1
|
Al-Taie A, Özcan Bülbül E. A paradigm use of monoclonal antibodies-conjugated nanoparticles in breast cancer treatment: current status and potential approaches. J Drug Target 2024; 32:45-56. [PMID: 38096045 DOI: 10.1080/1061186x.2023.2295803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/12/2023] [Indexed: 12/30/2023]
Abstract
Monoclonal antibodies (mAbs) are integral to cancer treatment over conventional non-specific therapy methods. This study provides a scoping review of the clinically approved mAbs, focusing on the current application of different nanocarrier technologies as drug delivery targets for mAb-conjugated nanoparticles (NPs) as potential features for breast cancer (BC) treatment. An extensive literature search was conducted between the years 2000 and 2023 using various sources of databases. The first part covered mAb classification, types, and mechanisms of action, pharmacokinetics and clinical applications in BC. The second part covered polymeric, lipid and inorganic-based NPs, which are a variety of mAb-conjugated NPs targeting BC. A total of 20 relevant studies were enrolled indicating there are three different types of nanoparticular systems (polymeric NPs, inorganic NPs and lipid-based NPs) that can be used for BC treatment by being loaded with various active substances and conjugated with these antibodies. While mAbs have altered the way in cancer treatment due to targeting cancer cells specifically, the delivery of mAbs with nanoparticulate systems is important in the treatment of BC, as NPs are still being investigated as distinctive and promising drug delivery methods that can be employed for effective treatment of BC.
Collapse
Affiliation(s)
- Anmar Al-Taie
- Clinical Pharmacy Department, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| | - Ece Özcan Bülbül
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| |
Collapse
|
2
|
Rodriguez SMB, Tataranu LG, Kamel A, Turliuc S, Rizea RE, Dricu A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int J Mol Sci 2024; 25:10765. [PMID: 39409094 PMCID: PMC11477435 DOI: 10.3390/ijms251910765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma is known to be one of the most aggressive and fatal human cancers, with a poor prognosis and resistance to standard treatments. In the last few years, many solid tumor treatments have been revolutionized with the help of immunotherapy. However, this type of treatment has failed to improve the results in glioblastoma patients. Effective immunotherapeutic strategies may be developed after understanding how glioblastoma achieves tumor-mediated immune suppression in both local and systemic landscapes. Biomarkers may help identify patients most likely to benefit from this type of treatment. In this review, we discuss the use of immunotherapy in glioblastoma, with an emphasis on immune checkpoint inhibitors and the factors that influence clinical response. A Pubmed data search was performed for all existing information regarding immune checkpoint inhibitors used for the treatment of glioblastoma. All data evaluating the ongoing clinical trials involving the use of ICIs either as monotherapy or in combination with other drugs was compiled and analyzed.
Collapse
Affiliation(s)
- Silvia Mara Baez Rodriguez
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Amira Kamel
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy “G. T. Popa”, 700115 Iasi, Romania;
| | - Radu Eugen Rizea
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania;
| |
Collapse
|
3
|
Rice SL, Muñoz FG, Benjamin J, Alnablsi MW, Pillai A, Osborne JR, Beets-Tan R. Transcatheter pseudo-vascular isolation for localization and concentration of a large molecule theranostic probe into a transgenic OncoPIG kidney tumor. Nucl Med Biol 2024; 136-137:108939. [PMID: 39003976 DOI: 10.1016/j.nucmedbio.2024.108939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/09/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Great strides have been made identifying molecular and genetic changes expressed by various tumor types. These molecular and genetic changes are used as pharmacologic targets for precision treatment using large molecule (LM) proteins with high specificity. Theranostics exploits these LM biomolecules via radiochemistry, creating sensitive diagnostic and therapeutic agents. Intravenous (i.v.) LM drugs have an extended biopharmaceutical half-life thus resulting in an insufficient therapeutic index, permitting only palliative brachytherapy due to unacceptably high rates of systemic nontarget radiation doses to normal tissue. We employ tumor arteriole embolization isolating a tumor from the systemic circulation, and local intra-arterial (i.a.) infusion to improve uptake of a LM drug within a porcine renal tumor (RT). METHODS In an oncopig RT we assess the in vivo biodistribution of 99mTc-labeled macroaggregated albumin (MAA) a surrogate for a LM theranostics agent in the RT, kidney, liver, spleen, muscle, blood, and urine. Control animals underwent i.v. infusion and experimental group undergoing arteriography with pseudovascular isolation (PVI) followed by direct i.a. injection. RESULTS Injected dose per gram (%ID/g) of the LM at 1 min was 86.75 ± 3.76 and remained elevated up to 120 min (89.35 ± 5.77) with i.a. PVI, this increase was statistically significant (SS) compared to i.v. (13.38 ± 1.56 and 12.02 ± 1.05; p = 0.0003 p = 0.0006 at 1 and 120 min respectively). The circulating distribution of LM in the blood was less with i.a. vs i.v. infusion (2.28 ± 0.31 vs 25.17 ± 1.84 for i.v. p = 0.033 at 1 min). Other organs displayed a trend towards less exposure to radiation for i.a. with PVI compared to i.v. which was not SS. CONCLUSION PVI followed by i.a. infusion of a LM drug has the potential to significantly increase the first pass uptake within a tumor. This minimally invasive technique can be translated into clinical practice, potentially rendering monoclonal antibody based radioimmunotherapy a viable treatment for renal tumors.
Collapse
Affiliation(s)
- Samuel L Rice
- Netherlands Cancer Institute-Antoni van Leeuwenhoekziekenhuis, Department of Radiology, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands; UT Southwestern Medical Center, Department of Radiology, Interventional Radiology Section, 5959 Harry Hines Blvd., Dallas, TX 75390-9061, Professional Office Building I (HP6.600) Mail Code 8834, United States of America.
| | - Fernando Gómez Muñoz
- Netherlands Cancer Institute-Antoni van Leeuwenhoekziekenhuis, Department of Radiology, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Jamaal Benjamin
- UT Southwestern Medical Center, Department of Radiology, Interventional Radiology Section, 5959 Harry Hines Blvd., Dallas, TX 75390-9061, Professional Office Building I (HP6.600) Mail Code 8834, United States of America
| | - Mhd Wisam Alnablsi
- UT Southwestern Medical Center, Department of Radiology, Interventional Radiology Section, 5959 Harry Hines Blvd., Dallas, TX 75390-9061, Professional Office Building I (HP6.600) Mail Code 8834, United States of America
| | - Anil Pillai
- UT Southwestern Medical Center, Department of Radiology, Interventional Radiology Section, 5959 Harry Hines Blvd., Dallas, TX 75390-9061, Professional Office Building I (HP6.600) Mail Code 8834, United States of America
| | - Joseph R Osborne
- New York-Presbyterian Weill Cornell Medical Center, Department of Radiology, 1305 York Avenue 3rd Floor, New York, NY 10021, United States of America
| | - Regina Beets-Tan
- Netherlands Cancer Institute-Antoni van Leeuwenhoekziekenhuis, Department of Radiology, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| |
Collapse
|
4
|
Al-Taie A, Sheta N. Clinically Approved Monoclonal Antibodies-based Immunotherapy: Association With Glycemic Control and Impact Role of Clinical Pharmacist for Cancer Patient Care. Clin Ther 2024; 46:e29-e44. [PMID: 37932154 DOI: 10.1016/j.clinthera.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/17/2023] [Accepted: 10/13/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Compared with more conventional, nonspecific therapy options, such as radiotherapy and chemotherapy, monoclonal antibodies (mAbs) constitute a crucial approach of cancer treatment. Multiple autoimmune diseases have been observed during treatment with mAb medications, including autoimmune diabetes mellitus (DM). This study provides a narrative review of clinically approved mAbs in cancer treatment and focuses on the development of hyperglycemia and DM arising from using these therapies. Furthermore, it highlights the critical role of oncology clinical pharmacists in the management of autoimmune DM and patient care while using these medications in an oncology setting. METHODS An extensive literature search was conducted using various sources of electronic databases, such as Scopus, Embase, Web of Science, and PubMed, and search engines, such as Google Scholar, for studies on mAb classification, types, mechanisms of action, pharmacokinetic properties, current clinical applications, and the associated common adverse effects with significant recommendations for patient care in an oncology setting, along with focusing on the proposed mechanisms and clinical studies that reported the association of DM after the use of these therapies. FINDINGS There are 4 types (murine, chimeric, humanized, and human) and 3 classes (unconjugated, conjugated, and bispecific) of mAbs with several mechanisms of action that can destroy cancer cells, including preventing tumor cell survival cascades, inhibiting tumor growth by interfering with tumor angiogenesis, evading programmed cell death, and bypassing immune checkpoints. However, multiple endocrinopathies, autoimmune diseases, and complications were reported from the use of these medications, including the development of autoimmune DM and diabetic ketoacidosis. These autoimmune disorders were reported most with the use of immune checkpoint inhibitors, including inhibitors of the programmed cell death protein 1 (nivolumab and pembrolizumab), its ligand (atezolizumab, avelumab, and durvalumab), and cytotoxic T-lymphocyte-associated protein 4 (ipilimumab). IMPLICATIONS mAbs are considered important approaches for the treatment of many cancer types. However, a high incidence of hyperglycemia, type 1 DM, and diabetic ketoacidosis is observed with the use of these medications, particularly immune checkpoint inhibitors. It is important for oncologic clinical pharmacists to be involved in addressing these autoimmune disorders from the use of these immunotherapies via the provision of patient education, medication adherence support, close monitoring, and follow-up, which will lead to better health-related outcomes and improved patient quality of life.
Collapse
Affiliation(s)
- Anmar Al-Taie
- Clinical Pharmacy Department, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye.
| | - Najat Sheta
- Clinical Pharmacy Department, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| |
Collapse
|
5
|
Ramapriyan R, Sun J, Curry A, Richardson LG, Ramesh T, Gaffey MA, Gedeon PC, Gerstner ER, Curry WT, Choi BD. The Role of Antibody-Based Therapies in Neuro-Oncology. Antibodies (Basel) 2023; 12:74. [PMID: 37987252 PMCID: PMC10660525 DOI: 10.3390/antib12040074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/16/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
This review explores the evolving landscape of antibody-based therapies in neuro-oncology, in particular, immune checkpoint inhibitors and immunomodulatory antibodies. We discuss their mechanisms of action, blood-brain barrier (BBB) penetration, and experience in neuro-oncological conditions. Evidence from recent trials indicates that while these therapies can modulate the tumor immune microenvironment, their clinical benefits remain uncertain, largely due to challenges with BBB penetration and tumor-derived immunosuppression. This review also examines emerging targets such as TIGIT and LAG3, the potential of antibodies in modulating the myeloid compartment, and tumor-specific targets for monoclonal antibody therapy. We further delve into advanced strategies such as antibody-drug conjugates and bispecific T cell engagers. Lastly, we explore innovative techniques being investigated to enhance antibody delivery, including CAR T cell therapy. Despite current limitations, these therapies hold significant therapeutic potential for neuro-oncology. Future research should focus on optimizing antibody delivery to the CNS, identifying novel biological targets, and discovering combination therapies to address the hostile tumor microenvironment.
Collapse
Affiliation(s)
- Rishab Ramapriyan
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| | - Jing Sun
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Annabel Curry
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Leland G. Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Tarun Ramesh
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| | - Matthew A. Gaffey
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Patrick C. Gedeon
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Elizabeth R. Gerstner
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - William T. Curry
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| | - Bryan D. Choi
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| |
Collapse
|
6
|
Zahavi D, Weiner L. Monoclonal Antibodies in Cancer Therapy. Antibodies (Basel) 2020; 9:E34. [PMID: 32698317 PMCID: PMC7551545 DOI: 10.3390/antib9030034] [Citation(s) in RCA: 307] [Impact Index Per Article: 76.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Accepted: 07/04/2020] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibody-based immunotherapy is now considered to be a main component of cancer therapy, alongside surgery, radiation, and chemotherapy. Monoclonal antibodies possess a diverse set of clinically relevant mechanisms of action. In addition, antibodies can directly target tumor cells while simultaneously promoting the induction of long-lasting anti-tumor immune responses. The multifaceted properties of antibodies as a therapeutic platform have led to the development of new cancer treatment strategies that will have major impacts on cancer care. This review focuses on the known mechanisms of action, current clinical applications for the treatment of cancer, and mechanisms of resistance of monoclonal antibody therapy. We further discuss how monoclonal antibody-based strategies have moved towards enhancing anti-tumor immune responses by targeting immune cells instead of tumor antigens as well as some of the current combination therapies.
Collapse
Affiliation(s)
- David Zahavi
- Tumor Biology Training Program, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, USA;
| | - Louis Weiner
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, USA
| |
Collapse
|
7
|
Rayahin JE, Buhrman JS, Gemeinhart RA. Melittin-glutathione S-transferase fusion protein exhibits anti-inflammatory properties and minimal toxicity. Eur J Pharm Sci 2014; 65:112-21. [PMID: 25240321 DOI: 10.1016/j.ejps.2014.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 09/09/2014] [Accepted: 09/10/2014] [Indexed: 12/11/2022]
Abstract
Although potent, proteins often require chemical modification for therapeutic use. Immunogenicity, difficult synthesis, and scale-up of these modifications are all engineering obstacles that stand in the way of expanding the use of these therapeutics. Melittin, a peptide derived from bee venom, has been shown to modulate inflammation. Although potentially therapeutic, the native peptide causes cell lysis and toxicity significantly hindering therapeutic application. Based upon the knowledge of the pore formation mechanism, we examined the toxicity and therapeutic effect of a melittin fusion protein with glutathione-S-transferase. The fusion of melittin and glutathione S-transferase results in diminished toxicity of the peptide and retained anti-inflammatory properties at doses that exceed toxic concentration of native melittin. Our results suggest that fusion proteins, particularly those of glutathione-S-transferase, may be facile modifications to control protein activity.
Collapse
Affiliation(s)
- Jamie E Rayahin
- Department of Biopharmaceutical Sciences, University of Illinois, Chicago, IL 60612-7231, USA
| | - Jason S Buhrman
- Department of Biopharmaceutical Sciences, University of Illinois, Chicago, IL 60612-7231, USA
| | - Richard A Gemeinhart
- Department of Biopharmaceutical Sciences, University of Illinois, Chicago, IL 60612-7231, USA; Department of Bioengineering, University of Illinois, Chicago, IL 60607-7052, USA; Department of Ophthalmology and Visual Sciences, University of Illinois, Chicago, IL 60612-4319, USA.
| |
Collapse
|
8
|
Abstract
Over a hundred years has passed since the discovery of the "magic bullet" serum therapy by Kitasato and Behring, the first ever therapeutic use of antibodies. More than 80 years later, the investigation of immunoglobulin structure and function and the development of cell and molecular biology introduced the production of monoclonal antibodies (MoAbs). In the 35 years since the first process for creating MoAbs was introduced, they have remained the centerpiece of the growing biotechnology and pharmaceutical industry. Herein, I review the history, development, and clinical settings of therapeutic MoAbs that have had a significant impact on life-saving medicine.
Collapse
|
9
|
Shin SU, Cho HM, Merchan J, Zhang J, Kovacs K, Jing Y, Ramakrishnan S, Rosenblatt JD. Targeted delivery of an antibody-mutant human endostatin fusion protein results in enhanced antitumor efficacy. Mol Cancer Ther 2011; 10:603-14. [PMID: 21393427 DOI: 10.1158/1535-7163.mct-10-0804] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The antiangiogenic protein endostatin showed considerable preclinical antitumor activity, but limited efficacy in phase I/II trials. Prior studies using an anti-HER2 antibody-murine endostatin fusion showed enhanced antitumor activity compared to anti-HER2 antibody or endostatin given alone, or in combination. We have generated two anti-HER2 human endostatin fusion proteins by fusing either wild-type or a mutant human endostatin (huEndo-P125A) to the 3' end of a humanized anti-HER2 IgG3 antibody. Antitumor efficacy was examined in murine and human breast tumor models. HuEndo-P125A antibody fusion protein (αHER2-huEndo-P125A) inhibited VEGF and bFGF induced endothelial cell proliferation, and tube formation in vitro, more efficiently than endostatin alone, wild-type endostatin fusion protein (αHER2-huEndo), or parental anti-HER2 antibody (αHER2 IgG3). Wild-type and mutant human endostatin was rapidly cleared from serum in mice (T½(2) = 2.0-2.1 hours), whereas αHER2-huEndo fusion proteins had a significantly prolonged half-life (T½(2) = 40.7-57.5 hours). Treatment of SK-BR-3 breast cancer xenografts with anti-HER2 IgG3-huEndo-P125A fusion resulted in greater inhibition of tumor growth and improved survival, compared to treatment with either αHER2 IgG3 (P = 0.025), human endostatin (P = 0.034), or anti-HER2 IgG3-huEndo (P = 0.016). αHER2-huEndo-P125A specifically inhibited tumors expressing HER2 in mice simultaneously implanted with murine mammary tumor EMT6 cells and with EMT6 engineered to express HER2 antigen (EMT6-HER2). Targeting of endostatin using antibody fusion proteins could improve antitumor activity of either anti-HER2 antibody and/or endostatin and provides a versatile approach that could be applied to other tumor targets with alternative antibody specificities.
Collapse
Affiliation(s)
- Seung-Uon Shin
- Department of Medicine, University of Miami School of Medicine and Sylvester Comprehensive Cancer Center, 1475 N.W. 12th Avenue (D8-4) Miami, FL 33136, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Cho HM, Rosenblatt JD, Tolba K, Shin SJ, Shin DS, Calfa C, Zhang Y, Shin SU. Delivery of NKG2D ligand using an anti-HER2 antibody-NKG2D ligand fusion protein results in an enhanced innate and adaptive antitumor response. Cancer Res 2011; 70:10121-30. [PMID: 21159634 DOI: 10.1158/0008-5472.can-10-1047] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
NKG2D ligands link the innate and adapative immune response by activating the receptors expressed on effector cells of both the innate (NK) and adaptive immune systems (CD8(+) T cells). In this study, we explored the potential therapeutic utility of this intersection by fusing the murine NKG2D ligand Rae-1β to the 3' end of an anti-HER2 IgG3 antibody containing an intact Fc domain (anti-HER2 IgG3-Rae-1β), thereby targeting an NK cell activation signal to HER2+ breast tumor cells. The antitumor efficacy of this anti-HER2-Rae-1β fusion protein was examined in a mouse mammary tumor model engineered to express HER2 (EMT6-HER2 cells). We observed an enhanced cytotoxic response of NK effectors against EMT-HER2 cells in vitro. Mice implanted on one flank with EMT6-HER2 cells and contralaterally with control EMT6 cells exhibited rapid regression of EMT6-HER2 tumors but delayed regression of contralateral EMT6 tumors. IFNγ was implicated, given a lack of antitumor efficacy in IFNγ(-/-) mice. Depletion of either NK cells or CD8(+) T cells abrogated tumor growth inhibition, suggesting essential roles for each in the observed antitumor activity. Mice rejecting EMT6-HER2 tumors after anti-HER2-Rae-1β treatment showed markedly decreased tumor growth when rechallenged with EMT6-HER2 or EMT6 cells, whereas both EMT6 and EMT6-HER2 cells grew in control mice, indicating the development of an adaptive memory response. Our findings demonstrate that administration of an antibody-NKG2D ligand fusion protein can enhance innate and adaptive immune antitumor responses, also evoking additional nontargeted antigens to enhance the potential clinical utility of this approach.
Collapse
Affiliation(s)
- Hyun-Mi Cho
- Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida 33136, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Development of human-murine chimeric immunoglobulin G for use in the serological detection of human flavivirus and alphavirus antibodies. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1617-23. [PMID: 20739503 DOI: 10.1128/cvi.00097-10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Diagnosis of human arboviral infections relies heavily on serological techniques such as the immunoglobulin M (IgM) antibody capture enzyme-linked immunosorbent assay (MAC-ELISA) and the indirect IgG ELISA. Broad application of these assays is hindered by the lack of standardized positive human control sera that react with a wide variety of flaviviruses (e.g., dengue, West Nile, yellow fever, Japanese encephalitis, Saint Louis encephalitis, and Powassan viruses), or alphaviruses (e.g., Eastern equine encephalitis, Western equine encephalitis, Venezuelan equine encephalitis, and chikungunya viruses) that can cause human disease. We have created human-murine chimeric monoclonal antibodies (cMAbs) by combining the variable regions of flavivirus (6B6C-1) or alphavirus (1A4B-6) broadly cross-reactive murine MAbs (mMAbs) with the constant region of human IgG1. These cMAbs may be used as standardized reagents capable of replacing human infection-immune-positive control sera in indirect IgG ELISA for diagnosis of all human flaviviral or alphaviral infections. The IgG cMAbs secreted from plasmid-transformed Sp2/0-Ag14 cells had serological activity identical to that of the parent mMAbs, as measured by ELISA using multiple flaviviruses or alphaviruses.
Collapse
|
12
|
Development of a human-murine chimeric immunoglobulin M antibody for use in the serological detection of human flavivirus antibodies. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:679-85. [PMID: 19297614 DOI: 10.1128/cvi.00354-08] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Current diagnosis of human flaviviral infections relies heavily on serological techniques such as the immunoglobulin M (IgM) antibody capture enzyme-linked immunosorbent assay (MAC-ELISA). Broad application of this assay is hindered by a lack of standardized human positive-control sera that react with the wide variety of flaviviruses that can cause human disease, e.g., dengue virus (DENV), West Nile virus (WNV), yellow fever virus (YFV), Japanese encephalitis virus (JEV), and St. Louis encephalitis virus (SLEV). We have created a human-murine chimeric antibody combining the variable regions of the broadly flavivirus cross-reactive murine monoclonal antibody (MAb) 6B6C-1 and the constant region of human IgM to produce a standardized reagent capable of replacing human positive-control sera in a MAC-ELISA for the diagnosis of all human flaviviral infections. The human-murine chimeric IgM antibody secreted from plasmid-transformed Sp2/0-Ag14 cells had a level of serological activity identical to that of 6B6C-1 as measured by ELISA, immunoblotting, and MAC-ELISA for multiple members of the flavivirus genus, including WNV, SLEV, YFV, DENV, and JEV.
Collapse
|
13
|
Zappe H, Snell ME, Bossard MJ. PEGylation of cyanovirin-N, an entry inhibitor of HIV. Adv Drug Deliv Rev 2008; 60:79-87. [PMID: 17884238 DOI: 10.1016/j.addr.2007.05.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2007] [Accepted: 05/05/2007] [Indexed: 02/06/2023]
Abstract
Cyanovirin-N (CV-N) is a potent inhibitor of human immunodeficiency virus and many other viruses. It has a high potential for use as a systemic compound to control viral load or in the development of microbicides to prevent primary viral infection. Due to its cyanobacterial origin it is likely to show the typical drawbacks associated with pharmaceutical use of foreign proteins such as short plasma half-life, proteolysis and immunogenicity. Several strategies were used to covalently bond poly(ethylene glycol) (PEGylate) to CV-N. Random PEGylation at lysine residues resulted in poor retention of antiviral activity. Many site-directed mutants were made to test site-specific PEGylation. One mutant, where glutamine 62 was replaced with cysteine (CV-N(Q62C)) and PEGylated with maleimide activated PEG, retained significant anti-HIV activity in vitro.
Collapse
Affiliation(s)
- H Zappe
- Nektar Therapeutics AL Corporation, 490 Discovery Drive, Huntsville, AL 35806, USA.
| | | | | |
Collapse
|
14
|
Cho HM, Rosenblatt JD, Kang YS, Iruela-Arispe ML, Morrison SL, Penichet ML, Kwon YG, Kim TW, Webster KA, Nechustan H, Shin SU. Enhanced inhibition of murine tumor and human breast tumor xenografts using targeted delivery of an antibody-endostatin fusion protein. Mol Cancer Ther 2005; 4:956-67. [PMID: 15956253 DOI: 10.1158/1535-7163.mct-04-0321] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endostatin can inhibit angiogenesis and tumor growth in mice. A potential limitation of endostatin as an antitumor agent in humans is the short serum half-life of the protein that may decrease effective concentration at the site of tumor and necessitate frequent dosing. In an effort to improve antitumor activity, endostatin was fused to an antibody specific for the tumor-selective HER2 antigen to create an antibody-endostatin fusion protein (anti-HER2 IgG3-endostatin). Normal endostatin rapidly cleared from serum in mice (T(1/2)(2), = 0.6-3.8 hours), whereas anti-HER2 IgG3-endostatin had a prolonged half-life (90% intact; T(1/2)(2), 40.2-44.0 hours). Antigen-specific targeting of anti-HER2 IgG3-endostatin was evaluated in BALB/c mice implanted with CT26 tumors or CT26 tumors engineered to express the HER2 antigen (CT26-HER2). Radio-iodinated anti-HER2 IgG3-endostatin preferentially localized to CT26-HER2 tumors relative to CT26 tumors. Administration of anti-HER2 IgG3-endostatin to mice showed preferential inhibition of CT26-HER2 tumor growth compared with CT26. Anti-HER2 IgG3-endostatin also markedly inhibited the growth of human breast cancer SK-BR-3 xenografts in severe combined immunodeficient mice. Anti-HER2 IgG3-endostatin inhibited tumor growth significantly more effectively than endostatin, anti-HER2 IgG3 antibody, or the combination of antibody and endostatin. CT26-HER2 tumors treated with the endostatin fusion protein had decreased blood vessel density and branching compared with untreated CT26-HER2 or CT26 treated with the fusion protein. The enhanced effectiveness of anti-HER2 IgG3-endostatin may be due to a longer half-life, improved serum stability, and selective targeting of endostatin to tumors, resulting in decreased angiogenesis. Linking of an antiangiogenic protein, such as endostatin, to a targeting antibody represents a promising and versatile approach to antitumor therapy.
Collapse
Affiliation(s)
- Hyun-Mi Cho
- Department of Medicine, Hematology-Oncology, University of Miami School of Medicine and Sylvester Comprehensive Cancer Center, FL 33136, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Maeda F, Takekoshi M, Nagatsuka Y, Aotsuka S, Tsukahara M, Ohshima A, Kido I, Ono Y, Ihara S. Production and characterization of recombinant human anti-HBs Fab antibodies. J Virol Methods 2005; 127:141-7. [PMID: 15896855 DOI: 10.1016/j.jviromet.2005.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Revised: 03/18/2005] [Accepted: 03/22/2005] [Indexed: 11/29/2022]
Abstract
Recombinant human Fab antibodies were generated with different reactivities against the hepatitis B virus surface (HBs) antigen. To isolate the antibodies, a method was used that combined transformation of human B cells by Epstein-Barr virus (EBV) infection with a primer-vector system developed for isolating DNA fragments of human Ig Fab portions. With this method, monoclonal and oligoclonal cell lines producing anti-HBs antibodies were established and three anti-HBs Fab antibodies were isolated from two of these cell lines. From analysis of affinity characteristics, immunohistochemical activity, and cytolysis activity, these three Fab antibodies were classified into three different groups. The first group had high affinity for HBs, the second had the ability to kill HBV-infected cells, and the third was applicable to immunohistochemical staining with HBV-infected cells. The combined effect of these antibodies was also investigated by complement-dependent cytotoxicity assay.
Collapse
Affiliation(s)
- F Maeda
- Department of Molecular Life Science, Tokai University School of Medicine, Bohseidai, Isehara 259-1193, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Helguera G, Dela Cruz JS, Lowe C, Ng PP, Trinh R, Morrison SL, Penichet ML. Vaccination with novel combinations of anti-HER2/neu cytokines fusion proteins and soluble protein antigen elicits a protective immune response against HER2/neu expressing tumors. Vaccine 2005; 24:304-16. [PMID: 16125282 DOI: 10.1016/j.vaccine.2005.07.073] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Accepted: 07/28/2005] [Indexed: 10/25/2022]
Abstract
We have previously demonstrated that anti-HER2/neu IgG3-(IL-2), (IL-12)-IgG3, or IgG3-(GM-CSF) antibody fusion proteins (mono-AbFPs) elicit anti-tumor activity against murine tumors expressing HER2/neu when used as adjuvants of extracellular domain of HER2/neu (ECD(HER2)) protein vaccination. We have now studied the effect of combinations of IL-2 and IL-12 or IL-12 and GM-CSF mono-AbFPs during vaccination with ECD(HER2). In addition, we developed two novel anti-HER2/neu IgG3-cytokine fusion proteins in which IL-2 and IL-12 or IL-12 and GM-CSF were fused to the same IgG3 molecule (bi-AbFPs). (IL-12)-IgG3-(IL-2) and (IL-12)-IgG3-(GM-CSF) were properly assembled and retained both cytokine activity and the ability to bind antigen. Vaccination of mice with ECD(HER2) and a combination of cytokines as either bi-AbFPs or two mono-AbFPs activated both Thl and Th2 immune responses and resulted in significant protection against challenge with a HER2/neu expressing tumor. Our results suggest that this approach will be effective in the prevention and/or treatment of HER2/neu expressing tumors.
Collapse
Affiliation(s)
- Gustavo Helguera
- Department of Microbiology, Immunology and Molecular Genetics and the Molecular Biology Institute, University of California, 405 Hilgard Avenue, Box 148906, Los Angeles, CA 90095-1489, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Tiroli AO, Tasic L, Oliveira CLP, Bloch C, Torriani I, Farah CS, Ramos CHI. Mapping contacts between regulatory domains of skeletal muscle TnC and TnI by analyses of single-chain chimeras. FEBS J 2005; 272:779-90. [PMID: 15670158 DOI: 10.1111/j.1742-4658.2004.04515.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The troponin (Tn) complex is formed by TnC, TnI and TnT and is responsible for the calcium-dependent inhibition of muscle contraction. TnC and TnI interact in an antiparallel fashion in which the N domain of TnC binds in a calcium-dependent manner to the C domain of TnI, releasing the inhibitory effect of the latter on the actomyosin interaction. While the crystal structure of the core cardiac muscle troponin complex has been determined, very little high resolution information is available regarding the skeletal muscle TnI-TnC complex. With the aim of obtaining structural information regarding specific contacts between skeletal muscle TnC and TnI regulatory domains, we have constructed two recombinant chimeric proteins composed of the residues 1-91 of TnC linked to residues 98-182 or 98-147 of TnI. The polypeptides were capable of binding to the thin filament in a calcium-dependent manner and to regulate the ATPase reaction of actomyosin. Small angle X-ray scattering results showed that these chimeras fold into compact structures in which the inhibitory plus the C domain of TnI, with the exception of residues 148-182, were in close contact with the N-terminal domain of TnC. CD and fluorescence analysis were consistent with the view that the last residues of TnI (148-182) are not well folded in the complex. MS analysis of fragments produced by limited trypsinolysis showed that the whole TnC N domain was resistant to proteolysis, both in the presence and in the absence of calcium. On the other hand the TnI inhibitory and C-terminal domains were completely digested by trypsin in the absence of calcium while the addition of calcium results in the protection of only residues 114-137.
Collapse
Affiliation(s)
- Ana O Tiroli
- Centro de Biologia Molecular Estrutural, Laboratório Nacional de Luz Síncrotron, Brazil
| | | | | | | | | | | | | |
Collapse
|
18
|
Asai T, Trinh R, Ng PP, Penichet ML, Wims LA, Morrison SL. A human biotin acceptor domain allows site-specific conjugation of an enzyme to an antibody-avidin fusion protein for targeted drug delivery. ACTA ACUST UNITED AC 2004; 21:145-55. [PMID: 15748688 DOI: 10.1016/j.bioeng.2004.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Revised: 10/21/2004] [Accepted: 10/22/2004] [Indexed: 11/19/2022]
Abstract
We have previously constructed an antibody-avidin (Av) fusion protein, anti-transferrin receptor (TfR) IgG3-Av, which can deliver biotinylated molecules to cells expressing the TfR. We now describe the use of the fusion protein for antibody-directed enzyme prodrug therapy (ADEPT). The 67 amino acid carboxyl-terminal domain (P67) of human propionyl-CoA carboxylase alpha subunit can be metabolically biotinylated at a fixed lysine residue. We genetically fused P67 to the carboxyl terminus of the yeast enzyme FCU1, a derivative of cytosine deaminase that can convert the non-toxic prodrug 5-fluorocytosine to the cytotoxic agent 5-fluorouracil. When produced in Escherichia coli cells overexpressing a biotin protein ligase, the FCU1-P67 fusion protein was efficiently mono-biotinylated. In the presence of 5-fluorocytosine, the biotinylated fusion protein conjugated to anti-rat TfR IgG3-Av efficiently killed rat Y3-Ag1.2.3 myeloma cells in vitro, while the same protein conjugated to an irrelevant (anti-dansyl) antibody fused to Av showed no cytotoxic effect. Efficient tumor cell killing was also observed when E. coli purine nucleoside phosphorylase was similarly targeted to the tumor cells in the presence of the prodrug 2-fluoro-2'-deoxyadenosine. These results suggest that when combined with P67-based biotinylation, anti-TfR IgG3-Av could serve as a universal delivery vector for targeted chemotherapy of cancer.
Collapse
Affiliation(s)
- Tsuneaki Asai
- Department of Microbiology, Molecular Genetics and Molecular Biology Institute, University of California Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
N-Linked carbohydrates are frequently found in the V region of Ig H chains and can have a positive or negative effect on Ag binding affinity. We have studied a murine anti-alpha(1-->6) dextran V(H) that contains a carbohydrate in complementarity-determining region 2 (CDR2). This carbohydrate remains high mannose rather than being processed to a complex form, as would be expected for glycans on exposed protein loops. We have shown that the glycan remained high mannose when murine-human chimeric Abs were produced in a variety of cell types. Also, when another carbohydrate was present in CDR1, CDR2, or CDR3 of the L chain, the V(H) CDR2 glycan remained high mannose. Importantly, we found that when the anti-dextran V(H) CDR2 replaced CDR2 of an anti-dansyl V(H), the glycosylation site was used, but H chains were withheld in the endoplasmic reticulum and did not traffic to the Golgi apparatus. These results suggest that inappropriate V region glycosylation could contribute to ineffective Ab production from expressed Ig genes. In some cases, a carbohydrate addition sequence generated by either V region rearrangement or somatic hypermutation may result in an Ab that cannot be properly folded and secreted.
Collapse
Affiliation(s)
- Françoise A Gala
- Department of Microbiology, Immunology, and Molecular Genetics, The Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | | |
Collapse
|
20
|
Trinh R, Gurbaxani B, Morrison SL, Seyfzadeh M. Optimization of codon pair use within the (GGGGS)3 linker sequence results in enhanced protein expression. Mol Immunol 2004; 40:717-22. [PMID: 14644097 DOI: 10.1016/j.molimm.2003.08.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Here, we report that a significant increase in recombinant fusion antibody expression can be accomplished by adjusting the nucleotide sequence to conform to certain codon pairing rules. We investigated the expression of a protein in which a single chain Fv specific for HER2/neu with VH and VL joined by a flexible (GGGGS)3 linker was linked to the CH3 of a human anti-rat transferrin receptor IgG3 heavy chain with the same flexible (GGGGS)3 linker. In initial experiments we failed to achieve significant expression of this protein. However, when we made a single nucleotide change in each (GGGGS)3 linker we were able to achieve expression The change of one nucleotide within each linker did not alter either the amino acid sequence or the frequency score of these codon triplets' usage in mammalian cells. Instead they removed two codon pairs predicted to be detrimental to expression. In a transient transfection assay we find that this change results in an over 30-fold increase in expression that is not the result of an increase in the level of accumulated mRNA. In addition, the changes made it possible to isolate stably transfected mammalian cell clones producing high levels of fusion protein, which had not been possible using the original gene.
Collapse
Affiliation(s)
- Ryan Trinh
- Department of Microbiology, Immunology and Molecular Genetics and the Molecular Biology Institute, University of California Los Angeles, 405 Hilgard Avenue, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
21
|
Kumar S, Kalsi JK, Ravirajan CT, Latchman DS, Pearl LH, Isenberg DA. Molecular expression systems for anti-DNA antibodies--2. Lupus 2003; 11:833-42. [PMID: 12529048 DOI: 10.1191/0961203302lu304rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Antibodies to double-stranded DNA are the best-known serological markers of systemic lupus erythematosus, and are closely associated with its renal pathogenesis. How these antibodies recognize DNA is not fully understood. An understanding of the relationship between the functional attributes of an antibody with the three-dimensional structure of its antigen-combining site would allow an insight into the rules that dictate auto-antibody-nucleic acid interaction and consequent pathogenicity of the autoantibody. Data from such studies could assist the development of novel drugs as an approach to specific therapies that can inhibit or disrupt protein-nucleic acid interactions. A full understanding of the binding specificities can be achieved only by experimental determination of detailed three-dimensional structure of these antibodies alone, and of their complexes with specific DNA antigens. A prerequisite of such a study is the ability to produce multimilligram quantities of the antibody protein. However, these antibodies are particularly difficult to express, probably due to their DNA-binding activity. This review attempts to focus on the recent developments on the over-expression of anti-DNA antibody fragments in heterologous cell expression systems and their purification to homogeneity that would in turn allow their structural studies via crystallization.
Collapse
Affiliation(s)
- S Kumar
- Centre for Rheumatology, Bloomsbury Rheumatology Unit, Department of Medicine, University College London Hospital, London, UK.
| | | | | | | | | | | |
Collapse
|
22
|
Chintalacharuvu KR, Chuang PD, Dragoman A, Fernandez CZ, Qiu J, Plaut AG, Trinh KR, Gala FA, Morrison SL. Cleavage of the human immunoglobulin A1 (IgA1) hinge region by IgA1 proteases requires structures in the Fc region of IgA. Infect Immun 2003; 71:2563-70. [PMID: 12704129 PMCID: PMC153282 DOI: 10.1128/iai.71.5.2563-2570.2003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2002] [Revised: 01/14/2003] [Accepted: 02/12/2003] [Indexed: 11/20/2022] Open
Abstract
Secretory immunoglobulin A (IgA) protects the mucosal surfaces against inhaled and ingested pathogens. Many pathogenic bacteria produce IgA1 proteases that cleave in the hinge of IgA1, thus separating the Fab region from the Fc region and making IgA ineffective. Here, we show that Haemophilus influenzae type 1 and Neisseria gonorrhoeae type 2 IgA1 proteases cleave the IgA1 hinge in the context of the constant region of IgA1 or IgA2m(1) but not in the context of IgG2. Both C(alpha)2 and C(alpha)3 but not C(alpha)1 are required for the cleavage of the IgA1 hinge by H. influenzae and N. gonorrhoeae proteases. While there was no difference in the cleavage kinetics between wild-type IgA1 and IgA1 containing only the first GalNAc residue of the O-linked glycans, the absence of N-linked glycans in the Fc increased the ability of the N. gonorrhoeae protease to cleave the IgA1 hinge. Taken together, these results suggest that, in addition to the IgA1 hinge, structures in the Fc region of IgA are required for the recognition and cleavage of IgA1 by the H. influenzae and N. gonorrhoeae proteases.
Collapse
Affiliation(s)
- Koteswara R Chintalacharuvu
- Department of Microbiology, Immunology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, California 90095, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chintalacharuvu KR, Yu LJ, Bhola N, Kobayashi K, Fernandez CZ, Morrison SL. Cysteine residues required for the attachment of the light chain in human IgA2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5072-7. [PMID: 12391223 DOI: 10.4049/jimmunol.169.9.5072] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In humans, there are two subclasses of IgA, IgA1 and IgA2, with IgA2 existing as three allotypes, IgA2m(1), IgA2m(2) and IgA2(n). In IgA1, Cys(133) in C(H)1 forms the disulfide bond to the L chain. Our previous studies indicated that in IgA2 lacking Cys(133), a disulfide bond forms between the alpha-chain and the L chain when Cys(220) is followed by Arg(221), but not when Cys(220) is followed by Pro(221), suggesting that the Cys in C(H)1 might be involved in disulfide bonding to the L chain. However, here we show that covalent assembly of the H and L chains in IgA2(n) requires hinge-proximal Cys(241) and Cys(242) in C(H)2 and not Cys(196) or Cys(220) in C(H)1. Using pulse-chase experiments, we have demonstrated that wild-type IgA2(n) with Arg(221) and Cys(241) and Cys(242) assembles through a disulfide-bonded HL intermediate. In contrast, the major intermediate for IgA2 m(1) with Pro(221) assembly was H(2) even though both Cys(241) and Cys(242) were present. Only a small fraction of IgA2 m(1) assembles through disulfide-bonded HL. Overall, our studies indicate that for IgA2 covalent assembly of the H and L chains requires the hinge-proximal cysteines in C(H)2 and that the structure of C(H)1 influences the efficiency with which this covalent bond forms.
Collapse
Affiliation(s)
- Koteswara R Chintalacharuvu
- Department of Microbiology, Immunology and Molecular Genetics and Molecular Biology Institute, University of California, Los Angeles 90095, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Gala FA, Morrison SL. The role of constant region carbohydrate in the assembly and secretion of human IgD and IgA1. J Biol Chem 2002; 277:29005-11. [PMID: 12023968 DOI: 10.1074/jbc.m203258200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immunoglobulins are glycoproteins, containing N- linked carbohydrates in the heavy chain constant regions of all isotypes and O-linked carbohydrates in the hinge regions of human IgA1 and IgD. A previous study showed that IgD synthesized in the presence of tunicamycin and lacking the three N-linked glycans on the heavy chain was not secreted (Shin, S. U., Wei, D. F., Amin, A. R., Thorbecke, G. J., and Morrison, S. L. (1992) Hum. Antibodies 3, 65-74). The contribution of each of the carbohydrates in the Fc of IgD to assembly and secretion was now analyzed by eliminating the carbohydrate addition sequence, Asn-X-Ser/Thr, through site-directed mutagenesis. Only the carbohydrate nearest the sole disulfide bond between heavy chains, which remained high mannose and appeared to be buried within the folded molecule, was found to be essential for secretion. When IgD lacked that glycan, assembly reached only the heavy/light chain half-molecule stage, and heavy chains were held inside the endoplasmic reticulum. Using benzyl 2-acetamido-2-deoxy-alpha-d-galactopyranoside (BADG) to inhibit complete O-linked glycosylation, we found that IgA1 and IgD with incomplete hinge carbohydrates were assembled and secreted from cells. Thus, one N-linked glycan plays a structural role in IgD and is required for proper assembly and secretion, but the O-linked carbohydrates in the hinge of IgD and IgA1 are not required for folding and export.
Collapse
Affiliation(s)
- Francoise A Gala
- Department of Microbiology, University of California, Los Angeles, California 90095-1489, USA
| | | |
Collapse
|
25
|
Morrison SL, Mohammed MS, Wims LA, Trinh R, Etches R. Sequences in antibody molecules important for receptor-mediated transport into the chicken egg yolk. Mol Immunol 2002; 38:619-25. [PMID: 11792430 DOI: 10.1016/s0161-5890(01)00095-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Large quantities of antibodies are transported into the yolk of the chicken's egg. We have identified several regions within the antibody molecule important for its uptake into the egg yolk. An intact Fc and hinge region but not the Fc-associated carbohydrate are required for transport. Our data suggest that the C(H)2/C(H)3 interface is recognized by the receptor responsible for immunoglobulin (Ig) transport. At this interface, residues 251-254 form an exposed loop on the surface of C(H)2. Chicken IgY (cIgY) has the sequence LYIS and human IgG (hIgG) has the sequence LMIS at these positions; mutation of MIS to glycines results in an IgG that is not transported. A second site important for transport is at positions 429-432 within C(H)3. All transported antibodies have the sequence HEAL, whereas, murine IgG2b (mIgG2b) with the sequence HEGL and cIgA with the sequence HDGI fail to be transported. hIgA has the HEAL sequence and is transported.
Collapse
Affiliation(s)
- Sherie L Morrison
- Department of Microbiology, Immunology and Molecular Genetics, Molecular Biology Institute, University of California, 405 Hilgard Avenue, Los Angeles, CA 90095, USA.
| | | | | | | | | |
Collapse
|
26
|
Chintalacharuvu KR, Vuong LU, Loi LA, Larrick JW, Morrison SL. Hybrid IgA2/IgG1 antibodies with tailor-made effector functions. Clin Immunol 2001; 101:21-31. [PMID: 11580223 DOI: 10.1006/clim.2001.5083] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Immunoglobulin (Ig) A and IgG are the principal immune effector molecules at mucosal surfaces and in blood, respectively. Mucosal IgA is polymeric and bound to secretory component, whereas serum IgG is monomeric. We have now produced IgA2/IgG1 hybrid antibodies that combine the properties of IgA and IgG. Antibodies with Calpha3 at the end of the IgG H chain resemble IgA and form polymers with J chain that bind the polymeric Ig receptor. Like IgG, the hybrid proteins activated complement and bound FcgammaRI and protein A. Though the hybrid proteins contained both Cgamma2 and Cgamma3, they have a short in vivo half-life. Surprisingly, this decreased half-life correlated with a higher avidity than that of IgG for murine FcRn. Interestingly, antibodies with Calpha1 replacing Cgamma1 were resistant to extremes of pH, suggesting that Calpha1 increases antibody stability. These results provide insights into engineering antibodies with novel combinations of effector functions.
Collapse
Affiliation(s)
- K R Chintalacharuvu
- Department of Microbiology, Immunology, and Molecular Genetics, Molecular Biology Institute, University of California Los Angeles, 405 Hilgard Avenue, Los Angeles, California 90095, USA.
| | | | | | | | | |
Collapse
|
27
|
Kumar S, Kalsi J, Latchman DS, Pearl LH, Isenberg DA. Expression of the Fabs of human auto-antibodies in Escherichia coli: optimization and determination of their fine binding characteristics and cross-reactivity. J Mol Biol 2001; 308:527-39. [PMID: 11327785 DOI: 10.1006/jmbi.2001.4534] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Fabs of three human auto-antibodies (B3/33H11, anti-DNA; UK4, anti-phospholipid) and six related hybrids have been cloned and expressed in Escherichia coli, and their relative binding to single-stranded or double-stranded DNA or to cardiolipin has been assessed in the presence of modulators (salts and serum). We describe optimized conditions that have led to significant improvement in the quality and quantity of the purified auto-antibodies. Protein expression of the assembled and functionally active Fabs was achievable with a yield of up to 5 to 9 mg/l of culture. The comparative DNA/cardiolipin-binding analyses of the nine Fabs in the presence of modulators demonstrated that B3 and 33H11 L chains possess both anti-DNA and anti-cardiolipin activities. This is the first report of the demonstration that both anti-DNA and anti-cardiolipin activities may lie on the same light chain of a human auto-antibody. We provide evidence that the auto-antibodies that appeared to be similar, in that they bound DNA or cardiolipin in conventional ELISA immunoassays, exhibited significant difference in their cross-reactivity and binding to the antigen in the presence of modulators. Such auto- antigen specificity and/or cross-reactivity may dictate the potential of an auto-antibody to cause pathogenicity and may provide an explanation as to why apparently similar auto-antibodies behave differently in vivo.
Collapse
Affiliation(s)
- S Kumar
- Centre for Rheumatology, and Department of Biochemistry and Molecular Biology, University College London, UK.
| | | | | | | | | |
Collapse
|
28
|
Dela Cruz JS, Trinh KR, Morrison SL, Penichet ML. Recombinant anti-human HER2/neu IgG3-(GM-CSF) fusion protein retains antigen specificity and cytokine function and demonstrates antitumor activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:5112-21. [PMID: 11046042 DOI: 10.4049/jimmunol.165.9.5112] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anti-HER2/neu therapy of human HER2/neu-expressing malignancies such as breast cancer has shown only partial success in clinical trials. To expand the clinical potential of this approach, we have genetically engineered an anti-HER2/neu IgG3 fusion protein containing GM-CSF. Anti-HER2/neu IgG3-(GM-CSF) expressed in myeloma cells was correctly assembled and secreted. It was able to target HER2/neu-expressing cells and to support growth of a GM-CSF-dependent murine myeloid cell line, FDC-P1. The Ab fusion protein activated J774.2 macrophage cells so that they exhibit an enhanced cytotoxic activity and was comparable to the parental Ab in its ability to effect Ab-dependent cellular cytotoxicity-mediated tumor cell lysis. Pharmacokinetic studies showed that anti-HER2/neu IgG3-(GM-CSF) is stable in the blood. Interestingly, the half-life of anti-HER2/neu IgG3-(GM-CSF) depended on the injected dose with longer in vivo persistence observed at higher doses. Biodistribution studies showed that anti-HER2/neu IgG3-(GM-CSF) is mainly localized in the spleen. In addition, anti-HER2/neu IgG3-(GM-CSF) was able to target the HER2/neu-expressing murine tumor CT26-HER2/neu and enhance the immune response against the targeted Ag HER2/neu. Anti-HER2/neu IgG3-(GM-CSF) is able to enhance both Th1- and Th2-mediated immune responses and treatment with this Ab fusion protein resulted in significant retardation in the growth of s.c. CT26-HER2/neu tumors. Our results suggest that anti-HER2/neu IgG3-(GM-CSF) fusion protein is useful in the treatment of HER2/neu-expressing tumors.
Collapse
MESH Headings
- Adenocarcinoma/immunology
- Adenocarcinoma/pathology
- Adenocarcinoma/prevention & control
- Animals
- Antibodies, Anti-Idiotypic/biosynthesis
- Antibodies, Neoplasm/administration & dosage
- Antibodies, Neoplasm/biosynthesis
- Antibodies, Neoplasm/chemistry
- Antibodies, Neoplasm/genetics
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/pharmacokinetics
- Cell Division/genetics
- Cell Division/immunology
- Cell Line
- Cell Membrane/genetics
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/prevention & control
- Cytokines/physiology
- Cytotoxicity, Immunologic/genetics
- Epitopes/immunology
- Epitopes/metabolism
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Half-Life
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/genetics
- Immunoglobulin Isotypes/biosynthesis
- Immunoglobulin Isotypes/classification
- Injections, Intravenous
- Injections, Subcutaneous
- Macrophages/immunology
- Mice
- Mice, Inbred BALB C
- Protein Binding/genetics
- Protein Binding/immunology
- Receptor, ErbB-2/administration & dosage
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/chemical synthesis
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacokinetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J S Dela Cruz
- Departments of Microbiology and Molecular Genetics, and The Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
29
|
Abstract
A novel method for the site-specific introduction of genes into eukaryotic cells using the prokaryotic Cre-LoxP recombination system is presented. Cre recombinase catalyzes recombination between two LoxP sites or between two mutant LoxP 511 sites. However, recombination is not catalyzed between a LoxP and a LoxP 511 site. We now demonstrate that it is possible to catalyze accurate exchange between two DNA segments each flanked by a LoxP and a LoxP 511 site. In the example presented, expression of the Cre recombinase resulted in the replacement of a murine IgA constant region gene with a LoxP site at the 5' end and a LoxP 511 site at the 3' end by a human IgA constant region gene flanked by the same wild type and mutant LoxP sites. This method provides a novel approach for the site-specific substitution of specific genes.
Collapse
Affiliation(s)
- K R Trinh
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles, CA 90095-1747, USA
| | | |
Collapse
|
30
|
Chan LA, Lyczak JB, Zhang K, Morrison SL, Saxon A. The novel human IgE epsilon heavy chain, epsilon tailpiece, is present in plasma as part of a covalent complex. Mol Immunol 2000; 37:241-52. [PMID: 10930631 DOI: 10.1016/s0161-5890(00)00042-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Several splice variants of the secreted human epsilon heavy chain have previously been identified by reverse transcription-PCR. The heavy chain of one isoform, IgE tailpiece, differs from the originally identified IgE, IgE classic, by the replacement of the 2 carboxy-terminal amino acids by 8 novel amino acids including a carboxy-terminal cysteine residue. Recombinant human epsilon tailpiece and epsilon classic heavy chains were expressed and secreted as H2L2 monomers in Sp2/0 murine myeloma cells. We have investigated the in vitro function and in vivo occurrence of epsilon tailpiece heavy chains using receptor binding assays, granule release assays, flow cytometry, half-life studies, immunoprecipitation, SDS-PAGE, two-dimensional SDS-PAGE, and Western blotting. IgE tailpiece and IgE classic exhibited similar in vivo half-lives in BALB/c mice, bound the human high- and low-affinity IgE receptors with similar affinities and triggered equivalent levels of high affinity IgE receptor induced degranulation. In humans, IgE classic is present as a 190 kD circulating protein in vivo. In contrast, we found that in plasma epsilon tailpiece was primarily present as part of covalent complexes of approximately 300 and 338 kD. Dissociation of the complexes revealed that two species of epsilon tailpiece heavy chains were present therein and surprisingly, these in vivo derived epsilon tailpiece heavy chains were approximately 5 and 10 kD smaller than the recombinant expressed epsilon tailpiece or epsilon classic heavy chains. These results show that epsilon tailpiece is present in novel covalent complexes in humans.
Collapse
Affiliation(s)
- L A Chan
- The Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles 90095, USA
| | | | | | | | | |
Collapse
|
31
|
Abstract
Our understanding of the rules relating sequence to structure in antibodies has led to the development of accurate knowledge-based procedures for antibody modeling. Information gained from the analysis of antibody structures has been successfully exploited to engineer antibody-like molecules endowed with prescribed properties, such as increased stability or different specificity, many of which have a broad spectrum of applications both in therapy and in research. Here we describe a knowledge-based procedure for the prediction of the antibody-variable domains, based on the canonical structures method for the antigen-binding site, and discuss its expected accuracy and limitations. The rational design of antibody-based molecules is illustrated using as an example one of the most widely employed modifications of antibody structures: the humanization of animal-derived antibodies to reduce their immunogenicity for serotherapy in humans.
Collapse
Affiliation(s)
- V Morea
- IRBM "P. Angeletti,", Via Pontina Km. 30.600, Pomezia, 00040, Italy
| | | | | |
Collapse
|
32
|
O'Connor KC, Ghatak S, Stollar BD. Use of hydrophobic interaction chromatography to separate recombinant antibody fragments from associated bacterial chaperone protein GroEL. Anal Biochem 2000; 278:239-41. [PMID: 10660472 DOI: 10.1006/abio.1999.4465] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- K C O'Connor
- Department of Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
33
|
Penichet ML, Kang YS, Pardridge WM, Morrison SL, Shin SU. An Antibody-Avidin Fusion Protein Specific for the Transferrin Receptor Serves as a Delivery Vehicle for Effective Brain Targeting: Initial Applications in Anti-HIV Antisense Drug Delivery to the Brain. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.8.4421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
In the present study a novel Ab-avidin fusion protein has been constructed to deliver biotinylated compounds across the blood brain barrier. This fusion molecule consists of an Ab specific for the transferrin receptor genetically fused to avidin. The Ab-avidin fusion protein (anti-TfR IgG3-CH3-Av) expressed in murine myeloma cells was correctly assembled and secreted and showed both Ab- and avidin-related activities. In animal models, it showed much longer serum half-life than the chemical conjugate between OX-26 and avidin. Most importantly, this fusion protein demonstrated superior [3H]biotin uptake into brain parenchyma in comparison with the chemical conjugate. We also delivered a biotinylated 18-mer antisense peptide-nucleic acid specific for the rev gene of HIV-1 to the brain. Brain uptake of the HIV antisense drug was increased at least 15-fold when it was bound to the anti-TfR IgG3-CH3-Av, suggesting its potential use in neurologic AIDS. This novel Ab fusion protein should have general utility as a universal vehicle to effectively deliver biotinylated compounds across the blood-brain barrier for diagnosis and/or therapy of a broad range of CNS disorders such as infectious diseases, brain tumors as well as Parkinson’s and Huntington’s diseases.
Collapse
Affiliation(s)
- Manuel L. Penichet
- *Department of Microbiology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Young-Sook Kang
- †College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - William M. Pardridge
- ‡Department of Medicine and Brain Research Institute, University of California at Los Angeles School of Medicine, Los Angeles, CA 90095; and
| | - Sherie L. Morrison
- *Department of Microbiology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Seung-Uon Shin
- *Department of Microbiology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, CA 90095
- §Institute of Environment and Life Science, Hallym Academy of Sciences, Hallym University, Kangwon-do, Korea
| |
Collapse
|
34
|
Tripathi PK, Qin H, Bhattacharya-Chatterjee M, Ceriani RL, Foon KA, Chatterjee SK. Construction and characterization of a chimeric fusion protein consisting of an anti-idiotype antibody mimicking a breast cancer-associated antigen and the cytokine GM-CSF. Hybridoma (Larchmt) 1999; 18:193-202. [PMID: 10380019 DOI: 10.1089/hyb.1999.18.193] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Anti-idiotype antibody, 11D10 mimics biologically and antigenically a distinct and specific epitope of the high molecular weight human milk fat globule (HMFG), a cancer-associated antigen present in over 90% of breast tumor samples. To augment the immunogenicity of 11D10 without the aid of a carrier protein or adjuvant, we made a chimeric 11D10-GM-CSF fusion protein for use as a vaccine. An expression plasmid for 11D10 was made by ligation of the DNA sequences of the 11D10 light-chain variable region upstream of the human kappa constant region. The heavy-chain plasmid carrying GM-CSF was made by ligation of the heavy-chain variable region sequences upstream of the human gamma1 constant region CH1 fused to the DNA fragment encoding the mature GM-CSF peptide 3' to the CH3 exon. NS1 plasmacytoma cells were transfected with the light and heavy-chain vectors by electroporation. Fusion protein secreted in the culture medium was purified and was characterized by gel electrophoresis as well as by determination of the biological activity of the fused GM-CSF. In nonreducing SDS-polyacrylamide gels, a single band approximately 200 Kd reacted with anti-human kappa, anti-human lambda1 and anti-GM-CSF antibodies. In reducing polyacrylamide gels, a approximately 74 kd protein reacted with anti-human lambda1 and anti-GM-CSF antibodies. The fusion protein induced proliferation of GM-CSF dependent NFS-60 cells. These results suggest that the protein is a chimeric anti-idiotype antibody consisting of 11D10 variable domains, human kappa and lambda1 constant domains and that the GM-CSF moiety fused to the constant region lambda1 is biologically active.
Collapse
Affiliation(s)
- P K Tripathi
- Department of Internal Medicine, and The Lucille Parker Markey Cancer Center, University of Kentucky Medical Center, Lexington 40536, USA
| | | | | | | | | | | |
Collapse
|
35
|
Gan J, Kendra K, Ricci M, Hank JA, Gillies SD, Sondel PM. Specific enzyme-linked immunosorbent assays for quantitation of antibody-cytokine fusion proteins. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 1999; 6:236-42. [PMID: 10066660 PMCID: PMC95693 DOI: 10.1128/cdli.6.2.236-242.1999] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Preliminary testing has shown in vitro and in vivo that antitumor activity can be obtained with fusion proteins linking tumor-reactive monoclonal antibodies to cytokines, such as granulocyte-macrophage colony-stimulating factor or interleukin 2 (IL-2). Preclinical and clinical testing of these reagents requires their in vitro and in vivo quantitation and pharmacokinetic evaluation. We have focused on the detection of a fusion protein which links one human IL-2 molecule to the carboxy terminus of each heavy chain of the tumor-reactive human-mouse chimeric anti-GD2 antibody, ch14.18. We have developed enzyme-linked immunosorbent assays (ELISAs) to evaluate intact tumor-reactive fusion proteins. By these ELISAs we can reliably measure nanogram quantities of intact ch14.18-IL-2 fusion protein and distinguish the intact protein from its components (ch14.18 and IL-2) in buffer, mouse serum, and human serum with specificity and reproducibility. The measurement of intact ch14.18-IL-2 fusion protein is not confounded by free IL-2 or free ch14.18 when 100 ng or less of total immunoglobulin per ml is used during the assay procedure. Our results indicate that these ELISAs are suitable for preclinical and clinical testing and with slight modifications are applicable to the analysis of a variety of other fusion proteins.
Collapse
Affiliation(s)
- J Gan
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin 53792, USA
| | | | | | | | | | | |
Collapse
|
36
|
Zhang HF, Yu J, Bajwa E, Morrison SL, Tomlinson S. Targeting of functional antibody-CD59 fusion proteins to a cell surface. J Clin Invest 1999; 103:55-61. [PMID: 9884334 PMCID: PMC407863 DOI: 10.1172/jci4607] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/1998] [Accepted: 11/03/1998] [Indexed: 11/17/2022] Open
Abstract
Complement is involved in the pathogenesis of many diseases, and there is great interest in developing inhibitors of complement for therapeutic application. CD59 is a natural membrane-bound inhibitor of the cytolytic complement membrane attack complex (MAC). In this study, the preparation and characterization of antibody-CD59 (IgG-CD59) chimeric fusion proteins are described. Constructs were composed of soluble CD59 fused to an antibody-combining site at the end of CH1, after the hinge (H), and after CH3 Ig regions. The antigen specificity of each construct was for the hapten 5-dimethylamino-naphthalene-1-sulfonyl (dansyl). Correct folding of each IgG-CD59 fusion partner was indicated by recognition with anti-CD59 antibodies specific for conformational determinants and by IgG-CD59 binding to dansyl. The IgG-CD59 fusion proteins all bound specifically to dansyl-labeled Chinese hamster ovary cells and provided targeted cells, but not untargeted cells, with effective protection from complement-mediated lysis. Data indicate that CD59 must be positioned in close proximity to the site of MAC formation for effective function, and that modes of membrane attachment other than glycophosphatidylinositol linkage can affect CD59 functional activity.
Collapse
Affiliation(s)
- H F Zhang
- Department of Pathology, New York University Medical Center, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
37
|
Penichet ML, Harvill ET, Morrison SL. An IgG3-IL-2 fusion protein recognizing a murine B cell lymphoma exhibits effective tumor imaging and antitumor activity. J Interferon Cytokine Res 1998; 18:597-607. [PMID: 9726441 DOI: 10.1089/jir.1998.18.597] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Antibody (Ab)-based tumor therapeutics use the tumor-binding specificity of the Ab to target Fc functions or associated molecules to the site of the tumor. We have used an Ab-interleukin-2 (IL-2) fusion protein to deliver IL-2 to a murine B cell lymphoma (38C13). This anti-Id IgG3-CH3-IL-2, which recognizes the idiotype present on the surface of the lymphoma has a half-life in mice approximately 17-fold longer than the half-life reported for IL-2. Gamma camera studies showed that anti-Id IgG3-CH3-IL-2 localizes at the site of a subcutaneous tumor in mice. The anti-Id IgG3-CH3-IL-2 also shows enhanced antitumor activity compared with the combination of Ab and IL-2 administered together. However, the mechanism of antitumor activity appears to depend on the dose and the treatment schedule used. A single dose of fusion protein prevented tumor in only 50% of the animals, although all the survivors showed some evidence of immunologic memory. Although multiple doses are more effective in preventing tumor growth (87% survivors), they are ineffective in generating protective immunologic memory. Our results suggest that Ab-IL-2 fusion proteins will be useful in the diagnosis and treatment of human B cell lymphomas and other related malignancies.
Collapse
Affiliation(s)
- M L Penichet
- Department of Microbiology and Molecular Genetics, Molecular Biology Institute, University of California, Los Angeles 90095-1489, USA
| | | | | |
Collapse
|
38
|
Hackett J, Hoff-Velk J, Golden A, Brashear J, Robinson J, Rapp M, Klass M, Ostrow DH, Mandecki W. Recombinant mouse-human chimeric antibodies as calibrators in immunoassays that measure antibodies to Toxoplasma gondii. J Clin Microbiol 1998; 36:1277-84. [PMID: 9574691 PMCID: PMC104814 DOI: 10.1128/jcm.36.5.1277-1284.1998] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the present study, we examined the feasibility of using recombinant antibodies containing murine variable regions and human constant regions as calibrators or controls in immunoassays. As a model system, we chose the Abbott IMx Toxo immunoglobulin M (IgM) and Toxo IgG assays designed to detect antibodies to Toxoplasma gondii. Two mouse monoclonal antibodies were selected based on their reactivity to the T. gondii antigens P30 and P66. Heavy- and light-chain variable-region genes were cloned from both hybridomas and transferred into immunoglobulin expression vectors containing human kappa and IgG1 or IgM constant regions. The constructs were stably transfected into Sp2/0-Ag14 cells. In the IMx Toxo IgG assay, immunoreactivity of the anti-P30 chimeric IgG1 antibody paralleled that of the positive human plasma-derived assay calibrators. Signal generated with the anti-P66 chimeric IgG1 antibody was observed to plateau below the maximal reactivity observed for the assay calibrator. Examination of the IgM chimeric antibodies in the IMx Toxo IgM assay revealed that both the anti-P30 and anti-P66 antibodies matched the assay index calibrator manufactured with human Toxo IgM-positive plasma. When evaluated with patient samples, the correlation between results obtained with the chimeric antibody calibrators and the positive human plasma calibrators was > or =0.985. These data demonstrate that chimeric mouse-human antibodies are a viable alternative to high-titer positive human plasma for the manufacture of calibrators and controls for diagnostic assays.
Collapse
Affiliation(s)
- J Hackett
- Abbott Diagnostics Division, Abbott Laboratories, North Chicago, Illinois 60064, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sensel MG, Kane LM, Morrison SL. Amino acid differences in the N-terminus of C(H)2 influence the relative abilities of IgG2 and IgG3 to activate complement. Mol Immunol 1997; 34:1019-29. [PMID: 9488053 DOI: 10.1016/s0161-5890(97)00112-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The four human IgG isotypes are highly conserved in amino acid sequence, but show differential ability to activate complement (C'): IgG3 and IgG1 are very active, IgG2 is active under certain conditions, and IgG4 is inactive. Although the second constant domain [C(H)2] is critical for C' activation, the individual amino acids that confer isotype-specific activity have not been identified. We have generated a series of mutants between IgG2 and IgG3, resulting in the exchange of the four N-terminal and six C-terminal polymorphic residues within C(H)2. Mutants containing the N-terminus of the C(H)2 of IgG3 were as effective as wildtype IgG3 in C1q binding, C1 activation and terminal complex (MAC) formation, but had reduced ability to effect C'-mediated lysis. IgG2 and mutants containing the N-terminal portion of the C(H)2 of IgG2 were reduced compared to IgG3 in activating C1, binding C1q and inducing assembly of the MAC, and were inactive in mediating lysis of target cells. Thus, the amino acid sequence differences in the N-terminus of C(H)2 play a critical role in determining the relative abilities of IgG2 and IgG3 to bind C1q and activate the C' cascade although additional residues of C(H)2 must be involved in mediating optimal target cells lysis. The sequence of the N-terminus of C(H)2 was less critical in determining C4 and C3 binding. Characterization of domain exchange mutants suggests that intermediate steps may be partly dependent on domains other than C(H)2. IgGs that do not direct target cell lysis nevertheless activate intermediate steps in the pathway, which may contribute to immune complex-associated disorders.
Collapse
Affiliation(s)
- M G Sensel
- Department of Microbiology and Molecular Genetics, Molecular Biology Institute, University of California, Los Angeles 90095-1489, USA
| | | | | |
Collapse
|
40
|
Abstract
We have produced novel bispecific antibodies by fusing the DNA encoding a single chain antibody (ScFv) after the C terminus (CH3-ScFv) or after the hinge (Hinge-ScFv) with an antibody of a different specificity. The fusion protein is expressed by gene transfection in the context of a murine variable region. Transfectomas secrete a homogeneous population of the recombinant antibody with two different specificities, one at the N terminus (anti-dextran) and one at the C terminus (anti-dansyl). The CH3-ScFv antibody, which maintains the constant region of human IgG3, has some of the associated effector functions such as long half-life and Fc receptor binding. The Hinge-ScFv antibody which lacks the CH2 and CH3 domains has no known effector functions.
Collapse
Affiliation(s)
- M J Coloma
- Department of Microbiology and Molecular Genetics, University of California at Los Angeles 90095, USA
| | | |
Collapse
|
41
|
Harvill ET, Morrison SL. An IgG3-IL-2 fusion protein has higher affinity than hrIL-2 for the IL-2R alpha subunit: real time measurement of ligand binding. Mol Immunol 1996; 33:1007-14. [PMID: 8960125 DOI: 10.1016/s0161-5890(96)00027-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The alpha subunit of the interleukin-2 (IL-2) receptor (IL-2R alpha)3 has the highest individual affinity for IL-2 and is the only subunit not known to bind other cytokines. The interactions between IL-2 and IL-2R alpha studied in cell binding assays have revealed a number of factors which may vary significantly in different cell lines used for these assays in different laboratories. In order to avoid the problems associated with cellular assays we used an optical biosensor to examine the interaction between IL-2R alpha and hrIL-2. Real-time measurement of association and dissociation resulted in a calculated KD of 1.9 x 10(-7) M for this interaction. We then examined the IL-2R alpha binding of a potentially bivalent IgG3-IL2 fusion protein previously shown to have a higher affinity than hrIL-2 for the high affinity IL-2R but not the intermediate affinity IL-2R. Biosensor measurements of association and dissociation of IgG3-IL2 to IL-2R alpha yielded a similar association rate but a decreased dissociation rate compared to hrIL-2, resulting in a KD of 5.3 x 10(-8) M. This system is applicable to the numerous IL-2 mutants with different affinities and activities and is generalizable to other cytokine/receptor interactions.
Collapse
Affiliation(s)
- E T Harvill
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles 90095, USA
| | | |
Collapse
|
42
|
Lyczak JB, Zhang K, Saxon A, Morrison SL. Expression of novel secreted isoforms of human immunoglobulin E proteins. J Biol Chem 1996; 271:3428-36. [PMID: 8631944 DOI: 10.1074/jbc.271.7.3428] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Four human IgE isoforms produced by alternative splicing of the epsilon primary transcript were expressed as chimeric mouse/human anti 5-dimethylamino-1-naphthalenesulfonyl antibodies in the murine myeloma cell line Sp2/0. The four isoforms include the classic secreted form and three novel isoforms with altered carboxyl termini. All of these isoforms lack the transmembrane region encoded by the M1/M1' exon and are therefore predicted to be secreted proteins. When expressed in Sp2/0 cells, three of the IgE isoforms are assembled into complete molecules of two Ig heavy chains and two Ig light chains, whereas the fourth isoform is predominately assembled into half-molecules of one Ig heavy chain and one Ig light chain. All four isoforms are secreted with similar kinetics. In contrast, when the isoform containing the C epsilon4 domain joined directly to the M2 exon (IgE grandé) is expressed in the J558L cell line, it is degraded intracellularly, suggesting a cell line-dependent regulation of secretion. These data show that these novel isoforms of human IgE, predicted to occur from in vivo and in vitro mRNA analysis, can be produced and secreted by mammalian cells. The different forms of IgE may have physiologically relevant but distinct roles in human IgE-mediated immune inflammation. The availability of purified recombinant human IgE isoforms makes it possible to analyze the functional differences among them.
Collapse
Affiliation(s)
- J B Lyczak
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
43
|
Harvill ET, Morrison SL. An IgG3-IL2 fusion protein activates complement, binds Fc gamma RI, generates LAK activity and shows enhanced binding to the high affinity IL-2R. IMMUNOTECHNOLOGY : AN INTERNATIONAL JOURNAL OF IMMUNOLOGICAL ENGINEERING 1995; 1:95-105. [PMID: 9373338 DOI: 10.1016/1380-2933(95)00009-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The therapeutic value of Interleukin 2 (IL-2) is limited by its short half life and systemic toxicity. One approach to overcoming these problems is to fuse this protein to an antibody, a protein with a long half life and the ability to target a unique antigen within the body. To examine the biochemical properties of such a molecule a fusion protein was constructed linking the N-terminus of human IL-2 to the C-terminus of IgG3. A similar fusion between IgG1 and IL-2 has previously been shown to bind antigen, generate antibody-dependent cellular cytotoxicity (ADCC) and stimulate T cell proliferation and cytotoxicity. We now extend these studies and show that the fusion protein, termed IgG3-IL2, is appropriately N-glycosylated within the IgG3 CH2 domain, binds the human high affinity Fc receptor (Fc gamma RI) with an affinity slightly lower than that of IgG3, and is able to activate complement via the classical pathway to lyse antigen coated sheep red blood cells (SRBC). When used to stimulate the proliferation of the IL-2 dependent cell line CTLL-2, IgG3-IL2 has a specific activity slightly lower than that of human recombinant IL-2 (hrIL-2). In marked contrast, when comparable unit concentrations, as defined by the standard CTLL-2 proliferation assay, are used to stimulate human peripheral blood lymphocytes (PBL), IgG3-IL2 generates significantly greater lymphokine activated killer (LAK) cell cytotoxicity than does hrIL-2. Competition studies show that IgG3-IL2 binds the intermediate affinity form of the IL-2 receptor (IL-2R), consisting of the beta and gamma subunits, with an affinity slightly less than that of hrIL-2. In contrast, IgG3-IL2 shows a greater affinity than hrIL-2 for the high affinity IL-2R, consisting of alpha, beta and gamma subunits. Our studies show that the IgG3-IL2 fusion protein possesses a combination of the biological properties of IgG3 and IL-2 including antigen binding, complement activation, Fc gamma RI binding, IL-2R binding and stimulation of both proliferation and LAK activity. This combination of activities may allow IgG3-IL2 to target humoral and cell-mediated immune activation to the site of an antigen of interest or target an antigen to IL-2R bearing cells or organs.
Collapse
Affiliation(s)
- E T Harvill
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles 90024-1489, USA
| | | |
Collapse
|
44
|
Nickerson KG, Tao MH, Chen HT, Larrick J, Kabat EA. Human and mouse monoclonal antibodies to blood group A substance, which are nearly identical immunochemically, use radically different primary sequences. J Biol Chem 1995; 270:12457-65. [PMID: 7759488 DOI: 10.1074/jbc.270.21.12457] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
A human monoclonal antibody (HuA) specific for blood group A substance with two fucose groups was found to be immunochemically almost identical with that of a previously characterized mouse monoclonal anti-A, AC-1001. The VH and VL chain cDNAs of HuA were sequenced and compared with those of AC-1001. The human and mouse antibodies used VH and Vk genes that came from different families and shared minimal nucleotide and amino acid sequence identity. Thus, two antibodies from two different species can use evolutionarily unrelated sequences to bind the same carbohydrate epitope. The cloned HuA VH and VL genes were then transfected into a mouse myeloma cell line and re-expressed, together, and each separately with an irrelevant VH or VL. Only the original HuA VH and Vk had anti-A activity, demonstrating that both the heavy and light chains contributed to specificity.
Collapse
Affiliation(s)
- K G Nickerson
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | |
Collapse
|
45
|
Poon PH, Morrison SL, Schumaker VN. Structure and function of several anti-dansyl chimeric antibodies formed by domain interchanges between human IgM and mouse IgG2b. J Biol Chem 1995; 270:8571-7. [PMID: 7721758 DOI: 10.1074/jbc.270.15.8571] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Two pairs of chimeric, domain-switched immunoglobulins with identical murine, anti-dansyl (5-dimethylaminonaphthalene-1-sulfonyl) variable domains have been generated, employing as parent antibodies a human IgM and a mouse IgG2b. The first pair of chimeric antibodies mu mu gamma mu and gamma gamma mu gamma was generated by switching the C mu 3 and C gamma 2 domains between IgM and IgG2b. The second pair of chimeras mu mu gamma gamma and gamma gamma mu mu were formed by switching both C mu 3 and C mu 4 with C gamma 2 and C gamma 3. SDS-polyacrylamide gel electrophoresis and analytical ultracentrifugation showed that over half (57 and 71%) of the two chimeric antibodies possessing the C mu 4 domain and tail piece formed disulfide-linked IgM-like polymers. In contrast, the two chimeric antibodies lacking the C mu 4 domain were almost entirely monomeric. Both monomeric chimeras had reduced ability to activate complement. The chimera gamma gamma mu gamma had no activity under any of the assay conditions, whereas mu mu gamma gamma caused only a small amount of cell lysis but was fully active in consuming complement at 4 degrees C. The polymeric chimera gamma gamma mu mu was much less active than IgM, bound C1 weakly and caused some cell lysis but consumed little complement with soluble antigen. The polymeric chimera mu mu gamma mu bound C1 strongly and was the most active antibody in all assays, even more active than the parental IgG2b and IgM antibodies; it was the only antibody that exhibited antigen-independent activity. The results suggest that C mu 3 alone does not constitute the complement binding site in IgM but requires both C mu 1-2 and C mu 4 for full activity.
Collapse
Affiliation(s)
- P H Poon
- Department of Chemistry and Biochemistry, University of California, Los Angeles 90024, USA
| | | | | |
Collapse
|
46
|
Shin SU, Friden P, Moran M, Olson T, Kang YS, Pardridge WM, Morrison SL. Transferrin-antibody fusion proteins are effective in brain targeting. Proc Natl Acad Sci U S A 1995; 92:2820-4. [PMID: 7708731 PMCID: PMC42310 DOI: 10.1073/pnas.92.7.2820] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In the present study, the receptor binding potential of transferrin (Tf) was linked to an antibody binding specificity. Human Tf was fused to mouse-human chimeric IgG3 at three positions: at the end of heavy chain constant region 1 (CH1), after the hinge, and after CH3. The resulting Tf-antibody fusion proteins were able to bind antigen and the Tf receptor. The CH3-Tf fusion protein showed no complement-mediated cytolysis but possessed IgG receptor I (Fc gamma RI) binding activity. Most importantly, all of the fusion proteins demonstrated significant uptake into brain parenchyma, with 0.3% of the injected dose of the hinge-Tf fusion protein rapidly targeted to the brain. Recovery of iodinated CH3-Tf fusion protein from the brain parenchyma demonstrated that the fusion proteins can cross the blood-brain barrier intact. The binding specificity of these fusion proteins can be used for brain delivery of noncovalently bound ligands, such as drugs and peptides, or for targeting antigens present within the brain.
Collapse
Affiliation(s)
- S U Shin
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles 90095, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Several cDNA expression vectors were constructed and tested by stable transfection into a murine lymphoid cell line in order to compare secretion rates of a human immunoglobulin (Ig) light chain (LC). When the cDNA was under transcriptional control of the SV40 promoter and enhancer and preceded by the SV40 19S late mRNA intron, a weak LC production was detected. Secretion rate was not improved by replacing the SV40 promoter and enhancer by a combination of a murine Ig heavy chain (HC) gene promoter and enhancer even with insertion of additional Ig enhancers. In contrast, replacement of the 19S intron by a large intron derived from a human Ig HC gene and containing the intronic enhancer dramatically increased the secretion rate. High-level production was also obtained with the same enhancer-containing intron placed downstream from the LC cDNA. Stable transfectants were obtained that secreted the human LC in amounts comparable to those obtained with Ig genes. Our results suggest that the SV40 19S late mRNA intron used in several expression vectors is not appropriate when the purpose is to produce large amounts of antibody molecules. By providing transcriptional, splicing and polyadenylation signals, the presently described vectors will be useful for easy cloning and high-level expression in lymphoid cells of cDNAs or PCR products encoding antibody molecules.
Collapse
Affiliation(s)
- A A Khamlichi
- Laboratoire d'Immunologie et Génétique, CNRS URA 1172, IBMIG, Faculté des Sciences, Poitiers, France
| | | | | |
Collapse
|
48
|
Wright A, Morrison SL. Effect of altered CH2-associated carbohydrate structure on the functional properties and in vivo fate of chimeric mouse-human immunoglobulin G1. J Exp Med 1994; 180:1087-96. [PMID: 8064227 PMCID: PMC2191655 DOI: 10.1084/jem.180.3.1087] [Citation(s) in RCA: 161] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Immunoglobulin G (IgG) molecules are glycosylated in CH2 at Asn297; the N-linked carbohydrates attached there have been shown to contribute to antibody (Ab) stability and various effector functions. The carbohydrate attached to the IgG constant region is a complex biantennary structure. Alterations in the structure of oligosaccharide have been associated with human diseases such as rheumatoid arthritis and osteoarthritis. To study the effects of altered carbohydrate structure on Ab effector function, we have used gene transfection techniques to produce mouse-human chimeric IgG1 Abs in the Chinese hamster ovary (CHO) cell line Lec 1, which is incapable of processing the high-mannose intermediate through the terminal glycosylation steps. We also produced IgG1 Abs in Pro-5, the wild-type CHO cell line that is the parent of Lec 1. The Pro-5-produced Ab (IgG1-Pro-5) was similar to IgG1-My 1, a myeloma-produced IgG1 Ab of the same specificity, in its biologic properties such as serum half-life, ability to effect complement-mediated cytolysis, and affinity for Fc gamma RI. Although the Lec 1-produced Ab, IgG1-Lec 1, was properly assembled and retained antigen specificity, it was incapable of complement-mediated hemolysis and was substantially deficient in complement consumption, C1q binding, and C1 activation. IgG1-Lec 1 also showed reduced but significant affinity for Fc gamma R1 receptors. The in vivo half-life of IgG1-Lec 1 was shorter than that of either the myeloma- or Pro-5-produced counterpart, with more being cleared during the alpha-phase and with more rapid clearance during the beta-phase. Clearance of IgG1-Lec 1 could be inhibited by the administration of yeast-derived mannan. Thus the uptake of IgG1-Lec 1 appears to be accelerated by the presence of terminally mannosylated oligosaccharide. Therefore, certain Ab functions as well as the in vivo fate of the protein are dramatically affected by altered carbohydrate structure. Expression of Igs in cell lines with defined glycosylation mutations is shown to be a useful technique for investigating the contribution of carbohydrate structure to Ab function.
Collapse
Affiliation(s)
- A Wright
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles 90024
| | | |
Collapse
|
49
|
Smith RI, Morrison SL. Recombinant polymeric IgG: an approach to engineering more potent antibodies. BIO/TECHNOLOGY (NATURE PUBLISHING COMPANY) 1994; 12:683-8. [PMID: 7764912 DOI: 10.1038/nbt0794-683] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
IgM and IgG are the only classes of immunoglobulin capable of initiating the classical complement cascade. IgM, however, fixes complement much more effectively than IgG making it a more attractive Fc for some immunotherapeutic strategies. IgG, on the other hand, triggers additional effector functions mediated through gamma specific Fc receptors. Using a generalizable technique we have produced IgM-like polymers of IgG that possess both the Fc gamma receptor binding properties of IgG and the more potent complement activity of IgM.
Collapse
Affiliation(s)
- R I Smith
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles 90024
| | | |
Collapse
|
50
|
Lyczak JB, Morrison SL. Biological and pharmacokinetic properties of a novel immunoglobulin-CD4 fusion protein. Arch Virol 1994; 139:189-96. [PMID: 7826209 DOI: 10.1007/bf01309464] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We report a CD4-immunoglobulin fusion protein in which the first two extracellular domains of human CD4 replace the Fc region of the immunoglobulin. When co-expressed with a gene encoding an immunoglobulin light chain, the protein was covalently assembled into a form having an M(r) consistent with that expected for two fusion heavy chains and two immunoglobulin light chains. The antigen specificity of the antibody was retained, however, binding to HIV gp120 was lost. Pharmacokinetic analysis revealed the in vivo half-life of the fusion protein to be 2.4 h in mice.
Collapse
Affiliation(s)
- J B Lyczak
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles
| | | |
Collapse
|