Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Eur Urol ; 72(1): 142-150, 2017 07.
Article in English | MEDLINE | ID: mdl-28408175

ABSTRACT

BACKGROUND: Although cisplatin-based neoadjuvant chemotherapy (NAC) improves survival of unselected patients with muscle-invasive bladder cancer (MIBC), only a minority responds to therapy and chemoresistance remains a major challenge in this disease setting. OBJECTIVE: To investigate the clinical significance of oncofetal chondroitin sulfate (ofCS) glycosaminoglycan chains in cisplatin-resistant MIBC and to evaluate these as targets for second-line therapy. DESIGN, SETTING, AND PARTICIPANTS: An ofCS-binding recombinant VAR2CSA protein derived from the malaria parasite Plasmodium falciparum (rVAR2) was used as an in situ, in vitro, and in vivo ofCS-targeting reagent in cisplatin-resistant MIBC. The ofCS expression landscape was analyzed in two independent cohorts of matched pre- and post-NAC-treated MIBC patients. INTERVENTION: An rVAR2 protein armed with cytotoxic hemiasterlin compounds (rVAR2 drug conjugate [VDC] 886) was evaluated as a novel therapeutic strategy in a xenograft model of cisplatin-resistant MIBC. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Antineoplastic effects of targeting ofCS. RESULTS AND LIMITATIONS: In situ, ofCS was significantly overexpressed in residual tumors after NAC in two independent patient cohorts (p<0.02). Global gene-expression profiling and biochemical analysis of primary tumors and cell lines revealed syndican-1 and chondroitin sulfate proteoglycan 4 as ofCS-modified proteoglycans in MIBC. In vitro, ofCS was expressed on all MIBC cell lines tested, and VDC886 eliminated these cells in the low-nanomolar IC50 concentration range. In vivo, VDC886 effectively retarded growth of chemoresistant orthotopic bladder cancer xenografts and prolonged survival (p=0.005). The use of cisplatin only for the generation of chemoresistant xenografts are limitations of our animal model design. CONCLUSIONS: Targeting ofCS provides a promising second-line treatment strategy in cisplatin-resistant MIBC. PATIENT SUMMARY: Cisplatin-resistant bladder cancer overexpresses particular sugar chains compared with chemotherapy-naïve bladder cancer. Using a recombinant protein from the malaria parasite Plasmodium falciparum, we can target these sugar chains, and our results showed a significant antitumor effect in cisplatin-resistant bladder cancer. This novel treatment paradigm provides therapeutic access to bladder cancers not responding to cisplatin.


Subject(s)
Antigens, Protozoan/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Chondroitin Sulfates/metabolism , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Oligopeptides/pharmacology , Urinary Bladder Neoplasms/drug therapy , Animals , Antigens, Protozoan/metabolism , Antineoplastic Agents/adverse effects , British Columbia , Cell Death/drug effects , Cell Line, Tumor , Cisplatin/adverse effects , Dose-Response Relationship, Drug , Europe , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Mice , Time Factors , Treatment Outcome , Tumor Burden/drug effects , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Cancer Cell ; 28(4): 500-514, 2015 10 12.
Article in English | MEDLINE | ID: mdl-26461094

ABSTRACT

Plasmodium falciparum engineer infected erythrocytes to present the malarial protein, VAR2CSA, which binds a distinct type chondroitin sulfate (CS) exclusively expressed in the placenta. Here, we show that the same CS modification is present on a high proportion of malignant cells and that it can be specifically targeted by recombinant VAR2CSA (rVAR2). In tumors, placental-like CS chains are linked to a limited repertoire of cancer-associated proteoglycans including CD44 and CSPG4. The rVAR2 protein localizes to tumors in vivo and rVAR2 fused to diphtheria toxin or conjugated to hemiasterlin compounds strongly inhibits in vivo tumor cell growth and metastasis. Our data demonstrate how an evolutionarily refined parasite-derived protein can be exploited to target a common, but complex, malignancy-associated glycosaminoglycan modification.


Subject(s)
Antigens, Protozoan/genetics , Chondroitin Sulfates/metabolism , Melanoma, Experimental/therapy , Placenta/metabolism , Recombinant Proteins/administration & dosage , Skin Neoplasms/therapy , Animals , Antigens, Protozoan/metabolism , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/metabolism , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronan Receptors/metabolism , Melanoma, Experimental/metabolism , Membrane Proteins/metabolism , Mice , Molecular Targeted Therapy , Oligopeptides/genetics , Oligopeptides/metabolism , Organ Specificity , Pregnancy , Recombinant Proteins/pharmacology , Skin Neoplasms/metabolism
3.
Cancer Immunol Res ; 3(9): 1052-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25943534

ABSTRACT

Programmed cell-death 1 ligand 1 (PD-L1) is a member of the B7/CD28 family of proteins that control T-cell activation. Many tumors can upregulate expression of PD-L1, inhibiting antitumor T-cell responses and avoiding immune surveillance and elimination. We have identified and characterized MEDI4736, a human IgG1 monoclonal antibody that binds with high affinity and specificity to PD-L1 and is uniquely engineered to prevent antibody-dependent cell-mediated cytotoxicity. In vitro assays demonstrate that MEDI4736 is a potent antagonist of PD-L1 function, blocking interaction with PD-1 and CD80 to overcome inhibition of primary human T-cell activation. In vivo MEDI4736 significantly inhibits the growth of human tumors in a novel xenograft model containing coimplanted human T cells. This activity is entirely dependent on the presence of transplanted T cells, supporting the immunological mechanism of action for MEDI4736. To further determine the utility of PD-L1 blockade, an anti-mouse PD-L1 antibody was investigated in immunocompetent mice. Here, anti-mouse PD-L1 significantly improved survival of mice implanted with CT26 colorectal cancer cells. The antitumor activity of anti-PD-L1 was enhanced by combination with oxaliplatin, which resulted in increased release of HMGB1 within CT26 tumors. Taken together, our results demonstrate that inhibition of PD-L1 function can have potent antitumor activity when used as monotherapy or in combination in preclinical models, and suggest it may be a promising therapeutic approach for the treatment of cancer. MEDI4736 is currently in several clinical trials both alone and in combination with other agents, including anti-CTLA-4, anti-PD-1, and inhibitors of IDO, MEK, BRAF, and EGFR.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , Binding, Competitive , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Melanoma/immunology , Melanoma/pathology , Melanoma/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Breast Cancer Res ; 17: 46, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25887862

ABSTRACT

INTRODUCTION: Podocalyxin (gene name PODXL) is a CD34-related sialomucin implicated in the regulation of cell adhesion, migration and polarity. Upregulated expression of podocalyxin is linked to poor patient survival in epithelial cancers. However, it is not known if podocalyxin has a functional role in tumor progression. METHODS: We silenced podocalyxin expression in the aggressive basal-like human (MDA-MB-231) and mouse (4T1) breast cancer cell lines and also overexpressed podocalyxin in the more benign human breast cancer cell line, MCF7. We evaluated how podocalyxin affects tumorsphere formation in vitro and compared the ability of podocalyxin-deficient and podocalyxin-replete cell lines to form tumors and metastasize using xenogenic or syngeneic transplant models in mice. Finally, in an effort to develop therapeutic treatments for systemic cancers, we generated a series of antihuman podocalyxin antibodies and screened these for their ability to inhibit tumor progression in xenografted mice. RESULTS: Although deletion of podocalyxin does not alter gross cell morphology and growth under standard (adherent) culture conditions, expression of PODXL is required for efficient formation of tumorspheres in vitro. Correspondingly, silencing podocalyxin resulted in attenuated primary tumor growth and invasiveness in mice and severely impaired the formation of distant metastases. Likewise, in competitive tumor engraftment assays where we injected a 50:50 mixture of control and shPODXL (short-hairpin RNA targeting PODXL)-expressing cells, we found that podocalyxin-deficient cells exhibited a striking decrease in the ability to form clonal tumors in the lung, liver and bone marrow. Finally, to validate podocalyxin as a viable target for immunotherapy, we screened a series of novel antihuman podocalyxin antibodies for their ability to inhibit tumor progression in vivo. One of these antibodies, PODOC1, potently blocked tumor growth and metastasis. CONCLUSIONS: We show that podocalyxin plays a key role in the formation of primary tumors and distant tumor metastasis. In addition, we validate podocalyxin as potential target for monoclonal antibody therapy to inhibit primary tumor growth and systemic dissemination.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Sialoglycoproteins/antagonists & inhibitors , Sialoglycoproteins/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Humans , Mammary Neoplasms, Animal , Mice , Neoplasm Metastasis , RNA Interference , RNA, Small Interfering/genetics , Sialoglycoproteins/genetics , Spheroids, Cellular , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Cancer Inform ; 13: 167-77, 2014.
Article in English | MEDLINE | ID: mdl-25506199

ABSTRACT

High-throughput transcriptome sequencing allows identification of cancer-related changes that occur at the stages of transcription, pre-messenger RNA (mRNA), and splicing. In the current study, we devised a pipeline to predict novel alternative splicing (AS) variants from high-throughput transcriptome sequencing data and applied it to large sets of tumor transcriptomes from The Cancer Genome Atlas (TCGA). We identified two novel tumor-associated splice variants of matriptase, a known cancer-associated gene, in the transcriptome data from epithelial-derived tumors but not normal tissue. Most notably, these variants were found in 69% of lung squamous cell carcinoma (LUSC) samples studied. We confirmed the expression of matriptase AS transcripts using quantitative reverse transcription PCR (qRT-PCR) in an orthogonal panel of tumor tissues and cell lines. Furthermore, flow cytometric analysis confirmed surface expression of matriptase splice variants in chinese hamster ovary (CHO) cells transiently transfected with cDNA encoding the novel transcripts. Our findings further implicate matriptase in contributing to oncogenic processes and suggest potential novel therapeutic uses for matriptase splice variants.

6.
Virology ; 361(1): 93-102, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17161858

ABSTRACT

Passive therapy with neutralizing human monoclonal antibodies (mAbs) could be an effective therapy against severe acute respiratory syndrome coronavirus (SARS-CoV). Utilizing the human immunoglobulin transgenic mouse, XenoMouse, we produced fully human SARS-CoV spike (S) protein specific antibodies. Antibodies were examined for reactivity against a recombinant S1 protein, to which 200 antibodies reacted. Twenty-seven antibodies neutralized 200TCID(50) SARS-CoV (Urbani). Additionally, 57 neutralizing antibodies were found that are likely specific to S2. Mapping of the binding region was achieved with several S1 recombinant proteins. Most S1 reactive neutralizing mAbs bound to the RBD, aa 318-510. However, two S1 specific mAbs reacted with a domain upstream of the RBD between aa 12 and 261. Immunoglobulin gene sequence analyses suggested at least 8 different binding specificities. Unique human mAbs could be used as a cocktail that would simultaneously target several neutralizing epitopes and prevent emergence of escape mutants.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Viral/biosynthesis , Antibodies, Viral/genetics , Antibody Specificity , Hemagglutinins, Viral/immunology , Humans , Hybridomas , Immunoglobulins/genetics , Immunoglobulins/metabolism , Membrane Glycoproteins/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Neutralization Tests , Sequence Alignment , Severe Acute Respiratory Syndrome , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/immunology
7.
J Immunol ; 174(8): 4768-78, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15814702

ABSTRACT

Most primates, including humans, are chronically infected with cospecifically evolved, potentially pathogenic CMV. Abs that bind a 10-aa linear epitope (antigenic determinant 2 site 1) within the extracellular domain of human CMV glycoprotein B neutralize viral infectivity. In this study, we show that genes generated by recombinations involving two well-conserved human germline V elements (IGHV3-30 and IGKV3-11), and IGHJ4, encode primary Ig molecules that bind glycoprotein B at this key epitope. These particular V(H), J(H), and V(kappa) genes enable humans to generate through recombination and N nucleotide addition, a useful frequency of primary Igs that efficiently target this critical site on human CMV and thus confer an innate foundation for a specific adaptive response to this pathogen.


Subject(s)
Antibodies, Viral/metabolism , Cytomegalovirus/immunology , Immunoglobulins/metabolism , Adaptation, Physiological , Amino Acid Sequence , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Antigens, Viral/genetics , Base Sequence , Codon/genetics , DNA, Complementary/genetics , Humans , Immunity, Innate , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulins/chemistry , Immunoglobulins/genetics , In Vitro Techniques , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...