Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 14(14): 4584-92, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18628473

ABSTRACT

PURPOSE: Invasion and metastasis of malignant epithelial cells into normal tissues is accompanied by adaptive changes in the mesenchyme-derived supporting stroma of the target organs. Altered gene expression in these nontransformed stromal cells provides potential targets for therapy. The present study was undertaken to determine the antitumor effects of an antibody-conjugate against fibroblast activation protein-alpha, a cell surface protease of activated tumor fibroblasts. EXPERIMENTAL DESIGN: A novel antibody-maytansinoid conjugate, monoclonal antibody (mAb) FAP5-DM1, was developed to target a shared epitope of human, mouse, and cynomolgus monkey fibroblast activation protein-alpha, enabling preclinical efficacy and tolerability assessments. We have used stroma-rich models in immunodeficient mice, which recapitulate the histotypic arrangement found in human epithelial cancers. RESULTS: Treatment with mAb FAP5-DM1 induced long-lasting inhibition of tumor growth and complete regressions in xenograft models of lung, pancreas, and head and neck cancers with no signs of intolerability. Analysis of chemically distinct conjugates, resistance models, and biomarkers implicates a unique mode of action, with mitotic arrest and apoptosis of malignant epithelial cells coupled to disruption of fibroblastic and vascular structures. CONCLUSIONS: We show that mAb FAP5-DM1 combines excellent efficacy and tolerability and provides a first assessment of the mode of action of a novel drug candidate for tumor stroma targeting, thus encouraging further development toward clinical testing of this treatment paradigm.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Fibroblasts/immunology , Immunoconjugates/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Serine Endopeptidases/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Endopeptidases , Gelatinases , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunohistochemistry , Macaca fascicularis , Maytansine/chemistry , Maytansine/immunology , Maytansine/therapeutic use , Membrane Proteins , Mice , Neoplasms/immunology , Surface Plasmon Resonance
2.
Cancer Res ; 68(12): 4774-82, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559524

ABSTRACT

Inhibition of tumor angiogenesis through blockade of the vascular endothelial growth factor (VEGF) signaling pathway is a novel treatment modality in oncology. Preclinical findings suggest that long-term clinical outcomes may improve with blockade of additional proangiogenic receptor tyrosine kinases: platelet-derived growth factor receptors (PDGFR) and fibroblast growth factor receptors (FGFR). BIBF 1120 is an indolinone derivative potently blocking VEGF receptor (VEGFR), PDGFR and FGFR kinase activity in enzymatic assays (IC(50), 20-100 nmol/L). BIBF 1120 inhibits mitogen-activated protein kinase and Akt signaling pathways in three cell types contributing to angiogenesis, endothelial cells, pericytes, and smooth muscle cells, resulting in inhibition of cell proliferation (EC(50), 10-80 nmol/L) and apoptosis. In all tumor models tested thus far, including human tumor xenografts growing in nude mice and a syngeneic rat tumor model, BIBF 1120 is highly active at well-tolerated doses (25-100 mg/kg daily p.o.), as measured by magnetic resonance imaging of tumor perfusion after 3 days, reducing vessel density and vessel integrity after 5 days, and inducing profound growth inhibition. A distinct pharmacodynamic feature of BIBF 1120 in cell culture is sustained pathway inhibition (up to 32 hours after 1-hour treatment), suggesting slow receptor off-kinetics. Although BIBF 1120 is rapidly metabolized in vivo by methylester cleavage, resulting in a short mean residence time, once daily oral dosing is fully efficacious in xenograft models. These distinctive pharmacokinetic and pharmacodynamic properties may help explain clinical observations with BIBF 1120, currently entering phase III clinical development.


Subject(s)
Blood Coagulation Factors/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Indoles/therapeutic use , Neoplasms, Experimental/drug therapy , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Indoles/chemical synthesis , Indoles/pharmacokinetics , Mice , Mice, Nude , Molecular Structure , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pericytes/cytology , Pericytes/drug effects , Pericytes/metabolism , Rats , Rats, Inbred F344 , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Tissue Distribution , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/immunology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
3.
Curr Biol ; 17(4): 316-22, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17291758

ABSTRACT

Fine-mapping of the cell-division cycle, notably the identification of mitotic kinase signaling pathways, provides novel opportunities for cancer-drug discovery. As a key regulator of multiple steps during mitotic progression across eukaryotic species, the serine/threonine-specific Polo-like kinase 1 (Plk1) is highly expressed in malignant cells and serves as a negative prognostic marker in specific human cancer types . Here, we report the discovery of a potent small-molecule inhibitor of mammalian Plk1, BI 2536, which inhibits Plk1 enzyme activity at low nanomolar concentrations. The compound potently causes a mitotic arrest and induces apoptosis in human cancer cell lines of diverse tissue origin and oncogenome signature. BI 2536 inhibits growth of human tumor xenografts in nude mice and induces regression of large tumors with well-tolerated intravenous dose regimens. In treated tumors, cells arrest in prometaphase, accumulate phosphohistone H3, and contain aberrant mitotic spindles. This mitotic arrest is followed by a surge in apoptosis, detectable by immunohistochemistry and noninvasive optical and magnetic resonance imaging. For addressing the therapeutic potential of Plk1 inhibition, BI 2536 has progressed into clinical studies in patients with locally advanced or metastatic cancers.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle/physiology , Enzyme Inhibitors/pharmacology , Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Signal Transduction/physiology , Animals , Body Weight , Cell Cycle Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Female , Flow Cytometry , HeLa Cells , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Microscopy, Fluorescence , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pteridines/metabolism , Spectrometry, Fluorescence , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
4.
Curr Biol ; 17(4): 304-15, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17291761

ABSTRACT

BACKGROUND: The mitotic kinases, Cdk1, Aurora A/B, and Polo-like kinase 1 (Plk1) have been characterized extensively to further understanding of mitotic mechanisms and as potential targets for cancer therapy. Cdk1 and Aurora kinase studies have been facilitated by small-molecule inhibitors, but few if any potent Plk1 inhibitors have been identified. RESULTS: We describe the cellular effects of a novel compound, BI 2536, a potent and selective inhibitor of Plk1. The fact that BI 2536 blocks Plk1 activity fully and instantaneously enabled us to study controversial and unknown functions of Plk1. Cells treated with BI 2536 are delayed in prophase but eventually import Cdk1-cyclin B into the nucleus, enter prometaphase, and degrade cyclin A, although BI 2536 prevents degradation of the APC/C inhibitor Emi1. BI 2536-treated cells lack prophase microtubule asters and thus polymerize mitotic microtubules only after nuclear-envelope breakdown and form monopolar spindles that do not stably attach to kinetochores. Mad2 accumulates at kinetochores, and cells arrest with an activated spindle-assembly checkpoint. BI 2536 prevents Plk1's enrichment at kinetochores and centrosomes, and when added to metaphase cells, it induces detachment of microtubules from kinetochores and leads to spindle collapse. CONCLUSIONS: Our results suggest that Plk1's accumulation at centrosomes and kinetochores depends on its own activity and that this activity is required for maintaining centrosome and kinetochore function. Our data also show that Plk1 is not required for prophase entry, but delays transition to prometaphase, and that Emi1 destruction in prometaphase is not essential for APC/C-mediated cyclin A degradation.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Enzyme Inhibitors/pharmacology , F-Box Proteins/metabolism , Microtubules/drug effects , Mitosis/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Spindle Apparatus/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Image Processing, Computer-Assisted , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pteridines/metabolism , Spindle Apparatus/metabolism , Polo-Like Kinase 1
5.
Cancer Immun ; 6: 10, 2006 Jul 31.
Article in English | MEDLINE | ID: mdl-16875435

ABSTRACT

Endosialin is a C-type lectin-like cell surface receptor of unknown function, with a distinctive pattern of endothelial expression in newly formed blood vessels in human cancers. The murine orthologue of endosialin has been identified, opening up the analysis of developmental regulation in the embryo and in aberrant tissue remodeling, notably cancer angiogenesis. To advance these studies we have generated an antibody to the extracellular domain of mouse endosialin and mapped protein expression from embryonic day E10.0 to the adult stage, complemented by mRNA quantification and co-typing for standard endothelial markers. Four main findings emerged. First, endosialin protein is restricted to vascular endothelium and fibroblast-like cells in developing organs, and largely disappears in the adult. Second, endothelial expression varies markedly between organs regarding spatial and temporal patterns. For instance, in the E10.0 embryo, endosialin is prominent in the endothelium of the dorsal aorta and, from E11.0 to E14.5, in vessels sprouting from the dorsal aorta, in perineural vascular plexuses, and in brain capillaries. Third, circumscribed mesenchymal expression in fibroblast-like cells was evident throughout development, most pronounced adjacent to certain budding epithelia, as exemplified by the lung and kidney glomeruli, but unrelated to the endothelial expression. The endosialin protein persists in the stromal fibroblasts of the adult uterus. Finally, in subcutaneous cancer xenograft models endosialin re-appears in the host-derived tumor stroma, both in neo-angiogenic vascular endothelium and in activated stromal fibroblasts. In future studies, the search for intrinsic or extrinsic signals contributing to endosialin induction in cancer stroma will be of interest.


Subject(s)
Antigens, CD/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic , Animals , Animals, Newborn , Antibodies/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , Embryonic Development , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Tumor Cells, Cultured
6.
Diagn Mol Pathol ; 15(1): 35-42, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16531767

ABSTRACT

Transcriptional profiling of cancer biopsies is used extensively to identify expression signatures for specific cancer types, diagnostic and prognostic subgroups, and novel molecular targets for therapy. To broaden these applications, several challenges remain. For example, the integrity of RNA extracted even from small tissue samples has to be insured and monitored. Moreover, total tumor RNA may hide the marked histologic heterogeneity of human cancers. A principle approach to this heterogeneity has been provided by laser capture microdissection performed on antibody-stained tissue sections (immuno-LCM; iLCM). In this study, we have established a procedure to assess the quality of RNA obtained from tissue sections, coupled with immunostaining using antibodies to different tumor stromal markers, and subsequent iLCM to selectively capture the cancer stroma compartments. The procedure was applied to 53 frozen specimens of human epithelial cancers. Sections were stained for histopathological evaluation, and RNA was isolated from adjacent serial sections. RNA quality was assessed by the Agilent-Bioanalyzer (Agilent, Palo Alto, CA) and by multiplex RT-PCR. Two thirds of the specimens were found to yield good to excellent RNA quality. For microdissection of the tumor stroma with reactive fibroblasts and tumor blood vessels, a rapid incubation protocol with antibodies against fibroblast activation protein (FAP) and against endosialin was developed to ensure RNA integrity for subsequent iLCM. Using these procedures, RNA from distinct tumor compartments can be isolated, analyzed, amplified, and used for transcription profiling.


Subject(s)
Carcinoma/diagnosis , Gelatinases/analysis , Membrane Proteins/analysis , Microdissection/methods , Neoplasm Proteins/analysis , RNA, Neoplasm/isolation & purification , Serine Endopeptidases/analysis , Antibodies, Neoplasm/immunology , Antigens, CD , Antigens, Neoplasm , Carcinoma/pathology , Endopeptidases , Frozen Sections , Gelatinases/immunology , Humans , Immunohistochemistry , Lasers , Membrane Proteins/immunology , Neoplasm Proteins/immunology , RNA, Neoplasm/analysis , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/immunology
7.
Cancer Immun ; 5: 10, 2005 Aug 03.
Article in English | MEDLINE | ID: mdl-16076089

ABSTRACT

Standardized, high-throughput RNA detection with microarray chips allows for the construction of genome-wide databases for tissue specimens suitable for in silico electronic Northern blot (eNorthern) analysis of marker genes. We used the BioExpress database, which contains transcriptional profiles of normal and cancer samples, to examine two putative markers of cancer stroma: fibroblast activation protein-alpha (FAP-alpha) and endosialin. Analyses for FAP-alpha showed that normal tissues generally lack RNA signals, with the exception of endometrium. Typing of tumors revealed prominent FAP-alpha signals in cancer types marked by desmoplasia, and localization of FAP-alpha in reactive cancer stroma was confirmed by immunohistochemistry. A subset of sarcomas displayed prominent FAP-alpha signals localizing to the malignant cells. For endosialin, eNorthern analyses showed low to moderate RNA signals in many normal organs, whereas immunohistochemistry revealed endosialin in only some tissues, such as endometrium. Endosialin was detected at the RNA and protein level in sarcomas, notably malignant fibrous histiocytomas. Low to moderate endosialin RNA signals were found in epithelial cancer types for which immunostaining identifies expression in subsets of tumor capillaries or fibroblasts. These findings extend the FAP-alpha and endosialin profiling in silico to an unbiased tumor database and place both molecules in a novel context of endometrial biology and sarcoma subtyping. Our findings suggest that BioExpress can be searched directly for tumor stroma markers but may need prior enrichment for markers with narrow cellular representation, such as endosialin. Constructing databases from microdissected cancer tissues may be an essential step for tumor stroma-targeted therapies.


Subject(s)
Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Databases, Genetic , Membrane Proteins/analysis , Neoplasm Proteins/analysis , Serine Endopeptidases/analysis , Adult , Antigens, CD , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Endopeptidases , Female , Gelatinases , Gene Expression Profiling , Gene Expression Regulation/genetics , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/analysis , Serine Endopeptidases/genetics , Up-Regulation
8.
Oncogene ; 23(32): 5435-46, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15133496

ABSTRACT

Fibroblast activation protein-alpha (FAP) is a cell surface serine protease expressed at sites of tissue remodeling in embryonic development. FAP is not expressed by mature somatic tissues except activated melanocytes and fibroblasts in wound healing or tumor stroma. FAP expression is specifically silenced in proliferating melanocytic cells during malignant transformation. To study the role of FAP as a tumor suppressor, the gene for mouse fap was cloned and mutated at the catalytic domain (FAP serine mutant, FSM). We found that expression of FAP or FSM at physiologic levels in mouse melanoma cells abrogated tumorigenicity. Remarkably, the mutant form FSM lacking specific serine protease activity was a more potent tumor suppressor. Tumor rejection was not due to adaptive immune responses because RAG1-/- mice challenged with melanoma cells expressing either FAP or FSM were not tumorigenic. In in vitro assays, FAP or FSM expression restored contact inhibition, led to cell cycle arrest at G0/G1 phase, and increased susceptibility to stress-induced apoptosis. Cell death in FAP+ or FSM+ melanoma cells was readily triggered by depletion of survival factors from the media, leading to subsequent activation of caspases via the intrinsic pathway. These results show that expression of FAP is a tumor suppressor that abrogates tumorigenicity through regulation of cell growth and survival.


Subject(s)
Gelatinases/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Contact Inhibition/genetics , Contact Inhibition/physiology , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Endopeptidases , Gelatinases/genetics , Membrane Proteins/genetics , Mice , Mutation , Serine/genetics , Serine/metabolism , Serine Endopeptidases/genetics , Transfection , Wound Healing/genetics
9.
Mol Cancer Ther ; 2(8): 729-37, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12939462

ABSTRACT

Antigens and receptors that are highly expressed on tumor stromal cells, such as fibroblast activation protein (FAP), are attractive targets for antibody-based therapies because the supporting stroma and vessel network is essential for a solid neoplasm to grow beyond a size of 1-2 mm. The in vivo characterization of antibodies targeting human stromal or vessel antigens is hindered by the lack of an appropriate mouse model system because xenografts in standard mouse models express stromal and vessels elements of murine origin. This limitation may be overcome by the development of a human skin/mouse chimeric model, which is established by transplanting human foreskin on to the lateral flank of severe combined immunodeficient mice. The subsequent inoculation of breast carcinoma MCF-7 cells within the dermis of the transplanted human skin resulted in the production of xenografts expressing stromal and vessel elements of human origin. Widespread expression of human FAP-positive reactive stromal fibroblasts within xenografts was seen up to 2 months posttransplantation and postinjection of cells. Human blood vessel antigen expression also persisted at 2 months posttransplantation and postinjection of cells with murine vessels coexisting with the human vascular supply. The model was subsequently used to evaluate the biodistribution properties of an iodine-131-labeled humanized anti-FAP monoclonal antibody (BIBH-7). The results showed high specific targeting of the stromal compartment of the xenograft, indicating that the model provides a useful and novel approach for the in vivo assessment of the immunotherapeutic potential of molecules targeting human stroma and angiogenic systems.


Subject(s)
Antigens, Neoplasm/analysis , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/analysis , Mice, SCID , Serine Endopeptidases/analysis , Animals , Antibodies, Monoclonal , Breast Neoplasms , Cell Line, Tumor , Endopeptidases , Gelatinases , Gene Targeting , H-2 Antigens/metabolism , HLA-A Antigens/analysis , HLA-B Antigens/analysis , HLA-C Antigens/analysis , HLA-C Antigens/metabolism , Humans , Membrane Proteins , Mice , Models, Animal , Neoplasm Transplantation , Skin/chemistry , Skin Transplantation , Stromal Cells/enzymology , Transplantation, Heterologous
10.
Clin Cancer Res ; 9(5): 1639-47, 2003 May.
Article in English | MEDLINE | ID: mdl-12738716

ABSTRACT

PURPOSE: The purpose of this research was to determine the safety, immunogenicity, pharmacokinetics, biodistribution, and tumor uptake of repeat infusions of a complementarity-determining region grafted humanized antibody (sibrotuzumab) directed against human fibroblast activation protein (FAP). EXPERIMENTAL DESIGN: A Phase I open-label dose escalation study was conducted in patients with cancers epidemiologically known to be FAP positive. Patients were entered into one of four dosage tiers of 5, 10, 25, or 50 mg/m(2) sibrotuzumab, administered weekly for 12 weeks, with trace labeling with 8-10 mCi of (131)I in weeks 1, 5, and 9. RESULTS: A total of 26 patients were entered into the trial (15 males and 11 females; mean age, 59.9 years; age range, 41-81 years). Twenty patients had colorectal carcinoma, and 6 patients had non-small cell lung cancer. A total of 218 infusions of sibrotuzumab were administered during the first 12 weeks of the study, with 24 patients being evaluable. One patient received an additional 96 infusions on continued-use phase for a total of 108 infusions over a 2-year period, and 1 patient received an additional 6 infusions on continued use. There were no objective tumor responses. Only one episode of dose-limiting toxicity was observed. Therefore, a maximum tolerated dose was not reached. Treatment-related adverse events were observed in 6 patients during the infusional monitoring period. Four of the 6 patients, 3 of whom had associated positive serum human antihuman antibody, were removed from the study because of clinical immune responses. Gamma camera images of [(131)I]sibrotuzumab demonstrated no normal organ uptake of sibrotuzumab, with tumor uptake evident within 24-48 h after infusion. Analysis of pharmacokinetics demonstrated a similar mean terminal t(1/2) of 1.4-2.6 days at the 5, 10, and 25 mg/m(2) dose levels, and with a longer mean t(1/2) of 4.9 days at the 50 mg/m(2) dose level. CONCLUSION: Repeat infusions of the humanized anti-FAP antibody sibrotuzumab can be administered safely to patients with advanced FAP-positive cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Serine Endopeptidases/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/secondary , Colorectal Neoplasms/blood , Colorectal Neoplasms/secondary , Dose-Response Relationship, Drug , Endopeptidases , Female , Follow-Up Studies , Gelatinases , Humans , Infusions, Intravenous , Iodine Radioisotopes , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Maximum Tolerated Dose , Membrane Proteins , Middle Aged , Radioimmunotherapy , Serine Endopeptidases/immunology , Treatment Outcome
11.
J Invest Dermatol ; 120(2): 182-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12542520

ABSTRACT

Growth and metastasis of solid neoplasms require the recruitment of a supporting tumor stroma. A highly consistent trait of tumor stromal fibroblasts in most epithelial cancers is the induction of fibroblast activation protein (FAP), a member of the serine protease family. Recently it was demonstrated that FAP has both dipeptidyl peptidase and collagenolytic activity capable of degrading gelatin and type I collagen. In this study, we describe the expression and enzyme activity of FAP in benign and malignant melanocytic skin tumors. FAP-positive fibroblasts were detected immunohistochemically in the reactive stroma of all melanocytic nevi tested. In primary and metastatic melanomas an upregulation of FAP expression in the reactive mesenchyme could be observed. Whereas 30% of the nevi revealed additional FAP expression on subsets of melanocytic cells, melanoma cells from primary and metastatic melanomas were FAP negative. This may indicate a possible role for FAP in the control of tumor cell growth and proliferation during melanoma carcinogenesis. Consistent with this in vivo expression pattern FAP enzyme activity could be detected by a specific immunocapture assay in extracts of melanocytic nevi and melanoma metastases, whereas no significant activity was detectable in normal adult skin. Strong protein expression of FAP was observed in patterned structures restricted to a subset of the melanoma metastases. Our findings that these FAP-positive structures showed no overlap with endothelial cell surface markers, nor with various melanoma antigens, suggest that FAP is a marker for specific stromal-cell-derived patterns in cutaneous melanoma metastases.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Nevus, Pigmented/metabolism , Serine Endopeptidases/metabolism , Skin Neoplasms/metabolism , Stromal Cells/enzymology , Antigens, Neoplasm/biosynthesis , Biomarkers, Tumor/biosynthesis , Endopeptidases , Fibroblasts/enzymology , Gelatinases , Humans , Melanoma/metabolism , Melanoma/secondary , Membrane Proteins , Neoplasms/metabolism , Neoplasms/pathology , Nevus, Pigmented/pathology , Serine Endopeptidases/biosynthesis , Skin Neoplasms/pathology
12.
J Biol Chem ; 277(18): 15566-72, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11854300

ABSTRACT

Telomerase, a ribonucleoprotein acting as a reverse transcriptase, has been identified as a target for cancer drug discovery. The synthetic, non-nucleosidic compound, BIBR1532, is a potent and selective telomerase inhibitor capable of inducing senescence in human cancer cells (). In the present study, the mode of drug action was characterized. BIBR1532 inhibits the native and recombinant human telomerase, comprising the human telomerase reverse transcriptase and human telomerase RNA components, with similar potency primarily by interfering with the processivity of the enzyme. Enzyme-kinetic experiments show that BIBR1532 is a mixed-type non-competitive inhibitor and suggest a drug binding site distinct from the sites for deoxyribonucleotides and the DNA primer, respectively. Thus, BIBR1532 defines a novel class of telomerase inhibitor with mechanistic similarities to non-nucleosidic inhibitors of HIV1 reverse transcriptase.


Subject(s)
Aminobenzoates , Enzyme Inhibitors/pharmacology , Naphthalenes , Telomerase/antagonists & inhibitors , Binding Sites , DNA Primers , Humans , Kinetics , RNA-Directed DNA Polymerase/metabolism , Recombinant Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...