Network


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

Hotspot


Dive into the research topics where Ryu Yoshida is active.

Publication


Featured researches published by Ryu Yoshida.


Antimicrobial Agents and Chemotherapy | 2011

Efficacy of Single Intravenous Injection of Peramivir against Influenza B Virus Infection in Ferrets and Cynomolgus Macaques

Mitsutaka Kitano; Yasushi Itoh; Makoto Kodama; Hirohito Ishigaki; Misako Nakayama; Hideaki Ishida; Kaoru Baba; Takahiro Noda; Kenji Sato; Yoichiro Nihashi; Takushi Kanazu; Ryu Yoshida; Ryuzo Torii; Akihiko Sato; Kazumasa Ogasawara

ABSTRACT We evaluated the efficacy of a single intravenous dose peramivir for treatment of influenza B virus infection in ferrets and cynomolgus macaques in the present study. A single dose of peramivir (60 mg/kg of body weight) given to ferrets on 1 day postinfection with influenza B virus significantly reduced median area under the curve (AUC) virus titers (peramivir, 8.3 log10 50% tissue culture infective doses [TCID50s]·day/ml; control, 10.7 log10 TCID50s·day/ml; P < 0.0001). Furthermore, nasal virus titers on day 2 postinfection in ferrets receiving a single injection of peramivir (30 mg/kg) and AUCs of the body temperature increase in ferrets receiving a single injection of peramivir (30 and 60 mg/kg) were lower than those in ferrets administered oral oseltamivir phosphate (30 and 60 mg/kg/day twice daily for 3 days). In macaques infected with influenza B virus, viral titers in the nasal swab fluid on days 2 and 3 postinfection and body temperature after a single injection of peramivir (30 mg/kg) were lower than those after oral administration of oseltamivir phosphate (30 mg/kg/day for 5 days). The two animal models used in the present study demonstrated that inhibition of viral replication at the early time point after infection was critical in reduction of AUCs of virus titers and interleukin-6 production, resulting in amelioration of symptoms. Our results shown in animal models suggest that the early treatment with a single intravenous injection of peramivir is clinically recommended to reduce symptoms effectively in influenza B virus infection.


Virology Journal | 2013

Antiviral activity of stachyflin on influenza A viruses of different hemagglutinin subtypes.

Yurie Motohashi; Manabu Igarashi; Masatoshi Okamatsu; Takeshi Noshi; Yoshihiro Sakoda; Naoki Yamamoto; Kimihito Ito; Ryu Yoshida; Hiroshi Kida

BackgroundThe hemagglutinin (HA) of influenza viruses is a possible target for antiviral drugs because of its key roles in the initiation of infection. Although it was found that a natural compound, Stachyflin, inhibited the growth of H1 and H2 but not H3 influenza viruses in MDCK cells, inhibitory activity of the compound has not been assessed against H4-H16 influenza viruses and the precise mechanism of inhibition has not been clarified.MethodsInhibitory activity of Stachyflin against H4-H16 influenza viruses, as well as H1-H3 viruses was examined in MDCK cells. To identify factors responsible for the susceptibility of the viruses to this compound, Stachyflin-resistant viruses were selected in MDCK cells and used for computer docking simulation.ResultsIt was found that in addition to antiviral activity of Stachyflin against influenza viruses of H1 and H2 subtypes, it inhibited replication of viruses of H5 and H6 subtypes, as well as A(H1N1)pdm09 virus in MDCK cells. Stachyflin also inhibited the virus growth in the lungs of mice infected with A/WSN/1933 (H1N1) and A/chicken/Ibaraki/1/2005 (H5N2). Substitution of amino acid residues was found on the HA2 subunit of Stachyflin-resistant viruses. Docking simulation indicated that D37, K51, T107, and K121 are responsible for construction of the cavity for the binding of the compound. In addition, 3-dimensional structure of the cavity of the HA of Stachyflin-susceptible virus strains was different from that of insusceptible virus strains.ConclusionAntiviral activity of Stachyflin was found against A(H1N1)pdm09, H5, and H6 viruses, and identified a potential binding pocket for Stachyflin on the HA. The present results should provide us with useful information for the development of HA inhibitors with more effective and broader spectrum.


Antimicrobial Agents and Chemotherapy | 2013

Efficacy of Repeated Intravenous Injection of Peramivir against Influenza A (H1N1) 2009 Virus Infection in Immunosuppressed Mice

Mitsutaka Kitano; Makoto Kodama; Yasushi Itoh; Takushi Kanazu; Masanori Kobayashi; Ryu Yoshida; Akihiko Sato

ABSTRACT The efficacy of intravenous peramivir against influenza A (H1N1) 2009 virus infection was evaluated in mice in which the immune system was suppressed by cyclophosphamide (CP) treatment. The mortality rate of the vehicle control group was 100%, and the mice lost 20% of their body weight on average by day 13 postinfection (p.i.). Repeated administration of peramivir (40 mg/kg of body weight once a day, given intravenously for 20 days), starting at 1 h p.i., significantly reduced mortality, body weight loss, viral titers, and cytokine production in infected mice compared with results for administration of vehicle (P < 0.01). In addition, repeated administration of peramivir, starting at 24 h, 48 h, or 72 h p.i., also resulted in increases in survival rates and reduction of viral titers in the lungs (P < 0.01). The mean days to death (MDD) of the vehicle group was 14.5 days, while in the groups treated with peramivir starting at 24 h, 48 h, and 72 h p.i., the MDDs were >23.0, 20.9, and 21.8 days, respectively. In comparison, repeated administration of oseltamivir phosphate (5 mg/kg twice a day, given orally for 20 days), starting at 24 h, 48 h, and 72 h p.i., also significantly prevented body weight loss, whereas no significant differences in mortality rates and viral titers in the lungs were observed compared with results for the vehicle group. These data indicated that repeated administration of peramivir was effective in promoting the survival and reducing virus replication in immunosuppressed mice infected with influenza A (H1N1) 2009 virus.


Virology | 2010

Establishment of a cynomolgus macaque model of influenza B virus infection

Mitsutaka Kitano; Yasushi Itoh; Makoto Kodama; Hirohito Ishigaki; Misako Nakayama; Tomoya Nagata; Hideaki Ishida; Hideaki Tsuchiya; Ryuzo Torii; Keiko Baba; Ryu Yoshida; Akihiko Sato; Kazumasa Ogasawara

Pathogenicity of influenza B virus was examined in cynomolgus macaques to establish a macaque model suitable for vaccine and antiviral drug development. We prepared influenza B viruses for inoculation with minimal passages after isolation from patients. Macaques inoculated with influenza B virus showed higher body temperature than that before infection for 6 to 12 days. Virus was detected in nasal, tracheal, and bronchial samples until 6 days after inoculation followed by an increase in neutralizing antibody. High levels of IL-6 and TNF-α in nasal swabs from the infected macaques were correlated with fever. Symptoms and duration of the viral replication would be sufficient to evaluate efficacy of vaccines and antiviral agents. In addition, measurement of immune responses including antibody and cytokine production would provide an immunological rationale in efficacy of vaccines and antiviral agents. The results suggest that cynomolgus macaques are appropriate model animals for research of influenza B virus.


The Journal of Infectious Diseases | 2011

Parenteral Peramivir Treatment for Oseltamivir-Resistant 2009 Pandemic Influenza A H1N1 Viruses

Yacine Abed; Guy Boivin; Ryu Yoshida; Makoto Kodama; Jaime E. Hernandez

To the Editor—In a recent issue of the Journal, Memoli and colleagues demonstrated , by using a relevant animal model (ie, ferrets), that viral fitness and transmissibility of 2009 pandemic influenza A H1N1 virus (pH1N1) isolates harboring the H275Y neuraminidase mutation were conserved [1]. Because the H275Y mutation was associated with a high level of resistance to oseltamivir and also conferred an intermediate level of resistance to peramivir [2], and given the fact that pH1N1 strains are naturally resistant to adamantanes, the search for antiviral alternatives against multidrug-resistant pH1N1 variants is highly relevant. In particular, these authors suggested that therapy with peramivir may not be an adequate treatment option in the case of infections with pH1N1 with the H275Y mutation. Although few currently circulating strains are H275Y mutants [3, 4], several lines of evidence suggest that the shift in susceptibility to peramivir in H275Y mutants may not be clinically significant: (1) The intrinsic potency of peramivir against pH1N1 strains is significantly higher [5]; (2) peramivir has a higher binding affinity and slower off rate from neuraminidase than those of oseltamivir carboxylate or zanamivir [6]; and (3) the peak levels achieved after administration of intravenous peramivir at the current dose (10 000 nmol/L) [7, 8] are much higher than the in vitro median inhibitory concentration for resistant isolates [9]. Mouse model data suggest that once daily dosing for 5–10 days may be superior to a single administration of peramivir for H275Y neuraminidase mutants [10]. The prophylactic activity of intramuscular peramivir was evaluated in mice infected with wild-type and H275Y mutant recombinant influenza A/WSN/33 (H1N1) viruses. Regimens used single (45 mg/kg or 90 mg/kg) or multiple (45 mg/kg daily for 5 days) intramuscular injections starting 1 hour before viral challenge. All peramivir regimens prevented mortality and weight loss while significantly reducing lung viral titers (LVTs) in mice infected with wild-type virus compared with untreated control mice. For the H275Y mutant, the multiple-dose regimen prevented mortality and weight loss and was associated with significant LVT reduction compared with untreated animals. Both single-dose regimens also reduced mortality and weight loss but not LVTs. In contrast, in a similar mouse model, prophylactic oral oseltamivir regimens did not prevent mortality or weight loss following infection with the same recombinant H275Y virus [10]. The activity of intravenous peramivir was also evaluated in a similar mouse model with H275Y mutant influenza A/WSN/33 (H1N1) viruses. Peramivir administered as a …


Antimicrobial Agents and Chemotherapy | 2014

Efficacy of Repeated Intravenous Administration of Peramivir against Highly Pathogenic Avian Influenza A (H5N1) Virus in Cynomolgus Macaques

Mitsutaka Kitano; Yasushi Itoh; Hirohito Ishigaki; Misako Nakayama; Hideaki Ishida; Van Loi Pham; Masahiko Arikata; Shintaro Shichinohe; Hideaki Tsuchiya; Naoko Kitagawa; Masanori Kobayashi; Ryu Yoshida; Akihiko Sato; Quynh Mai Le; Yoshihiro Kawaoka; Kazumasa Ogasawara

ABSTRACT Highly pathogenic avian influenza A (H5N1) viruses cause severe and often fatal disease in humans. We evaluated the efficacy of repeated intravenous dosing of the neuraminidase inhibitor peramivir against highly pathogenic avian influenza virus A/Vietnam/UT3040/2004 (H5N1) infection in cynomolgus macaques. Repeated dosing of peramivir (30 mg/kg/day once a day for 5 days) starting immediately after infection significantly reduced viral titers in the upper respiratory tract, body weight loss, and cytokine production and resulted in a significant body temperature reduction in infected macaques compared with that of macaques administered a vehicle (P < 0.05). Repeated administration of peramivir starting at 24 h after infection also resulted in a reduction in viral titers and a reduction in the period of virus detection in the upper respiratory tract, although the body temperature change was not statistically significant. The macaque model used in the present study demonstrated that inhibition of viral replication at an early time point after infection by repeated intravenous treatment with peramivir is critical for reduction of the production of cytokines, i.e., interleukin-6 (IL-6), tumor necrosis factor α, gamma interferon, monocyte chemotactic protein 1, and IL-12p40, resulting in amelioration of symptoms caused by highly pathogenic avian influenza virus infection.


Antiviral Research | 2014

The relationship between in vivo antiviral activity and pharmacokinetic parameters of peramivir in influenza virus infection model in mice

Makoto Kodama; Ryu Yoshida; Takahiro Hasegawa; Masaaki Izawa; Mitsutaka Kitano; Kaoru Baba; Takeshi Noshi; Takahiro Seki; Kenichi Okazaki; Masakatsu Tsuji; Takushi Kanazu; Hiroshi Kamimori; Tomoyuki Homma; Masanori Kobayashi; Yoshihiro Sakoda; Hiroshi Kida; Akihiko Sato; Yoshinori Yamano

The purpose of this study was to investigate the relationship between pharmacokinetic (PK) parameters of intravenous (IV) peramivir and in vivo antiviral activity pharmacodynamic (PD) outcomes in a mouse model of influenza virus infection. Peramivir was administrated to mice in three dosing schedules; once, twice and four times after infection of A/WS/33 (H1N1). The survival rate at day 14 after virus infection was employed as the antiviral activity outcome for analysis. The relationship between day 14 survival and PK parameters, including area under the concentration-time curve (AUC), maximum concentration (Cmax) and time that drug concentration exceeds IC95 (T(>IC95)), was estimated using a logistic regression model, and model fitness was evaluated by calculation of the Akaike information criterion (AIC) index. The AIC indices of AUC, Cmax and T(>IC95) were about 114, 151 and 124, respectively. The AIC of AUC and T(>IC95) were smaller than that of Cmax. Therefore, both AUC and T(>IC95) were the PK parameters that correlated best with the antiviral activity of peramivir IV against influenza virus infection in mice.


Antiviral Research | 2017

Therapeutic efficacy of peramivir against H5N1 highly pathogenic avian influenza viruses harboring the neuraminidase H275Y mutation

Masanori Kobayashi; Makoto Kodama; Takeshi Noshi; Ryu Yoshida; Takushi Kanazu; Naoki Nomura; Kosuke Soda; Norikazu Isoda; Masatoshi Okamatsu; Yoshihiro Sakoda; Yoshinori Yamano; Akihiko Sato; Hiroshi Kida

&NA; High morbidity and mortality associated with human cases of highly pathogenic avian influenza (HPAI) viruses, including H5N1 influenza virus, have been reported. The purpose of the present study was to evaluate the antiviral effects of peramivir against HPAI viruses. In neuraminidase (NA) inhibition and virus replication inhibition assays, peramivir showed strong inhibitory activity against H5N1, H7N1 and H7N7 HPAI viruses with sub‐nanomolar activity in enzyme assays. In H5N1 viruses containing the NA H275Y mutation, the antiviral activity of peramivir against the variant was lower than that against the wild‐type. Evaluation of the in vivo antiviral activity showed that a single intravenous treatment of peramivir (10 mg/kg) prevented lethality in mice infected with wild‐type H5N1 virus and also following infection with H5N1 virus with the H275Y mutation after a 5 day administration of peramivir (30 mg/kg). Furthermore, mice injected with peramivir showed low viral titers and low levels of proinflammatory cytokines in the lungs. These results suggest that peramivir has therapeutic activity against HPAI viruses even if the virus harbors the NA H275Y mutation. HighlightsPeramivir showed strong inhibitory activity against H5N1, H7N1 and H7N7 avian influenza viruses.Anti‐viral activity of peramivir was decreased in H5N1 virus containing the H275Y mutation.Peramivir prevented lethality in mice infected with H5N1 virus and also H5N1 virus harboring the H275Y mutation.Peramivir reduced viral titers and levels of proinflammatory cytokines in the lungs of mice infected with H5N1 viruses.


Open Forum Infectious Diseases | 2017

Delayed Dosing of S-033188, a Novel Inhibitor of Influenza Virus Cap-dependent Endonuclease, Exhibited Significant Reduction of Viral Titer and Mortality in Mice Infected with Influenza A Virus

Keita Fukao; Yoshinori Ando; Takeshi Noshi; Makoto Kawai; Ryu Yoshida; Takao Shishido; Akira Naito

Abstract Background Both epidemic and pandemic influenza are major public health concerns, but current standard treatment limits its usage by 48 hours from onsets. Furthermore, no antiviral drug has been shown to definitively reduce serious complications, hospitalization, or mortality in a randomized clinical trial. S-033188 is an orally available small molecule inhibitor of cap-dependent endonuclease that is essential for transcription and replication of influenza A and B virus. In this study, we evaluated the efficacy of delayed dosing of S-033188, either as single agent or in combination with oseltamivir, in mice infected with lethal doses of influenza A virus. Methods BALB/c mice were intranasally inoculated with A/PR/8/34 strain at 8.0 × 102 tissue culture infectious dose 50 (TCID50)/mouse. Mice were orally treated with S-033188 (0.5, 1.5, 15 or 50 mg/kg), oseltamivir phosphate (10 or 50 mg/kg), S-033188 (0.5 or 1.5 mg/kg) in combination with oseltamivir phosphate (10 or 50 mg/kg), or vehicle BID for 5 days, beginning at 96 hours after virus infection. Survival and body weight were then monitored through a 28-day period after infection. In addition, viral titer in the lung was determined during the treatment. Mice were euthanized and regarded as dead if their body weights were lower than 70% of the initial body weights according to humane endpoints. Results S-033188 monotherapy (15 or 50 mg/kg, BID for 5 days) completely eliminated mortality in mice, whereas oseltamivir monotherapy (10 or 50 mg/kg, BID for 5 days) exhibited only 10% or 40% survival (Figure1), respectively. S-033188 monotherapy also significantly reduced viral titer and prevented body weight loss, consistent with the prolonged survival. Furthermore, S-033188 (0.5 or 1.5 mg/kg) in combination with oseltamivir phosphate (10 or 50 mg/kg) exhibited significant improvement of mortality as compared with each oseltamivir phosphate monotherapy. Conclusion Delayed dosing of S-033188 exhibited significant efficacy in mice infected with lethal doses of influenza A virus compared with clinically equivalent or supratheraputic dosing of oseltamivir phosphate. Furthermore, delayed dosing of S-033188 in combination with oseltamivir phosphate exhibited significant improvement of mortality as compared with each oseltamivir phosphate monotherapy. Disclosures All authors: No reported disclosures.


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery of novel 5-hydroxy-4-pyridone-3-carboxy acids as potent inhibitors of influenza Cap-dependent endonuclease

Masayoshi Miyagawa; Toshiyuki Akiyama; Minako Mikamiyama-Iwata; Kazunari Hattori; Naoko Kurihara; Yoshiyuki Taoda; Chika Takahashi-Kageyama; Noriyuki Kurose; Hidenori Mikamiyama; Naoyuki Suzuki; Kenji Takaya; Kenji Tomita; Kenji Matsuo; Kenji Morimoto; Ryu Yoshida; Takao Shishido; Tomokazu Yoshinaga; Akihiko Sato; Makoto Kawai

We report the discovery of a novel series of influenza Cap-dependent EndoNuclease (CEN) inhibitors based on the 4-pyridone-carboxylic acid (PYXA) scaffold, which were found from our chelate library. Our SAR research revealed the lipophilic domain to be the key to CEN inhibition. In particular, the position between the chelate and the lipophilic domain in the derivatives was essential for enhancing the potency. Our study, based on virtual modeling, led to the identification of 2y as a potent CEN inhibitor with an IC50 of 5.12nM.

Collaboration


Dive into the Ryu Yoshida's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mitsutaka Kitano

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasushi Itoh

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Makoto Kawai

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hideaki Ishida

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Hirohito Ishigaki

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Kazumasa Ogasawara

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge