51
|
Schuster M, Umana P, Ferrara C, Brünker P, Gerdes C, Waxenecker G, Wiederkum S, Schwager C, Loibner H, Himmler G, Mudde GC. Improved effector functions of a therapeutic monoclonal Lewis Y-specific antibody by glycoform engineering. Cancer Res 2005; 65:7934-41. [PMID: 16140965 DOI: 10.1158/0008-5472.can-04-4212] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of the present study was to produce glycosylation variants of the therapeutic Lewis Y-specific humanized IgG1 antibody IGN311 to enhance cell-killing effector function. This was achieved via genetic engineering of the glycosylation machinery of the antibody-producing host. Antibody genes were transiently cotransfected with acetyl-glycosaminyltransferase-III genes into human embryonic kidney-EBV nuclear antigen cells. A control wild-type antibody, IGN311wt, was expressed in the same host using identical expression vectors, but without cotransfection of genes for acetyl-glycosaminyltransferase-III expression. Both expression products were purified to homogeneity and characterized. The glyco-engineered expression product (IGN312-Glyco-I) showed a remarkably homogenous N-linked glycosylation pattern consisting of one major hybrid-type, non-fucosylated and agalactosylated form carrying a bisecting GlcNAc-group. Wild-type expression product (IGN311wt) on the other hand was glycosylated by a multitude of different core-fucosylated complex-type structures of variable degrees of galactosylation. Target affinity of the glyco-engineered antibody as well as heavy and light chain assembly were not affected by acetyl-glycosaminyltransferase-III expression. In vitro experiments showed a approximately 10-fold increase of antibody-dependent cellular cytotoxicity of the glyco-engineered antibody using different Lewis Y-positive target cancer cell lines (SK-BR-3, SK-BR-5, OVCAR-3, and Kato-III). Complement-mediated cytotoxicity of IGN312-Glyco-I was 0.4-fold reduced using SK-BR-5 as target cell line. The reduction of complement activation could be prevented and even converted into a slight increase of activity by using a different molecular-biological approach directing the glycosylation towards increased levels of complex N-linked oligosaccharides of bisected, non-fucosylated type, as a result of cotransfection of mannosidase II together with acetyl-glycosaminyltransferase-III.
Collapse
|
52
|
Wang Z, Mårtensson L, Nilsson R, Bendahl PO, Lindgren L, Ohlsson T, Sjögren HO, Strand SE, Tennvall J. Blood Pharmacokinetics of Various Monoclonal Antibodies Labeled with a New Trifunctional Chelating Reagent for Simultaneous Conjugation with 1,4,7,10-Tetraazacyclododecane-N,N′,N″,N‴-Tetraacetic Acid and Biotin before Radiolabeling. Clin Cancer Res 2005; 11:7171s-7177s. [PMID: 16203818 DOI: 10.1158/1078-0432.ccr-1004-0001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Knowledge of the blood pharmacokinetics of monoclonal antibodies is crucial in deciding the optimal time for starting the administration of a "clearing agent" or using a "clearing device." The primary purpose was to investigate whether the pharmacokinetics of various antibodies labeled with the same chelator and (111)In differed significantly after i.v. injection in immunocompetent rats. A new trifunctional chelator called "1033" containing a biotin and a radiometal chelation moiety is introduced, making it possible to use only one conjugation procedure for the antibody. EXPERIMENTAL DESIGN Sixty-five non-tumor-bearing rats were included and divided into four groups (I-IV). The blood pharmacokinetics was investigated for rituximab, BR96, and trastuzumab labeled with 1033 and (111)In (I-III). The whole-body activity and activity uptake in muscle, liver, and kidney, which might explain differences in the early pharmacokinetics in blood, were also measured. hMN14 labeled with another chelator [1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA)], but with the same radionuclide ((111)In-biotin-DOTA-hMN14), was studied (IV). The blood pharmacokinetics from another 15 tumor-bearing rats was compared with those of non-tumor-bearing rats (III) by injection of (111)In-1033-BR96. RESULTS No statistical difference was detected between the groups regarding the blood pharmacokinetics of rituximab, BR96, or trastuzumab. The pharmacokinetics and biodistribution of (111)In-biotin-DOTA-hMN14 exhibited a clear difference compared with others. There were no significant differences in the blood pharmacokinetics of (111)In-1033-BR96 between tumor-bearing rats and non-tumor-bearing rats. CONCLUSIONS Different antibodies labeled with the trifunctional chelator 1033 and (111)In did not exhibit different blood pharmacokinetics, which means that the pharmacokinetics could be predicted irrespective of the IgG1 antibody chosen. A small tumor burden did not change the pharmacokinetics of the radioimmunoconjugates.
Collapse
Affiliation(s)
- Zhongmin Wang
- Department of Oncology, Lund University Hospital, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Abstract
Strategies for restoring beta-cell function in diabetic patients would be greatly aided by the ability to target genes, proteins, or small molecules specifically to these cells. Furthermore, the ability to direct imaging agents specifically to beta-cells would facilitate diagnosis and monitoring of disease progression. To isolate ligands that can home to beta-cells in vivo, we have panned a random phage-displayed 20-mer peptide library on freshly isolated rat islets. We have isolated two 20-mer peptides that bind to islets ex vivo. One of these peptides preferentially homes to the islets of Langerhans in a normal rat with clear differentiation between the endocrine and exocrine cells of the pancreas. Furthermore, this peptide does not target beta-cells in a type 2 diabetes animal model, suggesting that the peptide can discriminate between glucose-stimulated insulin secretion-functional and -dysfunctional beta-cells.
Collapse
Affiliation(s)
- Kausar N Samli
- Center for Biomedical Inventions, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9185, USA
| | | | | | | | | |
Collapse
|
54
|
De J, Chang YC, Samli KN, Schisler JC, Newgard CB, Johnston SA, Brown KC. Isolation of a mycoplasma-specific binding peptide from an unbiased phage-displayed peptide library. MOLECULAR BIOSYSTEMS 2005; 1:149-57. [PMID: 16880978 DOI: 10.1039/b504572j] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An important goal in medicine is the development of methods for cell-specific targeting of therapeutic molecules to pathogens or pathogen-infected cells. However, little progress has been made in cell-specific targeting of bacterially infected cells. Using a phage display approach, we have isolated a 20-mer peptide that binds to Mycoplasma arginini infected pancreatic beta-cells in tissue culture. This peptide binds to M. arginini infected beta-cells 200 times better than a control phage and is specific for the infected cells. Furthermore, transferring the M. arginini contamination to another cell line renders the newly infected cell line susceptible to peptide binding. Immunolocalization experiments suggest that the peptide is binding to M. arginini adhered to the cell surface. The free synthetic peptide retains its binding in the absence of the phage vehicle and tetramerization of the peptide increases its affinity for the infected cells. Efforts have been made to use this peptide to eliminate Mycoplasma from infected cell lines using ferromagnetic beads coated with the selected peptide. A ten-fold reduction of infection was accomplished with one fractionation via this approach. Our results suggest that this peptide, isolated from an unbiased selection, may be of utility for the detection and reduction of Mycoplasma infection in cultured cells. Furthermore, a general implication of our findings is that phage display methods may be useful for identifying peptides that target a broad array of other biological pathogens in a specific fashion.
Collapse
Affiliation(s)
- Jitakshi De
- Center for Translational Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9185, USA
| | | | | | | | | | | | | |
Collapse
|
55
|
Riddle DS, Sanz L, Chong H, Thompson J, Vile RG. Tumor Cell Surface Display of Immunoglobulin Heavy Chain Fc by Gene Transfer as a Means to Mimic Antibody Therapy. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.16.ft-88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
56
|
Kukowska-Latallo JF, Candido KA, Cao Z, Nigavekar SS, Majoros IJ, Thomas TP, Balogh LP, Khan MK, Baker JR. Nanoparticle Targeting of Anticancer Drug Improves Therapeutic Response in Animal Model of Human Epithelial Cancer. Cancer Res 2005; 65:5317-24. [PMID: 15958579 DOI: 10.1158/0008-5472.can-04-3921] [Citation(s) in RCA: 563] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prior studies suggested that nanoparticle drug delivery might improve the therapeutic response to anticancer drugs and allow the simultaneous monitoring of drug uptake by tumors. We employed modified PAMAM dendritic polymers <5 nm in diameter as carriers. Acetylated dendrimers were conjugated to folic acid as a targeting agent and then coupled to either methotrexate or tritium and either fluorescein or 6-carboxytetramethylrhodamine. These conjugates were injected i.v. into immunodeficient mice bearing human KB tumors that overexpress the folic acid receptor. In contrast to nontargeted polymer, folate-conjugated nanoparticles concentrated in the tumor and liver tissue over 4 days after administration. The tumor tissue localization of the folate-targeted polymer could be attenuated by prior i.v. injection of free folic acid. Confocal microscopy confirmed the internalization of the drug conjugates into the tumor cells. Targeting methotrexate increased its antitumor activity and markedly decreased its toxicity, allowing therapeutic responses not possible with a free drug.
Collapse
|
57
|
Reich O, Liegl B, Tamussino K, Regauer S. p185HER2 overexpression and HER2 oncogene amplification in recurrent vulvar Paget's disease. Mod Pathol 2005; 18:354-7. [PMID: 15272283 DOI: 10.1038/modpathol.3800243] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We evaluated p186Her2 overexpression and HER2 oncogene amplification in recurrent vulvar Paget's disease. We identified six patients with recurrent vulvar Paget's disease in our archives. The number of recurrences ranged from 1 to 11 over a time period of 1-168 months. Recurrences were evaluated immunohistochemically for p185Her2 overexpression with the HercepTest and for HER2 oncogene amplification with fluorescence in situ hybridization. p185Her2 overexpression was scored as 3 in two patients, as 2 in two patients, and as 1 in two patients. All patients with scores 2 and 3 showed HER2 oncogene amplification. Overexpression of p185Her2 and HER2 oncogene amplification appears to be common in recurrent vulvar Paget's disease.
Collapse
Affiliation(s)
- Olaf Reich
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria.
| | | | | | | |
Collapse
|
58
|
Vulfovich M, Saba N. Molecular biological design of novel antineoplastic therapies. Expert Opin Investig Drugs 2005; 13:577-607. [PMID: 15174947 DOI: 10.1517/13543784.13.6.577] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Novel therapies represent a new strategy for the development of anticancer agents. New targets derived from the knowledge of the molecular structure and genetic defects has been useful in developing anticancer drugs that prolong or stabilise the progression of tumours with minimal systemic toxicities. In this review, the mechanism of action and the most significant trials regarding monoclonal antibodies, tyrosine kinase inhibitors, angiogenesis and cyclooxygenase inhibitor-based therapies, farnesyl transferase inhibitors and proteasome inhibitors are discussed. The potential biological end points and toxicities are also described. In conclusion, novel therapies present a promising class of anticancer agents, acting through different mechanisms and offering a new perspective in the treatment of cancer.
Collapse
Affiliation(s)
- Michel Vulfovich
- Emory University School of Medicine and Winship Cancer Institute, Crawford Long Hospital, 550 Peachtree Street, Glenn Building, Atlanta, GA, USA
| | | |
Collapse
|
59
|
Xu C, Jung M, Burkhardt M, Stephan C, Schnorr D, Loening S, Jung K, Dietel M, Kristiansen G. Increased CD59 protein expression predicts a PSA relapse in patients after radical prostatectomy. Prostate 2005; 62:224-32. [PMID: 15389793 DOI: 10.1002/pros.20134] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Human protectin (CD59) is a regulator of complement activation that inhibits complement-mediated cell lysis, and thus might confer immune resistance to tumor cells. CD59 expression has been described in a variety of human malignancies, including breast cancer. Since a comprehensive investigation of CD59 expression in prostate cancer has not been conducted yet, we aimed to determine the significance of CD59 expression in prostate cancer. METHODS Eighty-six primary adenocarcinomas of the prostate were immunostained using a monoclonal CD59 antibody (clone MEM-43) and a standard detection system. The immunoreactivity of the tumor was evaluated as low versus high for statistical analysis. Additionally, CD59 mRNA levels were determined by real-time PCR in matched (tumor/normal) microdissected tissues from 26 cases. RESULTS Cytoplasmic CD59 immunoreactivity was found in epithelia of prostate cancer, prostatic intraepithelial neoplasia, benign hyperplasia, atrophic, and normal glands. High rates of CD59 expression were noted in 36% of prostate cancer cases and were significantly associated with tumor pT stage (P = 0.043), Gleason grade (P = 0.013) and earlier biochemical (PSA) relapse in Kaplan-Meier analysis (P = 0.0013). On RNA level, we found an upregulation in 19.2% (five cases), although the general rate of CD59 transcript was significantly lower in tumor tissue (P = 0.03). CONCLUSION CD59 protein is strongly expressed in 36% of adenocarcinomas of the prostate and and is associated with disease progression and adverse patient prognosis.
Collapse
Affiliation(s)
- Chuanliang Xu
- Department of Urology, Charité University Hospital, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Sznol M, Davis T. Antibodies and Recombinant Cytokines. THE CANCER HANDBOOK 2005. [DOI: 10.1002/0470025077.chap87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
61
|
Abstract
Colorectal cancer is one of the leading causes of cancer-related mortality. After a series of clinical trials, the adjuvant 5-FU-based chemotherapy has established a definitive role in the management of stage III colon cancer. While the precise role for chemotherapy in stage II disease remains under investigation, less toxic treatment modalities such as active specific immunotherapy (ASI) have emerged as potentially attractive alternatives. In a randomized trial it was demonstrated that ASI, using autologous tumor cells and BCG, had a significant clinical benefit in the adjuvant treatment of stage II colon cancer. In addition, it was found that the delayed-type cutaneous hypersensitivity reactions (DTH) against autologous cells correlated significantly with survival, confirming its suitability for immunomonitoring. Nowadays, 5-FU-based chemotherapy is a standard treatment for stage III colon cancer and because there is preclinical evidence for synergism between chemotherapy and ASI, we performed a phase II study to investigate whether the combined treatment with 5-FU and ASI is tolerable and feasible. We demonstrated that 5-FU treatment hardly affected the DTH response against autologous tumor cells and that the combination treatment was well tolerated. A randomized phase III trial is now warranted to show that the combination of ASI and chemotherapy is superior to chemotherapy alone.
Collapse
Affiliation(s)
- A J M van den Eertwegh
- Department of Medical Oncology, Vrije Universiteit Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
62
|
Abstract
The convergence of genomic technologies and the development of drugs designed against specific molecular targets provides many opportunities for using bioinformatics to bridge the gap between biological knowledge and clinical therapy. Identifying genes that have properties similar to known targets is conceptually straightforward. Additionally, genes can be linked to cancer via recurrent genomic or genetic abnormalities. Finally, by integrating large and disparate datasets, gene-level distinctions can be made between the different biological states that the data represents. These bioinformatics approaches and their associated methodologies, which can be applied across a range of technologies, facilitate the rapid identification of new target leads for further experimental validation.
Collapse
Affiliation(s)
- Brian Desany
- Department of Bioinformatics, Genentech, 1 DNA Way, M.S. 93, South San Francisco, CA 94080, USA
| | | |
Collapse
|
63
|
Zhou X, Chang YC, Oyama T, McGuire MJ, Brown KC. Cell-Specific Delivery of a Chemotherapeutic to Lung Cancer Cells. J Am Chem Soc 2004; 126:15656-7. [PMID: 15571383 DOI: 10.1021/ja0446496] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report that lung cancer-targeting peptides isolated from a peptide library can be used to deliver an active chemotherapeutic in a cell-specific fashion. The peptides were removed from the context of the phage and placed on a pegylated tetrameric scaffold. The tetrameric peptides were shown to block uptake of their cognate phage. The tetrameric peptides were coupled to doxorubicin, and their cytotoxicity against a panel of different cell lines was tested. Our data demonstrate that these targeting peptides can deliver an active anticancer agent in a cell-specific fashion, resulting in an increase of the therapeutic index of the targeted drug compared to systemic delivery. The efficacy of the peptide conjugate correlates to the affinity of the targeting peptide for a particular cell line. As such, we have demonstrated that cell-specific targeted drugs can be synthesized, even when the cell surface target is unknown.
Collapse
Affiliation(s)
- Xin Zhou
- Center for Biomedical Inventions and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9185, USA
| | | | | | | | | |
Collapse
|
64
|
McGuire MJ, Sykes KF, Samli KN, Timares L, Barry MA, Stemke-Hale K, Tagliaferri F, Logan M, Jansa K, Takashima A, Brown KC, Johnston SA. A Library-Selected, Langerhans Cell-Targeting Peptide Enhances an Immune Response. DNA Cell Biol 2004; 23:742-52. [PMID: 15585132 DOI: 10.1089/dna.2004.23.742] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The ability to deliver antigens and immunomodulators specifically to Langerhans cells (LCs) in the skin could impact vaccine development. However, cell-specific targeting of therapeutic molecules remains a challenge in biomedicine. Using phage display technologies, we have developed a protocol that identifies peptides that mediate uptake into target cell types. Employing this approach, we have isolated a 20-mer peptide that mediates specific uptake by immunopotent LCs. The peptide is functional outside the context of the phage and is able to deliver a nanoparticle to LCs in vitro. Although selected on cells in vitro, the peptide is able to direct antigens and genes to LCs in vivo. Liposomes bearing the LC targeting peptide are able to deliver a transcriptionally active gene to LCs in a mouse model. Furthermore, we demonstrate that a low-dose injection into mice of phage bearing the LC-targeting peptide yields faster and higher immune responses against phage-associated antigens than control-phage injections.
Collapse
Affiliation(s)
- Michael J McGuire
- Center for Biomedical Inventions, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Osta WA, Chen Y, Mikhitarian K, Mitas M, Salem M, Hannun YA, Cole DJ, Gillanders WE. EpCAM is overexpressed in breast cancer and is a potential target for breast cancer gene therapy. Cancer Res 2004; 64:5818-24. [PMID: 15313925 DOI: 10.1158/0008-5472.can-04-0754] [Citation(s) in RCA: 394] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
EpCAM (epithelial cell adhesion molecule) is a cell surface molecule that is known to be highly expressed in colon and other epithelial carcinomas. EpCAM is involved in cell-to-cell adhesion and has been the target of antibody therapy in several clinical trials. To assess the value of EpCAM as a novel target for breast cancer gene therapy, we performed real-time reverse transcription-PCR to quantify the level of EpCAM mRNA expression in normal breast tissue and primary and metastatic breast cancers. We found that EpCAM is overexpressed 100- to 1000-fold in primary and metastatic breast cancer. Silencing EpCAM gene expression with EpCAM short interfering RNA (siRNA) resulted in a 35-80% decrease in the rate of cell proliferation in four different breast cancer cell lines. EpCAM siRNA treatment decreased cell migration by 91.8% and cell invasion by 96.4% in the breast cancer cell line MDA-MB-231 in vitro. EpCAM siRNA treatment was also associated with an increase in the detergent-insoluble protein fraction of E-cadherin, alpha-catenin, and beta-catenin, consistent with the known biology of EpCAM as a regulator of cell adhesion. Our hypothesis is that modulation of EpCAM expression can affect cell migration, invasion, and proliferation by enhancing E-cadherin-mediated cell-to-cell adhesion. These data provide compelling evidence that EpCAM is a potential novel target for breast cancer gene therapy and offer insights into the mechanisms associated with EpCAM gene silencing.
Collapse
Affiliation(s)
- Walid A Osta
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Dermime S, Gilham DE, Shaw DM, Davidson EJ, Meziane EK, Armstrong A, Hawkins RE, Stern PL. Vaccine and antibody-directed T cell tumour immunotherapy. Biochim Biophys Acta Rev Cancer 2004; 1704:11-35. [PMID: 15238242 DOI: 10.1016/j.bbcan.2004.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2003] [Revised: 01/22/2004] [Accepted: 03/31/2004] [Indexed: 10/26/2022]
Abstract
Clearer evidence for immune surveillance in malignancy and the identification of many new tumour-associated antigens (TAAs) have driven novel vaccine and antibody-targeted responses for therapy in cancer. The exploitation of active immunisation may be particularly favourable for TAA where tolerance is incomplete but passive immunisation may offer an additional strategy where the immune repertoire is affected by either tolerance or immune suppression. This review will consider how to utilise both active and passive types of therapy delivered by T cells in the context of the failure of tumour-specific immunity by presenting cancer patients. This article will outline the progress, problems and prospects of several different vaccine and antibody-targeted approaches for immunotherapy of cancer where proof of principle pre-clinical studies have been or will soon be translated into the clinic. Two examples of vaccination-based therapies where both T cell- and antibody-mediated anti-tumour responses are likely to be relevant and two examples of oncofoetal antigen-specific antibody-directed T cell therapies are described in the following sections: (1) therapeutic vaccination against human papillomavirus (HPV) antigens in cervical neoplasia; (2) B cell lymphoma vaccines including against immunoglobulin idiotype; (3) oncofoetal antigens as tumour targets for redirecting T cells with antibody strategies.
Collapse
Affiliation(s)
- Said Dermime
- Immunology, Cancer Research UK Groups, Paterson Institute for Cancer Research and University of Manchester, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Fonsatti E, Maio M. Highlights on endoglin (CD105): from basic findings towards clinical applications in human cancer. J Transl Med 2004; 2:18. [PMID: 15193152 PMCID: PMC441416 DOI: 10.1186/1479-5876-2-18] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Accepted: 06/11/2004] [Indexed: 11/19/2022] Open
Abstract
Antibody targeting of tumor-associated vasculature is a promising therapeutic approach in human cancer; however, a specific cell membrane marker for endothelial cells of tumor vasculature has not been discovered yet. Endoglin (CD105) is a cell-surface glycoprotein most recently identified as an optimal indicator of proliferation of human endothelial cells. The finding that CD105 is over-expressed on vascular endothelium in angiogenetic tissues has prompted several pre-clinical studies designed to get a deeper understanding on the role of CD105 in angiogenesis, and to evaluate the most appropriate clinical setting(s) to utilize CD105 as a therapeutic target. In this review, the foreseeable clinical applications of CD105 in human cancer are discussed.
Collapse
Affiliation(s)
- Ester Fonsatti
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, I.R.C.C.S., 33081 Aviano, Italy
| | - Michele Maio
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, I.R.C.C.S., 33081 Aviano, Italy
- Division of Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, 53100 Siena, Italy
| |
Collapse
|
68
|
Sier CFM, Gelderman KA, Prins FA, Gorter A. Beta-glucan enhanced killing of renal cell carcinoma micrometastases by monoclonal antibody G250 directed complement activation. Int J Cancer 2004; 109:900-8. [PMID: 15027124 DOI: 10.1002/ijc.20029] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metastases from renal cell carcinomas (RCC) are resistant to radiation and chemotherapy but are relatively immunogenic. We have investigated the possibility to eliminate human RCC micrometastases using MAb G250. G250 penetrates human micrometastases completely in a spheroid model and induces complement deposition rapidly on the outmost cell layers. However, complement dependent cytotoxicity (CDC) was barely detected using either (51)chromium release assays or confocal microscopy, due to relatively low expression of the G250 antigen and the effect of membrane bound complement regulatory proteins. Addition of blocking anti-CD59 MAbs enhanced formation of C5b-9 and consequently complement mediated lysis (13%). Complement assisted cellular cytotoxicity (CACC) was not detectable, although the iC3b ligand and CR3 receptor were present on respectively target and effector cells. Addition of soluble beta-glucan induced the killing of MAb and iC3b opsonized spheroids by effector cells (6-21%). Despite a lower affinity for G250 antigen, a bispecific anti-G250*anti-CD55 MAb enhanced cell killing in spheroids comparable to the parental G250 MAb. Our results suggest that complement-activating G250 in combination with anti-mCRP MAbs is able to kill human RCC cells in micrometastasis in vitro. For CACC the presence of CR3-priming beta-glucan seems to be obligatory. In vivo, bi-MAb may be more effective as therapeutic agent due to its increased C5a generating properties.
Collapse
Affiliation(s)
- Cornelis F M Sier
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | |
Collapse
|
69
|
Usener D, Schadendorf D, Koch J, Dübel S, Eichmüller S. cTAGE: a cutaneous T cell lymphoma associated antigen family with tumor-specific splicing. J Invest Dermatol 2003; 121:198-206. [PMID: 12839582 DOI: 10.1046/j.1523-1747.2003.12318.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
cTAGE-1 is a cutaneous-T-cell-lymphoma-specific tumor antigen recently identified by serologic identification of antigens by recombinant expression cloning. This study was aimed at identifying and characterizing related genes. Rapid amplification of cDNA ends and DNA screening led to five new members of the cTAGE gene family belonging to four different genes, two of which were differentially spliced (cTAGE-1/2 and cTAGE-5). Expression analysis using reverse transcription polymerase chain reaction revealed that cTAGE-1, cTAGE-1B, and cTAGE-5A expression was restricted to testis and tumor tissues, whereas the other cTAGE members were found in two to eight other normal tissues (of 27 tissues tested). Tumor-specific protein expression of cTAGE-5 was confirmed by Western blotting. Sero-reactivity against cTAGE-1, cTAGE-4, cTAGE-5A, and cTAGE-5B was found only in tumor patients (cutaneous T cell lymphoma and melanoma). The immunogenic epitope of cTAGE-1 was determined by using epitope mapping and sera of two cutaneous T cell lymphoma patients. Moreover, cTAGE-1, cTAGE-4, cTAGE-5A, and cTAGE-5B could be detected in most types of tumor tissues and cell lines at variable frequencies, including those of cutaneous T cell lymphoma, melanoma, head and neck squamous cell carcinoma, breast carcinoma, and colon carcinoma. We conclude that cTAGE-1 and cTAGE-5 are new cancer germline antigens and that tumor-specific splicing of cTAGE genes may lead to further candidate proteins for specific immunotherapy of cutaneous T cell lymphoma and other malignancies.
Collapse
MESH Headings
- Amino Acid Sequence
- Antibodies/blood
- Antigens, Differentiation, T-Lymphocyte
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Blotting, Northern
- Blotting, Southern
- Blotting, Western
- Chromosomes, Human, Pair 18
- Epitope Mapping
- Gene Expression Regulation, Neoplastic
- Humans
- Lymphoma, T-Cell, Cutaneous/genetics
- Membrane Glycoproteins
- Molecular Sequence Data
- Neoplasm Proteins
- Nucleic Acid Amplification Techniques
- RNA Splicing
- Reverse Transcriptase Polymerase Chain Reaction
- Skin Neoplasms/genetics
Collapse
Affiliation(s)
- Dirk Usener
- Skin Cancer Unit, German Cancer Research Center, Heidelberg
| | | | | | | | | |
Collapse
|
70
|
Chosy EJ, Nakamura M, Melnik K, Comella K, Lasky LC, Zborowski M, Chalmers JJ. Characterization of antibody binding to three cancer-related antigens using flow cytometry and cell tracking velocimetry. Biotechnol Bioeng 2003; 82:340-51. [PMID: 12599261 DOI: 10.1002/bit.10581] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Proper antibody labeling is a fundamental step in the positive selection/isolation of rare cancer cells using immunomagnetic cell separation technology. Using either a two-step or single-step labeling protocol, we examined a combination of six different antibodies specific for three different antigens (epithelial specific antigen, epithelial membrane antigen, and HER-2/Neu) on two different breast cancer cell lines (HCC1954 and MCF-7). When a two-step labeling protocol was used (i.e., anti-surface marker-fluoroscein-isothiocyanate [FITC] [primary Ab], anti-FITC magnetic colloid [secondary Ab]) saturation of the primary antibody was determined using fluorescence intensity measurements from flow cytometry (FCM). The saturation of the secondary antibody (or saturation of a single-step labeling) was determined using magnetophoretic mobility measurements from cell tracking velocimetry (CTV). When the maximum magnetophoretic mobility was the primary objective, our results demonstrate that the quantities necessary for antibody saturation with respect to fluorescence intensity were generally higher than those recommended by the manufacturer. The results demonstrate that magnetophoretic mobility varies depending on the types of cell lines, primary antibodies, and concentration of secondary magnetic colloid-conjugated antibody. It is concluded that saturation studies are a vital preparatory step in any separation method involving antibody labeling, especially those that require the specificity of rare cell detection.
Collapse
Affiliation(s)
- E Julia Chosy
- Department of Chemical Engineering, Ohio State University, 125 Koffolt Laboratories, 140 West 19th Avenue, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Lah J, Marianovsky I, Glaser G, Engelberg-Kulka H, Kinne J, Wyns L, Loris R. Recognition of the intrinsically flexible addiction antidote MazE by a dromedary single domain antibody fragment. Structure, thermodynamics of binding, stability, and influence on interactions with DNA. J Biol Chem 2003; 278:14101-11. [PMID: 12533537 DOI: 10.1074/jbc.m209855200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Escherichia coli mazEF operon defines a chromosomal addiction module that programs cell death under various stress conditions. It encodes the toxic and long-lived MazF and the labile antidote MazE. The denaturation of MazE is a two-state reversible dimer-monomer transition. At lower concentrations the denatured state is significantly populated. This leads to a new aspect of the regulation of MazE concentration, which may decide about the life and death of the cell. Interactions of MazE with a dromedary antibody domain, cAbMaz1 (previously used as a crystallization aid), as well as with promoter DNA were studied using microcalorimetric and spectroscopic techniques. Unique features of cAbMaz1 enable a specific enthalpy-driven recognition of MazE and, thus, a significant stabilization of its dimeric native conformation. The MazE dimer and the MazE dimer-cAbMaz1 complex show very similar binding characteristics with promoter DNA, i.e. three binding sites with apparent affinities in micromolar range and highly exothermic binding accompanied by large negative entropy contributions. A working model for the MazE-DNA assembly is proposed on the basis of the structural and binding data. Both binding and stability studies lead to a picture of MazE solution structure that is significantly more unfolded than the structure observed in a crystal of the MazE-cAbMaz1 complex.
Collapse
Affiliation(s)
- Jurij Lah
- Department of Ultrastructure, Vrije Universiteit Brussel, Paardenstraat 65, B-1640 St. Genesius Rode, Belgium.
| | | | | | | | | | | | | |
Collapse
|
72
|
Dalerba P, Maccalli C, Casati C, Castelli C, Parmiani G. Immunology and immunotherapy of colorectal cancer. Crit Rev Oncol Hematol 2003; 46:33-57. [PMID: 12672517 DOI: 10.1016/s1040-8428(02)00159-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This review critically discusses data on immunology of colorectal cancer, starting from pathology and molecular biology, and then considering the molecular characterisation of colon cancer antigens and the clinical trials of immunotherapy. A careful evaluation of histopathological studies on intra-epithelial infiltration by T cells in primary tumours, together with the analysis of HLA expression by colorectal cancer cells, suggest that anti-tumour T cell immune responses may take place in vivo in those patients, influencing prognosis and shaping the tumour immunological profile. Moreover, the molecular characterisation of tumour antigens expressed by colorectal carcinomas, together with improved understanding of mechanisms of the immune response and more sensitive methods for the in vivo detection of T cell responses, are now allowing researchers to design new and more effective vaccination protocols, with encouraging preliminary results. By drawing together the experimental evidence from different research fields, this review provides support for the concept that colorectal carcinoma is immunogenic and may reasonably be considered as a target for immunotherapy, and attempts to address critical issues and envisage future developments in this challenging research field.
Collapse
Affiliation(s)
- Piero Dalerba
- Unit of Immunotherapy of Human Tumours, Istituto Nazionale per lo Studio e la Cura dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | | | | | | | | |
Collapse
|
73
|
Abstract
PURPOSE/OBJECTIVES To provide an overview of the biology of lung cancer with respect to genetic carcinogenesis and specific mutations and to discuss new therapies being developed to target lung cancer's biologic processes. DATA SOURCES Published articles, abstracts, book chapters, lectures, and personal experiences with experimental agents. DATA SYNTHESIS Lung cancer is the number one cause of cancer deaths for men and women in the United States, with minimal changes in the five-year survival rate during the past decade. New understanding of the biologic process of lung cancer is providing potential new therapies that many hope will lead to increased survival for patients with lung cancer. CONCLUSIONS Exciting new therapies for lung cancer are being developed that target specific biologic processes of lung cancer. IMPLICATIONS FOR NURSING When nurses are familiar with the rationale behind biologic therapies, they can understand the drugs, assess toxicities, and help patients make educated decisions about therapeutic alternatives.
Collapse
|
74
|
Donin N, Jurianz K, Ziporen L, Schultz S, Kirschfink M, Fishelson Z. Complement resistance of human carcinoma cells depends on membrane regulatory proteins, protein kinases and sialic acid. Clin Exp Immunol 2003; 131:254-63. [PMID: 12562385 PMCID: PMC1808622 DOI: 10.1046/j.1365-2249.2003.02066.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Nucleated cells employ several strategies to evade killing by homologous complement. We studied complement resistance in the human carcinoma cell lines (CA) T47D (mammary), SKOV3 (ovarian), and PC-3 (prostate) with emphasis on the following mechanisms of defense: 1. Expression and shedding of the membrane complement regulatory proteins (mCRP) CD46, CD55 and CD59; 2. Resistance based on protein phosphorylation; 3. Cell surface expression of sialic acid residues; 4. Desensitization to complement upon exposure to sublytic complement doses. Anti-mCRP antibody blocking experiments demonstrated that CD59 is the main mCRP protecting these CA from complement. Soluble CD59 was also found in supernates of PC-3> SKOV3 > T47D cells. Second, inhibitors of PKC, PKA and MEK sensitized the CA to lysis, thus implicating these protein kinases in CA complement resistance. Third, removal of sialic acid residues with neuraminidase also sensitized CA to lysis. Finally, exposure of CA to sublytic doses of complement conferred on them enhanced resistance to lytic complement doses in a PKC-dependent process. Combined treatment of CA with anti-CD59 antibodies, PD98059 (a MEK inhibitor) and neuraminidase produced a large enhancement in CA sensitivity to complement. Our results show that CD59 and sialic acid residues present on the cell surface, and intracellular processes involving protein phosphorylation act additively to secure CA resistance to complement-mediated lysis. Therefore, the effectiveness of antibody- and complement-based cancer immunotherapy will markedly improve by suppression of the various complement resistance mechanisms.
Collapse
Affiliation(s)
- N Donin
- Department of Cell Biology and Histology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
75
|
Imakiire T, Kuroki M, Shibaguchi H, Abe H, Yamauchi Y, Ueno A, Hirose Y, Yamada H, Yamashita Y, Shirakusa T, Ishida I, Kuroki M. Generation, immunologic characterization and antitumor effects of human monoclonal antibodies for carcinoembryonic antigen. Int J Cancer 2003; 108:564-70. [PMID: 14696121 DOI: 10.1002/ijc.11608] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We generated fully human mAbs (HmAbs) to carcinoembryonic antigen (CEA) using the KM mouse, which carries a human chromosome 14 fragment containing the entire Ig H chain loci and human kappa L chain segments in the mouse genome. Forty-six hybridoma clones producing HmAbs to CEA were thus obtained by fusing the P3-U1 mouse myeloma cells with splenocytes of the KM mice immunized with CEA. Among them, 22 clones produced HmAbs that reacted with CEA but not with 3 other CEA-related cell adhesion molecule (CEACAM) family members, CEACAM1, CEACAM6 and CEACAM8. In 12 HmAbs examined, 8 were IgG4, 2 were IgG3, 1 was IgG2, and the other was IgG1. The affinity constants for CEA of these HmAbs were comparable to those of the previously prepared mouse anti-CEA mAbs (MmAbs). BIAcore analyses revealed that 1 and 2 of the 22 HmAbs react with 2 epitopes defined by MmAbs on the domain N and the domain A1 or B1 of CEA, respectively. In the presence of human complement in vitro, 2 HmAbs tested showed substantial cytotoxicity, namely, 50-65%, against CEA-expressing tumor cells. With human lymphokine-activated killer cells in vitro, 3 HmAbs tested exhibited 40-65% Ab-dependent cell-mediated cytotoxicity against the tumor cells. Moreover, one of the HmAbs induced a significant inhibition of tumor growth when administered to mice xenografted with the CEA-expressing cells. Considering their lack of immunogenicity to humans, these CEA-specific HmAbs may be useful for immunotherapeutic approaches as well as for immunodiagnosis.
Collapse
Affiliation(s)
- Takayuki Imakiire
- Department of Biochemistry, Fukuoka University School of Medicine, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Meyer DL, Senter PD. Chapter 23. Recent advances in antibody drug conjugates for cancer therapy. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2003. [DOI: 10.1016/s0065-7743(03)38024-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
77
|
Palapattu GS, Reiter RE. Monoclonal antibody therapy for genitourinary oncology: promise for the future. J Urol 2002; 168:2615-23. [PMID: 12441996 DOI: 10.1016/s0022-5347(05)64230-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Metastatic or recurrent cancer continues to be the bane of the urological oncologist. Despite recent improvements in therapeutic strategies and outcomes for clinically localized disease overall survival in patients with the majority of metastatic and recurrent genitourinary malignancies remains relatively unchanged. Modern advances in the field of immunotherapy hold the promise of providing the clinical urologist/oncologist with new tools to fight urological cancer. In this review we discuss the various mAb based strategies currently under investigation for urological oncology as well as the lessons learned from similar approaches in other fields. MATERIALS AND METHODS We reviewed the literature on mAb based immunotherapy with a particular emphasis on target antigens, mAb design and potential applications in the field of urology. RESULTS Early trials with mAb therapy for solid tumor oncology met with limited success due to difficulty with mAb design and production, the development of host immunological responses against murine monoclonal antibodies (that is human anti-mouse antibodies), suboptimal target antigen selection, and poor monoclonal antibody pharmacokinetics and tumor tissue penetration. Recent advances in the fields of immunology and oncology have sought to circumvent these obstacles. Today several preliminary studies have shown the effectiveness and usefulness of mAb based strategies for urological oncology. CONCLUSIONS The field of mAb based immunotherapy continues to evolve. New discoveries in this burgeoning area of cancer therapy show promise for the future.
Collapse
|
78
|
|
79
|
Abstract
In the past two decades, the worldwide steadily rising incidence of melanoma, its dismal prognosis when locally advanced or metastatic, and the absence of clinically effective therapeutic options have prompted studies that generated extensive preclinical knowledge on the biology of melanoma cells and their interaction with the host's immune system. As a consequence, among solid tumors, melanoma represents a "model malignancy" to design and apply in the clinic new bioimmunotherapeutic approaches, that are eventually translated to solid tumors of different histotypes. Despite its waxing and waning appeal as a therapeutic strategy, antibody treatment still represents a promising clinical approach to melanoma. Europe is no exception in the clinical interest for antibodies as therapeutic tools in melanoma patients; European researchers have also focused on preclinical issues that may improve current antibody-based therapeutic approaches on the one hand, while providing novel clinical strategies on the other hand.
Collapse
Affiliation(s)
- Maresa Altomonte
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | | |
Collapse
|
80
|
De Pascalis R, Iwahashi M, Tamura M, Padlan EA, Gonzales NR, Santos AD, Giuliano M, Schuck P, Schlom J, Kashmiri SVS. Grafting of "abbreviated" complementarity-determining regions containing specificity-determining residues essential for ligand contact to engineer a less immunogenic humanized monoclonal antibody. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:3076-84. [PMID: 12218124 DOI: 10.4049/jimmunol.169.6.3076] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Murine mAb COL-1 reacts with carcinoembryonic Ag (CEA), expressed on a wide range of human carcinomas. In preclinical studies in animals and clinical trials in patients, murine COL-1 showed excellent tumor localization. To circumvent the problem of immunogenicity of the murine Ab in patients, a humanized COL-1 (HuCOL-1) was generated by grafting the complementarity-determining regions (CDRs) of COL-1 onto the frameworks of the variable light and variable heavy regions of human mAbs. To minimize anti-V region responses, a variant of HuCOL-1 was generated by grafting onto the human frameworks only the "abbreviated" CDRs, the stretches of CDR residues that contain the specificity-determining residues that are essential for the surface complementarity of the Ab and its ligand. In competition RIAs, the recombinant variant completely inhibited the binding of radiolabeled murine and humanized COL-1 to CEA. The HuCOL-1 and its variant showed no difference in their binding ability to the CEA expressed on the surface of a CEA-transduced tumor cell line. Compared with HuCOL-1, the HuCOL-1 variant showed lower reactivity to patients' sera carrying anti-V region Abs to COL-1. The final variant of the HuCOL-1, which retains its Ag-binding reactivity and shows significantly lower serum reactivity than that of the parental Ab, can serve as a prototype for the development of a potentially useful clinical reagent.
Collapse
Affiliation(s)
- Roberto De Pascalis
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Witz IP. Presence and functions of immune components in the tumor microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2002; 495:317-24. [PMID: 11774586 DOI: 10.1007/978-1-4615-0685-0_44] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- I P Witz
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
82
|
Savage P, Cowburn P, Clayton A, Man S, Lawson T, Ogg G, Lemoine N, McMichael A, Epenetos A. Anti-viral cytotoxic T cells inhibit the growth of cancer cells with antibody targeted HLA class I/peptide complexes in SCID mice. Int J Cancer 2002; 98:561-6. [PMID: 11920616 DOI: 10.1002/ijc.10219] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A number of experimental antibody mediated cancer therapies aim to redirect cytotoxic T cells (CTLs) of non-tumour specificity to cancer cells. It has been previously demonstrated that cancer cells targeted with recombinant HLA-class I/viral peptide complexes via antibody delivery systems can be killed by virus specific CTLs. This novel therapeutic system has been developed with a simple pre-clinical model using the recombinant anti-CD20 B9E9 scFvSA fusion protein to target HLA-A2/peptide complexes to CD20 +ve Daudi lymphoma cells. In vitro data confirmed that, although binding of the B9E9 scFvSA fusion protein alone to Daudi cells had no effect on their growth, effective CTL mediated killing of Daudi cells could be achieved by targeting with B9E9 sfvScSA and recombinant HLA-A2/MI complexes at dilutions as low as 100 pg/ml. In contrast the free HLA-A2/MI complexes only significantly inhibited CTL activity at concentrations in excess of 100 ng/ml. The in vivo tumour protection assays in SCID mice demonstrated that only 1 of the 4 mice that received anti-HLA-A2/M1 CTLs and Daudi cells targeted with the B9E9 scFvSA fusion protein and HLA-A2/M1 complexes developed a tumour. In contrast in the control mice that received CTL and native Daudi cells all 4 developed tumours, as did all 4 that received targeted Daudi cells but no CTLs. Similar results were obtained in a parallel experiment using Daudi cells targeted with B9E9 scFvSA and HLA-A2/BMLF1 complexes and a CTL line to HLA-A2/BMLF1. The demonstration of in vivo activity for targeted HLA class I/peptide complexes combined with anti-viral T cells, supports the further clinical development of the system where it may be combined with autologous CTLs produced by vaccination or ex vivo expansion.
Collapse
|
83
|
Xiong D, Xu Y, Liu H, Peng H, Shao X, Lai Z, Fan D, Yang M, Han J, Xie Y, Yang C, Zhu Z. Efficient inhibition of human B-cell lymphoma xenografts with an anti-CD20 x anti-CD3 bispecific diabody. Cancer Lett 2002; 177:29-39. [PMID: 11809528 DOI: 10.1016/s0304-3835(01)00758-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bispecific antibodies have been exploited both as cancer immunodiagnostics and as cancer therapeutics, and have shown promise in several clinical trials in cancer imaging and therapy. A number of bispecific antibodies against B-cell markers have been shown to be effective in vitro in mediating tumor cell lysis and in vivo in inhibiting tumor growth in animal models. We have constructed a bispecific diabody from the variable genes encoding two hybridoma-derived monoclonal antibodies directed against human CD20 on B cells and CD3 on T cells. The anti-CD20 x anti-CD3 diabody was expressed in a single Escherichia coli host and purified by a one-step affinity chromatography. The bispecific diabody bound as efficiently to both CD20- and CD3-positive cells as the respective parental antibodies, and was capable of cross-linking CD20-positive tumor cells and human T lymphocytes as shown by cellular rosetting. The diabody effectively lysed human B-lymphoma cells in the presence of T-enriched human peripheral blood lymphocytes (PBL). Further, when combined with human PBL and interleukin-2, the diabody significantly prolonged the survival of nude mice inoculated with human B-lymphoma cells. Taken together, our results suggest that an anti-CD20 x anti-CD3 diabody may have significant clinical application in the treatment of human CD20-positive B-cell malignancies.
Collapse
Affiliation(s)
- Dongsheng Xiong
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Abstract
Despite improvements in care of patients with breast cancer, up to half develop refractory or resistant disease. There is therefore a need for new, modified anticancer therapies with greater effectiveness, tolerability to patients, and tumour specificity. Trastuzumab (Herceptin) is the first clinically available oncogene-targeted therapeutic agent for treatment of solid tumours. Clinical trials in patients positive for HER2 (human epidermal-growth-factor receptor 2) show that trastuzumab is effective and well tolerated; as a single-agent second-line or third-line treatment, the drug produced durable tumour responses. First-line trastuzumab in combination with chemotherapy, particularly paclitaxel, significantly improved time to disease progression, duration of response, and time to treatment failure. Combination therapy resulted in a 25% improvement in overall survival compared with chemotherapy alone. Patients with HER2 gene amplification, high overexpression of HER2 (3+ on immunohisto-chemistry), or both features, obtained the greatest clinical benefit. Trastuzumab is the first monoclonal antibody with efficacy in breast cancer and the first gene-product-targeted therapy to produce a significant survival advantage in this disease. Trastuzumab is likely to find its ultimate role in the adjuvant setting. Its development provides a model for the integration of other gene-targeted therapies into breast-cancer management to improve survival and quality of life.
Collapse
|
85
|
Vaquero C, Sack M, Schuster F, Finnern R, Drossard J, Schumann D, Reimann A, Fischer R. A carcinoembryonic antigen-specific diabody produced in tobacco. FASEB J 2002; 16:408-10. [PMID: 11790722 DOI: 10.1096/fj.01-0363fje] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The feasibility of using tobacco for production of a recombinant antibody (T84.66/GS8 diabody) directed against the carcinoembryonic antigen (CEA) and used for tumor imaging was investigated. Two constructs were generated for targeting the protein either to the apoplast or to the endoplasmic reticulum. Expression of the diabody in tobacco leaves after vacuum-assisted infiltration of engineered Agrobacteria (agro-infiltration) and in regenerated transgenic tobacco plants was analyzed and compared. Results in terms of protein expression and accumulation between both systems showed a good correlation. His6-tagged T84.66 diabody was readily purified from agro-infiltrated tobacco leaves and from transgenic plants by immobilized metal ion affinity chromatography. The purified protein was analyzed by polyacrylamide gel electrophoresis, Western blot, gel filtration, electrospray mass spectrometry, direct and competition ELISA, electrophoretic mobility shift assay, and staining of CEA-positive colon adenocarcinoma cell line LS174T. Our results demonstrate that tobacco is a competent production system for this clinically relevant diabody.
Collapse
Affiliation(s)
- Carmen Vaquero
- Institut für Biologie VII (Molekulare Biotechnologie, RWTH Aachen, 52074 Aachen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Dumoulin M, Conrath K, Van Meirhaeghe A, Meersman F, Heremans K, Frenken LGJ, Muyldermans S, Wyns L, Matagne A. Single-domain antibody fragments with high conformational stability. Protein Sci 2002; 11:500-15. [PMID: 11847273 PMCID: PMC2373476 DOI: 10.1110/ps.34602] [Citation(s) in RCA: 466] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2001] [Revised: 11/13/2001] [Accepted: 11/16/2001] [Indexed: 10/17/2022]
Abstract
A variety of techniques, including high-pressure unfolding monitored by Fourier transform infrared spectroscopy, fluorescence, circular dichroism, and surface plasmon resonance spectroscopy, have been used to investigate the equilibrium folding properties of six single-domain antigen binders derived from camelid heavy-chain antibodies with specificities for lysozymes, beta-lactamases, and a dye (RR6). Various denaturing conditions (guanidinium chloride, urea, temperature, and pressure) provided complementary and independent methods for characterizing the stability and unfolding properties of the antibody fragments. With all binders, complete recovery of the biological activity after renaturation demonstrates that chemical-induced unfolding is fully reversible. Furthermore, denaturation experiments followed by optical spectroscopic methods and affinity measurements indicate that the antibody fragments are unfolded cooperatively in a single transition. Thus, unfolding/refolding equilibrium proceeds via a simple two-state mechanism (N <--> U), where only the native and the denatured states are significantly populated. Thermally-induced denaturation, however, is not completely reversible, and the partial loss of binding capacity might be due, at least in part, to incorrect refolding of the long loops (CDRs), which are responsible for antigen recognition. Most interestingly, all the fragments are rather resistant to heat-induced denaturation (apparent T(m) = 60-80 degrees C), and display high conformational stabilities (DeltaG(H(2)O) = 30-60 kJ mole(-1)). Such high thermodynamic stability has never been reported for any functional conventional antibody fragment, even when engineered antigen binders are considered. Hence, the reduced size, improved solubility, and higher stability of the camelid heavy-chain antibody fragments are of special interest for biotechnological and medical applications.
Collapse
Affiliation(s)
- Mireille Dumoulin
- Laboratoire d'Enzymologie, Centre d'Ingénerie des Protéines, Institut de Chimie B6, Université de Liége, B-4000 Liége, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Cook-Bruns N. Retrospective analysis of the safety of Herceptin immunotherapy in metastatic breast cancer. Oncology 2002; 61 Suppl 2:58-66. [PMID: 11694789 DOI: 10.1159/000055403] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Approximately 25,000 patients have been treated to date with the humanized anti-HER2 monoclonal antibody, Herceptin. This therapy has proved effective and well tolerated in patients with HER2-positive metastatic breast cancer; adverse events were generally infusion-related fever and chills of mild-to-moderate severity. Cardiotoxicity and infusion-related reactions emerged as the two main safety concerns with the use of Herceptin. Retrospective analysis revealed a higher incidence of heart failure when Herceptin was combined with anthracyclines than that expected with anthracyclines alone. Age, anthracycline exposure and cardiac risk factors were found to be predictors of cardiac adverse events. Patients experiencing cardiac dysfunction responded well to standard cardiac medication and the majority improved. Cardiac function should be monitored regularly and Herceptin should be discontinued if significant heart failure develops unless the benefits for an individual patient outweigh the risks. Of 25,000 patients, 74 (0.3%) were reported to have experienced a serious infusion-related reaction. The majority occurred during or shortly after the first infusion and were characterized by respiratory symptoms. Most patients were successfully treated; a total of 33 patients continued Herceptin therapy with no recurrence of infusion reactions. Although the benefit to risk ratio of Herceptin remains favorable, physicians must be vigilant and aggressive in managing cardiotoxicity and infusion-related reactions.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Cardiovascular Agents/therapeutic use
- Chills/chemically induced
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Drug Interactions
- Female
- Fever/chemically induced
- Heart Diseases/chemically induced
- Heart Diseases/drug therapy
- Heart Diseases/epidemiology
- Heart Failure/chemically induced
- Heart Failure/drug therapy
- Humans
- Immunotherapy
- Infusions, Intravenous/adverse effects
- Infusions, Intravenous/mortality
- Neoplasm Metastasis
- Pain/chemically induced
- Palliative Care
- Respiratory Insufficiency/etiology
- Respiratory Insufficiency/mortality
- Retrospective Studies
- Risk Factors
- Safety
- Salvage Therapy
- Trastuzumab
- Treatment Outcome
Collapse
Affiliation(s)
- N Cook-Bruns
- Pharmaceuticals Development Group, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
88
|
|
89
|
Ferreira CG, Huisman C, Giaccone G. Novel approaches to the treatment of non-small cell lung cancer. Crit Rev Oncol Hematol 2002; 41:57-77. [PMID: 11796232 DOI: 10.1016/s1040-8428(01)00197-4] [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: 11/30/2022] Open
Abstract
Prognosis of non-small cell lung carcinomas (NSCLC) remains poor, especially in advanced disease. The introduction of new cytotoxic agents in the past decade did only attain minor improvements in survival. It is rather clear that chemotherapy may have reached a plateau, and that it will be difficult to obtain better results in advanced NSCLC by chemotherapy alone. Novel treatment modalities are urgently needed in advanced NSCLC. Backed-up by advances in the understanding of tumor cell biology, a new generation of anticancer agents specifically directed at targets such as tyrosine kinases, farnesyl transferase, angiogenesis factors, matrixmetalloproteinases and oncogenes has been developed in recent years. In this review, we give a brief summary of the state-of-the-art treatment of NSCLC, highlighting its limitations. Novel systemic approaches are then discussed in detail with focus on their mechanistic rationale, stage of clinical development and possible drawbacks. Finally, perspectives of future applications and impact on the treatment of NSCLC are also discussed.
Collapse
Affiliation(s)
- Carlos G Ferreira
- Department of Medical Oncology, Academic Hospital Vrije Universiteit, Amsterdam, The Netherlands
| | | | | |
Collapse
|
90
|
|
91
|
Eccles SA. Monoclonal antibodies targeting cancer: 'magic bullets' or just the trigger? Breast Cancer Res 2001; 3:86-90. [PMID: 11250751 PMCID: PMC138676 DOI: 10.1186/bcr276] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2000] [Revised: 11/27/2000] [Accepted: 12/01/2000] [Indexed: 11/14/2022] Open
Abstract
The first monoclonal antibodies (mAbs) approved for cancer therapy are now in Phase II and III trials, but the critical mechanism(s) determining efficacy and response in patients are still largely undefined. Both the direct antigen-binding (Fab) and constant (Fc) regions of mAbs can contribute to their biological activity. However, Clynes et al (Nat Med 2000, 6:443) recently suggested that the latter (at least in experimental models) might be the dominant component in vivo, triggering host responses to destroy cancer cells. Those workers showed that in mice lacking 'activation' Fc receptors (Fc(gamma)RI and Fc(gamma)RIII), anti-tumour effects of certain mAbs were significantly reduced. In contrast, mice deficient in the 'inhibitory' receptor Fc(gamma)RIIB responded with tumour growth inhibition and enhanced antibody-dependent cellular cytotoxicity (ADCC). These observations suggest that mAbs might be engineered for preferential binding to Fc(gamma)RIII to maximise therapeutic benefit. However, further work is needed to establish a definitive cause-effect relationship in experimental models that are more clinically relevant, to determine whether human Fc(gamma)R isoforms behave in a similar fashion, and to confirm that therapeutic mAbs and host cells can adequately access solid tumour deposits to mediate effective ADCC in situ. Finally, the 'cost-benefit' ratio of such modified macromolecules will need to be measured against mini-mAb constructs, antisense oligonucleotides, peptidomimetics and emerging drugs capable of inhibiting key tumour cell signalling pathways.
Collapse
Affiliation(s)
- S A Eccles
- Institute of Cancer Research, Belmont, Sutton, Surrey, UK.
| |
Collapse
|
92
|
Wang Y, Yuan H, Wright SC, Wang H, Larrick JW. Synthesis and preliminary cytotoxicity study of a cephalosporin-CC-1065 analogue prodrug. BMC CHEMICAL BIOLOGY 2001; 1:4. [PMID: 11710971 PMCID: PMC59845 DOI: 10.1186/1472-6769-1-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2001] [Accepted: 11/02/2001] [Indexed: 11/10/2022]
Abstract
BACKGROUND: Antibody-directed enzyme prodrug therapy (ADEPT) is a promising new approach to deliver anticancer drugs selectively to tumor cells. In this approach, an enzyme is conjugated to a tumor-specific antibody. The antibody selectively localizes the enzyme to the tumor cell surface. Subsequent administration of a prodrug substrate of the enzyme leads to the enzyme-catalyzed release of the free drug at the tumor site. The free drug will destroy the tumor cells selectively, thus, reducing side effects. RESULTS: A CC-1065 analogue was conjugated to a cephalosporin affording prodrug 2. The prodrug and its corresponding free drug, 1, have IC50 values of 0.9 and 0.09 nM, respectively, against U937 leukemia cells in vitro. CONCLUSIONS: For the first time, a prodrug comprised of a cephalosporin and a CC-1065 analogue has been synthesized. The preliminary in vitro studies show that the prodrug was 10-fold less toxic than the free drug. Prodrug 2 has the potential to be useful in cancer treatment using the ADEPT approach.
Collapse
Affiliation(s)
- Yuqiang Wang
- Panorama Research, Inc., 2462 Wyandotte Stree, Mountain View, California, 94043, USA
| | - Huiling Yuan
- Panorama Research, Inc., 2462 Wyandotte Stree, Mountain View, California, 94043, USA
| | - Susan C Wright
- Panorama Research, Inc., 2462 Wyandotte Stree, Mountain View, California, 94043, USA
| | - Hong Wang
- Panorama Research, Inc., 2462 Wyandotte Stree, Mountain View, California, 94043, USA
| | - James W Larrick
- Panorama Research, Inc., 2462 Wyandotte Stree, Mountain View, California, 94043, USA
| |
Collapse
|