Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(7): 887-96, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-25961924

ABSTRACT

Human Mena (hMENA), an actin regulatory protein of the ENA/VASP family, cooperates with ErbB receptor family signaling in breast cancer. It is overexpressed in high-risk preneoplastic lesions and in primary breast tumors where it correlates with HER2 overexpression and an activated status of AKT and MAPK. The concomitant overexpression of hMENA and HER2 in breast cancer patients is indicative of a worse prognosis. hMENA is expressed along with alternatively expressed isoforms, hMENA(11a) and hMENAΔv6 with opposite functions. A novel role for the epithelial-associated hMENA(11a) isoform in sustaining HER3 activation and pro-survival pathways in HER2-overexpressing breast cancer cells has been identified by reverse phase protein array and validated in vivo in a series of breast cancer tissues. As HER3 activation is crucial in mechanisms of cell resistance to PI3K inhibitors, we explored whether hMENA(11a) is involved in these resistance mechanisms. The specific hMENA(11a) depletion switched off the HER3-related pathway activated by PI3K inhibitors and impaired the nuclear accumulation of HER3 transcription factor FOXO3a induced by PI3K inhibitors, whereas PI3K inhibitors activated hMENA(11a) phosphorylation and affected its localization. At the functional level, we found that hMENA(11a) sustains cell proliferation and survival in response to PI3K inhibitor treatment, whereas hMENA(11a) silencing increases molecules involved in cancer cell apoptosis. As shown in three-dimensional cultures, hMENA(11a) contributes to resistance to PI3K inhibition because its depletion drastically reduced cell viability upon treatment with PI3K inhibitor BEZ235. Altogether, these results indicate that hMENA(11a) in HER2-overexpressing breast cancer cells sustains HER3/AKT axis activation and contributes to HER3-mediated resistance mechanisms to PI3K inhibitors. Thus, hMENA(11a) expression can be proposed as a marker of HER3 activation and resistance to PI3K inhibition therapies, to select patients who may benefit from these combined targeted treatments. hMENA(11a) activity could represent a new target for antiproliferative therapies in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Microfilament Proteins/metabolism , Receptor, ErbB-3/genetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Microfilament Proteins/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Isoforms , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/genetics , Transfection
2.
Oncogene ; 34(19): 2493-504, 2015 May 07.
Article in English | MEDLINE | ID: mdl-24998848

ABSTRACT

The TP53 tumor-suppressor gene is frequently mutated in human cancer. Missense mutations can add novel functions (gain-of-function, GOF) that promote tumor malignancy. Here we report that mutant (mut) p53 promotes tumor malignancy by suppressing the expression of a natural occurring anti-inflammatory cytokine, the secreted interleukin-1 receptor antagonist (sIL-1Ra, IL1RN). We show that mutp53 but not wild-type (wt) p53 suppresses the sIL-1Ra production in conditioned media of cancer cells. Moreover, mutp53, but not wtp53, binds physically the sIL-1Ra promoter and the protein-protein interaction with the transcriptional co-repressor MAFF (v-MAF musculoaponeurotic fibrosarcoma oncogene family, protein F) is required for mutp53-induced sIL-1Ra suppression. Remarkably, when exposed to IL-1 beta (IL-1ß) inflammatory stimuli, mutp53 sustains a ready-to-be-activated in vitro and in vivo cancer cells' response through the sIL-1Ra repression. Taken together, these results identify sIL-1Ra as a novel mutp53 target gene, whose suppression might be required to generate a chronic pro-inflammatory tumor microenvironment through which mutp53 promotes tumor malignancy.


Subject(s)
DNA-Binding Proteins/genetics , Inflammation/genetics , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , HT29 Cells , Hep G2 Cells , Humans , Inflammation/immunology , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/pharmacology , MCF-7 Cells , MafF Transcription Factor/metabolism , Mutation , Neoplasms/genetics , Neoplasms/mortality , Nuclear Proteins/metabolism , Prognosis , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , RNA, Small Interfering , Tumor Microenvironment/immunology
3.
J Immunother ; 24(3): 221-31, 2001.
Article in English | MEDLINE | ID: mdl-11394499

ABSTRACT

The HER2 oncogene and its relative oncoprotein, gp185HER2, a transmembrane glycoprotein belonging to the epidermal growth factor receptor family, are overexpressed in a wide range of solid tumors including breast and ovarian cancer. In patients with breast cancer, both humoral and cell-mediated HER2 immune responses have been found as well as in some patients with gp185HER2 nonoverexpressing tumors. To establish whether peptide sequences identified as HLA-A2-restricted T-cell epitopes are expressed in breast tumor cell lines and tissues, we produced and characterized by different methodologic approaches polyclonal antibodies raised against four gp185HER2 peptides. Two of the antibodies recognized peptides eluted from the HLA-A2 groove of the mDAmB231 breast cancer cell line expressing a basal level of gp185HER2. Paraffin-embedded primary and metastatic breast tumors were specifically immunostained by all four reagents, thereby showing an overlapping reactivity. When this immunoreactivity was compared with that obtained using two different monoclonal antibodies, in 105 breast primary tumors and 36 corresponding lymph node metastases, we identified a subset of tumors that were negative with anti-gp185HER2 monoclonal antibodies and positive with the four antipeptide antibodies. Our novel observations provide in vivo evidence of the complexity involved in evaluating HER2 expression, and open a new path for understanding the biologic significance of HER2 status in breast tumors.


Subject(s)
Antibodies, Neoplasm , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Amino Acid Sequence , Animals , Antibodies, Neoplasm/biosynthesis , Antibody Specificity , Binding Sites , Female , Gene Expression , Genes, erbB-2 , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , Humans , Immunohistochemistry , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/immunology , Prognosis , Rabbits , Tumor Cells, Cultured
4.
Int J Cancer ; 84(6): 598-603, 1999 Dec 22.
Article in English | MEDLINE | ID: mdl-10567905

ABSTRACT

Overexpression of gp185(erbB-2) has been associated with reduced survival in breast-cancer patients. Our earlier results, now confirmed in a larger cohort of patients (798), evidenced that the HLA-A2 allele may participate in the modulation of the erbB-2 tumor phenotype in vivo. In the present study, we evaluated other clinico-biopathologic parameters possibly involved in the host immune response against erbB-2. Localization of the CD3(+) T-cell infiltrate was taken into consideration in 705 primary breast tumors, and expression of HLA-class-I and HLA-A2 antigens was evaluated in a subgroup of 170 frozen primary tumors of HLA-A2-positive patients. The presence or the absence of HLA-class-I and HLA-A2 antigens in primary tumors did not correlate with erbB-2 expression. However, HLA-A2-positive tumors preferentially showed intratumoral lymphocyte localization, whereas the lesions displaying undetectable HLA-class-I expression showed peritumoral CD3(+) T-cell localization. Taking into account erbB-2 immunoreactivity, we found that the relationship between HLA-A2 expression and intratumoral CD3(+) T-lymphocyte localization is significant only in the erbB-2 negative subset, whereas the relationship between lack of HLA-class-I expression and peritumoral CD3(+) T-lymphocyte localization is significant only in the erbB-2-positive subset. These data provide novel in vivo evidence of the possible contribution of the host immune system to control of erbB-2 oncogene overexpression in breast cancer. Int. J. Cancer (Pred. Oncol.) 84:598-603, 1999.


Subject(s)
Breast Neoplasms/immunology , HLA-A2 Antigen/immunology , Immunologic Surveillance/immunology , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/immunology , Adult , Age Factors , Aged , Aged, 80 and over , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , CD3 Complex/immunology , Female , HLA-A1 Antigen/immunology , Humans , Immunohistochemistry , Middle Aged , Phenotype , Prognosis , T-Lymphocytes/immunology
5.
J Immunother ; 20(6): 431-6, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409448

ABSTRACT

Small peptides, 8-10 amino acids long, derived from degradation of cytoplasmic proteins by a proteasome-proteinase complex, are usually presented and recognized by CD8+ cytolytic T lymphocytes (CTLs) associated with major histocompatibility complex (MHC) class I molecules. Recently synthetic peptides were used for the in vitro induction of tumor-specific CTLs, offering another strategy in the study of the immune-response repertoire and providing a new tool in cancer vaccination and immunotherapy. Peptides derived from otherwise normal proteins, overexpressed in many tumors as products of the protooncogene, may represent a target for an immune response. This is the case of HER-2/neu gene (also known as ErbB-2), encoding a cysteine-rich glycoprotein transmembrane receptor with tyrosine kinase activity (gp185neu). Recent data, demonstrating that HLA-A2.1-related peptides are able to stimulate in vitro CD8+ lymphocytes, Prompted us to study the binding to HLA-A2.1 molecules of several gp185 synthetic peptides containing a cystein residue and to define the relevance of this amino acid residue in the reduced or oxidated form of the sulfhydryl group. We found that monomers and their homodimers, linked by a disulfide bridge, bind to HLA-A2.1 molecules with overlapping affinity. These results suggest that additional amino acids of the nonapeptide do not prevent the binding and the HLA refolding through chemical or sterical interactions. This might be of particular relevance for the in vivo processing of cysteine-rich proteins. Because ErbB-2 molecules, as tumor-differentiation antigens in melanoma, are cysteine-rich molecules, it may be relevant to evaluate the possible role of the cystine residues interacting with the T-cell receptor. The recognition of these heterodimers by CD8+ lymphocytes will require functional in vivo studies.


Subject(s)
Cysteine/metabolism , HLA-A2 Antigen/metabolism , Histocompatibility Antigens Class I/metabolism , Oligopeptides/metabolism , Amino Acid Sequence , Dimerization , Glutathione/metabolism , Humans , Peptide Fragments/metabolism , Receptor, ErbB-2/chemistry
6.
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
7.
Exp Cell Res ; 236(1): 76-85, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9344587

ABSTRACT

Growth factors modulate integrin-mediated cell adhesion and motility, and their receptors are thought to share proteins that mediate intracellular signaling with integrin receptors. The crosstalk between these receptors is thought to play a relevant role in transformation and tumor progression. To highlight possible interactions between growth factors and cell adhesion receptors we investigated whether integrins associate with tyrosine kinase receptors in tumor cells. By affinity chromatography and Western blot analyses of purified immune complexes, we studied the association of laminin receptors (alpha 6 beta 1 and alpha 6 beta 4) with ErbB-2 tyrosine kinase in human carcinoma cell lines. We demonstrated that the alpha 6 beta 4 and alpha 6 beta 1 integrins coprecipitated with ErbB-2 in lysates from carcinoma or NIH3T3 cells overexpressing ErbB-2. Integrin-mediated activation of ErbB-2 receptors suggested that this association is functionally meaningful. Indeed, carcinoma cells treated with a monoclonal antibody to the alpha 6 integrin subunit showed a ligand-dependent increase of ErbB-2-phosphorylated molecules coprecipitated with integrins and an increased DNA synthesis. The interaction between growth factor receptors and integrins was also studied in NIH3T3 cells overexpressing alpha 6 beta 4 receptors and ErbB-2 protein. We report that cells overexpressing both receptors, but not those overexpressing a crippled ErbB-2, showed enhanced proliferation rates and invasiveness, further suggesting that alpha 6 beta 4 integrin and ErbB-2 receptor interaction might contribute to generate a more malignant phenotype in carcinoma cells.


Subject(s)
Antigens, Surface/metabolism , Breast Neoplasms , Integrins/metabolism , Ovarian Neoplasms , Receptor, ErbB-2/metabolism , 3T3 Cells/chemistry , 3T3 Cells/metabolism , Animals , Antibodies, Monoclonal , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antigens, Surface/genetics , Antigens, Surface/immunology , Cell Division/physiology , Female , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha6beta1 , Integrin alpha6beta4 , Integrins/genetics , Integrins/immunology , Ligands , Mice , Neoplasm Invasiveness , Phosphorylation , Receptor, ErbB-2/genetics , Receptors, Laminin/genetics , Receptors, Laminin/immunology , Receptors, Laminin/metabolism , Thymidine/metabolism , Transfection , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/metabolism
9.
J Clin Invest ; 94(4): 1426-31, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7929817

ABSTRACT

Cell-mediated immune response to breast tumor has only been marginally investigated. To gain insight into this issue we have developed two clones of distinct phenotype, CD3+ alpha/beta, CD4+, CD8-, CD16-, and CD3+ alpha/beta, CD4-, CD8+, CD16-, respectively, from peripheral blood lymphocytes (PBL) of a breast cancer patient. These effectors, selected on the basis of their cytolytic activity against autologous tumor cells and lack of lysis on NK-sensitive cell lines, preferentially recognize autologous tumor cells. The two clones' cytotoxic activity, while inhibited by anti-LFA-1 mAb, could not be abolished by mAbs to CD3, to class I and class II MHC molecules, and by mAbs to molecules involved in T cell function (i.e., CD4, CD8, CD2). The molecular structure of the alpha and beta T cell receptor chains of the two effector cells, confirmed their clonality and showed that, despite an overlapping killing pattern, they possess distinct TCR alpha and beta chains. These findings demonstrate that breast tumor-specific CTL clones can be generated through current technology and that a alpha/beta effector cell population operating through a HLA-unrestricted and TCR/CD3-independent pathway may be involved in the identification and killing of this tumor.


Subject(s)
Breast Neoplasms/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigens, Differentiation, T-Lymphocyte/immunology , Base Sequence , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunity, Cellular , Lymphocyte Culture Test, Mixed , Middle Aged , Molecular Sequence Data , RNA, Messenger/analysis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Sequence Alignment , Sequence Analysis, DNA , Tumor Cells, Cultured
10.
J Clin Invest ; 90(3): 1093-9, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1387883

ABSTRACT

Intratumor heterogeneity for susceptibility to cytotoxic T lymphocytes (CTL)-mediated lysis represents a major obstacle to cancer adoptive immunotherapy. To overcome the heterogeneity observed in terms of susceptibility of target cells to cell-mediated lysis, in this study we used two purified bispecific monoclonal antibodies (bsmAbs) that recognize molecules expressed by cytotoxic effector cells (CD3 and IgG Fc receptorial molecules), as well as one high molecular weight melanoma-associated antigen (HMW-MAA). The ability of these reagents to enhance or induce a relevant in vitro cytotoxic activity by a CTL clone (CTL 49) isolated from PBL of a melanoma patient was tested on a large panel of autologous and allogeneic melanoma cell lines and clones. Functional studies revealed that the CTL 49 clone lysed all the HMW-MAA+ tumor lines in the presence of bsmAbs and that these reagents affected the target lysis in a cooperative fashion. The effectiveness of bsmAbs in overcoming the heterogeneous susceptibility of human melanoma cells to cell-mediated lysis may find practical implications in cancer adoptive immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Cytotoxicity, Immunologic , Melanoma/immunology , Animals , Antigens, Differentiation/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Neoplasm , CD3 Complex , Humans , Immunotherapy, Adoptive , Melanoma/pathology , Melanoma/therapy , Melanoma-Specific Antigens , Mice , Mice, Inbred BALB C , Neoplasm Proteins/analysis , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Fc/immunology , Receptors, IgG , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
11.
Ann Ist Super Sanita ; 27(1): 133-7, 1991.
Article in English | MEDLINE | ID: mdl-1835569

ABSTRACT

Mouse monoclonal antibodies specific for CD3, FcR and a melanoma associated antigen have been produced. Drug resistence of such hibrydomas has been obtained tranfecting them with plasmids containing genes conferring specific resistence. Retrovirus derived shuttle vectors with high transfection efficiency have been used for transfection. Hybrydomas were than fused and bispecific antibody producing cells selected. Purification was performed by HPLC. Such bispecific antibodies can be used in ex vivo and in vivo immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Genetic Vectors , Retroviridae/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Neoplasm , CD3 Complex , Cell Fusion , Drug Resistance , Hybridomas/drug effects , Hybridomas/immunology , Immunotherapy , Melanoma-Specific Antigens , Mice , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Fc/immunology , Selection, Genetic
12.
Cancer Res ; 50(23): 7422-9, 1990 Dec 01.
Article in English | MEDLINE | ID: mdl-1701342

ABSTRACT

Twenty-five early-passage (less than or equal to 8) melanoma cell lines, isolated from ten patients with metastatic melanoma, were analyzed by a combination of serological, immunochemical, and molecular methods for mRNA levels, synthesis, and surface expression of MHC class I and class II antigens prior to and following exposure to recombinant human leukocyte (IFN-alpha A), fibroblast (IFN-beta), and immune (IFN-gamma) interferon. All the cell lines expressed variable levels of HLA class I gene products that were up-regulated to different extents upon exposure to specific interferons (IFNs). HLA class II antigens were expressed in 22 of the 25 melanoma lines and IFN-gamma increased the levels of class II mRNA, protein synthesis, and surface expression in all cultures displaying baseline expression. A significant up-regulation of class II antigen expression by IFN-alpha or -beta, associated with higher levels of class II transcripts and enhanced synthesis, was found only in two early-passage human melanoma cell lines. In three lesions from the same patient which did not constitutively express class II antigens, no expression of these glycoproteins could be induced with any of the IFNs. These results indicate that IFN-gamma does not act as a de novo inducer of class II antigen expression in early-passage human melanoma cell lines. This hypothesis is further supported by analysis of class II-associated invariant chain (Ii) expression, which is expressed and induced by IFNs in a manner similar to that of class II antigens. The present study also indicates that early-passage metastatic melanoma lesions from the same patient are heterogeneous in their de novo expression of major histocompatibility antigens and in their modulation by IFNs.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , Histocompatibility Antigens Class II/biosynthesis , Interferons/pharmacology , Major Histocompatibility Complex , Melanoma/metabolism , Blotting, Northern , Cell Line , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Neoplastic , HLA-DR Antigens/biosynthesis , Histocompatibility Antigens Class I/biosynthesis , Humans , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , RNA/analysis , Recombinant Proteins
13.
Int J Cancer ; 46(3): 539-45, 1990 Sep 15.
Article in English | MEDLINE | ID: mdl-2118485

ABSTRACT

We have investigated the relationship between in vitro cultivation of autologous melanoma metastases derived from different patients and their levels of expression of class-I and -II major histocompatibility complex (MHC) antigens and melanoma-associated antigens (MAAs). Cell cultures were established from 23 individual metastatic melanoma lesions from 10 patients and were tested early after isolation (between 3rd and 10th passages) for both constitutive expression and modulation by recombinant human leukocyte (IFN-alpha), fibroblast (IFN-beta) or immune (IFN-gamma) interferon of MHC antigens and MAA. All of the melanoma cell lines displayed altered antigen expression following IFN treatment. While in vitro cultures derived from different individuals varied in both constitutive and IFN-modified antigenic expression, cultures of autologous metastases derived from the same patient were very similar. In addition, differences in antigenic profile were apparent when early-passage in vitro cultures were compared with the same melanoma lesion, not established in culture, from which they were derived. The unique de novo and IFN-modified antigenic phenotype of cultures derived from different patients indicates that the antigenic phenotype displayed by melanoma cultures grown in vitro is genetically determined. The differences found between in vitro cultures and their corresponding in vivo lesions, as well as the antigenic heterogeneity displayed by multiple autologous melanoma lesions in vivo, suggest that the in vivo antigenic phenotype may be determined, at least in part, at an epigenetic level.


Subject(s)
Antigens, Neoplasm/analysis , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Melanoma/immunology , Antigens, Neoplasm/genetics , Enzyme-Linked Immunosorbent Assay , HLA Antigens/analysis , Humans , Melanoma/pathology , Neoplasm Metastasis/pathology , Phenotype , Recombinant Proteins/pharmacology , Tumor Cells, Cultured/drug effects
14.
Proc Natl Acad Sci U S A ; 87(8): 2941-5, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2326256

ABSTRACT

The present study reports on the use of gene transfer by retrovirus-derived shuttle vectors in the generation of hybrid hybridomas secreting bispecific monoclonal antibodies. neo- and dhfr- genes were infected into distinct murine hybridomas, thus conferring a dominant resistance trait to geneticin (G418) and to methotrexate. The vectors employed were replication-deficient and dependent on complementation by a helper virus provided by the irradiated packaging lines. After cocultivation with the relevant packaging cell lines, stable hybridoma lines expressing the selectable markers were easily obtained and were then suitable for conventional somatic fusion. This high-efficiency method was used to generate two bispecific monoclonal antibodies simultaneously targeting molecules expressed on cytotoxic cells (i.e., T lymphocytes and natural killer cells) against a human melanoma-associated antigen.


Subject(s)
Antibodies, Monoclonal/genetics , Genetic Vectors , Transfection , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/isolation & purification , Cell Line , Cells, Cultured , Chromatography, High Pressure Liquid , Cytotoxicity, Immunologic , Drug Resistance/genetics , Gentamicins/pharmacology , Humans , Hybridomas/immunology , Methotrexate/pharmacology , Mice , Retroviridae/genetics , T-Lymphocytes, Cytotoxic/immunology
15.
Dev Biol Stand ; 71: 15-22, 1990.
Article in English | MEDLINE | ID: mdl-2401381

ABSTRACT

This report deals with the use of gene transfer by retrovirus-derived shuttle vectors in a novel model aimed at the generation of hybrid hybridomas secreting bispecific monoclonal antibodies (biMAbs). Following this approach, two genes conferring dominant resistance trait to the neomycine analogue geneticin (G418) and to methotrexate (MTX) respectively, were infected in two established hybridoma lines, each producing a well characterized MAb. The vectors used here were replication-deficient, being dependent on the complementation of helper virus provided by packaging lines. The infection procedure involved co-cultivation of the hybridomas with irradiated packaging cell lines, previously transfected with the vectors and producing the recombinant retroviruses, not inclusive of helper virus in their genome. The packaging lines used were psi2 ecotropic cells made able to produce high titers of virus. Further, the vector pMV7 was carrying G418 resistance while the pSDHT render the cells able to survive MTX. Easy and fast transfer of the dominant selection markers yielded lines of hybridomas to be fused according to the conventional somatic fusions. The resulting double hybridomas were tested for the production of hybrid molecules retaining parental specificity and successively underwent extensive cloning. The purification system featuring the most efficiently between the true biMAbs and the parental immunoglobulins (or other combination products) proved to be HPLC on hydroxylapatite column. The method described above was successful in producing two biMAbs targeting simultaneously molecules expressed on cytotoxic cells (such as CD3 on T-lymphocytes and CD16 on NK cells) and the melanoma-associated antigen Ep2.


Subject(s)
Antibodies, Monoclonal/genetics , Genetic Vectors , Retroviridae/genetics , Transfection , Animals , Antibodies, Monoclonal/biosynthesis , Antibody Specificity , Cell Line , Genetic Markers , Hybridomas , Mice , Mice, Inbred BALB C , Models, Genetic
17.
Anticancer Res ; 6(5): 877-84, 1986.
Article in English | MEDLINE | ID: mdl-2432832

ABSTRACT

The ability of interferons to modulate the antigenic phenotype of tumor cells may involve alterations in the transcription, translation, membrane expression and shedding of Major Histocompatibility Complex (MHC) and Tumor Associated Antigens (TAAs). In the present study we have investigated possible mechanisms by which recombinant human interferons, IFN-alpha, -beta and -gamma, alter the antigenic profile of long- and short-term human melanoma cultures. IFN-alpha and -beta induced similar changes in the synthesis, expression and shedding of two TAAs, a HMW-MAA and a Cyt-MAA, in the established melanoma cell line Colo 38, whereas IFN-gamma exerted a differential effect on these melanoma associated antigens. Moreover, IFN-gamma was relatively more effective than IFN-alpha or -beta in upregulating the synthesis, expression and shedding of class I MHC antigens. At the same time a dramatic differential effect of the interferons was observed with class II MHC antigens. IFN-alpha or -beta induced a modest increase in the synthesis and expression of these antigens, whereas IFN-gamma was greater than 3-fold more active in inducing the synthesis and expression of DR/DP antigens and greater than 4- and greater than 10-fold more effective in increasing the synthesis and expression, respectively, of DQ antigens. Analysis of the levels of cytoplasmic mRNA for the DR-alpha and DQ-beta genes indicated no significant difference between IFN-alpha, beta or -gamma treated cells suggesting that IFN-gamma enhancement of the synthesis of DR and DQ antigens may occur at a posttranscriptional level. In the case of a newly established human melanoma cell line (MG-3) IFN-gamma enhanced the synthesis but not the expression of DR antigens and did not alter either the synthesis or expression of DQ antigens. Our studies indicate that the effects of interferons on the antigenic phenotype of melanoma cells will vary depending on the type of interferon employed, the antigen monitored and the target cell studied. In addition, it is also apparent that some of the biosynthetic steps involved in regulating the synthesis, expression and shedding of antigens may be coordinately regulated in some melanoma cells, whereas these processes may be under independent control in other melanoma populations.


Subject(s)
Antigens, Neoplasm/biosynthesis , Interferons/pharmacology , Major Histocompatibility Complex , Melanoma/immunology , Neoplasm Proteins/biosynthesis , Recombinant Proteins/pharmacology , Antigens, Neoplasm/genetics , Cell Line , Gene Expression Regulation/drug effects , Humans , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Melanoma-Specific Antigens , Neoplasm Proteins/genetics , Phenotype , RNA, Messenger/metabolism , Time Factors
18.
Chemioterapia ; 5(1): 33-6, 1986 Feb.
Article in English | MEDLINE | ID: mdl-3955781

ABSTRACT

The authors evaluated the results of a study using clindamycin phosphate plus gentamicin in short-term therapy in patients with tumors submitted to surgery for removal of the primary lesion. Only 6.6% of these patients became infected, and these good results are most likely due to the synergic activity of clindamycin with the physiological immune response. This agent, in fact, was able to interfere with IgM/IgA immunoregulatory balance by enhancing IgM production and, consequently, phagocytic mechanisms.


Subject(s)
Clindamycin/therapeutic use , Neoplasms/surgery , Premedication , Female , Gentamicins/therapeutic use , Humans , Immunoglobulins/analysis , Male , Middle Aged , Neoplasms/immunology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...