Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
1.
BMC Cancer ; 23(1): 907, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37752419

ABSTRACT

BACKGROUND: Heterozygous isocitrate dehydrogenase (IDH) mutations occur in about half of conventional central bone chondrosarcomas (CCBC). Aim of this study was to assess the frequency and prognostic impact of IDH mutations in high grade CCBC patients. METHODS: 64 patients with G2 and G3 CCBC were included. DNA extraction, PCR amplification of IDH1/2 exon 4s, and sequencing analysis with Sanger were performed. RESULTS: IDH mutations were detected in 24/54 patients (44%): IDH1 in 18, IDH2 in 4, and both IDH1/2 in 2 patients. The frequency of mutations was 37% in G2 vs. 69% in G3 (p = 0.039), and 100% in three Ollier disease associated chondrosarcoma. 5-year overall survival (OS) at 124 months (range 1-166) was 51%, with no significant difference based on the IDH mutational status: 61% in IDHmut vs. 44% in IDH wild type (IDHwt). The 5-year relapse free survival (RFS) was 33% (95% CI:10-57) for IDHmut vs. 57% (95%CI: 30-77) for IDHwt. Progression free survival (PFS) was 25% (95%CI:1-65) IDHmut vs. 16% (95%CI: 0.7-52) IDHwt. 55% (5/9) of IDHmut G2 became higher grade at the recurrence, as compared with 25% (3/12) of G2 IDHwt. CONCLUSIONS: This study shows a higher frequency of IDH mutations in G3 CCBC as compared with G2. No significant differences in OS, RFS, and PFS by mutational status were detected. After relapse, a higher rate of G3 for IDH mutated CCBC was observed.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Chondrosarcoma/genetics , Exons , Bone Neoplasms/genetics
2.
Oncogene ; 36(12): 1721-1732, 2017 03 23.
Article in English | MEDLINE | ID: mdl-27641338

ABSTRACT

We have previously shown that the d16HER2 splice variant is linked to HER2-positive breast cancer (BC) tumorigenesis, progression and response to Trastuzumab. However, the mechanisms by which d16HER2 contributes to HER2-driven aggressiveness and targeted therapy susceptibility remain uncertain. Here, we report that the d16HER2-positive mammary tumor cell lines MI6 and MI7, derived from spontaneous lesions of d16HER2 transgenic (tg) mice and resembling the aggressive features of primary lesions, are enriched in the expression of Wnt, Notch and epithelial-mesenchymal transition pathways related genes compared with full-length wild-type (WT) HER2-positive cells (WTHER2_1 and WTHER2_2) derived from spontaneous tumors arising in WTHER2 tg mice. MI6 cells exhibited increased resistance to anoikis and significantly higher mammosphere-forming efficiency (MFE) and self-renewal capability than the WTHER2-positive counterpart. Furthermore, d16HER2-positive tumor cells expressed a higher fraction of CD29High/CD24+/SCA1Low cells and displayed greater in vivo tumor engraftment in serial dilution conditions than WTHER2_1 cells. Accordingly, NOTCH inhibitors impaired mammosphere formation only in MI6 cells. A comparative analysis of stemness-related features driven by d16HER2 and WTHER2 in ad hoc engineered human BC cells (MCF7 and T47D) revealed a higher MFE and aldehyde dehydrogenase-positive staining in d16HER2- vs WTHER2-infected cells, sustaining consistent BC-initiating cell enrichment in the human setting. Moreover, marked CD44 expression was found in MCF7_d16 and T47D_d16 cells vs their WTHER2 and Mock counterparts. Clinically, BC cases from two distinct HER2-positive cohorts characterized by high levels of expression of the activated-d16HER2 metagene were significantly enriched in the Notch family and signal transducer genes vs those with low levels of the metagene.


Subject(s)
Alternative Splicing , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Receptor, ErbB-2/genetics , Animals , Apoptosis/genetics , Biomarkers , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cluster Analysis , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Gene Expression Profiling , Humans , Mice , Mutation , Receptor, ErbB-2/metabolism , Receptors, Notch/metabolism , Signal Transduction
3.
Oncogene ; 33(15): 1912-21, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-23644663

ABSTRACT

CD99, a transmembrane protein encoded by MIC2 gene is involved in multiple cellular events including cell adhesion and migration, apoptosis, cell differentiation and regulation of protein trafficking either in physiological or pathological conditions. In osteosarcoma, CD99 is expressed at low levels and functions as a tumour suppressor. The full-length protein (CD99wt) and the short-form harbouring a deletion in the intracytoplasmic domain (CD99sh) have been associated with distinct functional outcomes with respect to tumour malignancy. In this study, we especially evaluated modulation of cell-cell contacts, reorganisation of the actin cytoskeleton and modulation of signalling pathways by comparing osteosarcoma cells characterised by different metastasis capabilities and CD99 expression, to identify molecular mechanisms responsible for metastasis. Our data indicate that forced expression of CD99wt induces recruitment of N-cadherin and ß-catenin to adherens junctions. In addition, transfection of CD99wt inhibits the expression of several molecules crucial to the remodelling of the actin cytoskeleton, such as ACTR2, ARPC1A, Rho-associated, coiled-coil containing protein kinase 2 (ROCK2) as well as ezrin, an ezrin/radixin/moesin family member that has been clearly associated with tumour progression and metastatic spread in osteosarcoma. Functional studies point to ROCK2 as a crucial intracellular mediator regulating osteosarcoma migration. By maintaining c-Src in an inactive conformation, CD99wt inhibits ROCK2 signalling and this leads to ezrin decrease at cell membrane while N-cadherin and ß-catenin translocate to the plasma membrane and function as main molecular bridges for actin cytoskeleton. Taken together, we propose that the re-expression of CD99wt, which is generally present in osteoblasts but lost in osteosarcoma, through inhibition of c-Src and ROCK2 activity, manages to increase contact strength and reactivate stop-migration signals that counteract the otherwise dominant promigratory action of ezrin in osteosarcoma cells.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , Neoplasm Invasiveness/genetics , Osteosarcoma/genetics , Osteosarcoma/metabolism , rho-Associated Kinases/metabolism , 12E7 Antigen , Actin Cytoskeleton/metabolism , Antigens, CD/genetics , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement/genetics , Cytoskeletal Proteins , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Osteosarcoma/pathology , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transfection , rho-Associated Kinases/genetics
4.
Br J Cancer ; 107(8): 1302-9, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22929887

ABSTRACT

BACKGROUND: Human immune system (HIS)-engrafted mice are new tools to investigate human immune responses. Here, we used HIS mice to study human immune responses against human HER-2-positive cancer cells and their ability to control tumour growth and metastasis. METHODS: BALB/c Rag2(-/-), Il2rg(-/-) mice were engrafted with CD34(+) or CD133(+) human cord blood hematopoietic stem cells (HSC) and vaccinated with human HER-2-positive cancer cells SK-OV-3 combined to human IL-12. RESULTS: Both CD34(+) or CD133(+) human HSC gave long-term engraftment and differentiation, both in peripheral blood and in lymphoid organs, and production of human antibodies. Vaccinated mice produced specific anti-HER-2 human IgG. An s.c. SK-OV-3 challenge was significantly inhibited (but not abolished) in both vaccinated and non-vaccinated HIS mice. Tumours were heavily infiltrated with human and murine cells, mice showed NK cells and production of human interferon-γ, that could contribute to tumour growth inhibition. Vaccinated HIS mice showed significantly inhibited lung metastases when compared with non-vaccinated HIS mice and to non-HIS mice, along with higher levels of tumour-infiltrating human dendritic cells. CONCLUSION: Anti-HER-2 responses were elicited through an adjuvanted allogeneic cancer cell vaccine in HIS mice. Human immune responses elicited in HIS mice effectively inhibited lung metastases.


Subject(s)
Antigens, CD34/immunology , Antigens, CD/immunology , Cancer Vaccines/immunology , Glycoproteins/immunology , Lung Neoplasms/immunology , Peptides/immunology , Receptor, ErbB-2/immunology , AC133 Antigen , Animals , Cell Line, Tumor , Disease Models, Animal , Hematopoietic Stem Cells/immunology , Humans , Immunoglobulin Isotypes/immunology , Lung Neoplasms/secondary , Lymphocytes/immunology , Mice , Mice, Inbred BALB C
5.
Oncogene ; 30(24): 2730-40, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21278796

ABSTRACT

Identification of patient selection criteria and understanding of the potential mechanisms involved in the development of resistance are crucial for an appropriate and successful design of clinical trials with anti-insulin-like growth factor (IGF)-1R therapies. Few Ewing's sarcomas are highly sensitive to IGF-1R targeting and understanding the reason why, may hold the secret to improve successful treatments. In this paper, we show that a major mechanism of resistance to highly specific inhibitors of IGF-1R, either antibodies or tyrosine kinase inhibitors may involve enhanced insulin receptor (IR)-A homodimer formation and IGF-2 production. Resistant cells are able to switch from IGF-1/IGF-1R to IGF-2/IR-A dependency to maintain sustained activation of AKT and ERK1/2, proliferation, migration and metastasis. These cells also showed higher proliferative response to insulin, in keeping with a switch towards insulin pathways sustaining proliferation and malignancy, rather than metabolism. Our findings demonstrate a role for IR-A in eliciting intrinsic and adaptive resistance to anti-IGF-1R therapies. Thus, we indicate that tumors with low IGF-1R:IR ratio are unlikely to greatly benefit from anti-IGF-1R therapies and that the efficacy of anti-IGF-1R therapies should be evaluated in relationship to the IR-A:IGF-1R ratio in cancer cells. Moreover, we provide evidences supporting IR-A as an important target in sarcoma therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, Insulin/physiology , Sarcoma, Ewing/drug therapy , Signal Transduction/physiology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Mice , Receptor, IGF Type 1/analysis , Receptor, Insulin/analysis
6.
Hum Gene Ther ; 20(5): 453-64, 2009 May.
Article in English | MEDLINE | ID: mdl-19215191

ABSTRACT

The highly aggressive cancer syndrome of female mice carrying a p53 knockout allele and a rat HER-2/neu (Neu) transgene (BALB-p53Neu) can be prevented by a cell vaccine presenting three components: Neu, interleukin (IL)-12 production, and allogeneic major histocompatibility complex (MHC) alleles (Triplex cell vaccine). Here we tested a second-generation Triplex DNA-based vaccine (Tri-DNA), consisting of the combination of three gene components (a transmembrane-extracellular domain fragment of the Neu gene, IL-12 genes, and the H-2D(q) allogeneic MHC gene), carried by separate plasmids. The Tri-DNA vaccine was at least as effective as the Triplex cell vaccine for cancer immunoprevention, giving a similar delay in the onset of mammary cancer and complete protection from salivary cancer. Both vaccines induced anti-Neu antibodies of the murine IgG2a isotype at similar levels. The Tri-DNA vaccine gave more restricted immunostimulation, consisting of a fully helper T cell type 1 (Th1)-polarized response, with effective production of interferon (IFN)-gamma in response to the vaccine but no spontaneous production, and no induction of anti-Neu IgG3 antibodies. On the other hand, the Triplex cell vaccine induced both Th1 and Th2 cytokines, a strong increase in spontaneous IFN-gamma production, and high levels of IgG3 antibodies recognizing Neu-positive syngeneic cells. In conclusion, the Tri-DNA vaccine is as effective as Triplex cell vaccine, exploiting a more restricted immune stimulation.


Subject(s)
Cancer Vaccines/immunology , Interleukin-12/immunology , Neoplastic Syndromes, Hereditary/prevention & control , Receptor, ErbB-2/genetics , Tumor Suppressor Protein p53/genetics , Vaccines, DNA/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Cytotoxicity, Immunologic , Female , Genetic Therapy , Immunoglobulin G/blood , Immunotherapy , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-12/metabolism , Major Histocompatibility Complex/immunology , Mammary Glands, Animal/immunology , Mammary Glands, Animal/pathology , Mice , Neoplastic Syndromes, Hereditary/therapy , Rats , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Salivary Glands/immunology , Salivary Glands/pathology , Transfection , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
7.
Clin Exp Med ; 9(3): 199-205, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19225718

ABSTRACT

The large use of target therapies in the treatment of gastrointestinal stromal tumors (GISTs) highlighted the urgency to integrate new molecular imaging technologies, to develop new criteria for tumor response evaluation and to reach a more comprehensive definition of the molecular target. These aspects, which come from clinical experiences, are not considered enough in preclinical research studies which aim to evaluate the efficacy of new drugs or new combination of drugs with molecular target. We developed a xenograft animal model GIST882 using nude mice. We evaluated both the molecular and functional characterization of the tumor mass. The mutational analysis of KIT receptor of the GIST882 cell lines and tumor mass showed a mutation on exon 13 that was still present after in vivo cell growth. The glucose metabolism and cell proliferation was evaluated with a small animal PET using both FDG and FLT. The experimental development of new therapies for GIST treatment requires sophisticated animal models in order to represent the tumor molecular heterogeneity already demonstrated in the clinical setting and in order to evaluate the efficacy of the treatment also considering the inhibition of tumor metabolism, and not only considering the change in size of tumors. This approach of cancer research on GISTs is crucial and essential for innovative perspectives that could cross over to other types of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Gastrointestinal Stromal Tumors/drug therapy , Animals , Glucose/metabolism , Mice , Mice, Nude , Positron-Emission Tomography , Transplantation, Heterologous , Treatment Outcome
8.
Ann Oncol ; 20(2): 213-26, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18842614

ABSTRACT

The epidermal growth factor receptor (EGFr) is one of the most studied molecules as a target for cancer therapy. Over these last few years, several studies attempting to identify predictive biomarkers of treatment response, such as the receptor status or other molecules related to the downstream signalling pathway, have been conducted. However, from a clinical point of view, the information obtained from ex vivo analyses still has various limitations that may be overcome by the combination with molecular imaging technologies which may provide a noninvasive, global, in vivo evaluation of the molecular tumour background. The aim of this review is to report the preclinical results of all positron emission tomography (PET) tracers synthesized until now for in vivo detection of EGFr in cancer. Two classes of PET compounds have been developed: labelled small molecules such as tyrosine kinase inhibitors and labelled monoclonal antibodies. The in vitro and in vivo results of these PET tracers are very different depending on the chemical properties, positron emission radionuclide, or animal models. As a consequence, various critical questions are still open, and the implications of a translation in the clinical setting for EGFr imaging in cancer patients is discussed.


Subject(s)
ErbB Receptors/analysis , Neoplasms/diagnostic imaging , Neoplasms/enzymology , Positron-Emission Tomography/methods , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Iodine Radioisotopes , K562 Cells , Mice , Protein Kinase Inhibitors/pharmacology , Radionuclide Imaging , Rats , Xenograft Model Antitumor Assays
9.
Oncogene ; 26(46): 6604-18, 2007 Oct 11.
Article in English | MEDLINE | ID: mdl-17471235

ABSTRACT

CD99 gene encodes two distinct proteins, produced by alternative splicing of CD99 gene transcript. Full-length CD99 isoform (CD99wt) is formed by an extracellular domain, followed by a transmembrane domain and a 36 amino-acid intracytoplasmic domain, which is partially deleted in the truncated, short form (CD99sh). A differential expression of these two CD99 molecules can lead to distinct functional outcomes in lymphocytes. To investigate the functional effects of CD99 molecules on malignancy, forced overexpression of the two CD99 isoforms was induced in osteosarcoma and prostate cancer cells. The two isoforms exhibited opposite functions: the major form dramatically inhibits anchorage-independent growth, anoikis resistance, migration and metastasis, whereas the CD99sh remarkably favours the phenomena. A mechanistic analysis of CD99-transfected osteosarcoma cells points to involvement of c-Src family kinase activity in regulating CD99 functions in malignancy. Ser168 residue of CD99 plays a pivotal role in the reversion of the malignant phenotype. Our findings highlight the involvement of CD99 in crucial processes of cancer malignancy, serving as a curtain raiser for this, so far neglected molecule. In addition, a dualistic role for the two CD99 isoforms was shown in agreement with what was observed for other cell adhesion molecules.


Subject(s)
Antigens, CD/physiology , Cell Adhesion Molecules/physiology , Cell Transformation, Neoplastic , Neoplasm Metastasis , Neoplasms/metabolism , Protein-Tyrosine Kinases/physiology , 12E7 Antigen , CSK Tyrosine-Protein Kinase , Gene Expression Regulation, Neoplastic , Genes, src , Humans , Male , Osteosarcoma/metabolism , Prostatic Neoplasms/metabolism , Protein Isoforms/physiology , Transfection , Tumor Cells, Cultured , src-Family Kinases
11.
Cell Immunol ; 244(2): 137-40, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17442286

ABSTRACT

We present a further, yet important, step in applying Catania Mouse Model & simulator (SimTriplex) of immune system response to vaccination. In particular we show that, as immune response can induce toxicity, one can calibrate the vaccine administrations in such a way to avoid toxicity effects, keeping immunoprevention from cancer. This result increases the model's potential applications.


Subject(s)
Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carcinoma in Situ/immunology , Immunization Schedule , Mammary Neoplasms, Animal/immunology , Models, Immunological , Vaccination/methods , Animals , Carcinoma in Situ/prevention & control , Computer Simulation , Female , Mammary Neoplasms, Animal/prevention & control , Mice , Mice, Inbred BALB C , Mice, Transgenic
12.
Transplant Proc ; 37(5): 2144-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15964362

ABSTRACT

Immunosuppressive therapies associated with organ transplantation produce an increased risk of cancer development. Malignancies are increased in transplant recipients because of the impaired immune system. Moreover, experimental data point to a tumor-promoting activity of various immunosuppressive agents. In this study, we compared the effects of 4 immunosuppressive agents with different mechanisms of action (cyclosporine, rapamycin, mycophenolic acid, and leflunomide) on the in vitro growth of various tumor cell lines and umbilical vein endothelial cells. To varying degrees rapamycin (10 ng/mL), mycophenolic acid (300 nmol/L), and leflunomide (30 micromol/L) highly inhibited the growth of human rhabdomyosarcoma, hepatocellular carcinoma, colorectal carcinoma, and endothelial cells. In contrast, cyclosporine (100 ng/mL) did not affect their growth. Our data suggest that regimens containing rapamycin, mycophenolic acid, or leflunomide, which have both immunosuppressive and antitumor activities, should be preferred in transplant recipients to minimize the risk of tumors.


Subject(s)
Antineoplastic Agents , Cyclosporine/pharmacology , Immunosuppressive Agents , Carcinoma, Hepatocellular , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms , Humans , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Isoxazoles/pharmacology , Jurkat Cells , Leflunomide , Liver Neoplasms , Mycophenolic Acid/pharmacology , Rhabdomyosarcoma , Sirolimus/pharmacology
13.
Int J Immunopathol Pharmacol ; 18(2): 351-63, 2005.
Article in English | MEDLINE | ID: mdl-15888257

ABSTRACT

Prevention of the progression of precancerous lesions by vaccines is virtually uncharted territory. Their potential, however, is being assessed in transgenic mice which develop autochthonous tumors with defined stages of progression. In this paper we show that the DNA micro-array technology significantly helps assessment of the preventive efficacy of a combined DNA and cell vaccine. All female rat Her-2/neu transgenic BALB/c (BALB-neuT) mice develop an invasive carcinoma in each of their mammary glands within 25 weeks of age. This is elicited by the activated transforming rat Her-2/neu oncogene embedded in their genome. We have previously shown that vaccination of mice bearing multiple in situ carcinomas with DNA plasmids which code for the extracellular and transmembrane domain of rat p185neu, the product of the rat Her-2/neu oncogene, followed by a boost with rat p185neu+ allogeneic cells engineered to secrete interferon-gamma, keeps 48% of mice tumor free until week 32. We have now extended our follow-up until mice reach one year of age and show that protection vanishes as time progresses. This observation suggests that the accuracy of the results studying immunotherapy against life-threatening tumors is a function of the length of the follow-up. The application of microarrays, and the concordance of morphologic and gene expression data led us to identify antibody as the main mechanism induced by vaccination. Protection is associated with a break of tolerance and a limited autoimmunity against the endogenous mouse p185neu.


Subject(s)
Autoimmunity/drug effects , Glycoproteins/genetics , Mammary Neoplasms, Experimental/prevention & control , Precancerous Conditions/therapy , Receptor, ErbB-2/genetics , Vaccines, DNA/therapeutic use , Animals , Cell Line, Tumor , Cloning, Molecular , Female , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Precancerous Conditions/immunology , Precancerous Conditions/pathology , Rats , Transgenes
14.
Curr Cancer Drug Targets ; 5(3): 221-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15892621

ABSTRACT

Prevention of cancer through the activation of the immune system has been explored in recent years in preclinical systems thanks to the availability of several new transgenic mouse models that closely mimic the natural history of human tumors. The most thoroughly investigated model of cancer immunoprevention is the mammary carcinoma of HER-2/neu transgenic mouse. In this system it has clearly been shown that the activation of immune defences in healthy individuals can effectively prevent the subsequent onset of highly aggressive mammary carcinomas. A complete prevention was obtained using a combination of three signals (the so called "triplex" vaccine) that included the specific antigen (p185, the product of HER-2/neu) and nonspecific signals like allogeneic histocompatibility antigens and interleukin 12. The analysis of protective immune responses in models of cancer immunoprevention revealed some unexpected features, in particular the central role of antibodies in immunoprevention, at variance with conventional immuno-therapy which is firmly based on cytotoxic T cells. In the HER-2/neu system anti-p185 antibodies, in addition to immunological functions leading to tumor cell lysis, inhibit p185 dimerization and induce its internalization, resulting in the inhibition of mitogenic signaling. Most current tumor antigens appear to be unsuitable targets for cancer immunoprevention. An ideal antigen should have a crucial pathogenetic role in tumor growth to avoid the selection of antigen loss variants. Downregulation of major histocompatibility complex (MHC) expression during tumor progression frequently limits antigen recognition by MHC-restricted T cells. Thus an ideal antigen for cancer immunoprevention should be recognized both by T cells and by antibodies. Antibody binding to cell surface oncogenic determinants, in addition to complement- and cell-mediated tumor cell lysis, can block mitogenic signaling and induce internalization, resulting in tumor growth arrest. A search for new tumor antigens should be conducted among molecules that are directly involved in neoplastic transformation and are recognizable by the immune response also in MHC loss variants. Novel tumor antigens fulfilling both conditions will be crucial for the development of cancer immunoprevention and will provide new targets also for cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Neoplasms/prevention & control , Animals , Humans , Mice , Mice, Transgenic
15.
Bioinformatics ; 21(12): 2891-7, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15817697

ABSTRACT

UNLABELLED: We present an in silico model that simulates the immune system responses to tumor cells in naive and vaccinated mice. We have demonstrated the ability of this model to accurately reproduce the experimental results. MOTIVATION: In vivo experiments on HER-2/neu mice have shown the effectiveness of Triplex vaccine in the protection of mice from mammary carcinoma. Full protection was conferred using chronic (prophylactic) vaccination protocol while therapeutic vaccination was less efficient. Our in silico model was able to closely reproduce the effects of various vaccination protocols. This model is the first step towards the development of in silico experiments searching for optimal vaccination protocols. RESULTS: In silico experiments carried out on two large statistical samples of virtual mice showed very good agreements with in vivo experiments for all experimental vaccination protocols. They also show, as supported by in vivo experiments, that the humoral response is fundamental in controlling the tumor growth and therefore suggest the selection and timing of experiments for measuring the activity of T cells. CONTACT: francesco@dmi.unict.it SUPPLEMENTARY INFORMATION: http://www.dmi.unict.it/CIG/suppdata_bioinf.html.


Subject(s)
Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Drug Therapy, Computer-Assisted/methods , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Models, Biological , Vaccination/methods , Animals , Computer Simulation , Dose-Response Relationship, Drug , Mice , Treatment Outcome
16.
Scand J Immunol ; 55(5): 484-92, 2002 May.
Article in English | MEDLINE | ID: mdl-11975760

ABSTRACT

Cytokine-immunoglobulin (Ig)-fusion proteins have attracted increasing interest as antitumour agents. Here, we have investigated the antimetastatic and antitumour responses elicited in vivo by mammary adenocarcinoma cells (TS/A) engineered to secrete interleukin (IL)-2-IgG fusion proteins. TS/A cells were transfected with DNA coding for IL-2-IgG2b, IgG2b or IL-2, and injected subcutaneously into syngeneic mice. Animals injected with TS/A-IL-2 or TS/A-IL-2-IgG2b both efficiently rejected tumours, whereas treatment with parental cells or TS/A-IgG2b was lethal. Interestingly, only mice vaccinated with IL-2-IgG2b fusion protein-secreting cells showed a long-lasting protective immunity against a later challenge with parental tumour cells. Moreover, the metastatic potential of TS/A-IL-2-IgG2b-transfected cells was dramatically decreased compared with TS/A-IL-2-cells, with a virtual absence of lung metastases after intravenous injection. Adenocarcinomas secreting IL-2-IgG2b exhibited a more prominent, early and persistent infiltration of CD4+, CD8+ and natural killer (NK) cells than TS/A-IL-2 cells. Therefore, upon transfection into adenocarcinoma cells, the IgG2b part of IL-2 fusion protein exerts intriguing added antitumour properties over IL-2 alone, thus contributing to a long-lasting tumour immunity, probably by the recruitment of specific immune effector cells. These findings suggest a promising new oncotherapeutic strategy for poorly immunogenic tumours: vaccination with tumour cells engineered to secrete IL-2-IgG2b fusion protein.


Subject(s)
Adenocarcinoma/immunology , Immunoglobulin G/immunology , Interleukin-2/immunology , Mammary Neoplasms, Experimental/immunology , Recombinant Fusion Proteins/immunology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Growth Inhibitors/genetics , Growth Inhibitors/immunology , Immunoglobulin G/genetics , Immunohistochemistry , Immunotherapy , Interleukin-2/genetics , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Transfection , Tumor Cells, Cultured
17.
J Exp Med ; 194(9): 1195-205, 2001 Nov 05.
Article in English | MEDLINE | ID: mdl-11696586

ABSTRACT

Transgenic Balb/c mice expressing the transforming rat HER-2/neu oncogene develop early and multifocal mammary carcinomas. Within the first 5 months of life the tissue-specific expression of HER-2/neu causes a progression in all their 10 mammary glands from atypical hyperplasia to invasive carcinoma. It was previously observed that chronic administration of interleukin (IL)-12 increased tumor latency, but every mouse eventually succumbed to multiple carcinomas. A significant improvement in tumor prevention was sought by administering allogeneic mammary carcinoma cells expressing HER-2/neu combined with systemic IL-12. This treatment reduced tumor incidence by 90% and more than doubled mouse lifetime. For the maximum prevention p185(neu) antigen must be expressed by allogeneic cells. IL-12 treatment strongly increased the cell vaccine efficacy. The mammary glands of mice receiving the combined treatment displayed a markedly reduced epithelial cell proliferation, angiogenesis, and HER-2/neu expression, while the few hyperplastic foci were heavily infiltrated by granulocytes, macrophages, and CD8(+) lymphocytes. Specific anti-HER-2/neu antibodies were produced and a nonpolarized activation of CD4(+) and CD8(+) cells secreting IL-4 and interferon (IFN)-gamma were evident. A central role for IFN-gamma in the preventive effect was proven by the lack of efficacy of vaccination in IFN-gamma gene knockout HER-2/neu transgenic Balb/c mice. A possible requirement for IFN-gamma is related to its effect on antibody production, in particular on IgG2a and IgG2b subclasses, that were not induced in IFN-gamma knockout HER-2/neu mice. In conclusion, our data show that an allogeneic HER-2/neu-expressing cell vaccine combined with IL-12 systemic treatment can prevent the onset of genetically determined tumors.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Interleukin-12/immunology , Mammary Neoplasms, Experimental/prevention & control , Receptor, ErbB-2/physiology , Animals , Breast/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Transplantation , Female , Immunity, Cellular/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/administration & dosage , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Rats , Receptor, ErbB-2/genetics , Transplantation, Homologous , Tumor Cells, Cultured , Vaccination/methods
18.
Gene ; 274(1-2): 139-49, 2001 Aug 22.
Article in English | MEDLINE | ID: mdl-11675006

ABSTRACT

Rhabdomyosarcoma is a soft tissue tumor committed to the myogenic lineage but arrested prior to terminal differentiation. To identify new genes implicated in the block in myogenic differentiation of rhabdomyosarcoma cells and those responsible for their proceeding along the myogenic pathway we used cDNA microarrays to compare gene expression profiles of two clones of the human embryonal rhabdomyosarcoma cell line RD with different myogenic potentials: RD/12, which is unable to differentiate, and RD/18, which shows elements able to terminally differentiate, as defined by the expression of myosin heavy chain (up to 50% of the population) and the formation of multinucleated myotube-like structures. We identified 80 genes differentially expressed by the two clones. Differentiating RD/18 cells overexpressed the myogenic transcription factor myogenin along with known myogenic markers; myogenin transfection into undifferentiated RD/12 cells was able to revert the phenotype giving rise to 94% of clones displaying a differentiated morphology. RD/18 cells also expressed several genes not known to be expressed in rhabdomyosarcoma or muscle cells, such as pigment-epithelium derived factor and endothelin-3. Poorly differentiated RD/12 cells, along with genes related to mesenchymal lineage or early myogenic commitment, also expressed genes not previously known to be related to the differentiation block of human rhabdomyosarcoma, such as monocyte chemotactic protein-1, connective tissue growth factor and insulin-like growth factor binding protein-5. Differential expression of these genes in a time course of differentiation suggested their potential roles as either new myogenic markers or repressors of differentiation. These results identify a cluster of new genes related to the aberrant myogenic differentiation program of human rhabdomyosarcoma cells.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins , Muscles/metabolism , Rhabdomyosarcoma/genetics , Chemokine CCL2/genetics , Connective Tissue Growth Factor , Dose-Response Relationship, Drug , Endothelin-3/genetics , Gene Expression Regulation, Neoplastic/drug effects , Growth Substances/genetics , Humans , Immediate-Early Proteins/genetics , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Muscles/cytology , Myogenin/genetics , Myosins/genetics , Oligonucleotide Array Sequence Analysis , Rhabdomyosarcoma/pathology , Time Factors , Tumor Cells, Cultured
19.
Eur J Cancer ; 37(13): 1719-25, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11527701

ABSTRACT

The expression and biological function of Nerve Growth Factor (NGF) receptors was studied in a panel of rhabdomyosarcoma cell lines derived from embryonal and alveolar histotype. All the cell lines expressed both the high affinity receptor TrkA and the low affinity receptor p75(NTR). Treatment with exogenous NGF did not considerably alter rhabdomyosarcoma cell growth or differentiation, but significantly inhibited spontaneous apoptosis as well as apoptosis, and induced by serum starvation or apoptosis induced by treatment with cycloheximide (CHX). Rhabdomyosarcoma cell lines expressed NGF and other neurotrophins and trace amounts of NGF protein were found in the supernatants of rhabdomyosarcoma cell cultures. Blocking the putative autocrine loop with an anti-NGF antibody resulted in an increase in apoptosis compared with control cultures. These data suggest that the simultaneous presence of both high and low affinity NGF receptors engaged by endogenous or exogenous NGF might contribute to the escape from apoptosis exhibited by the rhabdomyosarcoma cells.


Subject(s)
Receptor, Nerve Growth Factor/physiology , Rhabdomyosarcoma/pathology , Apoptosis/physiology , Humans , Neoplasm Proteins/metabolism , Nerve Growth Factors/metabolism , RNA, Messenger/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Rhabdomyosarcoma/metabolism , Tumor Cells, Cultured
20.
Clin Cancer Res ; 7(6): 1790-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11410521

ABSTRACT

Innovative treatment modalities are needed for Ewing's sarcoma (ES), a neoplasm with a disappointingly low survival rate despite the use of aggressive multimodal therapeutic approaches. We and others (D. Yee et al., J. Clin. Investig., 86: 1806-1814, 1990; K. Scotlandi et al., Cancer Res., 56: 4570-4574, 1996) have previously shown the existence and the pathogenetic relevance of an autocrine loop, mediated by the insulin-like growth factor-I receptor (IGF-IR), which is crucial for survival and proliferation of ES cells in vitro. Moreover, we reported that the IGF-IR-blocking monoclonal antibody (MAb), alphaIR3, as well as suramin, a drug that can interfere with growth factor by binding to the receptors, inhibited both the tumorigenic and the metastatic ability of ES cells in athymic mice. In this study, we analyzed whether agents that can block the IGF-IR-mediated loop are of value in association with conventional cytotoxic drugs for the design of more effective therapeutic regimens. Both alphaIR3 MAb and suramin treatment significantly increased the antitumor in vitro effects of doxorubicin and vincristine, two drugs with a leader action on ES. These findings were obtained by both simultaneous and sequential treatments. Analysis of the proliferation rate and of apoptosis revealed that alphaIR3 MAb and suramin significantly enhanced the G(1)-phase rate induced by doxorubicin, without substantially affecting doxorubicin-G(2)-M-blockage of cell cycle, and significantly increased the induction of apoptosis, which confirmed that the specific blockage of IGF-IR deprives ES cells of an important tool for the prevention of drug-induced apoptosis. Moreover, combination treatments of doxorubicin plus alphaIR3 MAb significantly increase the doxorubicin-induced impairment of the ability of ES cells to form colonies in soft agar. In conclusion, we showed that, in ES, the blockage of IGF-IR by a neutralizing MAb or by suramin may greatly potentiate the antitumor activity of conventional chemotherapeutic drugs.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Doxorubicin/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Vincristine/pharmacology , Antibodies, Monoclonal/metabolism , Apoptosis , Bromodeoxyuridine/metabolism , Cell Cycle , Cell Division , Cell Nucleus/drug effects , Dose-Response Relationship, Drug , Humans , Suramin/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL