Network


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

Hotspot


Dive into the research topics where Zane C. Neal is active.

Publication


Featured researches published by Zane C. Neal.


Journal of Immunology | 2001

Anti-CD40 Antibody Induces Antitumor and Antimetastatic Effects: The Role of NK Cells

Joel G. Turner; Alexander L. Rakhmilevich; Lyudmila Burdelya; Zane C. Neal; Michael Imboden; Paul M. Sondel; Hua Yu

We assessed the effect of the stimulatory anti-CD40 Ab on NK cell activation in vivo and the therapeutic potential of activated NK cells in tumor-bearing mice. Single-dose i.p. injection of the anti-CD40 Ab resulted in production of IL-12 and IFN-γ in vivo, followed by a dramatic increase in NK cell cytolytic activity in PBLs. NK cell activation by anti-CD40 Ab was also observed in CD40 ligand knockout mice. Because NK cells express CD40 ligand but not CD40, our results suggest that NK activation is mediated by increased cytokine production upon CD40 ligation of APCs. Treatment of tumor-bearing mice with anti-CD40 Ab resulted in substantial antitumor and antimetastatic effects in three tumor models. Depletion of NK cells with anti-asialo GM1 Ab reduced or abrogated the observed antitumor effects in all the tested models. These results indicate that a stimulatory CD40 Ab indirectly activates NK cells, which can produce significant antitumor and antimetastatic effects.


Clinical Cancer Research | 2004

Enhanced activity of hu14.18-IL2 immunocytokine against murine NXS2 neuroblastoma when combined with interleukin 2 therapy.

Zane C. Neal; Jeannie C. Yang; Alexander L. Rakhmilevich; Ilia N. Buhtoiarov; Hillary E. Lum; Michael Imboden; Jacquelyn A. Hank; Holger N. Lode; Ralph A. Reisfeld; Stephen D. Gillies; Paul M. Sondel

Established s.c. NXS2 murine neuroblastoma tumors exhibited transient resolution after suboptimal therapy using the hu14.18-IL2 immunocytokine (IC). The hu14.18-IL2 IC is a fusion protein that has linked a molecule of interleukin 2 (IL-2) to the COOH terminus of each of the IgG heavy chains on the humanized anti-GD2 monoclonal antibody hu14.18. To induce more potent and longer lasting in vivo antitumor effects, we tested hu14.18-IL2 IC in a regimen combining it with constant infusion IL-2 in NXS2 tumor-bearing mice. The addition of the constant infusion IL-2 augmented the antitumor response induced by treatment with the hu14.18-IL2 IC in animals with experimentally induced hepatic metastases and in animals bearing localized s.c. tumors. The combined treatment induced prolonged tumor eradication in most animals bearing s.c. tumors and involved both natural killer cells and T cells. The enhanced ability of this combined treatment to prevent tumor recurrence was not observed when a larger dose of hu14.18-IL2 IC, similar in IL-2 content to the IC plus systemic IL-2 regimen, was tested as single-agent therapy. Animals showing prolonged tumor eradication of established tumors after the combined hu14.18-IL2 plus IL-2 regimen exhibited a protective T-cell-dependent antitumor memory response against NXS2 rechallenge.


BioTechniques | 2006

Genetic immunization for antibody generation in research animals by intravenous delivery of plasmid DNA

Mary Kay Bates; Guofeng Zhang; Magdolna G. Sebestyén; Zane C. Neal; Jon A. Wolff; Hans Herweijer

Genetic immunization is an attractive approach to generate antibodies because native proteins are expressed in vivo with normal posttranscriptional modifications, avoiding time-consuming and costly antigen isolation or synthesis. Hydrodynamic tail or limb vein delivery of naked plasmid DNA expression vectors was used to induce antigen-specific antibodies in mice, rats, and rabbits. Both methods allowed the efficient generation of high-titer, antigen-specific antibodies with an overall success rate of Western detectable antibodies of 78% and 92%, respectively. High-titer antibodies were typically present after 3 hydrodynamic tail vein plasmid DNA deliveries, 5 weeks after the initial injection (i.e., prime). For hydrodynamic limb vein plasmid DNA delivery, two deliveries were sufficient to induce high-titer antibody levels. Tail vein delivery was less successful at generating antibodies directed against secreted proteins as compared with limb vein delivery. Material for screening was generated by transfection of the immunization vector into mammalian cell lines. The cell line (COS-7) that produced the highest level of antigen expression performed best in Western blot analysis screens. In summary, intravenous delivery of antigen-expressing plasmid DNA vectors is an effective genetic immunization method for the induction of antigen-specific antibodies in small and large research animals.


Journal of Leukocyte Biology | 2011

Differential internalization of hu14.18‐IL2 immunocytokine by NK and tumor cell: impact on conjugation, cytotoxicity, and targeting

Ilia N. Buhtoiarov; Zane C. Neal; Jacek Gan; Tatiana N. Buhtoiarova; Manish S. Patankar; Jennifer A. A. Gubbels; Jacquelyn A. Hank; Brett Yamane; Alexander L. Rakhmilevich; Ralph A. Reisfeld; Stephen D. Gillies; Paul M. Sondel

The hu14.18‐IL2 (EMD 273063) IC, consisting of a GD2‐specific mAb genetically engineered to two molecules of IL‐2, is in clinical trials for treatment of GD2‐expressing tumors. Anti‐tumor activity of IC in vivo and in vitro involves NK cells. We studied the kinetics of retention of IC on the surface of human CD25+CD16– NK cell lines (NKL and RL12) and GD2+ M21 melanoma after IC binding to the cells via IL‐2R and GD2, respectively. For NK cells, ∼50% of IC was internalized by 3 h and ∼90% by 24 h of cell culture. The decrease of surface IC levels on NK cells correlated with the loss of their ability to bind to tumor cells and mediate antibody‐dependent cellular cytotoxicity in vitro. Unlike NK cells, M21 cells retained ∼70% of IC on the surface following 24 h of culture and maintained the ability to become conjugated and lysed by NK cells. When NKL cells were injected into M21‐bearing SCID mice, IT delivery of IC augmented NK cell migration into the tumor. These studies demonstrate that once IC binds to the tumor, it is present on the tumor surface for a prolonged time, inducing the recruitment of NK cells to the tumor site, followed by tumor cell killing.


Molecular Cancer Therapeutics | 2018

HIF2α targeted RNAi therapeutic inhibits clear cell renal cell carcinoma

So C. Wong; Weijun Cheng; Holly Hamilton; Anthony L. Nicholas; Darren H. Wakefield; Aaron Almeida; Andrei V. Blokhin; Jeffrey C. Carlson; Zane C. Neal; Vladimir Subbotin; Guofeng Zhang; Julia Hegge; Stephanie L. Bertin; Vladimir S. Trubetskoy; David B. Rozema; David L. Lewis; Steven B. Kanner

Targeted therapy against VEGF and mTOR pathways has been established as the standard-of-care for metastatic clear cell renal cell carcinoma (ccRCC); however, these treatments frequently fail and most patients become refractory requiring subsequent alternative therapeutic options. Therefore, development of innovative and effective treatments is imperative. About 80%–90% of ccRCC tumors express an inactive mutant form of the von Hippel-Lindau protein (pVHL), an E3 ubiquitin ligase that promotes target protein degradation. Strong genetic and experimental evidence supports the correlate that pVHL functional loss leads to the accumulation of the transcription factor hypoxia-inducible factor 2α (HIF2α) and that an overabundance of HIF2α functions as a tumorigenic driver of ccRCC. In this report, we describe an RNAi therapeutic for HIF2α that utilizes a targeting ligand that selectively binds to integrins αvβ3 and αvβ5 frequently overexpressed in ccRCC. We demonstrate that functional delivery of a HIF2α-specific RNAi trigger resulted in HIF2α gene silencing and subsequent tumor growth inhibition and degeneration in an established orthotopic ccRCC xenograft model. Mol Cancer Ther; 17(1); 140–9. ©2017 AACR.


Cancer Research | 2001

The Level of MHC Class I Expression on Murine Adenocarcinoma Can Change the Antitumor Effector Mechanism of Immunocytokine Therapy

Michael Imboden; Kristopher R. Murphy; Alexander L. Rakhmilevich; Zane C. Neal; Rong Xiang; Ralph A. Reisfeld; Stephen D. Gillies; Paul M. Sondel


Cancer Immunology, Immunotherapy | 2004

NXS2 murine neuroblastomas express increased levels of MHC class I antigens upon recurrence following NK-dependent immunotherapy

Zane C. Neal; Michael Imboden; Alexander L. Rakhmilevich; KyungMann Kim; Jacquelyn A. Hank; Jean E. Surfus; John R. Dixon; Holger N. Lode; Ralph A. Reisfeld; Stephen D. Gillies; Paul M. Sondel


Cancer Immunology, Immunotherapy | 2007

Flt3-L gene therapy enhances immunocytokine-mediated antitumor effects and induces long-term memory.

Zane C. Neal; Paul M. Sondel; Mary Kay Bates; Stephen D. Gillies; Hans Herweijer


Journal of Surgical Research | 2004

IL-12 cDNA Direct Injection: Antimetastatic Effect from a Single Injection in a Murine Hepatic Metastases Model

Sharon M. Weber; Chen Qi; Zane C. Neal; Paul M. Sondel; David M. Mahvi


Archive | 2005

Therapeutic fusion protein transgenes

Zane C. Neal; Hans Herweijer; Jon A. Wolff

Collaboration


Dive into the Zane C. Neal's collaboration.

Top Co-Authors

Avatar

Paul M. Sondel

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hans Herweijer

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jacquelyn A. Hank

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Imboden

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ralph A. Reisfeld

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Guofeng Zhang

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ilia N. Buhtoiarov

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge