Network


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

Hotspot


Dive into the research topics where Vladimir Ponomarev is active.

Publication


Featured researches published by Vladimir Ponomarev.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Imaging transcriptional regulation of p53-dependent genes with positron emission tomography in vivo

Michael Doubrovin; Vladimir Ponomarev; Tatiana Beresten; Julius Balatoni; William Bornmann; Ronald D. Finn; John L. Humm; Steven M. Larson; Michel Sadelain; Ronald G. Blasberg; Juri Gelovani Tjuvajev

A noninvasive method for molecular imaging of the activity of different signal transduction pathways and the expression of different genes in vivo would be of considerable value. It would aid in understanding the role specific genes and signal transduction pathways have in various diseases, and could elucidate temporal dynamics and regulation at different stages of disease and during various therapeutic interventions. We developed and assessed a method for monitoring the transcriptional activation of endogenous genes by positron-emission tomography (PET) imaging. The HSV1-tk/GFP (TKGFP) dual reporter gene was used to monitor transcriptional activation of p53-dependent genes. A retrovirus bearing the Cis-p53/TKGFP reporter system was constructed in which the TKGFP reporter gene was placed under control of an artificial cis-acting p53-specific enhancer. U87 glioma and SaOS-2 osteosarcoma cells were transduced with this retrovirus and used to establish xenografts in rats. We demonstrated that DNA damage-induced up-regulation of p53 transcriptional activity correlated with the expression of p53-dependent downstream genes, such as p21, in U87 (wild-type p53), but not in SaOS-2 osteosarcoma (p53 −/−) cells. We showed that PET, with [124I]FIAU (2′-fluoro-2′-deoxy-1-β-d-arabinofuranosyl-5-[124I]iodouracil) and the Cis-p53TKGFP reporter system, is sufficiently sensitive to image the transcriptional regulation of genes in the p53 signal transduction pathway. These imaging results were confirmed by independent measurements of p53 activity and the expression levels of downstream genes (e.g., p21) by using conventional molecular-biological assays. PET imaging of p53 transcriptional activity in tumor xenografts by using the Cis-p53TKGFP reporter system may be useful in assessing novel therapeutic approaches.


Nature Medicine | 2007

T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection

Matthias Stephan; Vladimir Ponomarev; Renier J. Brentjens; Alex H Chang; Konstantin Dobrenkov; Glenn Heller; Michel Sadelain

To reject tumors, T cells must overcome poor tumor immunogenicity and an adverse tumor microenvironment. Providing agonistic costimulatory signals to tumor-infiltrating T cells to augment T cell function remains a challenge for the implementation of safe and effective immunotherapy. We hypothesized that T cells overexpressing selected costimulatory ligands could serve as cellular vehicles mediating powerful, yet constrained, anatomically targeted costimulation. Here, we show that primary human T cells expressing CD80 and 4-1BB ligand (4-1BBL) vigorously respond to tumor cells lacking costimulatory ligands and provoke potent rejection of large, systemic tumors in immunodeficient mice. In addition to showing costimulation of bystander T cells (transcostimulation), we show the effect of CD80 and 4-1BBL binding to their respective receptors in the immunological synapse of isolated single cells (autocostimulation). This new strategy of endowing T cells with constitutively expressed costimulatory ligands could be extended to other ligand-receptor pairs and used to enhance any targeted adoptive transfer therapy.


