Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Aging Cell ; 22(5): e13806, 2023 05.
Article in English | MEDLINE | ID: mdl-36967480

ABSTRACT

Accumulation of senescent cells (SNCs) with a senescence-associated secretory phenotype (SASP) has been implicated as a major source of chronic sterile inflammation leading to many age-related pathologies. Herein, we provide evidence that a bifunctional immunotherapeutic, HCW9218, with capabilities of neutralizing TGF-ß and stimulating immune cells, can be safely administered systemically to reduce SNCs and alleviate SASP in mice. In the diabetic db/db mouse model, subcutaneous administration of HCW9218 reduced senescent islet ß cells and SASP resulting in improved glucose tolerance, insulin resistance, and aging index. In naturally aged mice, subcutaneous administration of HCW9218 durably reduced the level of SNCs and SASP, leading to lower expression of pro-inflammatory genes in peripheral organs. HCW9218 treatment also reverted the pattern of key regulatory circadian gene expression in aged mice to levels observed in young mice and impacted genes associated with metabolism and fibrosis in the liver. Single-nucleus RNA Sequencing analysis further revealed that HCW9218 treatment differentially changed the transcriptomic landscape of hepatocyte subtypes involving metabolic, signaling, cell-cycle, and senescence-associated pathways in naturally aged mice. Long-term survival studies also showed that HCW9218 treatment improved physical performance without compromising the health span of naturally aged mice. Thus, HCW9218 represents a novel immunotherapeutic approach and a clinically promising new class of senotherapeutic agents targeting cellular senescence-associated diseases.


Subject(s)
Cellular Senescence , Senescence-Associated Secretory Phenotype , Mice , Animals , Cellular Senescence/genetics , Aging , Inflammation , Immunotherapy , Phenotype
2.
Cancer Immunol Res ; 9(9): 1071-1087, 2021 09.
Article in English | MEDLINE | ID: mdl-34244297

ABSTRACT

Natural killer (NK) cells are a promising cellular therapy for cancer, with challenges in the field including persistence, functional activity, and tumor recognition. Briefly, priming blood NK cells with recombinant human (rh)IL-12, rhIL-15, and rhIL-18 (12/15/18) results in memory-like NK cell differentiation and enhanced responses against cancer. However, the lack of available, scalable Good Manufacturing Process (GMP)-grade reagents required to advance this approach beyond early-phase clinical trials is limiting. To address this challenge, we developed a novel platform centered upon an inert tissue factor scaffold for production of heteromeric fusion protein complexes (HFPC). The first use of this platform combined IL-12, IL-15, and IL-18 receptor engagement (HCW9201), and the second adds CD16 engagement (HCW9207). This unique HFPC expression platform was scalable with equivalent protein quality characteristics in small- and GMP-scale production. HCW9201 and HCW9207 stimulated activation and proliferation signals in NK cells, but HCW9207 had decreased IL-18 receptor signaling. RNA sequencing and multidimensional mass cytometry revealed parallels between HCW9201 and 12/15/18. HCW9201 stimulation improved NK cell metabolic fitness and resulted in the DNA methylation remodeling characteristic of memory-like differentiation. HCW9201 and 12/15/18 primed similar increases in short-term and memory-like NK cell cytotoxicity and IFNγ production against leukemia targets, as well as equivalent control of leukemia in NSG mice. Thus, HFPCs represent a protein engineering approach that solves many problems associated with multisignal receptor engagement on immune cells, and HCW9201-primed NK cells can be advanced as an ideal approach for clinical GMP-grade memory-like NK cell production for cancer therapy.


Subject(s)
Interleukin-12/pharmacology , Interleukin-15/pharmacology , Interleukin-18/pharmacology , Killer Cells, Natural/immunology , Leukemia/therapy , Animals , Cell Line, Tumor , Humans , Immunologic Memory/drug effects , Leukemia/immunology , Mice , Receptors, Natural Killer Cell/metabolism , Recombinant Fusion Proteins/pharmacology , Remission Induction , Xenograft Model Antitumor Assays
3.
Math Biosci Eng ; 18(4): 4586-4602, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34198455

ABSTRACT

Semi-supervised learning has always been a hot topic in machine learning. It uses a large number of unlabeled data to improve the performance of the model. This paper combines the co-training strategy and random forest to propose a novel semi-supervised regression algorithm: semi-supervised random forest regression model based on co-training and grouping with information entropy (E-CoGRF), and applies it to the evaluation of depression symptoms severity. The algorithm inherits the ensemble characteristics of random forest, and combines well with co-training. In order to balance the accuracy and diversity of co-training random forests, the algorithm proposes a grouping strategy to decision trees. Moreover, the information entropy is used to measure the confidence, which avoids unnecessary repeated training and improves the efficiency of the model. In the practical application of evaluation of depression symptoms severity, we collect cognitive behavioral data of emotional conflict based on the depressive affective disorder. And on this basis, feature construction and normalization preprocessing are carried out. Finally, the test is conducted on 35 labeled and 80 unlabeled depression patients. The result shows that the proposed algorithm obtains MAE (Mean Absolute Error) = 3.63 and RMSE (Root Mean Squared Error) = 4.50, which is better than other semi-supervised regression algorithms. The proposed method effectively solves the modeling difficulties caused by insufficient labeled samples, and has important reference value for the diagnosis of depression symptoms severity.


Subject(s)
Depression , Supervised Machine Learning , Algorithms , Depression/diagnosis , Entropy , Humans , Machine Learning
4.
Mol Ther ; 29(10): 2949-2962, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34091051

ABSTRACT

Advances in immunostimulatory and anti-immunosuppressive therapeutics have revolutionized cancer treatment. However, novel immunotherapeutics with these dual functions are not frequently reported. Here we describe the creation of a heterodimeric bifunctional fusion molecule, HCW9218, constructed using our soluble tissue factor (TF)-based scaffold technology. This complex comprises extracellular domains of the human transforming growth factor-ß (TGF-ß) receptor II and a human interleukin-15 (IL-15)/IL-15 receptor α complex. HCW9218 can be readily expressed in CHO cells and purified using antibody-based affinity chromatography in a large-scale manufacturing setting. HCW9218 potently activates mouse natural killer (NK) cells and CD8+ T cells in vitro and in vivo to enhance cell proliferation, metabolism, and antitumor cytotoxic activities. Similarly, human immune cells become activated with increased cytotoxicity following incubation with HCW9218. This fusion complex also exhibits TGF-ß neutralizing activity in vitro and sequesters plasma TGF-ß in vivo. In a syngeneic B16F10 melanoma model, HCW9218 displayed strong antitumor activity mediated by NK cells and CD8+ T cells and increased their infiltration into tumors. Repeat-dose subcutaneous administration of HCW9218 was well tolerated by mice, with a half-life sufficient to provide long-lasting biological activity. Thus, HCW9218 may serve as a novel therapeutic to simultaneously provide immunostimulation and lessen immunosuppression associated with tumors.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Interleukin-15/genetics , Killer Cells, Natural/metabolism , Melanoma, Experimental/drug therapy , Receptor, Transforming Growth Factor-beta Type II/chemistry , Receptors, Interleukin-15/genetics , Recombinant Fusion Proteins/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Injections, Subcutaneous , Interleukin-15/metabolism , Melanoma, Experimental/immunology , Mice , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Interleukin-15/metabolism , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
5.
Viruses ; 11(2)2019 01 22.
Article in English | MEDLINE | ID: mdl-30678320

ABSTRACT

Zika virus (ZIKV) infection can cause severe congenital diseases, such as microcephaly, ocular defects and arthrogryposis in fetuses, and Guillain⁻Barré syndrome in adults. Efficacious therapeutic treatments for infected patients, as well as prophylactic treatments to prevent new infections are needed for combating ZIKV infection. Here, we report that ZIKV-specific human polyclonal antibodies (SAB-155), elicited in transchromosomal bovine (TcB), provide significant protection from infection by ZIKV in STAT2 knockout (KO) golden Syrian hamsters both prophylactically and therapeutically. These antibodies also prevent testicular lesions in this hamster model. Our data indicate that antibody-mediated immunotherapy is effective in treating ZIKV infection. Because suitable quantities of highly potent human polyclonal antibodies can be quickly produced from the TcB system against ZIKV and have demonstrated therapeutic efficacy in a small animal model, they have the potential as an effective countermeasure against ZIKV infection.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Immunization, Passive , STAT2 Transcription Factor/genetics , Zika Virus Infection/prevention & control , Zika Virus Infection/therapy , Animals , Animals, Genetically Modified , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cattle , Cricetinae , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Male , Testis/pathology , Testis/virology , Zika Virus
6.
J Infect Dis ; 218(suppl_5): S636-S648, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30010950

ABSTRACT

Transchromosomic bovines (Tc-bovines) adaptively produce fully human polyclonal immunoglobulin (Ig)G antibodies after exposure to immunogenic antigen(s). The National Interagency Confederation for Biological Research and collaborators rapidly produced and then evaluated anti-Ebola virus IgG immunoglobulins (collectively termed SAB-139) purified from Tc-bovine plasma after sequential hyperimmunization with an Ebola virus Makona isolate glycoprotein nanoparticle vaccine. SAB-139 was characterized by several in vitro production, research, and clinical level assays using wild-type Makona-C05 or recombinant virus/antigens from different Ebola virus variants. SAB-139 potently activates natural killer cells, monocytes, and peripheral blood mononuclear cells and has high-binding avidity demonstrated by surface plasmon resonance. SAB-139 has similar concentrations of galactose-α-1,3-galactose carbohydrates compared with human-derived intravenous Ig, and the IgG1 subclass antibody is predominant. All rhesus macaques infected with Ebola virus/H.sapiens-tc/GIN/2014/Makona-C05 and treated with sufficient SAB-139 at 1 day (n = 6) or 3 days (n = 6) postinfection survived versus 0% of controls. This study demonstrates that Tc-bovines can produce pathogen-specific human Ig to prevent and/or treat patients when an emerging infectious disease either threatens to or becomes an epidemic.


Subject(s)
Antibodies, Viral/therapeutic use , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/drug therapy , Immunoglobulin G/therapeutic use , Animals , Cattle , Chlorocebus aethiops , Female , Humans , Macaca mulatta , Male , Vero Cells
7.
J Infect Dis ; 218(suppl_5): S658-S661, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30053153

ABSTRACT

Antibody therapy has been used to treat a variety of diseases and the success of ZMapp and other monoclonal antibody-based therapies during the 2014-2016 West African Ebola outbreak has shown this countermeasure can be a successful therapy for Ebola hemorrhagic fever. This study utilized transchromosomal bovines (TcB) vaccinated with a DNA plasmid encoding Ebola virus glycoprotein sequence to produce human polyclonal antibodies directed against Ebola virus glycoprotein. When administered 1 day postinfection, these TcB polyclonal antibodies provided partial protection and resulted in a 50% survival rate following a lethal challenge of Ebola virus Makona in rhesus macaques.


Subject(s)
Antibodies, Viral/therapeutic use , Hemorrhagic Fever, Ebola/prevention & control , Animals , Cattle , Hemorrhagic Fever, Ebola/mortality , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Humans , Macaca mulatta , RNA, Viral/analysis
8.
Lancet Infect Dis ; 18(4): 410-418, 2018 04.
Article in English | MEDLINE | ID: mdl-29329957

ABSTRACT

BACKGROUND: Middle East respiratory syndrome (MERS) is a severe respiratory illness with an overall mortality of 35%. There is no licensed or proven treatment. Passive immunotherapy approaches are being developed to prevent and treat several human medical conditions where alternative therapeutic options are absent. We report the safety of a fully human polyclonal IgG antibody (SAB-301) produced from the hyperimmune plasma of transchromosomic cattle immunised with a MERS coronavirus vaccine. METHODS: We did a phase 1 double-blind, placebo-controlled, single-dose escalation trial at the National Institutes of Health Clinical Center. We recruited healthy participants aged 18-60 years who had normal laboratory parameters at enrolment, a body-mass index of 19-32 kg/m2, and a creatinine clearance of 70 mL/min or more, and who did not have any chronic medical problems that required daily oral medications, a positive rheumatoid factor (≥15 IU/mL), IgA deficiency (<7 mg/dL), or history of allergy to intravenous immunoglobulin or human blood products. Participants were randomly assigned by a computer-generated table, made by a masked pharmacist, to one of six cohorts (containing between three and ten participants each). Cohorts 1 and 2 had three participants, randomly assigned 2:1 to receive active drug SAB-301 versus normal saline placebo; cohorts 3 and 4 had six participants randomised 2:1; and cohorts 5 and 6 had ten participants, randomised 4:1. Participants received 1 mg/kg, 2·5 mg/kg, 5 mg/kg, 10 mg/kg, 20 mg/kg, or 50 mg/kg of SAB-301, or equivalent volume placebo (saline control), on day 0, and were followed up by clinical, laboratory, and pharmacokinetic assessments on days 1, 3, 7, 21, 42, and 90. The primary outcome was safety, and immunogenicity was a secondary outcome. We analysed the intention-to-treat population. This trial is registered with ClinicalTrials.gov, number NCT02788188. FINDINGS: Between June 2, 2016, and Jan 4, 2017, we screened 43 participants, of whom 38 were eligible and randomly assigned to receive SAB-301 (n=28) or placebo (n=10). 97 adverse events were reported: 64 adverse events occurred in 23 (82%) of 28 participants receiving SAB-301 (mean 2·3 adverse events per participant). 33 adverse events occurred in all ten participants receiving placebo (mean 3·3 adverse events per participant). The most common adverse events were headache (n=6 [21%] in participants who received SAB-301 and n=2 [20%] in those receiving placebo), albuminuria (n=5 [18%] vs n=2 [20%]), myalgia (n=3 [11%] vs n=1 [10%]), increased creatine kinase (n=3 [11%] vs 1 [10%]), and common cold (n=3 [11%] vs n=2 [20%]). There was one serious adverse event (hospital admission for suicide attempt) in one participant who received 50 mg/kg of SAB-301. The area under the concentration-time curve (AUC) in the 50 mg/kg dose (27 498 µg × days per mL) is comparable to the AUC that was associated with efficacy in a preclinical model. INTERPRETATION: Single infusions of SAB-301 up to 50 mg/kg appear to be safe and well tolerated in healthy participants. Human immunoglobulin derived from transchromosomic cattle could offer a new platform technology to produce fully human polyclonal IgG antibodies for other medical conditions. FUNDING: National Institute of Allergy and Infectious Diseases, National Institutes of Health, and Biomedical Advanced Research and Development Authority.


Subject(s)
Antibodies, Viral/administration & dosage , Antibodies, Viral/adverse effects , Immunization, Passive/adverse effects , Immunization, Passive/methods , Middle East Respiratory Syndrome Coronavirus/immunology , Adult , Animals , Animals, Genetically Modified , Cattle , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Follow-Up Studies , Healthy Volunteers , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/adverse effects , Infusions, Intravenous , Male , Middle Aged , National Institutes of Health (U.S.) , Placebos/administration & dosage , United States , Young Adult
10.
Antiviral Res ; 146: 164-173, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28893603

ABSTRACT

Zika virus (ZIKV) is rapidly spreading throughout the Americas and is associated with significant fetal complications, most notably microcephaly. Treatment with polyclonal antibodies for pregnant women at risk of ZIKV-related complications could be a safe alternative to vaccination. We found that large quantities of human polyclonal antibodies could be rapidly produced in transchromosomal bovines (TcB) and successfully used to protect mice from lethal infection. Additionally, antibody treatment eliminated ZIKV induced tissue damage in immunologically privileged sites such as the brain and testes and protected against testicular atrophy. These data indicate that rapid development and deployment of human polyclonal antibodies could be a viable countermeasure against ZIKV.


Subject(s)
Antibodies, Viral/therapeutic use , Testis/pathology , Zika Virus Infection/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Viral/administration & dosage , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Atrophy/prevention & control , Cattle , Disease Models, Animal , Female , Humans , Male , Mice , Pregnancy , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/virology
11.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28468884

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a mosquito-borne RNA virus that causes low mortality but high morbidity rates in humans. In addition to natural outbreaks, there is the potential for exposure to VEEV via aerosolized virus particles. There are currently no FDA-licensed vaccines or antiviral therapies for VEEV. Passive immunotherapy is an approved method used to protect individuals against several pathogens and toxins. Human polyclonal antibodies (PAbs) are ideal, but this is dependent upon serum from convalescent human donors, which is in limited supply. Non-human-derived PAbs can have serious immunoreactivity complications, and when "humanized," these antibodies may exhibit reduced neutralization efficiency. To address these issues, transchromosomic (Tc) bovines have been created, which can produce potent neutralizing human antibodies in response to hyperimmunization. In these studies, we have immunized these bovines with different VEEV immunogens and evaluated the protective efficacy of purified preparations of the resultant human polyclonal antisera against low- and high-dose VEEV challenges. These studies demonstrate that prophylactic or therapeutic administration of the polyclonal antibody preparations (TcPAbs) can protect mice against lethal subcutaneous or aerosol challenge with VEEV. Furthermore, significant protection against unrelated coinfecting viral pathogens can be conferred by combining individual virus-specific TcPAb preparations.IMPORTANCE With the globalization and spread or potential aerosol release of emerging infectious diseases, it will be critical to develop platforms that are able to produce therapeutics in a short time frame. By using a transchromosomic (Tc) bovine platform, it is theoretically possible to produce antigen-specific highly neutralizing therapeutic polyclonal human antibody (TcPAb) preparations in 6 months or less. In this study, we demonstrate that Tc bovine-derived Venezuelan equine encephalitis virus (VEEV)-specific TcPAbs are highly effective against VEEV infection that mimics not only the natural route of infection but also infection via aerosol exposure. Additionally, we show that combinatorial TcPAb preparations can be used to treat coinfections with divergent pathogens, demonstrating that the Tc bovine platform could be beneficial in areas where multiple infectious diseases occur contemporaneously or in the case of multipathogen release.


Subject(s)
Animals, Genetically Modified , Antibodies, Viral/administration & dosage , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Encephalomyelitis, Venezuelan Equine/therapy , Immunization, Passive , Animals , Antibodies, Viral/isolation & purification , Cattle , Disease Models, Animal , Humans , Mice , Treatment Outcome
12.
Sci Rep ; 6: 24897, 2016 04 25.
Article in English | MEDLINE | ID: mdl-27109916

ABSTRACT

Polyclonal antibodies, derived from humans or hyperimmunized animals, have been used prophylactically or therapeutically as countermeasures for a variety of infectious diseases. SAB Biotherapeutics has successfully developed a transchromosomic (Tc) bovine platform technology that can produce fully human immunoglobulins rapidly, and in substantial quantities, against a variety of disease targets. In this study, two Tc bovines expressing high levels of fully human IgG were hyperimmunized with a recombinant glycoprotein (GP) vaccine consisting of the 2014 Ebola virus (EBOV) Makona isolate. Serum collected from these hyperimmunized Tc bovines contained high titers of human IgG against EBOV GP as determined by GP specific ELISA, surface plasmon resonance (SPR), and virus neutralization assays. Fully human polyclonal antibodies against EBOV were purified and evaluated in a mouse challenge model using mouse adapted Ebola virus (maEBOV). Intraperitoneal administration of the purified anti-EBOV IgG (100 mg/kg) to BALB/c mice one day after lethal challenge with maEBOV resulted in 90% protection; whereas 100% of the control animals succumbed. The results show that hyperimmunization of Tc bovines with EBOV GP can elicit protective and potent neutralizing fully human IgG antibodies rapidly and in commercially viable quantities.


Subject(s)
Animals, Genetically Modified , Antibodies, Viral/blood , Antibodies, Viral/therapeutic use , Cattle , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunization, Passive , Immunoglobulin G/blood , Immunoglobulin G/therapeutic use , Mice, Inbred BALB C , Neutralization Tests , Surface Plasmon Resonance , Treatment Outcome
13.
Sci Transl Med ; 8(326): 326ra21, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26888429

ABSTRACT

As of 13 November 2015, 1618 laboratory-confirmed human cases of Middle East respiratory syndrome coronavirus (MERS-CoV) infection, including 579 deaths, had been reported to the World Health Organization. No specific preventive or therapeutic agent of proven value against MERS-CoV is currently available. Public Health England and the International Severe Acute Respiratory and Emerging Infection Consortium identified passive immunotherapy with neutralizing antibodies as a treatment approach that warrants priority study. Two experimental MERS-CoV vaccines were used to vaccinate two groups of transchromosomic (Tc) bovines that were genetically modified to produce large quantities of fully human polyclonal immunoglobulin G (IgG) antibodies. Vaccination with a clade A γ-irradiated whole killed virion vaccine (Jordan strain) or a clade B spike protein nanoparticle vaccine (Al-Hasa strain) resulted in Tc bovine sera with high enzyme-linked immunosorbent assay (ELISA) and neutralizing antibody titers in vitro. Two purified Tc bovine human IgG immunoglobulins (Tc hIgG), SAB-300 (produced after Jordan strain vaccination) and SAB-301 (produced after Al-Hasa strain vaccination), also had high ELISA and neutralizing antibody titers without antibody-dependent enhancement in vitro. SAB-301 was selected for in vivo and preclinical studies. Administration of single doses of SAB-301 12 hours before or 24 and 48 hours after MERS-CoV infection (Erasmus Medical Center 2012 strain) of Ad5-hDPP4 receptor-transduced mice rapidly resulted in viral lung titers near or below the limit of detection. Tc bovines, combined with the ability to quickly produce Tc hIgG and develop in vitro assays and animal model(s), potentially offer a platform to rapidly produce a therapeutic to prevent and/or treat MERS-CoV infection and/or other emerging infectious diseases.


Subject(s)
Chromosomes, Mammalian/genetics , Immunoglobulin G/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibody-Dependent Enhancement , Cattle , Dipeptidyl Peptidase 4/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Mice, Inbred BALB C , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/physiology , Neutralization Tests , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transduction, Genetic , Vaccination , Virus Replication
14.
PLoS One ; 10(9): e0137786, 2015.
Article in English | MEDLINE | ID: mdl-26422247

ABSTRACT

DNA vaccination of transchromosomal bovines (TcBs) with DNA vaccines expressing the codon-optimized (co) glycoprotein (GP) genes of Ebola virus (EBOV) and Sudan virus (SUDV) produce fully human polyclonal antibodies (pAbs) that recognize both viruses and demonstrate robust neutralizing activity. Each TcB was vaccinated by intramuscular electroporation (IM-EP) a total of four times and at each administration received 10 mg of the EBOV-GPco DNA vaccine and 10 mg of the SUDV-GPco DNA vaccine at two sites on the left and right sides, respectively. After two vaccinations, robust antibody responses (titers > 1000) were detected by ELISA against whole irradiated EBOV or SUDV and recombinant EBOV-GP or SUDV-GP (rGP) antigens, with higher titers observed for the rGP antigens. Strong, virus neutralizing antibody responses (titers >1000) were detected after three vaccinations when measured by vesicular stomatitis virus-based pseudovirion neutralization assay (PsVNA). Maximal neutralizing antibody responses were identified by traditional plaque reduction neutralization tests (PRNT) after four vaccinations. Neutralizing activity of human immunoglobulins (IgG) purified from TcB plasma collected after three vaccinations and injected intraperitoneally (IP) into mice at a 100 mg/kg dose was detected in the serum by PsVNA up to 14 days after administration. Passive transfer by IP injection of the purified IgG (100 mg/kg) to groups of BALB/c mice one day after IP challenge with mouse adapted (ma) EBOV resulted in 80% protection while all mice treated with non-specific pAbs succumbed. Similarly, interferon receptor 1 knockout (IFNAR(-/-)) mice receiving the purified IgG (100 mg/kg) by IP injection one day after IP challenge with wild type SUDV resulted in 89% survival. These results are the first to demonstrate that filovirus GP DNA vaccines administered to TcBs by IM-EP can elicit neutralizing antibodies that provide post-exposure protection. Additionally, these data describe production of fully human IgG in a large animal system, a system which is capable of producing large quantities of a clinical grade therapeutic product.


Subject(s)
Antibodies, Viral/metabolism , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Post-Exposure Prophylaxis , Vaccines, DNA/immunology , Animals , Animals, Genetically Modified , Antibodies, Neutralizing/immunology , Cattle/genetics , Cattle/immunology , Chromosomes, Artificial, Human/genetics , Democratic Republic of the Congo , Ebola Vaccines/immunology , Female , Hemorrhagic Fever, Ebola/virology , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Post-Exposure Prophylaxis/methods , Receptor, Interferon alpha-beta/genetics , Sudan , Vaccination/methods
15.
PLoS One ; 10(6): e0130699, 2015.
Article in English | MEDLINE | ID: mdl-26107496

ABSTRACT

Large-scale production of fully human IgG (hIgG) or human polyclonal antibodies (hpAbs) by transgenic animals could be useful for human therapy. However, production level of hpAbs in transgenic animals is generally very low, probably due to the fact that evolutionarily unique interspecies-incompatible genomic sequences between human and non-human host species may impede high production of fully hIgG in the non-human environment. To address this issue, we performed species-specific human artificial chromosome (HAC) engineering and tested these engineered HAC in cattle. Our previous study has demonstrated that site-specific genomic chimerization of pre-B cell receptor/B cell receptor (pre-BCR/BCR) components on HAC vectors significantly improves human IgG expression in cattle where the endogenous bovine immunoglobulin genes were knocked out. In this report, hIgG1 class switch regulatory elements were subjected to site-specific genomic chimerization on HAC vectors to further enhance hIgG expression and improve hIgG subclass distribution in cattle. These species-specific modifications in a chromosome scale resulted in much higher production levels of fully hIgG of up to 15 g/L in sera or plasma, the highest ever reported for a transgenic animal system. Transchromosomic (Tc) cattle containing engineered HAC vectors generated hpAbs with high titers against human-origin antigens following immunization. This study clearly demonstrates that species-specific sequence differences in pre-BCR/BCR components and IgG1 class switch regulatory elements between human and bovine are indeed functionally distinct across the two species, and therefore, are responsible for low production of fully hIgG in our early versions of Tc cattle. The high production levels of fully hIgG with hIgG1 subclass dominancy in a large farm animal species achieved here is an important milestone towards broad therapeutic applications of hpAbs.


Subject(s)
Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal/biosynthesis , Chromosomes, Artificial, Human/immunology , Genetic Vectors/metabolism , Immunoglobulin G/biosynthesis , Pre-B Cell Receptors/immunology , Animals , Animals, Genetically Modified , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antigens/chemistry , Antigens/immunology , Cattle , Cell Line, Tumor , Chickens , Chromosome Mapping , Chromosomes, Artificial, Human/chemistry , Gene Knockout Techniques , Genetic Engineering , Genetic Vectors/chemistry , Humans , Immunization , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Pre-B Cell Receptors/genetics , Species Specificity
16.
Sci Transl Med ; 6(264): 264ra162, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25429055

ABSTRACT

Polyclonal immunoglobulin-based medical products have been used successfully to treat diseases caused by viruses for more than a century. We demonstrate the use of DNA vaccine technology and transchromosomal bovines (TcBs) to produce fully human polyclonal immunoglobulins (IgG) with potent antiviral neutralizing activity. Specifically, two hantavirus DNA vaccines [Andes virus (ANDV) DNA vaccine and Sin Nombre virus (SNV) DNA vaccine] were used to produce a candidate immunoglobulin product for the prevention and treatment of hantavirus pulmonary syndrome (HPS). A needle-free jet injection device was used to vaccinate TcB, and high-titer neutralizing antibodies (titers >1000) against both viruses were produced within 1 month. Plasma collected at day 10 after the fourth vaccination was used to produce purified α-HPS TcB human IgG. Treatment with 20,000 neutralizing antibody units (NAU)/kg starting 5 days after challenge with ANDV protected seven of eight animals, whereas zero of eight animals treated with the same dose of normal TcB human IgG survived. Likewise, treatment with 20,000 NAU/kg starting 5 days after challenge with SNV protected immunocompromised hamsters from lethal HPS, protecting five of eight animals. Our findings that the α-HPS TcB human IgG is capable of protecting in animal models of lethal HPS when administered after exposure provides proof of concept that this approach can be used to develop candidate next-generation polyclonal immunoglobulin-based medical products without the need for human donors, despeciation protocols, or inactivated/attenuated vaccine antigen.


Subject(s)
Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/prevention & control , Immunoglobulin G/biosynthesis , Vaccines, DNA/immunology , Animals , Animals, Genetically Modified , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Biological Availability , Cattle , Cricetinae , Disease Models, Animal , Female , Orthohantavirus/immunology , Hantavirus Pulmonary Syndrome/virology , Humans , Sin Nombre virus/immunology , Vaccination
17.
PLoS One ; 9(3): e90383, 2014.
Article in English | MEDLINE | ID: mdl-24603704

ABSTRACT

Towards the goal of producing fully human polyclonal antibodies (hpAbs or hIgGs) in transchromosomic (Tc) cattle, we previously reported that Tc cattle carrying a human artificial chromosome (HAC) comprising the entire unrearranged human immunoglobulin (Ig) heavy-chain (hIGH), kappa-chain (hIGK), and lambda-chain (hIGL) germline loci produced physiological levels of hIgGs when both of the bovine immunoglobulin mu heavy-chains, bIGHM and bIGHML1, were homozygously inactivated (bIGHM-/-, bIGHML1-/-; double knockouts or DKO). However, because endogenous bovine immunoglobulin light chain loci are still intact, the light chains are produced both from the hIGK and hIGL genomic loci on the HAC and from the endogenous bovine kappa-chain (bIGK) and lambda-chain (bIGL) genomic loci, resulting in the production of fully hIgGs (both Ig heavy-chains and light-chains are of human origin: hIgG/hIgκ or hIgG/hIgλ) and chimeric hIgGs (Ig heavy-chains are of human origin while the Ig light-chains are of bovine origin: hIgG/bIgκ or hIgG/bIgλ). To improve fully hIgG production in Tc cattle, we here report the deletion of the entire bIGL joining (J) and constant (C) gene cluster (bIGLJ1-IGLC1 to bIGLJ5-IGLC5) by employing Cre/loxP mediated site-specific chromosome recombination and the production of triple knockout (bIGHM-/-, bIGHML1-/- and bIGL-/-; TKO) Tc cattle. We further demonstrate that bIGL cluster deletion greatly improves fully hIgGs production in the sera of TKO Tc cattle, with 51.3% fully hIgGs (hIgG/hIgκ plus hIgG/hIgλ).


Subject(s)
Antibodies, Monoclonal/immunology , Chromosomes, Artificial, Human/genetics , Gene Knockout Techniques , Immunoglobulins/genetics , Immunoglobulins/immunology , Animals , Animals, Newborn , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Base Sequence , CHO Cells , Cattle , Cell Line , Cricetinae , Cricetulus , Female , Flow Cytometry , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Immunoglobulin lambda-Chains/genetics , Immunoglobulin lambda-Chains/immunology , Immunoglobulin mu-Chains/genetics , Immunoglobulin mu-Chains/immunology , Male , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
18.
PLoS One ; 8(10): e78119, 2013.
Article in English | MEDLINE | ID: mdl-24205120

ABSTRACT

Therapeutic human polyclonal antibodies (hpAbs) derived from pooled plasma from human donors are Food and Drug Administration approved biologics used in the treatment of a variety of human diseases. Powered by the natural diversity of immune response, hpAbs are effective in treating diseases caused by complex or quickly-evolving antigens such as viruses. We previously showed that transchromosomic (Tc) cattle carrying a human artificial chromosome (HAC) comprising the entire unrearranged human immunoglobulin heavy-chain (hIGH) and kappa-chain (hIGK) germline loci (named as κHAC) are capable of producing functional hpAbs when both of the bovine immunoglobulin mu heavy-chains, bIGHM and bIGHML1, are homozygously inactivated (double knockouts or DKO). However, B lymphocyte development in these Tc cattle is compromised, and the overall production of hpAbs is low. Here, we report the construction of an improved HAC, designated as cKSL-HACΔ, by incorporating all of the human immunoglobulin germline loci into the HAC. Furthermore, for avoiding the possible human-bovine interspecies incompatibility between the human immunoglobulin mu chain protein (hIgM) and bovine transmembrane α and ß immunoglobulins (bIgα and bIgß) in the pre-B cell receptor (pre-BCR) complex, we partially replaced (bovinized) the hIgM constant domain with the counterpart of bovine IgM (bIgM) that is involved in the interaction between bIgM and bIgα/Igß; human IgM bovinization would also improve the functionality of hIgM in supporting B cell activation and proliferation. We also report the successful production of DKO Tc cattle carrying the cKSL-HACΔ (cKSL-HACΔ/DKO), the dramatic improvement of B cell development in these cattle and the high level production of hpAbs (as measured for the human IgG isotype) in the plasma. We further demonstrate that, upon immunization by tumor immunogens, high titer tumor immunogen-specific human IgG (hIgG) can be produced from such Tc cattle.


Subject(s)
Immunoglobulins/metabolism , Animals , B-Lymphocytes/metabolism , Cattle , Cell Proliferation , Chromosomes, Artificial, Human/genetics , Chromosomes, Artificial, Human/immunology , Chromosomes, Artificial, Human/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Immunoglobulin mu-Chains/genetics , Immunoglobulin mu-Chains/immunology , Immunoglobulin mu-Chains/metabolism , Immunoglobulins/genetics , Immunoglobulins/immunology
19.
Thromb Haemost ; 103(1): 224-33, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20062929

ABSTRACT

Tissue factor (TF) antagonists targeting the factor VII (FVII) binding domain have been shown to interrupt acute vascular thrombus formation without impairing haemostasis in non-human primates. In this study, we evaluate whether a human/mouse chimeric monoclonal antibody (ALT-836, formerly known as Sunol-cH36) blocking the factor X/factor IX (FX/FIX) binding site of tissue factor could achieve similar clinical benefits in an arterial thrombosis model induced by surgical endarterectomy in chimpanzees. In this model, sequential surgical endarterectomies on right and left superficial femoral arteries were performed 30 days apart in five chimpanzees. A bolus (1 mg/kg) of ALT-836 was injected intravenously immediately preceding the restoration of flow in the endarterectomised femoral artery. Pre-surgical labelling of autologous platelets using (111)In-Oxine and post-surgical gamma camera imaging of (111)In-platelet deposition at endarterectomy sites was performed. The manipulated arterial segments were harvested for patency analysis 30 days following surgery. The results indicate that ALT-836 was highly effective at reducing acute vascular thrombosis, with no significant variations in surgical blood loss and template-bleeding time in the treated group compared to the control animals. These data suggest that ALT-836 is an effective and safe antithrombotic agent in preventing TF-initiated vascular thrombogenesis without compromising haemostasis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Factor X/metabolism , Fibrinolytic Agents/pharmacology , Thromboplastin/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity , Binding Sites , Blood Coagulation/drug effects , Blood Loss, Surgical/prevention & control , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Endarterectomy , Factor IX/metabolism , Factor VIIa/metabolism , Female , Femoral Artery/surgery , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/adverse effects , Fibrinolytic Agents/pharmacokinetics , Hemorrhage/chemically induced , Humans , Injections, Intravenous , Mice , Mice, Inbred BALB C , Pan troglodytes , Radionuclide Imaging , Recombinant Fusion Proteins/pharmacology , Thromboplastin/immunology , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/diagnostic imaging , Thrombosis/etiology
20.
Nat Biotechnol ; 27(2): 173-81, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19151699

ABSTRACT

Antigen-specific human polyclonal antibodies (hpAbs), produced by hyperimmunization, could be useful for treating many human diseases. However, yields from available transgenic mice and transchromosomic (Tc) cattle carrying human immunoglobulin loci are too low for therapeutic applications. We report a Tc bovine system that produces large yields of hpAbs. Tc cattle were generated by transferring a human artificial chromosome vector carrying the entire unrearranged, human immunoglobulin heavy (hIGH) and kappa-light (hIGK) chain loci to bovine fibroblasts in which two endogenous bovine IgH chain loci were inactivated. Plasma from the oldest animal contained >2 g/l of hIgG, paired with either human kappa-light chain (up to approximately 650 microg/ml, fully human) or with bovine kappa- or lambda-light chain (chimeric), with a normal hIgG subclass distribution. Hyperimmunization with anthrax protective antigen triggered a hIgG-mediated humoral immune response comprising a high proportion of antigen-specific hIgG. Purified, fully human and chimeric hIgGs were highly active in an in vitro toxin neutralization assay and protective in an in vivo mouse challenge assay.


Subject(s)
Animals, Genetically Modified , Immunoglobulin G/biosynthesis , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin kappa-Chains/biosynthesis , Animals , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , CHO Cells , Cattle/genetics , Chromosomes, Artificial, Human/genetics , Cricetinae , Cricetulus , Gene Knockout Techniques , Glycosylation , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Mice , Neutralization Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...