1
|
Sanz L, Ibáñez-Pérez R, Guerrero-Ochoa P, Lacadena J, Anel A. Antibody-Based Immunotoxins for Colorectal Cancer Therapy. Biomedicines 2021; 9:1729. [PMID: 34829955 PMCID: PMC8615520 DOI: 10.3390/biomedicines9111729] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 01/21/2023] Open
Abstract
Monoclonal antibodies (mAbs) are included among the treatment options for advanced colorectal cancer (CRC). However, while these mAbs effectively target cancer cells, they may have limited clinical activity. A strategy to improve their therapeutic potential is arming them with a toxic payload. Immunotoxins (ITX) combining the cell-killing ability of a toxin with the specificity of a mAb constitute a promising strategy for CRC therapy. However, several important challenges in optimizing ITX remain, including suboptimal pharmacokinetics and especially the immunogenicity of the toxin moiety. Nonetheless, ongoing research is working to solve these limitations and expand CRC patients' therapeutic armory. In this review, we provide a comprehensive overview of targets and toxins employed in the design of ITX for CRC and highlight a wide selection of ITX tested in CRC patients as well as preclinical candidates.
Collapse
Affiliation(s)
- Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute, Hospital Universitario Puerta de Hierro, 28222 Madrid, Spain
| | - Raquel Ibáñez-Pérez
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| | - Patricia Guerrero-Ochoa
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| | - Javier Lacadena
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
| | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| |
Collapse
|
2
|
Plunk MA, Quintana JM, Darden CM, Lawrence MC, Naziruddin B, Kane RR. Design and Catalyzed Activation of Mycophenolic Acid Prodrugs. ACS Med Chem Lett 2021; 12:812-816. [PMID: 34055230 DOI: 10.1021/acsmedchemlett.1c00079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/05/2021] [Indexed: 01/17/2023] Open
Abstract
Mycophenolic acid (MPA) and its morpholino ester prodrug mycophenolate mofetil (MMF) are widely used in solid organ transplantation. These drugs prevent rejection due to their potent inhibition of inosine-5'-monophosphate dehydrogenase (IMPDH), an enzyme vital for lymphocyte proliferation. As a strategy to provide localized immunosuppression in cell transplantation, four mycophenolic acid prodrugs designed to release MPA by two distinct mechanisms were synthesized and characterized. A nitrobenzyl ether prodrug was effectively converted to MPA upon exposure to bacterial nitroreductase, while a propargyl ether was converted to the active drug by immobilized Pd0 nanoparticles. In vitro, both prodrugs were inactive against IMPDH and exhibited reduced toxicity relative to the active drug, suggesting their potential for providing localized immunosuppression.
Collapse
Affiliation(s)
- Michael A. Plunk
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Jeremy M. Quintana
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Carly M. Darden
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
| | - Michael C. Lawrence
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, Texas 75204, United States
| | - Bashoo Naziruddin
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
- Baylor Simmons Transplant Institute, Baylor University Medical Center, Dallas, Texas 75204, United States
| | - Robert R. Kane
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
| |
Collapse
|
3
|
Wei D, Tao Z, Shi Q, Wang L, Liu L, She T, Yi Q, Wen X, Liu L, Li S, Yang H, Jiang X. Selective Photokilling of Colorectal Tumors by Near-Infrared Photoimmunotherapy with a GPA33-Targeted Single-Chain Antibody Variable Fragment Conjugate. Mol Pharm 2020; 17:2508-2517. [PMID: 32396000 DOI: 10.1021/acs.molpharmaceut.0c00210] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Antibody-based near-infrared photoimmunotherapy (NIR-PIT) is an attractive strategy for cancer treatment. Tumor cells can be selectively and efficiently killed by the targeted delivery of an antibody-photoabsorber complex followed by exposure to NIR light. Glycoprotein A33 antigen (GPA33) is highly expressed in most human colorectal cancers (CRCs) and is an ideal diagnostic and therapeutic target. We previously produced a single-chain fragment of a variable antibody against GPA33 (A33scFv antibody). Here, we investigate the efficacy of NIR-PIT by combining A33scFv with the NIR photoabsorber IR700 (A33scFv-IR700). In vitro, recombinant A33scFv displayed specific binding and delivery of an NIR dye to GPA33-positive tumor cells. Furthermore, A33scFv-IR700-mediated NIR-PIT was successful in rapidly and specifically killing GPA33-positive colorectal tumor cells. NIR-PIT treatment induced the release of lactate dehydrogenase from tumor cells, followed by cell necrosis, rather than apoptosis, through the promotion of reactive oxygen species accumulation in tumor cells. In mice bearing LS174T tumor grafts, A33scFv selectively accumulated in GPA33-positive tumors. Following only a single injection of the conjugate and subsequent illumination, A33scFv-IR700-mediated NIR-PIT induced a significant increase in therapeutic response in LS174T-tumor mice compared with that in the non-NIR-PIT groups (p < 0.001). Because the GPA33 antigen is specifically expressed in CRC tumors, A33scFv-IR700 might be a promising antibody fragment-photoabsorber conjugate for NIR-PIT of CRC.
Collapse
Affiliation(s)
- Danfeng Wei
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China.,Medical Research Center, The Third People's Hospital of Chengdu, The Second Affiliated Chengdu Clinical College of Chongqing Medical University, Chengdu 610031, China.,Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ze Tao
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiuxiao Shi
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lijun Wang
- Department of Ophthalmology, The Third People's Hospital of Chengdu, The Second Affiliated Chengdu Clinical College of Chongqing Medical University, Chengdu 610031, China
| | - Lei Liu
- Medical Research Center, The Third People's Hospital of Chengdu, The Second Affiliated Chengdu Clinical College of Chongqing Medical University, Chengdu 610031, China
| | - Tianshan She
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qin Yi
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiang Wen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lian Liu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shengfu Li
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Plunk MA, Alaniz A, Olademehin OP, Ellington TL, Shuford KL, Kane RR. Design and Catalyzed Activation of Tak-242 Prodrugs for Localized Inhibition of TLR4-Induced Inflammation. ACS Med Chem Lett 2020; 11:141-146. [PMID: 32071680 DOI: 10.1021/acsmedchemlett.9b00518] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/03/2020] [Indexed: 11/30/2022] Open
Abstract
Tak-242 (resatorvid), a Toll-like Receptor 4 (TLR4) inhibitor, has been identified as a potent suppressor of innate inflammation. As a strategy to target Tak-242 to select tissue, four TLR4-inactive prodrugs were synthesized for activation via two different release mechanisms. Two nitrobenzyl Tak-242 prodrugs released the parent drug upon exposure to the exogenous enzyme nitroreductase, while the two propargyl prodrugs were converted to Tak-242 in the presence of Pd0.
Collapse
Affiliation(s)
- Michael A. Plunk
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Alyssa Alaniz
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76798, United States
| | - Olatunde P. Olademehin
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Thomas L. Ellington
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Kevin L. Shuford
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Robert R. Kane
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76798, United States
| |
Collapse
|
5
|
Roy S, Brasino M, Beirne JM, Harguindey A, Chapnick DA, Liu X, Cha JN, Goodwin AP. Enzymes Photo-Cross-Linked to Live Cell Receptors Retain Activity and EGFR Inhibition after Both Internalization and Recycling. Bioconjug Chem 2019; 31:104-112. [PMID: 31840981 DOI: 10.1021/acs.bioconjchem.9b00781] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this work, we show that a prodrug enzyme covalently photoconjugated to live cell receptors survives endosomal proteolysis and retains its catalytic activity over multiple days. Here, a fusion protein was designed with both an antiepidermal growth factor receptor (EGFR) affibody and the prodrug enzyme cytosine deaminase, which can convert prodrug 5-fluorocytosine to the anticancer drug 5-fluorouracil. A benzophenone group was added at a site-specific mutation within the affibody, and the fusion protein was selectively photoconjugated to EGFR receptors expressed on membranes of MDA-MB-468 breast cancer cells. The fusion protein was next labeled with two dyes for tracking uptake: AlexaFluor 488 and pH-sensitive pHAb. Flow cytometry showed that fusion proteins photo-cross-linked to EGFR first underwent receptor-mediated endocytosis within 12 h, followed by recycling back to the cell membrane within 24 h. These findings were also confirmed by confocal microscopy. The unique cross-linking of the affibody-enzyme fusion proteins was utilized for two anticancer treatments. First, the covalent linking of the protein to the EGFR led to inhibition of ERK signaling over a two-day period, whereas conventional antibody therapy only led to 6 h of inhibition. Second, when the affibody-CodA fusion proteins were photo-cross-linked to EGFR overexpressed on MDA-MB-468 breast cancer cells, prodrug conversion was found even 48 h postincubation without any apparent decrease in cell killing, while without photo-cross-linking no cell killing was observed 8 h postincubation. These studies show that affinity-mediated covalent conjugation of the affibody-enzymes to cell receptors allows for prolonged expression on membranes and retained enzymatic activity without genetic engineering.
Collapse
|
6
|
Bakhtiary Z, Saei AA, Hajipour MJ, Raoufi M, Vermesh O, Mahmoudi M. Targeted superparamagnetic iron oxide nanoparticles for early detection of cancer: Possibilities and challenges. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 12:287-307. [PMID: 26707817 DOI: 10.1016/j.nano.2015.10.019] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/22/2015] [Accepted: 10/25/2015] [Indexed: 02/07/2023]
Abstract
UNLABELLED Nanomedicine, the integration of nanotechnological tools in medicine demonstrated promising potential to revolutionize the diagnosis and treatment of various human health conditions. Nanoparticles (NPs) have shown much promise in diagnostics of cancer, especially since they can accommodate targeting molecules on their surface, which search for specific tumor cell receptors upon injection into the blood stream. This concentrates the NPs in the desired tumor location. Furthermore, such receptor-specific targeting may be exploited for detection of potential metastases in an early stage. Some NPs, such as superparamagnetic iron oxide NPs (SPIONs), are also compatible with magnetic resonance imaging (MRI), which makes their clinical translation and application rather easy and accessible for tumor imaging purposes. Furthermore, multifunctional and/or theranostic NPs can be used for simultaneous imaging of cancer and drug delivery. In this review article, we will specifically focus on the application of SPIONs in early detection and imaging of major cancer types. FROM THE CLINICAL EDITOR Super-paramagnetic iron oxide nanoparticles (SPIONs) have been reported by many to be useful as an MRI contrast agent in the detection of tumors. To further enhance the tumor imaging, SPIONs can be coupled with tumor targeting motifs. In this article, the authors performed a comprehensive review on the current status of using targeted SPIONS in tumor detection and also the potential hurdles to overcome.
Collapse
Affiliation(s)
- Zahra Bakhtiary
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ata Saei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Mohammad J Hajipour
- Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad Raoufi
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Stuttgart, Germany; Department of Nanotechnology & Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ophir Vermesh
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, CA, USA
| | - Morteza Mahmoudi
- Department of Nanotechnology & Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
7
|
Tomé-Amat J, Herrero-Galán E, Oñaderra M, Martínez-Del-Pozo Á, Gavilanes JG, Lacadena J. Preparation of an engineered safer immunotoxin against colon carcinoma based on the ribotoxin hirsutellin A. FEBS J 2015; 282:2131-41. [PMID: 25752204 DOI: 10.1111/febs.13262] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 03/03/2015] [Accepted: 03/09/2015] [Indexed: 11/27/2022]
Abstract
Immunotoxins are chimeric proteins composed of an antibody domain that specifically directs the action of the toxic domain, resulting in the death of the targeted cells. Over recent years, immunotoxins have been widely studied and the number of different constructions has increased exponentially. Protein engineering has allowed the design of optimized versions of immunotoxins with an improved tumor binding affinity, stability or cytotoxic efficacy, although sometimes this has compromised the safety of the patient in terms of undesirable adverse secondary reactions. A triple mutant at three Trp residues (HtA3ΔW) of the ribotoxin hirsutellin A retains its specific ribonucleolytic activity, although cell internalization capacity is lacking. This toxin variant has been fused to the single chain variable fragment A33 (scFvA33). This immunoconjugate (IMTXA33HtA3ΔW) was produced in the methylotrophic yeast Pichia pastoris and purified using nickel-nitrilotriacetic acid affinity chromatography. Both target and toxic domains were characterized. The immunotoxin showed an exquisite specific binding against GPA33-positive culture cells, which results in the death of the targeted cells because of specific ribonucleolytic activity against ribosomes of the engineered hirsutellin A variant. IMTXA33HtA3ΔW represents a promising structure in the search for an improved immunotoxin without compromising the safety of patients.
Collapse
Affiliation(s)
- Jaime Tomé-Amat
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, Madrid, Spain.,Department of Food Science, Cornell University, Ithaca, NY, USA
| | - Elías Herrero-Galán
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mercedes Oñaderra
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, Madrid, Spain
| | | | - José G Gavilanes
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, Madrid, Spain
| | - Javier Lacadena
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, Madrid, Spain
| |
Collapse
|
8
|
Tomé-Amat J, Ruiz-de-la-Herrán J, Martínez-del-Pozo Á, Gavilanes JG, Lacadena J. α-sarcin and RNase T1 based immunoconjugates: the role of intracellular trafficking in cytotoxic efficiency. FEBS J 2014; 282:673-84. [PMID: 25475209 DOI: 10.1111/febs.13169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 11/07/2014] [Accepted: 12/02/2014] [Indexed: 01/16/2023]
Abstract
Toxins have been thoroughly studied for their use as therapeutic agents in search of an improvement in toxic efficiency together with a minimization of their undesired side effects. Different studies have shown how toxins can follow different intracellular pathways which are connected with their cytotoxic action inside the cells. The work herein presented describes the different pathways followed by the ribotoxin α-sarcin and the fungal RNase T1, as toxic domains of immunoconjugates with identical binding domain, the single chain variable fragment of a monoclonal antibody raised against the glycoprotein A33. According to the results obtained both immunoconjugates enter the cells via early endosomes and, while α-sarcin can translocate directly into the cytosol to exert its deathly action, RNase T1 follows a pathway that involves lysosomes and the Golgi apparatus. These facts contribute to explaining the different cytotoxicity observed against their targeted cells, and reveal how the innate properties of the toxic domain, apart from its catalytic features, can be a key factor to be considered for immunotoxin optimization.
Collapse
Affiliation(s)
- Jaime Tomé-Amat
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Spain; Department of Food Science, Cornell University, Ithaca, NY, USA
| | | | | | | | | |
Collapse
|
9
|
Chester KA, Baker M, Mayer A. Overcoming the immunologic response to foreign enzymes in cancer therapy. Expert Rev Clin Immunol 2014; 1:549-59. [DOI: 10.1586/1744666x.1.4.549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
10
|
Kwon HS, Jung SG, Kim HY, Parker SA, Batt CA, Kim YR. A multi-functional polyhydroxybutyrate nanoparticle for theranostic applications. J Mater Chem B 2014; 2:3965-3971. [DOI: 10.1039/c4tb00304g] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
One-step enzymatic synthesis of theranostic PHB nanoparticles using PHA synthase fused with A33scFv and GFP.
Collapse
Affiliation(s)
- Hee-Su Kwon
- Institute of Life Sciences and Resources & Department of Food Science and Biotechnology
- College of Life Sciences
- Kyung Hee University
- Yongin, Republic of Korea
| | - Sung-Geun Jung
- Institute of Life Sciences and Resources & Department of Food Science and Biotechnology
- College of Life Sciences
- Kyung Hee University
- Yongin, Republic of Korea
| | - Hae-Yeong Kim
- Institute of Life Sciences and Resources & Department of Food Science and Biotechnology
- College of Life Sciences
- Kyung Hee University
- Yongin, Republic of Korea
| | | | - Carl A. Batt
- Graduate field of Biomedical Engineering
- Cornell University
- Ithaca, USA
- Department of Food Science
- Cornell University
| | - Young-Rok Kim
- Institute of Life Sciences and Resources & Department of Food Science and Biotechnology
- College of Life Sciences
- Kyung Hee University
- Yongin, Republic of Korea
| |
Collapse
|
11
|
Kirui DK, Khalidov I, Wang Y, Batt CA. Targeted near-IR hybrid magnetic nanoparticles for in vivo cancer therapy and imaging. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:702-11. [DOI: 10.1016/j.nano.2012.11.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/04/2012] [Accepted: 11/18/2012] [Indexed: 11/25/2022]
|
12
|
Tomé-Amat J, Menéndez-Méndez A, García-Ortega L, Batt CA, Oñaderra M, Martínez-del-Pozo A, Gavilanes JG, Lacadena J. Production and characterization of scFvA33T1, an immunoRNase targeting colon cancer cells. FEBS J 2012; 279:3022-32. [PMID: 22748038 DOI: 10.1111/j.1742-4658.2012.08683.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Within the last 10 years, the use of different RNases as therapeutic agents for various diseases has been pursued. Furthermore, the advancements of recombinant technology have allowed the assembly of proteins with different functions. In this regard, immunoribonucleases (immunoRNases) stand out as some of the most promising therapeutic candidates given their enzymatic and non-mutagenic character. Accordingly, the work reported here describes fusing RNase T1, one of the most studied members of the microbial RNase family, to the single-chain variable fragment (scFv) of a monoclonal antibody that targets the glycoprotein A33 antigen (GPA33) from human colon cancer cells. A heterologous production system, which employs the yeast Pichia pastoris, has been optimized to produce this immunoRNase (scFvA33T1) with yields of ∼ 5-10 mg · L(-1). The purified protein appears to be correctly folded as it retains its antigen specificity and ribonucleolytic activity. Finally, it also shows specific binding to, internalization into and toxicity against GPA33-positive cell lines compared with the control, GPA33-negative cells. Overall, it can be concluded that scFvA33T1 is a promising therapeutic fusion protein with the additional advantage that presumably it can be produced and purified in large amounts using an easily scalable yeast-based system.
Collapse
Affiliation(s)
- Jaime Tomé-Amat
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Carreras-Sangrà N, Tomé-Amat J, García-Ortega L, Batt CA, Oñaderra M, Martínez-del-Pozo A, Gavilanes JG, Lacadena J. Production and characterization of a colon cancer-specific immunotoxin based on the fungal ribotoxin α-sarcin. Protein Eng Des Sel 2012; 25:425-35. [PMID: 22718791 DOI: 10.1093/protein/gzs032] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A single-chain fusion protein that directed the cytolytic activity of α-sarcin to A33 tumor antigen expressing cells was constructed and shown to effectively kill targeted cells. Glycoprotein A33 (GPA33) is a well-known colon cancer marker and a humanized antibody against it was used to target the α-sarcin. The fungal ribotoxin α-sarcin is one of the most potent and specific toxins known. It is small, protease resistant, thermostable and highly efficient towards the inactivation of ribosomes. This work describes the production and characterization of an immunotoxin resulting from fusing the single-chain variable fragment (scFv) of the monoclonal antibody that targets GPA33 to fungal α-sarcin. This chimeric protein (scFvA33αsarcin), produced in Pichia pastoris and purified in high yield was proven to be properly folded, active, specific and stable. It showed high specific toxicity against GPA33-positive tumoral cell lines providing scientific evidence to sustain that scFvA33αsarcin is a good immunotherapeutic candidate against GPA33-positive colon carcinomas.
Collapse
Affiliation(s)
- Nelson Carreras-Sangrà
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, 28040 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Andrady C, Sharma SK, Chester KA. Antibody-enzyme fusion proteins for cancer therapy. Immunotherapy 2011; 3:193-211. [PMID: 21322759 DOI: 10.2217/imt.10.90] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Advances in biomolecular technology have allowed the development of genetically fused antibody-enzymes. Antibody-enzyme fusion proteins have been used to target tumors for cancer therapy in two ways. In one system, an antibody-enzyme is pretargeted to the tumor followed by administration of an inactive prodrug that is converted to its active form by the pretargeted enzyme. This system has been described as antibody-directed enzyme prodrug therapy. The other system uses antibody-enzyme fusion proteins as direct therapeutics, where the enzyme is toxic in its own right. The key feature in this approach is that the antibody is used to internalize the toxic enzyme into the tumor cell, which activates cell-death processes. This antibody-enzyme system has been largely applied to deliver ribonucleases. This article addresses these two antibody-enzyme targeting strategies for cancer therapy from concept to (pre)clinical trials.
Collapse
Affiliation(s)
- Carima Andrady
- Cancer Research UK Targeting & Imaging Group, Department of Oncology, UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London WC1E6BT, UK.
| | | | | |
Collapse
|
15
|
Kirui DK, Rey DA, Batt CA. Gold hybrid nanoparticles for targeted phototherapy and cancer imaging. NANOTECHNOLOGY 2010; 21:105105. [PMID: 20154383 DOI: 10.1088/0957-4484/21/10/105105] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Gold and iron oxide hybrid nanoparticles (HNPs) synthesized by the thermal decomposition technique are bio-functionalized with a single chain antibody, scFv, that binds to the A33 antigen present on colorectal cancer cells. The HNP-scFv conjugates are stable in aqueous solution with a magnetization value of 44 emu g(-1) and exhibit strong optical absorbance at 800 nm. Here we test this material in targeting, imaging and selective thermal killing of colorectal cancer cells. Cellular uptake studies showed that A33-expressing cells take up the A33scFv-conjugated HNPs at a rate five times higher than cells that do not express the A33 antigen. Laser irradiation studies showed that approximately 53% of the A33-expressing cells exposed to targeted HNPs are killed after a six-minute laser treatment at 5.1 W cm(-2) using a 808 nm continuous wave laser diode while < 5% of A33-nonexpressing cells are killed. At a higher intensity, 31.5 W cm(-2), the thermal destruction increases to 99 and 40% for A33-expressing cells and A33 nonexpressing cells, respectively, after 6 min exposure. Flow cytometric analyses of the laser-irradiated A33 antigen-expressing cells show apoptosis-related cell death to be the primary mode of cell death at 5.1 W cm(-2), with increasing necrosis-related cell death at higher laser power. These results suggest that this new class of bio-conjugated hybrid nanoparticles can potentially serve as an effective antigen-targeted photothermal therapeutic agent for cancer treatment as well as a probe for magnetic resonance-based imaging.
Collapse
Affiliation(s)
- Dickson K Kirui
- Graduate Field of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| | | | | |
Collapse
|
16
|
Park JI, Cao L, Platt VM, Huang Z, Stull RA, Dy EE, Sperinde JJ, Yokoyama JS, Szoka FC. Antitumor therapy mediated by 5-fluorocytosine and a recombinant fusion protein containing TSG-6 hyaluronan binding domain and yeast cytosine deaminase. Mol Pharm 2009; 6:801-12. [PMID: 19265397 DOI: 10.1021/mp800013c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Matrix attachment therapy (MAT) is an enzyme prodrug strategy that targets hyaluronan in the tumor extracellular matrix to deliver a prodrug converting enzyme near the tumor cells. A recombinant fusion protein containing the hyaluronan binding domain of TSG-6 (Link) and yeast cytosine deaminase (CD) with an N-terminal His(x6) tag was constructed to test MAT on the C26 colon adenocarcinoma in Balb/c mice that were given 5-fluorocytosine (5-FC) in the drinking water. LinkCD was expressed in Escherichia coli and purified by metal-chelation affinity chromatography. The purified LinkCD fusion protein exhibits a K(m) of 0.33 mM and V(max) of 15 microM/min/microg for the conversion of 5-FC to 5-fluorouracil (5-FU). The duration of the enzyme activity for LinkCD was longer than that of CD enzyme at 37 degrees C: the fusion protein retained 20% of its initial enzyme activity after 24 h, and 12% after 48 h. The LinkCD fusion protein can bind to a hyaluronan oligomer (12-mer) at a K(D) of 55 microM at pH 7.4 and a K(D) of 5.32 microM at pH 6.0 measured using surface plasmon resonance (SPR). To evaluate the antitumor effect of LinkCD/5-FC combination therapy in vivo, mice received intratumoral injections of LinkCD on days 11 and 14 after C26 tumor implantation and the drinking water containing 10 mg/mL of 5-FC starting on day 11. To examine if the Link domain by itself was able to reduce tumor growth, we included treatment groups that received LinkCD without 5-FC and Link-mtCD (a functional mutant that lacks cytosine deaminase activity) with 5-FC. Animals that received LinkCD/5-FC treatment showed significant tumor size reduction and increased survival compared to the CD/5-FC treatment group. Treatment groups that were unable to produce 5-FU had no effect on the tumor growth despite receiving the fusion protein that contained the Link domain. The results indicate that a treatment regime consisting of a fusion protein containing the Link domain, the active CD enzyme, and the prodrug 5-FC is sufficient to produce an antitumor effect. Thus, the LinkCD fusion protein is an alternative to antibody-directed prodrug enzyme therapy (ADEPT) approaches for cancer treatment.
Collapse
Affiliation(s)
- Joshua I Park
- Department of Pharmaceutical Chemistry and Biopharmaceutical Sciences, University of California, San Francisco, California 94143-0912, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sun PH, Wu YL, Dong WJ, Qiao MM, Zhu LM, Tu SP. Construction of a survivin promoter-mediated adeno-associated virus expression vector carrying a fusion gene constituted with cytosine deaminase and survivin mutant. Shijie Huaren Xiaohua Zazhi 2009; 17:1085-1090. [DOI: 10.11569/wcjd.v17.i11.1085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct a recombinant survivin promoter (SurP)-mediated adeno-associated virus expression vector carrying a fusion gene constituted with cytosine deaminase (CD) gene and glycin-linked survivin mutant (glySurMut), and to examine the expression of CDglySurMut fusion protein in cancer cells transfected with this vector.
METHODS: The CD gene and glySurMut fragments were amplified by polymerase chain reaction (PCR), and cloned into the adeno-associated virus expression vector PAM with survivin promoter (PAM/SurP). The recombinant vector was transformed into E.coli DH5α. and the positive clones were selected. The plasmid DNA was identified using restriction enzyme analysis and sequencing. The recombinant vector pAM/SurP-CDglySurMut was transfected into gastric cancer cells MKN45. The expression of fusion protein in cancer cells was detected using Western blot.
RESULTS: The CD gene and glySurMut were cloned from E.coli and gastric cancer cells, respectively. The restrictive enzyme analysis and sequencing showed that the fusion gene of CD and glySurMut was successfully inserted into the PAM/SurP vector. Western blot showed that the recombinant vector expressed the CDglySurMut fusion protein in gastric cancer cells.
CONCLUSION: Our results show the pAM/SurP-CDglySurMut vector can efficiently express the recombinant CDglySurMut fusion protein in gastric cancer cells, thus provides experiment evidence for targeting cancer gene therapy using this pAM/SurP-CDglySurMut vector.
Collapse
|
18
|
Damasceno LM, Lee F, Ritter G, Old L, Batt C. High-level expression of a phage display-derived scFv in Pichia pastoris. Methods Mol Biol 2009; 562:225-236. [PMID: 19554300 DOI: 10.1007/978-1-60327-302-2_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Numerous techniques are available for investigating protein-ligand interactions. The phage display technique is one such method routinely used to identify antibody-antigen interactions and has the benefit of being easily adaptable to high-throughput screening platforms. Once identified, antigen-binding domains on fragment antibodies or single-chain fragment antibodies (scFv) can be expressed and purified for further studies. In this chapter, we describe a method for high-level expression of a phage display-derived scFv in Pichia pastoris. The phage display-derived antibody A33scFv recognizes a cell surface glycoprotein (designated A33) expressed in colon cancer that serves as a target antigen for radioimmunoimaging and/or immunotherapy of human colon cancer. The expression and purification of A33scFv was optimized for the methylotrophic yeast P. pastoris. P. pastoris with a Mut(S) phenotype was selected to express A33scFv under regulation of the methanol-inducible AOX1 promoter. Here we describe a large-scale fed-batch fermentation process with an efficient online closed-loop methanol control for the production of the recombinant protein. Purification of A33scFv from clarified culture medium was done using a two-step chromatographic procedure using anion exchange and hydrophobic interaction chromatography, resulting in a final product with more than 90% purity. This chapter provides protocols that can be used as a base for process development of recombinant protein expression in P. pastoris and purification of these proteins for use in further functionality studies and in diagnostic and therapeutic applications.
Collapse
|
19
|
Frey D, Coelho V, Petrausch U, Schaefer M, Keilholz U, Thiel E, Deckert PM. Surface expression of gpA33 is dependent on culture density and cell-cycle phase and is regulated by intracellular traffic rather than gene transcription. Cancer Biother Radiopharm 2008; 23:65-73. [PMID: 18298330 DOI: 10.1089/cbr.2007.0407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The cell-surface marker, gpA33, a new member of the immunoglobulin superfamily, is expressed by gastrointestinal cells and by 95% of colon cancers. It has become a promising target of immunologic therapy strategies, but its biologic function and potential role in tumorigenesis are unknown. In this study, we have investigated the expression of gpA33 on the mRNA and cell-surface protein levels by quantitative reverse transcriptase polymerase chain reaction and flow cytometry, respectively, in response to cell density in the culture and to cell-cycle arrest in the G1, S, or G2/M phases. As internalization of the surface protein had previously been reported, we also investigated the binding and intracellular migration of an anti-gpA33 fluobody with green fluorescent protein (A33scFv::GFP) by laser confocal microscopy. Contrary to intuition, we found that gpA33 surface expression and mRNA levels do only partly correlate under the conditions tested. Dependent on cell density in culture, gpA33 surface expression peaked at the point of confluence. Dependent on cell-cycle phase, it peaked in the G2/M phase but was lowest in the S phase, whereas mRNA levels were highest in S, but almost absent in G1. Laser confocal microscopy clearly demonstrated the intracellular uptake of A33scFv::GFP and showed the migration of microvesicles over time. These findings are, in part, concordant with the putative role of gpA33 as an adhesion molecule. However, intracellular traffic and recycling to the cell surface appear to play a major role in its function and to have an influence on its surface density superseding translational regulation.
Collapse
Affiliation(s)
- Dietmar Frey
- Centrum für Tumormedizin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
20
|
Panjideh H, Coelho V, Dernedde J, Fuchs H, Keilholz U, Thiel E, Deckert PM. Production of bifunctional single-chain antibody-based fusion proteins in Pichia pastoris supernatants. Bioprocess Biosyst Eng 2008; 31:559-68. [PMID: 18253756 DOI: 10.1007/s00449-008-0203-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 01/16/2008] [Indexed: 11/26/2022]
Abstract
Recombinant antibody fusion constructs with heterologous functional domains are a promising approach to new therapeutic targeting strategies. However, expression of such constructs is mostly limited to cost and labor-intensive mammalian expression systems. Here we report on the employment of Pichia pastoris for the expression of heterologous antibody fusion constructs with green fluorescent protein, A33scFv::GFP, or with cytosine deaminase, A33scFv::CDy, their production in a biofermenter and a modified purification strategy. Combined, these approaches improved production yields by about thirty times over established standard protocols, with extracellular secretion of the fusion construct reaching 12.0 mg/l. Bifunctional activity of the fusion proteins was demonstrated by flow cytometry and an in-vitro cytotoxicity assay. With equal amounts of purified protein, the modified purification method lead to higher functional results. Our results demonstrate the suitability of methylotrophic Pichia expression systems and laboratory-scale bioreactors for the production of high quantities of bifunctionally active heterologous single-chain fusion proteins.
Collapse
Affiliation(s)
- Hossein Panjideh
- Medizinische Klinik III, Hematology, Oncology und Transfusion Medicine, Charité, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
21
|
Ou-Yang F, Lan KL, Chen CT, Liu JC, Weng CL, Chou CK, Xie X, Hung JY, Wei Y, Hortobagyi GN, Hung MC. Endostatin-cytosine deaminase fusion protein suppresses tumor growth by targeting neovascular endothelial cells. Cancer Res 2006; 66:378-84. [PMID: 16397252 DOI: 10.1158/0008-5472.can-05-1578] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endostatin, an angiogenesis inhibitor tested in multiple clinical trials, selectively targets neovascular endothelial cells, suppressing tumor growth. To enhance the therapeutic efficacy of endostatin, we fused endostatin with cytosine deaminase, which converts a prodrug 5-flucytosine into a cytotoxic 5-fluorouracil. This therapeutic strategy was developed based on the observation that the endostatin-green fluorescence protein gene and endostatin-luciferase gene selectively target to endothelial cells in vitro and to the tumor site in vivo, respectively. When we used the endostatin-cytosine deaminase fusion protein to treat s.c. grafted tumors or experimental metastasis tumors, our results showed that endostatin-cytosine deaminase treatment provided stronger tumor growth suppression and increased mean survival time of the mice compared with the treatments of endostatin alone, cytosine deaminase alone, or endostatin plus cytosine deaminase. The endostatin-cytosine deaminase protein significantly inhibited the growth of endothelial cells and preferentially induced tumor cell apoptosis. This endostatin-cytosine deaminase fusion approach opens an avenue for cancer-targeting therapy.
Collapse
Affiliation(s)
- Fu Ou-Yang
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hillemann A, Brandenburg B, Schmidt U, Roos M, Smirnow I, Lemken ML, Lauer UM, Hildt E. Protein transduction with bacterial cytosine deaminase fused to the TLM intercellular transport motif induces profound chemosensitivity to 5-fluorocytosine in human hepatoma cells. J Hepatol 2005; 43:442-50. [PMID: 15922474 DOI: 10.1016/j.jhep.2005.02.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2004] [Revised: 02/12/2005] [Accepted: 02/16/2005] [Indexed: 01/09/2023]
Abstract
BACKGROUND/AIMS This study investigates the application of protein based therapeutic suicide enzyme/prodrug approaches providing novel means for both safe and effective local therapeutic regimes in solid tumors. METHODS Employing a novel cell permeable peptide, known as the translocation motif (TLM) of hepatitis B virus, E. coli cytosine deaminase (BCDase) suicide fusion proteins were generated. RESULTS TLM fusion proteins formed hexamers (as do parental wtBCDase proteins) and retained the specific enzymatic activity of cytosine conversion to uracil also being comparable to parental wtBCDase proteins. However, only BCDase-TLM fusion proteins, but not TLM-BCDase fusion nor parental wtBCDase proteins were found to be taken up to the cytoplasm of target cells as demonstrated both by confocal laser scanning microscopy and cell fractionation. Uptake of BCDase-TLM worked both efficiently and rapidly and was found to be independent from the endosomal pathway. Since BCDase-TLM fusion proteins completely retained their suicide enzymatic activity in the course of translocation across the plasma membrane their usage as profound inducers of chemo-sensitivity to 5-FC strongly is suggested. CONCLUSIONS Future therapeutic local application of cell-permeable BCDase-TLM fusion proteins together with a systemic 5-FC prodrug application could result in profound antitumor activities without apparent side effects.
Collapse
|
23
|
Damasceno LM, Pla I, Chang HJ, Cohen L, Ritter G, Old LJ, Batt CA. An optimized fermentation process for high-level production of a single-chain Fv antibody fragment in Pichia pastoris. Protein Expr Purif 2005; 37:18-26. [PMID: 15294276 DOI: 10.1016/j.pep.2004.03.019] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Revised: 03/09/2004] [Indexed: 10/26/2022]
Abstract
The expression of a humanized single-chain variable domain fragment antibody (A33scFv) was optimized for Pichia pastoris with yields exceeding 4 g L(-1). A33scFv recognizes a cell surface glycoprotein (designated A33) expressed in colon cancer that serves as a target antigen for immunotherapy of colon cancer. P. pastoris with a MutS phenotype was selected to express A33scFv, which was cloned under regulation of the methanol-inducible AOX1 promoter. We report the optimization of A33scFv production by examining methanol concentrations using fermentation technology with an on-line methanol control in fed-batch fermentation of P. pastoris. In addition, we examined the effect of pH on A33scFv production and biomass accumulation during the methanol induction phase. A33scFv production was found to increase with higher methanol concentrations, reaching 4.3 g L(-1) after 72 h induction with 0.5% (v/v) methanol. Protein production was also greatly affected by pH, resulting in higher yields (e.g., 4.88 g L(-1)) at lower pH values. Biomass accumulation did not seem to vary when cells were induced at different pH values, but was greatly affected by lower concentration of methanol. Purification of A33scFv from clarified medium was done using a two-step chromatographic procedure using anion-exchange and hydrophobic interaction chromatography, resulting in 25% recovery and >90% purity. Pure A33scFv was tested for functionality using surface plasmon resonance and showed activity against immobilized A33 antigen. Our results demonstrate that functional A33scFv can be produced in sufficient quantities using P. pastoris for use in further functionality studies and diagnostic applications.
Collapse
|
24
|
Ton GN, Weichert JP, Longino MA, Fine JP, Kwon GS. Methoxypoly(ethylene glycol)-conjugated carboxypeptidase A for solid tumor targeting. J Control Release 2005; 104:155-66. [PMID: 15866342 DOI: 10.1016/j.jconrel.2005.01.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2004] [Revised: 01/25/2005] [Accepted: 01/26/2005] [Indexed: 11/16/2022]
Abstract
We have evaluated effects of mPEG modification on pharmacokinetic properties of carboxypeptidase A (CPA) in normal rats. Attachment of two or three mPEG chains to CPA resulted in the generation of mPEG2-CPA and mPEG3-CPA analogs with significantly enhanced plasma half-lives, especially during the distribution phase. Moreover, the assessment of real-time whole-body kinetics in CT26 tumor-bearing mice showed both mPEG2-CPA and mPEG3-CPA exhibited increased body retention at 48 h post-injection. In addition, tumor localization of mPEG3-CPA at 72 h was visualized and confirmed by fusion of the gamma-scintigraphy and microCT data sets. Results from the imaging studies support our hypothesis of a correlation between tumor uptake and enhanced circulatory half-life. Tissue distribution data indicated the combination of increased tumor extravasation and effective renal elimination observed with mPEG2-CPA at 48 h following administration led to the highest observed tumor-to-blood ratio of 4.8:1. Although the total concentration of mPEG3-CPA accumulated in tumor was higher than that of mPEG2-CPA and CPA at predetermined time intervals, a higher tumor-to-blood ratio was not obtained owing to a higher level of blood activity. Clearly, the attachment of an appropriate number of mPEG chains can facilitate tumor localization as effectively as can the use of a tumor-specific antibody.
Collapse
Affiliation(s)
- Giangthy N Ton
- Department of Radiology, University of Wisconsin, K4/644 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792-3252, USA
| | | | | | | | | |
Collapse
|
25
|
Asai T, Wims LA, Morrison SL. An interaction between S*tag and S*protein derived from human ribonuclease 1 allows site-specific conjugation of an enzyme to an antibody for targeted drug delivery. J Immunol Methods 2005; 299:63-76. [PMID: 15914191 DOI: 10.1016/j.jim.2005.01.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Revised: 01/14/2005] [Accepted: 01/15/2005] [Indexed: 11/20/2022]
Abstract
We have previously demonstrated that an antibody-avidin fusion protein could be used to deliver biotinylated enzymes to tumor cells for antibody-directed enzyme prodrug therapy. However, the presence of the chicken protein avidin suggests that immunogenicity may be a problem. To address this concern, we developed a new delivery system consisting of human proteins. The amino-terminal 15-amino-acid peptide derived from human ribonuclease 1 (human S*tag) can bind with high affinity to human S*protein (residues 21-124 of the same ribonuclease). We constructed an antibody-S*protein fusion protein in which S*protein was genetically linked to an anti-rat transferrin receptor IgG3 at the carboxyl terminus of the heavy chain. We also constructed an enzyme-S*tag fusion protein in which S*tag was genetically linked to the carboxyl terminus of Escherichia coli purine nucleoside phosphorylase (PNP). When these two fusion proteins were mixed, S*tag and S*protein interacted specifically and produced homogeneous antibody/PNP complexes that retained the ability to bind antigen. Furthermore, in the presence of the prodrug 2-fluoro-2'-deoxyadenosine in vitro, the complex efficiently killed rat myeloma cells overexpressing the transferrin receptor. These results suggest that human ribonuclease-based site-specific conjugation can be used in vivo for targeted chemotherapy of cancer.
Collapse
Affiliation(s)
- Tsuneaki Asai
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, 405 Hilgard Avenue, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
26
|
Abstract
In the year 2003 there was a 17% increase in the number of publications citing work performed using optical biosensor technology compared with the previous year. We collated the 962 total papers for 2003, identified the geographical regions where the work was performed, highlighted the instrument types on which it was carried out, and segregated the papers by biological system. In this overview, we spotlight 13 papers that should be on everyone's 'must read' list for 2003 and provide examples of how to identify and interpret high-quality biosensor data. Although we still find that the literature is replete with poorly performed experiments, over-interpreted results and a general lack of understanding of data analysis, we are optimistic that these shortcomings will be addressed as biosensor technology continues to mature.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
27
|
Asai T, Trinh R, Ng PP, Penichet ML, Wims LA, Morrison SL. A human biotin acceptor domain allows site-specific conjugation of an enzyme to an antibody-avidin fusion protein for targeted drug delivery. ACTA ACUST UNITED AC 2004; 21:145-55. [PMID: 15748688 DOI: 10.1016/j.bioeng.2004.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Revised: 10/21/2004] [Accepted: 10/22/2004] [Indexed: 11/19/2022]
Abstract
We have previously constructed an antibody-avidin (Av) fusion protein, anti-transferrin receptor (TfR) IgG3-Av, which can deliver biotinylated molecules to cells expressing the TfR. We now describe the use of the fusion protein for antibody-directed enzyme prodrug therapy (ADEPT). The 67 amino acid carboxyl-terminal domain (P67) of human propionyl-CoA carboxylase alpha subunit can be metabolically biotinylated at a fixed lysine residue. We genetically fused P67 to the carboxyl terminus of the yeast enzyme FCU1, a derivative of cytosine deaminase that can convert the non-toxic prodrug 5-fluorocytosine to the cytotoxic agent 5-fluorouracil. When produced in Escherichia coli cells overexpressing a biotin protein ligase, the FCU1-P67 fusion protein was efficiently mono-biotinylated. In the presence of 5-fluorocytosine, the biotinylated fusion protein conjugated to anti-rat TfR IgG3-Av efficiently killed rat Y3-Ag1.2.3 myeloma cells in vitro, while the same protein conjugated to an irrelevant (anti-dansyl) antibody fused to Av showed no cytotoxic effect. Efficient tumor cell killing was also observed when E. coli purine nucleoside phosphorylase was similarly targeted to the tumor cells in the presence of the prodrug 2-fluoro-2'-deoxyadenosine. These results suggest that when combined with P67-based biotinylation, anti-TfR IgG3-Av could serve as a universal delivery vector for targeted chemotherapy of cancer.
Collapse
Affiliation(s)
- Tsuneaki Asai
- Department of Microbiology, Molecular Genetics and Molecular Biology Institute, University of California Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|