Skip to main content
Erschienen in: Vascular Cell 1/2011

Open Access 01.12.2011 | Review

Anti-DLL4, a cancer therapeutic with multiple mechanisms of action

verfasst von: Austin Gurney, Timothy Hoey

Erschienen in: Vascular Cell | Ausgabe 1/2011

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

DLL4 is a ligand for the Notch family of receptors. DLL4 has many important functions in normal development and tissue homeostasis, including roles in the immune system, the gastro-intestinal tract, and in vascular development. Because of the importance of Notch signaling in stem cell biology, DLL4 has been investigated for its role in the maintenance and proliferation of cancer stem cells (CSC). In addition, its important role in angiogenesis has been investigated for utility as an anti-angiogenic agent. Preclinical studies have highlighted that both anti-CSC and anti-angiogenic activities contribute to its anti-tumor efficacy, and have supported the clinical development of anti-DLL4 antibody for the treatment of cancer.
Hinweise

Competing interests

AG and TH are employees of OncoMed Pharmaceuticals which funds research described in this manuscript.

Authors' contributions

AG and TH wrote the manuscript. All authors read and approved the final manuscript.

Introduction

Cancer can be considered as an aberrant and mutant recapitulation of normal tissue development and homeostasis. Tumors are typically highly heterogeneous at the cellular level, and this heterogeneity frequently mirrors the cellular heterogeneity of the normal tissue. Normal tissue development and homeostasis is driven by an organized hierarchy of stem and progenitor populations which give rise to various differentiated cell types with specialized functions. Long term tissue maintenance is enabled by the unique ability of the stem cell to exhibit self-renewal, which is defined as the ability to proliferate while maintaining pluripotency. Similarly, cancer cells inappropriately activate self-renewal pathways and this enables their ability to grow indefinitely. Thus, the potential connections between normal stem cells and cancer have great importance for understanding tumor biology and also for developing new therapeutic strategies. In the past decade it has become increasingly clear that this self-renewal property is not possessed by all cells within a tumor, but that there exists a subpopulation of cells, often referred to as "cancer stem cells" or "tumor initiating cells" which possesses the ability to undergo self-renewal and thereby drive the growth of the tumor [13]. These cells also possess the ability to initiate the growth of new tumors that recapitulate the heterogeneity of the parent tumor. Thus, these cells have hallmark capabilities analogous to normal stem cells. This relationship has been strengthened by genetic studies which have shown that normal stem cells can be the cell of origin for tumors [4]. Additionally, cancer initiating mutations can originate in more differentiated cells and confer stem-like properties on the tumor cells. These connections have led to a careful examination of stem cell signaling pathways and their role in cancer. Intriguingly, several of these pathways, including the Notch and Wnt pathways, have long been recognized to be activated by oncogenic mutations and disregulated in cancer.
In addition to cancer's need to achieve the stem cell-like property of self-renewal, a cancer must also recruit a support system of stroma and vasculature. The development of vasculature is a complex developmental process, analogous to the development of organs, and so it is not surprising to note that here too signaling pathways important to stem cells, including Notch, have important role in cell fate decisions. There are roles recognized for multiple Notch receptors and multiple Notch ligands within this process [58]. These molecules play roles both within the endothelial cell layer, where they are involved in vessel branching and maturation, and in the surrounding pericyte and smooth muscle layers. Jagged1 and Notch3 are of particular importance in pericyte function [911]. DLL4 acting through Notch1 and Notch4 appears to play key roles regulating endothelial cells and bone marrow-derived endothelial cell progenitors during normal and tumor angiogenesis [12, 13].
These two lines of research, the role of the Notch pathway in the maintenance of cancer stem cells, and the activity of Notch in tumor vasculature, have led to intense research interest in targeting components of this pathway for the development of novel therapeutics. Through this effort, DLL4 has emerged as a compelling target. Indeed, an antibody to DLL4, OMP-21M18, was the first therapeutic entity that selectively targeted the Notch pathway to enter human clinical trials. Gamma-secretase inhibitors (GSIs) that inhibit the ligand-dependent cleavage of Notch receptors have also been developed as anti-cancer therapies. Treatment with GSIs has been found to result in severe gastrointestinal toxicity, limiting their therapeutic utility, due to the combined inhibition of both Notch1 and Notch2 within the stem-progenitor compartment of the intestinal crypt [14, 15]. In addition to processing Notch proteins, gamma-secretase cleaves many other membrane proteins and is involved in a large number of signaling pathways apart from Notch, and these pleiotropic effects are also likely to contribute to the toxicity of GSIs [16, 17].
DLL4 is one of three delta-like ligands in the mammalian genome [18]. Similar to Drosophila Delta, it acts as an agonist ligand to modulate the activity of Notch receptors. Binding studies and in vitro signal transduction studies indicate that DLL4 is readily able to signal through each of the four human Notch receptors, demonstrating its potential to participate in many of the diverse functions that have been described for the Notch family. Elucidating the full range of its activities is an ongoing area of investigation, and is somewhat complicated by the potential for compensatory action by other Notch ligands. Further complicating this analysis is the potential for DLL4 to be expressed in minor subsets of cells, such as stem cells and progenitors, which may lead to an under-appreciation of DLL4 expression and its relevance to a particular tissue. LacZ reporter studies in developing mouse embryos have revealed expression with the vascular system, multiple structures within the nervous system, the gastrointestinal system, the glomeruli of the kidney, and the thymus [19] Gene disruption studies indicate that DLL4 plays an important role in angiogenesis - haploinsufficiency results in embryonic lethality due to angiogenic defects [2022]. Conditional gene disruption studies have also demonstrated critical roles for DLL4 in T cell development [23, 24]. In addition, DLL4 is also expressed by myeloid cells including macrophage and dendritic cells and plays a role in modulating adaptive immune response [2527]. DLL4 is one of the ligands important for lineage commitment within the epithelial cell lining of the gastrointestinal tract. Interestingly, DLL1 and DLL4 appear to have redundant functions in normal intestinal development and homeostasis [28], which accounts for the fact that selective inhibition of DLL4 has minimal impact on the function of the GI tract.

Mechanism of DLL4 in Angiogenesis

Several groups have reported the discovery of antibodies which block the ability of DLL4 to activate Notch [2931]. This work contributed to the discovery that DLL4-Notch signaling is part of a negative feedback loop in the angiogenic process. Blockade of DLL4-Notch binding or loss of function of DLL4 leads to an up-regulation of VEGF signaling resulting in deregulated, hyperproliferation of the tip cells ultimately leading to immature vessels that lack a functional lumen. It has been shown that DLL4 inhibition can have a widespread effect in reducing tumor growth in a number of different xenograft models through this angiogenic mechanism [29, 30]. Inhibition of VEGF signaling is now a well established strategy for anti-cancer therapeutics [32, 33]. The work on DLL4 shows that up-regulation of VEGF in tumor vasculature can also have a therapeutic benefit. Tumors responsive to anti-DLL4 include those that are resistant to inhibition with anti-VEGF highlighting the potential utility of this approach.
We now have a fairly detailed understanding of the mechanism of action for how disrupting DLL4-Notch signaling leads to non-functional vasculature and thereby reduces tumor growth. Normally, DLL4-Notch signaling restricts the numbers of tip cells in response to VEGF, whereas inhibition of DLL4 leads to increased tip cell formation and reduced numbers of stalk cells in angiogenic regions [34]. Inhibition of DLL4 up-regulates VEGF expression as well as its receptors VEGFR2 and VEGFR3. The effect of DLL4 on sprouting angiogenesis is also mediated through regulation of matrix metalloproteinase (MMP) expression [35].

Role of DLL4 in Tumor Initiating Cells

In addition to its role in the tumor vasculature, evidence has accumulated indicating an important role for DLL4-Notch signaling in tumor cells [36, 37]. Across a range of major tumor types, including colorectal, kidney, breast and lung cancer, DLL4 protein expressed by tumor cells can be detected in a high percentage of patient samples [38]. An anti-DLL4 antibody, OMP-21M18, was found to have anti-tumor activity in patient-derived xenograft models independent of any effect on angiogenesis [27]. This antibody does not cross-react with murine DLL4 and therefore has no effect on the vasculature in murine xenografts. The combination of blocking DLL4 in tumor and vascular cells was shown to have an additive effect. Importantly, inhibition of DLL4 not only reduced tumor growth but also reduced tumor initiating cells as shown by serial transplantation experiments [31]. Treatment of colon tumors with anti-DLL4 up-regulates markers of more differentiated colon cells (for example, ATOH1 and Chromogranin A) indicating that DLL4-Notch inhibition limits the stem/progenitor-like properties of colon tumor cells and promotes a more differentiated phenotype. Potential insight into how DLL4-Notch signaling might function in colon cancer is provided by the fact that in normal colon development DLL4 is expressed on the paneth cells [39] which are adjacent to the stem cells and thought to play an important role as the niche to support stem cell maintenance and self-renewal through activation of Notch signaling.
Anti-DLL4 treatment was shown to have synergistic activity with various chemotherapeutic agents in reducing tumor volume and tumor initiating cell frequency [31]. These data are consistent with the hypothesis that the less differentiated, stem-like cancer cells are resistant to conventional cancer treatments including chemotherapy whereas promoting differentiation sensitizes tumor cells to the cytotoxic effects of chemotherapeutic drugs. More recently, anti-DLL4 was found to be active in a number of colon tumor xenografts including those harboring KRAS mutations which are insensitive to anti-EGFR treatment [40]. Since tumorigenic CSCs are thought to mediate tumor recurrence after treatment and the metastatic spread of the disease, agents that block key CSC self-renewal hold tremendous promise as improved cancer treatments.

Perspective

Anti-DLL4 attacks cancers through two distinct mechanisms, an anti-angiogenic effect and a reduction of CSCs, each fundamental for malignant tumor growth. This target offers tremendous potential for improved efficacy in cancer treatment. As discussed above, inhibition of DLL4 signaling has been shown to result endothelial cell hyperproliferation and increased vascular sprouting in tumor angiogenesis. In rodents, DLL4 inhibition or loss of function has been associated with certain adverse events including vascular hyperproliferation in the liver and non-malignant vascular neoplasms [41, 42]. These data raise concerns about potential toxicities associated with anti-DLL4 therapy in the clinic linked to its mechanisms of action in the vasculature and/or in stem cell biology. At least two anti-DLL4 antibodies have entered clinical testing (OMP-21M18 from OncoMed and REGN421 from Regeneron). To date there have not been reported incidents of either liver toxicity or vascular neoplasms in these clinical programs. None-the-less, DLL4 is an important ligand within a fundamental signaling pathway, and a molecule that robustly impacts vascular development, so it would not be surprising if adverse consequences to DLL4 inhibition are observed during clinical testing. Ongoing clinical studies will determine if this approach in blocking DLL4 can be developed into an effective therapeutic. As with all cancer therapeutics, identifying the right disease setting, dosing regimen, and combination strategy will be critical for successful drug development. It has been nearly 25 years since Notch was implicated in cancer by the discovery of Notch4/int-3 as a mammary proto-oncogene following activation by mouse mammary tumor virus (MMTV) integration [43, 44]. With our increasing understanding of the function of this pathway in both normal cell fate decisions and in cancer, anti-DLL4 may become the prototype member of a new class of therapeutic agents.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

AG and TH are employees of OncoMed Pharmaceuticals which funds research described in this manuscript.

Authors' contributions

AG and TH wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Clevers H: The cancer stem cell: premises, promises and challenges. Nat Med. 2011, 17: 313-319.CrossRefPubMed Clevers H: The cancer stem cell: premises, promises and challenges. Nat Med. 2011, 17: 313-319.CrossRefPubMed
2.
Zurück zum Zitat O'Brien CA, Kreso A, Jamieson CH: Cancer stem cells and self-renewal. Clin Cancer Res. 2010, 16: 3113-3120. 10.1158/1078-0432.CCR-09-2824.CrossRefPubMed O'Brien CA, Kreso A, Jamieson CH: Cancer stem cells and self-renewal. Clin Cancer Res. 2010, 16: 3113-3120. 10.1158/1078-0432.CCR-09-2824.CrossRefPubMed
3.
Zurück zum Zitat Clarke MF: Self-renewal and solid-tumor stem cells. Biol Blood Marrow Transplant. 2005, 11: 14-16.CrossRefPubMed Clarke MF: Self-renewal and solid-tumor stem cells. Biol Blood Marrow Transplant. 2005, 11: 14-16.CrossRefPubMed
4.
Zurück zum Zitat Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M, Danenberg E, Clarke AR, Sansom OJ, Clevers H: Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 2009, 457: 608-611. 10.1038/nature07602.CrossRefPubMed Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M, Danenberg E, Clarke AR, Sansom OJ, Clevers H: Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 2009, 457: 608-611. 10.1038/nature07602.CrossRefPubMed
5.
Zurück zum Zitat Kume T: Novel insights into the differential functions of Notch ligands in vascular formation. J Angiogenes Res. 2009, 1: 8-10.1186/2040-2384-1-8.PubMedCentralCrossRefPubMed Kume T: Novel insights into the differential functions of Notch ligands in vascular formation. J Angiogenes Res. 2009, 1: 8-10.1186/2040-2384-1-8.PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Roca C, Adams RH: Regulation of vascular morphogenesis by Notch signaling. Genes Dev. 2007, 21: 2511-2524. 10.1101/gad.1589207.CrossRefPubMed Roca C, Adams RH: Regulation of vascular morphogenesis by Notch signaling. Genes Dev. 2007, 21: 2511-2524. 10.1101/gad.1589207.CrossRefPubMed
8.
Zurück zum Zitat Hofmann JJ, Iruela-Arispe ML: Notch signaling in blood vessels: who is talking to whom about what?. Circ Res. 2007, 100: 1556-1568. 10.1161/01.RES.0000266408.42939.e4.CrossRefPubMed Hofmann JJ, Iruela-Arispe ML: Notch signaling in blood vessels: who is talking to whom about what?. Circ Res. 2007, 100: 1556-1568. 10.1161/01.RES.0000266408.42939.e4.CrossRefPubMed
9.
Zurück zum Zitat Liu H, Zhang W, Kennard S, Caldwell RB, Lilly B: Notch3 is critical for proper angiogenesis and mural cell investment. Circ Res. 2010, 107: 860-870. 10.1161/CIRCRESAHA.110.218271.PubMedCentralCrossRefPubMed Liu H, Zhang W, Kennard S, Caldwell RB, Lilly B: Notch3 is critical for proper angiogenesis and mural cell investment. Circ Res. 2010, 107: 860-870. 10.1161/CIRCRESAHA.110.218271.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Liu H, Kennard S, Lilly B: NOTCH3 expression is induced in mural cells through an autoregulatory loop that requires endothelial-expressed JAGGED1. Circ Res. 2009, 104: 466-475. 10.1161/CIRCRESAHA.108.184846.PubMedCentralCrossRefPubMed Liu H, Kennard S, Lilly B: NOTCH3 expression is induced in mural cells through an autoregulatory loop that requires endothelial-expressed JAGGED1. Circ Res. 2009, 104: 466-475. 10.1161/CIRCRESAHA.108.184846.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Dufraine J, Funahashi Y, Kitajewski J: Notch signaling regulates tumor angiogenesis by diverse mechanisms. Oncogene. 2008, 27: 5132-5137. 10.1038/onc.2008.227.PubMedCentralCrossRefPubMed Dufraine J, Funahashi Y, Kitajewski J: Notch signaling regulates tumor angiogenesis by diverse mechanisms. Oncogene. 2008, 27: 5132-5137. 10.1038/onc.2008.227.PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Real C, Remedio L, Caiado F, Igreja C, Borges C, Trindade A, Pinto-do OP, Yagita H, Duarte A, Dias S: Bone marrow-derived endothelial progenitors expressing delta-like 4 (dll4) regulate tumor angiogenesis. PLoS One. 2011, 6: e18323-10.1371/journal.pone.0018323.PubMedCentralCrossRefPubMed Real C, Remedio L, Caiado F, Igreja C, Borges C, Trindade A, Pinto-do OP, Yagita H, Duarte A, Dias S: Bone marrow-derived endothelial progenitors expressing delta-like 4 (dll4) regulate tumor angiogenesis. PLoS One. 2011, 6: e18323-10.1371/journal.pone.0018323.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Milano J, McKay J, Dagenais C, Foster-Brown L, Pognan F, Gadient R, Jacobs RT, Zacco A, Greenberg B, Ciaccio PJ: Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation. Toxicol Sci. 2004, 82: 341-358. 10.1093/toxsci/kfh254.CrossRefPubMed Milano J, McKay J, Dagenais C, Foster-Brown L, Pognan F, Gadient R, Jacobs RT, Zacco A, Greenberg B, Ciaccio PJ: Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation. Toxicol Sci. 2004, 82: 341-358. 10.1093/toxsci/kfh254.CrossRefPubMed
15.
Zurück zum Zitat Imbimbo BP: Therapeutic potential of gamma-secretase inhibitors and modulators. Curr Top Med Chem. 2008, 8: 54-61. 10.2174/156802608783334015.CrossRefPubMed Imbimbo BP: Therapeutic potential of gamma-secretase inhibitors and modulators. Curr Top Med Chem. 2008, 8: 54-61. 10.2174/156802608783334015.CrossRefPubMed
16.
Zurück zum Zitat Beel AJ, Sanders CR: Substrate specificity of gamma-secretase and other intramembrane proteases. Cell Mol Life Sci. 2008, 65: 1311-1334. 10.1007/s00018-008-7462-2.PubMedCentralCrossRefPubMed Beel AJ, Sanders CR: Substrate specificity of gamma-secretase and other intramembrane proteases. Cell Mol Life Sci. 2008, 65: 1311-1334. 10.1007/s00018-008-7462-2.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Hemming ML, Elias JE, Gygi SP, Selkoe DJ: Proteomic profiling of gamma-secretase substrates and mapping of substrate requirements. PLoS Biol. 2008, 6: e257-10.1371/journal.pbio.0060257.PubMedCentralCrossRefPubMed Hemming ML, Elias JE, Gygi SP, Selkoe DJ: Proteomic profiling of gamma-secretase substrates and mapping of substrate requirements. PLoS Biol. 2008, 6: e257-10.1371/journal.pbio.0060257.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Bray SJ: Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006, 7: 678-689. 10.1038/nrm2009.CrossRefPubMed Bray SJ: Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006, 7: 678-689. 10.1038/nrm2009.CrossRefPubMed
19.
Zurück zum Zitat Benedito R, Duarte A: Expression of Dll4 during mouse embryogenesis suggests multiple developmental roles. Gene Expr Patterns. 2005, 5: 750-755. 10.1016/j.modgep.2005.04.004.CrossRefPubMed Benedito R, Duarte A: Expression of Dll4 during mouse embryogenesis suggests multiple developmental roles. Gene Expr Patterns. 2005, 5: 750-755. 10.1016/j.modgep.2005.04.004.CrossRefPubMed
20.
Zurück zum Zitat Duarte A, Hirashima M, Benedito R, Trindade A, Diniz P, Bekman E, Costa L, Henrique D, Rossant J: Dosage-sensitive requirement for mouse Dll4 in artery development. Genes Dev. 2004, 18: 2474-2478. 10.1101/gad.1239004.PubMedCentralCrossRefPubMed Duarte A, Hirashima M, Benedito R, Trindade A, Diniz P, Bekman E, Costa L, Henrique D, Rossant J: Dosage-sensitive requirement for mouse Dll4 in artery development. Genes Dev. 2004, 18: 2474-2478. 10.1101/gad.1239004.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Gale NW, Dominguez MG, Noguera I, Pan L, Hughes V, Valenzuela DM, Murphy AJ, Adams NC, Lin HC, Holash J, et al: Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci USA. 2004, 101: 15949-15954. 10.1073/pnas.0407290101.PubMedCentralCrossRefPubMed Gale NW, Dominguez MG, Noguera I, Pan L, Hughes V, Valenzuela DM, Murphy AJ, Adams NC, Lin HC, Holash J, et al: Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci USA. 2004, 101: 15949-15954. 10.1073/pnas.0407290101.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Krebs LT, Shutter JR, Tanigaki K, Honjo T, Stark KL, Gridley T: Haploinsufficient lethality and formation of arteriovenous malformations in Notch pathway mutants. Genes Dev. 2004, 18: 2469-2473. 10.1101/gad.1239204.PubMedCentralCrossRefPubMed Krebs LT, Shutter JR, Tanigaki K, Honjo T, Stark KL, Gridley T: Haploinsufficient lethality and formation of arteriovenous malformations in Notch pathway mutants. Genes Dev. 2004, 18: 2469-2473. 10.1101/gad.1239204.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Koch U, Fiorini E, Benedito R, Besseyrias V, Schuster-Gossler K, Pierres M, Manley NR, Duarte A, Macdonald HR, Radtke F: Delta-like 4 is the essential, nonredundant ligand for Notch1 during thymic T cell lineage commitment. J Exp Med. 2008, 205: 2515-2523. 10.1084/jem.20080829.PubMedCentralCrossRefPubMed Koch U, Fiorini E, Benedito R, Besseyrias V, Schuster-Gossler K, Pierres M, Manley NR, Duarte A, Macdonald HR, Radtke F: Delta-like 4 is the essential, nonredundant ligand for Notch1 during thymic T cell lineage commitment. J Exp Med. 2008, 205: 2515-2523. 10.1084/jem.20080829.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Hozumi K, Mailhos C, Negishi N, Hirano K, Yahata T, Ando K, Zuklys S, Hollander GA, Shima DT, Habu S: Delta-like 4 is indispensable in thymic environment specific for T cell development. J Exp Med. 2008, 205: 2507-2513. 10.1084/jem.20080134.PubMedCentralCrossRefPubMed Hozumi K, Mailhos C, Negishi N, Hirano K, Yahata T, Ando K, Zuklys S, Hollander GA, Shima DT, Habu S: Delta-like 4 is indispensable in thymic environment specific for T cell development. J Exp Med. 2008, 205: 2507-2513. 10.1084/jem.20080134.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Ito T, Schaller M, Hogaboam CM, Standiford TJ, Sandor M, Lukacs NW, Chensue SW, Kunkel SL: TLR9 regulates the mycobacteria-elicited pulmonary granulomatous immune response in mice through DC-derived Notch ligand delta-like 4. J Clin Invest. 2009, 119: 33-46.PubMedCentralPubMed Ito T, Schaller M, Hogaboam CM, Standiford TJ, Sandor M, Lukacs NW, Chensue SW, Kunkel SL: TLR9 regulates the mycobacteria-elicited pulmonary granulomatous immune response in mice through DC-derived Notch ligand delta-like 4. J Clin Invest. 2009, 119: 33-46.PubMedCentralPubMed
26.
Zurück zum Zitat Kassner N, Krueger M, Yagita H, Dzionek A, Hutloff A, Kroczek R, Scheffold A, Rutz S: Cutting edge: Plasmacytoid dendritic cells induce IL-10 production in T cells via the Delta-like-4/Notch axis. J Immunol. 2010, 184: 550-554. 10.4049/jimmunol.0903152.CrossRefPubMed Kassner N, Krueger M, Yagita H, Dzionek A, Hutloff A, Kroczek R, Scheffold A, Rutz S: Cutting edge: Plasmacytoid dendritic cells induce IL-10 production in T cells via the Delta-like-4/Notch axis. J Immunol. 2010, 184: 550-554. 10.4049/jimmunol.0903152.CrossRefPubMed
27.
Zurück zum Zitat Reynolds ND, Lukacs NW, Long N, Karpus WJ: Delta-Like Ligand 4 Regulates CNS T Cell Accumulation during Experimental Autoimmune Encephalomyelitis. J Immunol. 2011 Reynolds ND, Lukacs NW, Long N, Karpus WJ: Delta-Like Ligand 4 Regulates CNS T Cell Accumulation during Experimental Autoimmune Encephalomyelitis. J Immunol. 2011
28.
Zurück zum Zitat Pellegrinet L, Rodilla V, Liu Z, Chen S, Koch U, Espinosa L, Kaestner KH, Kopan R, Lewis J, Radtke F: Dll1- and dll4-mediated notch signaling are required for homeostasis of intestinal stem cells. Gastroenterology. 2011, 140: 1230-1240 e1237. 10.1053/j.gastro.2011.01.005.PubMedCentralCrossRefPubMed Pellegrinet L, Rodilla V, Liu Z, Chen S, Koch U, Espinosa L, Kaestner KH, Kopan R, Lewis J, Radtke F: Dll1- and dll4-mediated notch signaling are required for homeostasis of intestinal stem cells. Gastroenterology. 2011, 140: 1230-1240 e1237. 10.1053/j.gastro.2011.01.005.PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Ridgway J, Zhang G, Wu Y, Stawicki S, Liang WC, Chanthery Y, Kowalski J, Watts RJ, Callahan C, Kasman I, et al: Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature. 2006, 444: 1083-1087. 10.1038/nature05313.CrossRefPubMed Ridgway J, Zhang G, Wu Y, Stawicki S, Liang WC, Chanthery Y, Kowalski J, Watts RJ, Callahan C, Kasman I, et al: Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature. 2006, 444: 1083-1087. 10.1038/nature05313.CrossRefPubMed
30.
Zurück zum Zitat Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland P, Gale NW, Lin HC, Yancopoulos GD, Thurston G: Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature. 2006, 444: 1032-1037. 10.1038/nature05355.CrossRefPubMed Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland P, Gale NW, Lin HC, Yancopoulos GD, Thurston G: Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature. 2006, 444: 1032-1037. 10.1038/nature05355.CrossRefPubMed
31.
Zurück zum Zitat Hoey T, Yen WC, Axelrod F, Basi J, Donigian L, Dylla S, Fitch-Bruhns M, Lazetic S, Park IK, Sato A, et al: DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency. Cell Stem Cell. 2009, 5: 168-177. 10.1016/j.stem.2009.05.019.CrossRefPubMed Hoey T, Yen WC, Axelrod F, Basi J, Donigian L, Dylla S, Fitch-Bruhns M, Lazetic S, Park IK, Sato A, et al: DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency. Cell Stem Cell. 2009, 5: 168-177. 10.1016/j.stem.2009.05.019.CrossRefPubMed
32.
Zurück zum Zitat Crawford Y, Ferrara N: VEGF inhibition: insights from preclinical and clinical studies. Cell Tissue Res. 2009, 335: 261-269. 10.1007/s00441-008-0675-8.CrossRefPubMed Crawford Y, Ferrara N: VEGF inhibition: insights from preclinical and clinical studies. Cell Tissue Res. 2009, 335: 261-269. 10.1007/s00441-008-0675-8.CrossRefPubMed
33.
Zurück zum Zitat Sharma PS, Sharma R, Tyagi T: VEGF/VEGFR Pathway Inhibitors as Anti-Angiogenic Agents: Present and Future. Curr Cancer Drug Targets. 2011 Sharma PS, Sharma R, Tyagi T: VEGF/VEGFR Pathway Inhibitors as Anti-Angiogenic Agents: Present and Future. Curr Cancer Drug Targets. 2011
34.
Zurück zum Zitat Hellstrom M, Phng LK, Hofmann JJ, Wallgard E, Coultas L, Lindblom P, Alva J, Nilsson AK, Karlsson L, Gaiano N, et al: Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature. 2007, 445: 776-780. 10.1038/nature05571.CrossRefPubMed Hellstrom M, Phng LK, Hofmann JJ, Wallgard E, Coultas L, Lindblom P, Alva J, Nilsson AK, Karlsson L, Gaiano N, et al: Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature. 2007, 445: 776-780. 10.1038/nature05571.CrossRefPubMed
35.
Zurück zum Zitat Funahashi Y, Shawber CJ, Sharma A, Kanamaru E, Choi YK, Kitajewski J: Notch modulates VEGF action in endothelial cells by inducing Matrix Metalloprotease activity. Vasc Cell. 2011, 3: 2-10.1186/2045-824X-3-2.PubMedCentralCrossRefPubMed Funahashi Y, Shawber CJ, Sharma A, Kanamaru E, Choi YK, Kitajewski J: Notch modulates VEGF action in endothelial cells by inducing Matrix Metalloprotease activity. Vasc Cell. 2011, 3: 2-10.1186/2045-824X-3-2.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Mullendore ME, Koorstra JB, Li YM, Offerhaus GJ, Fan X, Henderson CM, Matsui W, Eberhart CG, Maitra A, Feldmann G: Ligand-dependent Notch signaling is involved in tumor initiation and tumor maintenance in pancreatic cancer. Clin Cancer Res. 2009, 15: 2291-2301. 10.1158/1078-0432.CCR-08-2004.PubMedCentralCrossRefPubMed Mullendore ME, Koorstra JB, Li YM, Offerhaus GJ, Fan X, Henderson CM, Matsui W, Eberhart CG, Maitra A, Feldmann G: Ligand-dependent Notch signaling is involved in tumor initiation and tumor maintenance in pancreatic cancer. Clin Cancer Res. 2009, 15: 2291-2301. 10.1158/1078-0432.CCR-08-2004.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Indraccolo S, Minuzzo S, Masiero M, Pusceddu I, Persano L, Moserle L, Reboldi A, Favaro E, Mecarozzi M, Di Mario G, et al: Cross-talk between tumor and endothelial cells involving the Notch3-Dll4 interaction marks escape from tumor dormancy. Cancer Res. 2009, 69: 1314-1323. 10.1158/0008-5472.CAN-08-2791.CrossRefPubMed Indraccolo S, Minuzzo S, Masiero M, Pusceddu I, Persano L, Moserle L, Reboldi A, Favaro E, Mecarozzi M, Di Mario G, et al: Cross-talk between tumor and endothelial cells involving the Notch3-Dll4 interaction marks escape from tumor dormancy. Cancer Res. 2009, 69: 1314-1323. 10.1158/0008-5472.CAN-08-2791.CrossRefPubMed
38.
Zurück zum Zitat Martinez JC, Muller MM, Turley H, Steers G, Choteau L, Li JL, Sainson R, Harris AL, Pezzella F, Gatter KC: Nuclear and membrane expression of the angiogenesis regulator delta-like ligand 4 (DLL4) in normal and malignant human tissues. Histopathology. 2009, 54: 598-606. 10.1111/j.1365-2559.2009.03279.x.CrossRefPubMed Martinez JC, Muller MM, Turley H, Steers G, Choteau L, Li JL, Sainson R, Harris AL, Pezzella F, Gatter KC: Nuclear and membrane expression of the angiogenesis regulator delta-like ligand 4 (DLL4) in normal and malignant human tissues. Histopathology. 2009, 54: 598-606. 10.1111/j.1365-2559.2009.03279.x.CrossRefPubMed
39.
Zurück zum Zitat Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born M, Barker N, Shroyer NF, van de Wetering M, Clevers H: Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011, 469: 415-418. 10.1038/nature09637.PubMedCentralCrossRefPubMed Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born M, Barker N, Shroyer NF, van de Wetering M, Clevers H: Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011, 469: 415-418. 10.1038/nature09637.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Fischer M, Yen WC, Kapoun AM, Wang M, O'Young G, Lewicki J, Gurney A, Hoey T: Anti-DLL4 inhibits growth and reduces tumor-initiating cell frequency in colorectal tumors with oncogenic KRAS mutations. Cancer Res. 2011, 71: 1520-1525. 10.1158/0008-5472.CAN-10-2817.CrossRefPubMed Fischer M, Yen WC, Kapoun AM, Wang M, O'Young G, Lewicki J, Gurney A, Hoey T: Anti-DLL4 inhibits growth and reduces tumor-initiating cell frequency in colorectal tumors with oncogenic KRAS mutations. Cancer Res. 2011, 71: 1520-1525. 10.1158/0008-5472.CAN-10-2817.CrossRefPubMed
41.
Zurück zum Zitat Yan M, Callahan CA, Beyer JC, Allamneni KP, Zhang G, Ridgway JB, Niessen K, Plowman GD: Chronic DLL4 blockade induces vascular neoplasms. Nature. 2010, 463: E6-7. 10.1038/nature08751.CrossRefPubMed Yan M, Callahan CA, Beyer JC, Allamneni KP, Zhang G, Ridgway JB, Niessen K, Plowman GD: Chronic DLL4 blockade induces vascular neoplasms. Nature. 2010, 463: E6-7. 10.1038/nature08751.CrossRefPubMed
42.
Zurück zum Zitat Djokovic D, Trindade A, Gigante J, Badenes M, Silva L, Liu R, Li X, Gong M, Krasnoperov V, Gill PS, Duarte A: Combination of Dll4/Notch and Ephrin-B2/EphB4 targeted therapy is highly effective in disrupting tumor angiogenesis. BMC Cancer. 2010, 10: 641-10.1186/1471-2407-10-641.PubMedCentralCrossRefPubMed Djokovic D, Trindade A, Gigante J, Badenes M, Silva L, Liu R, Li X, Gong M, Krasnoperov V, Gill PS, Duarte A: Combination of Dll4/Notch and Ephrin-B2/EphB4 targeted therapy is highly effective in disrupting tumor angiogenesis. BMC Cancer. 2010, 10: 641-10.1186/1471-2407-10-641.PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Jhappan C, Gallahan D, Stahle C, Chu E, Smith GH, Merlino G, Callahan R: Expression of an activated Notch-related int-3 transgene interferes with cell differentiation and induces neoplastic transformation in mammary and salivary glands. Genes Dev. 1992, 6: 345-355. 10.1101/gad.6.3.345.CrossRefPubMed Jhappan C, Gallahan D, Stahle C, Chu E, Smith GH, Merlino G, Callahan R: Expression of an activated Notch-related int-3 transgene interferes with cell differentiation and induces neoplastic transformation in mammary and salivary glands. Genes Dev. 1992, 6: 345-355. 10.1101/gad.6.3.345.CrossRefPubMed
44.
Zurück zum Zitat Gallahan D, Kozak C, Callahan R: A new common integration region (int-3) for mouse mammary tumor virus on mouse chromosome 17. J Virol. 1987, 61: 218-220.PubMedCentralPubMed Gallahan D, Kozak C, Callahan R: A new common integration region (int-3) for mouse mammary tumor virus on mouse chromosome 17. J Virol. 1987, 61: 218-220.PubMedCentralPubMed
Metadaten
Titel
Anti-DLL4, a cancer therapeutic with multiple mechanisms of action
verfasst von
Austin Gurney
Timothy Hoey
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Vascular Cell / Ausgabe 1/2011
Elektronische ISSN: 2045-824X
DOI
https://doi.org/10.1186/2045-824X-3-18

Weitere Artikel der Ausgabe 1/2011

Vascular Cell 1/2011 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

„Jeder Fall von plötzlichem Tod muss obduziert werden!“

17.05.2024 Plötzlicher Herztod Nachrichten

Ein signifikanter Anteil der Fälle von plötzlichem Herztod ist genetisch bedingt. Um ihre Verwandten vor diesem Schicksal zu bewahren, sollten jüngere Personen, die plötzlich unerwartet versterben, ausnahmslos einer Autopsie unterzogen werden.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Schlechtere Vorhofflimmern-Prognose bei kleinem linken Ventrikel

17.05.2024 Vorhofflimmern Nachrichten

Nicht nur ein vergrößerter, sondern auch ein kleiner linker Ventrikel ist bei Vorhofflimmern mit einer erhöhten Komplikationsrate assoziiert. Der Zusammenhang besteht nach Daten aus China unabhängig von anderen Risikofaktoren.

Semaglutid bei Herzinsuffizienz: Wie erklärt sich die Wirksamkeit?

17.05.2024 Herzinsuffizienz Nachrichten

Bei adipösen Patienten mit Herzinsuffizienz des HFpEF-Phänotyps ist Semaglutid von symptomatischem Nutzen. Resultiert dieser Benefit allein aus der Gewichtsreduktion oder auch aus spezifischen Effekten auf die Herzinsuffizienz-Pathogenese? Eine neue Analyse gibt Aufschluss.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.