1
|
Antibody–cytokine fusion proteins. Arch Biochem Biophys 2012; 526:194-205. [DOI: 10.1016/j.abb.2012.03.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 01/01/2023]
|
2
|
Xiang J, Munegowda MA, Deng Y. Transgene expression of alpha tumor necrosis factor with mutations D142N and A144R under control of human telomerase reverse transcriptase promoter eradicates well-established tumors and induces long-term antitumor immunity. Cancer Gene Ther 2008; 16:430-8. [PMID: 19096444 DOI: 10.1038/cgt.2008.94] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant adenoviral vectors (AdVTNF-alpha) expressing alpha tumor necrosis factor (TNF-alpha) under control of cytomegalovirus (CMV) promoter have been used in cancer gene therapy. To reduce its cytotoxicity, we constructed a recombinant AdV(TERT)mTNF-alpha expressing a mutant TNF-alpha (mTNF-alpha) with mutations at D142N and A144R under control of human telomerase reverse transcriptase (hTERT) promoter for treatment of well-established ovalbumin (OVA)-expressing murine B16 melanoma (BL6-10(OVA)) (6 mm in diameter). We demonstrated that the mTNF-alpha with mutations at D142N and A144R has less in vitro cytotoxicity, but maintains its functional effect in the stimulation of T-cell proliferation. The in vitro and in vivo transgene expressions under control of hTERT promoter are highly restricted in tumor cells compared with those under the control of the CMV promoter. AdV(TERT)mTNF-alpha gene therapy by intratumoral injection of AdV(TERT)mTNF-alpha vector (2 x 10(9) PFU) expressing the mutant mTNF-alpha under control of hTERT promoter reduces its in vivo toxicity, eradicates well-established BL6-10(OVA) tumors in 4/10 tumor-bearing mice, and induces OVA-specific CD8(+) T-cell-mediated long-term antitumor immunity. Therefore, AdV(TERT)mTNF-alpha gene therapy may be very useful in the immunotherapy of cancer.
Collapse
Affiliation(s)
- J Xiang
- Cancer Research Unit, Saskatchewan Cancer Agency, Departments of Oncology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | | | | |
Collapse
|
3
|
Bauer S, Adrian N, Fischer E, Kleber S, Stenner F, Wadle A, Fadle N, Zoellner A, Bernhardt R, Knuth A, Old LJ, Renner C. Structure-activity profiles of Ab-derived TNF fusion proteins. THE JOURNAL OF IMMUNOLOGY 2006; 177:2423-30. [PMID: 16888004 DOI: 10.4049/jimmunol.177.4.2423] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TNF application in humans is limited by severe side effects, including life-threatening symptoms of shock. Therefore, TNF can be successfully applied as a tumor therapeutic reagent only under conditions that prevent its systemic action. To overcome this limitation, genetic fusion of TNF to tumor-selective Abs is a favored strategy to increase site-specific cytokine targeting. Because wild-type TNF displays its bioactivity as noncovalently linked homotrimer, the challenge is to define structural requirements for a TNF-based immunokine format with optimized structure-activity profile. We compared toxicity and efficacy of a dimerized CH2/CH3 truncated IgG1-TNF fusion protein and a single-chain variable fragment-coupled TNF monomer recognizing fibroblast-activating protein. The former construct preserves its dimeric structure stabilized by the natural disulfide bond IgG1 hinge region, while the latter trimerizes under native conditions. Analysis of complex formation of wild-type TNF and of both fusion proteins with TNFR type 1 (TNF-R1) using surface plasmon resonance correlated well with in vitro and in vivo toxicity data. There is strong evidence that TNF subunits in a trimeric state display similar toxicity profiles despite genetic fusion to single-chain variable fragment domains. However, LD(50) of either immunodeficient BALB/c nu/nu or immunocompetent BALB/c mice was significantly decreased following administration of TNF in the formation of IgG1-derived dimeric fusion protein. Reduction of unspecific peripheral complexation of TNF-R1 resulted in higher anticancer potency by immunotargeting of fibroblast-activating protein-expressing xenografts. The broader therapeutic window of the IgG1-derived TNF fusion protein favors the dimeric TNF-immunokine format for systemic TNF-based tumor immunotherapy.
Collapse
Affiliation(s)
- Stefan Bauer
- Oncology Department, Department of Internal Medicine, UniversitätsSpital Zürich, Rämistrasse 100, CH-8091 Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Liu Y, Zhang W, Cheung LH, Niu T, Wu Q, Li C, Van Pelt CS, Rosenblum MG. The antimelanoma immunocytokine scFvMEL/TNF shows reduced toxicity and potent antitumor activity against human tumor xenografts. Neoplasia 2006; 8:384-93. [PMID: 16790087 PMCID: PMC1592449 DOI: 10.1593/neo.06121] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The immunocytokine scFvMEL/TNF, a fusion protein composed of human tumor necrosis factor (TNF) and a single-chain Fv antibody (scFv) scFvMEL targeting the melanoma gp240 antigen, demonstrates impressive cytotoxic effects against human melanoma cell lines in vitro. Pharmacokinetic studies of 125I-scFvMEL/TNF in BALB/c mice showed that the construct clears from the circulation with a terminal-phase half-life of 17.6 hours after intravenous administration. The maximum tolerated dose of scFvMEL/TNF in nude mice was 4 mg/kg, i.v., on a daily x5 schedule. There were no changes in gross pathology, clinical chemistry, or hematologic parameters in mice treated at doses of up to 3 mg/kg. Therapeutic studies at a dose of 2.5 mg/kg on athymic mice bearing established (approximately 50 mm3) human melanoma A375GFP xenograft tumors transfected with green fluorescent protein demonstrated potent tumor suppression and complete tumor regression of all lesions. There was no subsequent outgrowth of tumors from mice rendered tumor-free. These data show that scFvMEL/TNF can target melanoma cells in vivo and can result in pronounced antimelanoma effects after systemic administration. Toxicology studies indicate the relative safety of this agent at doses that are therapeutically effective and provide guidance to projected phase I starting doses on patients at this schedule.
Collapse
Affiliation(s)
- Yuying Liu
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Chan T, Sami A, El-Gayed A, Guo X, Xiang J. HER-2/neu-gene engineered dendritic cell vaccine stimulates stronger HER-2/neu-specific immune responses compared to DNA vaccination. Gene Ther 2006; 13:1391-402. [PMID: 16724093 DOI: 10.1038/sj.gt.3302797] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
HER-2/neu is a candidate for developing breast cancer-targeted immunotherapeutics. Although DNA-based and HER-2/neu transgene-modified dendritic cell (DC)-based vaccines are potent at eliciting HER-2/neu-specific antitumor immunity, there has been no side-by-side study comparing them directly. The present study utilizes an in vivo murine tumor model expressing HER-2/neu antigen to compare the efficacy between adenovirus (AdVneu)-transfected dendritic cells (DC(neu)) and plasmid DNA (pcDNAneu) vaccine. Our data showed that DC(neu) upregulated the expression of immunologically important molecules and inflammatory cytokines and partially converted regulatory T (Tr)-cell suppression through interleukin-6 (IL-6) secretion. Vaccination of DC(neu) induced stronger HER-2/neu-specific humoral and cellular immune responses than DNA vaccination, which downregulated HER-2/neu expression and lysed HER-2/neu-positive tumor cells in vitro, respectively. In two HER-2/neu-expressing tumor models, DC(neu) completely protected mice from tumor cell challenge compared to partial or no protection observed in DNA-immunized mice. In addition, DC(neu) significantly delayed breast cancer development in transgenic mice in comparison to DNA vaccine (P<0.05). Taken together, we have demonstrated that HER-2/neu-gene-modified DC vaccine is more potent than DNA vaccine in both protective and preventive animal tumor models. Therefore, DCs genetically engineered to express tumor antigens such as HER-2/neu represent a new direction in DC vaccine of breast cancer.
Collapse
Affiliation(s)
- T Chan
- Cancer Research Unit, Saskatchewan Cancer Agency, University of Saskatchewan, 20 Campus Drive, Saskatoon, Saskatchewan, Canada S7N 4H4
| | | | | | | | | |
Collapse
|
6
|
Liu Y, Cheung LH, Marks JW, Rosenblum MG. Recombinant single-chain antibody fusion construct targeting human melanoma cells and containing tumor necrosis factor. Int J Cancer 2004; 108:549-57. [PMID: 14696119 DOI: 10.1002/ijc.11524] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Fusion constructs targeting tumor cells have significant potential applications against both solid tumors and hematologic malignancies. We developed a fusion construct of tumor necrosis factor (TNF) and a single-chain antibody (scFvMEL) recognizing the gp240 antigen on human melanoma cells. The scFvMEL/TNF construct, like TNF itself, was found to exist in solution primarily as a trimer of 45 kDa monomers (trimeric molecular weight = 135 kDa). The fusion construct bound specifically to gp240 antigen-positive but not to antigen-negative cells. The TNF component of the construct was biologically active (specific activity = 1 x 10(7) U/mg) compared with free TNF (specific activity = 2.6 x 10(7) U/mg) and was more cytotoxic to antigen-positive A375-M melanoma cells (IC(50) = 100 pM) than TNF alone (IC(50) = 1,000 pM) and, additionally, was active against AAB-527 melanoma cells (IC(50) = 20 nM) resistant to TNF itself (IC(50) > 1,000 nM). The augmented cytotoxicity was mediated by antibody-specific binding to the cell surface. Both A375-M and AAB-527 cells were shown to express TNFR1 and TNFR2 on the cell surface. The TNF moiety of the fusion construct was efficiently delivered into cells in time-dependent increase in cytosol as assessed by immunofluorescent staining of human melanoma cells. Radiolabeled scFvMEL/TNF localized effectively in human melanoma xenografts in nude (nu/nu) mice with a tumor:blood ratio of approximately 8 at 72 hr after administration. Our studies suggest that because of its unique biologic activity and low antigenic potential, scFvMEL/TNF makes an excellent cytotoxic protein for potential clinical treatment of human melanoma.
Collapse
Affiliation(s)
- Yuying Liu
- Immunopharmacology and Targeted Therapy Section, Department of Bioimmunotherapy, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
7
|
Hamilton S, Odili J, Gundogdu O, Wilson GD, Kupsch JM. Improved production by domain inversion of single-chain Fv antibody fragment against high molecular weight proteoglycan for the radioimmunotargeting of melanoma. HYBRIDOMA AND HYBRIDOMICS 2002; 20:351-60. [PMID: 11839253 DOI: 10.1089/15368590152740752] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Melanoma is among the few cancers with rising incidence. Currently there is no effective treatment for metastatic disease, but improved detection of melanoma has the potential to benefit the management of patients with early disease. Radioimmunodection by imaging with single-chain Fv (scFv) antibody fragments is one such emerging diagnostic method. However, the amount of scFv that can be produced at a scale suitable for use in patients is limiting. We have previously shown that the bacterial expression of a scFv derived from a monoclonal antibody (MAb) specific for melanoma-associated proteoglycan can be increased by light chain shuffling. In this report we show that a further increase in expression yield can be obtained by reversing the usual V(H)-V(L) orientation of scFvs to V(L)-V(H). Such seemingly minor changes have previously been reported to have unexpected effects on the in vitro and in vivo binding properties of recombinant antibodies. Our results show that reversal of the V domain orientation of the scFv improves expression by 150% without an adverse effect on melanoma binding in vitro and tumor targeting in vivo. Therefore, our results show that alteration of V domain orientation can improve the production yield of clinically useful antibody fragments. When used in combination with other antibody engineering approaches for increased antibody production changing the domain orientation is a simple strategy to achieve significant improvements in the production of scFvs for tumor radioimmunodetection for patient studies.
Collapse
Affiliation(s)
- S Hamilton
- RAFT Institute of Plastic Surgery and Cancer Research Trust, Mount Vernon Hospital, Northwood, Middlesex HA6 2RN, UK
| | | | | | | | | |
Collapse
|
8
|
Kriangkum J, Xu B, Nagata LP, Fulton RE, Suresh MR. Bispecific and bifunctional single chain recombinant antibodies. BIOMOLECULAR ENGINEERING 2001; 18:31-40. [PMID: 11535414 DOI: 10.1016/s1389-0344(01)00083-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bispecific and bifunctional monoclonal antibodies as second generation monoclonals, produced by conventional chemical or somatic methods, have proved useful in the immunodiagnosis and immunotherapy of cancer and other diseases. Recombinant antibodies produced by genetic engineering techniques have also become available for use in preclinical and clinical studies. Furthermore, through genetic engineering, it is possible to remove or add on key protein domains in order to create designer antibody molecules with two or more desired functions. This review summarizes the strategies for development of single chain variable fragment (scFv) bifunctional and bispecific antibodies. The advantages and disadvantages as well as the problems of generating the various bispecific and bifunctional antibody constructs are reported and discussed. Since conventionally prepared bispecific and bifunctional monoclonal antibodies have already shown promise in clinical trials and results from preclinical studies of recombinant bispecific antibodies are encouraging, clinical trials in humans of recombinant bispecific and bifunctional antibodies, as a new generation of biologicals, are likely to be the thrust in the next decade and beyond.
Collapse
Affiliation(s)
- J Kriangkum
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2N8
| | | | | | | | | |
Collapse
|
9
|
Abstract
Tumour Necrosis Factor (TNF) is a cytokine initially discovered for its capacity to induce haemorrhagic necrosis of experimental tumours and later found to be endowed with potent proinflammatory activities. It was soon realised that these latter properties were at the origin of unacceptable systemic toxicity in all trials aimed at exploiting the anti-tumour activities of TNF. The present review intends to reconsider the efforts that have been devoted over the past ten years to increase the therapeutic index of TNF so to make it a useful drug for the treatment of malignancies. Overall, attempts to achieve this goal with systemically administered TNF have met little success so far. On the other hand, impressive results have been obtained with locoregional administration of TNF. Although of relatively limited clinical utility, these observations have indicated a realistic possibility for a therapeutic exploitation of TNF in tumour therapy: the delivery of systemically administered TNF to the site of tumour growth. On this basis, different targeting and pre-targeting strategies have been developed to achieve this goal. While still in their infancy, these approaches have yielded encouraging results in experimental tumour models. In the forthcoming years it will be possible to evaluate if they represent a practicable means of delivering high doses of TNF to the tumour while sparing the organism from systemic, toxic effects.
Collapse
Affiliation(s)
- A Corti
- Department of Biological and Technological Research, San Raffaele H Scientific Institute, Milan, Italy
| | | |
Collapse
|
10
|
Yang J, Moyana T, MacKenzie S, Xia Q, Xiang J. One hundred seventy-fold increase in excretion of an FV fragment-tumor necrosis factor alpha fusion protein (sFV/TNF-alpha) from Escherichia coli caused by the synergistic effects of glycine and triton X-100. Appl Environ Microbiol 1998; 64:2869-74. [PMID: 9687443 PMCID: PMC106785 DOI: 10.1128/aem.64.8.2869-2874.1998] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
To target tumor necrosis factor alpha (TNF-alpha) to tumor cells, recombinant DNA techniques were used to construct and express the fused gene VKLVH-TNF-alpha, which encodes the secreted form of single-chain fusion protein sFV/TNF-alpha in Escherichia coli. sFV/TNF-alpha was secreted into the culture medium and purified by affinity chromatography. The production of the fusion protein in the culture medium under the optimal conditions of 30 degrees C and 37 micromol of isopropyl-beta-D-thiogalactopyranoside (IPTG) per liter was 16- and 5-fold higher than that under the standard conditions of 37 degrees C and 1 mmol of IPTG per liter. Fusion protein excretion into culture medium with 2% glycine, 1% Triton X-100, or both of these two chemicals was either 14-, 38-, or 170-fold higher, respectively than that without the two chemicals. The final yield of sFV/TNF-alpha was estimated to be 50 mg/liter. The loss of integrity of the cellular membrane may be a potential mechanism for enhancement of fusion protein production and excretion by treatment with glycine and Triton X-100. This study thus provides a practical, large-scale method for more efficient production of the heterologous fusion protein sFV/TNF-alpha in E. coli by using glycine and Triton X-100.
Collapse
Affiliation(s)
- J Yang
- Departments of Microbiology, Saskatoon Cancer Center, College of Medicine, University of Saskatchewan, Canada
| | | | | | | | | |
Collapse
|
11
|
Pietersz GA, Toohey B, McKenzie IF. In vitro and in vivo evaluation of human tumor necrosis factor-alpha (hTNFalpha) chemically conjugated to monoclonal antibody. J Drug Target 1998; 5:109-20. [PMID: 9588867 DOI: 10.3109/10611869808995864] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human tumor necrosis-alpha (hTNF-alpha) was chemically conjugated to the murine anti-Ly-2.1 T cell antibody using heterobifunctional crosslinking agents SAMSA and SPDP. SDS-PAGE analysis of the affinity purified conjugate consisted mainly of 1:1 and 1:2 (Ly-2.1:TNF) complexes. Conjugated hTNF retained 50% of its cytotoxic activity by the L929 cytolytic assay, with an IC50 = 0.12 ng/ml. hTNF-Ly-2.1 was also cytotoxic to E3 cells (Ly-2.1+ve) with an IC50 = 1.7 microg/ml - 3 times more cytotoxic to these cells than non-conjugated hTNF in vitro. However in vivo hTNF-Ly-2.1 conjugates were more toxic to mice than hTNF. In vivo blood clearance studies in E3 tumor bearing CBF1 mice demonstrated that the half life of the conjugate was 2 hr, compared to 20 min for hTNF. In biodistribution studies, tumor accumulation of 3% was seen for hTNF-Ly-2.1 while for unconjugated hTNF no activity in tumor was detected 24hr post injection. A single dose of hTNF-Ly-2.1 increased the accumulation of 125I-anti-Ly-2.1 by 3 fold compared to controls. However, the antitumor effect of hTNF-Ly-2.1 on E3 cells in vivo was marginal with some tumor growth retardation at day 1-3. The results of these in vitro and in vivo studies on chemically conjugated h-TNF-MoAb will be helpful in the design of novel recombinant fusion proteins for targeting the biologic activity of TNF to tumours.
Collapse
Affiliation(s)
- G A Pietersz
- The Austin Research Institute, Austin Hospital, Heidelberg Vic, Australia
| | | | | |
Collapse
|
12
|
Hoogenboom HR, Henderikx P. Creating and engineering human antibodies for immunotherapy. Adv Drug Deliv Rev 1998; 31:5-31. [PMID: 10837615 DOI: 10.1016/s0169-409x(97)00091-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Targeting in immunotherapy has traditionally been achieved by using monoclonal rodent antibodies. Despite gene-engineering, there are many problems and limitations associated with the non-human origin, the targeting specificity and the binding strength of these molecules. Now these issues may be addressed in a more rational way, by designing and then shaping, in vitro, the desired human antibodies. This review addresses how this may be achieved by the selection of monoclonal human antibodies from phage display libraries and the engineering of affinity and specificity thereafter. Phage display of antibody fragments has allowed access to large collections of different phage antibodies, created by cloning antibody V-genes from B-cells. Antibodies against any type of antigen may be derived from such repertoires, by rounds of enrichment on antigen and re-amplification. This review presents the state of the art in rational antibody design and creation. It will highlight the strengths of this increasingly important field, which will aid in the generation of tailor-made targeting entities for immunotherapy.
Collapse
Affiliation(s)
- HR Hoogenboom
- CESAME at Department of Pathology, University Hospital Maastricht, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | | |
Collapse
|
13
|
Song Z, Cai Y, Song D, Xu J, Yuan H, Wang L, Zhu X, Lin H, Breitling F, Dübel S. Primary structure and functional expression of heavy- and light-chain variable region genes of a monoclonal antibody specific for human fibrin. Hybridoma (Larchmt) 1997; 16:235-41. [PMID: 9219033 DOI: 10.1089/hyb.1997.16.235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The immunoglobulin heavy- and light-chain variable region (VH and VK) genes were isolated from 8E5 hybridoma cells, which secreted monoclonal antibody against human fibrin by RT-PCR. An expression vector pOPE51-8E5 was constructed for the recombinant VH-VK scFv expression. The primary sequence of the variable regions was determined. Expression product was found in the periplasmic space and inclusion bodies by SDS-PAGE and immunoblotting. It was a 30 KDa single chain fragment (scFv) with the antigen-binding specificity of the parental monoclonal antibody. A light chain shuffling with an unspecific VL did not result in a loss of fibrin binding specificity.
Collapse
Affiliation(s)
- Z Song
- Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Xiang J, Moyana T, Qi Y. Genetic engineering of a recombinant fusion possessing anti-tumor F(ab')2 and tumor necrosis factor. J Biotechnol 1997; 53:3-12. [PMID: 9165756 DOI: 10.1016/s0168-1656(96)01639-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The construction, synthesis and expression of a genetically engineered bifunctional antibody/cytokine fusion protein is described. In order to target alpha-tumor necrosis factor (TNF) to tumor cells, recombinant antibody techniques were used to construct an RM4/TNF fusion protein containing the chimeric anti-tumor F(ab')2 (RM4) as well as the TNF moiety. The recombinant cDNA of human TNF was linked to the 3' end of the chimeric heavy-chain gene fragment (M4) containing the VH, the CH1 and the hinge region to form the fused heavy-chain gene fragment M4-TNF. Transfection of the M4-TNF gene fragment into a VKCK cell line producing the chimeric light-chain of the same antibody allowed the transfectant secreting the bifunctional fusion protein RM4/TNF. The RM4/TNF was purified by affinity chromatography. Our data showed that RM4/TNF retained the TAG72 antigen-binding reactivity as well as TNF activity as measured by ELISA, Western blotting, flow cytometry analysis, immunohistochemistry and cytotoxicity assays using the human colon cancer cell line LS174T. Therefore, the bifunctional fusion protein RM4/TNF may prove useful in targeting the biological effects of TNF to tumor cells, and in this way stimulate the immune destruction of tumor cells.
Collapse
Affiliation(s)
- J Xiang
- Saskatoon Cancer Center, Department of Microbiology, University of Saskatchewan, Canada
| | | | | |
Collapse
|
15
|
Hayden MS, Grosmaire LS, Norris NA, Gilliland LK, Winberg G, Tritschler D, Tsu TT, Linsley PS, Mittler RS, Senter PD, Fell HP, Ledbetter JA. Costimulation by CD28 sFv expressed on the tumor cell surface or as a soluble bispecific molecule targeted to the L6 carcinoma antigen. TISSUE ANTIGENS 1996; 48:242-54. [PMID: 8946677 DOI: 10.1111/j.1399-0039.1996.tb02642.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Interaction of the CD80 (B7-1) and CD86 (B7-2) molecules on antigen presenting cells with the receptors CD28 and CTLA-4 on T cells generates signals important in the regulation of immune responses. Because this receptor system involves multiple receptor-ligand interactions, determining the function for individual receptors has been difficult. One approach is the use of antibodies and their derivatives with singular specificity as substitute ligands to explore the activities of these molecules. We have constructed recombinant mono- and bi-specific sFv molecules specific for the CD28 receptor that are capable of binding and generating costimulatory signals to activate T cells. We demonstrate that these soluble molecules are capable of higher levels of costimulation than soluble CD80Ig at equivalent concentrations. We also constructed artificial adhesion receptors on the cell surface using two different CD28-specific sFvIgs fused to the CD80 cytoplasmic and transmembrane domains. In this report, we compared costimulation by a soluble bispecific (alpha CD28-alpha L6) single chain sFvIg fusion protein to that generated by L6 antigen positive (L6+) H3347 tumor cells transduced with cell surface expressed forms of alpha CD28 sFv's. We show that the bispecific protein can target potent CD28 costimulatory activity to L6+ tumor cells in vitro. We also show that transfection of the cell surface forms of the two different CD28 sFvIgs into H3347 tumor cells allows them to generate significant costimulatory signals to activated T cells. Finally, we demonstrate that tumor cell presentation of either the soluble bispecific or transduced cell surface sFv generate similar costimulatory effects resulting in T cell activation.
Collapse
Affiliation(s)
- M S Hayden
- Bristol-Myers Squibb Pharmaceutical Research Institute, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|