1
|
Wang Q, Chen Y, Park J, Liu X, Hu Y, Wang T, McFarland K, Betenbaugh MJ. Design and Production of Bispecific Antibodies. Antibodies (Basel) 2019; 8:antib8030043. [PMID: 31544849 PMCID: PMC6783844 DOI: 10.3390/antib8030043] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/18/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
With the current biotherapeutic market dominated by antibody molecules, bispecific antibodies represent a key component of the next-generation of antibody therapy. Bispecific antibodies can target two different antigens at the same time, such as simultaneously binding tumor cell receptors and recruiting cytotoxic immune cells. Structural diversity has been fast-growing in the bispecific antibody field, creating a plethora of novel bispecific antibody scaffolds, which provide great functional variety. Two common formats of bispecific antibodies on the market are the single-chain variable fragment (scFv)-based (no Fc fragment) antibody and the full-length IgG-like asymmetric antibody. Unlike the conventional monoclonal antibodies, great production challenges with respect to the quantity, quality, and stability of bispecific antibodies have hampered their wider clinical application and acceptance. In this review, we focus on these two major bispecific types and describe recent advances in the design, production, and quality of these molecules, which will enable this important class of biologics to reach their therapeutic potential.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yiqun Chen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jaeyoung Park
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xiao Liu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yifeng Hu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tiexin Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kevin McFarland
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
2
|
Melief CJM, Scheper RJ, de Vries IJM. Scientific contributions toward successful cancer immunotherapy in The Netherlands. Immunol Lett 2014; 162:121-6. [PMID: 25455598 DOI: 10.1016/j.imlet.2014.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This historical overview shows that immunologists and clinicians from The Netherlands have contributed in a major way to better insights in the nature of cancer immunity. This work involved elucidation of the nature of cancer-associated antigens in autologous and allogeneic settings in addition to understanding of the cellular basis of natural immune responses against cancers and of important immune evasion mechanisms. Insight into such basic immunological mechanisms has contributed to the development of innovating therapies.
Collapse
Affiliation(s)
- Cornelis J M Melief
- Leiden University Medical Center, The Netherlands; ISA Pharmaceuticals, The Netherlands.
| | - Rik J Scheper
- Department of Pathology, Free University Hospital, Amsterdam, The Netherlands
| | | |
Collapse
|
3
|
Zhang L, Hou Y, Zhang J, Hu J, Zhang K. Cytotoxicity of cytokine-induced killer cells targeted by a bispecific antibody to gastric cancer cells. Oncol Lett 2013; 5:1826-1832. [PMID: 23833649 PMCID: PMC3701003 DOI: 10.3892/ol.2013.1281] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 12/20/2012] [Indexed: 01/22/2023] Open
Abstract
The aim of the present study was to investigate the cytotoxic activity of cytokine-induced killer (CIK) cells targeted by an epidermal growth factor receptor (EGFR)/CD3 bispecific antibody (BsAb) to the gastric cancer cell line SGC7901. A BsAb was constructed by chemically cross-linking a monoclonal antibody (McAb) against human CD3 with another McAb against human EGFR. An immunocytochemistry assay was performed to detect the expression of EGFR in SGC7901 cells. The cytotoxic activity of CIK cells targeted by the EGFR/CD3 BsAb was analyzed by the 51Cr release assay, Subsequently, a comparison of the cytotoxic activity between CIK cells targeted by EGFR/CD3 BsAb, CIK cells targeted by EGFR McAb or/and CD3 McAb and CIK cells was performed. The antineoplastic activity of the CIK cells directed using the EGFR/CD3 BsAb in vivo was analyzed by tumor growth and tumor reduction assays. The cell lysis rate of CIK cells targeted by the EGFR/CD3 BsAb was higher compared with those of CIK cells targeted by CD3 McAb only or by CD3 McAb and EGFR McAb. The lysis rates of the latter two groups were significantly higher than those of CIK cells targeted by EGFR McAb only and CIK cells (P<0.05). The mean tumor reduction using the administration of CIK cells directed by the EGFR/CD3 BsAb was higher than those of the other groups (P<0.05). The results indicate that the EGFR/CD3 BsAb is able to enhance the ability of CIK cells to bind to and kill gastric cancer cells in vitro and in vivo.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Gastroenterology, The 309 Hospital of the PLA, Beijing 100091
| | | | | | | | | |
Collapse
|
4
|
Abstract
Monoclonal antibodies (mAbs) appear to offer many benefits for the treatment of cancer and in particular lymphoma (1). They are natural products that can be made with precise specificity and in almost unlimited amounts. In addition, mAbs can be selected or engineered to efficiently recruit the body's effector systems, such as complement and natural killer cells, against the unwanted cells in much the same way as they might destroy an invading pathogen. Unfortunately, progress in the clinic has been slow, and the cytotoxic activity achieved with mAb in vitro has failed to be transferred into patients. Despite this rather disappointing outcome, recent results in treating non-Hodgkin's lymphoma (NHL) and chronic lymphocytic leukemia (CLL) with anti-CD20 and anti-CD52 (CAMPATH 1) mAb suggest, at least for certain neoplasms, that the situation may be changing (2,3). Stevenson and colleagues (personal communication) have recently achieved more than 70% complete responses in posttransplant lymphoma treated with a chimeric anti-CD20 mAb, and Maloney and co-workers (2) recently reported a 50% response rate in relapsed, low-grade, NHL, with a 10---11 mo duration. Encouragingly, patients in these studies did not raise antibody responses to the treatment anti-CD20 mAb and, unlike the situation following therapy in many lymphomas with anti-idiotype (Id) mAb, the emergence of antigen-negative tumors has not been seen (4). To underline its clinical success, anti-CD20 mAb (Rituximab) has now become the first anticancer mAb to become licensed by the FDA for lymphoma treatment. One of the most encouraging aspects of Rituximab treatment is that, in addition to its therapeutic activity, which appears to match that of more conventional chemotherapy in a similar setting, it has very few adverse effects and can be given to patients who are in poor condition with advanced disease. Early experience suggests that it will be this lack of adverse effects that will be its most attractive feature.
Collapse
|
5
|
Sharma HS, Patnaik R, Patnaik S, Mohanty S, Sharma A, Vannemreddy P. Antibodies to serotonin attenuate closed head injury induced blood brain barrier disruption and brain pathology. Ann N Y Acad Sci 2008; 1122:295-312. [PMID: 18077582 DOI: 10.1196/annals.1403.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Closed head injury (CHI) often results in profound brain swelling and instant death of the victims due to compression of the vital centers. However, the neurochemical basis of edema formation in CHI is still obscure. Previous studies from our laboratory show that blockade of serotonin synthesis prior to CHI in a rat model attenuates brain edema, indicating a prominent role for serotonin in head injury. Thus, neutralization of endogenous serotonin activity and/or blocking of its receptors will induce neuroprotection in CHI. Since serotonin has more than 14 receptors and selective serotonin antagonists are still not available, we used serotonin antiserum to neutralize its in vivo effects before or after CHI in a rat model. CHI was produced by an impact of 0.224 N on the right parietal skull bone under Equithesin anesthesia by dropping a weight of 114.6 g from a height of 20 cm through a guide tube. This concussive brain injury resulted in blood-brain barrier (BBB) disruption, brain edema formation, and volume swelling at 5 h that were most pronounced in the contralateral cerebral hemisphere. The plasma and brain serotonin levels were increased several-fold at this time. Intracerebroventricular administration of serotonin antiserum (1:20, monoclonal) into the left lateral cerebral ventricle (30 microL in PBS) 30 min before or 30 min (but not 60 min) after CHI significantly attenuated BBB disruption, brain edema formation, volume swelling, and brain pathology. The plasma and brain serotonin levels continued to remain high. These observations are the first to suggest that antiserum to serotonin when administered into the CSF during the early phase of CHI are capable of inducing neuroprotection.
Collapse
Affiliation(s)
- H S Sharma
- Department of Surgical Sciences, University Hospital, Uppsala University, SE-75421 Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
6
|
Verneris MR, Arshi A, Edinger M, Kornacker M, Natkunam Y, Karimi M, Karami M, Cao YA, Marina N, Contag CH, Negrin RS. Low levels of Her2/neu expressed by Ewing's family tumor cell lines can redirect cytokine-induced killer cells. Clin Cancer Res 2005; 11:4561-70. [PMID: 15958642 DOI: 10.1158/1078-0432.ccr-05-0157] [Citation(s) in RCA: 26] [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
PURPOSE To identify novel treatments for pediatric solid tumors and/or for malignancies with low-level Her2/neu expression. EXPERIMENTAL DESIGN Using fluorescence-activated cell sorting and immunohistochemistry, Her2/neu expression was determined on cell lines derived vfrom Ewing's family tumors (EFT) and neuroblastoma. Sensitivity to trastuzumab treatment was investigated using an in vitro proliferation assay. Cytotoxicity against EFT cell lines was done with either freshly isolated or ex vivo activated and expanded T cells (cytokine-induced killer cells, CIK cells), with or without addition of a CD3xHer2/neu bispecific antibody. The effects of either trastuzumab, CIK cells alone, or CD3xHer2/neu bispecific antibody redirected CIK cells was determined using a SCID/hu model of EFTs and serial, noninvasive bioluminescent imaging. RESULTS EFT cell lines express 5- to 10-fold lower levels of her2/neu than either breast (BT-474) or ovarian (SK-OV-3) cell lines. Treatment of EFT cell lines with trastuzumab did not induce growth inhibition either in vitro or in vivo. In contrast, Her2/neu could be used to redirect CIK cell to mediate cytotoxicity against EFTs both in vitro and in vivo (using two different treatment schemas). CONCLUSIONS CD3xHer2/neu bispecific antibody and CIK cells may be a suitable approach to treat malignancies with low-level Her2/neu expression not responsive to trastuzumab.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cytokines/pharmacology
- Cytotoxicity, Immunologic/drug effects
- Humans
- Immunohistochemistry
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Mice
- Mice, SCID
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
- Receptor, ErbB-2/metabolism
- Survival Analysis
- Trastuzumab
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- Michael R Verneris
- Department of Pediatrics, Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Schröder CP, Ruiters MHJ, de Jong S, Tiebosch ATMG, Wesseling J, Veenstra R, de Vries J, Hoekstra HJ, de Leij LFMH, de Vries EGE. Detection of micrometastatic breast cancer by means of real time quantitative RT-PCR and immunostaining in perioperative blood samples and sentinel nodes. Int J Cancer 2003; 106:611-618. [PMID: 12845661 DOI: 10.1002/ijc.11295] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The aim of our study was to detect micrometastatic breast cancer by epithelial glycoprotein-2 (EGP-2) and cytokeratin 19 (CK19), using immunostaining and real time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). Fifty-eight breast cancer patients, 52 primary tumors, 75 sentinel nodes (SN) and 149 peripheral blood (PB) samples (from before, during and 4 days after operation) were examined. Immunostaining was performed with antibodies directed against EGP-2 and CK19. Detection limits were one Michigan Cancer Foundation-7 (MCF-7) breast cancer cell line cell/2.10(6) leukocytes (immunostaining) and one MCF-7 cell/10(6) leukocytes qRT-PCR. Control noncancer lymph nodes (n = 10) showed nonspecific CK19 staining, but were qRT-PCR negative; control healthy volunteer PB (n = 11) was always negative. Primary tumor samples, all positive with immunostaining, showed a wide variation of EGP-2 (>10(4) fold) and CK19 mRNA expression (>10(3) fold). SN (n = 19) from 16 patients were tumor-positive with routine haematoxylin-eosin (H&E) and/or immunostaining. SN tumor presence was positively correlated to qRT-PCR expression, but 3 tumor-positive SN were false negative with qRT-PCR. Three SN were qRT-PCR positive, while tumor negative with H&E and/or immunostaining. No immunostaining positive PB was observed, but 19 patients (33%) had one or more qRT-PCR positive PB samples. We concluded that primary tumors have varying expressions of EGP-2 and CK19 mRNA. Both markers can be used in qRT-PCR to obtain adequate sensitivity for single tumor cell detection. In SN, immunostaining appears more sensitive/specific than H&E or qRT-PCR for tumor detection. No immunostaining positivity was found in PB, while 33% of patients had qRT-PCR positive PB. The clinical value of these findings will have to be clarified.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/blood
- Breast Neoplasms/diagnosis
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/blood
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Intraductal, Noninfiltrating/blood
- Carcinoma, Intraductal, Noninfiltrating/diagnosis
- Carcinoma, Intraductal, Noninfiltrating/secondary
- Carcinoma, Lobular/blood
- Carcinoma, Lobular/diagnosis
- Carcinoma, Lobular/secondary
- Case-Control Studies
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Differentiation
- Cell Nucleus/metabolism
- DNA Primers
- Epithelial Cell Adhesion Molecule
- Female
- Humans
- Keratins/genetics
- Keratins/metabolism
- Lymphatic Metastasis
- Middle Aged
- Neoplasm Staging
- Neoplastic Cells, Circulating/pathology
- RNA, Messenger/analysis
- RNA, Neoplasm/blood
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Sensitivity and Specificity
- Sentinel Lymph Node Biopsy
Collapse
Affiliation(s)
- Carolien P Schröder
- Department of Medical Oncology, University Hospital Groningen, The Netherlands
| | - Marcel H J Ruiters
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, The Netherlands
| | - Steven de Jong
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, The Netherlands
| | - Anton T M G Tiebosch
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, The Netherlands
| | - Jelle Wesseling
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, The Netherlands
| | - Rense Veenstra
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, The Netherlands
| | - Jaap de Vries
- Department of Surgical Oncology, University Hospital Groningen, The Netherlands
| | - Harold J Hoekstra
- Department of Surgical Oncology, University Hospital Groningen, The Netherlands
| | - Lou F M H de Leij
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, The Netherlands
| | | |
Collapse
|
8
|
Abstract
Bispecific monoclonal antibodies have drawn considerable attention from the research community due to their unique structure against two different antigens. The two-arm structure of bsMAb allows researchers to place a therapeutic agent on one arm while allowing the other to specifically target the disease site. The therapeutic agent can be a drug, toxin, enzyme, DNA, radionuclide, etc. Furthermore, bsMAb may redirect the cytotoxicity of immune effector cells towards the diseased cells or induce a systemic immune response against the target. BsMAb holds great promise for numerous therapeutic needs in the light of: (1) recent breakthroughs in recombinant DNA technology, (2) the increased number of identified disease targets as the result of the completion of human genomic map project, and (3) a better understanding of the mechanism of human immune system. This review focuses on therapeutic applications and production of bsMAb while providing the up-to-date clinical trial information.
Collapse
Affiliation(s)
- Ying Cao
- Abbott Laboratories, Dept. 04A6, Bldg. AP8B, 100 Abbott Park Road, Abbott Park, IL 60064, USA.
| | | |
Collapse
|
9
|
Kroesen BJ, McLaughlin PMJ, Schuilenga-Hut PHL, Jacobs SC, Molema G, Helfrich W, De Leij LFMH. Tumor-targeted immune complex formation: effects on myeloid cell activation and tumor-directed immune cell migration. Int J Cancer 2002; 98:857-63. [PMID: 11948463 DOI: 10.1002/ijc.10245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The effectiveness of cellular immunotherapy of solid tumors is often hampered by the lack of specific infiltration of immune effector cells into the tumor mass. Therefore, we studied the potential of tumor antigen-specific antibodies to elicit tumor-specific myeloid cell activation, to induce or enhance tumor infiltration by immune cells. To this end, we developed an in vitro model system using the human myeloid cell line MonoMac-6. Incubation of IFN-gamma-primed MonoMac-6 cells with serum-opsonized zymosan or EGP-2-directed, mouse IgG2a-opsonized, EGP-2-positive tumor cells resulted in the production of ROS and TNF-alpha and induced E-selectin and ICAM-1 expression on HUVECs. FcR-mediated MonoMac-6 cell activation was strictly dependent on the activation of MonoMac-6 cells with IFN-gamma. In addition, no myeloid cell activation was observed in the presence of human serum or using tumor antigen-specific mouse antibody subclasses other than IgG2a, suggesting the crucial involvement of CD64 (FcgammaR1) in the effects observed. However, serum-inhibited myeloid cell activation was completely restored employing a 2-step targeting approach in which tumor cell opsonization with mouse anti-EGP-2 antibodies was followed by incubation with human antimouse Ig antibodies. Moreover, using this 2-step approach, not only anti-EGP-2-directed mouse IgG2a but also mouse IgG1 antibodies effectively induced tumor-specific myeloid cell activation. In conclusion, we describe a method to induce efficient and tumor-specific activation of myeloid cells based on the sequential use of mouse tumor antigen-specific and human antimouse Ig antibodies. Targeted myeloid cell activation may provide a means to aid in the induction of a tumor-directed immune response and as such, the method described here could be of clinical significance.
Collapse
Affiliation(s)
- Bart-Jan Kroesen
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
10
|
Sen M, Wankowski DM, Garlie NK, Siebenlist RE, Van Epps D, LeFever AV, Lum LG. Use of anti-CD3 x anti-HER2/neu bispecific antibody for redirecting cytotoxicity of activated T cells toward HER2/neu+ tumors. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2001; 10:247-60. [PMID: 11359672 DOI: 10.1089/15258160151134944] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Relapse after adjuvant chemotherapy or high-dose chemotherapy with stem cell transplant for high-risk breast cancer remains high and new strategies that provide additional antitumor effects are needed. This report describes methods to generate highly effective HER2/neu-specific cytotoxic T cells by arming activated T cells with anti-CD3 x anti-HER2/neu bispecific antibody (BsAb). OKT3 and 9184 (anti-HER2) monoclonal antibodies (mAb) were conjugated and used to arm T cells that were subsequently tested in binding, cytotoxicity, and cytokine secretion assays. Armed T cells aggregated and specifically killed HER2/neu(+) breast cancer cells. Cytotoxicity emerged after 6 days of culture, was higher in armed T cells than unarmed T cells at all effector to target ratios (E/T) tested, and increased as the arming dose was increased. At an E/T of 20:1, the mean cytotoxicity of armed activated T cells (ATC) from 10 normal subjects increased by 59 +/- 11% (+/-SD) over that seen in unarmed ATC (p < 0.001) and the mean cytotoxicity of armed ATC from 6 cancer patients increased by 32 +/- 9% above that seen for unarmed ATC (p < 0.0004). After arming, the BsAb persisted on ATC up to 72 h and armed ATC continued to be cytotoxic up to 54 h. The amount of interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha), and granulocyte-macrophage colony-stimulating factor (GM-CSF) secreted was 1699, 922, and 3092 pg/ml/10(6) cells per 24 h, respectively, when armed T cells were exposed to a HER2/neu(+) breast carcinoma cell line. These studies show the feasibility and clinical adaptability of this approach for generating large numbers of anti-HER2-specific, cytotoxic T cells for clinical trials.
Collapse
Affiliation(s)
- M Sen
- Blood Center of Southeastern Wisconsin, Milwaukee, WI 53201, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Withoff S, Bijman MN, Stel AJ, Delahaye L, Calogero A, Jonge MW, Kroesen BJ, Leij L. Characterization of BIS20x3, a bi-specific antibody activating and retargeting T-cells to CD20-positive B-cells. Br J Cancer 2001; 84:1115-21. [PMID: 11308263 PMCID: PMC2363855 DOI: 10.1054/bjoc.2000.1707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This paper describes a bi-specific antibody, which was called BIS20x3. It retargets CD3varepsilon-positive cells (T-cells) to CD20-positive cells and was obtained by hybrid-hybridoma fusion. BIS20x3 could be isolated readily from quadroma culture supernatant and retained all the signalling characteristics associated with both of its chains. Cross-linking of BIS20x3 on Ramos cells leads to DNA fragmentation percentages similar to those obtained after Rituximab-cross-linking. Cross-linking of BIS20x3 on T-cells using cross-linking F(ab')2-fragments induced T-cell activation. Indirect cross-linking of T-cell-bound BIS20x3 via Ramos cells hyper-activated the T-cells. Furthermore, it was demonstrated that BIS20x3 effectively re-targets T-cells to B-cells, leading to high B-cell cytotoxicity. The results presented in this paper show that BIS20x3 is fully functional in retargeting T-cells to B-cells and suggest that B-cell lymphomas may represent ideal targets for T-cell retargeting bi-specific antibodies, because the retargeted T-cell is maximally stimulated in the presence of B-cells. Additionally, since B-cells may up-regulate CD95/ Fas expression upon binding of CD20-directed antibodies, B-cells will become even more sensitive for T-cell mediated killing via CD95L/ Fas L, and therefore supports the intention to use T-cell retargeting bi-specific antibodies recognizing CD20 on B-cell malignancies as a treatment modality for these diseases.
Collapse
Affiliation(s)
- S Withoff
- Department of Pathology and Laboratory Medicine, University Hospital Groningen, Section Medical Biology - Laboratory Tumor Immunology, Hanzeplein 1, GZ Groningen, 9713, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Molema G, Kroesen BJ, Helfrich W, Meijer DK, de Leij LF. The use of bispecific antibodies in tumor cell and tumor vasculature directed immunotherapy. J Control Release 2000; 64:229-39. [PMID: 10640660 DOI: 10.1016/s0168-3659(99)00137-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To overcome dose limiting toxicities and to increase efficacy of immunotherapy of cancer, a number of strategies are under development for selectively redirecting effector cells/molecules towards tumor cells. Many of these strategies exploit the specificity of tumor associated antigen recognition by monoclonal antibodies. Using either hybridoma fusion, chemical derivatization or molecular biology technology, antibodies with dual specificity can be constructed. These so called biospecific antibodies (BsAbs) have been used to redirect the cytolytic activity of a variety of immune effector cells such as cytotoxic T lymphocytes, natural killer cells, neutrophils and monocytes/macrophages to tumor cells. Local administration of BsAbs, either alone or in combination with autologous effector cells, is highly effective in eradicating tumor cells. In contrast, systemic application of BsAb at present is only suitable for adjuvant treatment of minimal residual disease due to poor tumor cell accessibility. As an alternative, angiogenesis related determinants on tumor blood vessels can be exploited for the selective delivery of effector cells/molecules apart from being used to inhibit angiogenesis. Important advantages of this strategy is that the endothelial cell associated target epitope(s) are easy accessible. The dependence of tumor growth on the tumor's blood supply also renders tumor endothelial cells an attractive target for therapy. Although still in its infancy, attacking the tumor's blood supply for example by delivering coagulation factors or toxins, or by BsAb directed immunotherapies holds great promise for antineoplastic therapy.
Collapse
Affiliation(s)
- G Molema
- Dept. Clinical Immunology, Groningen University Institute for Drug Exploration (GUIDE), Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
13
|
Ren-Heidenreich L, Hayman GT, Trevor KT. Specific targeting of EGP-2+ tumor cells by primary lymphocytes modified with chimeric T cell receptors. Hum Gene Ther 2000; 11:9-19. [PMID: 10646635 DOI: 10.1089/10430340050016111] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A promising strategy for cancer treatment is adoptive immunotherapy with gene-modified lymphocytes expressing a chimeric T cell receptor (cTCR) that directs tumor targeting and stimulates T cell effector functions. In this study, the activities of two novel cTCR molecules (GAgamma and GAHgamma) were investigated. Both encode a single-chain variable fragment (scFv) derived from the monoclonal antibody (MAb) GA733.2, which binds the epithelial glycoprotein 2 (EGP-2) overexpressed on a variety of human carcinomas. In the GAgamma cTCR, the scFv is directly fused to the transmembrane/cytoplasmic portions of the immunoglobulin Fc receptor (Ig FcRI) gamma subunit, which mediates T cell signaling. GAHgamma possesses an extracellular spacer composed of the CD8alpha immunoglobulin hingelike domain inserted between the scFv and gamma chain. Activated T cells (ATCs), stimulated ex vivo using anti-CD3 MAb, were derived from either healthy donors or patients and transduced with recombinant retrovirus encoding the respective GA cTCR molecules. After culture expansion for 14 days, GAgamma-modified ATCs demonstrated enhanced targeting and lysis of EGP-2+ colon cancer cells and increased cytokine secretion. Cells transduced with the GAHgamma cTCR displayed specific lytic activity that was about twofold greater than that of GAgamma-ATCs and produced significantly more cytokine. In addition, reactivation of GAHgamma-ATC with anti-CD3 MAb prior to addition to EGP-2+ tumor target induced a further increase in lytic activity. Because the activation status influences T cell antitumor functions, our data suggest that reactivation prior to adoptive transfer would improve the clinical efficacy of GAHgamma-modified ATCs.
Collapse
Affiliation(s)
- L Ren-Heidenreich
- Vince Lombardi Gene Therapy Laboratory, Immunotherapy Research and Treatment Institute, St. Luke's Medical Center, Milwaukee, WI 53215, USA
| | | | | |
Collapse
|
14
|
Molema G, Meijer DK, de Leij LF. Tumor vasculature targeted therapies: getting the players organized. Biochem Pharmacol 1998; 55:1939-45. [PMID: 9714313 DOI: 10.1016/s0006-2952(98)00011-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Based on their location and central role in solid tumor growth, tumor vascular endothelial cells may present an attractive target for the delivery of therapeutic drugs or cells. The potency of blocking the tumor blood supply in eradicating solid tumors was demonstrated recently in a mouse model of tumor vasculature targeting (Huang et al., Science 275: 547-550, 1997). For clinical application of such strategies, tumor endothelium specific target epitopes need to be identified. Recent studies on angiogenesis have identified angiogenesis-related molecules as potential target epitopes. Among these are vascular endothelial growth factor (VEGF)/VEGF-receptor complex, alpha(v) integrins, and Tie receptor tyrosine kinases. Besides blockade of their signalling cascades leading to inhibition of angiogenesis, these epitopes may also be instrumental in tumor vessel specific delivery of therapeutics. Data on the efficacy of therapeutic modalities aimed at these, mostly heterogeneously distributed tumor endothelial epitopes are scarce, and sophisticated experimentation is required to rationalize the development of new therapeutic strategies. Importantly, only detailed evaluations in cancer patients will provide the blueprint for the development of clinically effective tumor vascular targeted therapies.
Collapse
Affiliation(s)
- G Molema
- Department of Clinical Immunology, Groningen Utrecht Institute for Drug Exploration, The Netherlands.
| | | | | |
Collapse
|