Network


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

Hotspot


Dive into the research topics where Hideki Garren is active.

Publication


Featured researches published by Hideki Garren.


The New England Journal of Medicine | 2017

Ocrelizumab versus Placebo in Primary Progressive Multiple Sclerosis

Xavier Montalban; Stephen L. Hauser; Ludwig Kappos; Douglas L. Arnold; Amit Bar-Or; Giancarlo Comi; Jérôme De Seze; Gavin Giovannoni; Hans-Peter Hartung; Bernhard Hemmer; Fred D. Lublin; K Rammohan; Krzysztof Selmaj; Anthony Traboulsee; Annette Sauter; Donna Masterman; Paulo Fontoura; Shibeshih Belachew; Hideki Garren; Nicole Mairon; Peter Chin; Jerry S. Wolinsky

BACKGROUND An evolving understanding of the immunopathogenesis of multiple sclerosis suggests that depleting B cells could be useful for treatment. We studied ocrelizumab, a humanized monoclonal antibody that selectively depletes CD20‐expressing B cells, in the primary progressive form of the disease. METHODS In this phase 3 trial, we randomly assigned 732 patients with primary progressive multiple sclerosis in a 2:1 ratio to receive intravenous ocrelizumab (600 mg) or placebo every 24 weeks for at least 120 weeks and until a prespecified number of confirmed disability progression events had occurred. The primary end point was the percentage of patients with disability progression confirmed at 12 weeks in a time‐to‐event analysis. RESULTS The percentage of patients with 12‐week confirmed disability progression was 32.9% with ocrelizumab versus 39.3% with placebo (hazard ratio, 0.76; 95% confidence interval [CI], 0.59 to 0.98; P=0.03). The percentage of patients with 24‐week confirmed disability progression was 29.6% with ocrelizumab versus 35.7% with placebo (hazard ratio, 0.75; 95% CI, 0.58 to 0.98; P=0.04). By week 120, performance on the timed 25‐foot walk worsened by 38.9% with ocrelizumab versus 55.1% with placebo (P=0.04); the total volume of brain lesions on T2‐weighted magnetic resonance imaging (MRI) decreased by 3.4% with ocrelizumab and increased by 7.4% with placebo (P<0.001); and the percentage of brain‐volume loss was 0.90% with ocrelizumab versus 1.09% with placebo (P=0.02). There was no significant difference in the change in the Physical Component Summary score of the 36‐Item Short‐Form Health Survey. Infusion‐related reactions, upper respiratory tract infections, and oral herpes infections were more frequent with ocrelizumab than with placebo. Neoplasms occurred in 2.3% of patients who received ocrelizumab and in 0.8% of patients who received placebo; there was no clinically significant difference between groups in the rates of serious adverse events and serious infections. CONCLUSIONS Among patients with primary progressive multiple sclerosis, ocrelizumab was associated with lower rates of clinical and MRI progression than placebo. Extended observation is required to determine the long‐term safety and efficacy of ocrelizumab. (Funded by F. Hoffmann–La Roche; ORATORIO ClinicalTrials.gov number, NCT01194570.)


The New England Journal of Medicine | 2017

Ocrelizumab versus Interferon Beta-1a in Relapsing Multiple Sclerosis

Stephen L. Hauser; Amit Bar-Or; Giancarlo Comi; Gavin Giovannoni; Hans-Peter Hartung; Bernhard Hemmer; Fred D. Lublin; Xavier Montalban; K Rammohan; Krzysztof Selmaj; Anthony Traboulsee; Jerry S. Wolinsky; Douglas L. Arnold; G. Klingelschmitt; Donna Masterman; Paulo Fontoura; Shibeshih Belachew; Peter Chin; Nicole Mairon; Hideki Garren; Ludwig Kappos; Opera I; Opera Ii Clinical Investigators

BACKGROUND B cells influence the pathogenesis of multiple sclerosis. Ocrelizumab is a humanized monoclonal antibody that selectively depletes CD20+ B cells. METHODS In two identical phase 3 trials, we randomly assigned 821 and 835 patients with relapsing multiple sclerosis to receive intravenous ocrelizumab at a dose of 600 mg every 24 weeks or subcutaneous interferon beta‐1a at a dose of 44 μg three times weekly for 96 weeks. The primary end point was the annualized relapse rate. RESULTS The annualized relapse rate was lower with ocrelizumab than with interferon beta‐1a in trial 1 (0.16 vs. 0.29; 46% lower rate with ocrelizumab; P<0.001) and in trial 2 (0.16 vs. 0.29; 47% lower rate; P<0.001). In prespecified pooled analyses, the percentage of patients with disability progression confirmed at 12 weeks was significantly lower with ocrelizumab than with interferon beta‐1a (9.1% vs. 13.6%; hazard ratio, 0.60; 95% confidence interval [CI], 0.45 to 0.81; P<0.001), as was the percentage of patients with disability progression confirmed at 24 weeks (6.9% vs. 10.5%; hazard ratio, 0.60; 95% CI, 0.43 to 0.84; P=0.003). The mean number of gadolinium‐enhancing lesions per T1‐weighted magnetic resonance scan was 0.02 with ocrelizumab versus 0.29 with interferon beta‐1a in trial 1 (94% lower number of lesions with ocrelizumab, P<0.001) and 0.02 versus 0.42 in trial 2 (95% lower number of lesions, P<0.001). The change in the Multiple Sclerosis Functional Composite score (a composite measure of walking speed, upper‐limb movements, and cognition; for this z score, negative values indicate worsening and positive values indicate improvement) significantly favored ocrelizumab over interferon beta‐1a in trial 2 (0.28 vs. 0.17, P=0.004) but not in trial 1 (0.21 vs. 0.17, P=0.33). Infusion‐related reactions occurred in 34.3% of the patients treated with ocrelizumab. Serious infection occurred in 1.3% of the patients treated with ocrelizumab and in 2.9% of those treated with interferon beta‐1a. Neoplasms occurred in 0.5% of the patients treated with ocrelizumab and in 0.2% of those treated with interferon beta‐1a. CONCLUSIONS Among patients with relapsing multiple sclerosis, ocrelizumab was associated with lower rates of disease activity and progression than interferon beta‐1a over a period of 96 weeks. Larger and longer studies of the safety of ocrelizumab are required. (Funded by F. Hoffmann–La Roche; OPERA I and II ClinicalTrials.gov numbers, NCT01247324 and NCT01412333, respectively.)


Immunity | 2001

Combination of gene delivery and DNA vaccination to protect from and reverse Th1 autoimmune disease via deviation to the Th2 pathway

Hideki Garren; Pedro J. Ruiz; Trent A. Watkins; Paulo Fontoura; Louis-Vu T. Nguyen; Einat R Estline; David L. Hirschberg; Lawrence Steinman

Using a combination of local gene delivery and tolerizing DNA vaccination, we demonstrate that codelivery of the interleukin-4 (IL-4) gene and a DNA vaccine encoding the self-peptide proteolipid protein 139-151 (PLP139-151) provides protective immunity against experimental autoimmune encephalomyelitis (EAE). We provide evidence for a mechanism whereby IL-4 expressed from the naked DNA is secreted and acts locally on autoreactive T cells via activation of STAT6 to shift their cytokine profile to T helper 2. We also show that DNA vaccines can be used to reverse established EAE by covaccination with the genes for myelin oligodendrocyte glycoprotein and IL-4. This treatment strategy combines the antigen-specific effects of DNA vaccination and the beneficial effects of local gene delivery.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Multiple elements of the allergic arm of the immune response modulate autoimmune demyelination

Rosetta Pedotti; Jason DeVoss; Sawsan Youssef; Dennis J. Mitchell; Jochen Wedemeyer; Rami Madanat; Hideki Garren; Paulo Fontoura; Mindy Tsai; Stephen J. Galli; Raymond A. Sobel; Lawrence Steinman

Analysis of mRNA from multiple sclerosis lesions revealed increased amounts of transcripts for several genes encoding molecules traditionally associated with allergic responses, including prostaglandin D synthase, histamine receptor type 1 (H1R), platelet activating factor receptor, Ig Fc ɛ receptor 1 (FcɛRI), and tryptase. We now demonstrate that, in the animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), mediated by T helper 1 (Th1) T cells, histamine receptor 1 and 2 (H1R and H2R) are present on inflammatory cells in brain lesions. Th1 cells reactive to myelin proteolipid protein expressed more H1R and less H2R than Th2 cells. Pyrilamine, an H1R antagonist, blocked EAE, and the platelet activating factor receptor antagonist CV6209 reduced the severity of EAE. EAE severity was also decreased in mice with disruption of the genes encoding Ig FcγRIII or both FcγRIII and FcɛRI. Prostaglandin D synthase and tryptase transcripts were elevated in EAE brain. Taken together, these data reveal extensive involvement of elements of the immune response associated with allergy in autoimmune demyelination. The pathogenesis of demyelination must now be viewed as encompassing elements of both Th1 responses and “allergic” responses.


Annals of Neurology | 2008

Phase 2 trial of a DNA vaccine encoding myelin basic protein for multiple sclerosis

Hideki Garren; William H. Robinson; Eva Krasulova; Eva Havrdova; Congor Nadj; Krzysztof Selmaj; Jacek Losy; Ilinka Nadj; Ernst-Wilhelm Radue; Brian A. Kidd; Jill Gianettoni; Karen Tersini; Paul J. Utz; Frank Valone; Lawrence Steinman

To evaluate the efficacy and safety of BHT‐3009 in relapsing‐remitting multiple sclerosis (MS) and to confirm that BHT‐3009 causes immune tolerance.


Journal of Immunology | 2003

An Immunomodulatory GpG Oligonucleotide for the Treatment of Autoimmunity via the Innate and Adaptive Immune Systems

Peggy P. Ho; Paulo Fontoura; Pedro J. Ruiz; Lawrence Steinman; Hideki Garren

Bacterial DNA and immunostimulatory CpG oligodeoxynucleotides (ODNs) activate the innate immune system to produce proinflammatory cytokines. Shown to be potent Th1-like adjuvants, stimulatory CpG motifs are currently used as effective therapeutic vaccines for various animal models of infectious diseases, tumors, allergies, and autoimmune diseases. In this study, we show that the application of an immunomodulatory GpG ODN, with a single base switch from CpG to GpG, can effectively inhibit the activation of Th1 T cells associated with autoimmune disease. Moreover, this immunomodulatory GpG ODN suppresses the severity of experimental autoimmune encephalomyelitis in mice, a prototypic Th1-mediated animal disease model for multiple sclerosis.


Science Translational Medicine | 2013

Plasmid-encoded proinsulin preserves C-peptide while specifically reducing proinsulin-specific CD8 + T cells in type 1 diabetes

Bart O. Roep; Nanette Solvason; Peter A. Gottlieb; Joana R. F. Abreu; Leonard C. Harrison; George S. Eisenbarth; Liping Yu; Michael Leviten; William Hagopian; John B. Buse; Matthias von Herrath; Joanne Quan; Robert S. King; William H. Robinson; Paul J. Utz; Hideki Garren; Lawrence Steinman

A plasmid encoding proinsulin reduces the frequency of CD8+ T cells reactive to proinsulin while preserving C-peptide. Diabetes Trial Hits Its Mark Type 1 diabetes is an autoimmune disease where an inflammatory response destroys the insulin-producing cells of the pancreas. One way to block this response is through immunosuppression; however, it has proven difficult to target the specific autoreactive cells without suppressing the rest of the immune response. Now, Roep et al. demonstrate that an engineered plasmid that expresses proinsulin can preserve β cell function in type 1 diabetes patients. The authors randomized patients recently diagnosed with type 1 diabetes to receive various doses of either a proinsulin-expressing engineered plasmid or PBS vehicle. They observed no serious adverse events. The subjects in the experimental group had improved C-peptide levels—a readout of β cell function. The authors then examined the immune responses in these patients and found that there was a decrease in proinsulin-specific CD8+ T cells, but not unrelated CD8+ T cells. No difference was observed in cytokine production by CD4+ T cells. If these data hold true in larger studies, a plasmid encoding proinsulin could serve as a targeted means of immunosuppression for type 1 diabetes. In type 1 diabetes (T1D), there is an intense inflammatory response that destroys the β cells in the pancreatic islets of Langerhans, the site where insulin is produced and released. A therapy for T1D that targets the specific autoimmune response in this disease while leaving the remainder of the immune system intact, has long been sought. Proinsulin is a major target of the adaptive immune response in T1D. We hypothesized that an engineered DNA plasmid encoding proinsulin (BHT-3021) would preserve β cell function in T1D patients through reduction of insulin-specific CD8+ T cells. We studied 80 subjects over 18 years of age who were diagnosed with T1D within the past 5 years. Subjects were randomized 2:1 to receive intramuscular injections of BHT-3021 or BHT-placebo, weekly for 12 weeks, and then monitored for safety and immune responses in a blinded fashion. Four dose levels of BHT-3021 were evaluated: 0.3, 1.0, 3.0, and 6.0 mg. C-peptide was used both as an exploratory efficacy measure and as a safety measure. Islet-specific CD8+ T cell frequencies were assessed with multimers of monomeric human leukocyte antigen class I molecules loaded with peptides from pancreatic and unrelated antigens. No serious adverse events related to BHT-3021 were observed. C-peptide levels improved relative to placebo at all doses, at 1 mg at the 15-week time point (+19.5% BHT-3021 versus −8.8% BHT-placebo, P < 0.026). Proinsulin-reactive CD8+ T cells, but not T cells against unrelated islet or foreign molecules, declined in the BHT-3021 arm (P < 0.006). No significant changes were noted in interferon-γ, interleukin-4 (IL-4), or IL-10 production in CD4 T cells. Thus, we demonstrate that a plasmid encoding proinsulin reduces the frequency of CD8+ T cells reactive to proinsulin while preserving C-peptide over the course of dosing.


Journal of Immunology | 2002

Late Pregnancy Suppresses Relapses in Experimental Autoimmune Encephalomyelitis: Evidence for a Suppressive Pregancy-Related Serum Factor

Annette Langer-Gould; Hideki Garren; Amy Slansky; Pedro J. Ruiz; Lawrence Steinman

Women with multiple sclerosis have significantly diminished disease activity during pregnancy. The purpose of our study was to identify the underlying mechanism for the diminished disease activity. We found that during the period of late pregnancy there is protection against paralysis, during both the induction and effector phases of relapsing experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. We did not find any changes in the cytokine secretion profiles or the proliferative activity of autoreactive T cells from mice induced during late pregnancy compared with virgin controls. In mice mated after disease onset, the inflammatory histologic lesions did not clear, despite marked clinical improvement during pregnancy. We found evidence for a serum factor present in late pregnancy that suppresses T cell activation. In the presence of sera taken from mice late in pregnancy, the proliferative response and IL-2 production of proteolipid protein p139–151-specific T cells were significantly diminished as compared with stimulation in the presence of normal mouse sera. In conclusion, serum from late pregnancy has the capacity to down-regulate T cell responses and might be associated with the amelioration of disease activity in experimental autoimmune encephalomyelitis.


Journal of Immunology | 2004

Immunity to the Extracellular Domain of Nogo-A Modulates Experimental Autoimmune Encephalomyelitis

Paulo Fontoura; Peggy P. Ho; Jason DeVoss; Binhai Zheng; Byung J. Lee; Brian A. Kidd; Hideki Garren; Raymond A. Sobel; William H. Robinson; Marc Tessier-Lavigne; Lawrence Steinman

Nogo-66, the extracellular 66 aa loop of the Nogo-A protein found in CNS myelin, interacts with the Nogo receptor and has been proposed to mediate inhibition of axonal regrowth. It has been shown that immunization with Nogo-A promotes recovery in animal models of spinal cord injury through induction of Ab production. In this report, studies were performed to characterize the immune response to Nogo-66 and to determine the role of Nogo in experimental autoimmune encephalomyelitis (EAE). Immunization of EAE-susceptible mouse strains with peptides derived from Nogo-66 induced a CNS immune response with clinical and pathological similarities to EAE. The Nogo-66 peptides elicited strong T cell responses that were not cross-reactive to other encephalitogenic myelin Ags. Using a large scale spotted microarray containing proteins and peptides derived from a wide spectrum of myelin components, we demonstrated that Nogo-66 peptides also generated a specific Ab response that spreads to several other encephalitogenic myelin Ags following immunization. Nogo-66-specific T cell lines ameliorated established EAE, via Nogo-66-specific Th2 cells that entered the CNS. These results indicate that some T cell and B cell immune responses to Nogo-66 are associated with suppression of ongoing EAE, whereas other Nogo-66 epitopes can be encephalitogenic.


Journal of Immunology | 2008

Improved Efficacy of a Tolerizing DNA Vaccine for Reversal of Hyperglycemia through Enhancement of Gene Expression and Localization to Intracellular Sites

Nanette Solvason; Ya-Ping Lou; Wendy Peters; Erica Evans; Josefina Martinez; Uriel Ramirez; Andrea Ocampo; Rui Yun; Saira Ahmad; Edwin Liu; Liping Yu; George S. Eisenbarth; Michael Leviten; Lawrence Steinman; Hideki Garren

Insulin is a major target for the autoimmune-mediated destruction of pancreatic β cells during the pathogenesis of type I diabetes. A plasmid DNA vaccine encoding mouse proinsulin II reduced the incidence of diabetes in a mouse model of type I diabetes when administered to hyperglycemic (therapeutic mode) or normoglycemic (prophylactic mode) NOD mice. Therapeutic administration of proinsulin DNA was accompanied by a rapid decrease in the number of insulin-specific IFN-γ-producing T cells, whereas prophylactic treatment was accompanied by enhanced IFN-γ-secreting cells and a decrease in insulin autoantibodies. Adoptive transfer experiments demonstrated that the protection was not mediated by induction of CD25+/CD4+ T regulatory cells. The efficacy of the DNA vaccine was enhanced by increasing the level of expression of the encoded Ag, more frequent dosing, increasing dose level, and localization of the protein product to the intracellular compartment. The efficacy data presented in this study demonstrate that Ag-specific plasmid DNA therapy is a viable strategy for preventing progression of type I diabetes and defines critical parameters of the dosing regime that influences tolerance induction.

Collaboration


Dive into the Hideki Garren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas L. Arnold

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William H. Robinson

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar

Jerry S. Wolinsky

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge