104 results on '"Pytowski B"'
Search Results
2. Blockade of VEGF receptor-3 ameliorates VEGF-A induced chronic cutaneous inflammation.: 010
- Author
-
Young, H, Halin, C, Janes, L, Min, M, Pytowski, B, Griffiths, C, and Detmar, M
- Published
- 2005
3. The anti-VEGFR3 antibody mF4-31C1 can inhibit inflammatory corneal lymphangiogenesis
- Author
-
Onderka, J, Bock, F, Dietrich, T, Pytowski, B, Kruse, FE, and Cursiefen, C
- Subjects
ddc: 610 - Published
- 2006
4. Anti-VEGF agents confer survival advantages to tumor-bearing mice by improving cancer-associated systemic syndrome
- Author
-
Xue, Y., Religa, P., Cao, R., Hansen, A. J., Lucchini, Franco, Jones, B., Wu, Y., Zhu, Z., Pytowski, B., Liang, Y., Zhong, W., Vezzoni, P., Rozell, B., Cao, Y., Lucchini, Franco (ORCID:0000-0003-0280-7062), Xue, Y., Religa, P., Cao, R., Hansen, A. J., Lucchini, Franco, Jones, B., Wu, Y., Zhu, Z., Pytowski, B., Liang, Y., Zhong, W., Vezzoni, P., Rozell, B., Cao, Y., and Lucchini, Franco (ORCID:0000-0003-0280-7062)
- Abstract
The underlying mechanism by which anti-VEGF agents prolong cancer patient survival is poorly understood. We show that in a mouse tumor model, VEGF systemically impairs functions of multiple organs including those in the hematopoietic and endocrine systems, leading to early death. Anti-VEGF antibody, bevacizumab, and anti-VEGF receptor 2 (VEGFR-2), but not anti-VEGFR-1, reversed VEGF-induced cancer-associated systemic syndrome (CASS) and prevented death in tumor-bearing mice. Surprisingly, VEGFR2 blockage improved survival by rescuing mice from CASS without significantly compromising tumor growth, suggesting that “off-tumor” VEGF targets are more sensitive than the tumor vasculature to anti-VEGF drugs. Similarly, VEGF-induced CASS occurred in a spontaneous breast cancer mouse model overexpressing neu. Clinically, VEGF expression and CASS severity positively correlated in various human cancers. These findings define novel therapeutic targets of anti-VEGF agents and provide mechanistic insights into the action of this new class of clinically available anti-VEGF cancer drugs.
- Published
- 2008
5. Der anti-VEGFR3 Antikörper mF2-31C1 inhibiert inflammatorisch induzierte Lymphangiogenese in der Hornhaut
- Author
-
Onderka, J, Bock, F, Dietrich, T, Pytowski, B, Kruse, FE, Cursiefen, C, Onderka, J, Bock, F, Dietrich, T, Pytowski, B, Kruse, FE, and Cursiefen, C
- Published
- 2006
6. 34 Efficacy of VEGFR2 Targeted mAb Therapy in Preclinical Colorectal Cancer Model Resistant to Anti-VEGF mAb Therapy
- Author
-
Surguladze, D., primary, Malaby, J., additional, Prewett, M., additional, Duignan, I., additional, Rutstein, M., additional, Yao, Y.M., additional, Schwartz, J.D., additional, and Pytowski, B., additional
- Published
- 2012
- Full Text
- View/download PDF
7. 354 Blocking VEGF Receptors 1 and 2 Prevents Inflammatory Response and Experimental Obliterative Airway Disease
- Author
-
Krebs, R., primary, Tikkanen, J., additional, Hollmén, M., additional, Wu, Y., additional, Pytowski, B., additional, and Lemström, K., additional
- Published
- 2012
- Full Text
- View/download PDF
8. Phase I study of the anti-VEGFR-3 monoclonal antibody IMC-3C5 in subjects with advanced solid tumors refractory to standard therapy or for which no standard therapy is available.
- Author
-
Martell, R. E., primary, Chiorean, E. G., additional, Youssoufian, H., additional, Rutstein, M. D., additional, Pytowski, B., additional, Benjamin, L., additional, Abad, L., additional, Patel, J., additional, Steele, T., additional, Dowd, M., additional, Qin, A., additional, Kukel, C. F., additional, Fox, F. E., additional, Dontabhaktuni, A., additional, and Morozov, A., additional
- Published
- 2011
- Full Text
- View/download PDF
9. Differentiation between the EGFR antibodies necitumumab, cetuximab, and panitumumab: Antibody internalization and EGFR degradation.
- Author
-
Topper, M. B., primary, Tonra, J. R., additional, Pytowski, B., additional, and Eastman, S. W., additional
- Published
- 2011
- Full Text
- View/download PDF
10. BLOCKING VEGF RECEPTORS 1 AND 2 PREVENTS INFLAMMATORY RESPONSE AND EXPERIMENTAL OBLITERATIVE AIRWAY DISEASE
- Author
-
Krebs, R., primary, Tikkanen, J., additional, Hollmé, M., additional, Wu, Y., additional, Pytowski, B., additional, Koskinen, P., additional, and Lemström, K., additional
- Published
- 2010
- Full Text
- View/download PDF
11. 454: Targeting Lymphatic Vessel CCL21 Production by VEGFR-3 Inhibition Prevents Cardiac Allograft Rejection and Arteriosclerosis
- Author
-
Nykanen, A.I., primary, Sandelin, H., additional, Krebs, R., additional, Keranen, M.A.I., additional, Tuuminen, R., additional, Karpanen, T., additional, Wu, Y., additional, Pytowski, B., additional, Koskinen, P.K., additional, Yla-Herttuala, S., additional, Alitalo, K., additional, and Lemstrom, K.B., additional
- Published
- 2009
- Full Text
- View/download PDF
12. VEGF receptor-2 (VEGFR-2) and VEGFR-3 mediate peri-implantation uterine angiogenesis during early pregnancy development
- Author
-
Douglas, N.C., primary, Tang, H., additional, Gomez, R., additional, Pytowski, B., additional, Sauer, M.V., additional, and Zimmermann, R.C., additional
- Published
- 2007
- Full Text
- View/download PDF
13. P-551
- Author
-
Douglas, N.C., primary, Tan, H., additional, Gomez, R., additional, Pytowski, B., additional, Sauer, M.V., additional, and Zimmermann, R.C., additional
- Published
- 2006
- Full Text
- View/download PDF
14. Complete and Specific Inhibition of Adult Lymphatic Regeneration by a Novel VEGFR-3 Neutralizing Antibody
- Author
-
Pytowski, B., primary, Goldman, J., additional, Persaud, K., additional, Wu, Y., additional, Witte, L., additional, Hicklin, D. J., additional, Skobe, M., additional, Boardman, K. C., additional, and Swartz, M. A., additional
- Published
- 2005
- Full Text
- View/download PDF
15. The Effects of Flk-2/flt3 Ligand as Compared with c-kit Ligand on Short-Term and Long-Term Proliferation of CD34+ Hematopoietic Progenitors Elicited from Human Fetal Liver, Umbilical Cord Blood, Bone Marrow, and Mobilized Peripheral Blood
- Author
-
SHAPIRO, F., primary, PYTOWSKI, B., additional, RAFII, S., additional, WITTE, L., additional, HICKLIN, D.J., additional, YAO, T.J., additional, and MOORE, M.A.S., additional
- Published
- 1996
- Full Text
- View/download PDF
16. Endosome acidification and receptor trafficking: bafilomycin A1 slows receptor externalization by a mechanism involving the receptor's internalization motif.
- Author
-
Johnson, L S, primary, Dunn, K W, additional, Pytowski, B, additional, and McGraw, T E, additional
- Published
- 1993
- Full Text
- View/download PDF
17. Vascular endothelial growth factor receptor-1 in human cancer: concise review and rationale for development of IMC-18F1 (Human antibody targeting vascular endothelial growth factor receptor-1)
- Author
-
Schwartz JD, Rowinsky EK, Youssoufian H, Pytowski B, and Wu Y
- Published
- 2010
- Full Text
- View/download PDF
18. Mutagenesis of the human transferrin receptor: two cytoplasmic phenylalanines are required for efficient internalization and a second-site mutation is capable of reverting an internalization-defective phenotype.
- Author
-
McGraw, T E, primary, Pytowski, B, additional, Arzt, J, additional, and Ferrone, C, additional
- Published
- 1991
- Full Text
- View/download PDF
19. Fc and C3bi receptors and the differentiation antigen BH2-Ag are randomly distributed in the plasma membrane of locomoting neutrophils.
- Author
-
Pytowski, B, primary, Maxfield, F R, additional, and Michl, J, additional
- Published
- 1990
- Full Text
- View/download PDF
20. Identification of the residues in the extracellular region of KDR important for interaction with vascular endothelial growth factor and neutralizing anti-KDR antibodies.
- Author
-
Lu, D, Kussie, P, Pytowski, B, Persaud, K, Bohlen, P, Witte, L, and Zhu, Z
- Abstract
The kinase domain receptor (KDR) of vascular endothelial growth factor (VEGF) is the main human receptor responsible for the angiogenic activity of VEGF. The extracellular region of KDR is comprised of seven immunoglobulin-like domains, of which the first three have been shown to be required for ligand binding. We have previously described antibodies directed against the extracellular region of KDR, including MAB383 and MAB664, which were shown to block the binding of VEGF to the receptor and to inhibit both VEGF-induced mitogenesis of human endothelial cells in vitro and tumor growth in vivo. Here we generated a series of KDR deletion mutants consisting of truncated extracellular regions and mapped out the domain(s) responsible for binding to VEGF and the neutralizing anti-KDR antibodies. All neutralizing antibodies were found to require domain 3 for efficient binding. Alanine-scanning mutagenesis of domain 3 identified two different sets of five residues, Ile(256), Asp(257), Glu(261), Leu(313), and Thr(315) and Tyr(262), Pro(263), Ser(264), Ser(265), and Lys(266), that were critical for binding to MAB383 and MAB664, respectively. Combination of alanine mutations affecting both MAB383 and MAB664 binding resulted in a variant that also lost binding to VEGF. These results suggest that the residues within this region of domain 3 are critical for VEGF binding. Our studies provide a basis for the mechanism of action of our anti-KDR antibodies and establish a functional foundation for the development of other classes of antagonists to the receptor.
- Published
- 2000
21. A monoclonal antibody to a human neutrophil-specific plasma membrane antigen. Effect of the antibody on the C3bi-mediated adherence by neutrophils and expression of the antigen during myelopoiesis.
- Author
-
Pytowski, B, Easton, T G, Valinsky, J E, Calderon, T, Sun, T, Christman, J K, Wright, S D, and Michl, J
- Abstract
We have used mice selectively tolerized to antigens of human lymphocytes by treatment with cyclophosphamide to raise an mAb, BH2-C6, that reacts with a plasma membrane antigen specific for human neutrophils. This specificity is demonstrated by indirect immunofluorescence microscopy, cytochemical analysis of fluorescence-positive and -negative cell populations separated by flow cytometry, and by the selective, complement-mediated killing of mAb BH2-C6-treated neutrophils. Additional evidence for the neutrophil specificity of mAb BH2-C6 is shown by immunoelectron microscopy, which demonstrates a lack of reactivity with human eosinophils. Immunoblotting of SDS-PAGE-separated proteins of polymorphonuclear leukocytes with 125I-labeled BH2-C6 identifies protein with an average molecular mass of 157 kD. Binding studies show that, at saturation, neutrophils bind 214,000 molecules of 125I-BH2-C6 per cell. Addition of mAb BH2-C6 to neutrophils significantly reduces the number of C3bi-opsonized sheep erythrocytes (EIgMC3bi) bound by these cells. This reduction is partly reversed by the presence of soybean trypsin inhibitor (SBTI), indicating that at least one part of this inhibition is due to BH2-C6-stimulated secretion of a serine protease that may affect ligand binding. Cytochemical analysis of normal human bone marrow cells sorted by cytofluorimetry identifies the promyelocyte as the precursor cell that first expresses BH2-Ag on the plasma membrane. Using the leukemic cell line HL-60, we demonstrate that only inducers of granulocytic differentiation, cis-retinoic acid, and dimethyloxazolidine stimulate the expression of BH2-Ag. These results show that the expression of BH2-Ag during myelomonocytic differentiation is a property uniquely possessed by cells committed to the neutrophilic lineage.
- Published
- 1988
- Full Text
- View/download PDF
22. An internalization motif is created in the cytoplasmic domain of the transferrin receptor by substitution of a tyrosine at the first position of a predicted tight turn.
- Author
-
Pytowski, B, Judge, T W, and McGraw, T E
- Abstract
Receptors are internalized from the plasma membrane at approximately 10 times the rate of bulk membrane. The predominant model for the motif that promotes rapid internalization proposes a requirement for a tyrosine located in the first position of a tight turn. In this report we show that an internalization motif can be created de novo by substituting a tyrosine for the first or last residues of a tetrapeptide GDNS (residues 31-34) that is predicted to form a tight turn within the cytoplasmic domain of the human transferrin receptor. These substitutions restore wild-type levels of internalization to transferrin receptors that are poorly internalized due to missense mutations in the native internalization motif. The introduction of a tyrosine at the first or last position of the GDNS tetrapeptide in a transferrin receptor containing an unmodified wild-type internalization motif significantly increases the internalization rate above that of the wild-type receptor. Our results indicate that a functional novel internalization motif can be created by placing specific aromatic amino acids within the overall structure of an existing beta-turn in a cytoplasmic domain of a receptor.
- Published
- 1995
23. P-551: Blockage of the VEGF-C/VEGF receptor 3 pathway disrupts early pregnancy development in the rodent
- Author
-
Douglas, N.C., Tan, H., Gomez, R., Pytowski, B., Sauer, M.V., and Zimmermann, R.C.
- Published
- 2006
- Full Text
- View/download PDF
24. 162 In vivo effects of a monoclonal antibody to the murine VEGFR-3 that antagonizes the binding of VEGF-C and receptor signaling
- Author
-
Persaud, K., Goldman, J., Prewett, M., Tonra, J., Bassi, R., Anderson, D., Hicklin, D., Witte, L., Swartz, M.A., and Pytowski, B.
- Published
- 2004
- Full Text
- View/download PDF
25. Targeting Lymphatic Vessel CCL21 Production by VEGFR-3 Inhibition Prevents Cardiac Allograft Rejection and Arteriosclerosis
- Author
-
Nykanen, A. I., Sandelin, H., Rainer Krebs, Keranen, M. A. I., Tuuminen, R., Karpanen, T., Wu, Y., Pytowski, B., Petri Koskinen, Yla-Herttuala, S., Kari Alitalo, and Lemstrom, K.
26. Regulation of lymphatic capillary regeneration by interstitial flow in skin
- Author
-
Goldman, J., Conley, K. A., Raehl, A., Bondy, D. M., Pytowski, B., Swartz, M. A., Rutkowski, J. M., Jaroch, D. B., and Ongstad, E. L.
- Subjects
Mice ,Skin/*blood supply/drug effects ,Lymphangiogenesis/drug effects/*physiology ,Animals ,Female ,Vascular Endothelial Growth Factor C/*administration ,Vascular Endothelial Growth Factor Receptor-3/metabolism ,Skin Physiology/drug effects ,Regeneration/drug effects/*physiology ,dosage ,Lymph Nodes/blood supply/drug effects/*physiology ,Inbred BALB C ,Lymphatic Vessels/drug effects/*physiology - Abstract
Decreased interstitial flow (IF) in secondary lymphedema is coincident with poor physiological lymphatic regeneration. However, both the existence and direction of causality between IF and lymphangiogenesis remain unclear. This is primarily because the role of IF and its importance relative to the action of the prolymphangiogenic growth factor vascular endothelial growth factor (VEGF)-C (which signals primarily through its receptor VEGFR-3) are poorly understood. To clarify this, we explored the cooperative roles of VEGFR-3 and IF in a mouse model of lymphangiogenesis in regenerating skin. Specifically, a region of lymphangiogenesis was created by substituting a portion of mouse tail skin with a collagen gel within which lymphatic capillaries completely regenerate over a period of 60 days. The relative importance of IF and VEGF-C signaling were evaluated by either inhibiting VEGFR-3 signaling with antagonistic antibodies or by reducing IF. In some cases, VEGF-C signaling was then increased with exogenous protein. To clarify the role of IF, the distribution of endogenous matrix metalloproteinases (MMPs) and VEGF-C within the regenerating region was determined. It was found that inhibition of either VEGFR-3 or IF suppressed endogenous lymphangiogenesis. Reduction of IF was found to decrease lymphatic migration and transport of endogenous MMP and VEGF-C through the regenerating region. Therapeutic VEGF-C administration restored lymphangiogenesis following inhibition of VEGFR-3 but did not increase lymphangiogenesis following inhibition of IF. These results identify IF as an important regulator of the pro-lymphangiogenic action of VEGF-C.
27. Correction: Involvement of the VEGF receptor 3 in tubular morphogenesis demonstrated with a human antihuman VEGFR-3 monoclonal antibody that antagonizes receptor activation by VEGF-C.
- Author
-
Persaud K, Tille JC, Liu M, Zhu Z, Jimenez X, Pereira DS, Miao HQ, Brennan LA, Witte L, Pepper MS, and Pytowski B
- Published
- 2023
- Full Text
- View/download PDF
28. Xerna™ TME Panel is a machine learning-based transcriptomic biomarker designed to predict therapeutic response in multiple cancers.
- Author
-
Uhlik M, Pointing D, Iyer S, Ausec L, Štajdohar M, Cvitkovič R, Žganec M, Culm K, Santos VC, Pytowski B, Malafa M, Liu H, Krieg AM, Lee J, Rosengarten R, and Benjamin L
- Abstract
Introduction: Most predictive biomarkers approved for clinical use measure single analytes such as genetic alteration or protein overexpression. We developed and validated a novel biomarker with the aim of achieving broad clinical utility. The Xerna™ TME Panel is a pan-tumor, RNA expression-based classifier, designed to predict response to multiple tumor microenvironment (TME)-targeted therapies, including immunotherapies and anti-angiogenic agents., Methods: The Panel algorithm is an artificial neural network (ANN) trained with an input signature of 124 genes that was optimized across various solid tumors. From the 298-patient training data, the model learned to discriminate four TME subtypes: Angiogenic (A), Immune Active (IA), Immune Desert (ID), and Immune Suppressed (IS). The final classifier was evaluated in four independent clinical cohorts to test whether TME subtype could predict response to anti-angiogenic agents and immunotherapies across gastric, ovarian, and melanoma datasets., Results: The TME subtypes represent stromal phenotypes defined by angiogenesis and immune biological axes. The model yields clear boundaries between biomarker-positive and -negative and showed 1.6-to-7-fold enrichment of clinical benefit for multiple therapeutic hypotheses. The Panel performed better across all criteria compared to a null model for gastric and ovarian anti-angiogenic datasets. It also outperformed PD-L1 combined positive score (>1) in accuracy, specificity, and positive predictive value (PPV), and microsatellite-instability high (MSI-H) in sensitivity and negative predictive value (NPV) for the gastric immunotherapy cohort., Discussion: The TME Panel's strong performance on diverse datasets suggests it may be amenable for use as a clinical diagnostic for varied cancer types and therapeutic modalities., Competing Interests: MU, SI, KC, VS, BP and LB are employees of and have financial interests in OncXerna Therapeutics Inc. DP, LA, RC, MŽ, and RR are employees of and have financial interests in Genialis Inc. HL and AK were employees of and had/have financial interests in Checkmate Pharmaceuticals which was recently acquired by Regeneron. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Uhlik, Pointing, Iyer, Ausec, Štajdohar, Cvitkovič, Žganec, Culm, Santos, Pytowski, Malafa, Liu, Krieg, Lee, Rosengarten and Benjamin.)
- Published
- 2023
- Full Text
- View/download PDF
29. Treatment with a VEGFR-2 antibody results in intra-tumor immune modulation and enhances anti-tumor efficacy of PD-L1 blockade in syngeneic murine tumor models.
- Author
-
Li Y, Amaladas N, O'Mahony M, Manro JR, Inigo I, Li Q, Rasmussen ER, Brahmachary M, Doman TN, Hall G, Kalos M, Novosiadly R, Puig O, Pytowski B, and Schaer DA
- Subjects
- Animals, B7-H1 Antigen metabolism, Cell Line, Tumor, Mice, Tumor Microenvironment, Vascular Endothelial Growth Factor A, Neoplasms therapy, Vascular Endothelial Growth Factor Receptor-2
- Abstract
Prolonged activation of vascular endothelial growth factor receptor-2 (VEGFR-2) due to mis-regulation of the VEGF pathway induces aberrant blood vessel expansion, which supports growth and survival of solid tumors. Therapeutic interventions that inhibit the VEGFR-2 pathway have therefore become a mainstay of cancer treatment. Non-clinical studies have recently revealed that blockade of angiogenesis can modulate the tumor microenvironment and enhance the efficacy of concurrent immune therapies. Ramucirumab is an FDA-approved anti-angiogenic antibody that inhibits VEGFR-2 and is currently being evaluated in clinical studies in combination with anti-programmed cell death (PD-1) axis checkpoint inhibitors (pembrolizumab, durvalumab, or sintilimab) across several cancer types. The purpose of this study is to establish a mechanistic basis for the enhanced activity observed in the combined blockade of VEGFR-2 and PD-1-axis pathways. Pre-clinical studies were conducted in murine tumor models known to be responsive to anti-PD-1 axis therapy, using monoclonal antibodies that block mouse VEGFR-2 and programmed death-ligand 1 (PD-L1). Combination therapy resulted in enhanced anti-tumor activity compared to anti-PD-L1 monotherapy. VEGFR-2 blockade at early timepoints post-anti-PD-L1 therapy resulted in a dose-dependent and transient enhanced infiltration of T cells, and establishment of immunological memory. VEGFR-2 blockade at later timepoints resulted in enhancement of anti-PD-L1-driven immune cell infiltration. VEGFR-2 and PD-L1 monotherapies induced both unique and overlapping patterns of immune gene expression, and combination therapy resulted in an enhanced immune activation signature. Collectively, these results provide new and actionable insights into the mechanisms by which concurrent VEGFR-2 and PD-L1 antibody therapy leads to enhanced anti-tumor efficacy., Competing Interests: Y.L. employee and shareholder of Eli Lilly at the time of this work and is currently an employee of Regeneron. N.A. employee and shareholder of Eli Lilly. M.O.M. employee and shareholder of Eli Lilly. J.R.M. employee and shareholder of Eli Lilly. I.I. employee and shareholder of Eli Lilly at the time of this work and is currently an employee of Astra Zeneca. Q.L. employee and shareholder of Eli Lilly. E.R.R. employee and shareholder of Eli Lilly. M.B. employee and shareholder of Eli Lilly at the time of this work and is currently an employee of Sanofi, US. T.N.D. employee and shareholder of Eli Lilly. G.H. employee and shareholder of Eli Lilly. M.K. employee and shareholder of Eli Lilly at the time of this work, and currently Managing Director of Next Pillar Consulting, LLC. M.K. reports stock ownership as a result of employment or advisory roles in: ArsenalBio, Immunai, Cue Biopharma, Nanocell therapeutics, IMV inc., SentiBio, AdicetBio, Orange Grove Bio. Issued patents in the field of cell therapy, licensed by the University of Pennsylvania to Novartis corporation, resulting in royalty distributions. R.N. employee and shareholder of Eli Lilly at the time of this work and is currently an employee of Bristol-Myers Squibb. O.P. employee and shareholder of Eli Lilly. B.P. employee and shareholder of Eli Lilly at the time of this work and is currently an employee of OncXerna Therapeutics, Inc. B.P. reports support for attending meetings and/or travel and stock/stock options from OncXerna Therapeutics, Inc. D.A.S. employee and shareholder of Eli Lilly at the time of this work and is currently an employee of Pfizer. D.A.S. reports support for attending meetings and/or travel from Pfizer Inc (employee of Pfizer). This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2022
- Full Text
- View/download PDF
30. Anti-VEGFR2 therapy delays growth of preclinical pediatric tumor models and enhances anti-tumor activity of chemotherapy.
- Author
-
Lowery CD, Blosser W, Dowless M, Renschler M, Perez LV, Stephens J, Pytowski B, Wasserstrom H, Stancato LF, and Falcon B
- Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) is an attractive therapeutic target in solid malignancies due to its central role in tumor angiogenesis. Ramucirumab (Cyramza
® , LY3009806) is a human monoclonal antibody specific for VEGFR2 approved for several adult indications and currently in a phase 1 clinical trial for pediatric patients with solid tumors (NCT02564198). Here, we evaluated ramucirumab in vitro and the anti-murine VEGFR2 antibody DC101 in vivo with or without chemotherapy across a range of pediatric cancer models. Ramucirumab abrogated in vitro endothelial cord formation driven by cancer cell lines representing multiple pediatric histologies; this response was independent of the origin of the tumor cell-line. Several pediatric cancer mouse models responded to single agent DC101-mediated VEGFR2 inhibition with tumor growth delay. Preclinical stable disease and partial xenograft regressions were observed in mouse models of Ewing's sarcoma, synovial sarcoma, neuroblastoma, and desmoplastic small round cell tumor treated with DC101 and cytotoxic chemotherapy. In contrast, DC101 treatment in osteosarcoma models had limited efficacy alone or in combination with chemotherapeutics. Our data indicate differential efficacy of targeting the VEGFR2 pathway in pediatric models and support the continued evaluation of VEGFR2 inhibition in combination with cytotoxic chemotherapy in multiple pediatric indications., Competing Interests: CONFLICTS OF INTEREST All authors are full-time employees of Eli Lilly and Company., (Copyright: © 2019 Lowery et al.)- Published
- 2019
- Full Text
- View/download PDF
31. Blockade of Vascular Endothelial Growth Factor Receptor-1 (Flt-1), Reveals a Novel Analgesic For Osteoarthritis-Induced Joint Pain.
- Author
-
Das V, Kc R, Li X, O-Sullivan I, van Wijnen AJ, Kroin JS, Pytowski B, Applegate DT, Votta-Velis G, Ripper RL, Park TJ, and Im HJ
- Abstract
Osteoarthritis (OA) is a painful and debilitating disease. A striking feature of OA is the dramatic increase in vascular endothelial growth factor (VEGF) levels and in new blood vessel formation in the joints, both of which correlate with the severity of OA pain. Our aim was to determine whether anti-VEGF monoclonal antibodies (mAbs) - MF-1 (mAb to VEGFR1) and DC101 (mAb to VEGFR2) - can reduce OA pain and can do so by targeting VEGF signaling pathways such as Flt-1 (VEGFR1) and Flk-1 (VEGFR2). After IACUC approval, OA was induced by partial medial meniscectomy (PMM) in C57/BL6 mice (20 g). ln the first experiment, for validation of VEGFR1 in DRG, the mouse dorsal root ganglion (DRG) was stimulated with NGF for 48 hours to find the relative gene induction for VEGFR1 vs. 18S by RT-PCR. In the second experiment, Biotin-conjugated VEGFA (1 µg/knee joint) was administered in the left knee joint of mice with advanced OA in order to characterization of VEGFR1 and VEGFR2. pVEGFR1/VEGFR2 was detected by immunostaining in DRGs. Finally, MF-1 and DC101 were administered in OA mice by both intrathecal (IT) and intraarticular (IA) injections, and the change in paw withdrawal threshold (PWT) was measured. Retrograde transport of VEGF was confirmed for detection of pVEGFR1/VEGFR2 in the DRG. PMM surgery led to development of OA and mechanical allodynia, with reduced paw withdrawal thresholds (PWT) ( P<0.0001 ). IT injection of MF-1 led to a reduction of allodynia in advanced OA, but injection of DC101 did not. IA injection of MF-1 or DC101 at one week after PMM injury did not reduce allodynia, but when injected in advanced OA mice joints at 12 weeks, both Mabs increased PWT an indicator of analgesia. Our data show that MF-1 (VEGR1 inhibition) decreases pain in advanced OA after IT or IA injection. Activation of MF-1 or DC101 may ameliorate OA-related joint pain., Competing Interests: Competing interests: None declared.
- Published
- 2018
- Full Text
- View/download PDF
32. VEGF-C promotes the development of lymphatics in bone and bone loss.
- Author
-
Hominick D, Silva A, Khurana N, Liu Y, Dechow PC, Feng JQ, Pytowski B, Rutkowski JM, Alitalo K, and Dellinger MT
- Subjects
- Animals, Bone Resorption metabolism, Bone and Bones metabolism, Cells, Cultured, Endothelium, Lymphatic metabolism, Humans, Lymphatic Vessels metabolism, Mice, Mice, Transgenic, Osteoclasts metabolism, Phenotype, Signal Transduction, Vascular Endothelial Growth Factor Receptor-3 metabolism, Bone Resorption pathology, Bone and Bones pathology, Endothelium, Lymphatic pathology, Lymphatic Vessels pathology, Osteoclasts pathology, Vascular Endothelial Growth Factor C physiology
- Abstract
Patients with Gorham-Stout disease (GSD) have lymphatic vessels in their bones and their bones gradually disappear. Here, we report that mice that overexpress VEGF-C in bone exhibit a phenotype that resembles GSD. To drive VEGF-C expression in bone, we generated Osx-tTA;TetO-Vegfc double-transgenic mice. In contrast to Osx-tTA mice, Osx-tTA;TetO-Vegfc mice developed lymphatics in their bones. We found that inhibition of VEGFR3, but not VEGFR2, prevented the formation of bone lymphatics in Osx-tTA;TetO-Vegfc mice. Radiological and histological analysis revealed that bones from Osx-tTA;TetO-Vegfc mice were more porous and had more osteoclasts than bones from Osx-tTA mice. Importantly, we found that bone loss in Osx-tTA;TetO-Vegfc mice could be attenuated by an osteoclast inhibitor. We also discovered that the mutant phenotype of Osx-tTA;TetO-Vegfc mice could be reversed by inhibiting the expression of VEGF-C. Taken together, our results indicate that expression of VEGF-C in bone is sufficient to induce the pathologic hallmarks of GSD in mice., Competing Interests: DH, AS, NK, YL, PD, JF, JR, MD No competing interests declared, BP During preparation of the manuscript, B Pytowski was an employee of Eli Lilly and continues to hold stock in the company. KA Reviewing editor, eLife, (© 2018, Hominick et al.)
- Published
- 2018
- Full Text
- View/download PDF
33. Phase 1 study of the anti-vascular endothelial growth factor receptor 3 monoclonal antibody LY3022856/IMC-3C5 in patients with advanced and refractory solid tumors and advanced colorectal cancer.
- Author
-
Saif MW, Knost JA, Chiorean EG, Kambhampati SR, Yu D, Pytowski B, Qin A, Kauh JS, and O'Neil BH
- Subjects
- Adult, Aged, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Cohort Studies, Disease-Free Survival, Drug Resistance, Neoplasm, Female, Humans, Kaplan-Meier Estimate, Male, Maximum Tolerated Dose, Middle Aged, Treatment Outcome, Vascular Endothelial Growth Factor Receptor-3 immunology, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Colorectal Neoplasms drug therapy, Neoplasms drug therapy, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors
- Abstract
Purpose: Metastasis of solid tumors to regional lymph nodes is facilitated by tumor lymphangiogenesis, which is primarily mediated by the vascular endothelial growth factor receptor 3 (VEGFR-3). We conducted a phase 1 dose-escalation (part A) study of the VEGFR-3 human immunoglobulin G subclass 1 monoclonal antibody LY3022856 in advanced solid tumors, followed by a colorectal cancer (CRC) expansion (part B)., Methods: Part A evaluated the safety profile and maximum tolerated dose (MTD) of LY3022856 in patients treated intravenously at doses of 5-30 mg/kg weekly (qwk). Part B further evaluated tolerability in CRC patients treated with 30 mg/kg. Secondary objectives were pharmacokinetics, anti-tumor activity, and pharmacodynamics (exploratory)., Results: A total of 44 patients (23 in part A; 21 in part B) were treated; only one dose-limiting toxicity was observed at the lowest dose level. The MTD was not reached. Treatment-emergent adverse events (TEAEs) of any grade included in ≥15 % of all patients were: nausea (41 %), fatigue (32 %), vomiting (30 %), decreased appetite (27 %), pyrexia (25 %), peripheral edema (23 %), and urinary tract infection (UTI, 20 %). The most common grade 3/4 TEAEs included UTI and small intestinal obstruction (7 % each). No radiographic responses were noted. Median progression-free survival in part B was 6.3 weeks (95 % confidence interval: 5.1, 14.4), and a best overall response of stable disease was observed in 4 CRC patients (19.0 %)., Conclusions: LY3022856 was well tolerated up to a dose of 30 mg/kg qwk, but with minimal anti-tumor activity in CRC. CLINICALTRIALS., Gov Identifier: NCT01288989.
- Published
- 2016
- Full Text
- View/download PDF
34. Antagonist antibodies to vascular endothelial growth factor receptor 2 (VEGFR-2) as anti-angiogenic agents.
- Author
-
Falcon BL, Chintharlapalli S, Uhlik MT, and Pytowski B
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Drug Resistance, Neoplasm physiology, Drug Therapy, Combination, Humans, Ramucirumab, Neoplasms drug therapy, Neoplasms physiopathology, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic physiopathology, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors
- Abstract
Interaction of numerous signaling pathways in endothelial and mesangial cells results in exquisite control of the process of physiological angiogenesis, with a central role played by vascular endothelial growth factor receptor 2 (VEGFR-2) and its cognate ligands. However, deregulated angiogenesis participates in numerous pathological processes. Excessive activation of VEGFR-2 has been found to mediate tissue-damaging vascular changes as well as the induction of blood vessel expansion to support the growth of solid tumors. Consequently, therapeutic intervention aimed at inhibiting the VEGFR-2 pathway has become a mainstay of treatment in cancer and retinal diseases. In this review, we introduce the concepts of physiological and pathological angiogenesis, the crucial role played by the VEGFR-2 pathway in these processes, and the various inhibitors of its activity that have entered the clinical practice. We primarily focus on the development of ramucirumab, the antagonist monoclonal antibody (mAb) that inhibits VEGFR-2 and has recently been approved for use in patients with gastric, colorectal, and lung cancers. We examine in-depth the pre-clinical studies using DC101, the mAb to mouse VEGFR-2, which provided a conceptual foundation for the role of VEGFR-2 in physiological and pathological angiogenesis. Finally, we discuss further clinical development of ramucirumab and the future of targeting the VEGF pathway for the treatment of cancer., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Stromal-Based Signatures for the Classification of Gastric Cancer.
- Author
-
Uhlik MT, Liu J, Falcon BL, Iyer S, Stewart J, Celikkaya H, O'Mahony M, Sevinsky C, Lowes C, Douglass L, Jeffries C, Bodenmiller D, Chintharlapalli S, Fischl A, Gerald D, Xue Q, Lee JY, Santamaria-Pang A, Al-Kofahi Y, Sui Y, Desai K, Doman T, Aggarwal A, Carter JH, Pytowski B, Jaminet SC, Ginty F, Nasir A, Nagy JA, Dvorak HF, and Benjamin LE
- Subjects
- Animals, Biomarkers, Tumor analysis, Biomarkers, Tumor genetics, Computational Biology methods, Disease Models, Animal, Gene Expression Profiling methods, Heterografts, Humans, Image Processing, Computer-Assisted, Immunohistochemistry, Mice, Neovascularization, Pathologic genetics, Oligonucleotide Array Sequence Analysis, Tissue Array Analysis, Vascular Endothelial Growth Factor A metabolism, Stomach Neoplasms classification, Stomach Neoplasms genetics, Transcriptome genetics, Tumor Microenvironment genetics
- Abstract
Treatment of metastatic gastric cancer typically involves chemotherapy and monoclonal antibodies targeting HER2 (ERBB2) and VEGFR2 (KDR). However, reliable methods to identify patients who would benefit most from a combination of treatment modalities targeting the tumor stroma, including new immunotherapy approaches, are still lacking. Therefore, we integrated a mouse model of stromal activation and gastric cancer genomic information to identify gene expression signatures that may inform treatment strategies. We generated a mouse model in which VEGF-A is expressed via adenovirus, enabling a stromal response marked by immune infiltration and angiogenesis at the injection site, and identified distinct stromal gene expression signatures. With these data, we designed multiplexed IHC assays that were applied to human primary gastric tumors and classified each tumor to a dominant stromal phenotype representative of the vascular and immune diversity found in gastric cancer. We also refined the stromal gene signatures and explored their relation to the dominant patient phenotypes identified by recent large-scale studies of gastric cancer genomics (The Cancer Genome Atlas and Asian Cancer Research Group), revealing four distinct stromal phenotypes. Collectively, these findings suggest that a genomics-based systems approach focused on the tumor stroma can be used to discover putative predictive biomarkers of treatment response, especially to antiangiogenesis agents and immunotherapy, thus offering an opportunity to improve patient stratification. Cancer Res; 76(9); 2573-86. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
36. Inhibition of VEGFR-3 activation in tumor-draining lymph nodes suppresses the outgrowth of lymph node metastases in the MT-450 syngeneic rat breast cancer model.
- Author
-
Quagliata L, Klusmeier S, Cremers N, Pytowski B, Harvey A, Pettis RJ, Thiele W, and Sleeman JP
- Subjects
- Animals, Breast Neoplasms pathology, Female, Humans, Lung Neoplasms pathology, Lung Neoplasms secondary, Lymph Nodes pathology, Lymphatic Metastasis pathology, Mammary Neoplasms, Animal pathology, Rats, Transcriptional Activation genetics, Vascular Endothelial Growth Factor C antagonists & inhibitors, Vascular Endothelial Growth Factor C metabolism, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors, Breast Neoplasms genetics, Lymphatic Metastasis genetics, Mammary Neoplasms, Animal genetics, Vascular Endothelial Growth Factor C genetics, Vascular Endothelial Growth Factor Receptor-3 genetics
- Abstract
For many types of human cancer, the expression of vascular endothelial growth factor-C (VEGF-C) correlates with enhanced tumor-associated lymphatic vessel density, metastasis formation and poor prognosis. In experimental animals, VEGF-C produced by primary tumors can induce lymphangiogenesis within and/or at the periphery of the tumor, and promotes metastasis formation. Tumor-induced lymphangiogenesis is therefore thought to expedite entry of tumor cells into the lymphatic vasculature and their trafficking to regional lymph nodes, thereby fostering metastatic dissemination. Tumour-produced VEGF-C can also drain to the regional lymph nodes and induce lymphangiogenesis there. Whether this activity promotes metastasis formation remains unclear. To address this issue we manipulated VEGF-C activity and VEGFR-3 activation in the lymph nodes draining syngeneic rat breast cancers using intra-dermal delivery of either recombinant VEGF-C or VEGFR-3 blocking antibodies to induce or suppress lymph node lymphangiogenesis, respectively. Recombinant VEGF-C induced lymph node lymphangiogenesis, but was not sufficient to promote metastasis formation by poorly metastatic NM-081 breast tumours. Conversely, inhibition of lymph node lymphangiogeneis induced by highly metastatic MT-450 breast tumours suppressed the outgrowth of lymph node metastases, but not the initial colonization of the lymph nodes. Lung metastasis was also not affected. We conclude that tumor-derived VEGF-C draining to regional lymph nodes promotes the outgrowth of lymph node metastases. VEGF-C may induce lung metastasis independently of its effects on lymph node metastasis.
- Published
- 2014
- Full Text
- View/download PDF
37. Pulmonary lymphangiectasia resulting from vascular endothelial growth factor-C overexpression during a critical period.
- Author
-
Yao LC, Testini C, Tvorogov D, Anisimov A, Vargas SO, Baluk P, Pytowski B, Claesson-Welsh L, Alitalo K, and McDonald DM
- Subjects
- Animals, Female, Humans, Infant, Lung Diseases genetics, Lung Diseases metabolism, Lung Diseases pathology, Lymphangiectasis genetics, Lymphangiectasis metabolism, Lymphangiectasis pathology, Lymphatic Vessels metabolism, Lymphatic Vessels pathology, Male, Mice, Mice, Inbred Strains, Mice, Transgenic, Pregnancy, Pulmonary Edema genetics, Pulmonary Edema metabolism, Pulmonary Edema pathology, Signal Transduction physiology, Trachea metabolism, Trachea pathology, Uteroglobin genetics, Uteroglobin metabolism, Vascular Endothelial Growth Factor C metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism, Vascular Endothelial Growth Factor Receptor-3 metabolism, Lung Diseases congenital, Lymphangiectasis congenital, Vascular Endothelial Growth Factor C genetics
- Abstract
Rationale: Lymphatic vessels in the respiratory tract normally mature into a functional network during the neonatal period, but under some pathological conditions they can grow as enlarged, dilated sacs that result in the potentially lethal condition of pulmonary lymphangiectasia., Objective: We sought to determine whether overexpression of the lymphangiogenic growth factor (vascular endothelial growth factor-C [VEGF-C]) can promote lymphatic growth and maturation in the respiratory tract. Unexpectedly, perinatal overexpression of VEGF-C in the respiratory epithelium led to a condition resembling human pulmonary lymphangiectasia, a life-threatening disorder of the newborn characterized by respiratory distress and the presence of widely dilated lymphatics., Methods and Results: Administration of doxycycline to Clara cell secretory protein-reverse tetracycline-controlled transactivator/tetracycline operator-VEGF-C double-transgenic mice during a critical period from embryonic day 15.5 to postnatal day 14 was accompanied by respiratory distress, chylothorax, pulmonary lymphangiectasia, and high mortality. Enlarged sac-like lymphatics were abundant near major airways, pulmonary vessels, and visceral pleura. Side-by-side comparison revealed morphological features similar to pulmonary lymphangiectasia in humans. The condition was milder in mice given doxycycline after age postnatal day 14 and did not develop after postnatal day 35. Mechanistic studies revealed that VEGF recptor (VEGFR)-3 alone drove lymphatic growth in adult mice, but both VEGFR-2 and VEGFR-3 were required for the development of lymphangiectasia in neonates. VEGFR-2/VEGFR-3 heterodimers were more abundant in the dilated lymphatics, consistent with the involvement of both receptors. Despite the dependence of lymphangiectasia on VEGFR-2 and VEGFR-3, the condition was not reversed by blocking both receptors together or by withdrawing VEGF-C., Conclusions: The findings indicate that VEGF-C overexpression can induce pulmonary lymphangiectasia during a critical period in perinatal development.
- Published
- 2014
- Full Text
- View/download PDF
38. Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF-VEGFR2 signalling.
- Author
-
Benedito R, Rocha SF, Woeste M, Zamykal M, Radtke F, Casanovas O, Duarte A, Pytowski B, and Adams RH
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Calcium-Binding Proteins, Cells, Cultured, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells metabolism, Female, HEK293 Cells, Human Umbilical Vein Endothelial Cells, Humans, Intracellular Signaling Peptides and Proteins deficiency, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Male, Membrane Proteins deficiency, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Models, Biological, Neovascularization, Physiologic drug effects, Protein Kinase Inhibitors pharmacology, Receptors, Notch antagonists & inhibitors, Transcription, Genetic, Vascular Endothelial Growth Factor Receptor-2 deficiency, Vascular Endothelial Growth Factor Receptor-2 genetics, Vascular Endothelial Growth Factor Receptor-3 biosynthesis, Vascular Endothelial Growth Factor Receptor-3 genetics, Neovascularization, Physiologic physiology, Receptors, Notch metabolism, Signal Transduction drug effects, Up-Regulation, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism, Vascular Endothelial Growth Factor Receptor-3 metabolism
- Abstract
Developing tissues and growing tumours produce vascular endothelial growth factors (VEGFs), leading to the activation of the corresponding receptors in endothelial cells. The resultant angiogenic expansion of the local vasculature can promote physiological and pathological growth processes. Previous work has uncovered that the VEGF and Notch pathways are tightly linked. Signalling triggered by VEGF-A (also known as VEGF) has been shown to induce expression of the Notch ligand DLL4 in angiogenic vessels and, most prominently, in the tip of endothelial sprouts. DLL4 activates Notch in adjacent cells, which suppresses the expression of VEGF receptors and thereby restrains endothelial sprouting and proliferation. Here we show, by using inducible loss-of-function genetics in combination with inhibitors in vivo, that DLL4 protein expression in retinal tip cells is only weakly modulated by VEGFR2 signalling. Surprisingly, Notch inhibition also had no significant impact on VEGFR2 expression and induced deregulated endothelial sprouting and proliferation even in the absence of VEGFR2, which is the most important VEGF-A receptor and is considered to be indispensable for these processes. By contrast, VEGFR3, the main receptor for VEGF-C, was strongly modulated by Notch. VEGFR3 kinase-activity inhibitors but not ligand-blocking antibodies suppressed the sprouting of endothelial cells that had low Notch signalling activity. Our results establish that VEGFR2 and VEGFR3 are regulated in a highly differential manner by Notch. We propose that successful anti-angiogenic targeting of these receptors and their ligands will strongly depend on the status of endothelial Notch signalling.
- Published
- 2012
- Full Text
- View/download PDF
39. Live imaging of newly formed lymphatic vessels in the cornea.
- Author
-
Yuen D, Wu X, Kwan AC, Ledue J, Zhang H, Ecoiffier T, Pytowski B, and Chen L
- Subjects
- Animals, Cornea pathology, Cornea ultrastructure, Diagnostic Imaging instrumentation, Lymphangiogenesis, Lymphatic Vessels pathology, Mice, Microscopy, Video, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-2 metabolism, Cornea blood supply, Diagnostic Imaging methods, Lymphatic Vessels cytology
- Published
- 2011
- Full Text
- View/download PDF
40. Combined blockade of VEGFR-2 and VEGFR-3 inhibits inflammatory lymphangiogenesis in early and middle stages.
- Author
-
Yuen D, Pytowski B, and Chen L
- Subjects
- Animals, Antibodies, Neutralizing pharmacology, Corneal Neovascularization metabolism, Disease Models, Animal, Fluorescent Antibody Technique, Indirect, Glycoproteins metabolism, Injections, Intraperitoneal, Lymphatic Vessels pathology, Male, Membrane Transport Proteins, Mice, Mice, Inbred BALB C, Microscopy, Fluorescence, Time Factors, Vascular Endothelial Growth Factor Receptor-2 immunology, Vascular Endothelial Growth Factor Receptor-3 immunology, Antibodies, Monoclonal pharmacology, Corneal Neovascularization prevention & control, Lymphangiogenesis drug effects, Lymphatic Vessels drug effects, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors
- Abstract
Purpose: Lymphangiogenesis (LG) accompanies many corneal diseases after inflammatory, infectious, or chemical insults and is a primary mediator of transplant rejection. The purpose of this study was to investigate whether there is a time window for therapeutic intervention of corneal LG and whether a combined blockade of VEGFR-2 and VEGFR-3 effectively suppresses early-, middle-, or late-stage LG., Methods: Corneal inflammatory neovascularization was induced by a standard suture placement model in mice. Neutralizing antibodies against VEGFR-3 and/or VEGFR-2 were administrated systemically with the treatment started at postoperative day 0, day 7, or day 14. Whole mount corneas were sampled for immunofluorescence microscopic studies using LYVE-1 (a lymphatic marker) antibodies. Digital images were analyzed by software., Results: Both VEGFR-3 and VEGFR-2 were involved in corneal suture-induced inflammatory LG. Their combined blockade led to a significant inhibition of both early- and middle-stage LG while demonstrating no effect on late-stage LG., Conclusions: Corneal inflammatory LG has a discrete time window for intervention therapy. Although it is important to start the treatment as soon as possible, interventions initiated in the middle of the LG process are still effective. These novel findings will shed some light on our understanding of inflammatory LG and the development of new therapeutic protocols for LG-related diseases at different stages.
- Published
- 2011
- Full Text
- View/download PDF
41. Vascular endothelial growth factor receptor 3 directly regulates murine neurogenesis.
- Author
-
Calvo CF, Fontaine RH, Soueid J, Tammela T, Makinen T, Alfaro-Cervello C, Bonnaud F, Miguez A, Benhaim L, Xu Y, Barallobre MJ, Moutkine I, Lyytikkä J, Tatlisumak T, Pytowski B, Zalc B, Richardson W, Kessaris N, Garcia-Verdugo JM, Alitalo K, Eichmann A, and Thomas JL
- Subjects
- Animals, Cells, Cultured, Enzyme-Linked Immunosorbent Assay, Immunohistochemistry, Lymphangiogenesis genetics, Lymphangiogenesis physiology, Mice, Mice, Mutant Strains, Microscopy, Electron, Transmission, Neovascularization, Physiologic genetics, Neovascularization, Physiologic physiology, Neural Stem Cells cytology, Neural Stem Cells metabolism, Neurogenesis genetics, Oligonucleotide Array Sequence Analysis, Reverse Transcriptase Polymerase Chain Reaction, Vascular Endothelial Growth Factor Receptor-3 genetics, Neurogenesis physiology, Vascular Endothelial Growth Factor Receptor-3 metabolism
- Abstract
Neural stem cells (NSCs) are slowly dividing astrocytes that are intimately associated with capillary endothelial cells in the subventricular zone (SVZ) of the brain. Functionally, members of the vascular endothelial growth factor (VEGF) family can stimulate neurogenesis as well as angiogenesis, but it has been unclear whether they act directly via VEGF receptors (VEGFRs) expressed by neural cells, or indirectly via the release of growth factors from angiogenic capillaries. Here, we show that VEGFR-3, a receptor required for lymphangiogenesis, is expressed by NSCs and is directly required for neurogenesis. Vegfr3:YFP reporter mice show VEGFR-3 expression in multipotent NSCs, which are capable of self-renewal and are activated by the VEGFR-3 ligand VEGF-C in vitro. Overexpression of VEGF-C stimulates VEGFR-3-expressing NSCs and neurogenesis in the SVZ without affecting angiogenesis. Conversely, conditional deletion of Vegfr3 in neural cells, inducible deletion in subventricular astrocytes, and blocking of VEGFR-3 signaling with antibodies reduce SVZ neurogenesis. Therefore, VEGF-C/VEGFR-3 signaling acts directly on NSCs and regulates adult neurogenesis, opening potential approaches for treatment of neurodegenerative diseases.
- Published
- 2011
- Full Text
- View/download PDF
42. Stimulation of lymphangiogenesis via VEGFR-3 inhibits chronic skin inflammation.
- Author
-
Huggenberger R, Ullmann S, Proulx ST, Pytowski B, Alitalo K, and Detmar M
- Subjects
- Animals, Antibodies, Blocking immunology, CD8-Positive T-Lymphocytes immunology, Chronic Disease, Dermatitis genetics, Dermatitis immunology, Dermatitis therapy, Keratin-14 genetics, Mice, Mice, Transgenic, Skin blood supply, Skin immunology, Skin metabolism, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor D administration & dosage, Vascular Endothelial Growth Factor D immunology, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-2 immunology, Vascular Endothelial Growth Factor Receptor-3 immunology, Vascular Endothelial Growth Factor Receptor-3 metabolism, Dermatitis metabolism, Lymphangiogenesis, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors
- Abstract
The role of lymphangiogenesis in inflammation has remained unclear. To investigate the role of lymphatic versus blood vasculature in chronic skin inflammation, we inhibited vascular endothelial growth factor (VEGF) receptor (VEGFR) signaling by function-blocking antibodies in the established keratin 14 (K14)-VEGF-A transgenic (Tg) mouse model of chronic cutaneous inflammation. Although treatment with an anti-VEGFR-2 antibody inhibited skin inflammation, epidermal hyperplasia, inflammatory infiltration, and angiogenesis, systemic inhibition of VEGFR-3, surprisingly, increased inflammatory edema formation and inflammatory cell accumulation despite inhibition of lymphangiogenesis. Importantly, chronic Tg delivery of the lymphangiogenic factor VEGF-C to the skin of K14-VEGF-A mice completely inhibited development of chronic skin inflammation, epidermal hyperplasia and abnormal differentiation, and accumulation of CD8 T cells. Similar results were found after Tg delivery of mouse VEGF-D that only activates VEGFR-3 but not VEGFR-2. Moreover, intracutaneous injection of recombinant VEGF-C156S, which only activates VEGFR-3, significantly reduced inflammation. Although lymphatic drainage was inhibited in chronic skin inflammation, it was enhanced by Tg VEGF-C delivery. Together, these results reveal an unanticipated active role of lymphatic vessels in controlling chronic inflammation. Stimulation of functional lymphangiogenesis via VEGFR-3, in addition to antiangiogenic therapy, might therefore serve as a novel strategy to treat chronic inflammatory disorders of the skin and possibly also other organs.
- Published
- 2010
- Full Text
- View/download PDF
43. VEGF receptor 2/-3 heterodimers detected in situ by proximity ligation on angiogenic sprouts.
- Author
-
Nilsson I, Bahram F, Li X, Gualandi L, Koch S, Jarvius M, Söderberg O, Anisimov A, Kholová I, Pytowski B, Baldwin M, Ylä-Herttuala S, Alitalo K, Kreuger J, and Claesson-Welsh L
- Subjects
- Animals, Cell Line, Cells, Cultured, Embryo, Mammalian metabolism, Endothelium, Vascular metabolism, Humans, Neovascularization, Physiologic, Protein Multimerization, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors, Endothelial Cells metabolism, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor C metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism, Vascular Endothelial Growth Factor Receptor-3 metabolism
- Abstract
The vascular endothelial growth factors VEGFA and VEGFC are crucial regulators of vascular development. They exert their effects by dimerization and activation of the cognate receptors VEGFR2 and VEGFR3. Here, we have used in situ proximity ligation to detect receptor complexes in intact endothelial cells. We show that both VEGFA and VEGFC potently induce formation of VEGFR2/-3 heterodimers. Receptor heterodimers were found in both developing blood vessels and immature lymphatic structures in embryoid bodies. We present evidence that heterodimers frequently localize to tip cell filopodia. Interestingly, in the presence of VEGFC, heterodimers were enriched in the leading tip cells as compared with trailing stalk cells of growing sprouts. Neutralization of VEGFR3 to prevent heterodimer formation in response to VEGFA decreased the extent of angiogenic sprouting. We conclude that VEGFR2/-3 heterodimers on angiogenic sprouts induced by VEGFA or VEGFC may serve to positively regulate angiogenic sprouting.
- Published
- 2010
- Full Text
- View/download PDF
44. Targeting lymphatic vessel activation and CCL21 production by vascular endothelial growth factor receptor-3 inhibition has novel immunomodulatory and antiarteriosclerotic effects in cardiac allografts.
- Author
-
Nykänen AI, Sandelin H, Krebs R, Keränen MA, Tuuminen R, Kärpänen T, Wu Y, Pytowski B, Koskinen PK, Ylä-Herttuala S, Alitalo K, and Lemström KB
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antigen-Presenting Cells, Arteriosclerosis drug therapy, Cell Movement immunology, Graft Rejection immunology, Mice, Mice, Knockout, Rats, Signal Transduction immunology, Vascular Endothelial Growth Factor Receptor-3 immunology, Arteriosclerosis prevention & control, Chemokine CCL21 biosynthesis, Heart Transplantation immunology, Immunomodulation drug effects, Lymphatic Vessels metabolism, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors
- Abstract
Background: Lymphatic network and chemokine-mediated signals are essential for leukocyte traffic during the proximal steps of alloimmune response. We aimed to determine the role of lymphatic vessels and their principal growth signaling pathway, vascular endothelial growth factor (VEGF)-C/D/VEGFR-3, during acute and chronic rejection in cardiac allografts., Methods and Results: Analysis of heterotopically transplanted rat cardiac allografts showed that chronic rejection increased VEGF-C(+) inflammatory cell and hyaluronan receptor-1 (LYVE-1)(+) lymphatic vessel density. Allograft lymphatic vessels were VEGFR-3(+), contained antigen-presenting cells, and produced dendritic cell chemokine CCL21. Experiments with VEGFR-3/LacZ mice or mice with green fluorescent protein-positive bone marrow cells as cardiac allograft recipients showed that allograft lymphatic vessels originated almost exclusively from donor cells. Intraportal adenoviral VEGFR-3-Ig (Ad.VEGFR-3-Ig/VEGF-C/D-Trap) perfusion was used to inhibit VEGF-C/D/VEGFR-3 signaling. Recipient treatment with Ad.VEGFR-3-Ig prolonged rat cardiac allograft survival. Ad.VEGFR-3-Ig did not affect allograft lymphangiogenesis but was linked to reduced CCL21 production and CD8(+) effector cell entry in the allograft. Concomitantly, Ad.VEGFR-3-Ig reduced OX62(+) dendritic cell recruitment and increased transcription factor Foxp3 expression in the spleen. In separate experiments, treatment with a neutralizing monoclonal VEGFR-3 antibody reduced arteriosclerosis, the number of activated lymphatic vessels expressing VEGFR-3 and CCL21, and graft-infiltrating CD4(+) T cells in chronically rejecting mouse cardiac allografts., Conclusions: These results show that VEGFR-3 participates in immune cell traffic from peripheral tissues to secondary lymphoid organs by regulating allograft lymphatic vessel CCL21 production and suggest VEGFR-3 inhibition as a novel lymphatic vessel-targeted immunomodulatory therapy for cardiac allograft rejection and arteriosclerosis.
- Published
- 2010
- Full Text
- View/download PDF
45. Inhibition of lymphangiogenesis and lymphatic drainage via vascular endothelial growth factor receptor 3 blockade increases the severity of inflammation in a mouse model of chronic inflammatory arthritis.
- Author
-
Guo R, Zhou Q, Proulx ST, Wood R, Ji RC, Ritchlin CT, Pytowski B, Zhu Z, Wang YJ, Schwarz EM, and Xing L
- Subjects
- Animals, Antibodies pharmacology, Arthritis pathology, Arthritis physiopathology, CD11b Antigen metabolism, Chronic Disease, Disease Models, Animal, Disease Progression, Inflammation pathology, Inflammation physiopathology, Lymph Nodes drug effects, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neovascularization, Physiologic drug effects, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Vascular Endothelial Growth Factor C metabolism, Vascular Endothelial Growth Factor Receptor-3 metabolism, Arthritis metabolism, Inflammation metabolism, Lymph Nodes blood supply, Lymph Nodes metabolism, Neovascularization, Physiologic physiology, Severity of Illness Index, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors
- Abstract
Objective: This study was undertaken to investigate the effect of lymphatic inhibition on joint and draining lymph node (LN) pathology during the course of arthritis progression in mice., Methods: Tumor necrosis factor (TNF)-transgenic mice were used as a model of chronic inflammatory arthritis. Mice were subjected to contrast-enhanced magnetic resonance imaging to obtain ankle and knee joint synovial volumes and draining popliteal LN volumes before and after 8 weeks of treatment with vascular endothelial growth factor receptor 3 (VEGFR-3) neutralizing antibody, VEGFR-2 neutralizing antibody, or isotype IgG. Animals were subjected to near-infrared lymphatic imaging to determine the effect of VEGFR-3 neutralization on lymph transport from paws to draining popliteal LNs. Histologic, immunohistochemical, and reverse transcriptase-polymerase chain reaction analyses were used to examine lymphatic vessel formation and the morphology of joints and popliteal LNs., Results: Compared with IgG treatment, VEGFR-3 neutralizing antibody treatment significantly decreased the size of popliteal LNs, the number of lymphatic vessels in joints and popliteal LNs, lymphatic drainage from paws to popliteal LNs, and the number of VEGF-C-expressing CD11b+ myeloid cells in popliteal LNs. However, it increased the synovial volume and area of inflammation in ankle and knee joints. VEGFR-2 neutralizing antibody, in contrast, inhibited both lymphangiogenesis and joint inflammation., Conclusion: These findings indicate that lymphangiogenesis and lymphatic drainage are reciprocally related to the severity of joint lesions during the development of chronic arthritis. Lymphatic drainage plays a beneficial role in controlling the progression of chronic inflammation.
- Published
- 2009
- Full Text
- View/download PDF
46. Vascular endothelial growth factor receptor 2 (VEGFR-2) functions to promote uterine decidual angiogenesis during early pregnancy in the mouse.
- Author
-
Douglas NC, Tang H, Gomez R, Pytowski B, Hicklin DJ, Sauer CM, Kitajewski J, Sauer MV, and Zimmermann RC
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Apoptosis, Cell Proliferation, Decidua drug effects, Decidua metabolism, Embryonic Development drug effects, Embryonic Development physiology, Female, Fluorescent Antibody Technique, Immunohistochemistry, Male, Mice, Neovascularization, Physiologic drug effects, Ovariectomy, Pregnancy, Reverse Transcriptase Polymerase Chain Reaction, Uterus drug effects, Uterus metabolism, Vascular Endothelial Growth Factor Receptor-1 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-1 metabolism, Vascular Endothelial Growth Factor Receptor-1 physiology, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-2 metabolism, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-3 metabolism, Vascular Endothelial Growth Factor Receptor-3 physiology, Decidua blood supply, Neovascularization, Physiologic physiology, Vascular Endothelial Growth Factor Receptor-2 physiology
- Abstract
Implantation of an embryo induces rapid proliferation and differentiation of uterine stromal cells, forming a new structure, the decidua. One salient feature of decidua formation is a marked increase in maternal angiogenesis. Vascular endothelial growth factor (VEGF)-dependent pathways are active in the ovary, uterus, and embryo, and inactivation of VEGF function in any of these structures might prevent normal pregnancy development. We hypothesized that decidual angiogenesis is regulated by VEGF acting through specific VEGF receptors (VEGFRs). To test this hypothesis, we developed a murine pregnancy model in which systemic administration of a receptor-blocking antibody would act specifically on uterine angiogenesis and not on ovarian or embryonic angiogenesis. In our model, ovarian function was replaced with exogenous progesterone, and blocking antibodies were administered prior to embryonic expression of VEGFRs. After administration of a single dose of the anti-VEGFR-2 antibody during the peri-implantation period, no embryos were detected on embryonic d 10.5. The pregnancy was disrupted because of a significant reduction in decidual angiogenesis, which under physiological conditions peaks on embryonic d 5.5 and 6.5. Inactivation of VEGFR-3 reduced angiogenesis in the primary decidual zone, whereas administration of VEGFR-1 blocking antibodies had no effect. Pregnancy was not disrupted after administration of anti-VEGFR-3 or anti-VEGFR-1 antibodies. Thus, the VEGF/VEGFR-2 pathway plays a key role in the maintenance of early pregnancy through its regulation of peri-implantation angiogenesis in the uterine decidua. This newly formed decidual vasculature serves as the first exchange apparatus for the developing embryo until the placenta becomes functionally active.
- Published
- 2009
- Full Text
- View/download PDF
47. Tumor necrosis factor-alpha and interleukin-1 antagonists alleviate inflammatory skin changes associated with epidermal growth factor receptor antibody therapy in mice.
- Author
-
Surguladze D, Deevi D, Claros N, Corcoran E, Wang S, Plym MJ, Wu Y, Doody J, Mauro DJ, Witte L, Busam KJ, Pytowski B, Rodeck U, and Tonra JR
- Subjects
- Animals, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Antibodies, Monoclonal therapeutic use, Dermatitis etiology, Dermatitis pathology, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, ErbB Receptors antagonists & inhibitors, Etanercept, Exanthema chemically induced, Exanthema prevention & control, Female, Humans, Immunoglobulin G pharmacology, Interleukin 1 Receptor Antagonist Protein pharmacology, Interleukin-1 genetics, Interleukin-1 metabolism, Mice, Mice, SCID, Receptors, Tumor Necrosis Factor, Reverse Transcriptase Polymerase Chain Reaction, Skin drug effects, Skin metabolism, Skin pathology, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Antibodies, Monoclonal adverse effects, Dermatitis prevention & control, ErbB Receptors immunology, Interleukin-1 antagonists & inhibitors, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Cancer patients receiving epidermal growth factor receptor (EGFR) antibody therapy often experience an acneiform rash of uncertain etiology in skin regions rich in pilosebaceous units. Currently, this condition is treated symptomatically with very limited, often anecdotal success. Here, we show that a monoclonal antibody targeting murine EGFR, ME1, caused a neutrophil-rich hair follicle inflammation in mice, similar to that reported in patients. This effect was preceded by the appearance of lipid-filled hair follicle distensions adjacent to enlarged sebaceous glands. The cytokine tumor necrosis factor-alpha (TNFalpha), localized immunohistochemically to this affected region of the pilosebaceous unit, was specifically up-regulated by ME1 in skin but not in other tissues examined. Moreover, skin inflammation was reduced by cotreatment with the TNFalpha signaling inhibitor, etanercept, indicating the involvement of TNFalpha in this inflammatory process. Interleukin-1, a cytokine that frequently acts in concert with TNFalpha, is also involved in this process given the efficacy of the interleukin-1 antagonist Kineret. Our results provide a mechanistic framework to develop evidence-based trials for EGFR antibody-induced skin rash in patients with cancer.
- Published
- 2009
- Full Text
- View/download PDF
48. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells.
- Author
-
Hooper AT, Butler JM, Nolan DJ, Kranz A, Iida K, Kobayashi M, Kopp HG, Shido K, Petit I, Yanger K, James D, Witte L, Zhu Z, Wu Y, Pytowski B, Rosenwaks Z, Mittal V, Sato TN, and Rafii S
- Subjects
- Animals, Ataxin-1, Ataxins, Blood Vessels physiology, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Endothelium, Vascular radiation effects, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Mice, Mice, Knockout, Nerve Tissue Proteins biosynthesis, Nuclear Proteins biosynthesis, Sequence Deletion, Signal Transduction, Vascular Endothelial Growth Factor Receptor-2 genetics, Whole-Body Irradiation, Bone Marrow blood supply, Endothelium, Vascular physiology, Hematopoiesis, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells physiology, Regeneration, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Myelosuppression damages the bone marrow (BM) vascular niche, but it is unclear how regeneration of bone marrow vessels contributes to engraftment of transplanted hematopoietic stem and progenitor cells (HSPCs) and restoration of hematopoiesis. We found that chemotherapy and sublethal irradiation induced minor regression of BM sinusoidal endothelial cells (SECs), while lethal irradiation induced severe regression of SECs and required BM transplantation (BMT) for regeneration. Within the BM, VEGFR2 expression specifically demarcated a continuous network of arterioles and SECs, with arterioles uniquely expressing Sca1 and SECs uniquely expressing VEGFR3. Conditional deletion of VEGFR2 in adult mice blocked regeneration of SECs in sublethally irradiated animals and prevented hematopoietic reconstitution. Similarly, inhibition of VEGFR2 signaling in lethally irradiated wild-type mice rescued with BMT severely impaired SEC reconstruction and prevented engraftment and reconstitution of HSPCs. Therefore, regeneration of SECs via VEGFR2 signaling is essential for engraftment of HSPCs and restoration of hematopoiesis.
- Published
- 2009
- Full Text
- View/download PDF
49. Anti-VEGF agents confer survival advantages to tumor-bearing mice by improving cancer-associated systemic syndrome.
- Author
-
Xue Y, Religa P, Cao R, Hansen AJ, Lucchini F, Jones B, Wu Y, Zhu Z, Pytowski B, Liang Y, Zhong W, Vezzoni P, Rozell B, and Cao Y
- Subjects
- Anemia physiopathology, Animals, Capillary Permeability, Humans, Immunohistochemistry, Liver physiopathology, Mice, Neoplasms, Experimental blood supply, Neoplasms, Experimental physiopathology, Neoplasms, Experimental prevention & control, Angiogenesis Inhibitors therapeutic use, Antineoplastic Agents therapeutic use, Neoplasms, Experimental drug therapy, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
The underlying mechanism by which anti-VEGF agents prolong cancer patient survival is poorly understood. We show that in a mouse tumor model, VEGF systemically impairs functions of multiple organs including those in the hematopoietic and endocrine systems, leading to early death. Anti-VEGF antibody, bevacizumab, and anti-VEGF receptor 2 (VEGFR-2), but not anti-VEGFR-1, reversed VEGF-induced cancer-associated systemic syndrome (CASS) and prevented death in tumor-bearing mice. Surprisingly, VEGFR2 blockage improved survival by rescuing mice from CASS without significantly compromising tumor growth, suggesting that "off-tumor" VEGF targets are more sensitive than the tumor vasculature to anti-VEGF drugs. Similarly, VEGF-induced CASS occurred in a spontaneous breast cancer mouse model overexpressing neu. Clinically, VEGF expression and CASS severity positively correlated in various human cancers. These findings define novel therapeutic targets of anti-VEGF agents and provide mechanistic insights into the action of this new class of clinically available anti-VEGF cancer drugs.
- Published
- 2008
- Full Text
- View/download PDF
50. Suppression of prostate cancer nodal and systemic metastasis by blockade of the lymphangiogenic axis.
- Author
-
Burton JB, Priceman SJ, Sung JL, Brakenhielm E, An DS, Pytowski B, Alitalo K, and Wu L
- Subjects
- Animals, Genetic Therapy, Humans, Immunotherapy, Lentivirus genetics, Lung Neoplasms prevention & control, Lung Neoplasms secondary, Lymph Nodes pathology, Lymphatic Metastasis, Male, Mice, Mice, SCID, Neovascularization, Pathologic pathology, Neovascularization, Pathologic therapy, Prostatic Neoplasms blood supply, Prostatic Neoplasms genetics, Solubility, Tumor Cells, Cultured, Vascular Endothelial Growth Factor C antagonists & inhibitors, Vascular Endothelial Growth Factor C physiology, Vascular Endothelial Growth Factor Receptor-2 immunology, Vascular Endothelial Growth Factor Receptor-3 genetics, Vascular Endothelial Growth Factor Receptor-3 immunology, Xenograft Model Antitumor Assays, Lymphangiogenesis genetics, Lymphangiogenesis physiology, Prostatic Neoplasms pathology, Prostatic Neoplasms therapy, Vascular Endothelial Growth Factor Receptor-3 therapeutic use
- Abstract
Lymph node involvement denotes a poor outcome for patients with prostate cancer. Our group, along with others, has shown that initial tumor cell dissemination to regional lymph nodes via lymphatics also promotes systemic metastasis in mouse models. The aim of this study was to investigate the efficacy of suppressive therapies targeting either the angiogenic or lymphangiogenic axis in inhibiting regional lymph node and systemic metastasis in subcutaneous and orthotopic prostate tumor xenografts. Both androgen-dependent and more aggressive androgen-independent prostate tumors were used in our investigations. Interestingly, we observed that the threshold for dissemination is lower in the vascular-rich prostatic microenvironment compared with subcutaneously grafted tumors. Both vascular endothelial growth factor-C (VEGF-C) ligand trap (sVEGFR-3) and antibody directed against VEGFR-3 (mF4-31C1) significantly reduced tumor lymphangiogenesis and metastasis to regional lymph nodes and distal vital organs without influencing tumor growth. Conversely, angiogenic blockade by short hairpin RNA against VEGF or anti-VEGFR-2 antibody (DC101) reduced tumor blood vessel density, significantly delayed tumor growth, and reduced systemic metastasis, although it was ineffective in reducing lymphangiogenesis or nodal metastasis. Collectively, these data clarify the utility of vascular therapeutics in prostate tumor growth and metastasis, particularly in the context of the prostate microenvironment. Our findings highlight the importance of lymphangiogenic therapies in the control of regional lymph node and systemic metastasis.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.