Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Nat Commun ; 14(1): 5249, 2023 08 28.
Article En | MEDLINE | ID: mdl-37640732

Low affinity is common for germline B cell receptors (BCR) seeding development of broadly neutralizing antibodies (bnAbs) that engage hypervariable viruses, including HIV. Antibody affinity selection is also non-homogenizing, insuring the survival of low affinity B cell clones. To explore whether this provides a natural window for expanding human B cell lineages against conserved vaccine targets, we deploy transgenic mice mimicking human antibody diversity and somatic hypermutation (SHM) and immunize with simple monomeric HIV glycoprotein envelope immunogens. We report an immunization regimen that focuses B cell memory upon the conserved CD4 binding site (CD4bs) through both conventional affinity maturation and reproducible expansion of low affinity BCR clones with public patterns in SHM. In the latter instance, SHM facilitates target acquisition by decreasing binding strength. This suggests that permissive B cell selection enables the discovery of antibody epitopes, in this case an HIV bnAb site.


AIDS Vaccines , HIV Infections , Humans , Animals , Mice , B-Lymphocytes , Memory B Cells , Receptors, Antigen, B-Cell/genetics , Broadly Neutralizing Antibodies , HIV Antigens , Mice, Transgenic , HIV Infections/prevention & control
2.
Cell Rep ; 41(6): 111628, 2022 11 08.
Article En | MEDLINE | ID: mdl-36351401

Pathogens evade host humoral responses by accumulating mutations in surface antigens. While variable, there are conserved regions that cannot mutate without compromising fitness. Antibodies targeting these conserved epitopes are often broadly protective but remain minor components of the repertoire. Rational immunogen design leverages a structural understanding of viral antigens to modulate humoral responses to favor these responses. Here, we report an epitope-enriched immunogen presenting a higher copy number of the influenza hemagglutinin (HA) receptor-binding site (RBS) epitope relative to other B cell epitopes. Immunization in a partially humanized murine model imprinted with an H1 influenza shows H1-specific serum and >99% H1-specific B cells being RBS-directed. Single B cell analyses show a genetically restricted response that structural analysis defines as RBS-directed antibodies engaging the RBS with germline-encoded contacts. These data show how epitope enrichment expands B cell responses toward conserved epitopes and advances immunogen design approaches for next-generation viral vaccines.


Influenza Vaccines , Influenza, Human , Humans , Mice , Animals , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Antibodies, Viral , Epitopes, B-Lymphocyte
3.
Immunity ; 55(9): 1693-1709.e8, 2022 09 13.
Article En | MEDLINE | ID: mdl-35952670

Human broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin stalk of group 1 influenza A viruses (IAVs) are biased for IGHV1-69 alleles that use phenylalanine (F54) but not leucine (L54) within their CDRH2 loops. Despite this, we demonstrated that both alleles encode for human IAV bnAbs that employ structurally convergent modes of contact to the same epitope. To resolve differences in lineage expandability, we compared F54 versus L54 as substrate within humanized mice, where antibodies develop with human-like CDRH3 diversity but are restricted to single VH genes. While both alleles encoded for bnAb precursors, only F54 IGHV1-69 supported elicitation of heterosubtypic serum bnAbs following immunization with a stalk-only nanoparticle vaccine. L54 IGHV1-69 was unproductive, co-encoding for anergic B cells and autoreactive stalk antibodies that were cleared from B cell memory. Moreover, human stalk antibodies also demonstrated L54-dependent autoreactivity. Therefore, IGHV1-69 polymorphism, which is skewed ethnically, gates tolerance and vaccine expandability of influenza bnAbs.


Influenza A virus , Influenza Vaccines , Influenza, Human , Alleles , Animals , Antibodies, Neutralizing , Antibodies, Viral , Broadly Neutralizing Antibodies , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza, Human/prevention & control , Mice
5.
Nat Rev Drug Discov ; 21(7): 509-528, 2022 07.
Article En | MEDLINE | ID: mdl-34937915

Cancer immunity, and the potential for cancer immunotherapy, have been topics of scientific discussion and experimentation for over a hundred years. Several successful cancer immunotherapies - such as IL-2 and interferon-α (IFNα) - have appeared over the past 30 years. However, it is only in the past decade that immunotherapy has made a broad impact on patient survival in multiple high-incidence cancer indications. The emergence of immunotherapy as a new pillar of cancer treatment (adding to surgery, radiation, chemotherapy and targeted therapies) is due to the success of immune checkpoint blockade (ICB) drugs, the first of which - ipilimumab - was approved in 2011. ICB drugs block receptors and ligands involved in pathways that attenuate T cell activation - such as cytotoxic T lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD1) and its ligand, PDL1 - and prevent, or reverse, acquired peripheral tolerance to tumour antigens. In this Review we mark the tenth anniversary of the approval of ipilimumab and discuss the foundational scientific history of ICB, together with the history of the discovery, development and elucidation of the mechanism of action of the first generation of drugs targeting the CTLA4 and PD1 pathways.


Immune Checkpoint Inhibitors , Neoplasms , CTLA-4 Antigen , Humans , Immunotherapy , Ipilimumab/pharmacology , Ipilimumab/therapeutic use , Programmed Cell Death 1 Receptor
6.
Front Immunol ; 12: 730471, 2021.
Article En | MEDLINE | ID: mdl-34566992

The ligand-binding surface of the B cell receptor (BCR) is formed by encoded and non-encoded antigen complementarity determining regions (CDRs). Genetically reproducible or 'public' antibodies can arise when the encoded CDRs play deterministic roles in antigen recognition, notably within human broadly neutralizing antibodies against HIV and influenza virus. We sought to exploit this by engineering virus-like-particle (VLP) vaccines that harbor multivalent affinity against gene-encoded moieties of the BCR antigen binding site. As proof of concept, we deployed a library of RNA bacteriophage VLPs displaying random peptides to identify a multivalent antigen that selectively triggered germline BCRs using the human VH gene IGVH1-2*02. This VLP selectively primed IGHV1-2*02 BCRs that were present within a highly diversified germline antibody repertoire within humanized mice. Our approach thus provides methodology to generate antigens that engage specific BCR configurations of interest, in the absence of structure-based information.


B-Lymphocytes/immunology , Protein Engineering , RNA Phages/immunology , Receptors, Antigen, B-Cell/immunology , Single-Domain Antibodies/immunology , Vaccines, Virus-Like Particle/immunology , Adoptive Transfer , Animals , Antibody Specificity , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/transplantation , Female , Gene Library , Humans , Ligands , Male , Mice, Transgenic , Proof of Concept Study , RNA Phages/genetics , RNA Phages/metabolism , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Vaccination , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/metabolism
7.
Cell Syst ; 11(6): 573-588.e9, 2020 12 16.
Article En | MEDLINE | ID: mdl-33031741

The antibody repertoire possesses near-limitless diversity, enabling the adaptive immune system to accommodate essentially any antigen. However, this diversity explores the antigenic space unequally, allowing some pathogens like influenza virus to impose complex immunodominance hierarchies that distract antibody responses away from key sites of virus vulnerability. We developed a computational model of affinity maturation to map the patterns of immunodominance that evolve upon immunization with natural and engineered displays of hemagglutinin (HA), the influenza vaccine antigen. Based on this knowledge, we designed immunization protocols that subvert immune distraction and focus serum antibody responses upon a functionally conserved, but immunologically recessive, target of human broadly neutralizing antibodies. We tested in silico predictions by vaccinating transgenic mice in which antibody diversity was humanized to mirror clinically relevant humoral output. Collectively, our results demonstrate that complex patterns in antibody immunogenicity can be rationally defined and then manipulated to elicit engineered immunity.


B-Lymphocytes/immunology , Broadly Neutralizing Antibodies/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Animals , Disease Models, Animal , Humans , Mice
8.
Cell Rep ; 32(8): 108065, 2020 08 25.
Article En | MEDLINE | ID: mdl-32846123

B cell receptors (BCRs) display a combination of variable (V)-gene-encoded complementarity determining regions (CDRs) and adaptive/hypervariable CDR3 loops to engage antigens. It has long been proposed that the former tune for recognition of pathogens or groups of pathogens. To experimentally evaluate this within the human antibody repertoire, we perform immune challenges in transgenic mice that bear diverse human CDR3 and light chains but are constrained to different human VH-genes. We find that, of six commonly deployed VH sequences, only those CDRs encoded by IGHV1-2∗02 enable polyclonal antibody responses against bacterial lipopolysaccharide (LPS) when introduced to the bloodstream. The LPS is from diverse strains of gram-negative bacteria, and the VH-gene-dependent responses are directed against the non-variable and universal saccrolipid substructure of this antigen. This reveals a broad-spectrum anti-LPS response in which germline-encoded CDRs naturally hardwire the human antibody repertoire for recognition of a conserved microbial target.


Immunoglobulin Variable Region/metabolism , Lipopolysaccharides/immunology , Animals , Humans , Mice , Mice, Transgenic
9.
Immunity ; 51(4): 735-749.e8, 2019 10 15.
Article En | MEDLINE | ID: mdl-31563464

Antibody paratopes are formed by hypervariable complementarity-determining regions (CDRH3s) and variable gene-encoded CDRs. The latter show biased usage in human broadly neutralizing antibodies (bnAbs) against both HIV and influenza virus, suggesting the existence of gene-endowed targeting solutions that may be amenable to pathway amplification. To test this, we generated transgenic mice with human CDRH3 diversity but simultaneously constrained to individual user-defined human immunoglobulin variable heavy-chain (VH) genes, including IGHV1-69, which shows biased usage in human bnAbs targeting the hemagglutinin stalk of group 1 influenza A viruses. Sequential immunization with a stalk-only hemagglutinin nanoparticle elicited group 1 bnAbs, but only in IGHV1-69 mice. This VH-endowed response required minimal affinity maturation, was elicited alongside pre-existing influenza immunity, and when IGHV1-69 B cells were diluted to match the frequency measured in humans. These results indicate that the human repertoire could, in principle, support germline-encoded bnAb elicitation using a single recombinant hemagglutinin immunogen.


Antibodies, Viral/metabolism , B-Lymphocytes/immunology , Broadly Neutralizing Antibodies/metabolism , Influenza A virus/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Receptors, Antigen, B-Cell/genetics , Animals , Antibodies, Viral/genetics , Antibody Affinity , Broadly Neutralizing Antibodies/genetics , Complementarity Determining Regions/genetics , Germ-Line Mutation/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunity, Humoral , Immunization, Secondary , Immunoglobulin Heavy Chains/genetics , Mice , Mice, Transgenic , Nanoparticles , Protein Engineering
10.
Cancer Immunol Res ; 5(4): 275-281, 2017 04.
Article En | MEDLINE | ID: mdl-28373215

Cancer therapeutics that target the immune system rather than the cancer cell itself are becoming standard of care in a growing number of different malignancies. Although cancer immunotherapy is not a new concept, the potential importance of this class of drugs was probably not fully appreciated as recently as a decade ago when much of the focus of cancer drug discovery was on cancer cell-targeted medicines. The authors were lucky enough to be able to witness and participate in the discovery and development of ipilimumab and nivolumab, two relatively early examples of immune system-targeted drugs. The challenges associated with discovering and developing these molecules may be of historical interest and instructive for moving cancer immunotherapy forward for greater numbers of patients. Cancer Immunol Res; 5(4); 275-81. ©2017 AACR.


Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Drug Discovery , Immunomodulation/drug effects , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Combined Modality Therapy , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/mortality , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , Treatment Outcome
11.
Cancer Immunol Res ; 4(4): 279-88, 2016 Apr.
Article En | MEDLINE | ID: mdl-27036972

With the recent FDA approvals of pembrolizumab and nivolumab, and a host of additional immunomodulatory agents entering clinical development each year, the field of cancer immunotherapy is changing rapidly. Strategies that can assist researchers in choosing the most promising drugs and drug combinations to move forward through clinical development are badly needed in order to reduce the likelihood of late-stage clinical trial failures. On October 5, 2014, the Cancer Immunotherapy Consortium of the Cancer Research Institute, a collaborative think tank composed of stakeholders from academia, industry, regulatory agencies, and patient interest groups, met to discuss strategies for de-risking immunotherapy development, with a focus on integrating preclinical and clinical studies, and conducting smarter early-phase trials, particularly for combination therapies. Several recommendations were made, including making better use of clinical data to inform preclinical research, obtaining adequate tissues for biomarker studies, and choosing appropriate clinical trial endpoints to identify promising drug candidates and combinations in nonrandomized early-phase trials.


Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Clinical Trials, Phase I as Topic , Combined Modality Therapy , Disease Models, Animal , Drug Discovery , Drug Evaluation, Preclinical , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Mice , Molecular Targeted Therapy
12.
Pharmacol Ther ; 148: 132-53, 2015 Apr.
Article En | MEDLINE | ID: mdl-25476108

The discovery and increased understanding of the complex interactions regulating the immune system have contributed to the pharmacologic activation of antitumor immunity. The activity of effector cells, such as T and NK cells, is regulated by an array of activating and attenuating receptors and ligands. Agents that target these molecules can modulate immune responses by exerting antagonistic or agonistic effects. Several T- or NK-cell modulators have entered clinical trials, and two have been approved for use. Ipilimumab (Yervoy®, Bristol-Myers Squibb) and nivolumab (OPDIVO, Ono Pharmaceutical Co., Ltd./Bristol-Myers Squibb) were approved for the treatment of metastatic melanoma, in March 2011 in the United States, and in July 2014 in Japan, respectively. The clinical activity of these two antibodies has not been limited to tumor types considered sensitive to immunotherapy, and promising activity has been reported in other solid and hematologic tumors. Clinical development of ipilimumab and nivolumab has presented unique challenges in terms of safety and efficacy, requiring the establishment of new evaluation criteria for adverse events and antitumor effects. Guidelines intended to help oncologists properly manage treatment in view of these non-traditional features have been implemented. The introduction of this new modality of cancer treatment, which is meant to integrate with or replace the current standards of care, requires additional efforts in terms of optimization of treatment administration, identification of biomarkers and application of new clinical trial designs. The availability of immune modulators with different mechanisms of action offers the opportunity to establish immunological combinations as new standards of care.


Antibodies, Monoclonal/therapeutic use , Immunologic Factors/therapeutic use , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/adverse effects , Humans , Immunologic Factors/adverse effects
13.
Ann N Y Acad Sci ; 1291: 1-13, 2013 Jul.
Article En | MEDLINE | ID: mdl-23772560

The immunotherapeutic agent ipilimumab has helped address a significant unmet need in the treatment of advanced melanoma. Ipilimumab is a fully human monoclonal antibody that targets cytotoxic T-lymphocyte antigen-4 (CTLA-4), thereby augmenting antitumor immune responses. After decades in which a number of clinical trials were conducted, ipilimumab was the first therapy to improve overall survival in a randomized, controlled phase III trial of patients with advanced melanoma. These results led to the regulatory approval of ipilimumab at 3 mg/kg for the treatment of unresectable or metastatic melanoma. More than 17,000 patients worldwide have received ipilimumab, either as a commercial drug at 3 mg/kg or in clinical trials and expanded access programs at different doses. Consistent with its proposed mechanism of action, the most common toxicities associated with ipilimumab therapy are inflammatory in nature. These immune-related adverse events were mostly reversible when effective treatment guidelines were followed. Importantly, long-term follow-up of patients who received ipilimumab in a phase III trial showed that 24% survived at least two years, and in phase II studies, a proportion of patients survived at least five years. Evaluation of ipilimumab is ongoing in the adjuvant setting for melanoma, and for advanced disease in nonsmall cell lung, small cell lung, prostate, ovarian, and gastric cancers.


Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Agents/immunology , Clinical Trials as Topic/methods , Humans , Immunotherapy/methods , Ipilimumab , Melanoma/immunology , Melanoma/mortality , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Survival Rate/trends , Treatment Outcome
16.
Curr Opin Immunol ; 20(4): 450-9, 2008 Aug.
Article En | MEDLINE | ID: mdl-18606226

Over the past two decades, technologies have emerged for generating monoclonal antibodies (MAbs) derived from human immunoglobulin gene sequences. These fully human MAbs provide an alternative to re-engineered, or de-immunized, rodent MAbs as a source of low immunogenicity therapeutic antibodies. There are now two marketed fully human therapeutic MAbs, adalimumab and panitumumab, and several dozen more in various stages of human clinical testing. Most of the drugs, including adalimumab and panitumumab, were generated using either phage display or transgenic mouse platforms. The reported clinical experience with fully human MAbs demonstrates that these two platforms are, and should continue to be, a significant source of active and well tolerated experimental therapeutics. While this body of reported clinical data does not yet provide a clear distinction between the platforms, the available descriptions of the drug discovery processes used to identify the clinical candidates highlight one difference. It appears that lead optimization is more commonly applied to phage display derived leads than transgenic mouse derived leads.


Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/drug therapy , Neoplasms/drug therapy , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, CD20/immunology , Antigens, CD20/metabolism , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Autoimmune Diseases/immunology , CD4 Antigens/immunology , CD4 Antigens/metabolism , CTLA-4 Antigen , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Epithelial Cell Adhesion Molecule , ErbB Receptors/immunology , ErbB Receptors/metabolism , Humans , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Mice , Mice, Transgenic , Neoplasms/immunology , Peptide Library , Protein Engineering , RANK Ligand/immunology , RANK Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
18.
Nat Biotechnol ; 23(9): 1117-25, 2005 Sep.
Article En | MEDLINE | ID: mdl-16151405

Laboratory mice provide a ready source of diverse, high-affinity and high-specificity monoclonal antibodies (mAbs). However, development of rodent antibodies as therapeutic agents has been impaired by the inherent immunogenicity of these molecules. One technology that has been explored to generate low immunogenicity mAbs for in vivo therapy involves the use of transgenic mice expressing repertoires of human antibody gene sequences. This technology has now been exploited by over a dozen different pharmaceutical and biotechnology companies toward developing new therapeutic mAbs, and currently at least 33 different drugs in clinical testing--including several in pivotal trials--contain variable regions encoded by human sequences from transgenic mice. The emerging data from these trials provide an early glimpse of the safety and efficacy issues for these molecules. Nevertheless, actual product approval, the biggest challenge so far, is required to fully validate this technology as a drug discovery tool. In the future, it may be possible to extend this technology beyond rodents and use transgenic farm animals to directly generate and produce human sequence polyclonal sera.


Animals, Genetically Modified , Antibodies, Monoclonal/chemistry , Antibodies/chemistry , Animals , Biotechnology/methods , Biotechnology/trends , Clinical Trials as Topic , Drug Delivery Systems , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Light Chains/chemistry , Immunoglobulins/chemistry , Mice , Mice, Transgenic
19.
J Immunol ; 171(11): 6251-9, 2003 Dec 01.
Article En | MEDLINE | ID: mdl-14634142

The immune modulatory molecule CTLA-4 (CD152), through interactions with the B7 costimulatory molecules, has been shown to be a negative regulator of T cell activation in various murine model systems. Abs that block CTLA-4 function can enhance immune responses that mediate potent antitumor activity. However, CTLA-4 blockade can also exacerbate autoimmune disease. The safety and activity of anti-CTLA-4 Abs in primates has not been addressed. To that end, we generated human Abs against CTLA-4 using transgenic mice expressing human Ig genes. A high affinity Ab (10D1) that blocked the binding of CTLA-4 to the B7-1 and B7-2 ligands and had cross-reactivity with macaque CTLA-4 was chosen for further development. Administration of 10D1 to cynomolgus macaques significantly enhanced Ab responses to hepatitis surface Ag and a human melanoma cell vaccine. Anti-self Ab responses as measured by immunoassays using lysate from melanocyte-rich tissues were elicited in those animals receiving the melanoma cell vaccine and anti-CTLA-4 Ab. Remarkably, chronic administration of 10D1 did not result in measurable polyclonal T cell activation, significant alteration of the lymphocyte subsets, or induce clinically observable autoimmunity. Repeated dosing of the 10D1 did not elicit monkey anti-human Ab responses in the monkeys. These observations support the development of CTLA-4 blockade for human immunotherapy.


Antibodies, Blocking/adverse effects , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/adverse effects , Antigens, Differentiation/immunology , Cancer Vaccines/pharmacology , Down-Regulation/immunology , Hepatitis B Vaccines/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Blocking/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/biosynthesis , Antigens, CD , CTLA-4 Antigen , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Drug Synergism , Female , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/immunology , Humans , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/immunology , Macaca fascicularis , Male , Mice , Mice, Transgenic
20.
Cloning Stem Cells ; 4(1): 91-102, 2002.
Article En | MEDLINE | ID: mdl-12006160

We have developed TransChromo (TC) technology, which enables the introduction of megabase-sized segments of DNA into cells. We have used this approach to derive mice that carry megabases of human DNA by the use of a human chromosome fragment (HCF) as a vector. TC technology has been applied to the construction of the TC Mouse,trade mark which incorporates entire human immunoglobulin (hIg) loci. TC Mouse expresses a fully diverse repertoire of hIgs, including all the subclasses of IgGs (IgG1-G4). Immunization of the TC Mouse with various human antigens produced antibody responses comprised of human antibodies. Furthermore, it was possible to obtain hybridoma clones expressing fully human antibodies specific for the target human antigen. However, because of the instability of the Igkappa locus-bearing HCF2, the efficiency of hybridoma production was less than one-tenth of that observed in normal mice. An instant solution to this problem was to cross-breed the Kirin TC Mouse carrying the HCF14, which was stable in mouse cells, with the Medarex YAC-transgenic mouse carrying about 50% of the hIgVkappa gene segments as a region that is stably integrated into the mouse genome. The resulting mouse, dubbed the KM Mouse, performed as well as normal mice with regard to immune responsiveness and efficiency of hybridoma production. Another application of TC technology is the production of polyclonal antibodies in large animals such as chickens and cows. To test the efficacy of human polyclonal antibodies derived from TC animals, feasibility studies were performed using antisera and purified gamma-globulin from TC mice immunized with Pseudomonas aeruginosa, methicillin-resistant Staphylococcus aureus (MRSA), or Japanese encephalitis virus (JEV). The TC mouse-derived antisera and gamma-globulin showed a much higher titer and efficacy in terms of the neutralizing activity of the pathogens in vitro and in vivo than either human serum or gamma-globulin prepared from human blood.


Animals, Genetically Modified , Antibodies, Monoclonal/biosynthesis , Immunoglobulins/metabolism , Animals , Genetic Engineering , Humans , Mice , Mice, Knockout , Mice, Transgenic , Models, Genetic
...