Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Crit Rev Immunol ; 34(5): 399-432, 2014.
Article in English | MEDLINE | ID: mdl-25404047

ABSTRACT

Anti-tumor immunotherapy using tumor lysate-based vaccines has made great advances over recent decades. Cancer vaccines aim to elicit adaptive immune responses through various pathways by providing tumor and tumor-associated antigens with an immune stimulant or adjuvant. These anti-tumor vaccines are therefore developed as personalized treatments. Utilizing tumors as a source of vaccine antigens in immunotherapy has demonstrated promising results with minimal toxicity. However, to date, researchers have failed to overcome the overpowering immune suppressive effects within the tumor microenvironment. Immune suppression occurs naturally via multiple mechanisms. These mechanisms serve an important homeostatic role restoring a normal tissue microenvironment following an inflammatory response. Due to these suppressive mechanisms and the inherent heterogeneity of tumors, it is imperative to then elicit and maintain a specific tumoricidal response if vaccine therapy or some other combination of reagents is chosen. In this review, we focus on the historical use of tumors as a source of antigens to elicit a tumoricidal response and the limitations encountered that prevent greater success in immunotherapy. We describe the advantages and disadvantages of various vaccines and their ineffectiveness due to tumor-induced immune suppression.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Dog Diseases/therapy , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Neoplasm/immunology , Brain Neoplasms/immunology , Brain Neoplasms/veterinary , Dog Diseases/immunology , Dogs , Humans , Immunosuppression Therapy , Precision Medicine , Remission Induction , Treatment Failure , Tumor Escape , Tumor Microenvironment
2.
Target Oncol ; 9(3): 239-49, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23900680

ABSTRACT

The discovery of chemoresistant cancer stem cells (CSCs) in carcinomas has created the need for therapies that specifically target these subpopulations of cells. Here, we characterized a bispecific targeted toxin that is composed of two antibody fragments and a catalytic protein toxin allowing it to bind two CSC markers on the same cell killing this resistant subpopulation. CD133 is a well-known CSC marker and has been successfully targeted and caused regression of head and neck squamous cell carcinoma (HNSCC) in vivo. To enable it to bind a broader range of CSCs, an anti-epithelial cell adhesion molecule (EpCAM) scFv was added to create dEpCAMCD133KDEL, a deimmunized bispecific targeted toxin on a single amino acid chain. This bispecific potently inhibited protein translation and proliferation in vitro in three different types of carcinoma. Furthermore, in a CSC spheroid model dEpCAMCD133KDEL eliminated Mary-X spheroids, an inflammatory breast carcinoma. Finally, this bispecific also caused tumor regression in an in vivo model of HNSCC. This represents the first bispecific CSC-targeted toxin and warrants further development as a possible therapy for carcinoma.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Carcinoma, Squamous Cell/therapy , Cell Adhesion Molecules/immunology , Glycoproteins/immunology , Head and Neck Neoplasms/therapy , Immunotoxins/pharmacology , Peptides/immunology , AC133 Antigen , Animals , Antibodies, Bispecific/immunology , Caco-2 Cells , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Epithelial Cell Adhesion Molecule , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Immunotoxins/immunology , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Squamous Cell Carcinoma of Head and Neck , Transfection
3.
J Immunother Cancer ; 2(1): 46, 2014.
Article in English | MEDLINE | ID: mdl-25598973

ABSTRACT

BACKGROUND: Immunological quiescence in the central nervous system (CNS) is a potential barrier to immune mediated anti-tumor response. One suppressive mechanism results from the interaction of parenchyma-derived CD200 and its receptor on myeloid cells. We suggest that CD200/CD200R interactions on myeloid cells expand the myeloid-derived suppressor cell (MDSC) population and that blocking tumor-derived CD200 will enhance the efficacy of immunotherapy. METHODS: CD200 mRNA expression levels in human brain tumor tissue samples were measured by microarray. The amount of circulating CD200 protein in the sera of patients with brain tumors was determined by ELISA and, when corresponding peripheral blood samples were available, was correlated quantitatively with MDSCs. CD200-derived peptides were used as competitive inhibitors in a mouse model of glioblastoma immunotherapy. RESULTS: CD200 mRNA levels were measured in human brain tumors, with different expression levels being noted among the sub groups of glioblastoma, medulloblastoma and ependymoma. Serum CD200 concentrations were highest in patients with glioblastoma and correlated significantly with MDSC expansion. Similarly, in vitro studies determined that GL261 cells significantly expanded a MDSC population. Interestingly, a CD200R antagonist inhibited the expansion of murine MDSCs in vitro and in vivo. Moreover, inclusion of CD200R antagonist peptide in glioma tumor lysate-derived vaccines slowed tumor growth and significantly enhanced survival. CONCLUSION: These data suggest that CNS-derived tumors can evade immune surveillance by engaging CD200. Because of the homology between mouse and human CD200, our data also suggest that blockade of CD200 binding to its receptor will enhance the efficacy of immune mediated anti-tumor strategies for brain tumors.

4.
Gynecol Oncol ; 130(3): 579-87, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23721800

ABSTRACT

OBJECTIVES: While most women with ovarian cancer will achieve complete remission after treatment, the majority will relapse within two years, highlighting the need for novel therapies. Cancer stem cells (CSC) have been identified in ovarian cancer and most other carcinomas as a small population of cells that can self-renew. CSC are more chemoresistant and radio-resistant than the bulk tumor cells; it is likely that CSC are responsible for relapse, the major problem in cancer treatment. CD133 has emerged as one of the most promising markers for CSC in ovarian cancer. The hypothesis driving this study is that despite their low numbers in ovarian cancer tumors, CSC can be eradicated using CD133 targeted therapy and tumor growth can be inhibited. METHODS: Ovarian cancer cell lines were evaluated using flow cytometry for expression of CD133. In vitro viability studies with an anti-CD133 targeted toxin were performed on one of the cell lines, NIH:OVCAR5. The drug was tested in vivo using a stably transfected luciferase-expressing NIH:OVCAR5 subline in nude mice, so that tumor growth could be monitored by digital imaging in real time. RESULTS: Ovarian cancer cell lines showed 5.6% to 16.0% CD133 expression. dCD133KDEL inhibited the in vitro growth of NIH:OVCAR5 cells. Despite low numbers of CD133-expressing cells in the tumor population, intraperitoneal drug therapy caused a selective decrease in tumor progression in intraperitoneal NIH:OVCAR5-luc tumors. CONCLUSIONS: Directly targeting CSC that are a major cause of drug resistant tumor relapse with an anti-CD133 targeted toxin shows promise for ovarian cancer therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Cytotoxins/therapeutic use , Glycoproteins/metabolism , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/drug therapy , Peptides/metabolism , AC133 Antigen , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytotoxins/pharmacology , Disease Models, Animal , Disease Progression , Female , Glycoproteins/immunology , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Peptides/immunology
5.
Drug Deliv Transl Res ; 3(2): 143-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-25787982

ABSTRACT

CD133, also known as Prominin-1, is expressed on stem cells present in many tissues and tumors. In this work, we have identified and characterized a single-chain variable fragment (scFv) for the efficient and specific recognition of CD133. Phage display was used to develop the scFv from a previously reported anti-CD133 hybridoma clone 7, which was capable of recognizing both glycosylated and non-glycosylated forms of human CD133. The scFv immunostained CD133(+) Caco-2 cells, but not CD133(-/low) U87 cells. Significantly, it immunostained CD133(-) cells transiently transfected with the mouse CD133 gene as well as CD133(+) mouse cells. Co-immunostaining studies in mouse bone marrow cells, using anti-CD133 scFv-FITC and anti-mouse CD133-PE (clone 13A4) commercial antibody, indicated that the epitopes recognized by these reagents partially overlap. Taken together, these results suggest that the scFv can recognize mouse CD133 protein in addition to recognizing human CD133. This new scFv is expected to be valuable both as a molecular diagnostic reagent for identifying CD133(+) cells and as a ligand for targeting therapeutics to CD133(+) tumor-initiating cells.

6.
Drug Deliv Transl Res ; 3(2): 195-204, 2013 Apr.
Article in English | MEDLINE | ID: mdl-25787984

ABSTRACT

CD133 expression enriches for tumor-initiating cells and is a negative prognostic factor in numerous cancers. We previously developed an immunotoxin against CD133 by fusing a gene fragment encoding the scFv portion of an anti-CD133 antibody to a gene fragment encoding deimmunized PE38KDEL. The resulting fusion protein, dCD133KDEL, demonstrated potent antitumor activity following intratumoral delivery into head neck cell carcinoma xenografts. However, the efficacy against other tumors and the tolerability of systemic administration remained unclear. The purpose of this study was to evaluate the tolerability and efficacy of dCD133KDEL in a systemic human breast carcinoma model. Time course viability studies showed that dCD133KDEL selectively inhibited MDA-MB-231 ductal breast carcinoma cells that contained a minority CD133(+) subpopulation, implicating CD133(+) cells as a source for self-renewal within this cell line. Furthermore, systemic administration of dCD133KDEL caused regression or inhibition of tumor growth in mice bearing an intrasplenic MDA-MB-231 tumor challenge as a model for metastatic disease. In the same model, combined therapy with dCD133KDEL and another immunotoxin designed to target the bulk tumor mass was the most effective therapy, supporting the idea that such combination therapies might better address tumor heterogeneity. dCD133KDEL shows promise as a therapeutic agent and as a biologic tool to study cancer stem cells.

7.
Oral Oncol ; 48(12): 1202-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22818892

ABSTRACT

OBJECTIVES: To investigate the efficacy of the bispecific targeted toxin, dEGFATFKDEL, on head and neck carcinoma cell lines in vitro and in vivo. MATERIALS AND METHODS: A deimmunized bispecific anti-cancer agent was constructed to simultaneously target both the overexpressed EGF receptor on carcinomas and the urokinase receptor (uPAR), that is found on the endothelial cells of the neovasculature within tumors. Flow cytometry assays were performed to determine the level of EGFR expressed on a variety of carcinoma lines. These lines were then tested in tritiated leucine incorporation assays to determine the efficacy of dEGFATFKDEL. Human vein endothelial primary cells were also tested to determine the effectiveness of the ATF portion of the molecule that binds uPAR. Furthermore, mouse studies were performed to determine whether dEGFATFKDEL was effective at inhibiting tumor growth in vivo. RESULTS: UMSCC-11B and NA, two head and neck squamous cell carcinomas, highly expressed EGFR. Both the carcinoma lines and the human vein endothelial cells were inhibited at sub-nanomolar concentrations by dEGFATFKDEL. The tumor studies showed that the tumors treated with dEGFATFKDEL were significantly inhibited whereas the negative control and untreated tumors progressed. In a separate in vivo study involving another carcinoma line, MDA-MB-231, the effectiveness of dEGFATFKDEL was confirmed. No toxicity was seen at the doses used in either of these mouse studies. CONCLUSIONS: This bispecific agent is effective in a mouse model of head and neck squamous cell carcinoma. Further study of this reagent for use in the treatment of carcinomas is warranted.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Disease Models, Animal , ErbB Receptors/drug effects , Head and Neck Neoplasms/metabolism , Oligopeptides/pharmacology , Receptors, Urokinase Plasminogen Activator/drug effects , Animals , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , ErbB Receptors/metabolism , Flow Cytometry , Head and Neck Neoplasms/pathology , Humans , Male , Mice , Mice, Nude , Oligopeptides/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism
8.
Mol Cancer Ther ; 10(10): 1829-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21862685

ABSTRACT

A novel anticancer agent was constructed by fusing a gene encoding the scFV that targets both glycosylated and unglycosylated forms of CD133 to a gene fragment encoding deimmunized PE38KDEL. The resulting fusion protein, dCD133KDEL, was studied to determine its ability to bind and kill tumor-initiating cells in vitro and in vivo. The anti-CD133 scFV selectively bound HEK293 cells transfected with the CD133 receptor gene. Time course viability studies showed that dCD133KDEL selectively inhibited NA-SCC and UMSCC-11B, 2 head and neck squamous cell carcinomas that contain a CD133 expressing subpopulation. Importantly, the drug did not inhibit the viability of hematopoietic lineages measured by long-term culture-initiating cell and colony-forming assays from sorted human CD34+ progenitor cells. In addition to in vitro studies, in vivo tumor initiation experiments confirmed that CD133-sorted cells implanted into the flanks of nude mice grew faster and larger than unsorted cells. In contrast, cells that were pretreated with dCD133KDEL before implantation showed the slowest and lowest incidence of tumors. Furthermore, UMSCC-11B-luc tumors treated with multiple intratumoral injections of dCD133KDEL showed marked growth inhibition, leading to complete degradation of the tumors that was not observed with an irrelevant control-targeted toxin. Experiments in immunocompetent mice showed that toxin deimmunization resulted in a 90% reduction in circulating antitoxin levels. These studies show that dCD133KDEL is a novel anticancer agent effective at inhibiting cell proliferation, tumor initiation, and eliminating established tumors by targeting the CD133 subpopulation. This agent shows significant promise for potential development as a clinically useful therapy.


Subject(s)
Antigens, CD/immunology , Carcinoma, Squamous Cell/drug therapy , Glycoproteins/immunology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Immunotoxins/pharmacology , Neoplastic Stem Cells/drug effects , Peptides/immunology , Recombinant Fusion Proteins/pharmacology , AC133 Antigen , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacokinetics , ADP Ribose Transferases/pharmacology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Bacterial Toxins/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Exotoxins/genetics , Exotoxins/pharmacokinetics , Exotoxins/pharmacology , Female , Glycoproteins/biosynthesis , Glycoproteins/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/pharmacology , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, Nude , Peptides/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Transfection , Virulence Factors/genetics , Virulence Factors/pharmacokinetics , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL
...