Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
1.
Ann Oncol ; 26(4): 798-803, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25538176

ABSTRACT

BACKGROUND: In the NIBIT-M1 study, we reported a promising activity of ipilimumab combined with fotemustine in metastatic melanoma (MM) patients with or without brain metastases. To corroborate these initial findings, we now investigated the long-term efficacy of this combination. PATIENTS AND METHODS: This analysis captured the 3-year outcome of MM patients who received ipilimumab combined with fotemustine as first- or second-line treatment. Median overall survival (OS), 3-year survival rates, immune-related (ir) progression-free survival (irPFS), brain PFS, and ir duration of response (irDOR) for the entire population and for patients with brain metastases were assessed. Clinical results were correlated with circulating CD3(+)CD4(+)ICOS(+)CD45RO(+) or CD45RA(+) T cells, neutrophil/lymphocyte (N/L) ratios, and tumorBRAF-V600 mutational status. RESULTS: Eighty-six MM patients, including 20 with asymptomatic brain metastases that had been pre-treated with radiotherapy in 7 subjects, were enrolled in the study. With a median follow-up of 39.9 months, median OS and 3-year survival rates were 12.9 months [95% confidence interval (CI) 7.1-18.7 months] and 28.5% for the whole study population, and 12.7 months (95% CI 2.7-22.7 months) and 27.8% for patients with brain metastases, respectively. Long-term ir adverse events consisting of G1 rush and pruritus occurred in 21% of patients. The absolute increase from baseline to week 12 in 'memory' but not in 'naïve' T cells identified patients with a better survival (P = 0.002). The N/L ratio correlated with a significantly better survival at early time points. BRAF status did not correlate with clinical outcome. CONCLUSIONS: Long-term analysis of the NIBIT-M1 trial continues to demonstrate efficacy of ipilimumab combined with fotemustine in MM patients. Fotemustine does not seem to impair the immunologic activity of ipilimumab. EUDRACT NUMBER: 2010-019356-50. CINICALTRIALSGOV: NCT01654692.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Therapy/mortality , Brain Neoplasms/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Female , Follow-Up Studies , Humans , Ipilimumab , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Neoplasm Staging , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Prognosis , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Rate
2.
Ann Oncol ; 24(11): 2911-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24067719

ABSTRACT

BACKGROUND: Patients with advanced uveal melanoma have a poor prognosis and limited treatment options. Ipilimumab is approved for pre-treated adult patients with advanced melanoma. However, because previous clinical trials with ipilimumab have excluded patients with uveal melanoma, data in this patient population are limited. PATIENTS AND METHODS: Pre-treated patients with advanced uveal melanoma received ipilimumab 3 mg/kg through an expanded access programme, every 3 weeks for four doses. Tumour assessments were conducted at baseline and after completion of treatment and patients were monitored throughout for adverse events. RESULTS: Among 82 assessable patients, 4 (5%) had an immune-related objective response and 24 (29%) had immune-related stable disease lasting ≥3 months for an immune-related disease control rate of 34%. With a median follow-up of 5.6 months, median overall survival (OS) was 6.0 months and median progression-free survival (PFS) was 3.6 months. The 1-year rates of OS and PFS were 31% and 11%, respectively. The safety profile of ipilimumab was similar to that in patients with cutaneous melanoma. CONCLUSIONS: These data suggest ipilimumab 3 mg/kg is a feasible option in pre-treated patients with metastatic uveal melanoma. Evidence of disease control and a 1-year survival rate of 31% indicate the need for further investigation in randomised, controlled trials to determine the optimal timing and use of ipilimumab in this patient population.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Uveal Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Disease-Free Survival , Female , Humans , Ipilimumab , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Treatment Outcome , Uveal Neoplasms/mortality , Uveal Neoplasms/pathology
3.
Expert Opin Biol Ther ; 9(8): 1043-55, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19591629

ABSTRACT

Since the identification of tumor associated antigens (TAA) in different tumor histotypes, many vaccination strategies have been investigated, including peptide-based vaccines. Results from the first decade of clinical experimentation, though demonstrating the feasibility and the good toxicity profile of this approach, provided evidence of clinical activity only in a minority of patients, despite inducing immunization in up to 50% of them. In this review, we discuss the different approaches recently developed in order to induce stronger peptide-induced immune-mediated tumor growth control, possibly translating into improved clinical response rates, with specific focus on multipeptide-based anti-cancer vaccines. This strategy offers many advantages, such as the possibility of bypassing tumor heterogeneity and selection of antigen (Ag)-negative clones escaping peptide-specific immune responses, or combining HLA class I- and class II-restricted epitopes, thus eliciting both CD4- and CD8-mediated immune recognition. Notably, advances in Ag discovery technologies permit further optimization of peptide selection, in terms of identification of tumor-specific and unique TAA as well as Ags derived from different tumor microenvironment cell components. With the ultimate goal of combining peptide selection with patient-specific immunogenic profile, peptide based anti-cancer vaccines remain a promising treatment for cancer patients, as attested by of pre-clinical and clinical studies.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasms/therapy , Vaccines, Subunit/therapeutic use , Cancer Vaccines/immunology , Clinical Trials as Topic , Epitopes/chemistry , Female , Histocompatibility Antigens Class II/chemistry , Humans , Male , Medical Oncology/trends , Melanoma/therapy , Peptides/chemistry , Prostatic Neoplasms/therapy , Skin Neoplasms/therapy , Treatment Outcome , Vaccines, Subunit/immunology
4.
Cell Death Differ ; 15(1): 80-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17932500

ABSTRACT

Tumour cells release vesicular structures, defined as microvesicles or exosomes, carrying a large array of proteins from their originating cell. The expression of antigenic molecules recognized by T cells has originally suggested a role for these organelles as a cell-free antigen source for anticancer vaccines. However, recent evidence shows that tumour exosomes may also exert a broad array of detrimental effects on the immune system, ranging from apoptosis in activated antitumour T cells to impairment of monocyte differentiation into dendritic cells and induction of myeloid suppressive cells. Immunosuppressive exosomes of tumour origin can be found in neoplastic lesions and sera from cancer patients, implying a potential role of this pathway in in vivo tumour progression. Through the expression of molecules involved in angiogenesis promotion, stromal remodelling, delivery of signalling pathways through growth factor/receptor transfer, chemoresistance and genetic intercellular exchange, tumour exosomes could represent a versatile tool for moulding host environment. Hence, their secretion by neoplastic cells may in the future become a novel pathway to target for therapeutic intervention in cancer patients.


Subject(s)
Cytoplasmic Vesicles/physiology , Neoplasms/immunology , Signal Transduction , Antigens, Neoplasm/immunology , Cytokines/immunology , Cytokines/metabolism , Cytoplasmic Vesicles/immunology , Dendritic Cells/immunology , Humans , Immunotherapy , Neoplasms/pathology , Neoplasms/physiopathology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology
5.
Ann Oncol ; 18(2): 226-32, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17116643

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been and is still widely used as an adjuvant in clinical trials of vaccination with autologous tumor cells, peptides and/or dendritic cells in a variety of human neoplasms. This cytokine was administered either as product of gene-transduced tumor cells or as recombinant protein together with the vaccine given subcutaneously or intradermally. Results of these trials were heterogeneous in terms of induction of vaccine-specific immune response and of clinical response. Though in some of these studies GM-CSF appeared to help in generating an immune response, in others no effect or even a suppressive effect was reported. Here, we review the literature dealing with the immune adjuvant activity of GM-CSF both in animal models and clinical trials. As a consequence of such analysis, we conclude that GM-CSF may increase the vaccine-induced immune response when administered repeatedly at relatively low doses (range 40-80 microg for 1-5 days) whereas an opposite effect was often reported at dosages of 100-500 microg. The potential mechanisms of the GM-CSF-mediated immune suppression are discussed at the light of studies describing the activation and expansion of myeloid suppressor cells by endogenous tumor-derived or exogenous GM-CSF.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Neoplasms/immunology , Animals , Humans , Neoplasms/therapy
6.
Oncogene ; 25(24): 3357-64, 2006 Jun 08.
Article in English | MEDLINE | ID: mdl-16462768

ABSTRACT

Activating BRAF or NRAS mutations have been found in 80% of human sporadic melanomas, but only one of these genetic alterations could be detected in each tumour. This suggests that BRAF and NRAS 'double mutants' may not provide advantage for tumour growth, or may even be selected against during tumorigenesis. However, by applying mutant-allele-specific-amplification-PCR method to short-term melanoma lines, one out of 14 tumours was found to harbour both BRAFV600E and the activating NRASQ61R mutations. On the other hand, analysis of 21 melanoma clones isolated by growth in soft agar from this tumour indicated that 16/21 clones harboured a BRAFV600E, but were wild-type for NRAS, whereas the remaining had the opposite genotype (NRASQ61R/wild-type BRAF). When compared to BRAFV600E clones, NRASQ61R clones displayed reduced growth in soft agar, but higher proliferative ability in vitro in liquid medium and even in vivo after grafting into SCID/SCID mice. These data suggest that NRAS and BRAF activating mutations can coexist in the same melanoma, but are mutually exclusive at the single-cell level. Moreover, the presence of NRASQ61R or BRAFV600E is associated with distinct in vitro and in vivo growth properties of neoplastic cells.


Subject(s)
Genes, ras , Melanoma/genetics , Melanoma/metabolism , Mutation , Proto-Oncogene Proteins B-raf/genetics , Animals , Base Sequence , Cell Line, Tumor , Female , Genotype , Humans , Mice , Mice, SCID , Molecular Sequence Data , Neoplasm Transplantation
7.
Ann Oncol ; 14(6): 817-24, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12796017

ABSTRACT

The molecular characterisation of human tumour antigens recognised by T cells has provided new impetus for immunisation of patients bearing tumours expressing well-defined antigens. After evaluating the immunogenicity of the new, molecularly characterised antigens in vitro, several clinical studies were conducted to assess the in vivo immunogenicity and the clinical efficacy of vaccines including these antigens. The findings generated by trials based on the administration of peptides or DNA-encoding antigens are discussed to highlight the limits of this therapeutic approach; however, this approach has resulted in some complete and durable regressions, although still in a unsatisfactory small number of cases (5-25%). The recent use of dendritic cells loaded ex vivo with tumour antigens suggests that a high frequency of tumour-specific immune responses can be achieved. Possible means of overcoming the clinical limits and improving the outcome of previous studies are also discussed.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasms/drug therapy , Vaccination , Animals , Clinical Trials as Topic , Humans , Neoplasms/immunology
8.
Tissue Antigens ; 59(4): 273-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12135425

ABSTRACT

Melanoma-associated peptides recognized by cytolytic T lymphocytes (CTL) in the context of several histocompatibility leukocyte antigens (HLA) are required for the development of specific immunotherapies. Using a transient transfection assay into COS-7 cells, we identified the gp100/pMel17 melanosomal protein as the shared antigen recognized by three independent CD8+ CTL clones in HLA-A*6801-restricted fashion. This finding was confirmed by the correlation between lack of gp100/pMel17 protein in a number of HLA-A*6801-positive melanomas and their resistance to lysis/cytokine production by the specific effectors. The gp100/pMel17 antigenic epitope was identified based on recognition of subfragments and on a computer-based prediction algorithm. Among a panel of gp100/pMel17-derived synthetic peptides only the 10-mer HTMEVTVYHR (gp100/pMel17182-191) induced tumor necrosis factor (TNF) release by CTL clones when pulsed on suitable target cells whereas both the 10-mer and the shorter 9-mer gp100/pMel17183-191 sensitized the same antigen-pulsed cells to lysis. In conclusion, the identification of the HTMEVTVYHR peptide will extend to HLA-A*6801 melanoma patients the possibility to exploit gp100/pMel17 melanosomal protein for experimental and clinical studies.


Subject(s)
HLA-A Antigens/immunology , Melanoma , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Skin Neoplasms , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antigen Presentation , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , COS Cells , Clone Cells , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Tumor Cells, Cultured , gp100 Melanoma Antigen
9.
Cancer Res ; 61(22): 8218-26, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719453

ABSTRACT

In this study, we investigated whether maturation of monocyte-derived myeloid dendritic cells (DCs) is differentially affected by the uptake of dying human melanoma cells in distinct phases of apoptosis. Maturation of monocyte-derived DCs, as documented by phenotype analysis and T-cell immunostimulatory activity, was inhibited by phagocytosis of dying melanoma cells containing a large fraction of cells in early apoptosis (Annexin-V+ and propidium iodide-) but promoted by the same tumors when in late apoptosis/secondary necrosis (Annexin-V+ and propidium iodide+) or when dying by primary necrosis. These opposite effects on DC maturation were observed after the uptake of early or late apoptotic cells from most vertical growth phase primary tumors and all metastases but not after the uptake of dying cells from a radial growth phase primary tumor or normal adult melanocytes. Inhibition of DC maturation by early apoptotic melanoma cells correlated with expression of interleukin-10 in neoplastic cells and was prevented by preincubating the tumor cells with a neutralizing antibody to interleukin-10 before tumor uptake by DCs. Cross-presentation of the melanoma-associated antigen gp100(209-217) to peptide-specific CTLs by HLA-A*0201+ DCs was achieved 48-72 h after phagocytosis of HLA-A*0201- melanoma cells in apoptosis, or primary necrosis, but only when tumor necrosis factor-alpha was added to DCs 4 h after the initiation of tumor phagocytosis. These results suggest that phases of apoptosis and neoplastic transformation affect maturation of myeloid DCs that take up dying cells of the melanocyte lineage. However, neoplastic cells in late apoptosis, or even in primary necrosis, induce only a partial DC differentiation not sufficient to achieve cross-presentation of tumor antigens to CTLs unless further DC maturation is promoted by additional signals. These results suggest a novel mechanism of tumor escape that may prevent the development of antitumor immunity through the maturation block induced in DCs by neoplastic cells in the early phase of apoptosis.


Subject(s)
Apoptosis/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Melanocytes/immunology , Melanoma/immunology , Adult , Antigen Presentation , Antigens, Neoplasm/immunology , Apoptosis/radiation effects , Cell Communication/immunology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Dendritic Cells/cytology , Humans , Interleukin-10/immunology , Melanocytes/cytology , Melanoma/pathology , Necrosis , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Ultraviolet Rays
10.
Br J Cancer ; 85(6): 836-44, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11556834

ABSTRACT

Physical interaction between CDKN2A/p16 and CDK4 proteins regulates the cell cycle progression through the G1 phase and dysfunction of these proteins by gene mutation is implicated in genetic predisposition to melanoma. We analysed 15 Italian melanoma families for germ line mutations in the coding region of the CDKN2A gene and exon 2 of the CDK4 gene. One novel disease-associated mutation (P48T), 3 known pathological mutations (R24P, G101W and N71S) and 2 common polymorphisms (A148T and Nt500 G>C) were identified in the CDKN2A gene. In a family harbouring the R24P mutation, an intronic variant (IVS1, +37 G>C) of uncertain significance was detected in a non-carrier melanoma case. The overall incidence of CDKN2A mutations was 33.3%, but this percentage was higher in families with 3 or more melanoma cases (50%) than in those with only 2 affected relatives (25%). Noteworthy, functional analysis established that the novel mutated protein, while being impaired in cell growth and inhibition assays, retains some in vitro binding to CDK4/6. No variant in the p16-binding region of CDK4 was identified in our families. Our results, obtained in a heterogeneous group of families, support the view that inactivating mutations of CDKN2A contribute to melanoma susceptibility more than activating mutations of CDK4 and that other genetic factors must be responsible for melanoma clustering in a high proportion of families. In addition, they indicate the need for a combination of functional assays to determine the pathogenetic nature of new CDKN2A mutations.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinases/genetics , Germ-Line Mutation , Melanoma/genetics , Proto-Oncogene Proteins , Skin Neoplasms/genetics , Adult , Aged , Cell Cycle , Cell Division , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinases/metabolism , DNA Mutational Analysis , DNA, Neoplasm/analysis , Disease Susceptibility , Exons , Female , G1 Phase , Genetic Linkage , Genetic Testing , Humans , Italy/epidemiology , Male , Middle Aged , Pedigree , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Transcription, Genetic
11.
Cancer Gene Ther ; 8(7): 491-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11498770

ABSTRACT

The effect of local and systemic delivery of the angiostatin gene on human melanoma growth was studied in nude mice. Liposome-coated plasmids carrying the cDNA coding for murine and human angiostatin (CMVang and BSHang) were injected weekly, locally or systemically, in mice transplanted with melanoma cells. The treatment reduced melanoma growth by 50% to 90% compared to that occurring in control animals treated with liposome-coated plasmid carrying the lacZ gene or in untreated controls. The growth of both locally injected and controlateral uninjected tumors in mice bearing two melanoma grafts was significantly suppressed after intratumoral treatment. Tumor growth inhibition was also observed in mice treated by intraperitoneal delivery, suggesting that angiostatin gene therapy acts through a systemic effect. Both melanoma growth suppression and delay in the onset of tumor growth were observed in treated mice. PCR performed on tumors and normal tissues showed that the lipofected DNA was present in tissues from treated mice, and angiostatin expression was demonstrated by RT-PCR. Histopathological analysis of melanoma nodules revealed an increase in apoptotic cells and a reduction in vessel density in tumors from treated mice. Our results suggest that systemic, liposome-mediated administration of genes coding for antiangiogenic factors represents a promising strategy for melanoma treatment in humans.


Subject(s)
Genetic Therapy/methods , Melanoma/therapy , Peptide Fragments/genetics , Plasminogen/genetics , Angiostatins , Animals , Cell Division , DNA/metabolism , DNA, Complementary/metabolism , Humans , Lac Operon/genetics , Liposomes/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic , Peptide Fragments/biosynthesis , Plasminogen/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured
12.
Cancer Immunol Immunother ; 50(4): 199-211, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11459172

ABSTRACT

In order to construct an immunogenic cellular vaccine, we transduced three HLA-A*0201 human melanoma lines, selected for expression of classes I and II HLA, adhesion molecules and the T cell-defined melanoma antigens Melan/MART-1, gp100 and tyrosinase, with both interleukin-2 (IL-2) and B7-1 genes by the use of a polycistronic retroviral vector. The lines were selected to share only the HLA-A*0201 allele to avoid generation of strong alloreactivity in case of their multiple in vivo use in HLA-A*0201 + patients. Phenotypic and functional analysis of B7-1-IL2 transduced melanoma lines in comparison with B7-1 transduced and/or parental untransduced counterparts were then carried out. Tumor cells expressing either B7-1 or both genes did not change their original antigenic profile. From a functional point of view, expression of both genes in melanoma lines: (1) improved the response of anti-melanoma cytotoxic T lymphocytes (CTL) over singly transduced or untransduced melanoma cells when subthreshold levels of MHC-peptide complexes were expressed by melanoma cells; (2) conferred a distinct advantage in the ability to stimulate cytotoxicity and interferon-gamma release by autologous and/or HLA-A*0201-compatible allogeneic lymphocytes; (3) allowed the generation of a high number of specific CTL by in vitro stimulation of lymphocytes of HLA-A*0201-melanoma patients. Thus, B7-IL2 gene-transduced melanoma lines appear to display a high immunogenicity and could be used as vaccine in melanoma patients.


Subject(s)
B7-1 Antigen/genetics , Cancer Vaccines/immunology , Interleukin-2/genetics , Lymphocyte Activation , Melanoma/immunology , Transduction, Genetic , B7-1 Antigen/metabolism , Cytotoxicity, Immunologic , Gene Expression , Genetic Therapy , HLA Antigens/metabolism , HLA-A2 Antigen/metabolism , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-2/metabolism , Melanoma/genetics , T-Lymphocytes, Cytotoxic/immunology
13.
Keio J Med ; 50(2): 86-90, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11450597

ABSTRACT

During the last 10 years many melanoma antigens recognized by T cells have been molecularly characterized. This review summarizes the main features of these antigens, including both classes I and II HLA-restricted peptides, and describes their classification into diverse groups according to the tissue distribution of the antigens. The different in vitro and in vivo immunogenicity of such antigens is then discussed leading to the conclusion that Melan-A/MART-1 is the strongest among those tested being frequently recognized by patients' T cells both in vitro and in vivo. However, no correlation was found between T-cell response of melanoma patients to Melan-A/MART-1 and clinical response when it was used for vaccination. Data are also presented that suggest, through an ex vivo analysis carried out with tetramers staining of melanoma-specific T cells, that only in a limited number of advanced patients does a specific immune response develop. This response, however, appears unable to effectively counteract metastatic melanoma growth.


Subject(s)
Antigens, Neoplasm , Melanoma/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/classification , Cancer Vaccines/pharmacology , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , In Vitro Techniques , Melanoma/therapy
15.
Cancer Res ; 61(1): 222-7, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196165

ABSTRACT

Members of the heat shock protein 70 (HSP70) family display a broad cellular localization and thus bind a repertoire of chaperoned peptides potentially derived from proteins of different cellular compartments. In this report, we show that HSP70 purified from human melanoma can activate T cells recognizing melanoma differentiation antigens in an antigen- and HLA class I-dependent fashion. HLA class I-restricted anti-melanoma T cells were susceptible to MHC-restricted, HSP70-dependent stimulation, indicating that HSP70 complexed peptides were able to gain access to the class I HLA presentation pathway. In addition, MHC matching between the melanoma cells used as a source of HSP and the responding T cells were not required, indicating that HSP70 activation may occur across MHC barriers. Besides the MHC-restricted and peptide-dependent activation pathway, HSP70 with no endogenous complexed peptides or HSP70 purified from antigen-negative cells was also able to induce IFN-gamma release by antimelanoma T cells by a MHC-independent mechanism. In this case, however, higher doses of HSP70 were required. The capacity to activate class I-restricted, antitumor T cells as well as antigen-presenting cells, together with the finding that the HSP70 chaperoned peptide repertoire includes melanoma-shared epitopes, holds promise for a HSP70-based cancer vaccine.


Subject(s)
HSP70 Heat-Shock Proteins/immunology , Melanoma/immunology , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Cross Reactions , Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , HSP70 Heat-Shock Proteins/isolation & purification , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/pharmacology , Histocompatibility Antigens Class I/immunology , Humans , Lymphocyte Activation/immunology , Melanoma/chemistry , Molecular Chaperones/metabolism , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology
16.
J Clin Oncol ; 19(2): 425-31, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11208835

ABSTRACT

PURPOSE: Because interferon gave promising results in the management of metastatic renal cell carcinoma in the 1980s, a multicentric randomized controlled trial was planned to compare adjuvant recombinant interferon alfa-2b (rIFNalpha2b) with observation after radical nephrectomy in patients with Robson stages II and III renal cell carcinoma. Overall and event-free survival were to be evaluated together with prognostic factors. PATIENTS AND METHODS: Overall and event-free survival curves for 247 patients (124 controls and 123 treated) were estimated by the Kaplan-Meier method and compared using the log-rank test. Cox's multiple regression models were adopted to perform a joint analysis of treatment and prognostic factors. RESULTS: The 5-year overall and event-free survival probabilities were 0.665 and 0.671, respectively, for controls and 0.660 and 0.567, respectively, for the treated group; the differences were not statistically significant (2P = .861 for overall and 2P = .107 for event-free survival with the log-rank test). Regarding prognostic factors, only grade, pT, and pN demonstrated a significant prognostic role. First-order interactions of treatment with pT and pN category were investigated; a significant interaction was found between pN and treatment. A harmful effect of rIFNalpha2b in the 97 treated pN0 patients and a protective effect in the 13 treated pN2/pN3 patients were statistically significant. CONCLUSION: Adjuvant rIFNalpha2b is not indicated after radical nephrectomy for renal cell carcinoma. The protective effect in the small group of pN2/pN3 patients requires further investigation.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Interferon-alpha/therapeutic use , Kidney Neoplasms/drug therapy , Adult , Aged , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/surgery , Chemotherapy, Adjuvant , Female , Humans , Interferon alpha-2 , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Male , Middle Aged , Neoplasm Staging , Nephrectomy , Prognosis , Proportional Hazards Models , Recombinant Proteins , Survival Analysis
19.
Methods Mol Med ; 61: 203-22, 2001.
Article in English | MEDLINE | ID: mdl-22323260

ABSTRACT

The identification of genes involved in different biologic functions and in the pathogenesis of diseases has paved the way to the possibility of either interfering with the role of such genes or replacing them in somatic cells in case of loss, which may occur in some genetic diseases or cancer. Such progress has been accomplished thanks to advances in molecular biology and applied technology that allow the transport and insertion of genes into recipient cells by viral or physical vectors as well as the inhibition of gene transcription by antisense oligonucleotides. Methods have also been devised to transfer genes not only in vitro but also in vivo, although this latter approach is still limited owing to poor selectivity and targeting of most vectors when given systemically. Viral and physical vectors have been employed; each of these vectors has distinct advantages and disadvantages, and, therefore, the appropriate vector should be selected according to the therapeutic system involved (1). Retro viral vectors have been used largely for their ability to selectively transfect proliferating cells, a feature that can be advantageous in case one wishes to target only proliferating tumor cells. Owing to the heterogeneous proliferation rate in different parts of a tumor, however, it could be desirable, under some circumstances, to be able to target even the fraction of nonproliferating tumor cells. This can now be obtained by the use of lentivirus (2) or by switching to the use of adenoviruses that can target both dividing and quiescent cells but also induce unwanted inflammmatory reactions from the host.

20.
Int J Cancer ; 88(6): 956-61, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11093821

ABSTRACT

Few cases of anti-colon cancer specific T lymphocytes have been described so far. Moreover, the majority of these effectors were generated in vitro by stimulating PBMC from patients or healthy donors with peptides that were derived from proteins expressed and/or secreted by colon cancer tissue such as CEA, Mucin or Her-2/neu. The aim of our study was to evaluate the immunogenicity of colorectal carcinomas in an autologous setting. We exploited the antigen processing and presentation capacity of dendritic cells (DC) to establish an in vitro autologous system that can bypass the need of obtaining cultured tumor cells. DC were generated from the adherent monocyte fraction of PBMC taken from stage II/III colorectal cancer patients. A single cell suspension was prepared by mechanical and enzymatic disruption of the surgical specimens immediately after resection. DC were loaded with autologous tumor lysate, obtained by repeated freezing and thawing, before being used as stimulators for autologous PBL. HLA-class II restricted T cells that recognize the autologous tumor could be generated in a proportion of patients. The fine specificity of the anti-tumor T cells indicates that differentiation as well as tumor restricted antigens are expressed in colon cancer and that these antigens can evoke a class II HLA-restricted response in an autologous setting. Altogether these findings may open a new perspective for a DC based vaccination of colon cancer patients.


Subject(s)
Antigens, Neoplasm/immunology , Colorectal Neoplasms/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocytes/immunology , Humans , Immunity, Cellular
SELECTION OF CITATIONS
SEARCH DETAIL
...