101
|
Ikeda Y, Taira K. Ligand-Targeted Delivery of Therapeutic siRNA. Pharm Res 2006; 23:1631-40. [PMID: 16850274 DOI: 10.1007/s11095-006-9001-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Accepted: 03/06/2006] [Indexed: 10/24/2022]
Abstract
RNA interference (RNAi) is a post-transcriptional gene-silencing phenomenon that is triggered by double-stranded RNA (dsRNA). Since many diseases are associated with the inappropriate production of specific proteins, attempts are being made to exploit RNAi in a clinical settings. However, before RNAi can be exploited as therapeutically, several obstacles must be overcome. For example, small interfering RNA (siRNA) is unstable in the blood stream so any effects of injected siRNA are only transient. Accordingly, methods must be developed to prolong its activity. Furthermore, the efficient and safe delivery of siRNA into target tissues and cells is critical for successful therapy. Any useful delivery method should be designed to target siRNA to specific cells and to promote gene-silencing activity once the siRNA is inside the cells. Recent chemical modifications of siRNA have overcome problems associated with the instability of siRNA, and various ligands, including glycosylated molecules, peptides, proteins, antibodies and engineered antibody fragments, appear to be very useful or have considerable potential for the targeted delivery of siRNA. The use of such ligands improves the efficiency, specificity and, as a consequence, the safety of the corresponding delivery systems.
Collapse
Affiliation(s)
- Yutaka Ikeda
- Gene Function Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-1 Higashi, 305-8562 Tsukuba Science City, Japan
| | | |
Collapse
|
102
|
Hu S, Li L, Qiao J, Guo Y, Cheng L, Liu J. Codon optimization, expression, and characterization of an internalizing anti-ErbB2 single-chain antibody in Pichia pastoris. Protein Expr Purif 2006; 47:249-57. [PMID: 16403645 DOI: 10.1016/j.pep.2005.11.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2005] [Revised: 11/10/2005] [Accepted: 11/17/2005] [Indexed: 10/25/2022]
Abstract
Anti-ErbB2 antibodies are used as convenient tools in exploration of ErbB2 functional mechanisms and in treatment of ErbB2-overexpressing tumors. When we employed the yeast Pichia pastoris to express an anti-ErbB2 single-chain antibody (scFv) derived from the tumor-inhibitory monoclonal antibody A21, the yield did not exceed 1-2 mg/L in shake flask cultures. As we considered that the poor codon usage bias may be one limiting factor leading to the inefficient translation and scFv production, we designed and synthesized the full-length scFv gene by choosing the P. pastoris preferred codons while keeping the G+C content at relatively low level. Codon optimization increased the scFv expression level 3- to 5-fold and up to 6-10 mg/L. Northern blotting further confirmed that the increase of scFv expression was mainly due to the enhancement of translation efficiency. Investigation of culture conditions revealed that the maximal cell growth and scFv expression were achieved at pH 6.5-7.0 with 2% casamino acids after 72 h methanol induction. Secreted scFv was easily purified (>95% homogeneous product) from culture supernatants in one step by using Ni2+ chelating affinity chromatography. The yield was approximately 10-15 mg/L. Functional studies showed that the A21 scFv could be internalized with high efficiency after binding to the ErbB2-overexpressing cells, suggesting this regent may prove especially useful for ErbB2-targeted immunotherapy.
Collapse
Affiliation(s)
- Siyi Hu
- Laboratory of Molecular and Cellular Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | | | | | | | | | | |
Collapse
|
103
|
Horak E, Heitner T, Robinson MK, Simmons HH, Garrison J, Russeva M, Furmanova P, Lou J, Zhou Y, Yuan QA, Weiner LM, Adams GP, Marks JD. Isolation of scFvs to in vitro produced extracellular domains of EGFR family members. Cancer Biother Radiopharm 2006; 20:603-13. [PMID: 16398612 DOI: 10.1089/cbr.2005.20.603] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The members of the epidermal growth factor receptor (EGFR) family are over expressed in a variety of malignancies and are frequently linked to aggressive disease and a poor prognosis. Although clinically effective monoclonal antibodies (MAbs) have been developed to target HER2 and EGFR, the remaining two family members, HER3 and HER4, have not been the subject of significant efforts. In this paper, we have taken the initial steps required to generate antibodies with potential clinically utility that target the members of the EGFR family. The genes for the extracellular domains (ECDs) of all four members of the EGFR family were cloned and used to stably transfect 293 (HEK) cells. Milligram quantities of each ECD were produced and characterized. The HER3, HER4, and EGFR ECDs were then employed as targets for the selection of antibodies from naïve human scFv (single-chain Fv) phage display libraries. Six unique scFv clones were isolated that bound specifically to HER3, 13 unique clones were isolated with specificity for HER4 and 52 unique anti-EGFR clones were isolated. These scFvs provide a valuable and potentially clinically relevant panel of agents to target the members of the EGFR family.
Collapse
Affiliation(s)
- Eva Horak
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Abstract
Antibodies constitute the most rapidly growing class of human therapeutics and the second largest class of drugs after vaccines. The generation of potent antibody therapeutics, which I review here, is an iterative design process that involves the generation and optimization of antibodies to improve their clinical potential.
Collapse
Affiliation(s)
- Paul J Carter
- Department of Antibody Technologies, Seattle Genetics Incorporated, 21823 30th Drive South East, Bothell, Washington 98021, USA.
| |
Collapse
|
105
|
Hackel BJ, Huang D, Bubolz JC, Wang XX, Shusta EV. Production of Soluble and Active Transferrin Receptor-Targeting Single-Chain Antibody using Saccharomyces cerevisiae. Pharm Res 2006; 23:790-7. [PMID: 16550469 DOI: 10.1007/s11095-006-9778-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Accepted: 12/16/2005] [Indexed: 11/25/2022]
Abstract
PURPOSE This study describes the soluble production, purification, and functional testing of an anti-transferrin receptor single-chain antibody (OX26 scFv) using the yeast Saccharomyces cerevisiae. METHODS The yeast secretion apparatus was optimized by modulating expression temperature, the folding environment of the endoplasmic reticulum, and gene dosage. Secreted scFv was purified using immobilized metal affinity chromatography, and tested for binding and internalization into the RBE4 rat brain endothelial cell line. RESULTS Secretion of OX26 scFv was optimal when expression was induced at 20 degrees C. Co-overexpression of heavy chain binding protein and protein disulfide isomerase elevated scFv expression levels by 10.4 +/- 0.3-fold. Optimization of scFv gene dosage increased secretion by 7.1 +/- 0.2-fold, but the overall benefits of binding protein and protein disulfide isomerase overexpression were diminished. Purified OX26 scFv yields of 0.5 mg/L secreted protein were achieved, and the scFv was actively internalized into RBE4 cells with a pattern similar to that observed with intact OX26 monoclonal antibody. CONCLUSIONS The optimized S. cerevisiae expression system is amenable to production of soluble and active brain targeting OX26 scFv, and the yeast-produced scFv has potential for the targeting and delivery of small molecules, proteins, or drug carriers across the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Benjamin J Hackel
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, Wisconsin 53706, USA
| | | | | | | | | |
Collapse
|
106
|
Hu S, Guo X, Xie H, Du Y, Pan Y, Shi Y, Wang J, Hong L, Han S, Zhang D, Huang D, Zhang K, Bai F, Jiang H, Zhai H, Nie Y, Wu K, Fan D. Phage display selection of peptides that inhibit metastasis ability of gastric cancer cells with high liver-metastatic potential. Biochem Biophys Res Commun 2006; 341:964-72. [PMID: 16458253 DOI: 10.1016/j.bbrc.2006.01.047] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2006] [Accepted: 01/10/2006] [Indexed: 11/28/2022]
Abstract
Organ-specific metastasis is an important character of cancer cells. Cancer cells that can metastasize to a special organ were thought to have different proteins in cell membrane, which might have potential utility as diagnostic markers and therapeutic targets. In the present work, based on high liver-metastatic gastric cancer cells, XGC9811-L, a screening approach with phage displayed peptide library, was successfully used to isolate 8-mer peptide ligands binding to the target cells. The phage20 had the highest binding efficiency to XGC9811-L cells, which also displayed remarkable cell specificity. Peptide20 that was displayed on phage20 could suppress the motility and invasion of XGC9811-L significantly. The adhesive ability of XGC9811-L to collagen IV was also inhibited by peptide20. Furthermore, phage20 could significantly reduce the incidence of liver metastasis of gastric cancer transplanted into nude mice and was also beneficial for the reduction the number of metastatic nodules in the liver. In conclusion, the phage display is an effective method to screen for the new molecules associated with organ-specific metastasis. The selected peptide20 can reverse the liver metastasis behavior of the gastric cancer cells.
Collapse
Affiliation(s)
- Shengjuan Hu
- State Key Laboratory of GI Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi Province 710032, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Noble CO, Kirpotin DB, Hayes ME, Mamot C, Hong K, Park JW, Benz CC, Marks JD, Drummond DC. Development of ligand-targeted liposomes for cancer therapy. Expert Opin Ther Targets 2006; 8:335-53. [PMID: 15268628 DOI: 10.1517/14728222.8.4.335] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The continued evolution of targeted liposomal therapeutics has resulted in new agents with remarkable antitumour efficacy and relatively mild toxicity profiles. A careful selection of the ligand is necessary to reduce immunogenicity, retain extended circulation lifetimes, target tumour-specific cell surface epitopes, and induce internalisation and subsequent release of the therapeutic substance from the liposome. Methods for assembling targeted liposomes, including a novel micellar insertion technology, for incorporation of targeting molecules that efficiently transforms a non-targeted liposomal therapeutic to a targeted one, greatly assist the translation of targeted liposome technology into the clinic. Targeting strategies with liposomes directed at solid tumours and vascular targets are discussed. The authors believe the development of ligand-targeted liposomes is now in the advanced stage and offers unique and important advantages among other targeted therapies. Anti-HER2 immunoliposomal doxorubicin is awaiting Phase I clinical trials, the results of which should provide new insights into the promise of ligand-targeted liposomal therapies.
Collapse
|
108
|
Fransson J, Tornberg UC, Borrebaeck CAK, Carlsson R, Frendéus B. Rapid induction of apoptosis in B-cell lymphoma by functionally isolated human antibodies. Int J Cancer 2006; 119:349-58. [PMID: 16477633 DOI: 10.1002/ijc.21829] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Novel panning and screening methodology was devised to isolate high affinity human recombinant scFv antibody fragments with functionally associated properties in B lymphoma cells. The approach was used to generate a panel of apoptosis-inducing antibodies specific for antigens differentially expressed in B lymphoma vs. T leukaemia cells. The selections resulted in an antibody pool with near perfect selectivity (>99%) for the B lymphoma target cells. Randomly picked clones (72) revealed 7 unique antibody genotypes. Six of these rapidly induced apoptosis in target cells. Following the conversion to full IgGs, the antibodies were shown to be specific for HLA-DR/DP, the B-cell receptor mu chain and for CD54/ICAM-1. The latter receptor was not previously associated with apoptotic properties in B-cell lymphomas. Anti-ICAM-1 IgG induced apoptosis in a broad range of B lymphoma cell lines and were shown by immunohistochemistry to bind strongly to B lymphoma tissue obtained from 5 different B lymphoma patients. The recombinant IgG antibodies had affinities in the subnanomolar (0.3 nM) to nanomolar (3 nM) range. The described technology is generally applicable for the rapid isolation of high affinity human antibodies with specificity for differentially expressed cell surface receptors with intrinsic negative or positive signalling properties from naïve phage libraries.
Collapse
Affiliation(s)
- Johan Fransson
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | | | | | | |
Collapse
|
109
|
Zanghi CN, Lankes HA, Bradel-Tretheway B, Wegman J, Dewhurst S. A simple method for displaying recalcitrant proteins on the surface of bacteriophage lambda. Nucleic Acids Res 2005; 33:e160. [PMID: 16224099 PMCID: PMC1258178 DOI: 10.1093/nar/gni158] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Bacteriophage lambda (λ) permits the display of many foreign peptides and proteins on the gpD major coat protein. However, some recombinant derivatives of gpD are incompatible with the assembly of stable phage particles. This presents a limitation to current λ display systems. Here we describe a novel, plasmid-based expression system in which gpD deficient λ lysogens can be co-complemented with both wild-type and recombinant forms of gpD. This dual expression system permits the generation of mosaic phage particles that contain otherwise recalcitrant recombinant gpD fusion proteins. Overall, this improved gpD display system is expected to permit the expression of a wide variety of peptides and proteins on the surface of bacteriophage λ and to facilitate the use of modified λ phage vectors in mammalian gene transfer applications.
Collapse
Affiliation(s)
- Christine N. Zanghi
- Department of Microbiology and Immunology, University of Rochester Medical Center601 Elmwood Avenue, Box 672, Rochester, NY 14642, USA
| | - Heather A. Lankes
- Department of Microbiology and Immunology, University of Rochester Medical Center601 Elmwood Avenue, Box 672, Rochester, NY 14642, USA
| | - Birgit Bradel-Tretheway
- Department of Microbiology and Immunology, University of Rochester Medical Center601 Elmwood Avenue, Box 672, Rochester, NY 14642, USA
| | - Jessica Wegman
- Department of Microbiology and Immunology, University of Rochester Medical Center601 Elmwood Avenue, Box 672, Rochester, NY 14642, USA
| | - Stephen Dewhurst
- Department of Microbiology and Immunology, University of Rochester Medical Center601 Elmwood Avenue, Box 672, Rochester, NY 14642, USA
- Cancer Center, University of Rochester School of Medicine and DentistryRochester, NY 14642, USA
- To whom correspondence should be addressed. Tel: +1 585 275 3216; Fax: +1 585 473 2361;
| |
Collapse
|
110
|
Askoxylakis V, Zitzmann S, Mier W, Graham K, Krämer S, von Wegner F, Fink RHA, Schwab M, Eisenhut M, Haberkorn U. Preclinical Evaluation of the Breast Cancer Cell-Binding Peptide, p160. Clin Cancer Res 2005; 11:6705-12. [PMID: 16166451 DOI: 10.1158/1078-0432.ccr-05-0432] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Selective delivery of drugs into the target tissue is expected to result in high drug concentrations in the tissue of interest and therefore enhanced drug efficacy. To develop a peptide-based radiopharmaceutical, we investigated the properties of a peptide with affinity for human breast cancer, which has been selected through phage display. EXPERIMENTAL DESIGN The bioactivity of the p160 peptide (VPWMEPAYQRFL) was evaluated in vitro and in vivo. The specific binding to human breast cancer MDA-MB-435 cells was confirmed in competition experiments. Internalization of the peptide was investigated with confocal microscopy. Furthermore, the biodistribution of (131)I-labeled p160 was studied in tumor-bearing mice. In vivo stability was evaluated at different periods after tracer administration using high-performance liquid chromatography analysis. RESULTS The binding of (125)I-labeled p160 was inhibited up to 95% by the unlabeled peptide with an IC(50) value of 0.6 micromol/L. In addition, 40% of the total bound activity was found to be internalized into the human breast cancer cells. Although a rapid degradation was seen, biodistribution studies in nude mice showed a higher uptake in tumor than in most of the organs. Perfusion of the animals caused a reduction of the radioligand accumulation in the healthy tissues, whereas the tumor uptake remained constant. A comparison of [(131)I]p160 with a (131)I-labeled Arg-Gly-Asp peptide revealed a higher tumor-to-organ ratio for [(131)I]p160. CONCLUSIONS p160 has properties that make it an attractive carrier for tumor imaging and the intracellular delivery of isotopes or chemotherapeutic drugs.
Collapse
Affiliation(s)
- Vasileios Askoxylakis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Razai A, Garcia-Rodriguez C, Lou J, Geren IN, Forsyth CM, Robles Y, Tsai R, Smith TJ, Smith LA, Siegel RW, Feldhaus M, Marks JD. Molecular Evolution of Antibody Affinity for Sensitive Detection of Botulinum Neurotoxin Type A. J Mol Biol 2005; 351:158-69. [PMID: 16002090 DOI: 10.1016/j.jmb.2005.06.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Revised: 05/26/2005] [Accepted: 06/01/2005] [Indexed: 10/25/2022]
Abstract
Botulism is caused by botulinum neurotoxin (BoNT), the most poisonous substance known. Potential use of BoNT as a biothreat agent has made development of sensitive assays for toxin detection and potent antitoxin for treatment of intoxication a high priority. To improve detection and treatment of botulism, molecular evolution and yeast display were used to increase the affinity of two neutralizing single chain Fv (scFv) antibodies binding BoNT serotype A (BoNT/A). Selection of yeast displayed scFv libraries was performed using methods to select for both increased association rate constant (k(on)) and decreased dissociation rate constants (k(off)). A single cycle of error prone mutagenesis increased the affinity of the 3D12 scFv 45-fold from a K(D) of 9.43x10(-10)M to a K(D) of 2.1x10(-11)M. Affinity of the HuC25 scFv was increased 37-fold from 8.44x10(-10)M to 2.26x10(-11)M using libraries constructed by both random and site directed mutagenesis. scFv variable region genes were used to construct IgG for use in detection assays and in vivo neutralization studies. While IgG had the same relative increases in affinity as scFv, (35-fold and 81-fold, respectively, for 3D12 and HuC25) higher solution equilibrium binding constants were observed for the IgG, with the 3D12 K(D) increasing from 6.07x10(-11)M to 1.71x10(-12)M and the HuC25 K(D) increasing from 4.51x10(-11)M to 5.54x10(-13)M. Affinity increased due to both an increase in k(on), as well as slowing of k(off). Higher affinity antibodies had increased sensitivity, allowing detection of BoNT/A at concentrations as low as 1x10(-13)M. The antibodies will also allow testing of the role of affinity in in vivo toxin neutralization and could lead to the generation of more potent antitoxin.
Collapse
Affiliation(s)
- A Razai
- Department of Anesthesia and Pharmaceutical Chemistry, University of California, San Francisco Rm 3C-38, San Francisco General Hospital, 1001 Potrero Ave, San Francisco, CA 94110, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Nellis DF, Ekstrom DL, Kirpotin DB, Zhu J, Andersson R, Broadt TL, Ouellette TF, Perkins SC, Roach JM, Drummond DC, Hong K, Marks JD, Park JW, Giardina SL. Preclinical manufacture of an anti-HER2 scFv-PEG-DSPE, liposome-inserting conjugate. 1. Gram-scale production and purification. Biotechnol Prog 2005; 21:205-20. [PMID: 15903260 DOI: 10.1021/bp049840y] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A GMP-compliant process is described for producing F5cys-PEG-lipid conjugate. This material fuses with preformed, drug-loaded liposomes, to form "immunoliposomes" that bind to HER2/neu overexpressing carcinomas, stimulates drug internalization, and ideally improves the encapsulated drug's therapeutic index. The soluble, single-chain, variable region antibody fragment, designated F5cys, was produced in E. coli strain RV308 using high-density cultures. Affinity adsorption onto horizontally tumbled Streamline rProtein-A resin robustly recovered F5cys from high-pressure-disrupted, whole-cell homogenates. Two product-related impurity classes were identified: F5cys with mid-sequence discontinuities and F5cys with remnants of a pelB leader peptide. Low-pressure cation exchange chromatography, conducted at elevated pH under reducing conditions, enriched target F5cys relative to these impurities and prepared a C-terminal cysteine for conjugation. Site-directed conjugation, conducted at pH 5.9 +/- 0.1 with reaction monitoring and cysteine quenching, yielded F5cys-MP-PEG(2000)-DSPE. Low-pressure size exclusion chromatography separated spontaneously formed, high-molecular-weight conjugate micelles from low-molecular-weight impurities. When formulated at 1-2 mg/mL in 10 mM trisodium citrate, 10% sucrose (w/v), at pH 6.4 (HCl), the conjugate was stable when stored below -70 degrees C. Six scale-up lots were compared. The largest 40-L culture produced enough F5cys to manufacture 2,085 mg of conjugate, enough to support planned preclinical and future clinical trials. The conjugate was 93% pure, as measured by polyacrylamide gel electrophoresis. Impurities were primarily identified as product-related. Residual endotoxin, rProtein A, and genomic DNA, were at acceptable levels. This study successfully addressed a necessary step in the scale-up of immunoliposome-encapsulated therapeutics.
Collapse
Affiliation(s)
- David F Nellis
- SAIC-Frederick, Inc., National Cancer Institute at Frederick, PO Box B, Frederick, Maryland 21702, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Shukla GS, Krag DN. Phage display selection for cell-specific ligands: development of a screening procedure suitable for small tumor specimens. J Drug Target 2005; 13:7-18. [PMID: 15848950 DOI: 10.1080/10611860400020464] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Phage display technology has been widely used for developing tumor-targeting agents. Most of the efforts were directed towards identifying phage-displayed ligands against cancer-relevant purified targets and cancer cell lines. Whole cell screening procedures typically use a relatively large sample size and are not ideally suited for complex tumor tissues. We describe here a screening protocol that is suitable for non-adherent tumor cells from biopsy specimens. It requires only approximately 20,000 cells/round for biopanning and approximately 10,000 cells/well for subsequent clone binding assessment by ELISA. We standardized the newly developed protocol using erbB2-overexpressing SKBR3 breast cancer cells and compared the results with conventional protocols employing about 10-times more plate-adhered fixed or live cells. The selection rate of SKBR3-binding clones from biopanning approximately 20,000 non-adherent SKBR3 cells by our filter cup protocol was comparable to that obtained from using approximately 200,000 plate-adhered cells. Assessment of clones selected from different phage libraries showed that clones from fixed or live cells, adherent or non-adherent cells, either biopanned in filter cup or plate share specific motifs and binding properties. Some of the clones from each biopanning protocol bound to purified erbB2 and shared motifs with erbB family of receptors and their known ligands. These results demonstrated that the protocol developed in this study was capable of selecting cell-specific ligands using relatively small numbers of cells. Screening cells from a fresh human breast cancer specimen using our protocol showed enrichment of tumor binding clones at successive rounds of selection and some of the selected clones were tumor-specific in comparison to normal breast cells. These protocols have direct application to screen for tumor-binding ligands with small tumor tissue specimens.
Collapse
Affiliation(s)
- Girja S Shukla
- Department of Surgery, Vermont Comprehensive Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | |
Collapse
|
114
|
Piazza T, Cha E, Bongarzone I, Canevari S, Bolognesi A, Polito L, Bargellesi A, Sassi F, Ferrini S, Fabbi M. Internalization and recycling of ALCAM/CD166 detected by a fully human single-chain recombinant antibody. J Cell Sci 2005; 118:1515-25. [PMID: 15769845 DOI: 10.1242/jcs.02280] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activated leukocyte cell adhesion molecule (ALCAM/CD166), a member of the immunoglobulin superfamily with five extracellular immunoglobulin-like domains, promotes heterophilic (ALCAM-CD6) and homophilic (ALCAM-ALCAM) cell-cell interactions. Here we describe a fully human single-chain antibody fragment (scFv) directed to ALCAM/CD166. We selected the I/F8 scFv from a phage display library of human V-gene segments by cell panning and phage internalization into IGROV-I human ovary carcinoma cells. The I/F8 specificity was identified as ALCAM/CD166 by matrix-assisted laser desorption/ionisation time-of-flight (MALDI-TOF) peptide mass fingerprinting of the I/F8-immunoprecipitated protein. The I/F8 scFv reacts with the human, monkey and murine ALCAM/CD166 molecule, indicating that the recognized epitope is highly conserved. The I/F8 scFv completely abolished binding of both ALCAM/Fc and CD6/Fc soluble ligands, whereas it did not compete with the anti-ALCAM/CD166 murine monoclonal antibodies J4-81 and 3A6 and therefore recognizes a different epitope. Engagement through I/F8 scFv, 3A6 monoclonal antibody or CD6/Fc ligand induced ALCAM/CD166 internalization, with a kinetics slower than that of transferrin in the same cells. Newly internalized I/F8-ALCAM complexes colocalized with clathrin but not with caveolin and we demonstrated, using surface biotinylation and recycling assays, that endocytosed ALCAM/CD166 recycles back to the cell surface. Such an endocytic pathway allows the efficient delivery of an I/F8 scFv-saporin immunotoxin into tumor cells, as the conjugates are able to selectively kill cell lines expressing ALCAM/CD166. Altogether these data provide evidence of the suitability of the I/F8 scFv for further functional analysis of ALCAM/CD166 and intracellular delivery of effector moieties.
Collapse
Affiliation(s)
- Tiziana Piazza
- Istituto Nazionale per la Ricerca sul Cancro, Largo R. Benzi 10, 16132 Genova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Abstract
Nanoscale drug delivery systems including liposomes, polymers, and other nanoparticles provide potential solutions for improved cancer therapeutics. Of these drug delivery systems, liposome-based agents, particularly liposomal anthracyclines, have had the greatest impact in oncology to date. Current liposomal drugs evolved from a number of design strategies for improved biodistribution over free drugs. Reticuloendothelial system-targeted formulations significantly reduce systemic exposure to high peak levels of free drug, but do not facilitate targeting to tumors. Passive or physiologic targeting of drugs to tumors is achievable using long-circulating liposomes, including pure lipid systems as well as surface-modified formulations designed to resist recognition and uptake by reticuloendothelial system cells. The latter, represented by pegylated or STEALTH liposomes, circulate for days as stable constructs and slowly extravasate in neoangiogenic vessels in tumors, providing a degree of passive targeting to tumor tissue. Future liposome therapeutics are building on these validated designs as well as on pharmacologic insights into their mechanisms of delivery. For example, camptothecin analogues, anti-angiogenesis agents, and antisense oligonucleotides each represent rational candidates for delivery in highly stabilized and long-circulating liposomes. For such agents, pegylated liposome delivery offers improved chemical stability of encapsulated drug, enhanced accumulation in tumors, and prolonged drug exposure. True molecular targeting can be achieved using liposomes linked to ligands such as monoclonal antibody fragments directed against cancer-associated antigens. Immunoliposomes combine antibody-mediated tumor recognition with liposomal delivery and, when designed for target cell internalization, provide intracellular drug release. Recent advances in immunoliposome design include rapid selection of phage antibody-derived scFv for targeting, and methods for conjugation of ligands to existing US Food and Drug Administration-approved liposomal drugs such as pegylated liposomal doxorubicin (Doxil/Caelxy [PLD]). An immunoliposome consisting of novel anti-HER2 scFv F5 conjugated to PLD, currently in development, selectively binds to and internalizes in HER2-overexpressing tumor cells. The modular organization of immunoliposome technology enables a combinatorial approach in which a repertoire of monoclonal antibody fragments can be used in conjunction with a series of liposomal drugs to yield a new generation of molecularly targeted agents.
Collapse
Affiliation(s)
- John W Park
- UCSF Comprehensive Cancer Center, San Francisco, CA 94115-1710, USA
| | | | | |
Collapse
|
116
|
Affiliation(s)
- Andrew R M Bradbury
- Bioscience Division, Los Alamos National Laboratory, TA-43, HRL-1, MS M888, NM 87545, USA.
| | | |
Collapse
|
117
|
Robinson P, Stuber D, Deryckère F, Tedbury P, Lagrange M, Orfanoudakis G. Identification using phage display of peptides promoting targeting and internalization into HPV-transformed cell lines. J Mol Recognit 2004; 18:175-82. [PMID: 15384175 DOI: 10.1002/jmr.723] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
'High-risk' human papilloma viruses (HPVs) cause cervical tumours. In order to treat these tumours therapeutic approaches must be developed that efficiently target the tumour cells. Using phage display, we selected tumour-targeting peptides from a library of constrained nonamer peptides presented multivalently on pVIII of M13. Three different consensus peptide sequences were isolated by biopanning on HPV16-transformed SiHa cells. The corresponding phage-peptides targeted and were internalized in HPV16 transformed SiHa and CaSki cells as well as in HPV18-transformed HeLa cells, but failed to bind a panel of normal or transformed cell lines. Two of the three selected peptides targeted cells only when presented on phage particles in a constrained conformation. However, all three peptides retained their targeting capacity when presented on the reporter protein enhanced green fluorescent protein (EGFP) in a monovalent form. These peptides may be useful for the design of drug or gene delivery vectors for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Philip Robinson
- UMR7100-CNRS Ecole Supérieure de Biotechnologie de Strasbourg, Université Louis Pasteur, boulevard Sébastien Brandt, 67400-Illkirch, France
| | | | | | | | | | | |
Collapse
|
118
|
Xu MY, Xu XH, Chen GZ, Deng XL, Li J, Yu XJ, Chen MZ. Production of a human single-chain variable fragment antibody against esophageal carcinoma. World J Gastroenterol 2004; 10:2619-23. [PMID: 15309706 PMCID: PMC4572180 DOI: 10.3748/wjg.v10.i18.2619] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To construct a phage display library of human single-chain variable fragment (scFv) antibodies associated with esophageal cancer and to preliminarily screen a scFv antibody against esophageal cancer.
METHODS: Total RNA extracted from metastatic lymph nodes of esophageal cancer patients was used to construct a scFv gene library. Rescued by M13K07 helper phage, the scFv phage display library was constructed. esophageal cancer cell line Eca109 and normal human esophageal epithelial cell line (NHEEC) were used for panning and subtractive panning of the scFv phage display library to obtain positive phage clones. Soluble scFv was expressed in E. coli HB2151 which was transfected with the positive phage clone, then purified by affinity chromatography. Relative molecular mass of soluble scFv was estimated by Western blotting, its bioactivity was detected by cell ELISA assay. Sequence of scFv was determined using the method of dideoxynucleotide sequencing.
RESULTS: The size of scFv gene library was approximately 9 × 106 clones. After four rounds of panning with Eca109 and three rounds of subtractive panning with NHEEC cells, 25 positive phage clones were obtained. Soluble scFv was found to have a molecular mass of 31 ku and was able to bind to Eca109 cells, but not to HeLa and NHEEC cells. Variable heavy (VH) gene from one of the positive clones was shown to be derived from the γ chain subgroup IV of immunoglobulin, and variable light (VL) gene from the κ chain subgroup I of immunoglobulin.
CONCLUSION: A human scFv phage display library can be constructed from the metastatic lymph nodes of esophageal cancer patients. A whole human scFv against esophageal cancer shows some bioactivity.
Collapse
Affiliation(s)
- Ming-Yan Xu
- Department of Forensic Medicine, Medical College, Shantou University, Shantou 515041, Guangdong Province, China.
| | | | | | | | | | | | | |
Collapse
|
119
|
Burg M, Ravey EP, Gonzales M, Amburn E, Faix PH, Baird A, Larocca D. Selection of internalizing ligand-display phage using rolling circle amplification for phage recovery. DNA Cell Biol 2004; 23:457-62. [PMID: 15294095 DOI: 10.1089/1044549041474760] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Selection of phage libraries against complex living targets such as whole cells or organs can yield valuable targeting ligands without prior knowledge of the targeted receptor. Our previous studies have shown that noninfective multivalent ligand display phagemids internalize into mammalian cells more efficiently than their monovalent counterparts suggesting that cell-based selection of internalizing ligands might be improved using multivalently displayed peptides, antibodies or cDNAs. However, alternative methods of phage recovery are needed to select phage from noninfective libraries. To this end, we reasoned that rolling circle amplification (RCA) of phage DNA could be used to recover noninfective phage. In feasibility studies, we obtained up to 1.5 million-fold enrichment of internalizing EGF-targeted phage using RCA. When RCA was applied to a large random peptide library, eight distinct human prostate carcinoma cell-internalizing peptides were isolated within three selection rounds. These data establish RCA as an alternative to infection for phage recovery that can be used to identify peptides from noninfective phage display libraries or infective libraries under conditions where there is the potential for loss of phage infectivity.
Collapse
Affiliation(s)
- Michael Burg
- Selective Genetics, Inc., San Diego, California 92121, USA
| | | | | | | | | | | | | |
Collapse
|
120
|
Fransson J, Ek S, Ellmark P, Söderlind E, Borrebaeck CAK, Furebring C. Profiling of internalizing tumor-associated antigens on breast and pancreatic cancer cells by reversed genomics. Cancer Lett 2004; 208:235-42. [PMID: 15142683 DOI: 10.1016/j.canlet.2003.11.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Revised: 11/26/2003] [Accepted: 11/28/2003] [Indexed: 11/23/2022]
Abstract
Human antibodies directed towards functionally associated tumor antigens have great potentials as adjuvant treatment in cancer therapy. Here we describe an efficient subtractive approach to select single chain Fv (scFv) antibodies, specifically binding to unknown rapidly internalizing tumor-associated antigens (TAA) on human breast and pancreatic carcinoma cell lines. After re-engineering the scFv into intact IgG molecules, these fully human antibodies displayed individual binding profiles to TAAs on breast, pancreatic, colorectal and prostate carcinomas, while showing no reactivity to lymphomas. The ability of the selected antibodies to undergo receptor-mediated internalization was verified by confocal microscopy, thus proving our strategy to provide a unique set of human antibodies, potentially useful in immunotherapy.
Collapse
Affiliation(s)
- Johan Fransson
- Department of Immunotechnology, Lund University, P.O. Box 7031, SE-220 07 Lund, Sweden
| | | | | | | | | | | |
Collapse
|
121
|
Liu B, Conrad F, Cooperberg MR, Kirpotin DB, Marks JD. Mapping Tumor Epitope Space by Direct Selection of Single-Chain Fv Antibody Libraries on Prostate Cancer Cells. Cancer Res 2004; 64:704-10. [PMID: 14744788 DOI: 10.1158/0008-5472.can-03-2732] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The identification of tumor-specific cell surface antigens is a critical step toward the development of targeted therapeutics for cancer. The epitope space at the tumor cell surface is highly complex, composed of proteins, carbohydrates, and other membrane-associated determinants including post-translational modification products, which are difficult to probe by approaches based on gene expression. This epitope space can be efficiently mapped by complementary monoclonal antibodies. By selecting human antibody gene diversity libraries directly on the surface of prostate cancer cells, we have taken a functional approach to identifying fully human, tumor-specific monoclonal antibodies without prior knowledge of their target antigens. Selection conditions have been optimized to favor tumor-specific antibody binding and internalization. To date, we have discovered >90 monoclonal antibodies that specifically bind and enter prostate cancer cells, with little or no binding to control cells. These antibodies are able to efficiently deliver intracellular payloads when attached to nanoparticles such as liposomes. In addition, a subset of the antibodies displayed intrinsic antiproliferative activity. These tumor-specific internalizing antibodies are likely to be useful for targeted therapeutics either alone or in combination with effector molecules. The antigens they bind constitute a tumor-specific internalizing epitope space that is likely to play a significant role in cancer cell homeostasis. Targeting components of this epitope space may facilitate development of immunotherapeutic and small molecule-based strategies as well as the use of other therapeutic agents that rely upon delivery to the interior of the tumor cell.
Collapse
Affiliation(s)
- Bin Liu
- Department of Anesthesia and Pharmaceutical Chemistry, University of California at San Francisco, San Francisco General Hospital, California, USA.
| | | | | | | | | |
Collapse
|
122
|
Manoutcharian K, Acero G, Munguia ME, Montero JA, Govezensky T, Cao C, Ugen K, Gevorkian G. Amyloid-beta peptide-specific single chain Fv antibodies isolated from an immune phage display library. J Neuroimmunol 2003; 145:12-7. [PMID: 14644026 DOI: 10.1016/j.jneuroim.2003.08.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A single-chain fragment variable (scFv) antibody library displayed on phage was constructed using spleen cells from mice immunized with human amyloid-beta peptide (Abeta42). This first anti-Abeta42 scFv immune antibody library was selected against human Abeta42. A number of positive clones were obtained, and sequences of VH and Vkappa genes were analyzed using ExPASy and BLAST computer tools. This analysis revealed that only two unique clones with identical VH and Vkappa complementarity determining region (CDR) (except HCDR2) and identical germline genes were selected, indicating that oligoclonal immune response was occurring in Abeta42-immunized mice. Abeta42-specific scFv antibodies selected from this first immune anti-Abeta42 phage antibody library may be an important tool for the development of therapeutic molecules for Alzheimer's disease (AD).
Collapse
Affiliation(s)
- K Manoutcharian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, México DF, 04510, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
Antibody or ligand-mediated targeting of liposomal anticancer drugs to antigens expressed selectively or over-expressed on tumor cells is increasingly being recognized as an effective strategy for increasing the therapeutic indices of anticancer drugs. This review summarizes some recent advances in the field of ligand-targeted liposomes (LTLs) for the delivery of anticancer drugs. New approaches used in the design and optimization of LTLs is discussed and the advantages and potential problems associated with their therapeutic applications are described. New technologies are widening the spectrum of ligands available for targeting and are allowing choices to be made regarding affinity, internalization and size. The time is rapidly approaching where we will see translation of anticancer drugs entrapped in LTLs to the clinic.
Collapse
Affiliation(s)
- P Sapra
- Department of Pharmacology, University of Alberta, Edmonton AB, Canada, T6G 2H7
| | | |
Collapse
|
124
|
Woiwode TF, Haggerty JE, Katz R, Gallop MA, Barrett RW, Dower WJ, Cwirla SE. Synthetic Compound Libraries Displayed on the Surface of Encoded Bacteriophage. ACTA ACUST UNITED AC 2003; 10:847-58. [PMID: 14522055 DOI: 10.1016/j.chembiol.2003.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We describe a technology for attaching libraries of synthetic compounds to coat proteins of bacteriophage particles such that the identity of the chemical structure is encoded in the genome of the phage, analogous to peptides displayed on phage surfaces by conventional phage-display techniques. This format allows a library of synthetic compounds to be screened very efficiently as a single pool. Encoded phage serve as extremely robust reporters of the presence of each compound, providing exquisite sensitivity for identification of active compounds engaged in complex biological processes such as receptor-mediated endocytosis and transcytosis. To evaluate this approach, we constructed a library of 980 analogs of folic acid displayed on T7 phage, and demonstrated rapid identification of compounds that bind to folate receptor and direct endocytosis of associated phage particles into cells that express the targeted receptor.
Collapse
Affiliation(s)
- Thomas F Woiwode
- XenoPort, Inc., 3410 Central Expressway, Santa Clara, CA 95051, USA
| | | | | | | | | | | | | |
Collapse
|
125
|
Nielsen UB, Cardone MH, Sinskey AJ, MacBeath G, Sorger PK. Profiling receptor tyrosine kinase activation by using Ab microarrays. Proc Natl Acad Sci U S A 2003; 100:9330-5. [PMID: 12876202 PMCID: PMC170918 DOI: 10.1073/pnas.1633513100] [Citation(s) in RCA: 182] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2002] [Indexed: 11/18/2022] Open
Abstract
Signal transduction in mammalian cells is mediated by complex networks of interacting proteins. Understanding these networks at a circuit level requires devices to measure the amounts and activities of multiple proteins in a rapid and accurate manner. Ab microarrays have previously been applied to the quantification of labeled recombinant proteins and proteins in serum. The development of methods to analyze intracellular signaling molecules on microarrays would make Ab arrays widely useful in systems biology. Here we describe the fabrication of multiplex Ab arrays sensitive to the amounts and modification states of signal transduction proteins in crude cell lysates and the integration of these arrays with 96-well microtiter plate technology to create microarrays in microplates. We apply the Ab arrays to monitoring the activation, uptake, and signaling of ErbB receptor tyrosine kinases in human tumor cell lines. Data obtained from multicolor ratiometric microarrays correlate well with data obtained by using traditional approaches, but the arrays are faster and simpler to use. The integration of microplate and microarray methods for crude cell lysates should make it possible to identify and analyze small molecule inhibitors of signal transduction processes with unprecedented speed and precision. We demonstrate the future potential of this approach by characterizing the action of the epidermal growth factor receptor inhibitor PD153035 on cells by using Ab arrays; direct scale-up to array-based screening in 96- and 384-well plates should allow small molecules to be identified with specific inhibitory profiles against a signaling network.
Collapse
Affiliation(s)
- Ulrik B Nielsen
- Department of Biology 68-371, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | |
Collapse
|
126
|
Rosenkranz AA, Lunin VG, Gulak PV, Sergienko OV, Shumiantseva MA, Voronina OL, Gilyazova DG, John AP, Kofner AA, Mironov AF, Jans DA, Sobolev AS. Recombinant modular transporters for cell-specific nuclear delivery of locally acting drugs enhance photosensitizer activity. FASEB J 2003; 17:1121-3. [PMID: 12692081 DOI: 10.1096/fj.02-0888fje] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The search for new pharmaceuticals that are specific for diseased rather than normal cells in the case of cancer and viral disease has raised interest in locally acting drugs that act over short distances within the cell and for which different cell compartments have distinct sensitivities. Thus, photosensitizers (PSs) used in anti-cancer therapy should ideally be transported to the most sensitive subcellular compartments in order for their action to be most pronounced. Here we describe the design, production, and characterization of the effects of bacterially expressed modular recombinant transporters for PSs comprising 1) alpha-melanocyte-stimulating hormone as an internalizable, cell-specific ligand; 2) an optimized nuclear localization sequence of the SV40 large T-antigen; 3) an Escherichia coli hemoglobin-like protein as a carrier; and 4) an endosomolytic amphipathic polypeptide, the translocation domain of diphtheria toxin. These modular transporters delivered PSs into the nuclei, the most vulnerable sites for the action of PSs, of murine melanoma cells, but not non-MSH receptor-overexpressing cells, to result in cytotoxic effects several orders of magnitude greater than those of nonmodified PSs. The modular fusion proteins described here for the first time, capable of cell-specific targeting to particular subcellular compartments to increase drug efficacy, represent new pharmaceuticals with general application.
Collapse
Affiliation(s)
- Andrey A Rosenkranz
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334, Moscow, Russia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Gao C, Mao S, Ronca F, Zhuang S, Quaranta V, Wirsching P, Janda KD. De novo identification of tumor-specific internalizing human antibody-receptor pairs by phage-display methods. J Immunol Methods 2003; 274:185-97. [PMID: 12609544 DOI: 10.1016/s0022-1759(02)00522-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Three tumor-specific, internalizing human single-chain Fvs (scFvs) were obtained by direct selection against tumor cells from a large, nonimmune scFv-phage library pre-subtracted with various normal human cells. After scFv selection and characterization for cell binding and internalization, the scFvs were also employed in immunoprecipitations to identify putative receptors. In the case of a prostate tumor-cell specific scFv PR5, the receptor that mediated endocytosis was shown to be the transferrin receptor. For two pancreatic adenocarcinoma specific scFvs SW1 and PAN10, the alpha(3)beta(1) integrin was identified. The scFv SW1 was studied in further detail and found to induce functional effects as a ligand-mimetic by mediating cell adhesion and migration. The results demonstrated the feasibility of utilizing enhanced phage-display methods as a rapid and general approach for not only direct isolation of human internalizing scFvs, but also for identifying tumor cell-surface receptors from various classes. The use of scFv constructs that target tumor cells and undergo internalization could have significant impact on the future of cancer and gene therapy.
Collapse
Affiliation(s)
- Changshou Gao
- Department of Chemistry, The Scripps Research Institute and the Skaggs Institute for Chemical Biology, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
128
|
Shadidi M, Sioud M. Identification of novel carrier peptides for the specific delivery of therapeutics into cancer cells. FASEB J 2003; 17:256-8. [PMID: 12490548 DOI: 10.1096/fj.02-0280fje] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cancer therapy is currently limited by the difficulty of achieving efficient delivery into target cells. To investigate whether therapeutics can be delivered specifically to cancer cells, we have explored the possibility of selecting small peptides that bind specifically, or preferentially, to breast cancer cell lines. By using random peptide phage libraries and an experimental approach that allows the selection of internalized peptides, cell-specific binding peptides have been identified. The peptides define a major core motif (LTVXPWY) that was not found in negative phages. Phage displaying LTVSPWY peptide sequence exhibited a specific binding to breast cancer cells. None of the selected peptides bound to human primary cells from different tissue origin (e.g., epithelial, endothelial, hematopoetic). The potential of the selected peptides to mediate cellular internalization in the context of phages and recombinant GFP-peptide fusions was demonstrated. By linking the LTVSPWY peptide to an antisense phosphorothioate oligonucleotide against the ErbB2 receptor, specific delivery to cancer cells was achieved. In contrast to free antisense, the peptide-antisense conjugates inhibited ErbB2 gene expression. Thus, efficient delivery of antisense oligonucleotides can be achieved by coupling them to cancer cell-specific peptides, identified by a method that did not require any knowledge about their corresponding receptors.
Collapse
Affiliation(s)
- Mohsen Shadidi
- Institute for Cancer Research, Department of Immunology, Molecular Medicine Group, The Norwegian Radium Hospital, Montebello, N-0310 Oslo, Norway
| | | |
Collapse
|
129
|
Radosević K, Voerman JSA, Hemmes A, Muskens F, Speleman L, de Weers M, Rosmalen JGM, Knegt P, van Ewijk W. Colony lift assay using cell-coated filters: a fast and efficient method to screen phage libraries for cell-binding clones. J Immunol Methods 2003; 272:219-33. [PMID: 12505726 DOI: 10.1016/s0022-1759(02)00208-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
For efficient screening of phage antibody libraries obtained by selection on whole cells, we have developed a modified colony lift assay using cell-coated filters. Both cells growing in suspension as well as adherent cells can be coated onto nitrocellulose filters and used to detect bacterial colonies responsible for the production of cell-binding (specific) single chain variable fragment (scFv) antibodies. We demonstrate, using a selected library developed in our laboratory (named "AB" library) as a model system, that the frequency of specific clones as detected by colony lift assay using cell-coated filter is comparable to the frequency of positive clones as detected by the "classical" method (i.e. random picking and flow cytometric analysis). However, the colony lift assay enables detection and isolation of a higher number of specific clones as compared to the random pick. This is due to screening of a much higher number of clones simultaneously (it is possible to screen at least 1000 clones plated on one 9-cm agar dish). Using this method, clones occurring at a low frequency (such as present in early selection rounds) can be detected and isolated efficiently. We clearly demonstrate the usefulness of the colony lift assay with cell-coated filter by applying it to screen the head-and-neck carcinoma (HN) library (a selected library generated in our laboratory). Using the assay, but not the random picking, we were able to isolate specific clones from 2nd to 3rd selection rounds of the HN library.
Collapse
Affiliation(s)
- Katarina Radosević
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Affiliation(s)
- Peter J Hudson
- CRC for Diagnostics at CSIRO Health Sciences and Nutrition, Parkville, Victoria, Australia.
| | | |
Collapse
|
131
|
Gao C, Mao S, Ditzel HJ, Farnaes L, Wirsching P, Lerner RA, Janda KD. A cell-penetrating peptide from a novel pVII-pIX phage-displayed random peptide library. Bioorg Med Chem 2002; 10:4057-65. [PMID: 12413859 DOI: 10.1016/s0968-0896(02)00340-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A novel random peptide library was constructed using a phage-display format on the coat proteins pVII and pIX of filamentous bacteriophage. Panning against B-lymphocyte WI-L2 cells yielded one unique peptide-phage, denoted CHL8, that specifically bound to and penetrated the cells. Studies of each peptide derived from CHL8, denoted pep7 and pep9, established that only pep7 mediated the observed activity and only as a homodimer. Peptide libraries displayed on pVII-pIX should serve as a novel source of bioactive ligands for a variety of applications.
Collapse
Affiliation(s)
- Changshou Gao
- Department of Chemistry, The Scripps Research Institute and the Skaggs Institute for Chemical Biology, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
132
|
Nielsen UB, Kirpotin DB, Pickering EM, Hong K, Park JW, Refaat Shalaby M, Shao Y, Benz CC, Marks JD. Therapeutic efficacy of anti-ErbB2 immunoliposomes targeted by a phage antibody selected for cellular endocytosis. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1591:109-118. [PMID: 12183061 DOI: 10.1016/s0167-4889(02)00256-2] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Many targeted cancer therapies require endocytosis of the targeting molecule and delivery of the therapeutic agent to the interior of the tumor cell. To generate single chain Fv (scFv) antibodies capable of triggering receptor-mediated endocytosis, we previously developed a method to directly select phage antibodies for internalization by recovering infectious phage from the cytoplasm of the target cell. Using this methodology, we reported the selection of a panel of scFv that were internalized into breast cancer cells from a nonimmune phage library. For this work, an immunotherapeutic was generated from one of these scFv (F5), which bound to ErbB2 (HER2/neu). The F5 scFv was reengineered with a C-terminal cysteine, expressed at high levels in Escherichia coli, and coupled to sterically stabilized liposomes. F5 anti-ErbB2 immunoliposomes were immunoreactive as determined by surface plasmon resonance (SPR) and were avidly internalized by ErbB2-expressing tumor cell lines in proportion to the levels of ErbB2 expression. F5-scFv targeted liposomes containing doxorubicin had antitumor activity and produced significant reduction in tumor size in xenografted mice compared to nontargeted liposomes containing doxorubicin. This strategy should be applicable to generate immunotherapeutics for other malignancies by selecting phage antibodies for internalization into other tumor types and using the scFv to target liposomes or other nanoparticles.
Collapse
Affiliation(s)
- Ulrik B Nielsen
- Department of Anesthesia and Pharmaceutical Chemistry, University of California-San Francisco, SF General Hospital, 1001 Potrero Avenue, San Francisco, CA 94110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
133
|
O'Connell D, Becerril B, Roy-Burman A, Daws M, Marks JD. Phage versus phagemid libraries for generation of human monoclonal antibodies. J Mol Biol 2002; 321:49-56. [PMID: 12139932 DOI: 10.1016/s0022-2836(02)00561-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Non-immune (naïve) phage antibody libraries have become an important source of antibodies for reagent, diagnostic, and therapeutic use. To date, reported naïve libraries have been constructed in phagemid vectors as fusions to pIII, yielding primarily single copy (monovalent) display of antibody fragments. For this work, we subcloned the single chain Fv (scFv) gene repertoire from a naïve phagemid antibody library into a true phage vector to create a multivalently displayed scFv phage library. Compared to monovalently displayed scFv, multivalent phage display resulted in improved efficiency of display as well as antibody selection. A greater number of antibodies were obtained and at earlier rounds of selection. Such increased efficiency allows the screening for binding antibodies after a single round of selection, greatly facilitating automation. Expression levels of antigen-binding scFv were also higher than from the phagemid library. In contrast, the affinities of scFv from the phage library were lower than from the phagemid library. This could be overcome by utilizing the scFv in a multivalent format, by affinity maturation, or by converting the library to monovalent display after the first round of selection.
Collapse
Affiliation(s)
- David O'Connell
- Department of Anesthesia and Pharmaceutical Chemistry, University of California, San Francisco, Rm 3C-38, San Francisco General Hospital, 1001 Potrero Ave, 94110, USA
| | | | | | | | | |
Collapse
|
134
|
Rasmussen UB, Schreiber V, Schultz H, Mischler F, Schughart K. Tumor cell-targeting by phage-displayed peptides. Cancer Gene Ther 2002; 9:606-12. [PMID: 12082461 DOI: 10.1038/sj.cgt.7700476] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2002] [Indexed: 11/09/2022]
Abstract
We isolated cancer cell-specific phages by subtracting and selecting complex peptide display phage libraries on cultured human cancer cells. The best candidate was selected by performing three rounds of subtraction before each of five selections on the human colorectal WiDr cell line. The phage showed more than 1000-fold higher binding efficiency for WiDr cells when compared to five other human cancer cell lines, including two of colorectal origin, and when compared to wild-type M13 phage. Fifty-fold higher binding efficiency was also seen for a human breast cancer cell line. We show that the WiDr cell binding of the selected phage was efficiently competed by the synthetic peptide HEWSYLAPYPWF, predicted from the phage sequence. This confirms that the specificity of the peptide is independent of the display by the phage coat proteins. The identified peptide may target biomarkers linked to colorectal cancer, and thus be useful for designing gene transfer vectors as well as diagnostic and prognostic tools for this disease.
Collapse
Affiliation(s)
- Ulla B Rasmussen
- Department of Molecular and Cellular Biology, Transgene, SA, 11 rue de Molsheim, F-67082 Strasbourg, France.
| | | | | | | | | |
Collapse
|
135
|
Legendre D, Fastrez J. Construction and exploitation in model experiments of functional selection of a landscape library expressed from a phagemid. Gene 2002; 290:203-15. [PMID: 12062815 DOI: 10.1016/s0378-1119(02)00562-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Phage display has evolved during the past 15 years as a powerful technique to select, from libraries of peptides or proteins, binders for various targets or to evolve new functions in proteins. In recent years, the knowledge acquired in phage display technology was exploited to engineer phages as vehicles for receptor-mediated gene delivery. The first vectors generated provided the proof of the concept that development of gene delivery vehicles based on phages was feasible. Results obtained showed that the level of receptor ligand display was an essential factor that determines the efficiency of transduction and suggested that phagemids might be more appropriate than phages for gene delivery. However, due to the limitations of the existing display systems, vectors constructed up to now allowed only relatively low levels of ligand display. The transduction efficiency of these vectors was relatively poor. Here, we describe the construction and optimization of a new phagemid display system that was designed to allow the functional selection of peptides that promote gene delivery from phagemids in a high display format. Peptides are displayed on every copy of the major coat protein pVIII and are expressed from the phagemid itself. The phagemid is rescued as particles by a modified R408 helper phage, deficient in pVIII production. Besides an expression cassette for pVIII, the phagemid also contains the SV40 origin of replication, the GFP gene and the neomycin resistance marker. As a model we constructed a library of octapeptides and showed that the library is amenable to selection on cos-7 cells. Several selection approaches were investigated and a preliminary analysis of the peptides selected was carried out.
Collapse
Affiliation(s)
- Daniel Legendre
- Laboratoire de Biochimie Physique et des Biopolymères, Institut des Sciences de la Vie, Université Catholique de Louvain, Place L. Pasteur, 1-1b, 1348, Louvain-la-Neuve, Belgium.
| | | |
Collapse
|
136
|
De Lorenzo C, Nigro A, Piccoli R, D'Alessio G. A new RNase-based immunoconjugate selectively cytotoxic for ErbB2-overexpressing cells. FEBS Lett 2002; 516:208-12. [PMID: 11959134 DOI: 10.1016/s0014-5793(02)02527-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We report a new tumor-directed immunoRNase, a chimeric protein made up of an antibody fragment (single-chain Fv fragment) directed to ErbB2, a cell surface receptor, and a non-toxic, human ribonuclease, which upon cell internalization becomes cytotoxic. The immunoRNase is active as a ribonuclease, specifically binds and selectively kills ErbB2-positive cells. ErbB2 is one of the most specific tumor-associated antigens identified so far, overexpressed on tumor cells of different origin. Its choice as target antigen and that of a non-toxic, human RNase as the killer moiety makes this immunoRNase a new, potentially attractive anticancer agent.
Collapse
Affiliation(s)
- Claudia De Lorenzo
- Dipartimento di Chimica Biologica, Università di Napoli Federico II, Via Mezzocannone 16, 80134, Naples, Italy
| | | | | | | |
Collapse
|
137
|
Molenaar TJM, Michon I, de Haas SAM, van Berkel TJC, Kuiper J, Biessen EAL. Uptake and processing of modified bacteriophage M13 in mice: implications for phage display. Virology 2002; 293:182-91. [PMID: 11853411 DOI: 10.1006/viro.2001.1254] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Internalization and degradation of filamentous bacteriophage M13 by a specific target cell may have major consequences for the recovery of phage in in vivo biopanning of phage libraries. Therefore, we investigated the pharmacokinetics and processing of native and receptor-targeted phage in mice. (35)S-radiolabeled M13 was chemically modified by conjugation of either galactose (lacM13) or succinic acid groups (sucM13) to the coat protein of the phage to stimulate uptake by galactose recognizing hepatic receptors and scavenger receptors, respectively. Receptor-mediated endocytosis of modified phage reduced the plasma half-life of native M13 (t(1/2) = 4.5 h) to 18 min for lactosylated and 1.5 min for succinylated bacterophage. Internalization of sucM13 was complete within 30 min after injection and resulted in up to 5000-fold reduction of bioactive phage within 90 min. In conclusion, these data provide information on the in vivo behavior of wild-type and receptor-targeted M13, which has important implications for future in vivo phage display experiments and for the potential use of M13 as a viral gene delivery vehicle.
Collapse
Affiliation(s)
- Tom J M Molenaar
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Leiden University, Sylvius Laboratory, 2300 RA Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
138
|
Arafat WO, Gómez-Navarro J, Buchsbaum DJ, Xiang J, Wang M, Casado E, Barker SD, Mahasreshti PJ, Haisma HJ, Barnes MN, Siegal GP, Alvarez RD, Hemminki A, Nettelbeck DM, Curiel DT. Effective single chain antibody (scFv) concentrations in vivo via adenoviral vector mediated expression of secretory scFv. Gene Ther 2002; 9:256-62. [PMID: 11896464 DOI: 10.1038/sj.gt.3301639] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2001] [Accepted: 11/20/2001] [Indexed: 11/09/2022]
Abstract
Single chain antibodies (scFv) represent powerful interventional agents for the achievement of targeted therapeutics. The practical utility of these agents have been limited, however, by difficulties related to production of recombinant scFv and the achievement of effective and sustained levels of scFv in situ. To circumvent these limitations, we have developed an approach to express scFv in vivo. An anti-erbB2 scFv was engineered for secretion by eukaryotic cells. The secreted scFv could bind to its target and specifically suppress cell growth of erbB2-positive cells in vitro. Adenoviral vectors expressing the cDNA for the secretory scFv likewise could induce target cells to produce an anti-tumor anti-erbB2 scFv. In vivo gene transfer via the anti-erbB2 scFv encoding adenovirus also showed anti-tumor effects. Thus, by virtue of engineering a secreted version of the anti-tumor anti-erbB-2 scFv, and in vivo expression via adenoviral vector, effective concentrations of scFv were achieved. In vivo gene transfer clearly represents a powerful means to realize effective scFv-based approaches. This method will likely have applicability for a range of disorders amenable to targeted therapeutic approaches.
Collapse
Affiliation(s)
- W O Arafat
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Hartley O. The use of phage display in the study of receptors and their ligands. J Recept Signal Transduct Res 2002; 22:373-92. [PMID: 12503628 DOI: 10.1081/rrs-120014608] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Phage display technology presents a rapid means by which proteins and peptides that bind specifically to predefined molecular targets can be isolated from extremely complex combinatorial libraries. There are several important ways by which phage display can provide impetus to receptor-based research. Firstly, phage display can be applied, alongside transcriptome and proteome expression profiling techniques, to the identification and characterisation of receptors whose expression is specific to either a cell lineage, a tissue or a disease state. Secondly, specific monoclonal antibodies that enable researchers to identify, localize and quantify receptors can be produced very rapidly (weeks). Thirdly, it should be possible to apply phage display to the matching of orphan ligands and receptors. Finally, phage display can be used to identify proteins and peptides that modulate receptor activity. As well as being useful in the study of receptor function, biologically active proteins and peptides could also be used therapeutically, or as leads for drug design. Hence phage display is ready to play a central role in the study of receptors in the post-genome era. This review outlines the ways in which phage display has been applied to the study of receptor-ligand systems, and discusses how new developments in the technology may be of even greater utility to the field in the next decade.
Collapse
Affiliation(s)
- Oliver Hartley
- Département de Biochimie Medicale, Centre Médicale Universitaire, 1 rue Michel Servet, CH-1211 Genève 4, Switzerland.
| |
Collapse
|
140
|
Abstract
A quarter of a century after their advent, monoclonal antibodies have become the most rapidly expanding class of pharmaceuticals for treating a wide variety of human diseases, including cancer. Although antibodies have yet to achieve the ultimate goal of curing cancer, many innovative approaches stand poised to improve the efficacy of antibody-based therapies.
Collapse
MESH Headings
- Animals
- Antibodies/immunology
- Antibodies/therapeutic use
- Antibodies, Bispecific/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/therapeutic use
- Antibody-Dependent Cell Cytotoxicity
- Antigens, Neoplasm/immunology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Combined Modality Therapy
- Cytokines/administration & dosage
- Cytokines/therapeutic use
- Drug Delivery Systems
- Humans
- Immunization, Passive
- Immunoconjugates/therapeutic use
- Immunotherapy/methods
- Immunotoxins/therapeutic use
- Liposomes
- Mice
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/immunology
- Neoplasm, Residual
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/radiotherapy
- Neoplasms/therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/therapy
- Prodrugs/administration & dosage
- Protein Engineering
- Radioimmunotherapy
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/therapeutic use
- Rituximab
- Trastuzumab
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- P Carter
- Immunex, Seattle, Washington 98101, USA.
| |
Collapse
|
141
|
Abstract
Phage display, which exploits fundamental tools and principles of immune repertoire diversity, antigen-antibody interactions, and clonal and immunologic selection, is used increasingly to advance experimental and clinical hematology. Phage display is based on the ability of bacteriophage to present engineered proteins on their surface coat. Diverse libraries of proteins such as peptides, antibody fragments, and protein domains corresponding to gene fragments or cDNAs may be displayed. Interactions between phage-displayed proteins and target antigens can be identified rapidly and characterized using high throughput methodologies. Peptide and gene fragment libraries are particularly useful to characterize binding interactions between proteins, such as ligand-receptor interactions. This approach allows rapid generation of human antibodies, often against nonimmunogenic, conserved proteins. Phage antibodies against surface and intracellular antigens are used as reagents for flow cytometry, in vivo imaging, and therapeutic targeting. Phage-derived antibodies also facilitate analyses of the humoral antibody response. Finally, cellular delivery of phage-displayed peptides and gene fragments can be used to modulate functional pathways and molecules in vitro and in vivo. The combinatorial power of phage display enables identification of candidate epitopes without knowledge of the protein interaction, a priori. Overall, these capabilities provide a versatile, high-throughput approach to develop tools and reagents useful for a plethora of experimental hematology applications. This paper focuses on current and future applications of antibody and epitope phage display technology in hematology.
Collapse
Affiliation(s)
- B P Mullaney
- Department of Laboratory Medicine, University of California at San Francisco, San Francisco, Calif., USA.
| | | |
Collapse
|
142
|
Larocca D, Baird A. Receptor-mediated gene transfer by phage-display vectors: applications in functional genomics and gene therapy. Drug Discov Today 2001; 6:793-801. [PMID: 11470588 DOI: 10.1016/s1359-6446(01)01837-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent studies have demonstrated targeted gene-delivery to mammalian cells using modified phage-display vectors. Specificity is determined by the choice of the genetically displayed targeting ligand. Without targeting, phage particles have virtually no tropism for mammalian cells. Thus, novel ligands can be selected from phage libraries by their ability to deliver a reporter gene to targeted cells. Together with advances in cDNA display technologies, these findings offer new opportunities for the use of phage-display technology in functional genomics. In addition, targeted phage particles have potential as alternative gene therapy vectors that can be further improved using directed evolution.
Collapse
Affiliation(s)
- D Larocca
- Selective Genetics, 11035 Roselle Street San Diego, 92121, San Diego, CA, USA
| | | |
Collapse
|
143
|
Abstract
We have isolated several organ- and tumor-homing peptides by using in vivo phage display. This technology involves the screening of peptide libraries in a living animal. The peptides that result from such a selection home to specific organs or tissues because they recognize molecular 'addresses', receptors that are differentially expressed in vascular beds. Targeted delivery of chemotherapeutics, pro-apoptotic peptides and cytokines to tumors using these peptides improved therapeutic efficacy in animal models. Translation of this technology into clinical applications will form the basis for targeting therapeutic and imaging agents in the context of cancer and other diseases.
Collapse
Affiliation(s)
- M Kolonin
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 427, 77030-4095, Houston, TX, USA
| | | | | |
Collapse
|
144
|
Heitner T, Moor A, Garrison JL, Marks C, Hasan T, Marks JD. Selection of cell binding and internalizing epidermal growth factor receptor antibodies from a phage display library. J Immunol Methods 2001; 248:17-30. [PMID: 11223066 DOI: 10.1016/s0022-1759(00)00340-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The first step in developing a targeted cancer therapeutic is generating a ligand that binds to a receptor which is either tumor specific or sufficiently overexpressed in tumors to provide targeting specificity. For this work, we generated human monoclonal antibodies to the EGF receptor (EGFR), an antigen overexpressed on many solid tumors. Single chain Fv (scFv) antibody fragments were directly selected by panning a phage display library on tumor cells (A431) overexpressing EGFR or Chinese hamster ovary cells (CHO/EGFR cells) transfected with the EGFR gene and recovering endocytosed phage from within the cell. Three unique scFvs were isolated, two from selections on A431 cells and two from selections on CHO/EGFR cells. All three scFv bound native receptor as expressed on a panel of tumor cells and did not bind EGFR negative cells. Phage antibodies and multivalent immunoliposomes constructed from scFv were endocytosed by EGFR expressing cells as shown by confocal microscopy. Native scFv primarily stained the cell surface, with less staining intracellularly. The results demonstrate how phage antibodies binding native cell surface receptors can be directly selected on overexpressing cell lines or transfected cells. Use of a transfected cell line allows selection of antibodies to native receptors without the need for protein expression and purification, significantly speeding the generation of targeting antibodies to genomic sequences. Depending upon the format used, the antibodies can be used to deliver molecules to the cell surface or intracellularly.
Collapse
Affiliation(s)
- T Heitner
- Department of Anesthesia, University of California, San Francisco, Room 3C-38, San Francisco General Hospital, 1001 Potrero Avenue, San Francisco, CA 94110, USA.
| | | | | | | | | | | |
Collapse
|
145
|
Neve RM, Nielsen UB, Kirpotin DB, Poul MA, Marks JD, Benz CC. Biological effects of anti-ErbB2 single chain antibodies selected for internalizing function. Biochem Biophys Res Commun 2001; 280:274-9. [PMID: 11162510 DOI: 10.1006/bbrc.2000.4104] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two internalizing monovalent single chain antibody fragments (scFv), C6.5 and F5, that recognize distinct ErbB2 extracellular domain (ECD) epitopes, and their bivalent forms dbC6.5 and F5(scFv')(2), were compared to the growth-inhibiting anti-ErbB2 antibody Herceptin/trastuzumab, in either its bivalent (Her) or monovalent (4D5Fab') form, for their abilities to induce biological responses in the ErbB2-overexpressing breast cancer cells, SkBr-3. Assays compared internalization by receptor-mediated endocytosis, effects on cell cycling and culture growth, and interference with intracellular MAPK and PI3K signaling pathways. We found no correlation between ErbB2 epitope affinity or valency on degree of antibody-induced endocytosis, since all the scFv were able to internalize better than Her. Unlike Her, neither the monovalent or bivalent forms of the internalizing scFv had any sustained effect on cell growth. Basal levels of MAPK and PI3K signaling in SkBr-3 cells were not inhibited by up to 8 h scFv treatment, while decreased MAPK and PI3K signals were noted within 8 h of Her treatment. In summary, antibody-induced ErbB2-mediated endocytosis is not a surrogate marker for resultant biological response, as it shows no correlation with cell cycle, culture proliferation, or intracellular kinase signal induction by internalizing antibodies. Thus, the enhanced endocytotic property of scFv like C6.6 and F5 in conjunction with their absence of any growth or signaling impact on ErbB2-overexpressing cells favors their choice as ErbB2 targeting moieties for intracellular delivery of novel cancer therapeutics.
Collapse
Affiliation(s)
- R M Neve
- Department of Medicine, University of California at San Francisco, San Francisco, California 94143, USA
| | | | | | | | | | | |
Collapse
|
146
|
Internalizing antibodies and targeted cancer therapy: direct selection from phage display libraries. PHARMACEUTICAL SCIENCE & TECHNOLOGY TODAY 2000; 3:282-291. [PMID: 10916148 DOI: 10.1016/s1461-5347(00)00280-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antibody internalization is required for the success of many targeted therapeutics, such as immunotoxins, immunoliposomes, antibody-drug conjugates and for the targeted delivery of genes or viral DNA into cells. Recently, it has become possible to directly select antibody fragments from phage display libraries for internalization into mammalian cells. Here we review the therapeutic applications of internalized antibodies and describe how phage display enables the isolation of internalizing antibodies to novel or known targets.
Collapse
|