Network


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

Hotspot


Dive into the research topics where Kosei Hasegawa is active.

Publication


Featured researches published by Kosei Hasegawa.


Gene Therapy | 2007

Evaluation of T cells as carriers for systemic measles virotherapy in the presence of antiviral antibodies.

Hooi-Tin Ong; Kosei Hasegawa; Allan B. Dietz; Stephen J. Russell; Kah-Whye Peng

Neutralizing antiviral antibodies (Abs) can hinder systemic virotherapy. Here, we used activated T cells as carriers to deliver oncolytic measles viruses (MV) to multiple myeloma xenografts in the presence of anti-MV antibodies (Abs). Virus-infected T cells expressing measles H/F fusogenic envelope glycoproteins could efficiently transfer MV infection by heterofusion, even after exposure to virus-inactivating anti-MV antisera. Severe-combined immunodeficiency (SCID) mice bearing subcutaneous or disseminated human myeloma xenografts were given MV-luciferase (MV-Luc) or MV-Luc-infected T cells intravenously. Indium111 labeling indicated that 1–2% of the virus-infected T cells trafficked to tumors. Preinfected T cells fused with tumor cells in vivo and transferred MV-Luc to tumor xenografts where intratumoral viral spread was monitored non-invasively using bioluminescent imaging. In mice passively immunized with high titers of measles immune serum, intravenous virus and cell delivery were both inhibited. Decreasing the amount of measles immune serum given to mice permitted tumor infection by virus-infected T cells and cell-free virus. In conclusion, virus-loaded T cells may facilitate systemic measles virotherapy in the presence of antiviral Abs and they warrant further investigation as potential MV cell carriers.


Clinical Cancer Research | 2006

Dual Therapy of Ovarian Cancer Using Measles Viruses Expressing Carcinoembryonic Antigen and Sodium Iodide Symporter

Kosei Hasegawa; Linh Pham; Michael K. O'Connor; Mark J. Federspiel; Stephen J. Russell; Kah Whye Peng

Purpose: MV-CEA is an oncolytic measles virus currently being tested in patients with ovarian cancer and whose propagation can be monitored by measuring blood carcinoembryonic antigen (CEA) levels. MV-NIS is an oncolytic measles virus coding for the thyroidal sodium iodide symporter (NIS) whose propagation can be mapped by serial radioiodine imaging. Expression of both CEA and NIS genes from a single virus would combine sensitive, quantitative expression monitoring (CEA) with radioisotopic expression mapping (NIS). Because of the unfavorable replication kinetics of measles viruses expressing both CEA and NIS, we explored the feasibility of combining MV-CEA with MV-NIS for comprehensive virotherapy monitoring in ovarian cancer. Experimental Design and Results: Mice implanted with i.p. SKOV3ip.1 ovarian cancer xenografts received MV-CEA alone, MV-NIS alone, or a combination of MV-CEA plus MV-NIS. Viral gene expression was monitored by measuring blood CEA levels, and the location of virus-infected cells was monitored by gamma camera imaging. Surprisingly, mice receiving the combination of MV-CEA plus MV-NIS showed greatly superior responses to therapy, but this was associated with 10-fold lower plasma levels of CEA compared with mice treated with MV-CEA alone. In vitro studies showed superior replication kinetics of MV-NIS relative to MV-CEA. The gamma camera scans were considerably less sensitive than the plasma CEA marker for monitoring virus infection. Conclusions: Dual therapy with MV-CEA and MV-NIS is superior to treatment with either virus alone, and it allows noninvasive monitoring of virotherapy via soluble marker peptide and gamma camera imaging. This has important implications for the clinical development of oncolytic measles viruses.


Clinical Cancer Research | 2006

The use of a tropism-modified measles virus in folate receptor-targeted virotherapy of ovarian cancer.

Kosei Hasegawa; Takafumi Nakamura; Mary Harvey; Yasuhiro Ikeda; Ann L. Oberg; Mariangela Figini; Silvana Canevari; Lynn C. Hartmann; Kah Whye Peng

Purpose: Attenuated measles viruses are promising experimental anticancer agents currently being evaluated in a phase I dose escalation trial for ovarian cancer patients. Virus attachment, entry, and subsequent intercellular fusion between infected and uninfected neighboring cells are mediated via the two measles receptors (CD46 and SLAM). To minimize potential toxicity due to measles virus–associated immunosuppression and infection of nontarget tissues, we sought to develop an ovarian cancer exclusive fully retargeted measles virus. Experimental Design and Results: Interactions of measles virus with its natural receptors were ablated, and a single-chain antibody (scFv) specific for α-folate receptor (FRα), a target overexpressed on 90% of nonmucinous ovarian cancer, was genetically engineered on the viral attachment protein (MV-αFR). Specificity of virus tropism was tested on tumor and normal cells. Biodistribution of measles virus infection was evaluated in measles-susceptible CD46 transgenic mice, whereas antitumor activity was monitored noninvasively by bioluminescence imaging in xenograft models. Tropism and fusogenic activity of MV-αFR was redirected exclusively to FRα without compromise to virus infectivity. In contrast to the parental virus, MV-αFR has no background infectivity on normal human cells. The antitumor activity of MV-αFR, as assessed by tumor volume reduction and overall survival increase, was equal to the parental virus in two models of human ovarian cancer (s.c. and i.p.). Conclusions: A FR-exclusive ovarian cancer targeted oncolytic virus was generated and shown to be therapeutically effective, thus introducing a new modality for FR targeting and a candidate measles virus for clinical testing.


Journal of Virology | 2007

Affinity Thresholds for Membrane Fusion Triggering by Viral Glycoproteins

Kosei Hasegawa; Chunling Hu; Takafumi Nakamura; James D. Marks; Stephen J. Russell; Kah Whye Peng

ABSTRACT Enveloped viruses trigger membrane fusion to gain entry into cells. The receptor affinities of their attachment proteins vary greatly, from 10−4 M to 10−9 M, but the significance of this is unknown. Using six retargeted measles viruses that bind to Her-2/neu with a 5-log range in affinity, we show that receptor affinity has little impact on viral attachment but is nevertheless a key determinant of infectivity and intercellular fusion. For a given cell surface receptor density, there is an affinity threshold above which cell-cell fusion proceeds efficiently. Suprathreshold affinities do not further enhance the efficiency of membrane fusion.


The Prostate | 2009

Prostate-specific membrane antigen retargeted measles virotherapy for the treatment of prostate cancer.

Chunsheng Liu; Kosei Hasegawa; Stephen J. Russell; Michel Sadelain; Kah Whye Peng

Live attenuated vaccine strain of measles virus (MV) has promising antitumor activity and is undergoing clinical testing in three different phase I cancer trials. The virus uses one of two receptors, CD46 which is ubiquitously expressed on all nucleated cells or CD150 which is expressed on immune cells, to infect cells. To minimize potential toxicity due to indiscriminate infection of normal cells, we have generated a fully retargeted MV that infects cells exclusively through the prostate‐specific membrane antigen (PSMA) receptor, which is overexpressed on prostate cancer cells and tumor neovasculature.


Molecular Therapy | 2007

Engineering oncolytic measles virus to circumvent the intracellular innate immune response.

Iana H. Haralambieva; Ianko Iankov; Kosei Hasegawa; Mary Harvey; Stephen J. Russell; Kah Whye Peng


Molecular Therapy | 2006

957. Effect of Ligand Binding Affinity and Receptor Density in Relation to Biology of Retargeted Viruses

Kosei Hasegawa; Takafumi Nakamura; James D. Marks; Stephen J. Russell; Kah-Whye Peng


Molecular Therapy | 2006

482. T Cells as Carriers for Systemic Measles Virotherapy of Multiple Myeloma

Hooi Tin Ong; Kosei Hasegawa; Mary Harvey; Allan B. Dietz; Stephen J. Russell; Kah-Whye Peng


Molecular Therapy | 2006

775. Folate Receptor Targeted Virotherapy of Ovarian Cancer Using a Tropism Modified Measles Virus

Kosei Hasegawa; Takafumi Nakamura; Mary Harvey; Yashiro Ikeda; Ann L. Oberg; Mariangela Figini; Silvana Canevari; Lynn C. Hartmann; Kah-Whye Peng


Cancer Research | 2006

Oncolytic virotherapy for targeting ovarian cancer using fully folate receptor targeted recombinant measles virus

Kosei Hasegawa; Takafumi Nakamura; Mary Harvey; Atsushi Hongo; Mariangela Figini; Silvana Canervari; Kah-Whye Peng

Collaboration


Dive into the Kosei Hasegawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James D. Marks

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge