Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-jbqgn Total loading time: 0 Render date: 2024-06-25T08:11:22.665Z Has data issue: false hasContentIssue false

7 - Therapeutic Antibodies from XenoMouse Transgenic Mice

from PART III - TRANSGENIC HUMAN ANTIBODY REPERTOIRES

Published online by Cambridge University Press:  15 December 2009

Melvyn Little
Affiliation:
Affimed Therapeutics AG
Get access

Summary

For close to two decades, realization of the promise of monoclonal antibody (mAb) technology for the generation of therapeutic “magic bullets” has been challenged primarily by limited efficacy and safety related to immunogenicity of mouse antibodies in human patients. Among the technologies developed to overcome these hurdles were transgenic mice genetically engineered with a “humanized” humoral immune system. One such transgenic technology, the XenoMouse, has succeeded in recapitulating the human antibody response in mice by introducing nearly the entire human immunoglobulin (Ig) loci into the germline of mice with inactivated mouse antibody machinery. XenoMouse strains have been used to generate a large array of high-affinity, potent, fully human antibodies directed to targets in multiple disease indications, many of which are advancing in clinical development. Full validation of the technology has been achieved with the recent regulatory approval of panitumumab, a fully human antibody directed against epidermal growth factor receptor, for the treatment of advanced colorectal cancer. The successful development of panitumumab, as the first antibody derived from human antibody transgenic mice, signifies an important milestone for XenoMouse and other human antibody transgenic technologies and points to their potential contributions for future therapeutics.

RATIONALE FOR DEVELOPING HUMAN ANTIBODY-PRODUCING TRANSGENIC MICE

The discovery of hybridoma technology in 1975 for the isolation of high-specificity and high-affinity mouse monoclonal antibodies (mAbs) opened the door to a new class of therapeutics with a potential to substantially impact both therapy and diagnosis of many human diseases.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Kohler, G. & Milstein, C.. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature, 256 (1975), 495–497.CrossRefGoogle ScholarPubMed
Pendley, C., Schantz, A., & Wagner, C.. Immunogenicity of therapeutic monoclonal antibodies. Curr Opin Mol Ther, 5 (2003), 172–179.Google ScholarPubMed
Winter, G. & Milstein, C.. Man-made antibodies. Nature, 349 (1991), 293–299.CrossRefGoogle ScholarPubMed
Morrison, S.L. & Oi, V.T.. Chimeric immunoglobulin genes, in Immunoglobulin genes (London, UK: Academic Press, 1975), pp. 260–274.Google Scholar
Riechmann, L., Clark, M., Waldmann, H., & Winter, G.. Reshaping human antibodies for therapy. Nature, 332 (1988), 323–327.CrossRefGoogle ScholarPubMed
Burton, D.R. & Barbas, C.F.. Human antibodies from combinatorial libraries, in Protein engineering of antibody molecules for prophylactic and therapeutic antibodies in man (ed. Clark, M.) (Nottingham, UK: Nottingham Academic Titles, 1993), pp. 65–82.Google Scholar
Hoogenboom, H.R.. Selecting and screening recombinant antibody libraries. Nat Biotechnol, 23 (2005), 1105–1116.CrossRefGoogle ScholarPubMed
Lonberg, N.. Human antibodies from transgenic animals. Nat Biotechnol, 23 (2005), 1117–1125.CrossRefGoogle ScholarPubMed
Jakobovits, A., et al. Analysis of homozygous mutant chimeric mice: deletion of the immunoglobulin heavy-chain joining region blocks B-cell development and antibody production. Proc Natl Acad Sci USA, 90 (1993), 2551–2555.CrossRefGoogle ScholarPubMed
Green, L.L. & Jakobovits, A.. Regulation of B cell development by variable gene complexity in mice reconstituted with human immunoglobulin yeast artificial chromosomes. J Exp Med, 188 (1998), 483–495.CrossRefGoogle ScholarPubMed
Green, L.L., et al. Antigen-specific human monoclonal antibodies from mice engineered with human Ig heavy and light chain YACs. Nat Genet, 7 (1994), 13–21.CrossRefGoogle ScholarPubMed
Matsuda, F. & Honjo, T.. Organization of the human immunoglobulin heavy-chain locus. Adv Immunol, 62 (1996), 1–29.CrossRefGoogle ScholarPubMed
Cook, G.P. & Tomlinson, I.M.. The human immunoglobulin VH repertoire. Immunol Today, 16 (1995), 237–242.CrossRefGoogle ScholarPubMed
Max, E.. Immunoglobulins: molecular genetics, in Fundamental immunology (ed. Paul, W.E.), (New York: Raven Press, 1993), pp. 315–382.Google Scholar
Mendez, M.J., et al. Analysis of the structural integrity of YACs comprising human immunoglobulin genes in yeast and in embryonic stem cells. Genomics, 26 (1995), 294–307.CrossRefGoogle ScholarPubMed
Mendez, M.J., et al. Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice. Nat Genet, 15 (1997), 146–156.CrossRefGoogle ScholarPubMed
Weichhold, G.M., Ohnheiser, R., & Zachau, H.G.. The human immunoglobulin kappa locus consists of two copies that are organized in opposite polarity. Genomics, 16 (1993), 503–511.CrossRefGoogle ScholarPubMed
Jakobovits, A., et al. Germ-line transmission and expression of a human-derived yeast artificial chromosome. Nature, 362 (1993), 255–258.CrossRefGoogle ScholarPubMed
Kellermann, S.A. & Green, L.L.. Antibody discovery: the use of transgenic mice to generate human monoclonal antibodies for therapeutics. Curr Opin Biotechnol, 13 (2002), 593–597.CrossRefGoogle ScholarPubMed
Gallo, M.L., Ivanov, V.E., Jakobovits, A., & Davis, C.G.. The human immunoglobulin loci introduced into mice: V (D) and J gene segment usage similar to that of adult humans. Eur J Immunol, 30 (2000), 534–540.3.0.CO;2-P>CrossRefGoogle Scholar
Fishwild, D.M., et al. High-avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice. Nat Biotechnol, 14 (1996), 845–851.CrossRefGoogle ScholarPubMed
Lonberg, N., et al. Antigen-specific human antibodies from mice comprising four distinct genetic modifications. Nature, 368 (1994), 856–859.CrossRefGoogle ScholarPubMed
Huang, S., et al. Fully humanized neutralizing antibodies to interleukin-8 (ABX-IL8) inhibit angiogenesis, tumor growth, and metastasis of human melanoma. Am J Pathol, 161 (2002), 125–134.CrossRefGoogle ScholarPubMed
McGary, E.C., et al. A fully human antimelanoma cellular adhesion molecule/MUC18 antibody inhibits spontaneous pulmonary metastasis of osteosarcoma cells in vivo. Clin Cancer Res, 9 (2003), 6560–6566.Google ScholarPubMed
Ostendorf, T., et al. A fully human monoclonal antibody (CR002) identifies PDGF-D as a novel mediator of mesangioproliferative glomerulonephritis. J Am Soc Nephrol, 14 (2003), 2237–2247.CrossRefGoogle ScholarPubMed
Cohen, B.D., et al. Combination therapy enhances the inhibition of tumor growth with the fully human anti-type 1 insulin-like growth factor receptor monoclonal antibody CP-751,871. Clin Cancer Res, 11 (2005), 2063–2073.CrossRefGoogle ScholarPubMed
Hanson, D.C.. Preclinical in vitro characterization of anti-CTLA4 therapeutic antibody CP-675-206. Proc Am Assoc Cancer Res, 45 (2004), 877.Google Scholar
Gladue, R.P. & Bedian, V.. Identification and characterization of a human CD40 agonist antibody with efficacy against human tumors SCID mice. Proc Am Assoc Cancer Res, 47 (2006), Abstract #1355.Google Scholar
Long, L., et al. Antagonist anti-CD40 monoclonal antibody, CHIR-12.12, inhibits growth of a rituximab-resistant NHL xenograft model and achieves synergistic activity when combined with ineffective rituximab. Blood, 104 (2004), Abstract #3281.Google Scholar
Burgess, T., et al. Fully human monoclonal antibodies to hepatocyte growth factor with therapeutic potential against hepatocyte growth factor/c-Met-dependent human tumors. Cancer Res, 66 (2006), 1721–1729.CrossRefGoogle ScholarPubMed
Bekker, P.J., et al. A single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res, 19 (2004), 1059–1066.CrossRefGoogle ScholarPubMed
Jakobovits, A., et al. A fully human monoclonal antibody to prostate stem cell antigen (PSCA) for the treatment prostate pancreatic cancers. J Clin Oncol, 23(16s) (2005), Abstract #4722.CrossRefGoogle Scholar
Mahler, D.A., Huang, S., Tabrizi, M., & Bell, G.M.. Efficacy and safety of a monoclonal antibody recognizing interleukin-8 in COPD: a pilot study. Chest, 126 (2004), 926–934.CrossRefGoogle ScholarPubMed
Ribas, A., Camacho, L.H., & Lopez-Berestein, G., et al. Antitumor activity in melanoma and anti-self responses in a phase I trial with the anti-cytotocix T lymphocyte-associated antigen 4 monoclonal antibody. J. Clin Oncol, 23 (2005), 8968–8977.CrossRefGoogle Scholar
McClung, M.R., et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med, 354 (2006), 821–831.CrossRefGoogle ScholarPubMed
Body, J.J., et al. A study of the biological receptor activator of nuclear factor-kappaB ligand inhibitor, denosumab, in patients with multiple myeloma or bone metastases from breast cancer. Clin Cancer Res, 12 (2006), 1221–1228.CrossRefGoogle ScholarPubMed
Yang, X.D., et al. Eradication of established tumors by a fully human monoclonal antibody to the epidermal growth factor receptor without concomitant chemotherapy. Cancer Res, 59 (1999), 1236–1243.Google ScholarPubMed
Mendelsohn, J. & Baselga, J.. Epidermal growth factor receptor targeting in cancer. Semin Oncol, 33 (2006), 369–385.CrossRefGoogle Scholar
Yarden, Y. & Sliwkowski, M.X.. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol, 2, (2001), 127–137.CrossRefGoogle ScholarPubMed
Yang, X.D., Roskos, L.K., Davis, C.G., & Schwab, G.. From XenoMouse® technology to panitumumab, in Cancer drug discovery and development, the oncogenomics handbook (eds. LaRochelle, W.J. & Shimkets, R.A.) (Totowa, NJ:Humana Press, 2005), 647–657.Google Scholar
Jakobovits, A., Amado, R.G., & Yang, X., et al. From XenoMouse technology to panitumumab, the first fully human antibody product from transgenic mice. Nat Biotechnol, 25 (2007), 1134–1143.CrossRefGoogle ScholarPubMed
Weiner, L.M., et al. Updated results from a dose and schedule study of Panitumumab (ABX-EGF) monotherapy, in patients with advanced solid malignancies. J Clin Oncol, 23 (suppl 16) (2005), Abstract #3059.CrossRefGoogle Scholar
Rowinsky, E.K., et al. Safety, pharmacokinetics, and activity of ABX-EGF, a fully human anti-epidermal growth factor receptor monoclonal antibody in patients with metastatic renal cell cancer. J Clin Oncol, 22 (2004), 3003–3015.CrossRefGoogle ScholarPubMed
Hecht, J.R., et al. Panitumumab monotherapy in patients with previously treated metastatic colorectal cancer. Cancer, 110 (2007), 980–987.CrossRefGoogle ScholarPubMed
Cutsem, E., et al. An open-label, randomized, phase 3 clinical trial of panitumumab plus best supportive care versus best supportive care in patients with chemotherapy-refractory metastatic colorectal cancer. J Clin Oncol, 25 (2007), 1658–1664.CrossRefGoogle Scholar
Cunningham, D., et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refactory metastatic colorectal cancer. N Engl J Med, 351 (2004), 337–345.CrossRefGoogle ScholarPubMed
Burtness, B., Goldwasser, M.A., Flood, W., Mattar, B., & Forastiere, A.A.. Phase III randomized trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent head and neck cancer: an Eastern Cooperative Oncology Group study. J Clin Oncol, 23 (2005), 8646–8654.CrossRefGoogle ScholarPubMed
Schwarz, E.M. & Ritchlin, C.T.. Clinical development of anti-RANKL therapy. Arthritis Res Ther, 9 (2007), S:7.CrossRefGoogle ScholarPubMed
Bekker, P.J., Holloway, D., & Ramussen, S., et al. A single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res, 19 (2004), 1059–1066.CrossRefGoogle ScholarPubMed
Korman, A.J., Peggs, K.S., & Allison, J.P.. Checkpoint blockade in cancer immunotherapy. Adv Immunol, 90 (2006), 297–339.CrossRefGoogle ScholarPubMed
Ribas, A., Glaspy, J.A., & Lee, Y., et al. Role of dendritic cell phenotype, determinant spreading, and negative costimulatory blockade in dendritic cell-based melanoma immunotherapy. J Immunother, 27 (2004), 354–367.CrossRefGoogle ScholarPubMed
Reuben, J.M., Lee, B.N., & Li, C., et al. Biologic and immunomodulatory events after CTLA-4 blockade with ticilimumab in patients with advanced malignant melanoma. Cancer, 106 (2006), 2437–2444.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×