Cancer Research | 2004

Molecular Imaging of Temporal Dynamics and Spatial Heterogeneity of Hypoxia-Inducible Factor-1 Signal Transduction Activity in Tumors in Living Mice

Inna Serganova; Michael Doubrovin; Jelena Vider; Vladimir Ponomarev; Suren Soghomonyan; Tatiana Beresten; Ludmila Ageyeva; Alexander Serganov; Shangde Cai; Julius Balatoni; Ronald G. Blasberg; Juri G. Gelovani

Tumor hypoxia is a spatially and temporally heterogeneous phenomenon, which results from several tumor and host tissue-specific processes. To study the dynamics and spatial heterogeneity of hypoxia-inducible factor-1 (HIF-1)-specific transcriptional activity in tumors, we used repetitive noninvasive positron emission tomography (PET) imaging of hypoxia-induced HIF-1 transcriptional activity in tumors in living mice. This approach uses a novel retroviral vector bearing a HIF-1–inducible “sensor” reporter gene (HSV1-tk/GFP fusion) and a constitutively expressed “beacon” reporter gene (DsRed2/XPRT). C6 glioma cells transduced with this multireporter system revealed dose-dependent patterns in temporal dynamics of HIF-1 transcriptional activity induced by either CoCl2 or decreased atmospheric oxygen concentration. Multicellular spheroids of C6 reporter cells developed a hypoxic core when >350 μm in diameter. 18F-2′-fluoro-2′deoxy-1β-D-arabionofuranosyl-5-ethyl-uracil (FEAU) PET revealed spatial heterogeneity of HIF-1 transcriptional activity in reporter xenografts in mice as a function of size or ischemia-reperfusion injury. With increasing tumor diameter (>3 mm), a marked increase in HIF-1 transcriptional activity was observed in the core regions of tumors. Even a moderate ischemia-reperfusion injury in small C6 tumors caused a rapid induction of HIF-1 transcriptional activity, which persisted for a long time because of the inability of C6 tumors to rapidly compensate acute changes in tumor microcirculation.


Nature Biotechnology | 2008

Peptide-conjugated antisense oligonucleotides for targeted inhibition of a transcriptional regulator in vivo

Erik Henke; Jonathan Perk; Jelena Vider; Paola de Candia; Yvette Chin; David B. Solit; Vladimir Ponomarev; Luca Cartegni; Katia Manova; Neal Rosen; Robert Benezra

Transcription factors are important targets for the treatment of a variety of malignancies but are extremely difficult to inhibit, as they are located in the cells nucleus and act mainly by protein-DNA and protein-protein interactions. The transcriptional regulators Id1 and Id3 are attractive targets for cancer therapy as they are required for tumor invasiveness, metastasis and angiogenesis. We report here the development of an antitumor agent that downregulates Id1 effectively in tumor endothelial cells in vivo. Efficient delivery and substantial reduction of Id1 protein levels in the tumor endothelium were effected by fusing an antisense molecule to a peptide known to home specifically to tumor neovessels. In two different tumor models, systemic delivery of this drug led to enhanced hemorrhage, hypoxia and inhibition of primary tumor growth and metastasis, similar to what is observed in Id1 knockout mice. Combination with the Hsp90 inhibitor 17-(allylamino)-17-demethoxygeldanamycin yielded virtually complete growth suppression of aggressive breast tumors.


The Journal of Nuclear Medicine | 2007

A Human-Derived Reporter Gene for Noninvasive Imaging in Humans: Mitochondrial Thymidine Kinase Type 2

Vladimir Ponomarev; Michael Doubrovin; Aleksander Shavrin; Inna Serganova; Tatiana Beresten; Ludmila Ageyeva; Changde Cai; Julius Balatoni; Mian M. Alauddin; Juri Gelovani

A human-derived intrinsically nonimmunogenic reporter gene was tested for PET imaging of different molecular–genetic processes for potential clinical use. Methods: The human mitochondrial thymidine kinase type 2 (hTK2) reporter gene truncated at the N terminus (ΔhTK2), alone or fused with green fluorescent protein (GFP), was used for preclinical evaluation in a mouse model. The levels of enzymatic activity of ΔhTK2 and ΔhTK2 GFP proteins were assessed using radiotracer accumulation and prodrug activation assays in vitro and in subcutaneous tumors grown from the corresponding cell lines in nude mice. Kinetic analyses of 124I-2′-fluoro-2′-deoxy-1-β-d-β-arabinofuranosyl-5-iodouracil (FIAU), 18F-2′-fluoro-2′-deoxy-1-β-d-β-arabinofuranosyl-5-ethyluracil (FEAU), or 18F-9-(4-18F-fluoro-3-hydroxymethylbutyl)guanine (FHBG) uptake in tumors and biodistribution studies were performed. Results: ΔhTK2 was successfully expressed in the cytoplasm of transduced cells. A new anti-hTK2 monoclonal antibody 8G2 was developed. The levels of FIAU and FEAU accumulation in cells expressing ΔhTK2 and ΔhTK2 GFP were at least 10-fold higher than in wild-type cells in vitro and about 6 times higher in vivo. We determined that FEAU is a more specific reporter substrate for ΔhTK2 than FIAU, whereas FHBG is not phosphorylated by this enzyme. In addition, we showed that ΔhTK2 transduced cells can be eliminated by treatment with d-arabinofuranosyl-cytosine. Conclusion: We have tested a human-derived reporter gene that is likely to be nonimmunogenic and potentially allows for long-term monitoring of different molecular–genetic processes by nuclear imaging techniques in humans. Using 124I-FIAU, 18F-FIAU, or 18F-FEAU, it should be possible to image ΔhTK2 reporter gene expression with PET in preclinical and clinical studies.


Medical Physics | 2009

In vivo bioluminescence tomography with a blocking-off finite-difference SP3 method and MRI/CT coregistration.

Alexander D. Klose; Bradley J. Beattie; Hamid Dehghani; Lena Vider; Carl Le; Vladimir Ponomarev; Ronald G. Blasberg

PURPOSE Bioluminescence imaging is a research tool for studying gene expression levels in small animal models of human disease. Bioluminescence light, however, is strongly scattered in biological tissue and no direct image of the light-emitting reporter probes location can be obtained. Therefore, the authors have developed a linear image reconstruction method for bioluminescence tomography (BLT) that recovers the three-dimensional spatial bioluminescent source distribution in small animals. METHODS The proposed reconstruction method uses third-order simplified spherical harmonics (SP3) solutions to the equation of radiative transfer for modeling the bioluminescence light propagation in optically nonuniform tissue. The SP3 equations and boundary conditions are solved with a finite-difference (FD) technique on a regular grid. The curved geometry of the animal surface was taken into account with a blocking-off region method for regular grids. Coregistered computed tomography (CT) and magnetic resonance (MR) images provide information regarding the geometry of the skin surface and internal organs. The inverse source problem is defined as an algebraic system of linear equations for the unknown source distribution and is iteratively solved given multiview and multispectral boundary measurements. The average tissue absorption parameters, which are used for the image reconstruction process, were calculated with an evolution strategy (ES) from in vivo measurements using an implanted pointlike source of known location and spectrum. Moreover, anatomical information regarding the location of the internal organs and other tissue structures within the animals body are provided by coregistered MR images. RESULTS First, the authors recovered the wavelength-dependent absorption coefficients (average error of 14%) with the ES under ideal conditions by using a numerical mouse model. Next, they reconstructed the average absorption coefficient of a small animal by using an artificial implanted light source and the validated ES. Last, they conducted two in vivo animal experiments and recovered the spatial location of the implanted light source and the spatial distribution of a bioluminescent reporter system located in the kidneys. The source reconstruction results were coregistered to CT and MR images. They further found that accurate bioluminescence image reconstructions could be obtained when segmenting a voidlike cyst with low-scattering and absorption parameters, whereas inaccurate image reconstructions were obtained when assuming a uniform optical parameter distribution instead. The image reconstructions were completed within 23 min on a 3 GHz Intel processor. CONCLUSIONS The authors demonstrated on in vivo examples that the combination of anatomical coregistration, accurate optical tissue properties, multispectral acquisition, and a blocking-off FD-SP3 solution of the radiative transfer model significantly improves the accuracy of the BLT reconstructions.


The Journal of Nuclear Medicine | 2008

Monitoring the Efficacy of Adoptively Transferred Prostate Cancer–Targeted Human T Lymphocytes with PET and Bioluminescence Imaging

Konstantin Dobrenkov; Malgorzata Olszewska; Yury Likar; Larissa Shenker; Gertrude Gunset; Shangde Cai; Nagavarakishore Pillarsetty; Hedvig Hricak; Michel Sadelain; Vladimir Ponomarev

Noninvasive imaging technologies have the potential to enhance the monitoring and improvement of adoptive therapy with tumor-targeted T lymphocytes. We established an imaging methodology for the assessment of spatial and temporal distributions of adoptively transferred genetically modified human T cells in vivo for treatment monitoring and prediction of tumor response in a systemic prostate cancer model. Methods: RM1 murine prostate carcinoma tumors transduced with human prostate-specific membrane antigen (hPSMA) and a Renilla luciferase reporter gene were established in SCID/beige mice. Human T lymphocytes were transduced with chimeric antigen receptors (CAR) specific for either hPSMA or human carcinoembryonic antigen (hCEA) and with a fusion reporter gene for herpes simplex virus type 1 thymidine kinase (HSV1tk) and green fluorescent protein, with or without click beetle red luciferase. The localization of adoptively transferred T cells in tumor-bearing mice was monitored with 2′-18F-fluoro-2′-deoxy-1-β-d-arabinofuranosyl-5-ethyluracil (18F-FEAU) small-animal PET and bioluminescence imaging (BLI). Results: Cotransduction of CAR-expressing T cells with the reporter gene did not affect CAR-mediated cytotoxicity. BLI of Renilla and click beetle red luciferase expression enabled concurrent imaging of adoptively transferred T cells and systemic tumors in the same animal. hPSMA-specific T lymphocytes persisted longer than control hCEA-targeted T cells in lung hPSMA-positive tumors, as indicated by both PET and BLI. Precise quantification of T-cell distributions at tumor sites by PET revealed that delayed tumor progression was positively correlated with the levels of 18F-FEAU accumulation in tumor foci in treated animals. Conclusion: Quantitative noninvasive monitoring of genetically engineered human T lymphocytes by PET provides spatial and temporal information on T-cell trafficking and persistence. PET may be useful for predicting tumor response and for guiding adoptive T-cell therapy.


Clinical Cancer Research | 2009

Zoledronic Acid Inhibits Both the Osteolytic and Osteoblastic Components of Osteosarcoma Lesions in a Mouse Model

Agatha Labrinidis; Shelley Hay; Vasilios Liapis; Vladimir Ponomarev; David M. Findlay; Andreas Evdokiou

Purpose: To evaluate the efficacy of zoledronic acid (ZOL) against osteosarcoma (OS) growth, progression, and metastatic spread using an animal model of human OS that closely resembles the human disease. Experimental Design: Human K-HOS or KRIB OS cells, tagged or untagged with a luciferase reporter construct, were transplanted directly into the tibial cavity of nude mice. ZOL was given as weekly, or a single dose of 100 μg/kg body weight, equivalent to the 4 mg i.v. dose used clinically. Tumor growth at the primary site and as pulmonary metastases was monitored by bioluminescence imaging and histology, and OS-induced bone destruction was measured using high-resolution micro–computed tomography. Results: Mice transplanted with OS cells exhibited aberrant bone remodeling in the area of cancer cell transplantation, with areas of osteolysis mixed with extensive new bone formation extending from the cortex. ZOL administration prevented osteolysis and significantly reduced the amount of OS-induced bone formation. However, ZOL had no effect on tumor burden at the primary site. Importantly, ZOL failed to reduce lung metastasis and in some cases was associated with larger and more numerous metastatic lesions. Conclusions: Our data show that clinically relevant doses of ZOL, while protecting the bone from OS-induced bone destruction, do not inhibit primary tumor growth. Moreover, lung metastases were not reduced and may even have been promoted by this treatment, indicating that caution is required when the clinical application of the bisphosphonate class of antiresorptives is considered in OS.


Molecular Cancer Therapeutics | 2009

Apomab, a fully human agonistic antibody to DR5, exhibits potent antitumor activity against primary and metastatic breast cancer

Irene Zinonos; Agatha Labrinidis; Michelle Lee; Vasilios Liapis; Shelley Hay; Vladimir Ponomarev; Peter Diamond; Andrew C.W. Zannettino; David M. Findlay; Andreas Evdokiou

Apomab, a fully human agonistic DR5 monoclonal antibody, triggers apoptosis through activation of the extrinsic apoptotic signaling pathway. In this study, we assessed the cytotoxic effect of Apomab in vitro and evaluated its antitumor activity in murine models of breast cancer development and progression. MDA-MB-231-TXSA breast cancer cells were transplanted into the mammary fat pad or directly into the tibial marrow cavity of nude mice. Apomab was administered early, postcancer cell transplantation, or after tumors progressed to an advanced stage. Tumor burden was monitored progressively using bioluminescence imaging, and the development of breast cancer–induced osteolysis was measured using microcomputed tomography. In vitro, Apomab treatment induced apoptosis in a panel of breast cancer cell lines but was without effect on normal human primary osteoblasts, fibroblasts, or mammary epithelial cells. In vivo, Apomab exerted remarkable tumor suppressive activity leading to complete regression of well-advanced mammary tumors. All animals transplanted with breast cancer cells directly into their tibiae developed large osteolytic lesions that eroded the cortical bone. In contrast, treatment with Apomab following an early treatment protocol inhibited both intraosseous and extraosseous tumor growth and prevented breast cancer–induced osteolysis. In the delayed treatment protocol, Apomab treatment resulted in the complete regression of advanced tibial tumors with progressive restoration of both trabecular and cortical bone leading to full resolution of osteolytic lesions. Apomab represents a potent immunotherapeutic agent with strong activity against the development and progression of breast cancer and should be evaluated in patients with primary and metastatic disease. [Mol Cancer Ther 2009;8(10):2969–80]


Nature Biotechnology | 2015

Therapeutic bispecific T-cell engager antibody targeting the intracellular oncoprotein WT1.

Tao Dao; Dmitry Pankov; Andrew M. Scott; Tatyana Korontsvit; Victoriya Zakhaleva; Yiyang Xu; Jingyi Xiang; Su Yan; Manuel Guerreiro; Nicholas Veomett; Leonid Dubrovsky; Michael J. Curcio; Ekaterina Doubrovina; Vladimir Ponomarev; Cheng Liu; Richard J. O'Reilly; David A. Scheinberg

Intracellular tumor antigens presented on the cell surface in the context of human leukocyte antigen (HLA) molecules have been targeted by T cell–based therapies, but there has been little progress in developing small-molecule drugs or antibodies directed to these antigens. Here we describe a bispecific T-cell engager (BiTE) antibody derived from a T-cell receptor (TCR)-mimic monoclonal antibody (mAb) ESK1, which binds a peptide derived from the intracellular oncoprotein WT1 presented on HLA-A*02:01. Despite the very low density of the complexes at the cell surface, ESK1-BiTE selectively activated and induced proliferation of cytolytic human T cells that killed cells from multiple leukemias and solid tumors in vitro and in mice. We also discovered that in an autologous in vitro setting, ESK1-BiTE induced a robust secondary CD8 T-cell response specific for tumor-associated antigens other than WT1. Our study provides an approach that targets tumor-specific intracellular antigens without using cell therapy and suggests that epitope spreading could contribute to the therapeutic efficacy of this BiTE.

Collaboration


Dive into the Vladimir Ponomarev's collaboration.

Top Co-Authors

Avatar

Ronald G. Blasberg

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Inna Serganova

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael Doubrovin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Larissa Shenker

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shelley Hay

University of Adelaide

View shared research outputs
Top Co-Authors

Avatar

Jelena Vider

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shangde Cai

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge