Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Thromb Haemost ; 122(3): 320-328, 2022 03.
Article in English | MEDLINE | ID: mdl-33930902

ABSTRACT

Coagulation factor X (F10) amplifies the clotting reaction in the middle of the coagulation cascade, and thus F10 deficiency leads to a bleeding tendency. Isolated acquired F10 deficiency is widely recognized in patients with immunoglobulin light-chain amyloidosis or plasma cell dyscrasias. However, its occurrence as an autoimmune disorder is extremely rare. The Japanese Collaborative Research Group has been conducting a nationwide survey on autoimmune coagulation factor deficiencies (AiCFDs) starting in the last decade; we recently identified three patients with autoimmune F10 deficiency (AiF10D). Furthermore, an extensive literature search was performed, confirming 26 AiF10D and 28 possible cases. Our study revealed that AiF10D patients were younger than patients with other AiCFDs; AiF10D patients included children and were predominantly male. AiF10D was confirmed as a severe type of bleeding diathesis, although its mortality rate was not high. As AiF10D patients showed only low F10 inhibitor titers, they were considered to have nonneutralizing anti-F10 autoantibodies rather than their neutralizing counterparts. Accordingly, immunological anti-F10 antibody detection is highly recommended. Hemostatic and immunosuppressive therapies may help arrest bleeding and eliminate anti-F10 antibodies, leading to a high recovery rate. However, further investigation is necessary to understand the basic characteristics and proper management of AiF10D owing to the limited number of patients.


Subject(s)
Autoimmune Diseases , Factor X Deficiency , Factor X/immunology , Hemorrhagic Disorders , Autoimmune Diseases/blood , Autoimmune Diseases/complications , Disease Management , Factor X Deficiency/complications , Factor X Deficiency/immunology , Hemorrhagic Disorders/etiology , Hemorrhagic Disorders/therapy , Humans
2.
Int J Hematol ; 112(5): 621-630, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32748217

ABSTRACT

Emicizumab shortens activated partial thromboplastin time (aPTT) greater than Factor (F)VIII. Clot waveform analysis triggered by ellagic acid and tissue factor trigger (Elg/TF) provided a useful means of assessing emicizumab activity. Thrombin generation assays (TGA) using this trigger reagent might also overcome the difficulties associated with aPTT by emicizumab. To compare TGA triggered by Elg/TF and other reagents (FXIa, TF) for evaluating emicizumab activity. Emicizumab, FVIII, or FVIII-bypassing agents (BPAs) were incubated with FVIII-deficient plasmas prior to TGA initiated by Elg/TF (0.2 µM/0.5 pM), FXIa (5.21 pM), or TF (PPP-Reagent LOW®). Emicizumab, FVIII, or BPAs increased peak thrombin generation (peak-Th) dose-dependently using Elg/TF-trigger and the other triggers. Low responses were evident with FXIa-trigger and the enhanced effects remained below normal levels with Elg/TF-trigger. Experiments using FVIII with emicizumab demonstrated an additive effect on peak-Th using Elg/TF-trigger, and this effect appeared to be less at FVIII ≥ 40 IU/dl. BPAs with emicizumab appeared to mediate additive effects, although its effects were variable. Parameters of thrombin generation from BPAs and emicizumab with Elg/TF-trigger were improved to normal level compared to low TF-trigger. Elg/TF-TGA could evaluate global coagulation potential during emicizumab prophylaxis including concomitant therapy with FVIII or BPAs.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Blood Coagulation Tests/methods , Factor VIII/therapeutic use , Factor X/immunology , Hemophilia A/drug therapy , Hemorrhage/prevention & control , Partial Thromboplastin Time , Thrombin/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Factor VIII/pharmacology , Hemophilia A/blood , Hemophilia A/complications , Hemorrhage/etiology , Humans
3.
Blood ; 136(6): 740-748, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32369559

ABSTRACT

The bispecific antibody emicizumab is increasingly used for hemophilia A treatment. However, its specificity for human factors IX and X (FIX and FX) has limited its in vivo functional analysis to primate models of acquired hemophilia. Here, we describe a novel mouse model that allows emicizumab function to be examined. Briefly, FVIII-deficient mice received IV emicizumab 24 hours before tail-clip bleeding was performed. A second infusion with human FIX and FX, administered 5 minutes before bleeding, generated consistent levels of emicizumab (0.7-19 mg/dL for 0.5-10 mg/kg doses) and of both FIX and FX (85 and 101 U/dL, respectively, after dosing at 100 U/kg). Plasma from these mice display FVIII-like activity in assays (diluted activated partial thromboplastin time and thrombin generation), similar to human samples containing emicizumab. Emicizumab doses of 1.5 mg/kg and higher significantly reduced blood loss in a tail-clip-bleeding model using FVIII-deficient mice. However, reduction was incomplete compared with mice treated with human FVIII concentrate, and no difference in efficacy between doses was observed. From this model, we deducted FVIII-like activity from emicizumab that corresponded to a dose of 4.5 U of FVIII per kilogram (ie, 9.0 U/dL). Interestingly, combined with a low FVIII dose (5 U/kg), emicizumab provided enough additive activity to allow complete bleeding arrest. This model could be useful for further in vivo analysis of emicizumab.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor IX/administration & dosage , Factor X/administration & dosage , Hemophilia A/drug therapy , Hemorrhage/drug therapy , Models, Animal , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/immunology , Drug Therapy, Combination , Factor IX/analysis , Factor IX/immunology , Factor VIII/administration & dosage , Factor VIII/analysis , Factor VIII/therapeutic use , Factor X/analysis , Factor X/immunology , Factor XIa/pharmacology , Female , Hemophilia A/blood , Hemophilia A/complications , Hemophilia A/immunology , Hemorrhage/etiology , Infusions, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Partial Thromboplastin Time , Tail/injuries , Thrombin/biosynthesis
4.
Front Immunol ; 11: 476, 2020.
Article in English | MEDLINE | ID: mdl-32265927

ABSTRACT

Replacement therapy with coagulation factor VIII (FVIII) represents the current clinical treatment for patients affected by hemophilia A (HA). This treatment while effective is, however, hampered by the formation of antibodies which inhibit the activity of infused FVIII in up to 30% of treated patients. Immune tolerance induction (ITI) protocols, which envisage frequent infusions of high doses of FVIII to confront this side effect, dramatically increase the already high costs associated to a patient's therapy and are not always effective in all treated patients. Therefore, there are clear unmet needs that must be addressed in order to improve the outcome of these treatments for HA patients. Taking advantage of preclinical mouse models of hemophilia, several strategies have been proposed in recent years to prevent inhibitor formation and eradicate the pre-existing immunity to FVIII inhibitor positive patients. Herein, we will review some of the most promising strategies developed to avoid and eradicate inhibitors, including the use of immunomodulatory drugs or molecules, oral or transplacental delivery as well as cell and gene therapy approaches. The goal is to improve and potentiate the current ITI protocols and eventually make them obsolete.


Subject(s)
Factor VIII/immunology , Hemophilia A/drug therapy , Immune Tolerance/drug effects , Immunologic Factors/therapeutic use , Isoantibodies/immunology , Adoptive Transfer , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Drug Carriers , Drug Compounding , Drug Evaluation, Preclinical , Factor IXa/immunology , Factor VIII/administration & dosage , Factor VIII/therapeutic use , Factor X/immunology , Female , Fetal Therapies , Genetic Therapy , Hemophilia A/immunology , Hemophilia A/therapy , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunotherapy, Adoptive , Isoantibodies/biosynthesis , Lymphoid Tissue/immunology , Mice , Models, Animal , Plant Cells , Pregnancy , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/transplantation
5.
Int J Hematol ; 111(1): 20-30, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30350119

ABSTRACT

Hemophilia A is a congenital disorder caused by deficiency or malfunction of coagulation factor (F) VIII. While exogenously provided FVIII effectively reduces bleeding complications in many hemophilia A patients, multiple efforts are underway to develop new drugs to meet the needs that conventional FVIII agents do not. We have been long engaged in creating and clinically developing a humanized anti-FIXa/FX asymmetric bispecific IgG antibody with a FVIIIa-cofactor activity. Since this project was born from a creative and unique idea, our group recognized from the first that it would face many difficulties in the course of research including establishment of industrial manufacturability of an asymmetric bispecific IgG antibody. The group actually faced various challenges, but addressed all of them during about 10 years of research, and successfully created the potent humanized bispecific antibody, emicizumab. Emicizumab has showed clinical benefits in the human trials among which the first one was started in 2012, and has been currently approved in US, EU, Japan, and some other countries. It is now expected to improve the quality of life of patients and their families. In this article, we review the course of the research and clinical development of emicizumab, and describe its molecular characteristics.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor IXa/immunology , Factor VIIIa/metabolism , Factor X/immunology , Hemophilia A/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Blood Coagulation/drug effects , Factor VIII/immunology , Factor VIII/metabolism , Hemophilia A/immunology , Humans , Protein Engineering
6.
Sci Immunol ; 4(39)2019 09 20.
Article in English | MEDLINE | ID: mdl-31541031

ABSTRACT

Immune evasion in the tumor microenvironment (TME) is a crucial barrier for effective cancer therapy, and plasticity of innate immune cells may contribute to failures of targeted immunotherapies. Here, we show that rivaroxaban, a direct inhibitor of activated coagulation factor X (FX), promotes antitumor immunity by enhancing infiltration of dendritic cells and cytotoxic T cells at the tumor site. Profiling FX expression in the TME identifies monocytes and macrophages as crucial sources of extravascular FX. By generating mice with immune cells lacking the ability to produce FX, we show that myeloid cell-derived FX plays a pivotal role in promoting tumor immune evasion. In mouse models of cancer, we report that the efficacy of rivaroxaban is comparable with anti-programmed cell death ligand 1 (PD-L1) therapy and that rivaroxaban synergizes with anti-PD-L1 in improving antitumor immunity. Mechanistically, we demonstrate that FXa promotes immune evasion by signaling through protease-activated receptor 2 and that rivaroxaban specifically targets this cell-autonomous signaling pathway to reprogram tumor-associated macrophages. Collectively, our results have uncovered the importance of FX produced in the TME as a regulator of immune cell activation and suggest translational potential of direct oral anticoagulants to remove persisting roadblocks for immunotherapy and provide extravascular benefits in other diseases.


Subject(s)
Factor X/immunology , Mammary Neoplasms, Animal/immunology , Myeloid Cells/immunology , Animals , Female , Humans , Immunotherapy , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred C57BL
7.
Viruses ; 11(7)2019 07 05.
Article in English | MEDLINE | ID: mdl-31284434

ABSTRACT

Human adenovirus 5 (HAdV-5) is used as a vector in gene therapy clinical trials, hence its interactions with the host immune system have been widely studied. Previous studies have demonstrated that HAdV-5 binds specifically to murine coagulation factor X (mFX), inhibiting IgM and complement-mediated neutralization. Here, we examined the physical binding of immune components to HAdV-5 by nanoparticle tracking analysis, neutralization assays, mass spectrometry analysis and in vivo experiments. We observed that purified mouse Immunoglobulin M (IgM) antibodies bound to HAdV-5 only in the presence of complement components. Active serum components were demonstrated to bind to HAdV-5 in the presence or absence of mFX, indicating that immune molecules and mFX might bind to different sites. Since binding of mFX to HAdV-5 blocks the neutralization cascade, these findings suggested that not all complement-binding sites may be involved in virion neutralization. Furthermore, the data obtained from serum neutralization experiments suggested that immune molecules other than IgM and IgG may trigger activation of the complement cascade in vitro. In vivo experiments were conducted in immunocompetent C57BL/6 or immuno-deficient Rag2-/- mice. HAdV-5T* (a mutant HAdV-5 unable to bind to human or mFX) was neutralized to some extent in both mouse models, suggesting that murine immunoglobulins were not required for neutralization of HAdV-5 in vivo. Liquid Chromatography-Mass Spectrometry (LC-MS/MS) analysis of HAdV-5 and HAdV-5T* after exposure to murine sera showed stable binding of C3 and C4b in the absence of mFX. In summary, these results suggest that HAdV-5 neutralization can be mediated by both the classical and alternative pathways and that, in the absence of immunoglobulins, the complement cascade can be activated by direct binding of C3 to the virion.


Subject(s)
Adenoviruses, Human/immunology , Antibodies, Viral/immunology , Complement System Proteins/immunology , Immunoglobulin M/immunology , Adenoviruses, Human/genetics , Animals , Cell Line , Complement Activation , DNA-Binding Proteins/deficiency , Factor X/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neutralization Tests , Serogroup , Virion/immunology
8.
Pediatr Blood Cancer ; 66(11): e27886, 2019 11.
Article in English | MEDLINE | ID: mdl-31348595

ABSTRACT

BACKGROUND: Emicizumab is a bispecific antibody that bridges factor IXa and factor X to restore hemostasis in patients with hemophilia A (HA). Its efficacy and safety have been proven in multicenter trials. However, real world data regarding its use in very young children are currently lacking. Ancillary test results for monitoring emicizumab's hemostatic effect and their clinical correlations are scarce. METHODS: Children with HA and inhibitors treated by emicizumab were prospectively followed at our center. Laboratory follow-up included rotational thromboelastometry (ROTEM) and thrombin generation (TG), prior to and during treatment. RESULTS: Eleven children whose median age was 26 months were treated by emicizumab and followed for a median of 36 weeks. During follow-up, none experienced hemarthrosis or any other spontaneous bleeds. For 7/11 patients, emicizumab prophylaxis was sufficient to maintain hemostasis without additional supplemental therapy. Only 4/11 patients were occasionally treated with recombinant activated FVII for trauma. Two minor surgeries were safely performed without supplemental therapy while another procedure was complicated by major bleeding. TG parameters improved for all patients, correlating with their clinical status. Interestingly, the lowest TG values were obtained for patients experiencing bleeding episodes, while ROTEM parameters in all patients were close to the normal range. CONCLUSIONS: This study confirms the safety and efficacy of emicizumab in reducing bleeds in young children with HA with inhibitors, including infants. However, surgeries warrant caution as emicizumab prophylaxis may not be sufficient for some procedures. TG may more accurately reflect the hemostasis state than ROTEM in pediatric patients treated with emicizumab.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Hemophilia A/drug therapy , Hemorrhage/prevention & control , Blood Loss, Surgical/prevention & control , Child, Preschool , Factor IXa/immunology , Factor X/immunology , Follow-Up Studies , Hemophilia A/blood , Hemophilia A/complications , Hemorrhage/etiology , Humans , Infant , Male , Prospective Studies , Thrombelastography , Thrombin/analysis , Treatment Outcome
9.
Infect Immun ; 87(5)2019 03.
Article in English | MEDLINE | ID: mdl-30782860

ABSTRACT

Coagulation and inflammation are interconnected, suggesting that coagulation plays a key role in the inflammatory response to pathogens. A phenome-wide association study (PheWAS) was used to identify clinical phenotypes of patients with a polymorphism in coagulation factor X. Patients with this single nucleotide polymorphism (SNP) were more likely to be hospitalized with hemostatic and infection-related disorders, suggesting that factor X contributes to the immune response to infection. To investigate this, we modeled infections by human pathogens in a mouse model of factor X deficiency. Factor X-deficient mice were protected from systemic Acinetobacter baumannii infection, suggesting that factor X plays a role in the immune response to A. baumannii Factor X deficiency was associated with reduced cytokine and chemokine production and alterations in immune cell population during infection: factor X-deficient mice demonstrated increased abundance of neutrophils, macrophages, and effector T cells. Together, these results suggest that factor X activity is associated with an inefficient immune response and contributes to the pathology of A. baumannii infection.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter Infections/physiopathology , Acinetobacter baumannii/immunology , Factor X/genetics , Factor X/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Phenotype , Polymorphism, Genetic
11.
Yakugaku Zasshi ; 137(7): 831-836, 2017.
Article in Japanese | MEDLINE | ID: mdl-28674296

ABSTRACT

Nearly fifty monoclonal antibodies have been approved to date, and the market for monoclonal antibodies is expected to continue to grow. Since global competition in the field of antibody therapeutics is intense, we need to establish novel antibody engineering technologies to provide true benefit for patients, with differentiated product values. Bispecific antibodies are among the next generation of antibody therapeutics that can bind to two different target antigens by the two arms of immunoglobulin G (IgG) molecule, and are thus believed to be applicable to various therapeutic needs. Until recently, large scale manufacturing of human IgG bispecific antibody was impossible. We have established a technology, named asymmetric re-engineering technology (ART)-Ig, to enable large scale manufacturing of bispecific antibodies. Three examples of next generation antibody therapeutics using ART-Ig technology are described. Recent updates on bispecific antibodies against factor IXa and factor X for the treatment of hemophilia A, bispecific antibodies against a tumor specific antigen and T cell surface marker CD3 for cancer immunotherapy, and bispecific antibodies against two different epitopes of soluble antigen with pH-dependent binding property for the elimination of soluble antigen from plasma are also described.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal , Immunoglobulin G , Protein Engineering/methods , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Epitopes/immunology , Factor IXa/immunology , Factor X/immunology , Hemophilia A/therapy , Humans , Hydrogen-Ion Concentration , Immunotherapy , Neoplasms/therapy , Protein Binding , Solubility
12.
Thromb Haemost ; 117(7): 1348-1357, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28451690

ABSTRACT

Emicizumab, a humanised bispecific antibody recognising factors (F) IX/IXa and X/Xa, can accelerate FIXa-catalysed FX activation by bridging FIXa and FX in a manner similar to FVIIIa. However, details of the emicizumab-antigen interactions have not been reported so far. In this study, we first showed by surface plasmon resonance analysis that emicizumab bound FIX, FIXa, FX, and FXa with moderate affinities (KD = 1.58, 1.52, 1.85, and 0.978 µM, respectively). We next showed by immunoblotting analysis that emicizumab recognised the antigens' epidermal growth factor (EGF)-like domains. We then performed KD-based simulation of equilibrium states in plasma for quantitatively predicting the ways that emicizumab would interact with the antigens. The simulation predicted that only a small part of plasma FIX, FX, and emicizumab would form antigen-bridging FIX-emicizumab-FX ternary complex, of which concentration would form a bell-shaped relationship with emicizumab concentration. The bell-shaped concentration dependency was reproduced by plasma thrombin generation assays, suggesting that the plasma concentration of the ternary complex would correlate with emicizumab's cofactor activity. The simulation also predicted that at 10.0-100 µg/ml of emicizumab-levels shown in a previous study to be clinically effective-the majority of plasma FIX, FX, and emicizumab would exist as monomers. In conclusion, emicizumab binds FIX/FIXa and FX/FXa with micromolar affinities at their EGF-like domains. The KD-based simulation predicted that the antigen-bridging ternary complex formed in circulating plasma would correlate with emicizumab's cofactor activity, and the majority of FIX and FX would be free and available for other coagulation reactions.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Factor VIIIa/immunology , Antibodies, Bispecific/blood , Antibodies, Monoclonal, Humanized/blood , Antibody Specificity , Antigen-Antibody Reactions , Binding Sites , Biomimetic Materials/pharmacology , Computer Simulation , Factor IX/antagonists & inhibitors , Factor IX/immunology , Factor IXa/antagonists & inhibitors , Factor IXa/immunology , Factor X/antagonists & inhibitors , Factor X/immunology , Factor Xa/immunology , Factor Xa Inhibitors/blood , Factor Xa Inhibitors/immunology , Factor Xa Inhibitors/pharmacology , Humans , Models, Immunological
13.
Haemophilia ; 22 Suppl 5: 36-41, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27405674

ABSTRACT

The development of inhibitors to factor VIII (FVIII) or factor IX (FIX) remains a major treatment complication encountered in the treatment of haemophilia. Not all patients with even the same severity and genotype develop inhibitors suggesting an underlying mechanism of tolerance against FVIII- or FIX-related immunity. One mechanism may be central tolerance observed in patients in whom the FVIII mutation enables some production of the protein. The other is a peripheral tolerance mechanism which may be evident in patients with null mutation. Recently, recombinant porcine FVIII (rpFVIII, Obixur, OBI-1, BAX801) has been developed for the haemostatic treatment of both congenital haemophilia with inhibitor (CHAWI) and acquired haemophilia A (AHA). In 28 subjects with AHA with life-/limb-threatening bleeding, rpFVIII reduced or stopped bleeding in all patients within 24 h. The cross-reactivity of anti-human FVIII antibodies to rpFVIII remains around 30-50%. Recently, new therapeutics based on the quite novel concepts have been developed and clinical studies are ongoing. These are humanized asymmetric antibody mimicking FVIIIa function by maintaining a suitable interaction between FIXa and FX (Emicizumab, ACE910), and small interfering RNAs (siRNA, ALN-AT3) suppress liver production of AT through post-transcriptional gene silencing and a humanized anti-TFPI monoclonal antibody (Concizumab). Their main advantages are longer half-life, subcutaneous applicability and efficacy irrespective of the presence of inhibitors which will make it easier to initiate more effective treatment especially early childhood.


Subject(s)
Factor VIII/immunology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/blood , Factor VIII/therapeutic use , Factor X/immunology , Factor X/metabolism , Factor Xa/immunology , Factor Xa/metabolism , Hemophilia A/drug therapy , Hemophilia A/immunology , Humans , Immune Tolerance , RNA Interference
14.
Thromb Res ; 141 Suppl 2: S34-5, 2016 May.
Article in English | MEDLINE | ID: mdl-27207420

ABSTRACT

There are some issues in the current factor (F)VIII replacement therapy for severe hemophilia A. One is mental and physical burden for the multiple intravenous infusions, and the other is difficulty in the hemostatic treatment for the patients with FVIII inhibitor. The development of novel drug with fully hemostatic effect, simply procedure, and long-acting reaction has been expected. Recently, FVIIIa-mimicking humanized recombinant bispecific antibody (ACE910) against FIXa and FX was developed. In the non-human clinical study, primate model of acquired hemophilia A demonstrated that the ACE910 was effective on both on-going and spontaneous bleedings. A phase I clinical study was conducted in healthy adults by single subcutaneous infusion of ACE910, followed by the patients' part study, Japanese patients with severe hemophilia A without or with inhibitor were treated with once-weekly subcutaneous injection of ACE910 at three dose levels for 12 successive weeks. There was no significant adverse event related to ACE910 in the clinical and laboratorial findings, and t1/2 of ACE910 was ∼30 days. The median annual bleeding rates were reduced very markedly dose-dependently, independently of inhibitor. Furthermore, among the patients with dose escalation, bleeding rate was decreased as ACE910 dose was increased. In conclusion, ACE910 would have a number of promising features: its high subcutaneous bioavailability and long half-life make the patients possible to be injected subcutaneously with a once-a-week or less frequency. In addition, ACE910 would provide the bleeding prophylactic efficacy, independently of inhibitor.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor VIII/metabolism , Hemophilia A/drug therapy , Hemophilia A/metabolism , Hemostatics/therapeutic use , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Clinical Trials as Topic , Factor IXa/immunology , Factor IXa/metabolism , Factor X/immunology , Factor X/metabolism , Hemophilia A/blood , Hemophilia A/immunology , Hemorrhage/blood , Hemorrhage/immunology , Hemorrhage/metabolism , Hemorrhage/prevention & control , Hemostatics/immunology , Hemostatics/pharmacology , Humans
15.
Gene Ther ; 23(7): 592-6, 2016 07.
Article in English | MEDLINE | ID: mdl-27014840

ABSTRACT

Intravenous delivery of adenoviruses is the optimal route for many gene therapy applications. Once in the blood, coagulation factor X (FX) binds to the adenovirus capsid and protects the virion from natural antibody and classical complement-mediated neutralisation in mice. However, to date, no studies have examined the relevance of this FX/viral immune protective mechanism in human samples. In this study, we assessed the effects of blocking FX on adenovirus type 5 (Ad5) activity in the presence of human serum. FX prevented human IgM binding directly to the virus. In individual human sera samples (n=25), approximately half of those screened inhibited adenovirus transduction only when the Ad5-FX interaction was blocked, demonstrating that FX protected the virus from neutralising components in a large proportion of human sera. In contrast, the remainder of sera tested had no inhibitory effects on Ad5 transduction and FX armament was not required for effective gene transfer. In human sera in which FX had a protective role, Ad5 induced lower levels of complement activation in the presence of FX. We therefore demonstrate for the first time the importance of Ad-FX protection in human samples and highlight subject variability and species-specific differences as key considerations for adenoviral gene therapy.


Subject(s)
Adenoviridae/immunology , Factor X/immunology , Gene Transfer Techniques , Genetic Therapy/methods , Adenoviridae/genetics , Cell Line, Tumor , Genetic Vectors/blood , Genetic Vectors/genetics , Genetic Vectors/immunology , HEK293 Cells , Humans , Immunoglobulin M/immunology , Injections, Intravenous/methods
16.
Hum Gene Ther ; 27(2): 193-201, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26757054

ABSTRACT

Adenovirus (Ad) infection in humans is associated with inflammatory responses and thrombocytopenia. Although several studies were conducted in mice models to understand molecular and cellular mechanisms of Ad-induced inflammatory responses, only few of them turned their interest toward the mechanisms of Ad-induced thrombocytopenia. Using different depletion methods, the present study ruled out any significant role of spleen, macrophages, and vitamin K-dependent factor in Ad-induced thrombocytopenia. Interestingly, mice displaying thrombocytopenia expressed high levels of cytokines/chemokines after Ad administration. Most importantly, pseudotyping adenovirus with the fiber protein from other serotypes was associated with reduction of both cytokine/chemokine production and thrombocytopenia. Altogether, our results suggest that capsid fiber protein (and more precisely its shaft) of Ad serotype 5 triggers the cytokine production that leads to Ad-induced thrombocytopenia.


Subject(s)
Adenoviridae Infections/virology , Adenoviruses, Human/immunology , Capsid Proteins/immunology , Serogroup , Thrombocytopenia/virology , Adenoviridae Infections/complications , Adenoviridae Infections/immunology , Adenoviridae Infections/pathology , Adenoviruses, Human/genetics , Animals , Capsid Proteins/genetics , Cytokines/agonists , Cytokines/biosynthesis , Cytokines/immunology , Factor X/immunology , Female , Gene Expression , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Spleen/immunology , Spleen/virology , Thrombocytopenia/etiology , Thrombocytopenia/immunology , Thrombocytopenia/pathology
17.
Rinsho Ketsueki ; 56(6): 623-31, 2015 Jun.
Article in Japanese | MEDLINE | ID: mdl-26256871

ABSTRACT

Unmet needs of current hemophilia A treatment include the requirement for frequent intravenous infusions, inhibitor development, and containment of high medical costs. In order to overcome these issues, we produced FVIII which mimics a bispecific antibody against FIXa/FX. ACE910 demonstrated hemostatic effects on both ongoing and spontaneous joint bleeding in the primate acquired hemophilia A model. Recently, a phase 1 study for PK, PD, and the safety of ACE910 was initiated. The t1/2 was approximately 30 days. There were no severe ACE910 related adverse events. Furthermore, bleeding was remarkably decreased by weekly subcutaneous administration in patients with severe hemophilia A, regardless of whether an inhibitor was used. ACE910 has the remarkable advantages of prophylactic efficacy which can be achieved by convenient subcutaneous administrations at a markedly reduced frequency.


Subject(s)
Antibodies/therapeutic use , Factor IXa/immunology , Factor X/immunology , Hemophilia A/drug therapy , Hemophilia B/drug therapy , Animals , Clinical Trials as Topic , Hemophilia A/immunology , Hemophilia B/immunology , Humans
18.
PLoS Pathog ; 11(2): e1004673, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25658827

ABSTRACT

Adenoviruses are common pathogens, mostly targeting ocular, gastrointestinal and respiratory cells, but in some cases infection disseminates, presenting in severe clinical outcomes. Upon dissemination and contact with blood, coagulation factor X (FX) interacts directly with the adenovirus type 5 (Ad5) hexon. FX can act as a bridge to bind heparan sulphate proteoglycans, leading to substantial Ad5 hepatocyte uptake. FX "coating" also protects the virus from host IgM and complement-mediated neutralisation. However, the contribution of FX in determining Ad liver transduction whilst simultaneously shielding the virus from immune attack remains unclear. In this study, we demonstrate that the FX protection mechanism is not conserved amongst Ad types, and identify the hexon hypervariable regions (HVR) of Ad5 as the capsid proteins targeted by this host defense pathway. Using genetic and pharmacological approaches, we manipulate Ad5 HVR interactions to interrogate the interplay between viral cell transduction and immune neutralisation. We show that FX and inhibitory serum components can co-compete and virus neutralisation is influenced by both the location and extent of modifications to the Ad5 HVRs. We engineered Ad5-derived HVRs into the rare, native non FX-binding Ad26 to create Ad26.HVR5C. This enabled the virus to interact with FX at high affinity, as quantified by surface plasmon resonance, FX-mediated cell binding and transduction assays. Concomitantly, Ad26.HVR5C was also sensitised to immune attack in the absence of FX, a direct consequence of the engineered HVRs from Ad5. In both immune competent and deficient animals, Ad26.HVR5C hepatic gene transfer was mediated by FX following intravenous delivery. This study gives mechanistic insight into the pivotal role of the Ad5 HVRs in conferring sensitivity to virus neutralisation by IgM and classical complement-mediated attack. Furthermore, through this gain-of-function approach we demonstrate the dual functionality of FX in protecting Ad26.HVR5C against innate immune factors whilst determining liver targeting.


Subject(s)
Adenoviruses, Human/immunology , Antibodies, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Factor X/immunology , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/prevention & control , Adenoviruses, Human/genetics , Animals , Antibodies, Neutralizing/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line, Tumor , Genetic Variation/genetics , Genetic Vectors/genetics , HEK293 Cells , HeLa Cells , Humans , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mice , Mice, Inbred C57BL , Surface Plasmon Resonance , Transduction, Genetic , Virus Attachment
20.
Blood ; 124(20): 3165-71, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25274508

ABSTRACT

ACE910 is a humanized anti-factor IXa/X bispecific antibody mimicking the function of factor VIII (FVIII). We previously demonstrated in nonhuman primates that a single IV dose of ACE910 exerted hemostatic activity against hemophilic bleeds artificially induced in muscles and subcutis, and that a subcutaneous (SC) dose of ACE910 showed a 3-week half-life and nearly 100% bioavailability, offering support for effective prophylaxis for hemophilia A by user-friendly SC dosing. However, there was no direct evidence that such SC dosing of ACE910 would prevent spontaneous bleeds occurring in daily life. In this study, we newly established a long-term primate model of acquired hemophilia A by multiple IV injections of an anti-primate FVIII neutralizing antibody engineered in mouse-monkey chimeric form to reduce its antigenicity. The monkeys in the control group exhibited various spontaneous bleeding symptoms as well as continuous prolongation of activated partial thromboplastin time; notably, all exhibited joint bleeds, which are a hallmark of hemophilia. Weekly SC doses of ACE910 (initial 3.97 mg/kg followed by 1 mg/kg) significantly prevented these bleeding symptoms; notably, no joint bleeding symptoms were observed. ACE910 is expected to prevent spontaneous bleeds and joint damage in hemophilia A patients even with weekly SC dosing, although appropriate clinical investigation is required.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor IXa/immunology , Factor X/immunology , Hemophilia A/complications , Hemorrhage/complications , Hemorrhage/prevention & control , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal, Humanized/immunology , Cell Line , Disease Models, Animal , Hemorrhage/pathology , Humans , Joints/drug effects , Joints/pathology , Macaca fascicularis , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...