Network


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

Hotspot


Dive into the research topics where Ivan D. Horak is active.

Publication


Featured researches published by Ivan D. Horak.


Molecular Cancer Therapeutics | 2006

Asparagine synthetase as a causal, predictive biomarker for l-asparaginase activity in ovarian cancer cells

Philip L. Lorenzi; William C. Reinhold; Martina Rudelius; Michele Gunsior; Uma Shankavaram; Kimberly J. Bussey; Uwe Scherf; Gabriel S. Eichler; Scott E. Martin; Koei Chin; Joe W. Gray; Elise C. Kohn; Ivan D. Horak; Daniel D. Von Hoff; Mark Raffeld; Paul Goldsmith; Natasha J. Caplen; John N. Weinstein

l-Asparaginase (l-ASP), a bacterial enzyme used since the 1970s to treat acute lymphoblastic leukemia, selectively starves cells that cannot synthesize sufficient asparagine for their own needs. Molecular profiling of the NCI-60 cancer cell lines using five different microarray platforms showed strong negative correlations of asparagine synthetase (ASNS) expression and DNA copy number with sensitivity to l-ASP in the leukemia and ovarian cancer cell subsets. To assess whether the ovarian relationship is causal, we used RNA interference to silence ASNS in three ovarian lines and observed 4- to 5-fold potentiation of sensitivity to l-ASP with two of the lines. For OVCAR-8, the line that expresses the least ASNS, the potentiation was >500-fold. Significantly, that potentiation was >700-fold in the multidrug-resistant derivative OVCAR-8/ADR, showing that the causal relationship between ASNS expression and l-ASP activity survives development of classical multidrug resistance. Tissue microarrays confirmed low ASNS expression in a subset of clinical ovarian cancers as well as other tumor types. Overall, this pharmacogenomic/pharmacoproteomic study suggests the use of l-ASP for treatment of a subset of ovarian cancers (and perhaps other tumor types), with ASNS as a biomarker for patient selection. [Mol Cancer Ther 2006;5(11):2613–23]


Cancer Research | 2011

ErbB3 Ablation Impairs PI3K/Akt-Dependent Mammary Tumorigenesis

Rebecca S. Cook; Joan T. Garrett; Violeta Sanchez; Jamie C. Stanford; Christian D. Young; Anindita Chakrabarty; Cammie Rinehart; Yixian Zhang; Yaming Wu; Lee M. Greenberger; Ivan D. Horak; Carlos L. Arteaga

The ErbB receptor family member ErbB3 has been implicated in breast cancer growth, but it has yet to be determined whether its disruption is therapeutically valuable. In a mouse model of mammary carcinoma driven by the polyomavirus middle T (PyVmT) oncogene, the ErbB2 tyrosine kinase inhibitor lapatinib reduced the activation of ErbB3 and Akt as well as tumor cell growth. In this phosphatidylinositol-3 kinase (PI3K)-dependent tumor model, ErbB2 is part of a complex containing PyVmT, p85 (PI3K), and ErbB3, that is disrupted by treatment with lapatinib. Thus, full engagement of PI3K/Akt by ErbB2 in this oncogene-induced mouse tumor model may involve its ability to dimerize with and phosphorylate ErbB3, which itself directly binds PI3K. In this article, we report that ErbB3 is critical for PI3K/Akt-driven tumor formation triggered by the PyVmT oncogene. Tissue-specific, Cre-mediated deletion of ErbB3 reduced Akt phosphorylation, primary tumor growth, and pulmonary metastasis. Furthermore, EZN-3920, a chemically stabilized antisense oligonucleotide that targets the ErbB3 mRNA in vivo, produced similar effects while causing no toxicity in the mouse model. Our findings offer further preclinical evidence that ErbB3 ablation may be therapeutically effective in tumors where ErbB3 engages PI3K/Akt signaling.


Clinical Cancer Research | 2010

Tumor Regression and Curability of Preclinical Neuroblastoma Models by PEGylated SN38 (EZN-2208), a Novel Topoisomerase I Inhibitor

Fabio Pastorino; Monica Loi; Puja Sapra; Pamela Becherini; Michele Cilli; Laura Emionite; Domenico Ribatti; Lee M. Greenberger; Ivan D. Horak; Mirco Ponzoni

Purpose: Treatment of neuroblastoma is successful in less than half of patients with high-risk disease. The antitumor activity of a water soluble pegylated SN38 drug conjugate, EZN-2208, was compared with CPT-11 (a prodrug for SN38) in preclinical models of human neuroblastoma. Experimental Design: The in vitro cytotoxicity of EZN-2208 was tested by counting trypan blue dye– and Annexin V–positive cells, whereas its therapeutic efficacy was evaluated, in terms of survival, and antitumor and antiangiogenic activities, in s.c. luciferase-transfected, pseudometastatic, and orthotopic neuroblastoma animal models. Results: EZN-2208 was about 100-fold more potent than CPT-11 in vitro, by inducing apoptosis/necrosis and p53 expression and by reducing hypoxia-inducible factor (HIF)-1α/HIF-2α expression. EZN-2208 gave superior antitumor effects compared with CPT-11 in neuroblastoma xenografts. EZN-2208 treatment always resulted in lack of tumor detection at the end of trials whereas only small therapeutic effects were observed with CPT-11, as assessed by luciferase assay or tumor size, or even by staining histologic sections of tumors with antibodies recognizing neuroblastoma cells and cell proliferation. In a neuroblastoma model resistant to doxorubicin, cisplatin, vincristine, fenretinide, and topotecan, EZN-2208 induced 100% curability. It also blocked tumor relapse after topotecan-vincristine-doxorubicin combined treatment. Mechanistic experiments showed statistically significantly enhanced terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling and Histone H2ax staining as well as decreased vascular endothelial growth factor, CD31, matrix metalloproteinase (MMP)-2, and MMP-9 expression in tumors removed from EZN-2208–treated mice and radiating vessels invading the tumor implanted onto the chorioallantoic membranes. Conclusions: EZN-2208 should be considered a most promising novel antineuroblastoma agent. An ongoing phase I study in pediatric patients should identify the optimal dose for a phase II study. Clin Cancer Res; 16(19); 4809–21. ©2010 AACR.


Nucleosides, Nucleotides & Nucleic Acids | 2010

Down-modulation of survivin expression and inhibition of tumor growth in vivo by EZN-3042, a locked nucleic acid antisense oligonucleotide.

Puja Sapra; Maoliang Wang; Raj Bandaru; Hong Zhao; Lee M. Greenberger; Ivan D. Horak

Survivin plays an important role in preventing apoptosis and permitting mitosis, and is highly expressed in various human cancers. EZN-3042 is a locked nucleic acid antisense oligonucleotide (LNA-AsODN) against survivin. We report the effects of EZN-3042 in animal models. In a chemical-induced liver regeneration model, treatment with a mouse homolog of EZN-3042 resulted in 80% down-modulation of survivin mRNA. In A549 and Calu-6 lung xenograft models, treatment with EZN-3042 single agent induced 60% down-modulation of survivin mRNA in tumors and 37–45% tumor growth inhibition (TGI). In Calu-6 model, when EZN-3042 was combined with paclitaxel, an 83% TGI was obtained. EZN-3042 is currently being evaluated in a Phase 1 clinical trial as a single agent and in combination with docetaxel.


Molecular Cancer Therapeutics | 2013

Downregulation of HER3 by a Novel Antisense Oligonucleotide, EZN-3920, Improves the Antitumor Activity of EGFR and HER2 Tyrosine Kinase Inhibitors in Animal Models

Yaming Wu; Yixian Zhang; Maoliang Wang; Qi Li; Zhengxing Qu; Victoria Shi; Patricia Kraft; Steve Kim; Ying Gao; Jenny Pak; Stephen Youngster; Ivan D. Horak; Lee M. Greenberger

Among the four human EGF receptor (HER) family members (EGFR, HER2, HER3, HER4), HER3 is of particular interest as it interacts with HER2 and EGFR via heterodimerization and is a key link to the phosphoinositide 3-kinase (PI3K)/AKT signal transduction axis. Recent studies indicate that HER3 plays a critical role in mediating resistance to agents that target EGFR or HER2. As HER3 lacks significant kinase activity and cannot be inhibited by tyrosine kinase inhibitors, neutralizing antibodies and alternative inhibitors of HER3 have been sought as cancer therapeutics. We describe here a locked nucleic acid (LNA)-based HER3 antisense oligonucleotide, EZN-3920, that specifically downmodulated the expression of HER3, which was associated with growth inhibition. EZN-3920 effectively downmodulated HER3 expression, HER3-driven PI3K/AKT signaling pathway, and growth in tumors derived from BT474M1 breast and HCC827 lung carcinoma cell lines, which overexpress HER2 and EGFR, respectively. Furthermore, when EZN-3920 was coadministered with gefitinib or lapatinib in xenograft tumor models, enhanced antitumor activity compared with the effect of monotherapy was found. The effect was associated with a blockade of induced HER3 mRNA expression caused by lapatinib or gefitinib treatment. Finally, EZN-3920 sustained its antiproliferative effect in trastuzumab-resistant cells and three independently derived gefitinib-resistant cells. Our findings show that downmodulation of HER3 by EZN-3920 leads to the suppression of tumor growth in vitro and in vivo, suggesting that HER3 can be an effective target for the treatment of various cancers that have been activated by HER3 alone or where HER3 activation is associated with EGFR or HER2 expression. Mol Cancer Ther; 12(4); 427–37. ©2013 AACR.


Mutation Research-dna Repair | 1996

Gene-specific repair in human CD4+ lymphocytes reflects transcription and proliferation

Zdena Bartośova; Miroslav Pirśel; William C. Reinhold; Maryalice Stetler-Stevenson; Maria Zajac-Kaye; Alfred May; Ivan D. Horak; Vilhelm A. Bohr

We have measured the gene-specific repair of ultraviolet irradiation (UV)-induced cyclobutane pyrimidine dimers (CPD) in freshly isolated human peripheral blood CD4+ T-lymphocytes. Two populations of CD4+ lymphocytes were assayed: resting and proliferating cells. DNA repair was assessed in the essential gene dihydrofolate reductase (DHFR) as well as in each of its strands, in the proliferation inducible c-myc gene and in the inactive delta-globin gene. Transcription rates in these genes were determined by nuclear run-on assay in the two cell populations. The rate of DHFR transcription increased 10-fold from resting to proliferating lymphocytes. Transcripts from c-myc were present only in proliferating cells, and we detected no delta-globin transcripts in either cell population. During the 24-h period after UV irradiation, there was little or no repair in any of the genes in the resting cells; there was some repair in the transcribed strand of the DHFR gene, but no repair in its nontranscribed strand. In the proliferating cells where the transcription of DHFR was much increased, the repair was efficient. The delta-globin gene was not expressed in either cell population, but it was more efficiently repaired in the proliferating than in the resting cells. We suggest that the gene-specific repair activity in CD4+ lymphocytes can reflect the proliferative state of the cells as well as the transcriptional state of the gene.


Cancer Research | 2011

Abstract SY31-02: Locked nucleic acid (LNA)-modified antisense oligonucleotides as anticancer agents: Using high-affinity antisense molecules in the laboratory and in the clinic

Arthur A. Levin; Lee M. Greenberger; Aby Buchbinder; Bo Hansen; Maj Hedtjärn; Sakari Kauppinen; Henrik Oerum; Ivan D. Horak

The inhibition of cellular processes associated with the malignant pheonotype is the goal many oncolytics. In the antisense oligonucleotide (ASO) approach, Watson and Crick base-pairing rules serve as the basis of rational drug design to create single-stranded oligonucleotides that are complementary and bind to RNAs critical for the malignant phenotype. The targets for antisense approaches can be mRNAs that encode for disease related proteins like those that control tumor growth, cell division, or survival. More recently, RNA targets in oncology have been expanded to include microRNAs. A successful oncolytic oligonucleotide must effectively bind and inhibit a target mRNA or miRNA that is critical for the malignant transformation or survival. Target identification presents the same challenge in antisense therapeutics as any other class of oncolytics, but drug design is based the known sequence of the target RNA. The first clinical trials to use antisense oligonucleotides in oncology produced only modest efficacy in clinical trials, but the therapeutic potential of these initial antisense drugs may have been hampered by their low stability and low binding affinity. Advancements in oligonucleotide chemistry have produced oligonucleotides with increased stability and increased affinities. This talk will focus on the use of oligonucleotides with the locked nucleic acid (LNA) modification. In these antisense constructs, some of the nucleotides in the sequence have a modification of the ribose sugar that includes a methylene bridge between the 2’ and 4’ positions. This bridge functions to lock the ribose into a (C3’-endo) configuration: a configuration that is optimized for binding to its cognate nucleotide. LNA-modified oligonucleotides bind to their target RNAs with higher affinities than most other oligonucleotide chemistries. One measure of affinity is the melting temperature (T m ) for binding to its complementary sequence. For each LNA-modified nucleotide added to an ASO sequence, T m can increase T m by over 5°. For example, an LNA-modified oligonucleotide currently in clinical development, miravirsen (SPC3649), with 9 LNA-modified nucleotides has a T m of approximately 80° demonstrating that LNA-modified oligonucleotides have sufficient affinities to bind to and inhibit RNA function. Affinities like these have translated into increase potency for the inhibition of target RNAs in nonclinical models and should yield therapeutic benefit more robust than those seen with earlier generations of oligonucleotide therapeutics. Other drug-like properties of antisense therapeutics have also been improved by including LNA-modified nucleotides. For example, the pharmacokinetic properties of antisense oligonucleotides support their use in clinical medicine. Antisense oligonucleotides are single stranded and bind to proteins in circulation and on cell surfaces. After parenteral administration, antisense oligonucleotide are bound first to plasma proteins, limiting glomerular filtration, and then later the antisense oligonucleotides appear bound to cell surface (proteins), allowing them to be internalized in cells. Delivery into cells and tissues is achieved without the need for formulations more complex than just aqueous solutions. LNA modifications increase the metabolic stability in both circulation and inside of cells resulting in stable tissue concentrations and prolonged activities. This long tissue residence and stability, allows for constant exposure in tumor cells with dosing as infrequent as weekly or fortnightly. Again, this represents an improvement over antisense drugs used in previous oncology trials. Using in vitro and in vivo laboratory models it has been possible to demonstrate that treatment with LNA-modified oligonucleotides produces reductions in target gene expression and ultimately tumor growth that are sequence-, concentration-, and duration-of-therapy-dependent. Taken together the properties of LNA-modified oligonucleotides make them excellent candidates for inhibiting key RNA targets and at this time there are multiple LNA-modified oligonucleotides in clinical trials in oncology. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr SY31-02. doi:10.1158/1538-7445.AM2011-SY31-02


Cancer Research | 2011

Abstract 232: Targeting HER3 mRNA by a locked nucleic antisense molecule enhances the antitumor activity of gefitinib in vivo

Yaming Wu; Maoliang Wang; Patricia Kraft; Zhengxing Qu; Yixian Zhang; Victoria Shi; Lee M. Greenberger; Ivan D. Horak

Background: The HER family consists of four tyrosine kinase receptors designated as EGFR, HER2, HER3 and HER4. HER3 is a unique member that plays a critical role in tumor growth since it 1) can heterodimerize with EGFR or HER2 and is the main link to the downstream PI3K/Akt signaling axis, 2) can mediate resistance to HER1/2-targeted therapeutics, and 3) unlike EGFR or HER2, is not typically overexpressed but still hyperphosphorylated in a variety of tumors. Resistance to the EGFR-tyrosine kinase inhibitors, such as gefitinib, can be associated with EGFR mutations and/or activation of HER3 ultimately leading to activation of the PI3K/AKT axis. We have used an RNA antagonist of HER3, designated EZN-3920, to explore the ability of this compound to inhibit tumor growth and overcome resistance to HER1/2 therapeutics. Methods: Tissue culture cells were treated with EZN-3920, a locked nucleic acid (LNA)-based oligonucleotide complementary to HER3. Compound was either added to tissue culture media (i.e. no transfection) in vitro or prepared in saline and given IV in vivo. Endpoints were measured by qRT-PCR, MTT, Western Blot analysis, Immunohistochemistry, and tumor size. Results: EZN-3920 down modulated HER3 mRNA, protein expression, PI3K/AKT signaling, and inhibited tumor cell proliferation. In vivo, systemic administration of EZN-3920, prepared in saline, resulted in specific down- modulation of HER3 mRNA and protein expression, as well as blockade in PI3K/AKT signaling pathways in NSCLC HCC827 associated with tumor growth inhibition. Similar results were also shown in tumor derived from BT-474-M1 breast carcinoma xenograft models. Interestingly, in the HCC827 tumor model, EZN-3920 at 30 mg/kg (biweekly for 4 weeks, i.v.) and gefitinib at 15 mg/kg (5 times a week for two weeks) completely shrank established tumors while either treatment alone only delayed tumor growth by 40-60%. Conclusions: The studies suggest that down regulation of HER3 by antisense molecule EZN-3920 inhibits human tumor growth in mice. Beyond this, antitumor effects of gefitinib can be enhanced by HER3 down modulation in gefitinib-sensitive tumors. On-going studies will determine if tumors that are resistant to HER1/2 therapeutics will either have enhanced sensitivity to EZN-3920 and/or the compound can restore sensitivity to such agents. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 232. doi:10.1158/1538-7445.AM2011-232


Cancer Research | 2011

Abstract 5394: Dual inhibition of the androgen receptor by ligand blockade and antisense-mediated downregulation is associated with synergistic antitumor activity model of prostate cancer

Yixian Zhang; Melissa Dumble; Stephen Castaneda; Mary Milesky; Steven Kim; Zhengxing Qu; Lee M. Greenberger; Ivan D. Horak

Background: Although new agents to treat prostate cancer have recently been approved, and new investigational drugs (Abiraterone and MDV3100) that mediate hormone deprivation show promising data from late-stage clinical trials, these therapies typically extend overall survival up to 4 months in patients with advanced disease who have failed conventional androgen deprivation therapy. Hence, there continues to be great medical need. EZN-4176 is a novel LNA-based antisense oligonucleotide (EZN-4176) that down regulates androgen receptor (AR) expression. Previously, antitumor activity of EZN-4176 in multiple xenograft models including both androgen-sensitive and castrate-resistant tumors was shown. More importantly, a synergistic effect was also found when EZN-4176 was combined with MDV3100. Further exploration of the mechanistic underpinning of the effect was explored here. Methods: The mRNA, growth, luciferase activity, protein, and prostate specific antigen (PSA) were evaluated by qRT-PCR, MTT, SteadyGlo, western blot analysis, and ELISA assay, respectively. The effect of EZN-4176 on AR transcriptional activity was evaluated in cells stably express luciferase gene regulated by AR. In vivo, the effect of EZN-4176 on AR transcriptional activity was evaluated in a bone tumor model developed by intratibia injection of C4-2b-AR-luc cells. Results: Combination of EZN-4176 with MDV3100 resulted in a much improved inhibitory effect in colony formation assay in vitro. This effect was not associated with enhanced efficiency of AR mRNA down-modulation as treatment with EZN-4176 with or without MDV3100 showed similar effects on the level of AR mRNA. However, when AR transcriptional activity was examined, we found that the combined effect was greater than each agent alone. In C4-2b-AR-luc bone tumor model, EZN-4176 potently and specifically down-modulated AR-luciferase reporter activity. Further examinations of the anti-tumor effect in this model will help understand the potential of the combinatorial effect in a highly relevant disease model. Conclusions: Our preliminary data suggest that the synergistic anti-tumor effect of EZN-4176 in combination with MDV3100 is in part associated with much reduced transcriptional activity of AR. EZN-4176 alone or in combination with anti-androgens offers great potential in treating patients with advanced prostate cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5394. doi:10.1158/1538-7445.AM2011-5394


Cancer Research | 2010

Abstract 2645: Customized PEG linkers improve the pharmaceutical properties of cytotoxic small molecules

Hong Zhao; Dechun Wu; Jing Xia; Yoany Lozanguiez; Syed M. Ali; Snehlata Tripathi; Prakash Sai; Charles D. Conover; Lee M. Greenberger; Ivan D. Horak

Introduction: PEGylation is an established delivery technology for proteins with benefits such as decreased immunogenicity and prolonged circulating half-life. The application of PEGylation to small molecules may improve the poor pharmaceutical properties including poor solubility, suboptimal pharmacokinetic (PK) profiles, and unwanted toxicities. Here we utilized Customized PEG Linkers to enhance the therapeutic index of several cytotoxic agents including doxorubicin, ara-C, gemcitabine, and SN38. Experimental procedures: drug molecules were either reacted with proper linker moieties first before PEGylation, or conjugated directly with PEG linkers. For those compounds thathave more than two reactive functional groups, the unwanted reactive sites were first protected before conjugation with PEG linkers. The PEG conjugates were incubated in PBS and plasma to study their stability, followed by in vivo PK studies in mice. Additionally, in vitro anti-proliferation assays with different human cancer cell lines were conducted to evaluate the PEG conjugates with different half-lives. Furthermore, the PEG conjugates were evaluated in vivo in multiple human cancer models in mice for their anticancer activities. Summary of data: a series of novel PEG conjugates were synthesized with different Customized PEG Linkers. In general, while these PEG conjugates were stable in PBS buffer, they demonstrated a broad range of half-lives in rat and human plasma, varying from minutes to days. PEG conjugates with longer half-lives have shown prolonged circulation time and increased AUC compared to native drugs in the PK studies in mice. In addition, PEGylation also greatly increased the water solubility for those insoluble molecules, for instance by about 1000-fold for SN38. In the cellular based studies, PEG conjugates showed different degrees of anticancer activities against a panel of human cancer cells. Remarkably, enhanced anticancer activities compared to the non-pegylated agent have been observed for many of the conjugates in a variety of cancer models, including both solid tumors and hematological malignancies. Conclusions: Customized PEG Linkers have greatly improved the solubility of certain small molecule drugs and enabled the systemic administration of these drugs. It allowed us to generate a variety of PEG-drug conjugates with broad half-life ranges in plasma which are clinically relevant. The combined effect of increased solubility, optimized PK profile, and passive accumulation of macromolecular PEG conjugates in tumors due to the enhanced permeation and retention effect may all contributed to the greatly enhanced anticancer efficacy of these PEG conjugates in animal models. In summary, customized PEG Linkers represent a promising technology to improve current cytotoxic agents. One agent, PEG-SN38 is undergoing Phase II evaluation in cancer patients with metastatic colorectal and breast cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2645.

Collaboration


Dive into the Ivan D. Horak's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Zhao

Enzon Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge