1
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
2
|
Shin J, Phelan PJ, Gjoerup O, Bachovchin W, Bullock PA. Characterization of a single chain variable fragment of nivolumab that targets PD-1 and blocks PD-L1 binding. Protein Expr Purif 2021; 177:105766. [PMID: 32987122 PMCID: PMC7518118 DOI: 10.1016/j.pep.2020.105766] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/15/2020] [Accepted: 09/19/2020] [Indexed: 12/28/2022]
Abstract
Activated T-cells express Programmed cell Death protein 1 (PD-1), a key immune checkpoint receptor. PD-1 functions primarily in peripheral tissues, where T cells may encounter tumor-derived immunosuppressive ligands. Monoclonal antibodies that disrupt the interaction between T-cell derived PD-1 and immunosuppressive ligands, such as PD-L1, have revolutionized approaches to cancer therapy. For instance, Nivolumab is a monoclonal Ab that targets human PD-1 and has played an important role in immune checkpoint therapy. Herein we report the purification and initial characterization of a ~27 kDa single chain variable fragment (scFv) of Nivolumab that targets human PD-1 and blocks binding by PD-L1. The possibility that the anti-PD-1 scFv can serve as both an anti-tumor agent and as an anti-viral agent is discussed. IMPORTANCE: The clinical significance of anti-PD-1 antibodies for treatment of a range of solid tumors is well documented (reviewed in [1-4]). In this report, we describe the results of studies that establish that an anti-PD-1 scFv purified from E. coli binds tightly to human PD-1. Furthermore, we demonstrate that upon binding, the anti-PD-1 scFv disrupts the interaction between PD-1 and PD-L1. Thus, the properties of this scFv, including its small size, stability and affinity for human PD-1, suggest that it has the potential to be a useful reagent in subsequent immunotherapeutic, diagnostic and anti-viral applications.
Collapse
Affiliation(s)
- Jong Shin
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Paul J Phelan
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Ole Gjoerup
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
| | - William Bachovchin
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Peter A Bullock
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA.
| |
Collapse
|
3
|
Kim EJ, Kim BS, Choi DB, Chi SG, Choi TH. Improved In Vivo Stability of Radioiodinated Rituximab Using an Iodination Linker for Radioimmunotherapy. Cancer Biother Radiopharm 2016; 31:287-294. [PMID: 27689933 DOI: 10.1089/cbr.2016.2047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Directly radioiodinated [131I]-rituximab has been developed as a radioimmunotherapeutic agent in patients with CD20-positive B cell non-Hodgkin's lymphoma. However, there are concerns over its in vivo catabolism and deiodination. A novel radioiodination linker, N-(4-isothiocyanatobenzyl)-2-(3-(tributylstannyl)phenyl) acetamide (IBPA), was synthesized for the preparation of stable radioiodinated proteins. METHODS The authors evaluated the potential of IBPA as a stable radioiodinated linker for rituximab. [125I]-IBPA was purified and conjugated with rituximab, and in vitro stability testing was performed in serum and liver microsomes. In vivo studies were performed after i.v. injection of [125I]-rituximab or [125I]-IBPA-rituximab to nude mice. RESULTS In in vitro studies, [125I]-IBPA-rituximab was stable in serum and liver microsomes. In static scans, high radioactivity was evident in the thyroid following injection of [125I]-rituximab, but low radioactivity was seen in the thyroid following injection of [125I]-IBPA-rituximab. In biodistribution studies, radioactivity uptake in thyroid glands of [125I]-IBPA-rituximab was decreased by approximately sevenfold compared to [125I]-rituximab. In pharmacokinetics, the half-life of [125I]-rituximab was shorter than that of [125I]-IBPA-rituximab in plasma of nude mice. CONCLUSIONS The authors demonstrate that [125I]-IBPA-rituximab is more stable to metabolic deiodination in vivo than is [125I]-rituximab. Radioiodination of rituximab using IBPA is thus preferable to direct labeling in terms of in vivo stability.
Collapse
Affiliation(s)
- Eun Jung Kim
- 1 Korea Drug Development Platform using Radio-Isotope (KDePRI), Korea Institute of Radiological and Medical Sciences , Seoul, Korea.,2 School of Life Sciences and Biotechnology, Korea University , Seoul, Korea
| | - Byoung Soo Kim
- 1 Korea Drug Development Platform using Radio-Isotope (KDePRI), Korea Institute of Radiological and Medical Sciences , Seoul, Korea.,3 Department of Molecular Imaging, Korea Institute of Radiological and Medical Sciences , Seoul, Korea
| | - Dan Bee Choi
- 1 Korea Drug Development Platform using Radio-Isotope (KDePRI), Korea Institute of Radiological and Medical Sciences , Seoul, Korea
| | - Sung-Gil Chi
- 2 School of Life Sciences and Biotechnology, Korea University , Seoul, Korea
| | - Tae Hyun Choi
- 3 Department of Molecular Imaging, Korea Institute of Radiological and Medical Sciences , Seoul, Korea
| |
Collapse
|
4
|
Bäck T, Haraldsson B, Hultborn R, Jensen H, Johansson ME, Lindegren S, Jacobsson L. Glomerular Filtration Rate After Alpha-Radioimmunotherapy with 211At-MX35-F(ab′)2: A Long-Term Study of Renal Function in Nude Mice. Cancer Biother Radiopharm 2009; 24:649-58. [DOI: 10.1089/cbr.2009.0628] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tom Bäck
- Department of Radiation Physics, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Börje Haraldsson
- Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ragnar Hultborn
- Department of Oncology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Holger Jensen
- Cyclotron and PET Unit, Rigshospitalet, Copenhagen, Denmark
| | - Martin E. Johansson
- Center for Molecular Pathology, Department of Laboratory Medicine, Lund University, University Hospital MAS, Malmö, Sweden
| | - Sture Lindegren
- Department of Radiation Physics, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lars Jacobsson
- Department of Radiation Physics, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Karacay H, Brard PY, Sharkey RM, Chang CH, Rossi EA, McBride WJ, Ragland DR, Horak ID, Goldenberg DM. Therapeutic advantage of pretargeted radioimmunotherapy using a recombinant bispecific antibody in a human colon cancer xenograft. Clin Cancer Res 2006; 11:7879-85. [PMID: 16278412 DOI: 10.1158/1078-0432.ccr-05-1246] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess if pretargeting, using a combination of a recombinant bispecific antibody (bsMAb) that binds divalently to carcinoembryonic antigen (CEA) and monovalently to the hapten histamine-succinyl-glycine and a (90)Y-peptide, improves therapeutic efficacy in a human colon cancer-nude mouse xenograft compared with control animals given (90)Y-humanized anti-CEA immunoglobulin G (IgG). EXPERIMENTAL DESIGN Clearance and biodistribution were monitored by whole-body readings and necropsy. Animals were monitored for 34 weeks with a determination of residual disease and renal pathology in survivors. Hematologic toxicity was assessed separately in non-tumor-bearing NIH Swiss mice. RESULTS Hematologic toxicity was severe at doses of 100 to 200 microCi of (90)Y-IgG, yet mild in the pretargeted animals given 500 or 700 microCi of the (90)Y-peptide. Evidence of end-stage renal disease was found at 900 microCi of the pretargeted (90)Y-peptide whereas animals given 700 microCi showed only mild renal pathology, similar to that seen in control animals given (90)Y-IgG. Biodistribution data indicated that the average amount of tumor radioactivity by a 700-microCi dose of the pretargeted peptide over a 96-hour period was increased 2.5-fold (48 microCi/g) compared with 150 microCi of (90)Y-IgG (18.9 microCi/g). At these doses, survival (i.e., time to progression to 2.5 cm(3)) was significantly improved (P < 0.04) compared with (90)Y-IgG, with ablation of about one third of the tumors, whereas viable tumor was present in all of the (90)Y-IgG-treated animals. CONCLUSION Pretargeting increases the amount of radioactivity delivered to colorectal tumors sufficiently to improve the therapeutic index and responses as compared with conventional radioimmunotherapy.
Collapse
Affiliation(s)
- Habibe Karacay
- Center for Molecular Medicine and Immunology and the Garden State Cancer Center, Belleville, NJ 07109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Abou-Jaoudé W, Dale R. A theoretical radiobiological assessment of the influence of radionuclide half-life on tumor response in targeted radiotherapy when a constant kidney toxicity is maintained. Cancer Biother Radiopharm 2005; 19:308-21. [PMID: 15285877 DOI: 10.1089/1084978041425089] [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: 11/12/2022] Open
Abstract
The potential of targeted radionuclide therapy may be limited if the antibody affinity to the tumor is relatively low and if significant normal tissue damage occurs while the tumor is sterilized. One way to increase the efficiency of the antibody-radionuclide complex might be to use knowledge of the radiobiological processes to select a near-optimal radionuclide half-life. In this paper, the role of physical half-life in targeted radiotherapy optimization is investigated using the linear quadratic (LQ) radiobiological model in conjunction with a range of radiobiological parameters relevant to the tumor. Five radionuclides ((211)At, (90)Y, (131)I, (86)Rb, and (114m)In) were selected, providing a half-life range from 0.3-49.5 days. The dose-limiting organ was assumed to be the kidney, with a simple fractional link between the initial (extrapolated) dose-rate to the tumor and the initial dose-rate to the kidney. The results suggest that short-lived radionuclides (half-life in the range of 1-10 days) have an advantage over medium- and long-lived radionuclides. Furthermore, for very rapid tumor uptake (uptake half-time of a few hours), very short-lived radionuclides (half-life of less than 1 day) could be efficiently employed. Ultimately, however, treatment outcome (in terms of tumor cell kill) is limited by the antibody affinity to the tumor.
Collapse
Affiliation(s)
- Wassim Abou-Jaoudé
- Department of Biological and Medical Systems, Imperial College, London, United Kingdom
| | | |
Collapse
|
7
|
Boskovitz A, Akabani GH, Pegram CN, Bigner DD, Zalutsky MR. Human/murine chimeric 81C6 F(ab')(2) fragment: preclinical evaluation of a potential construct for the targeted radiotherapy of malignant glioma. Nucl Med Biol 2004; 31:345-55. [PMID: 15028247 DOI: 10.1016/j.nucmedbio.2003.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2003] [Revised: 10/03/2003] [Accepted: 10/05/2003] [Indexed: 11/17/2022]
Abstract
We have obtained encouraging responses in recent Phase I studies evaluating (131)I-labeled human/murine chimeric 81C6 anti-tenascin monoclonal antibody (ch81C6) administered into surgically-created tumor resection cavities in brain tumor patients. However, because the blood clearance is slow, hematologic toxicity has been higher than seen with murine 81C6 (mu81C6). In the current study, a series of paired-label experiments were performed in athymic mice bearing subcutaneous D-245 MG human glioma xenografts to compare the biodistribution of the fragment ch81C6 F(ab')(2) labeled using Iodogen to a) intact ch81C6, b) mu81C6, and c) ch81C6 F(ab')(2) labeled using N-succinimidyl 3-[(131)I]iodobenzoate. Tumor retention of radioiodine activity for the F(ab')(2) fragment was comparable to that for intact ch81C6 for the first 24 h and to that for mu81C6 for the first 48 h; as expected, blood and other normal tissue levels declined faster for ch81C6 F(ab')(2.) Radiation dosimetry calculations suggest that (131)I-labeled ch81C6 F(ab')(2) may warrant further evaluation as a targeted radiotherapeutic for the treatment of brain tumors.
Collapse
Affiliation(s)
- Abraham Boskovitz
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
8
|
Deyev SM, Waibel R, Lebedenko EN, Schubiger AP, Plückthun A. Design of multivalent complexes using the barnase*barstar module. Nat Biotechnol 2003; 21:1486-92. [PMID: 14634668 DOI: 10.1038/nbt916] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Accepted: 09/25/2003] [Indexed: 01/01/2023]
Abstract
The ribonuclease barnase (12 kDa) and its inhibitor barstar (10 kDa) form a very tight complex in which all N and C termini are accessible for fusion. Here we exploit this system to create modular targeting molecules based on antibody scFv fragment fusions to barnase, to two barnase molecules in series and to barstar. We describe the construction, production and purification of defined dimeric and trimeric complexes. Immobilized barnase fusions are used to capture barstar fusions from crude extracts to yield homogeneous, heterodimeric fusion proteins. These proteins are stable, soluble and resistant to proteolysis. Using fusions with anti-p185(HER2-ECD) 4D5 scFv, we show that the anticipated gain in avidity from monomer to dimer to trimer is obtained and that favorable tumor targeting properties are achieved. Many permutations of engineered multispecific fusion proteins become accessible with this technology of quasi-covalent heterodimers.
Collapse
Affiliation(s)
- Sergey M Deyev
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry and Institute of Gene Biology, Russian Academy of Sciences, Miklukho-Maklaya str.16/10, 117997 Moscow, Russia.
| | | | | | | | | |
Collapse
|
9
|
Flynn AA, Pedley RB, Green AJ, Dearling JL, El-Emir E, Boxer GM, Boden R, Begent RHJ. The nonuniformity of antibody distribution in the kidney and its influence on dosimetry. Radiat Res 2003; 159:182-9. [PMID: 12537523 DOI: 10.1667/0033-7587(2003)159[0182:tnoadi]2.0.co;2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The therapeutic efficacy of radiolabeled antibody fragments can be limited by nephrotoxicity, particularly when the kidney is the major route of extraction from the circulation. Conventional dose estimates in kidney assume uniform dose deposition, but we have shown increased antibody localization in the cortex after glomerular filtration. The purpose of this study was to measure the radioactivity in cortex relative to medulla for a range of antibodies and to assess the validity of the assumption of uniformity of dose deposition in the whole kidney and in the cortex for these antibodies with a range of radionuclides. Storage phosphor plate technology (radioluminography) was used to acquire images of the distributions of a range of antibodies of various sizes, labeled with 125I, in kidney sections. This allowed the calculation of the antibody concentration in the cortex relative to the medulla. Beta-particle point dose kernels were then used to generate the dose-rate distributions from 14C, 131I, 186Re, 32P and 90Y. The correlation between the actual dose-rate distribution and the corresponding distribution calculated assuming uniform antibody distribution throughout the kidney was used to test the validity of estimating dose by assuming uniformity in the kidney and in the cortex. There was a strong inverse relationship between the ratio of the radioactivity in the cortex relative to that in the medulla and the antibody size. The nonuniformity of dose deposition was greatest with the smallest antibody fragments but became more uniform as the range of the emissions from the radionuclide increased. Furthermore, there was a strong correlation between the actual dose-rate distribution and the distribution when assuming a uniform source in the kidney for intact antibodies along with medium- to long-range radionuclides, but there was no correlation for small antibody fragments with any radioisotope or for short-range radionuclides with any antibody. However, when the cortex was separated from the whole kidney, the correlation between the actual dose-rate distribution and the assumed dose-rate distribution, if the source was uniform, increased significantly. During radioimmunotherapy, the extent of nonuniformity of dose deposition in the kidney depends on the properties of the antibody and radionuclide. For dosimetry estimates, the cortex should be taken as a separate source region when the radiopharmaceutical is small enough to be filtered by the glomerulus.
Collapse
Affiliation(s)
- Aiden A Flynn
- CRC Targeting and Imaging Group, Department of Oncology, Royal Free and University College Medical School, University College London, London, NW3 2PF, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Reff ME, Hariharan K, Braslawsky G. Future of monoclonal antibodies in the treatment of hematologic malignancies. Cancer Control 2002; 9:152-66. [PMID: 11965235 DOI: 10.1177/107327480200900207] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The approval of monoclonal antibodies (MAbs) as antibody-targeted therapy in the management of patients with hematologic malignancies has led to new treatment options for this group of patients. The ability to target antibodies to novel functional receptors can increase their therapeutic efficacy. METHODS The authors reviewed improvements in MAb design to enhance their effectiveness over the existing therapeutic MAb currently approved for treating hematologic malignancies. RESULTS Three classes of therapeutic MAbs showing promise in human clinical trials for treatment of hematologic malignancies include unconjugated MAb, drug conjugates in which the antibody preferentially delivers a potent cytotoxic drug to the tumor, and radioactive immunotherapy in which the antibody delivers a sterilizing dose of radiation to the tumor. CONCLUSIONS A better appreciation of how MAbs are metabolized in the body and localized to tumors is resulting in the development of new antibody constructs with improved biodistribution profiles.
Collapse
Affiliation(s)
- Mitchell E Reff
- Molecular Biology and Tumor Immunology Departments, IDEC Pharmaceuticals Corp, San Diego, CA 92191-9080, USA.
| | | | | |
Collapse
|
11
|
Oshikiri T, Hida Y, Miyamoto M, Hashida H, Katoh K, Suzuoki M, Nakakubo Y, Hiraoka K, Shinohara T, Itoh T, Kondo S, Katoh H. RCAS1 as a tumour progression marker: an independent negative prognostic factor in gallbladder cancer. Br J Cancer 2001; 85:1922-7. [PMID: 11747335 PMCID: PMC2364016 DOI: 10.1054/bjoc.2001.2192] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Receptor-binding cancer antigen expressed on SiSo cells (RCAS1) induces apoptosis in immune cells bearing the RCAS1 receptor. We sought to determine RCAS1 involvement in the origin and progression of gallbladder cancer, and also implications of RCAS1 for patient survival. RCAS1 expression was examined immunohistochemically in 110 surgically resected gallbladder specimens. The gallbladders represented 20 cases of cholecystitis with no associated pancreaticobiliary maljunction; 23 cases of cholecystitis with pancreaticobiliary maljunction; 14 cases of adenomyomatosis; 7 adenomas; and 46 cancers. High expression of RCAS1 (immunoreactivity in over 25% of cells) was observed in 32 of the 46 cancers (70%), but not in other diseases, including pre-cancerous conditions. RCAS1 immunoreactivity was associated with depth of tumour invasion (P = 0.0180), lymph node metastasis (P = 0.0033), lymphatic involvement (P = 0.0104), venous involvement (P = 0.0224), perineural involvement (P = 0.0351) and stage by the tumour, nodes and metastases (TNM) classification (P = 0.0026). Thus, RCAS1 expression may be a relatively late event in gallbladder carcinogenesis, possibly promoting tumour progression. Cox regression multivariate analysis demonstrated RCAS1 positivity to be an independent negative predictor for survival (P = 0.0337; risk ratio, 12.690; 95% confidence interval, 1.216-132.423). High expression of RCAS1 significantly correlated with tumour progression and predicted poor outcome in gallbladder cancer.
Collapse
Affiliation(s)
- T Oshikiri
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University Graduate School of Medicine, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kortt AA, Dolezal O, Power BE, Hudson PJ. Dimeric and trimeric antibodies: high avidity scFvs for cancer targeting. BIOMOLECULAR ENGINEERING 2001; 18:95-108. [PMID: 11566601 DOI: 10.1016/s1389-0344(01)00090-9] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Recombinant antibody fragments can be engineered to assemble into stable multimeric oligomers of high binding avidity and specificity to a wide range of target antigens and haptens. This review describes the design and expression of diabodies (dimers), triabodies (trimers) and tetrabodies (tetramers). In particular we discuss the role of linker length between V-domains and the orientation of the V-domains to direct the formation of either diabodies (60 kDa), triabodies (90 kDa) or tetrabodies (120 kDa), and how the size, flexibility and valency of each molecules is suited to different applications for in vivo imaging and therapy. Single chain Fv antibody fragments joined by polypeptide linkers of at least 12 residues irrespective of V-domains orientation predominantly form monomers with varying amounts of dimer and higher molecular mass oligomers in equilibrium. A scFv molecule with a linker of 3-12 residues cannot fold into a functional Fv domain and instead associates with a second scFv molecule to form a bivalent dimer (diabody, approximately 60 kDa). Reducing the linker length below three residues can force scFv association into trimers (triabodies, approximately 90 kDa) or tetramers ( approximately 120 kDa) depending on linker length, composition and V-domain orientation. A particular advantage for tumour targeting is that molecules of 60-100 kDa have increased tumour penetration and fast clearance rates compared with the parent Ig (150 kDa). We highlight a number of cancer-targeting scFv diabodies that have undergone successful pre-clinical trials for in vivo stability and efficacy. We also briefly review the design of multi-specific Fv modules suited to cross-link two or more different target antigens. Bi-specific diabodies formed by association of different scFv molecules have been designed as cross-linking reagents for T-cell recruitment into tumours (immunotherapy), viral retargeting (gene therapy) and as red blood cell agglutination reagents (immunodiagnostics). The more challenging trispecific multimers (triabodies) remain to be described.
Collapse
Affiliation(s)
- A A Kortt
- CSIRO Health Science and Nutrition, 343 Royal Parade, Vic. 3052, Parkville, Australia.
| | | | | | | |
Collapse
|
13
|
Storto G, Buchegger F, Waibel R, Kuenzi G, Offord RE, Schubiger PA, Gillet M, Delaloye AB. Biokinetics of a F(ab')3 iodine-131 labeled antigen binding construct (Mab 35) directed against CEA in patients with colorectal carcinoma. Cancer Biother Radiopharm 2001; 16:371-9. [PMID: 11776754 DOI: 10.1089/108497801753354276] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
UNLABELLED An 131I labeled trivalent antigen binding construct, formed from 3 Fab' fragments of murine anti-CEA monoclonal antibody (Mab) 35, has shown favorable biokinetics in animal studies. OBJECTIVES The aim of this study was to evaluate biodistribution and tumor uptake of 131I-F(ab')3 in patients and its potential utility for radioimmunotherapy of CEA expressing tumors. PATIENTS AND METHODS Six patients (5 M, 1 F; age 62 +/- 13 y) with liver metastases of colorectal cancer, scheduled for hepatic surgery were studied by 2-3 whole body scans immediately post infusion of 111-137 MBq of 131I labeled Mab 35 F(ab')3 and up to 72 h. Circulating CEA ranged from 1.2 to 1930 ng/ml. We evaluated plasma and whole body clearance, activity accumulation by post-surgical ex-vivo tissue measurement in primary tumor (T) and metastases (M), and calculated M to blood (M/B) and M to liver (M/L) ratios. RESULTS All known tumor sites were detected by immunoscintigraphy and confirmed at surgery. Whole body effective T1/2 calculated in two patients was 51.5 h and 55.6 h respectively. Effective serum T1/2 was mono-exponential in 3 patients (short observation interval) with 20.9 +/- 7 h and bi-exponential in three with alpha T1/2 of 6.3 +/- 1 h and beta T1/2 of 38.6 +/- 5 h. In a patient with concomitant colic and hepatic lesions uptake of primary tumor was 0.0071% injected dose per gram of tissue (%ID/g) and mean metastases activity was 0.0275 %ID/g at 48 h. In the 3 patients who had surgery at 48 h, mean uptake in metastases and normal liver was 0.0182 %ID/g and 0.0021 %ID/g, respectively (M/L 8.67). In the single subject followed until 7 days post infusion, residual activity in liver metastases was 10 times higher than in normal parenchyma. CONCLUSIONS Tumor uptake and tumor to blood ratio, as well as serum clearance of the triconstruct are similar to those observed with intact iodinated anti-CEA antibodies. In the patient studied for 7 days the tumor residence time was favorable. Further improvements, however, need to be obtained before considering this approach for radioimmunotherapy.
Collapse
Affiliation(s)
- G Storto
- Department of Nuclear Medicine, University Hospital Lausanne, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Recombinant antibodies now represent over 30% of biopharmaceuticals in clinical trials, highlighted by the recent approvals for cancer immunotherapy from the FDA which has awoken the biotechnology industry. Sales of these antibodies are increasing very rapidly to a predicted US$ 3 billion per annum worldwide by 2002. Since the development of new therapeutic reagent into commercial product takes 10 years, the recent FDA-approved antibodies are based on early antibody designs which are now considered primitive. Emerging technologies have created a vast range of novel, recombinant, antibody-based reagents which specifically target clinical biomarkers of disease. In the past year, radiolabelling of antibodies has increased their potential for cancer imaging and targeting. Recombinant antibodies have also been reduced in size and rebuilt into multivalent molecules for higher affinity. In addition, antibodies have been fused with many molecules including toxins, enzymes and viruses for prodrug therapy, cancer treatment and gene delivery. Recombinant antibody technology has enabled clever manipulations in the construction of complex antibody library repertoires for the selection of high-affinity reagents against refractory targets. Although phage display remains the most extensively used method, this year high affinity reagents have been isolated using alternative display and selection systems such as ribosome display and yeast display confirming the emergence of new display methods. Furthermore, innovative affinity maturation strategies have been developed to obtain high affinity reagents. This review focuses on developments in the last 12 months and describes the latest developments in the design, production and clinical use of recombinant antibodies for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- P J Hudson
- CRC for Diagnostics at CSIRO Health Sciences and Nutrition, 343 Royal Parade, Parkville, Victoria, 3052, Australia.
| | | |
Collapse
|
15
|
Goel A, Batra SK. Antibody constructs for radioimmunodiagnosis and treatment of human pancreatic cancer. TERATOGENESIS, CARCINOGENESIS, AND MUTAGENESIS 2001; 21:45-57. [PMID: 11135320 DOI: 10.1002/1520-6866(2001)21:1<45::aid-tcm5>3.0.co;2-a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pancreatic cancer (PC) is a common disease that is seldom cured. Current approaches to the treatment of PC are not effective because the non-specific nature of both chemotherapy and external beam radiation results in toxicity to normal tissue. Monoclonal antibodies (MAbs) can be used as selective carriers for delivering radionuclides, toxins, or cytotoxic drugs to malignant cell populations. Therefore, MAb-technology has led to a significant amount of research in targeted therapy. Targeted therapy would generally allow the concentration of cytotoxic agents in tumors and would markedly lessen the toxicity to normal tissues, which limits the dosage and effectiveness of systemically administered drugs. A variety of MAbs are being pre-clinically evaluated for the diagnosis and treatment of PC. Novel recombinant antibody constructs hold a promising future in both the diagnosis and treatment of cancer. By genetic-engineering methods, several high affinity antibody fragments with optimum tumor targeting properties, such as higher functional affinity (divalent and multivalent scFvs) and blood residence time (good tumor localization with high radiolocalization index), have been generated. Animal models have permitted the in vivo assessment of these antibody-based reagents, therapeutic/diagnostic radionuclide, radiolabeling conditions, and efficacy of administration regimes. For PC, immunoscintigraphy using MAbs has taken new strides. The use of MAbs and their fragments for radioimmunoguided surgery and therapy of PC has shown encouraging results at preclinical levels and warrants further attention.
Collapse
Affiliation(s)
- A Goel
- Department of Biochemistry and Molecular Biology, Eppley Institute of Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | |
Collapse
|
16
|
Todorovska A, Roovers RC, Dolezal O, Kortt AA, Hoogenboom HR, Hudson PJ. Design and application of diabodies, triabodies and tetrabodies for cancer targeting. J Immunol Methods 2001; 248:47-66. [PMID: 11223068 DOI: 10.1016/s0022-1759(00)00342-2] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Multivalent recombinant antibody fragments provide high binding avidity and unique specificity to a wide range of target antigens and haptens. This review describes the design and expression of diabodies, triabodies and tetrabodies using examples of scFv molecules that target viruses (influenza neuraminidase) and cancer (Ep-CAM; epithelial cell adhesion molecule). We discuss the preferred choice of linker length between V-domains to direct the formation of either diabodies (60 kDa), triabodies (90 kDa) or tetrabodies (120 kDa), each with size, flexibility and valency suited to different applications for in vivo imaging and therapy. The increased binding valency of these scFv multimers results in high avidity (low off-rates). A particular advantage for tumour targeting is that molecules of 60-100 kDa have increased tumour penetration and fast clearance rates compared to the parent Ig (150 kDa). We highlight a number of cancer-targeting scFv multimers that have recently successfully undergone pre-clinical trials for in vivo stability and efficacy. We also review the design of multi-specific Fv modules suited to cross-link two or more different target antigens. These bi- and tri-specific multimers can be formed by association of different scFv molecules and, in the first examples, have been designed as cross-linking reagents for T-cell recruitment into tumours (immunotherapy), viral retargeting (gene therapy) and as red blood cell agglutination reagents (immunodiagnostics).
Collapse
Affiliation(s)
- A Todorovska
- CSIRO Health Science and Nutrition and CRC for Diagnostic Technologies, 343 Royal Parade, Victoria 3052, Parkville, Australia
| | | | | | | | | | | |
Collapse
|
17
|
Hudson PJ. Recombinant antibodies: a novel approach to cancer diagnosis and therapy. Expert Opin Investig Drugs 2000; 9:1231-42. [PMID: 11060739 DOI: 10.1517/13543784.9.6.1231] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Recombinant antibodies and their fragments currently represent over 30% of all biological proteins undergoing clinical trials for diagnosis and therapy. These reagents dominate the cancer-targeting field, as highlighted by the recent approval of the first engineered therapeutic antibodies by the Food and Drugs Administration (FDA). Last year, important advances have been made in the design, selection and production of recombinant antibodies. The natural immune repertoire and somatic cell affinity maturation has been superseded by large antibody display libraries and rapid molecular evolution strategies. These novel libraries and selection methods have enabled the rapid isolation of high-affinity cancer targeting and antiviral antibodies, the latter capable of redirecting viruses for gene therapy applications. In alternative strategies for cancer diagnosis and therapy, recombinant antibody fragments have been fused to radioisotopes, drugs, toxins, enzymes and biosensor surfaces. Antibody-directed cancer pre-targeting followed by prodrug activation (ADEPT) has proved a most promising therapeutic strategy. Multi-specific antibodies have been effective for cytotoxic T-cell recruitment and antibody-fusion proteins have delivered enhanced immunotherapeutic and vaccination strategies. The new millennium is indeed an exciting time for the design, selection and formulation of a range of new antibody-based products for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- P J Hudson
- CRC for Diagnostic Technologies, CSIRO Health Sciences and Nutrition, 343 Royal Parade, Parkville, Victoria, Australia 3052.
| |
Collapse
|
18
|
Abstract
Recombinant antibodies and their fragments now represent over 30% of all biological proteins undergoing clinical trials for diagnosis and therapy. The focus on antibodies as the ideal cancer-targeting reagents recently culminated in approval by the Food and Drugs Administration for the first engineered therapeutic antibodies. In the past year, important advances have been made in the design, selection and production of new types of engineered antibodies. Innovative selection methods have enabled the isolation of high-affinity cancer-targeting and antiviral antibodies, the latter capable of redirecting viruses for gene therapy applications. In other strategies for cancer diagnosis and therapy, recombinant antibody fragments have been fused to radioisotopes, drugs, toxins, enzymes and biosensor surfaces. Bispecific antibodies and related fusion proteins have been produced for cancer immunotherapy, effectively enhancing the human immune response in anticancer vaccines and T cell recruitment strategies.
Collapse
Affiliation(s)
- P J Hudson
- Cooperative Research Centre for Diagnostic Technologies, The Commonwealth Scientific and Industrial Research Organisation, MolecularScience Unit, 343 Royal Parade, Parkville, Victoria, 3052, Australia.
| |
Collapse
|