P < 0.05). In B16F10 tumor–bearing mice, 64Cu-CB-TE2A-LLP2A had high uptake in the VLA-4–rich organs marrow, spleen, and tumor (11.26% ± 2.59%, 8.36% ± 2.15%, and 3.09% ± 0.58% injected dose/g, respectively). Cumulative standardized uptake value data from 2 independent studies (n = 7 and 8 mice) on nude mice implanted with VLA-4–negative MDA-MB-231/fluc human breast tumor cells suggested an influx of VLA-4–positive BMD cells that corresponded to metastasis (P < 0.05). Immunohistochemical analysis and flow cytometry also showed upregulation of VLA-4–positive cell clusters and BMD cells at the metastatic sites, providing evidence for noninvasive imaging of BMD cells in the premetastatic niche. Conclusion: The results of the study demonstrated the potential of PET with VLA-4–targeted 64Cu-CB-TE2A-LLP2A to visualize BMD cell reorganization and expansion noninvasively in vivo.


PLOS ONE | 2013

Very Late Antigen-4 (α4β1 Integrin) Targeted PET Imaging of Multiple Myeloma

Deepti Soodgupta; Michelle A. Hurchla; Majiong Jiang; Alexander Zheleznyak; Katherine N. Weilbaecher; Carolyn J. Anderson; Michael H. Tomasson; Monica Shokeen

Biomedical imaging techniques such as skeletal survey and 18F-fluorodeoxyglucose (FDG)/Positron Emission Tomography (PET) are frequently used to diagnose and stage multiple myeloma (MM) patients. However, skeletal survey has limited sensitivity as it can detect osteolytic lesions only after 30–50% cortical bone destruction, and FDG is a marker of cell metabolism that has limited sensitivity for intramedullary lesions in MM. Targeted, and non-invasive novel probes are needed to sensitively and selectively image the unique molecular signatures and cellular processes associated with MM. Very late antigen-4 (VLA-4; also called α4β1 integrin) is over-expressed on MM cells, and is one of the key mediators of myeloma cell adhesion to the bone marrow (BM) that promotes MM cell trafficking and drug resistance. Here we describe a proof-of-principle, novel molecular imaging strategy for MM tumors using a VLA-4 targeted PET radiopharmaceutical, 64Cu-CB-TE1A1P-LLP2A. Cell uptake studies in a VLA-4-positive murine MM cell line, 5TGM1, demonstrated receptor specific uptake (P<0.0001, block vs. non-block). Tissue biodistribution at 2 h of 64Cu-CB-TE1A1P-LLP2A in 5TGM1 tumor bearing syngeneic KaLwRij mice demonstrated high radiotracer uptake in the tumor (12±4.5%ID/g), and in the VLA-4 rich organs, spleen (8.8±1.0%ID/g) and marrow (11.6±2.0%ID/g). Small animal PET/CT imaging with 64Cu-CB-TE1A1P-LLP2A demonstrated high uptake in the 5TGM1 tumors (SUV 6.6±1.1). There was a 3-fold reduction in the in vivo tumor uptake in the presence of blocking agent (2.3±0.4). Additionally, 64Cu-CB-TE1A1P-LLP2A demonstrated high binding to the human MM cell line RPMI-8226 that was significantly reduced in the presence of the cold targeting agent. These results provide pre-clinical evidence that VLA-4-targeted imaging using 64Cu-CB-TE1A1P-LLP2A is a novel approach to imaging MM tumors.


Journal of drug delivery | 2012

Application of Collagen-Model Triple-Helical Peptide-Amphiphiles for CD44-Targeted Drug Delivery Systems

Margaret W Ndinguri; Alexander Zheleznyak; Janelle L. Lauer; Carolyn J. Anderson; Gregg B. Fields

Cancer treatment by chemotherapy is typically accompanied by deleterious side effects, attributed to the toxic action of chemotherapeutics on proliferating cells from nontumor tissues. The cell surface proteoglycan CD44 has been recognized as a cancer stem cell marker. The present study has examined CD44 targeting as a way to selectively deliver therapeutic agents encapsulated inside colloidal delivery systems. CD44/chondroitin sulfate proteoglycan binds to a triple-helical sequence derived from type IV collagen, α1(IV)1263–1277. We have assembled a peptide-amphiphile (PA) in which α1(IV)1263–1277 was sandwiched between 4 repeats of Gly-Pro-4-hydroxyproline and conjugated to palmitic acid. The PA was incorporated into liposomes composed of DSPG, DSPC, cholesterol, and DSPE-PEG-2000 (1 : 4 : 5 : 0.5). Doxorubicin-(DOX-)loaded liposomes with and without 10% α1(IV)1263–1277 PA were found to exhibit similar stability profiles. Incubation of DOX-loaded targeted liposomes with metastatic melanoma M14#5 and M15#11 cells and BJ fibroblasts resulted in IC50 values of 9.8, 9.3, and >100 μM, respectively. Nontargeted liposomes were considerably less efficacious for M14#5 cells. In the CD44+ B16F10 mouse melanoma model, CD44-targeted liposomes reduced the tumor size to 60% of that of the untreated control, whereas nontargeted liposomes were ineffective. These results suggest that PA targeted liposomes may represent a new class of nanotechnology-based drug delivery systems.


Nuclear Medicine and Biology | 2013

Initial characterization of a dually radiolabeled peptide for simultaneous monitoring of protein targets and enzymatic activity

Efrem Mebrahtu; Alexander Zheleznyak; Minjun A. Hur; Richard Laforest; Suzanne E. Lapi

OBJECTIVE The goal of this study was to develop dually radiolabeled peptides for simultaneous imaging of cancer cell localization by targeting the α(v)β(3) integrin and their pathophysiology by targeting the activity of the proteolytic enzyme MMP2, involved in the metastatic process. METHODS A hybrid peptide c(RGDfE)K(DOTA)PLGVRY containing an RGD motif for binding to the α(v)β(3)integrin, a metal chelator (DOTA) for radiolabeling with [(64)Cu], and the MMP2 substrate cleavage sequence PLGVRY with terminal tyrosine for labeling with [(123)I] was synthesized, labeled with [(64)Cu] and [(123)I], and evaluated in vitro as a potential imaging agent. RESULTS The peptide was synthesized and labeled with [(64)Cu] and [(123)I] with 300 and 40 μCi/μg (542 and 72.2 mCi/μmol) specific activities, respectively, and radiochemical purity of >98%. c(RGDfE)K(DOTA)PLGVRY demonstrated high affinity for α(v)β(3) integrins (Kd=83.4+13.2 nM) in both substrate competition and cell binding assays. c(RGDfE)K(DOTA)PLGVRY peptide, but not the scrambled version, c(RGDfE)K(DOTA)GRPLVY was specifically cleaved by MMP2. CONCLUSIONS These results demonstrate the feasibility of developing dually radiolabeled peptides for the simultaneous imaging of cancer cells and their pathophysiologic activity.

Collaboration


Dive into the Alexander Zheleznyak's collaboration.

Top Co-Authors

Avatar

Suzanne E. Lapi

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric J. Brown

University of California

View shared research outputs
Top Co-Authors

Avatar

Monica Shokeen

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Katherine N. Weilbaecher

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Samuel Achilefu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Thaddeus J. Wadas

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Bernadette V. Marquez

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Kexian Liang

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Majiong Jiang

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge