Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Br J Cancer ; 95(1): 49-55, 2006 Jul 03.
Article in English | MEDLINE | ID: mdl-16755292

ABSTRACT

Multiple defects in apoptotic pathways have been described in peripheral neuroblastic tumours (NTs). Mitosis-karyorrhexis index (MKI) is a reliable morphological marker identifying favourable and unfavourable NTs. The extent to which apoptotic processes contribute to determine the clinical significance of MKI is still undefined. Apoptosis was investigated in a series of 110 peripheral NTs by comparing MKI to immunohistochemical and molecular apoptotic features. High MKI was found in 55 out of 110 NTs (50%) and was associated with advanced stage (P = 0.007), neuroblastoma (NB) histological category (P = 0.024), MYCN amplification (P < 0.001), and poor outcome (P = 0.011). Overall survival probability was 45% in patients with high MKI compared to 73% in patients with low MKI. In the same 110 NTs, the expression of Bcl-2, Bcl-XL, Bax and Mcl-1 was studied by immunohistochemistry, but no significant associations were found with clinicohistological features. Microarray analysis of apoptotic genes was performed in 40 out of 110 representative tumours. No significant association was found between the expression of apoptotic genes and MKI or clinicohistological features. Proliferative activity was assessed in 60 out of 110 representative tumours using Ki67 immunostaining, but no significant correlations with MKI or clinicobiological features were found. In NTs, the combination of apoptosis and proliferation as expressed by MKI is a significant prognostic parameter, although neither of them is per se indicative of the clinicobiological behaviour and outcome.


Subject(s)
Apoptosis , Neuroblastoma/diagnosis , Neuroblastoma/metabolism , Peripheral Nervous System Neoplasms/diagnosis , Peripheral Nervous System Neoplasms/metabolism , Adolescent , Biomarkers, Tumor/biosynthesis , Cell Proliferation , Child , Child, Preschool , Female , Gene Expression Profiling , Humans , Infant , Infant, Newborn , Male , Mitotic Index , Neuroblastoma/genetics , Oligonucleotide Array Sequence Analysis , Peripheral Nervous System Neoplasms/genetics , Predictive Value of Tests , Prognosis , Survival Analysis
2.
Allergol Immunopathol (Madr) ; 31(5): 259-64, 2003.
Article in English | MEDLINE | ID: mdl-14572414

ABSTRACT

Although sublingual allergen-specific immunotherapy has been proved to be effective in the treatment of allergic diseases, controversy surrounds the means by which such a local therapy can induce systemic immunological changes. Adhesion molecules are critical in the regulation of leukocyte traffic. It has been hypothesized that allergenic extract, administered locally, may induce an up-regulation of the mucosal vessel vascular adhesion molecules (CAMs) resulting in local recruitment of circulating inflammatory cells. In the present study we investigated whether the mite antigens, Der p1 and Der p2, can modulate CAM expression of human endothelial cells (HEC). To do this, slices of whole human umbilical cord vein underwent short-term (8 hours) cultures in the presence or absence of mite antigen (baseline, unstimulated controls). Cryostatic sections of the specimens were then evaluated immunohistochemically for expression of intercellular adhesion molecule (ICAM-1) and vascular cell adhesion molecule (VCAM-1) molecules. The results revealed that while Der p1 is capable of significantly up-regulating ICAM-1 and VCAM-1 on HEC, Der p2 antigen moderately up-regulates ICAM-1 expression but is ineffective in modulating VCAM-1. Although preliminary, these results clearly support the hypothesis that at least some of the effects of sublingual immunotherapy may derive from inflammatory cell recruitment at the site of allergen release.


Subject(s)
Antigens, Dermatophagoides/immunology , Desensitization, Immunologic , Endothelial Cells/immunology , Endothelium, Vascular/immunology , Gene Expression Regulation/immunology , Intercellular Adhesion Molecule-1/biosynthesis , Mites/immunology , Vascular Cell Adhesion Molecule-1/biosynthesis , Administration, Sublingual , Animals , Arthropod Proteins , Cysteine Endopeptidases , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Organ Culture Techniques , Umbilical Veins , Vascular Cell Adhesion Molecule-1/genetics , Vasculitis/etiology
3.
Gynecol Oncol ; 85(2): 305-10, 2002 May.
Article in English | MEDLINE | ID: mdl-11972392

ABSTRACT

OBJECTIVES: Cyclooxygenase-2 (COX-2) seems to be involved in critical steps of cancer onset and progression. Abnormalities of epidermal growth factor receptor (EGFR) and Her-2/neu have been actively investigated in ovarian cancer and associated with unfavorable clinical outcome. The involvement of COX-2 in ErbB family pathways has been proposed. We investigated by immunohistochemistry the expression of COX-2, EGFR, and Her-2/neu in a series of advanced primary ovarian cancers. METHODS: The study included 76 consecutive stage IIIC-IV ovarian cancer patients with measurable disease after first surgery. Immunohistochemistry was performed on paraffin-embedded sections with rabbit antiserum against COX-2, murine monoclonal antibody (MoAb) 300G9 against Her-2/neu, and monoclonal antibody 108 against EGFR. RESULTS: No association among COX-2, EGFR, and HER-2/neu was found. COX-2 positivity was found in a statistically significant higher percentage of unresponsive cases (80.0%) than in patients responding to chemotherapy (35.7%) (P = 0.0008). The association between COX-2 positivity and poor chance of response to treatment was retained in multivariate analysis. In the subgroup of patients who underwent explorative laparotomy COX-2-positive cases showed a shorter overall survival (P = 0.049). CONCLUSIONS: COX-2 could represent a possible new marker of sensitivity to platin-based chemotherapy in ovarian cancer. The lack of association of COX-2 with EGFR or Her-2/neu suggests that the ability of COX-2 to predict tumor sensitivity to chemotherapy is not dependent on EGFR or Her-2/neu status and could be independently associated with prognosis. In this context, the availability of agents able to specifically interfere with COX-2, Her-2/neu, or EGFR tyrosine kinase is of potential interest.


Subject(s)
ErbB Receptors/biosynthesis , Isoenzymes/biosynthesis , Ovarian Neoplasms/metabolism , Prostaglandin-Endoperoxide Synthases/biosynthesis , Receptor, ErbB-2/biosynthesis , Cyclooxygenase 2 , Female , Humans , Immunohistochemistry , Membrane Proteins , Neoplasm Staging , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology
4.
J Cell Physiol ; 188(1): 1-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11382917

ABSTRACT

Endoglin (CD105) is a cell membrane glycoprotein over-expressed on highly proliferating endothelial cells in culture, and on endothelial cells of angiogenetic blood vessels within benign and malignant tissues. CD105 binds several factors of the Transforming Growth Factor (TGF)-beta superfamily, and its over-expression modulates cellular responses to TGF-beta1. The complex of experimental findings accumulated in the last few years strongly indicate that CD105 is a powerful marker of angiogenesis, and that it might play a critical role in the pathogenesis of vascular diseases and in tumor progression. In this paper, we will review the structural, biological and functional features of CD105, as well as its distribution within normal and neoplastic tissues, emphasizing its foreseeable role as a molecular target for new diagnostic and bioimmunotherapeutic approaches in human malignancies.


Subject(s)
Neoplasms/metabolism , Neovascularization, Pathologic/physiopathology , Transforming Growth Factor beta/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Antigens, CD , Biomarkers, Tumor/metabolism , Endoglin , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Macromolecular Substances , Neoplasms/blood supply , Neoplasms/pathology , Neoplasms/therapy , Neovascularization, Physiologic/physiology , Receptors, Cell Surface , Transforming Growth Factor beta/genetics , Vascular Cell Adhesion Molecule-1/chemistry , Vascular Cell Adhesion Molecule-1/genetics
5.
Am J Pathol ; 158(3): 841-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238033

ABSTRACT

Endothelin-1 (ET-1) has been shown to be mitogenic for endothelial and several tumor cells through an autocrine mechanism. In this study we evaluated whether the tumorigenic KS IMM cell line deriving from Kaposi's sarcoma (KS), a highly angiogenic tumor, is susceptible to ET-1 mitogenic activity. By reverse transcriptase-polymerase chain reaction, we detected ET-1 mRNA expression and both ET(A) receptor (ET(A)R) and ET(B)R mRNA transcripts in the KS IMM cells. High concentrations of ET-1 are released from the KS IMM cells and competition-binding studies demonstrated that these cells also express functional ET(A)R and ET(B)R with high affinity for ET-1 and ET-1/ET-3, respectively. Expression of ET-1 and cognate receptors could be detected by immunohistochemical method in vitro, in KS IMM xenograft, and in tissue sections of a human KS lesion. Furthermore ET-1 induces a marked and dose-dependent increase in [3H]thymidine incorporation comparable to that elicited by vascular endothelial growth factor. Addition of both selective ET(B)R antagonist (BQ 788) and ET(A)R antagonist (BQ 123), completely blocked ET-1-induced mitogenic response and reduced the basal growth rate of unstimulated cells, suggesting that both receptors mediated the proliferative signal. Such findings demonstrate that ET-1 participates on KS pathogenesis acting as an autocrine growth factor and that ET-1 receptor antagonists may thus be novel candidates for therapeutic intervention.


Subject(s)
Endothelin Receptor Antagonists , Sarcoma, Kaposi/etiology , Animals , Autocrine Communication , Cell Division/drug effects , Cells, Cultured , Endothelin-1/biosynthesis , Endothelin-1/genetics , Endothelin-1/pharmacology , Humans , Mice , Mice, Nude , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Piperidines/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Receptors, Endothelin/biosynthesis , Receptors, Endothelin/genetics , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Transcription, Genetic , Tumor Cells, Cultured
6.
Am J Pathol ; 157(5): 1537-47, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073813

ABSTRACT

Endothelin-1 (ET-1) is overexpressed in ovarian carcinomas and acts, via ET(A) receptors (ET(A)R), as an autocrine growth factor. In this study we investigate the role of ET-1 in the neovascularization of ovarian carcinoma. Archival specimens of primary (n = 40) and metastatic (n = 8) ovarian tumors were examined by immunohistochemistry for angiogenic factor and receptor expression and for microvessel density using antibodies against CD31, ET-1, vascular endothelial growth factor (VEGF), and their receptors. ET-1 expression correlated with neovascularization and with VEGF expression. The localization of functional ET(A)R and ET(A)R mRNA expression, as detected by autoradiography and in situ hybridization, was evident in tumors and in intratumoral vessels, whereas ET(B)R were expressed mainly in endothelial cells. High levels of ET-1 were detected in the majority of ascitic fluids of patients with ovarian carcinoma and significantly correlated with VEGF ascitic concentration. Furthermore ET-1, through ET(A)R, stimulated VEGF production in an ovarian carcinoma cell line, OVCA 433, by an extent comparable to hypoxia. Finally, conditioned media from OVCA 433 as well as ascitic fluids caused an increase in endothelial cell migration and the ET-1 receptor blockade significantly inhibited this angiogenic response. These findings indicate that ET-1 could modulate tumor angiogenesis, acting directly and in part through VEGF.


Subject(s)
Adenocarcinoma/blood supply , Carcinoma/blood supply , Endothelin-1/physiology , Neovascularization, Pathologic/physiopathology , Ovarian Neoplasms/blood supply , Adenocarcinoma/metabolism , Adult , Aged , Ascitic Fluid/metabolism , Blood Vessels/pathology , Carcinoma/metabolism , Cell Movement/physiology , Endothelial Growth Factors/metabolism , Endothelin-1/pharmacology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Humans , Lymphokines/metabolism , Middle Aged , Ovarian Neoplasms/metabolism , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Endothelin/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Tissue Antigens ; 56(1): 30-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10958353

ABSTRACT

HLA-G is an effective ligand of natural killer (NK) inhibitory receptors, HLA-G transcripts have been detected in several human tumors, and cytokines like gamma interferon (IFN) enable HLA-G molecules to be expressed. These findings are particularly upsetting in case of melanomas: IFN treatment is frequently included in melanoma therapeutic protocols, and downregulation of classical class I molecules occurs in nearly half of these tumors. Therefore, a melanoma cell downregulating classical class I and de novo expressing HLA-G, either constitutively or upon IFN treatment, is probably a stealthy target for the immune system, having inhibited both the cytotoxic T lymphocyte (CTL) and the NK activity. To elucidate this point we have investigated the expression of HLA-G molecules in 45 melanoma cell lines before and after gammaIFN treatment. Analysis was performed by immunofluorescence and flow cytometry, using the anti-HLA-G MoAbs 87G and G233, by Western blot, using the anti-HLA-G MEM/G1 MoAb and PAG1 antiserum, and by RT-PCR analysis. In addition, 8 melanoma tissues from patients free from therapy and 6 nevi were studied by immunohistochemistry using the 87G MoAb. No evidence was gathered of HLA-G expression, neither constitutive nor, in cell lines, after gammaIFN treatment. We therefore conclude that HLA-G expression is an uncommon event in melanomas, and that a therapy including IFNs cannot harm the patient by inducing the de novo expression of HLA-G molecules at least in its G1 isoform.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Interferon-gamma/pharmacology , Melanoma/immunology , Antibodies, Monoclonal/immunology , Blotting, Western , Flow Cytometry , HLA Antigens/genetics , HLA Antigens/immunology , HLA-G Antigens , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , RNA, Messenger/analysis , Tumor Cells, Cultured
8.
Clin Cancer Res ; 6(5): 2037-43, 2000 May.
Article in English | MEDLINE | ID: mdl-10815930

ABSTRACT

Increasing evidence suggests that endoglin (CD105) is a new powerful marker of neovascularization in solid malignancies; thus, using breast cancer as a model, we investigated whether targeting of CD105 by monoclonal antibody (mAb) MAEND3 can be used for in vivo imaging of solid tumors. Immunohistochemistry and flow cytometry identified differential expression of CD105 on breast cancer and endothelial cells; in fact, neoplastic cells were weakly and rarely stained by mAb MAEND3, which in contrast, strongly and invariably stained blood vessel endothelia within the breast adenocarcinomas investigated and cultured endothelial cells. Moreover, in contrast to CD31, which currently represents the reference marker to assess angiogenetic activity, CD105 expression was highest in semiconfluent and actively proliferating endothelial cells, and it progressively decreased as cells reached tight confluency and low [3H]thymidine uptake. i.v. administration of 18 MBq of 125I-labeled mAb MAEND3 efficiently imaged spontaneous mammary adenocarcinomas in two dogs; the uptake of radiolabeled mAb was rapid and intense because tumor: background ratios of 8.2:1 and 9.3:1 were reached 8 h after mAb administration, in the absence of immediate and/or long-term clinical side effects. Altogether, our present data suggest that targeting of CD105 on tumor-associated blood vessels may represent a new strategy for in vivo imaging of solid malignancies, regardless of their histological origin.


Subject(s)
Neoplasms/diagnostic imaging , Vascular Cell Adhesion Molecule-1/analysis , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Animals , Antibodies, Monoclonal/immunology , Antigens, CD , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Count , Cell Division , Cell Line , Disease Models, Animal , Dogs , Endoglin , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Gamma Cameras , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Iodine Radioisotopes , Mammary Neoplasms, Animal/diagnostic imaging , Mammary Neoplasms, Animal/metabolism , Neoplasms/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/analysis , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Radionuclide Imaging , Receptors, Cell Surface , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
9.
Cancer Res ; 59(11): 2657-67, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10363989

ABSTRACT

The down-regulation of human leukocyte antigen (HLA) class I molecules, especially the selective down-regulation of certain allelic products, is believed to represent a major mechanism of tumor escape from immune surveillance. In the present report, an original approach is described to precisely evaluate and classify HLA class I epitope losses in 30 cancer patients with malignant melanoma and lung, breast, endometrium, ovary, and colon carcinoma tumors. Early-passage tumor cell lines were established in culture from the corresponding metastatic tumor lesions obtained in each patient. Both the cell lines and the tumor lesions were compared, in their HLA-A and -B expression, to the peripheral blood mononuclear cells (PBMCs) obtained from the same patient (autologous PBMCs). On the basis of HLA-genotyping data, the appropriate monoclonal antibodies identifying mono- and poly-morphic HLA-A and HLA-B epitopes were selected from a panel of 34 antibodies for a total of 24 testable alleles. The selected antibodies were used not only in immunohistochemical assays on cryostatic tumor sections and cytospins of PBMCs but also in quantitative, sensitive flow cytometry assays on early-passage tumor cells and PBMC suspensions. With this latter method, a low overall HLA expression was detected in 26 tumor cell explants and a complete, generalized HLA-A, HLA-B, HLA-C loss in the remaining 4 cases. However, no complete, selective loss of any of the 45 tested HLA-A and HLA-B allomorphs was observed. Sequences from all of the HLA class I alleles could be detected at the genomic DNA level in tumor cells and tissues. At variance from the literature and the results of immunohistochemical experiments performed in parallel on the corresponding tumor lesions, the relative proportions of the various HLA epitopes were relatively preserved in each early-passage cell line/PBMC pair, and selective increases, rather than decreases, in the expression of polymorphic HLA epitopes had the highest prevalence and greatest magnitude. Our data suggest an alternative tumor stealth strategy in which up- and down-regulation are equally important. This alternative model of tumor-host interaction better fits the available models of tumor cell recognition by CTLs and natural killer cells bearing activatory and inhibitory receptors for HLA-A, HLA-B, HLA-C molecules.


Subject(s)
Down-Regulation , Epitopes/metabolism , HLA-A Antigens/metabolism , HLA-B Antigens/metabolism , Neoplasms/immunology , Antibodies, Monoclonal/immunology , Antibody Specificity , Epitopes/immunology , Flow Cytometry , Genotype , HLA-A Antigens/genetics , HLA-A Antigens/immunology , HLA-B Antigens/genetics , HLA-B Antigens/immunology , Humans , Immunohistochemistry , Immunologic Surveillance , Polymorphism, Genetic , Tumor Cells, Cultured
10.
Cancer Res ; 59(3): 720-7, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9973223

ABSTRACT

In the present study, we have investigated the expression of endothelin 1 (ET-1) and the ET(A) receptor (ET(A)R) and ET(B) receptor (ET(B)R) in primary (n = 30) and metastatic (n = 8) ovarian carcinomas and their involvement in tumor growth. By reverse transcription-PCR and Northern blot analysis, we detected ET-1 mRNA in 90% of primary and 100% of metastatic ovarian carcinomas. ET-1 mRNA expression was significantly higher in tumors than in normal ovarian tissues (n = 12; P < 0.01). ET(A)R mRNA was also detected in 84% of the carcinomas examined, whereas ET(B)R mRNA was expressed in 50% of the tumors. The in vivo presence of mature ET-1 and ET(A)R was confirmed by immunohistochemistry, demonstrating a higher expression in primary and metastatic cells. Ten primary cultures of ovarian tumors secreted ET-1 and were positive for ET-1 and ET(A)R mRNA, whereas only 40% expressed ET(B)R mRNA. Radioligand binding studies showed that ET-1-producing cells also expressed functional ET(A)R, whereas no specific ET(B)R could be demonstrated. ET-1 stimulated dose-dependent [3H]thymidine incorporation and enhanced the mitogenic effect of epidermal growth factor. The ET(A)R-selective antagonist BQ 123 strongly inhibited ET-1-stimulated growth and substantially reduced the basal growth rate of unstimulated cells, whereas the ET(B)R-selective antagonist BQ 788 had no effect. In conclusion, the present data demonstrate a novel mechanism in the growth control of ovarian carcinoma in vivo mediated by the ET-1 autocrine loop that selectively occurs via the ET(A)R.


Subject(s)
Endothelin-1/biosynthesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Endothelin/biosynthesis , Adult , Aged , Blotting, Northern , Cell Division/drug effects , Cell Division/physiology , Endothelin Receptor Antagonists , Endothelin-1/pharmacology , Female , Humans , Middle Aged , Peptides, Cyclic/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Receptor, Endothelin A , Receptor, Endothelin B , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Article in English | MEDLINE | ID: mdl-10664934

ABSTRACT

We have recently shown the expression of lymphoid early developmental markers, including CD104, Thy 1, CD1a, Pgp-1 and TdT, by the cells constituting atopic dermatitis skin infiltrates. To further characterize the cellular phenotypes we used an indirect immunoperoxidase assay to analyze sections from two atopic dermatitis lesion skin biopsies using the following as first step monoclonal antibodies (MAB): anti-CD34, CD2, CD5 and CD7. CD34+ mononuclear cells and endothelial cells were identified. A strong immunoreaction was observed for the T-lineage marker CD2 and CD5, but a poor reaction, if any, was seen for the CD7. Since CD34+ marrow and blood cells are currently believed to be the major source of the hemopoietic precursors, our data provide further substantial evidence supporting the hypothesis that the atopic dermatitis skin cell infiltrate represents an ongoing T-lineage in situ differentiation process regulated by the skin epithelial microenvironment. The observed defective expression of the CD7 antigen requires further investigation for its confirmation as a possible constant feature in atopic dermatitis.


Subject(s)
Antigens, CD34/immunology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Acute Disease , Antibodies, Monoclonal , Antibody Specificity , Cell Differentiation/immunology , Endothelium/pathology , Humans , Skin/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
Int J Cancer ; 77(2): 200-4, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9650552

ABSTRACT

Human melanoma cells express several antigens which are recognized by autologous and specific CTL clones in association with HLA-class-I molecules. Many of these antigens represent suitable targets for tumor immunotherapy, since their expression in human melanoma cells is common and highly specific. In order to achieve real clinical success with therapeutic vaccination strategies, one important requirement is the expression of the target antigen by all the tumor lesions of a patient. We have studied this issue by assessing, through an RT-PCR approach, the expression of MAGE-1, MAGE-2, MAGE-3, BAGE, GAGE-1/2, Tyrosinase and Melan-A/MART-1 genes in 17 clusters of simultaneous in-transit or regional lymph-node metastases collected from 15 stage-III and 1 stage-IV (AJCC/UICC pTNM system) melanoma patients. In 14 out of 17 clusters of simultaneous metastatic lesions (82%), the homogeneity in the pattern of gene expression within the cluster was complete. Heterogeneity within the same cluster was observed in only 3 out of 17 clusters (18%) and represented only minor features. Our data reveal that, in AJCC-stage-III melanoma patients, different but simultaneous metastatic lesions express the same pattern of antigen-coding genes. These observations have 2 main clinical implications: (i) the antigenic characterization of one single and easily accessible lesion allows identification of optimal targets for an active antigen-specific immunotherapy treatment; (ii) almost all the metastatic lesions are expected to be hit by the immune response eventually induced against the tumor antigen. Moreover, these data suggest that active specific immunotherapy directed against MAGE-1, MAGE-3, BAGE, GAGE-1/2, Melan-A/MART-1 and Tyrosinase antigens could be exploited as an adjuvant treatment to surgery in high-risk AJCC-stage-III-melanoma patients.


Subject(s)
Antigens, Neoplasm/analysis , Antigens, Neoplasm/metabolism , Melanoma/genetics , Female , Gene Expression , Humans , Lymphatic Metastasis , Male , Melanoma/secondary , Melanoma-Specific Antigens , Neoplasm Metastasis , Neoplasm Proteins/metabolism
13.
Int J Cancer ; 75(2): 171-5, 1998 Jan 19.
Article in English | MEDLINE | ID: mdl-9462703

ABSTRACT

Increasing experimental evidence indicates that stem-cell factor (SCF) and its cognate receptor c-Kit may participate in the growth control of various solid human malignancies. In the present study, we have extended this analysis to non-small-cell lung carcinomas (NSCLC). The results of an immunohistochemical analysis demonstrated that c-Kit/SCF are expressed by 30%/58% of adenocarcinomas, 15%/37% of squamous-cell carcinomas and by 40%/30% of undifferentiated carcinomas respectively. In 15% of primary and 18% of metastatic tumors, co-expression of the receptor and its ligand was documented. Western-blot assays of tumor extracts demonstrated that both molecules exhibit features of the normal receptor and ligand. Since biologically active SCF is physiologically present in the bloodstream, our data indicate that SCF is available to c-kit-expressing NSCLC cells via autocrine, paracrine or endocrine mechanisms. Thus activation of c-Kit in these tumors may contribute critically to their progression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Lung/chemistry , Proto-Oncogene Proteins c-kit/analysis , Stem Cell Factor/analysis , Carcinoma, Non-Small-Cell Lung/secondary , Humans , Lung Neoplasms/secondary , Molecular Weight , Tumor Cells, Cultured
14.
Article in English | MEDLINE | ID: mdl-10028483

ABSTRACT

The association between the atopic dermatitis, eczema and T-cell immunodeficiency disorders are well known, thus suggesting that bone marrow T-precursors could use the micro-environment of the skin as an extrathymic site for compensatory ontogenesis. In keeping with this hypothesis, we analyzed the atopic dermatitis skin lymphocytic infiltrate phenotypes to establish their ontogenetic stage of development. Cryostatic sections (4 microns) obtained from acute lesional skin biopsies of six patients with extrinsic atopic dermatitis were processed with indirect immunoperoxidase, using a panel of first-step monoclonal antibodies (mAb) specific to CD104 (integrin beta 4 chain), CD90w (Thy 1 antigen), CD44 (phagocytic glycoprotein-1; Pgp-1), CD1a and the DNA polymerase terminal deoxynucleotidyl transferase (TdT). Within the lymphocytic dermal infiltrate different levels of immunoreactivity were observed with respect to CD104, CD90w and CD1a. A strong, spread staining was also detected for mAb specific to Pgp-1 and TdT. Together, the reported features indicate that the atopic dermatitis skin-homing lymphocytes express immunophenotypes which are distinctive of the early T-ontogeny.


Subject(s)
Dermatitis, Atopic/immunology , Skin/immunology , T-Lymphocyte Subsets/immunology , Adult , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/analysis , Biomarkers , Cell Differentiation , DNA Nucleotidylexotransferase/metabolism , Dermatitis, Atopic/pathology , Humans , Immunophenotyping , Mice , Skin/pathology
15.
J Immunother ; 20(6): 466-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409452

ABSTRACT

The human immune repertoire appears to be capable of recognizing normal antigens expressed by tumor cells. Among these antigens, those of differentiation, characterized by a restricted tissue expression, could be of clinical interest since they may represent a target for immunotherapeutic protocols. In this context we have evaluated, in benign and malignant lesions of the melanocytic lineage, the expression of the Melan-A/MART-1 antigen, which has been shown to be recognized by T cells, of HLA-A2 melanoma patients. The immunohistochemical analysis conducted with a Melan-A/MART-1 monoclonal antibody demonstrated that the antigen expression does not correlate with transformation or tumor progression. At variable levels Melan-A/MART-1, differently from other differentiation antigens, is homogeneously expressed by multiple autologous metastases and by melanoma metastases at different body sites. This tissue distribution adds further biological support to the ongoing use of Melan-A/MART-1-related peptides in active immunotherapy.


Subject(s)
Eye Neoplasms/immunology , Melanoma/immunology , Neoplasm Proteins/analysis , Skin Neoplasms/immunology , Antibodies, Monoclonal , Antigens, Neoplasm , HLA-A2 Antigen/analysis , Humans , Immunoenzyme Techniques , MART-1 Antigen , Neoplasm Metastasis , Nevus/immunology
16.
Cancer Res ; 57(8): 1554-60, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9108459

ABSTRACT

Several lines of experimental evidence in in vitro and animal model systems suggest that the integrin alpha(v)beta3 plays a role in the tumorigenicity of human melanoma cells and that the blocking of alpha(v)beta3 ligand binding can inhibit tumor progression. However, there is only scanty information about the role of alpha(v)beta3 in malignant melanoma in a clinical setting. Therefore, in the present study, we have analyzed the distribution in lesions of melanocyte origin and in normal tissues of the alpha(v) integrin subunit and of the alpha(v)beta3 complex and their association with histopathological and clinical parameters of malignant melanoma. We have used as probes the monoclonal antibodies (mAbs) TP36.1 and VF27.263.15, which we have shown with a combination of serological and immunochemical assays to be specific for the alpha(v) subunit and for the alpha(v)beta3 complex, respectively. In immunohistochemical assays, mAb TP36.1 stained both benign and malignant lesions of melanocyte origin. In contrast, the reactivity of mAb VF27.263.15 was restricted to malignant lesions. Both mAbs displayed differential reactivity with primary melanoma lesions of different histotypes because they stained about 50% of acral lentiginous melanoma and superficial spreading melanoma lesions, at least 80% of nodular melanoma lesions, and none of the uveal melanoma lesions tested. Both mAbs TP36.1 and VF27.263.15 stained about 60% of lymph node metastases and 80% of cutaneous metastases. Expression of the alpha(v)beta3 complex in melanocytic lesions resembles that of intercellular adhesion molecule-1 (ICAM-1) in several respects: (a) both are expressed in a significantly (P < 0.004) larger proportion of malignant than of benign lesions; (b) expression of both molecules in primary melanoma lesions is significantly (P < 0.05) associated with lesion thickness; and (c) expression of both molecules in primary lesions from patients with stage I melanoma is significantly (P < 0.05) associated with an increased probability of disease recurrence following surgical excision. alpha(v)beta3 and ICAM-1 in primary melanoma lesions complement each other in predicting the outcome of the disease, because the association with prognosis was enhanced when primary lesions were stained by both anti-alpha(v)beta3 mAb VF27.263.15 and anti-ICAM-1 mAb CL203.4 or by neither mAb. Because alpha(v)beta3 has been suggested as a potential target of immunotherapy, its distribution in normal tissues was investigated. alpha(v)beta3 expression is restricted because it was only detected in ductal epithelium of parotid glands, thyrocytes, basal glands of the stomach, colonic and rectal epithelium glomeruli, Bowman's capsules and proximal and distal tubules of kidneys, and endometrial epithelium. These findings suggest that renal function will be a critical clinical parameter to monitor in therapies of malignant diseases relying on systemic administration of anti-alpha(v)beta3 mAb.


Subject(s)
Antigens, CD/metabolism , Integrins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Skin Neoplasms/metabolism , Antibodies, Monoclonal , Antibody Specificity , Disease-Free Survival , Humans , Integrin alpha3 , Integrin alphaV , Melanoma/pathology , Melanoma/secondary , Skin Neoplasms/pathology , Tumor Cells, Cultured
17.
Br J Cancer ; 76(10): 1255-61, 1997.
Article in English | MEDLINE | ID: mdl-9374368

ABSTRACT

Levels of circulating soluble intercellular adhesion molecule-1 (sICAM-1) are elevated in patients affected by solid malignancies; however, the cellular sources generating high levels of sICAM-1 remain to be characterized. Using conditioned media (CM) from seven ICAM-1-positive or -negative neoplastic cells, we demonstrate that tumour-derived interleukin 1alpha (IL-1alpha) significantly (P < 0.05) up-regulates the release of sICAM-1 by human umbilical vein endothelial cells. The intensity of the effect correlated with the amounts of IL-1alpha detectable in CM. Levels of ICAM-1 mRNA were also up-regulated by tumour-secreted IL-1alpha. The up-regulation of the shedding of sICAM-1 and of its expression at protein and mRNA level were completely reversed by the addition of anti-IL-1alpha neutralizing antibodies. Consistent with the in vitro data, tumour endothelia were strongly stained for ICAM-1 compared with autologous normal tissue endothelia. Taken altogether, our observations reveal an IL-1alpha-mediated tumour-endothelium relationship sustaining the shedding of sICAM-1 by endothelial cells. This is a general phenomenon in solid malignancies that correlates with the ability of neoplastic cells to secrete IL-1alpha rather than with their expression of ICAM-1 and/or histological origin. sICAM-1 has been previously shown to inhibit LFA-1/ICAM-1-mediated cell-cell interactions; therefore, the ability of neoplastic cells to secrete IL-1alpha is likely to represent a mechanism for their escape from immune interaction.


Subject(s)
Endothelium, Vascular/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1/physiology , Neoplasms/immunology , Cells, Cultured , Culture Media, Conditioned , Humans , Intercellular Adhesion Molecule-1/analysis , Up-Regulation , Urinary Bladder Neoplasms/chemistry
18.
J Neurooncol ; 31(1-2): 57-64, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9049831

ABSTRACT

In neuroblastoma, high levels of mRNA for p140trkA and p75LNGFR neurotrophin receptors are predictive of favorable outcome. Their evaluation by Northern blot, however, requires substantial amounts of tissue and this prevents their routine evaluation as well as the possibility for multicenter studies to be easily carried out. In an attempt to overcome these limitations, the feasibility and reliability of determining both neurotrophin receptors on cryostat sections by immunohistochemistry were assessed, and these findings were compared to those obtained from Northern blot analysis. Primary tumor samples from 28 untreated patients at all stages were evaluated by using H10 anti-p140trkA and ME20.4 anti-p75LNGFR mAbs. Although weak, positive immunostaining was found in 9 of 28 tumors for p140trkA and in 5 of 28 tumors for p75LNGFR. As compared to Northern blot, the concordance rate was 79% (22 of 28 cases) for p140trkA (p < 0.05) and 71% (20 of 28 cases) for p75LNGFR (p < 0.05). No case negative for Northern blot was found to be positive with immunohistochemistry. Since only high mRNA levels for both receptors have been shown to be clinically relevant, their immunohistochemical detection, although less sensitive than Northern blot, can be just as sufficient and reliable as a prognostic tool, and possibly with a better cost-benefit ratio.


Subject(s)
Neuroblastoma , Proto-Oncogene Proteins/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Nerve Growth Factor/analysis , Antibodies, Monoclonal , Blotting, Northern , Child , Humans , Immunohistochemistry , Neuroblastoma/diagnosis , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Receptor, Nerve Growth Factor , Receptor, trkA , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/immunology , Tumor Cells, Cultured/chemistry
19.
Tissue Antigens ; 50(6): 555-66, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9458108

ABSTRACT

Lacking monospecific antibodies to HLA-C, the expression and synthesis of these molecules have been difficult to evaluate. Using biochemical and flow cytometry approaches, the present report demonstrates that the reactivity of the murine monoclonal antibody L31 is restricted to naturally occurring HLA-C (HLA-Cw1 through -Cw8), HLA-B8 and HLA-B51 heavy chains not associated with beta2-microglobin (beta2m). This is due to two properties of HLA-C heavy chains: (a) they share the L31 epitope which distinguishes them from all the HLA-A and most HLA-B molecules; (b) they accumulate intracellularly, in a beta2m-free form, in much greater amounts than most L31-reacting HLA-B heavy chains. On the basis of this restricted reactivity, a representative panel of normal and neoplastic human tissues and cells derived from HLA-B8- B51- individuals was selected and employed to assess the tissue distribution, surface expression and IFN-gamma responsiveness of beta2m-free HLA-C heavy chains. At variance from antibody W6/32 to beta2m-associated heavy chains, L31 stains normal and neoplastic tissues with a ground-glass pattern and weakly binds to the surface of viable cells, even after treatment with interferon gamma (IFN-gamma). Thus, beta2m-free HLA-C heavy chains are, for the most part, located intracellularly. In spite of their distinct cellular localization, L31- and W6/32-reacting molecules have an overlapping tissue distribution, undergo concordant changes upon transformation and are upregulated in their synthesis by IFN-gamma to a similar extent. These observations demonstrate a coordinate regulation of HLA-C with HLA-A and -B molecules. In addition, they indicate that the assembly of HLA-C is impaired in most body districts and IFN-gamma is unable to completely reverse this impairment. The present results are consistent with a low surface expression of HLA-C and with a privileged role of these molecules in signaling class I loss to cytotoxic effectors in pathological conditions.


Subject(s)
HLA-C Antigens/immunology , Interferon-gamma/immunology , beta 2-Microglobulin/immunology , Adult , Animals , Cell Line, Transformed , Flow Cytometry , Gene Deletion , HLA-B Antigens/genetics , HLA-B Antigens/immunology , HLA-B51 Antigen , HLA-B8 Antigen/genetics , HLA-B8 Antigen/immunology , HLA-C Antigens/genetics , HT29 Cells , Humans , Interferon-gamma/pharmacology , Lymphoid Tissue , Mice , Neoplasms/immunology , Precipitin Tests , Recombinant Proteins , Tissue Distribution , Tissue Fixation , Tumor Cells, Cultured
20.
Br J Cancer ; 74(12): 1862-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8980383

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) stimulates the invasive growth of epithelial cells via the c-MET oncogene-encoded receptor. In normal lung, both the receptor and the ligand are detected, and the latter is known to be a mitogenic and a motogenic factor for both cultured bronchial epithelial cells and non-small-cell carcinoma lines. Here, ligand and receptor expression was examined in 42 samples of primary human non-small-cell lung carcinoma of different histotype. Each carcinoma sample was compared with adjacent normal lung tissue. The Met/HGF receptor was found to be 2 to 10-fold increased in 25% of carcinoma samples (P = 0.0113). The ligand, HGF/SF, was found to be 10 to 100-fold overexpressed in carcinoma samples (P < 0.0001). Notably, while HGF/SF was occasionally detectable and found exclusively as a single-chain inactive precursor in normal tissues, it was constantly in the biologically-active heterodimeric form in carcinomas. Immunohistochemical staining showed homogeneous expression of both the receptor and the ligand in carcinoma samples, whereas staining was barely detectable in their normal counterparts. These data show that HGF/SF is overexpressed and consistently activated in non-small-cell lung carcinomas and may contribute to the invasive growth of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Hepatocyte Growth Factor/biosynthesis , Lung Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Antibodies, Monoclonal/analysis , Blotting, Western/methods , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Female , Humans , Lung/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Proto-Oncogene Proteins c-met
SELECTION OF CITATIONS
SEARCH DETAIL
...