Network


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

Hotspot


Dive into the research topics where Walter Hans Meisen is active.

Publication


Featured researches published by Walter Hans Meisen.


Scientific Reports | 2015

CAR-Engineered NK Cells Targeting Wild-Type EGFR and EGFRvIII Enhance Killing of Glioblastoma and Patient-Derived Glioblastoma Stem Cells

Jianfeng Han; Jianhong Chu; Wing Keung Chan; Jianying Zhang; Youwei Wang; Justus B. Cohen; Aaron R. Victor; Walter Hans Meisen; Sung-Hak Kim; Paola Grandi; Qi-En Wang; Xiaoming He; Ichiro Nakano; E. Antonio Chiocca; Joseph C. Glorioso; Balveen Kaur; Michael A. Caligiuri; Jianhua Yu

Glioblastoma (GB) remains the most aggressive primary brain malignancy. Adoptive transfer of chimeric antigen receptor (CAR)-modified immune cells has emerged as a promising anti-cancer approach, yet the potential utility of CAR-engineered natural killer (NK) cells to treat GB has not been explored. Tumors from approximately 50% of GB patients express wild-type EGFR (wtEGFR) and in fewer cases express both wtEGFR and the mutant form EGFRvIII; however, previously reported CAR T cell studies only focus on targeting EGFRvIII. Here we explore whether both wtEGFR and EGFRvIII can be effectively targeted by CAR-redirected NK cells to treat GB. We transduced human NK cell lines NK-92 and NKL, and primary NK cells with a lentiviral construct harboring a second generation CAR targeting both wtEGFR and EGFRvIII and evaluated the anti-GB efficacy of EGFR-CAR-modified NK cells. EGFR-CAR-engineered NK cells displayed enhanced cytolytic capability and IFN-γ production when co-cultured with GB cells or patient-derived GB stem cells in an EGFR-dependent manner. In two orthotopic GB xenograft mouse models, intracranial administration of NK-92-EGFR-CAR cells resulted in efficient suppression of tumor growth and significantly prolonged the tumor-bearing mice survival. These findings support intracranial administration of NK-92-EGFR-CAR cells represents a promising clinical strategy to treat GB.


PLOS ONE | 2013

Thymoquinone Inhibits Autophagy and Induces Cathepsin-Mediated, Caspase-Independent Cell Death in Glioblastoma Cells

Ira O. Racoma; Walter Hans Meisen; Qi-En Wang; Balveen Kaur; Altaf A. Wani

Glioblastoma is the most aggressive and common type of malignant brain tumor in humans, with a median survival of 15 months. There is a great need for more therapies for the treatment of glioblastoma. Naturally occurring phytochemicals have received much scientific attention because many exhibit potent tumor killing action. Thymoquinone (TQ) is the bioactive compound of the Nigella sativa seed oil. TQ has anti-oxidant, anti-inflammatory and anti-neoplastic actions with selective cytotoxicity for human cancer cells compared to normal cells. Here, we show that TQ selectively inhibits the clonogenicity of glioblastoma cells as compared to normal human astrocytes. Also, glioblastoma cell proliferation could be impaired by chloroquine, an autophagy inhibitor, suggesting that glioblastoma cells may be dependent on the autophagic pathway for survival. Exposure to TQ caused an increase in the recruitment and accumulation of the microtubule-associated protein light chain 3-II (LC3-II). TQ also caused an accumulation of the LC3-associated protein p62, confirming the inhibition of autophagy. Furthermore, the levels of Beclin-1 protein expression were unchanged, indicating that TQ interferes with a later stage of autophagy. Finally, treatment with TQ induces lysosome membrane permeabilization, as determined by a specific loss of red acridine orange staining. Lysosome membrane permeabilization resulted in a leakage of cathepsin B into the cytosol, which mediates caspase-independent cell death that can be prevented by pre-treatment with a cathepsin B inhibitor. TQ induced apoptosis, as determined by an increase in PI and Annexin V positive cells. However, apoptosis appears to be caspase-independent due to failure of the caspase inhibitor z-VAD-FMK to prevent cell death and absence of the typical apoptosis related signature DNA fragmentation. Inhibition of autophagy is an exciting and emerging strategy in cancer therapy. In this vein, our results describe a novel mechanism of action for TQ as an autophagy inhibitor selectively targeting glioblastoma cells.


Molecular Therapy | 2013

Systemic Delivery of SapC-DOPS Has Antiangiogenic and Antitumor Effects Against Glioblastoma

Jeffrey Wojton; Zhengtao Chu; Haritha Mathsyaraja; Walter Hans Meisen; Nicholas Denton; Chang-Hyuk Kwon; Lionel M.L. Chow; Mary B. Palascak; Robert S. Franco; Tristan Bourdeau; Sherry Thornton; Michael C. Ostrowski; Balveen Kaur; Xiaoyang Qi

Saposin C-dioleoylphosphatidylserine (SapC-DOPS) nanovesicles are a nanotherapeutic which effectively target and destroy cancer cells. Here, we explore the systemic use of SapC-DOPS in several models of brain cancer, including glioblastoma multiforme (GBM), and the molecular mechanism behind its tumor-selective targeting specificity. Using two validated spontaneous brain tumor models, we demonstrate the ability of SapC-DOPS to selectively and effectively cross the blood-brain tumor barrier (BBTB) to target brain tumors in vivo and reveal the targeting to be contingent on the exposure of the anionic phospholipid phosphatidylserine (PtdSer). Increased cell surface expression of PtdSer levels was found to correlate with SapC-DOPS-induced killing efficacy, and tumor targeting in vivo was inhibited by blocking PtdSer exposed on cells. Apart from cancer cell killing, SapC-DOPS also exerted a strong antiangiogenic activity in vitro and in vivo. Interestingly, unlike traditional chemotherapy, hypoxic cells were sensitized to SapC-DOPS-mediated killing. This study emphasizes the importance of PtdSer exposure for SapC-DOPS targeting and supports the further development of SapC-DOPS as a novel antitumor and antiangiogenic agent for brain tumors.


Clinical Cancer Research | 2015

The Impact of Macrophage- and Microglia-Secreted TNFα on Oncolytic HSV-1 Therapy in the Glioblastoma Tumor Microenvironment

Walter Hans Meisen; Eric S. Wohleb; Alena Cristina Jaime-Ramirez; Chelsea Bolyard; Ji Young Yoo; Luke Russell; Jayson Hardcastle; Samuel Dubin; Kamaldeen Muili; Jianhua Yu; Michael A. Caligiuri; Jonathan P. Godbout; Balveen Kaur

Purpose: Oncolytic herpes simplex viruses (oHSV) represent a promising therapy for glioblastoma (GBM), but their clinical success has been limited. Early innate immune responses to viral infection reduce oHSV replication, tumor destruction, and efficacy. Here, we characterized the antiviral effects of macrophages and microglia on viral therapy for GBM. Experimental Design: Quantitative flow cytometry of mice with intracranial gliomas (±oHSV) was used to examine macrophage/microglia infiltration and activation. In vitro coculture assays of infected glioma cells with microglia/macrophages were used to test their impact on oHSV replication. Macrophages from TNFα-knockout mice and blocking antibodies were used to evaluate the biologic effects of TNFα on virus replication. TNFα blocking antibodies were used to evaluate the impact of TNFα on oHSV therapy in vivo. Results: Flow-cytometry analysis revealed a 7.9-fold increase in macrophage infiltration after virus treatment. Tumor-infiltrating macrophages/microglia were polarized toward a M1, proinflammatory phenotype, and they expressed high levels of CD86, MHCII, and Ly6C. Macrophages/microglia produced significant amounts of TNFα in response to infected glioma cells in vitro and in vivo. Using TNFα-blocking antibodies and macrophages derived from TNFα-knockout mice, we discovered TNFα-induced apoptosis in infected tumor cells and inhibited virus replication. Finally, we demonstrated the transient blockade of TNFα from the tumor microenvironment with TNFα-blocking antibodies significantly enhanced virus replication and survival in GBM intracranial tumors. Conclusions: The results of these studies suggest that FDA approved TNFα inhibitors may significantly improve the efficacy of oncolytic virus therapy. Clin Cancer Res; 21(14); 3274–85. ©2015 AACR.


Cancer Research | 2015

TGFβ Treatment Enhances Glioblastoma Virotherapy by Inhibiting the Innate Immune Response

Jianfeng Han; Xilin Chen; Jianhong Chu; Bo Xu; Walter Hans Meisen; Lichao Chen; Lingling Zhang; Jianying Zhang; Xiaoming He; Qi-En Wang; E. Antonio Chiocca; Balveen Kaur; Michael A. Caligiuri; Jianhua Yu

Oncolytic viruses, including oncolytic herpes simplex virus (oHSV), have produced provocative therapeutic responses in patients with glioblastoma, the most aggressive brain tumor. Paradoxically, innate immune responses mediated by natural killer (NK) cells and macrophages/microglia appear to limit oHSV efficacy. Therefore, we investigated whether pretreatment with an immunosuppressive cytokine, TGFβ, might reverse these effects and thereby potentiate oHSV efficacy. TGFβ treatment of NK cells rendered them less cytolytic against oHSV-infected glioblastoma cells and stem-like cells in vitro. Furthermore, TGFβ treatment of NK cells, macrophages, or microglia increased viral titers of oHSV in cocultures with glioblastoma cells. In a syngeneic mouse model of glioblastoma, administering TGFβ prior to oHSV injection inhibited intracranial infiltration and activation of NK cells and macrophages. Notably, a single administration of TGFβ prior to oHSV therapy was sufficient to phenocopy NK-cell depletion and suppress tumor growth and prolong survival in both xenograft and syngeneic models of glioblastoma. Collectively, our findings show how administering a single dose of TGFβ prior to oncolytic virus treatment of glioblastoma can transiently inhibit innate immune cells that limit efficacy, thereby improving therapeutic responses and survival outcomes.


Cancer Research | 2014

Aurora-A inhibition offers a novel therapy effective against intracranial glioblastoma

James R. Van Brocklyn; Jeffrey Wojton; Walter Hans Meisen; David A. Kellough; Jeffrey Ecsedy; Balveen Kaur; Norman L. Lehman

Glioblastoma remains a devastating disease for which novel therapies are urgently needed. Here, we report that the Aurora-A kinase inhibitor alisertib exhibits potent efficacy against glioblastoma neurosphere tumor stem-like cells in vitro and in vivo. Many glioblastoma neurosphere cells treated with alisertib for short periods undergo apoptosis, although some regain proliferative activity upon drug removal. Extended treatment, however, results in complete and irreversible loss of tumor cell proliferation. Moreover, alisertib caused glioblastoma neurosphere cells to partially differentiate and enter senescence. These effects were also observed in glioma cells treated with the Aurora-A inhibitor TC-A2317 or anti-Aurora-A siRNA. Furthermore, alisertib extended median survival of mice bearing intracranial human glioblastoma neurosphere tumor xenografts. Alisertib exerted similar effects on glioblastoma neurosphere cells in vivo and resulted in markedly reduced activated phosphoThr288Aurora-A and increased abnormal mitoses and cellular ploidy, consistent with on-target activity. Our results offer preclinical proof-of-concept for alisertib as a new therapeutic for glioma treatment.


Molecular Therapy | 2014

Role of Cysteine-rich 61 Protein (CCN1) in Macrophage-mediated Oncolytic Herpes Simplex Virus Clearance

Amy Haseley Thorne; Walter Hans Meisen; Luke Russell; Ji Young Yoo; Chelsea Bolyard; Justin D. Lathia; Jeremy N. Rich; Vinay K. Puduvalli; Hsiaoyin Mao; Jianhua Yu; Michael A. Caligiuri; Susheela Tridandapani; Balveen Kaur

Glioblastoma is a devastating disease, and there is an urgent need to develop novel therapies, such as oncolytic HSV1 (OV) to effectively target tumor cells. OV therapy depends on tumor-specific replication leading to destruction of neoplastic tissues. Host responses that curtail virus replication limit its efficacy in vivo. We have previously shown that cysteine-rich 61 protein (CCN1) activates a type 1 IFN antiviral defense response in glioblastoma cells. Incorporating TCGA data, we found CCN1 expression to be a negative prognostic factor for glioblastoma patients. Based on this, we used neutralizing antibodies against CCN1 to investigate its effect on OV therapy. Use of an anti-CCN1 antibody in mice bearing glioblastomas treated with OV led to enhanced virus expression along with reduced immune cell infiltration. OV-induced CCN1 increases macrophage migration toward infected glioblastoma cells by directly binding macrophages and also by enhancing the proinflammatory activation of macrophages inducing MCP-1 expression in glioblastoma cells. Activation of macrophages by CCN1 also increases viral clearance. Neutralization of integrin αMβ2 reversed CCN1-induced macrophage activation and migration, and reduced MCP-1 expression by glioblastoma cells. Our findings reveal that CCN1 plays a novel role in pathogen clearance; increasing macrophage infiltration and activation resulting in increased virus clearance in tumors.


Expert Review of Neurotherapeutics | 2013

How can we trick the immune system into overcoming the detrimental effects of oncolytic viral therapy to treat glioblastoma

Walter Hans Meisen; Balveen Kaur

Although China approved the use of the H101 adenovirus for the treatment of various solid cancers in 2005, similar viruses have not obtained clinical approval in the USA [1]. While there are several Phase III trials testing the therapeutic efficacy of oncolytic viruses (OVs) in patients diagnosed with melanoma, bladder and advanced head and neck cancers (ClinicalTrials.gov identifiers: NCT01438112, NCT00769704, NCT01166542 [101]), there are currently only Phase I/II OV trials for patients with glioblastoma (GB), which highlights the difficulty associated with treating this particular disease (ClinicalTrials.gov identif iers: NCT00157703, NCT01174537, NCT00528684, NCT01301430, NCT01582516 [101]). The uniqueness of the brain tumor microenvironment – the extracellular matrix, leaky blood vessels and immune responses – has collectively been shown to limit OV delivery, replication, spread and efficacy. Of these factors, the impact of the host immune response on OV efficacy is perhaps the most complicated to understand. In the context of OV therapy, the immune response is a double-edged sword. On one hand, innate immune responses result in rapid viral clearance and decreased OV efficacy; on the other hand, immune responses elicited after viral infection also have the potential to activate an adaptive antitumor immune response to promote tumor eradication. Here, we summarize some of the challenges and recent progress made by investigators in manipulating the immune response with respect to OV therapies for GB.


Molecular Cancer Therapeutics | 2015

Changes in BAI1 and Nestin Expression Are Prognostic Indicators for Survival and Metastases in Breast Cancer and Provide Opportunities for Dual Targeted Therapies

Walter Hans Meisen; Samuel Dubin; Steven T. Sizemore; Haritha Mathsyaraja; Katie Thies; Norman L. Lehman; Peter Boyer; Alena Cristina Jaime-Ramirez; J. Bradley Elder; Kimerly A. Powell; Arnab Chakravarti; Michael C. Ostrowski; Balveen Kaur

The 2-year survival rate of patients with breast cancer brain metastases is less than 2%. Treatment options for breast cancer brain metastases are limited, and there is an unmet need to identify novel therapies for this disease. Brain angiogenesis inhibitor 1 (BAI1) is a GPCR involved in tumor angiogenesis, invasion, phagocytosis, and synaptogenesis. For the first time, we identify that BAI1 expression is significantly reduced in breast cancer and higher expression is associated with better patient survival. Nestin is an intermediate filament whose expression is upregulated in several cancers. We found that higher Nestin expression significantly correlated with breast cancer lung and brain metastases, suggesting both BAI1 and Nestin can be therapeutic targets for this disease. Here, we demonstrate the ability of an oncolytic virus, 34.5ENVE, to target and kill high Nestin-expressing cells and deliver Vstat120 (extracellular fragment of BAI1). Finally, we created two orthotopic immune-competent murine models of breast cancer brain metastases and demonstrated 34.5ENVE extended the survival of immune-competent mice bearing intracranial breast cancer tumors. Mol Cancer Ther; 14(1); 307–14. ©2014 AACR.


Cancer Research | 2016

Abstract C30: The CSF1-PU.1 pathway in tumor associated macrophages promotes breast cancer growth and progression

Katie Thies; David A. Taffany; Haritha Mathsyaraja; Sudarshana M. Sharma; Walter Hans Meisen; Tom Liu; Cynthia Timmers; Jose Otero; Balveen Kaur; Michael C. Ostrowski

Purpose of Study: Research over the past decade has established a critical role for the tumor microenvironment in facilitating tumor growth and promoting invasiveness. Cells of the myeloid lineage, including macrophages, have been known to be key mediators of tumor progression and facilitate metastasis. Recent work from our lab highlights the Ets transcription factors, PU.1 and ETS2, as important players in tumor associated macrophages (TAMs) during breast cancer progression. Both transcription factors are downstream effectors of the Colony Stimulating Factor 1 (CSF1) signaling pathway, which is not only important for myeloid cell survival, but also has a well-established role within the tumor microenvironment. Here, we investigated the requirement of PU.1 in TAMs in a mouse mammary tumor model and have begun to address the role of PU.1 during metastasis. Research Method: We employed cre-loxP technology to conditionally delete PU.1 in the myeloid cell compartment. Syngeneic mice were orthotopically injected with mammary tumor cells directly into the fat pad. To model breast cancer brain metastasis in the mouse, we employed an intracranial injection model which recapitulates the biology of the human disease. We were able to isolate tumor associated macrophages from either site (mammary or brain tumor) for downstream applications. Novel Findings: Macrophage-specific deletion of PU.1 resulted in a significant reduction in mammary tumor growth as well as tumor angiogenesis. Our results suggest that ETS2 and PU.1, acting downstream of the CSF1 signaling pathway, cooperatively regulate the expression of pro-tumor genes along with ‘oncogenic’ microRNA. Conventional ChIP assays show that both transcription factors occupy enhancer regions adjacent to a set of selected genes and cooperatively regulate expression in tumor infiltrating macrophages. We focused on miR-21 and miR-29a for a more detailed investigation as both are expressed in CSF1R+ myeloid cells within the brain metastatic microenvironment. In our intracranial injection model, macrophages (F4/80 positive cells) are recruited to the growing tumor, and both miR-21 and miR-29a are upregulated in TAMs isolated by FACS from tumor-bearing mice as compared to controls. Conclusions and Implications: Given our present data, we believe that the Ets factors, PU.1 and Ets2, regulate a transcriptional program, including microRNAs, that is critical for CSF1 action in tumor infiltrating macrophages. Currently, clinical trials are underway using CSF1R inhibitors for several solid tumor types [clinicaltrials.gov]. The macrophage-specific microRNA, including miR-21 and miR-29a, may serve as indicators for to the efficacy of CSF1R inhibition. Citation Format: Katie Thies, David A. Taffany, Haritha Mathsyaraja, Sudarshana M. Sharma, Walter Hans Meisen, Tom Liu, Cynthia Timmers, Jose Otero, Balveen Kaur, Michael C. Ostrowski. The CSF1-PU.1 pathway in tumor associated macrophages promotes breast cancer growth and progression. [abstract]. In: Proceedings of the AACR Special Conference: Function of Tumor Microenvironment in Cancer Progression; 2016 Jan 7–10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2016;76(15 Suppl):Abstract nr C30.

Collaboration


Dive into the Walter Hans Meisen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

E. Antonio Chiocca

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge