101
|
Cardillo TM, Karacay H, Goldenberg DM, Yeldell D, Chang CH, Modrak DE, Sharkey RM, Gold DV. Improved targeting of pancreatic cancer: experimental studies of a new bispecific antibody, pretargeting enhancement system for immunoscintigraphy. Clin Cancer Res 2004; 10:3552-61. [PMID: 15161715 DOI: 10.1158/1078-0432.ccr-03-0340] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The early detection and diagnosis of pancreatic cancer remains a major clinical challenge in which imaging procedures have a central role. The purpose of this study was to evaluate a pretargeting method with a bispecific PAM4 (bsPAM4; anti-MUC1) antibody for radioimmunoscintigraphy of experimental human pancreatic cancer. EXPERIMENTAL DESIGN A bispecific F(ab')(2) antibody was generated from chimeric PAM4 Fab' and murine 734 (anti-indium-diethylenetriaminepentaacetic acid) Fab' fragments and then used in conjunction with 2 peptide haptens ((111)In-IMP-156 and (99m)Tc-IMP-192). Biodistribution studies and radioimmunoscintigraphic imaging properties of the radiolabeled bsPAM4, and pretargeted, radiolabeled peptides were examined in the CaPan1 human pancreatic tumor grown as s.c. xenografts in athymic nude mice. Tumor uptake and tumor:nontumor ratios were compared with a nontargeting irrelevant anti-CD20, bispecific rituximab, radiolabeled peptides alone, and with directly labeled PAM4. RESULTS Biodistribution results indicated significantly greater tumor uptake of radiolabeled peptides at 3 h after injection when pretargeting was performed with bsPAM4 as compared with the bispecific rituximab [20.2 +/- 5.5 percentage of injected dose per gram of tissue (%ID/g) versus 0.9 +/- 0.1%ID/g, respectively, for (111)In-IMP-156, and 16.8 +/- 4.8%ID/g versus 1.1 +/- 0.2%ID/g, respectively, for (99m)Tc-IMP-192]. Similar results were obtained at the 24-h time point. Tumor:nontumor ratios were >30 for all of the tissues except the kidneys, where a ratio of 7.8 +/- 2.8 was observed. By immunoscintigraphy, tumors could be visualized as early as 30 min after injection of the radiolabeled peptide. CONCLUSIONS These studies demonstrate the feasibility of using the pretargeted, bispecific antibody technology for nuclear imaging of pancreatic cancer. The advantage of pretargeted bsPAM4 antibody as an imaging platform is the high specificity for pancreatic cancer as compared with the physicochemical parameters identified by current imaging technologies.
Collapse
|
102
|
Herbert LM, Grosso JF, Dorsey M, Fu T, Keydar I, Cejas MA, Wreschner DH, Smorodinski N, Lopez DM. A unique mucin immunoenhancing peptide with antitumor properties. Cancer Res 2004; 64:8077-84. [PMID: 15520219 DOI: 10.1158/0008-5472.can-04-0853] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Implantation of DA-3 mammary tumor cells into BALB/c mice results in tumor growth, metastatic lesions, and death. These cells were transfected with genes encoding for either the transmembrane (DA-3/TM) or secreted (DA-3/sec) form of human mucin 1 (MUC1). Although the gene for the secreted form lacks the transmembrane and cytoplasmic domains, the 5' sequences of these mucins are identical; however, the gene for the secreted mucin isoform ends with a sequence encoding for a unique 11 amino acid peptide. The DA-3/TM or DA-3 cells transfected with the neomycin vector only (DA-3/neo) have the same in vivo growth characteristics as the parent cell line. In contrast, DA-3/sec cells fail to grow when implanted in immunocompetent BALB/c animals. DA-3/sec cells implanted in nude mice resulted in tumor development verifying the tumorigenic potential of these cells. Pre-exposure of BALB/c mice to DA-3/sec cells afforded protection against challenge with DA-3/TM or DA-3/neo mammary tumors and the unrelated tumors K7, an osteosarcoma, and RENCA, a renal cell carcinoma. Partial protection against subsequent tumor challenges was also achieved by substituting the 11 amino acid peptide found only in the secreted MUC1 isoform, for the live DA-3/sec cells. Notably, the efficacy of this peptide is not strain restricted because it also retarded the growth of Lewis lung carcinoma cells in C57 BL/6 mice. These findings reveal that a unique peptide present in the secreted MUC1 has immunoenhancing properties and may be a potential agent for use in immunotherapy.
Collapse
Affiliation(s)
- Lynn M Herbert
- Department of Microbiology & Immunology, University of Miami School of Medicine, Miami, Florida 33101, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Grosso JF, Herbert LM, Owen JL, Lopez DM. MUC1/sec-expressing tumors are rejected in vivo by a T cell-dependent mechanism and secrete high levels of CCL2. THE JOURNAL OF IMMUNOLOGY 2004; 173:1721-30. [PMID: 15265901 DOI: 10.4049/jimmunol.173.3.1721] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
MUC1/sec is a secreted form of the glycoprotein mucin 1 (MUC1). To characterize the role that MUC1 and MUC1/sec have in tumor progression, these genes were expressed in DA-3 mammary tumor cells. DA-3 cells and DA-3 cells expressing the transmembrane MUC1 gene (DA-3/TM) grow with similar kinetics in BALB/c mice. Surprisingly, DA-3 cells expressing and secreting MUC1/sec (DA-3/sec) fail to form tumors in vivo. The mechanism of rejection was evaluated using mice deficient in constituents of the immune system. All mice lacking IFN-gamma, NK, NKT, or macrophages formed DA-3/sec tumors that regressed shortly after implantation. However, progressively growing DA-3/sec tumors developed in mice devoid of T lymphocytes. The importance of T lymphocytes in the rejection of DA-3/sec tumors was further supported by detection of DA-3-specific CTL in mice challenged with the DA-3/sec tumor. Recruitment of appropriate APC and effector cells is an important first step in the tumor clearance. Indeed, DA-3/sec cells or cell supernatants recruited 3-4 times as many macrophages as DA-3/TM cells in vivo, suggesting that a secreted chemotactic product is produced from DA-3/sec cells. RNA and protein analysis of DA-3/sec cells revealed that several genes are up-regulated by MUC1/sec expression, including MCP-1 (CCL-2). These results suggest DA-3/sec cells are capable of recruiting immune cells, and that rejection of DA-3/sec tumors, although aided by cells of the innate immune response, is ultimately due to T cell-mediated events.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigens, Neoplasm/immunology
- Carrageenan/pharmacology
- Cell Line, Tumor/metabolism
- Chemokine CCL2/metabolism
- Chemotaxis, Leukocyte
- Disease Progression
- Female
- Gene Expression Regulation, Neoplastic
- Graft Rejection/immunology
- Immunologic Deficiency Syndromes/immunology
- Interferon-gamma/deficiency
- Killer Cells, Natural/immunology
- Lymphocyte Depletion
- Macrophages/drug effects
- Macrophages/immunology
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mucin-1/genetics
- Mucin-1/physiology
- Neoplasm Proteins/metabolism
- Neoplasm Proteins/physiology
- Neoplasm Transplantation/immunology
- Protein Isoforms/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Transfection
Collapse
Affiliation(s)
- Joseph F Grosso
- Department of Microbiology and Immunology, University of Miami School of Medicine, FL 33136, USA
| | | | | | | |
Collapse
|
104
|
Baniyash M. TCR zeta-chain downregulation: curtailing an excessive inflammatory immune response. Nat Rev Immunol 2004; 4:675-87. [PMID: 15343367 DOI: 10.1038/nri1434] [Citation(s) in RCA: 260] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The T-cell receptor (TCR) functions in both antigen recognition and signal transduction, which are crucial initial steps of antigen-specific immune responses. TCR integrity is vital for the induction of optimal and efficient immune responses, including the routine elimination of invading pathogens and the elimination of modified cells and molecules. Of the TCR subunits, the zeta-chain has a key role in receptor assembly, expression and signalling. Downregulation of TCR zeta-chain expression and impairment of T-cell function have been shown for T cells isolated from hosts with various chronic pathologies, including cancer, and autoimmune and infectious diseases. This review summarizes studies of the various pathologies that show this phenomenon and provides new insights into the mechanism responsible for downregulation of zeta-chain expression, its relevance and its clinical implications.
Collapse
Affiliation(s)
- Michal Baniyash
- The Lautenberg Center for General and Tumor Immunology, The Hebrew University-Hadassah Medical School, Post Office Box 12272, Jerusalem 91120, Israel.
| |
Collapse
|
105
|
Bohnenkamp HR, Coleman J, Burchell JM, Taylor-Papadimitriou J, Noll T. Breast carcinoma cell lysate-pulsed dendritic cells cross-prime MUC1-specific CD8+ T cells identified by peptide-MHC-class-I tetramers. Cell Immunol 2004; 231:112-25. [PMID: 15919376 DOI: 10.1016/j.cellimm.2004.12.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Revised: 12/14/2004] [Accepted: 12/21/2004] [Indexed: 02/02/2023]
Abstract
For cancer immunotherapy the loading of dendritic cells (DCs) with whole tumor cell lysate preparations represents a simple and promising approach for presentation of tumor-associated antigens (TAAs), avoiding the disadvantages of HLA-matching and definition of TAAs. The aim of this study was to investigate whether lysate-pulsed DCs efficiently cross-prime CD8+ T cells and induce a strong T(H)1 cell response, as compared to DCs pulsed with specific peptides (FLU M1 and Melan-A/Mart-1). As a model system breast carcinoma cell lysate from either MCF-7 or MDA-MB-231 cell lines (both HLA-A*0201+) expressing the TAA MUC1 were selected. Both cell lines expressed MUC1, the epithelial mucin, which is a large molecular weight O-glycosylated protein expressed in the majority of breast, ovarian, and other epithelial malignancies and is under evaluation as a target antigen in cancer immunotherapy. We developed a simple lysate preparation method to solubilize all cell proteins without degradation. For loading of monocyte-derived dendritic cells, 100 microgmL(-1) of breast carcinoma cell lysate was used, accompanied by an adjuvant consisting of tumor necrosis factor-alpha (TNF-alpha) and prostaglandin-E2. T cells were co-cultivated with lysate or peptide pulsed DCs and were restimulated weekly. Before cultivation, and after the 3rd stimulation, tetramer frequencies for the MUC1 epitopes M1.2 and F7 as well as for the FLU M1 and Melan-A/Mart-1 epitopes were determined. After stimulation with lysate, higher frequencies for M1.2-specific T cells were observed compared with the F7 epitope. Furthermore, we found expansion factors for M1.2-specific T cells that had been stimulated with MCF-7 lysate-pulsed DCs of up to 43-fold. The analysis of typical T(H)1/T(H)2 cytokines (IFN-gamma, TNF-alpha, IL-12p70, IL-2, IL-4, IL-5, and IL-10) revealed a strong T(H)1 response. These results provide evidence for a strong T(H)1 polarization and cross-priming of MUC1-specific CD8+ T cells and demonstrate the feasibility of using lysate-pulsed dendritic cells in breast cancer immunotherapy.
Collapse
Affiliation(s)
- Hermann R Bohnenkamp
- Institute of Biotechnology 2, Research Center Jülich GmbH, Leo-Brandt-Strasse 1, 52428 Jülich, Germany.
| | | | | | | | | |
Collapse
|
106
|
ten Dam GB, Poels LG, Pullens R, Jap PHK, van de Molengraft FJJM. Expression of a colorectal antigen defined by a new monoclonal antibody, CO-TL1. J Transl Med 2004; 84:1160-73. [PMID: 15195115 DOI: 10.1038/labinvest.3700130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A murine monoclonal antibody (MoAb CO-TL1, IgG1) has been raised by differential screening of hybridoma supernatants on sections of human large and small intestines, followed by screening on colon adenomas as well as on colorectal carcinomas. In both paraffin sections and cryostat sections, the antibody stained strongly all cell types in adult, neonatal and fetal human colorectal epithelium, that is, the goblet cells, the columnar cells and the endocrine cells. No staining was observed in the remaining parts of the normal gastrointestinal tract and other tissues. As revealed by immuno electron microscopy the epitope was present in the apical and basolateral cell membranes, the Golgi complex, secretory vesicles of goblet and columnar cells, and also in granules of the endocrine cells. The epitope in colorectal tissue sections was resistant to the deglycosylation enzymes neuramidase, diastase and hyaluronidase indicating its proteinaceous nature. This colorectal antigen remained expressed in 100% of colorectal adenomas (n = 39) and 86% (n = 29) of colorectal carcinomas. The expression was reduced in undifferentiated carcinomas. The CO-TL1 antibody detected also most other gastrointestinal adenocarcinomas and a few carcinomas of the ovary, uterus, breast, gallbladder and pancreas. However, it never detected carcinomas derived from the thyroid, lung, liver, bladder, kidney, prostate, testis, serous membranes of body cavities and skin. A wild-type variant protein of > 300 kDa of the colorectal antigen was identified in normal colorectal epithelium. In colorectal tumours, however, two tumour variant forms were found of 160-200 and 115-140 kDa, respectively. Our data indicate that this new MoAb CO-TL1 can be considered as a useful marker, which identifies normal colorectal epithelium and gastrointestinal tumours and especially colorectal tumours with high accuracy and excludes tumours originated from thyroid, lung, liver, bladder, kidney, prostate, testis, mesothelium and skin.
Collapse
Affiliation(s)
- Gerdy B ten Dam
- Department of Biochemistry, Nijmegen Center for Molecular Life Sciences, University Medical Center, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
107
|
Rahbarizadeh F, Rasaee MJ, Forouzandeh Moghadam M, Allameh AA, Sadroddiny E. Production of Novel Recombinant Single-Domain Antibodies against Tandem Repeat Region of MUC1 Mucin. ACTA ACUST UNITED AC 2004; 23:151-9. [PMID: 15312305 DOI: 10.1089/1536859041224334] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recently, the existence of "heavy-chain" antibody in Camelidae has been described. However, as yet there is no data on the binding of this type of antibody to peptides. In addition, there was not any report of production of single-domain antibodies in two-humped camels (Camelus bactrianus). In the present study, these questions are addressed. We showed the feasibility of immunizing old world camels, cloning the repertoire of the variable domain of their heavy-chain antibodies, panning and selection, leading to the successful identification of minimum-sized antigen binders. Antigen-specific fragments of the heavy-chain IgGs (V(HH)) are of great interest in biotechnology because they are very stable, highly soluble, and react specifically and with high affinity to the antigens. In this study, we immunized two camels (Camelus dromedarius and Camelus bactrianus) with homogenized cancerous tissues, synthetic peptide, and human milk fat globule membrane (HMFG), and generated two V(HH) libraries displayed on phage particles. Some single-domain antibody fragments have been isolated that specifically recognize the tandem repeat region of MUC1. The camels' single-domain V(HH) harbor the original, intact antigen binding site and reacted specifically and with high affinity to the tandem repeat region of MUC1. Indeed soluble, specific antigen binders and good affinities (in the range of 0.2 x 10(9) M(-1) to 0.6 x 10(9) M(-1)) were identified from these libraries. This is the first example of the isolation of camel anti-peptide V(HH) domains.
Collapse
Affiliation(s)
- F Rahbarizadeh
- Department of Clinical Biochemistry, School of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | | | | | | | | |
Collapse
|
108
|
Abstract
MUCs are glycoproteins with various roles in homeostasis and carcinogenesis. Among other actions, MUC1 may inhibit cell-cell and cell-stroma interactions and function as a signal transducer, participating in cancer progression. In contrast, MUC2 is normally found only in goblet cells, where it contributes to the protective barrier function of these cells. Recently, a tumour suppressor role has been demonstrated for MUC2, and both MUC1 and MUC2 appear to have important roles in pancreatic neoplasia. MUC1 appears to be a marker of aggressive phenotype and may facilitate the vascular spread of carcinoma cells. In contrast, MUC2 is rarely detectable in aggressive pancreatic tumours, but is commonly expressed in intraductal papillary mucinous neoplasms (IPMNs), which are rare, indolent tumours, in intestinal IPMNs, and in indolent colloid carcinomas. MUC2 appears to be not only a marker of this indolent pathway, but also partly responsible for its less aggressive nature. Thus, in pancreatic neoplasia, MUC1 and MUC2 have potential diagnostic and prognostic value as markers of aggressive and indolent phenotypes, respectively, and have potential as therapeutic targets.
Collapse
Affiliation(s)
- E Levi
- John Dingell Veterans Administration Medical Center, 4646 John Road, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
109
|
Plunkett T, Graham R, Correa I, Sewell R, Miles D, Burchell J, Taylor-Papadimitriou J. Protection against MUC1 expressing mouse tumours by intra-muscular injection of MUC1 cDNA requires functional CD8+ and CD4+ T cells but does not require the MUC1 tandem repeat domain. Int J Cancer 2004; 109:691-7. [PMID: 14999776 DOI: 10.1002/ijc.20040] [Citation(s) in RCA: 15] [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
Using a C57Bl/6 mouse model system, where intramuscular (i.m.) injection of full length (FL) MUC1 cDNA protects against subsequent challenge with MUC1-expressing syngeneic tumour cells, we have investigated the importance of the tandem repeat (TR) domain in the induction of T cell-dependent tumour rejection. A MUC1 construct engineered to remove the TR domain (MUC1 0TR) was found to be as effective as the full length MUC1 cDNA in inhibiting the growth of RMA MUC1 cells in C57Bl/6 mice. Protection by i.m. injection of either the FL-MUC1 cDNA or the MUC1 0TR construct depended on the presence of functional CD4+ and CD8+ T cells. Specific CD8+ T cell responses, however, could not be detected in vitro using mouse spleen cells taken after only cDNA injection, but only after challenge in vivo with MUC1-expressing tumour cells. To attempt to enhance the responses of CD4+ T cells, a cDNA construct was developed, where the extracellular domain of MUC1 was fused to the transmembrane and cytoplasmic domain of Lamp1 (MUC1/Lamp1). This construct was equally effective in inducing tumour rejection but did not induce MUC1-specific CTL in mice before challenge with MUC1-expressing tumour cells. Our results indicate that, in this model, T cell responses necessary for protection against MUC1-expressing tumours that are induced by IM injection of MUC1 cDNA are independent of the tandem repeat domain as well as the transmembrane and cytoplasmic domains. A low level of protection was seen with all constructs in BALB/c mice, which show a defect in Th1 responses. C57Bl/6xBALB/c hybrids were, however, well protected against both H2(d) and H2(b) expressing tumour challenge, emphasizing the importance of the host background.
Collapse
Affiliation(s)
- Tim Plunkett
- Breast Cancer Biology Group, Cancer Research UK, 3rd Floor Thomas Guy House, Guy's Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
110
|
Dziadek S, Kunz H. Synthesis of tumor-associated glycopeptide antigens for the development of tumor-selective vaccines. CHEM REC 2004; 3:308-21. [PMID: 14991920 DOI: 10.1002/tcr.10074] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In contrast to normal cells, the glycoprotein profile on epithelial tumor cells is distinctly altered. Due to an incomplete formation of the glycan side-chains resulting from a premature sialylation, additional peptide epitopes become accessible to the immune system in mucin-type glycoproteins on tumor cells. These tumor-associated structure alterations constitute the basis for a selective immunological attack on cancer cells. For the construction of immunostimulating antigens, glycopeptide partial structures from the mucins MUC1 and MUC4 carrying the tumor-associated sialyl-T(N), alpha2,6-sialyl-T and alpha2,3-sialyl-T antigens have been synthesized. Employing different linkers such as the allylic HYCRON or the fluoride-sensitive PTMSEL anchor, the antigenic glycopeptide structures were constructed on the solid phase utilizing pre-assembled glycosyl amino acid building blocks prepared in solution by convergent chemical or chemoenzymatic strategies. The proliferation of cytotoxic T cells has been induced applying a construct composed of a sialyl-T(N) MUC1-glycopeptide conjugated with a tetanus toxin T cell peptide epitope.
Collapse
Affiliation(s)
- Sebastian Dziadek
- Institut für Organische Chemie, Johannes-Gutenberg-Universität Mainz, Duesbergwerg 10-14, D-55128 Mainz, Germany
| | | |
Collapse
|
111
|
Martinson J, Bae J, Klingemann HG, Tam Y. Activated platelets rapidly up-regulate CD40L expression and can effectively mature and activate autologous ex vivo differentiated DC. Cytotherapy 2004; 6:487-97. [PMID: 15512915 DOI: 10.1080/14653240410005249] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background DC are a promising immunotherapeutic for treatment of infectious/malignant disease. For clinical trials, immature DC generated from precursor cells such as monocytes, using serum-free media containing GM-CSF and IL-4, can be matured with specific cytokines/factors. CD40 ligand (CD40L) plays an important role in DC activation/maturation but is not available for clinical applications. These studies evaluated the feasibility of using activated platelets with elevated CD40L surface expression to stimulate autologous DC maturation. Methods Pilot and clinical scale studies were executed using magnetic/centrifugal separation. Monocyte precursors were differentiated to immature DC with GM-CSF and IL-4 and the ability of activated autologous platelets to mature these cells was evaluated on the basis of phenotype and function. Results In small-scale studies, DC cultures stimulated with activated autologous platelets (CD40L-AP), tumor necrosis factor-alpha (TNF-alpha) or soluble CD40L (sCD40L) up-regulated expression of phenotype markers indicative of activation and maturation. CD86 expression was significantly enhanced (P<0.05) by stimulation with either CD40L-AP (75.5+/-14.5%) or sCD40L (80.5%+/-5.3%) compared with immature DC (55.2+/-14.8%), as were CD80 and CD83. Similarly, in large-scale studies using Isolex 300I to enrich for monocytes and platelets for DC generation/maturation on a clinical scale, stimulation with CD40L-AP increased CD86 and CD80 expression as well as the ability to stimulate allogeneic lymphocytes compared with control cultures. Discussion These results demonstrate that thrombin-activated platelets express CD40L and are effective at inducing matured DC with potent immunogenic activity. Furthermore, these studies demonstrate the feasibility of this approach for clinical immunotherapeutic interventions.
Collapse
Affiliation(s)
- Ja Martinson
- Section of Bone Marrow Transplant and Cell Therapy, Rush Presbyterian-St. Luke's Medical Center, Rush Medical School, Chicago, IL, USA
| | | | | | | |
Collapse
|
112
|
Affiliation(s)
- Jeffrey W Pollard
- Center for the Study of Reproductive Biology and Women's Health and the Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York 10461, USA.
| |
Collapse
|
113
|
Gum JR, Hicks JW, Crawley SC, Dahl CM, Yang SC, Roberton AM, Kim YS. Initiation of transcription of the MUC3A human intestinal mucin from a TATA-less promoter and comparison with the MUC3B amino terminus. J Biol Chem 2003; 278:49600-9. [PMID: 12958310 DOI: 10.1074/jbc.m305769200] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human intestinal mucin genes MUC3A and MUC3B are members of a membrane mucin gene family residing at chromosome 7q22. In this paper, we utilized genomic and cDNA cloning to elucidate the sequence of the 5'-region of the MUC3A gene including the gene promoter and the amino terminus coding sequence. Following its 21-residue signal peptide, the amino terminus of the mucin consists of a 233-residue Thr-, Ser-, and Pro-rich nonrepetitive sequence that is contiguous with its hypervariable domain of 375-residue repeats. RNase protection analysis and 5'-GeneRacer PCR indicated that MUC3A gene transcripts initiate from multiple start sites along a region spanning approximately 180 bases. The 5'-flanking region of the gene had promoter activity when fused to a luciferase reporter gene in all of the tested cell lines. This region contained binding sites for several transcription factors, including those implicated in the regulation of intestinal genes, but lacked a cognate TATA box. These features of the gene promoter may enable the gene to be expressed at variable levels in several cell types with different repertoires of transcription factors. We also utilized 5'-GeneRacer PCR to determine the sequence of the 5'-terminus of the MUC3B message. The amino termini of the MUC3A and MUC3B mucins are 91% conserved at the amino acid level. Thus, MUC3A and MUC3B have highly conserved amino and carboxyl termini, suggesting a recent duplication of the entire ancestral gene. It remains to be determined whether other members of the 7q22 membrane mucin gene family have amino-terminal domains similar to MUC3A and MUC3B.
Collapse
Affiliation(s)
- James R Gum
- Department of Veterans Affairs Medical Center, San Francisco, California 94121, USA.
| | | | | | | | | | | | | |
Collapse
|
114
|
Brokx RD, Revers L, Zhang Q, Yang S, Mal TK, Ikura M, Gariépy J. Nuclear magnetic resonance-based dissection of a glycosyltransferase specificity for the mucin MUC1 tandem repeat. Biochemistry 2003; 42:13817-25. [PMID: 14636048 DOI: 10.1021/bi0353070] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The human glycoprotein MUC1 mucin plays a critical role in cancer progression. Breast, ovarian, and colon cancer cells often display unique cell-surface antigens corresponding to aberrantly glycosylated forms of the MUC1 tandem repeat. In this report, (15)N- and (13)C-labeled forms of a recombinant MUC1 construct containing five tandem repeats were used as substrates to define the order and kinetics of addition of N-acetylgalactosamine (GalNAc) moieties by a recombinant active form of the human enzyme UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase I (ppGalNAc-T1; residues 40-559). Heteronuclear NMR experiments were performed to assign resonances associated with the two serines (Ser5 and Ser15) and three threonines (Thr6, Thr14, and Thr19) present in the 20-residue long MUC1 repeat. The kinetics and order of addition of GalNAc moieties (Tn antigen) on the MUC1 construct by human ppGalNAc-T1 were subsequently dissected by NMR spectroscopy. Threonine 14 was shown to be rapidly glycosylated by ppGalNAc-T1 with an initial rate of 25 microM/min, followed by Thr6 (8.6 microM/min). The enzyme also modified Ser5 at a slower rate (1.7 microM/min), an event that started only after the glycosylation of Thr14 and Thr6 side chains was mostly completed. Ser15 and Thr19 remained unglycosylated by ppGalNAc-T1. Corresponding O-glycosylation sites within all five tandem repeats were simultaneously modified by ppGalNAc-T1, suggesting that each repeat behaves as an independent substrate unit. This study demonstrated that the hydroxyl oxygens of Thr14 and to a lesser extent Thr 6 represent the two dominant substrates modified by ppGalNAc-T1 within the context of a complex MUC1 peptide substrate. More importantly, the availability of defined isotopically labelled MUC1 glycopeptide substrates and the relative simplicity of their NMR spectra will facilitate the analysis of other transferases within the O-glycosylation pathways and the rational design of tumor-associated MUC1 antigens.
Collapse
Affiliation(s)
- Richard D Brokx
- Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | |
Collapse
|
115
|
Scholl S, Squiban P, Bizouarne N, Baudin M, Acres B, von Mensdorff-Pouilly S, Shearer M, Beuzeboc P, Van Belle S, Uzielly B, Pouillart P, Taylor-Papadimitriou J, Miles D. Metastatic Breast Tumour Regression Following Treatment by a Gene-Modified Vaccinia Virus Expressing MUC1 and IL-2. J Biomed Biotechnol 2003; 2003:194-201. [PMID: 12975534 PMCID: PMC400208 DOI: 10.1155/s111072430320704x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
MUC1 is expressed by glandular epithelial cells. It is overexpressed in the majority of breast tumours, making it a potential target for immune therapy. The objectives of the present study were to evaluate the anti-tumour activity and tolerance of repeated administration of TG1031 (an attenuated recombinant vaccinia virus containing sequences coding for human MUC1 and the immune stimulatory cytokine IL-2) in patients with MUC1-positive metastatic breast cancer. This was an open-label, randomised study comparing two dose levels, 5 x 10E6 and 5 x 10E7, with 14 patients in each arm. The treatment was administered intramuscularly every 3 weeks for the first 4 doses and every 6 weeks thereafter, until progression. Two patients had a partial tumour regression ( > 50%), and 15 patients had stable disease as their best overall response until at least the 5th injection. Partial regression lasted for 11 months in one patient and for 12 months in the second patient who then underwent surgical resection of her hepatic metastases. The most frequent adverse events included inflammation at injection site: 7 patients, itching or pain at injection site: 5 patients, and moderate fever: 6 patients. One responding patient developed antinuclear, anti-DNA, and increased anti-TPO antibodies after the fifth injection, and which resolved at the end of treatment. The treatment regimes were well tolerated with a low toxicity profile. Although clinical efficacy remains limited, this study demonstrates the potential use of MUC1-based immune therapy in breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Moira Shearer
- Cancer Research UK Breast Cancer
Biology Group, Guy's Hospital, London, UK
| | | | | | | | | | | | - David Miles
- Cancer Research UK Breast Cancer
Biology Group, Guy's Hospital, London, UK
| |
Collapse
|
116
|
Gipson IK, Argüeso P. Role of Mucins in the Function of the Corneal and Conjunctival Epithelia. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 231:1-49. [PMID: 14713002 DOI: 10.1016/s0074-7696(03)31001-0] [Citation(s) in RCA: 213] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The surface of the eye is covered by a tear film, which is held in place by a wet-surfaced, stratified, corneal and conjunctival epithelia. Both are vital for light refraction and protection of vision. Maintenance of tear film on the ocular surface, lubrication, and provision of a pathogen barrier on this wet surface is facilitated by a class of large, highly glycosylated, hydrophilic glycoproteins--the mucins. In the past 15 years, a number of mucin genes have been cloned, and based on protein sequence, categorized as either secreted or membrane associated. Both types of mucins are expressed by ocular surface epithelia. Goblet cells intercalated within the stratified epithelium of the conjunctiva secrete the large gel-forming mucin MUC5AC, and lacrimal gland epithelia secrete the small soluble mucin MUC7. Apical cells of the stratified epithelium of both corneal and conjunctival epithelium express at least three membrane-associated mucins (MUCs 1, 4, and 16), which extend from their apical surface to form the thick glycocalyx at the epithelium-tear film interface. The current hypothesis regarding mucin function and tear film structure is that the secreted mucins form a hydrophilic blanket that moves over the glycocalyx of the ocular surface to clear debris and pathogens. Mucins of the glycocalyx prevent cell-cell and cell-pathogen adherence. The expression and glycosylation of mucins are altered in drying, keratinizing ocular surface diseases.
Collapse
Affiliation(s)
- Ilene K Gipson
- Schepens Eye Research Institute and the Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|