31 results on '"Vegetti C"'
Search Results
2. NFATc2 is an intrinsic regulator of melanoma dedifferentiation
- Author
-
Perotti, V, Baldassari, P, Molla, A, Vegetti, C, Bersani, I, Maurichi, A, Santinami, M, Anichini, A, and Mortarini, R
- Published
- 2016
- Full Text
- View/download PDF
3. Mutually exclusive NRASQ61R and BRAFV600E mutations at the single-cell level in the same human melanoma
- Author
-
Sensi, M, Nicolini, G, Petti, C, Bersani, I, Lozupone, F, Molla, A, Vegetti, C, Nonaka, D, Mortarini, R, Parmiani, G, Fais, S, and Anichini, A
- Published
- 2006
- Full Text
- View/download PDF
4. Identification of a novel gp100/pMel17 peptide presented by HLA-A*6801 and recognized on human melanoma by cytolytic T cell clones
- Author
-
Sensi, M., Pellegatta, S., Vegetti, C., Nicolini, G., Parmiani, G., and Anichini, A.
- Published
- 2002
5. Peripheral burst of tumor-specific cytotoxic T lymphocytes and infiltration of metastatic lesions by memory CD8+ T cells in melanoma patients receiving interleukin 12
- Author
-
Mortarini, R, Borri, A, Tragni, G, Bersani, I, Vegetti, C, Bajetta, E, Pilotti, S, Cerundolo, V, and Anichini, A
- Abstract
Systemic effects on T-cell-mediated antitumor immunity, on expression of T-cell adhesion/homing receptors, and on the promotion of T-cell infiltration of neoplastic tissue may represent key steps for the efficacy of immunological therapies of cancer. In this study, we investigated whether these processes can be promoted by s.c. administration of low-dose (0.5 microg/kg) recombinant human interleukin-12 (rHuIL-12) to metastatic melanoma patients. A striking burst of HLA-restricted CTL precursors (CTLp) directed to autologous tumor was documented in peripheral blood by a high-efficiency limiting dilution analysis technique within a few days after rHuIL-12 injection. A similar burst in peripheral CTLp frequency was observed even when looking at response to a single tumor-derived peptide, as documented by an increase in Melan-A/Mart-1(27-35)-specific CTLp in two HLA-A*0201+ patients by limiting dilution analysis and by staining peripheral blood lymphocytes (PBLs) with HLA-A*0201-melanoma antigen-A/melanoma antigen recognized by T cells (Melan-A/Mart)-1 tetrameric complexes. The CTLp burst was associated, in PBLs, with enhanced expression of T-cell adhesion/homing receptors CD11a/CD18, CD49d, CD44, and with increased proportion of cutaneous lymphocyte antigen (CLA)-positive T cells. This was matched by a marked increase, in serum, of soluble forms of the endothelial cell adhesion molecules E-selectin, vascular cell adhesion molecules (VCAM)-1 and intercellular adhesion molecules (ICAM)-1. Infiltration of neoplastic tissue by CDS+ T cells with a memory and cytolytic phenotype was found by immunohistochemistry in eight of eight posttreatment metastatic lesions but not in five of five pretreatment metastatic lesions from three patients. Increased tumor necrosis and/or fibrosis were also found in several posttherapy lesions of two of three patients in comparison with pretherapy metastases. These results provide the first evidence that rHuIL-12 can boost the frequency of circulating antitumor CTLp in tumor patients, enhances expression of ligand receptor pairs contributing to the lymphocyte function-associated antigen-1/ICAM-1, very late antigen-4/VCAM-1, and CLA/E-selectin adhesion pathways, and promotes infiltration of neoplastic lesions by CD8+ memory T cells in a clinical setting.
- Published
- 2016
6. NFATc2 is an intrinsic regulator of melanoma dedifferentiation
- Author
-
Perotti, V, primary, Baldassari, P, additional, Molla, A, additional, Vegetti, C, additional, Bersani, I, additional, Maurichi, A, additional, Santinami, M, additional, Anichini, A, additional, and Mortarini, R, additional
- Published
- 2015
- Full Text
- View/download PDF
7. Synergistic anti-tumor activity and inhibition of angiogenesis by cotargeting of oncogenic and death receptor pathways in human melanoma
- Author
-
Grazia, G, primary, Vegetti, C, additional, Benigni, F, additional, Penna, I, additional, Perotti, V, additional, Tassi, E, additional, Bersani, I, additional, Nicolini, G, additional, Canevari, S, additional, Carlo-Stella, C, additional, Gianni, A M, additional, Mortarini, R, additional, and Anichini, A, additional
- Published
- 2014
- Full Text
- View/download PDF
8. Peptides with dual binding specificity for HLA-A2 and HLA-E are encoded by alternatively spliced isoforms of the antioxidant enzyme peroxiredoxin 5
- Author
-
Sensi, M., primary, Pietra, G., additional, Molla, A., additional, Nicolini, G., additional, Vegetti, C., additional, Bersani, I., additional, Millo, E., additional, Weiss, E., additional, Moretta, L., additional, Mingari, M. C., additional, and Anichini, A., additional
- Published
- 2009
- Full Text
- View/download PDF
9. The effect of artificial antigen-presenting cells with preclustered anti-CD28/-CD3/-LFA-1 monoclonal antibodies on the induction of ex vivo expansion of functional human antitumor T cells
- Author
-
Zappasodi, R., primary, Di Nicola, M., additional, Carlo-Stella, C., additional, Mortarini, R., additional, Molla, A., additional, Vegetti, C., additional, Albani, S., additional, Anichini, A., additional, and Gianni, A. M., additional
- Published
- 2008
- Full Text
- View/download PDF
10. Novel isoforms of peroxiredoxin 5 encode a unique melanoma antigen and can provide dual-binding peptides for classical and nonclassical HLA molecules
- Author
-
Sensi, M., primary, Pietra, G., additional, Nicolini, G., additional, Vegetti, C., additional, Molla, A., additional, Zanon, M., additional, Mingari, M., additional, Moretta, L., additional, and Anichini, A., additional
- Published
- 2006
- Full Text
- View/download PDF
11. Multiple melanoma-associated epitopes recognized by HLA-A3-restricted CTLs and shared by melanomas but not melanocytes.
- Author
-
Mazzocchi, A, primary, Storkus, W J, additional, Traversari, C, additional, Tarsini, P, additional, Maeurer, M J, additional, Rivoltini, L, additional, Vegetti, C, additional, Belli, F, additional, Anichini, A, additional, Parmiani, G, additional, and Castelli, C, additional
- Published
- 1996
- Full Text
- View/download PDF
12. Mutually exclusive NRASQ61R and BRAFV600E mutations at the single-cell level in the same human melanoma.
- Author
-
Sensi, M., Nicolini, G., Petti, C., Bersani, I., Lozupone, F., Molla, A., Vegetti, C., Nonaka, D., Mortarini, R., Parmiani, G., Fais, S., and Anichini, A.
- Subjects
GENETICS ,TUMORS ,POLYMERASE chain reaction ,LABORATORY mice ,CELLS ,MELANOMA - 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 BRAF
V600E 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.Oncogene (2006) 25, 3357–3364. doi:10.1038/sj.onc.1209379; published online 6 February 2006 [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
13. Differential loss of T cell signaling molecules in metastatic melanoma patients' T lymphocyte subsets expressing distinct TCR variable regions
- Author
-
Maccalli, C., Pisarra, P., Vegetti, C., Sensi, M., Parmiani, G., and Andrea Anichini
- Subjects
Adult ,Male ,CD3 Complex ,Receptors, Antigen, T-Cell, alpha-beta ,Immunology ,Receptors, Antigen, T-Cell ,Down-Regulation ,Lymphocyte Activation ,Immunophenotyping ,CD28 Antigens ,HLA Antigens ,T-Lymphocyte Subsets ,Suppressor Factors, Immunologic ,Tumor Cells, Cultured ,Humans ,Immunology and Allergy ,Melanoma ,Aged ,Aged, 80 and over ,ZAP-70 Protein-Tyrosine Kinase ,Cell-Free System ,Histocompatibility Antigens Class I ,Antibodies, Monoclonal ,Membrane Proteins ,Middle Aged ,Protein-Tyrosine Kinases ,Solubility ,Lymphocyte Specific Protein Tyrosine Kinase p56(lck) ,Disease Progression ,Female ,Signal Transduction - Abstract
In this study we tested the hypothesis that loss of T cell signaling molecules in metastatic melanoma patients’ T cells may affect differently T cell subsets characterized by distinct TCR variable regions. By a two-color immunofluorescence technique, expression of ζ-chain, lck, and ZAP-70 was evaluated in CD3+ T cells and in three representative T cell subsets expressing TCRAV2, TCRBV2, or TCRBV18. Partial loss of lck and ZAP-70 was found in CD3+ T cells from PBL of most melanoma patients, but not of healthy donors. The extent of ζ-chain, lck, and ZAP-70 loss depended on the TCRV region expressed by the T cells, and this association was maintained or increased during progression of disease. Coculture of patients’ or donors’ T cell with melanoma cells, or with their supernatants, but not with normal fibroblasts or their supernatants, down-modulated expression of ζ-chain, lck, and ZAP-70 in a TCRV region-dependent way. Immunodepletion of soluble HLA class I molecules present in tumor supernatants, but not of soluble ICAM-1, blocked the suppressive effect on T cell signaling molecule expression. T cell activation with mAbs to a single TCRV region and to CD28 led to significant and TCRV region-specific re-induction of ζ-chain expression. These findings indicate that extent of TCR signaling molecules loss in T lymphocytes from metastatic melanoma patients depends on the TCRV region and suggest that tumor-derived HLA class I molecules may contribute to induce such alterations.
14. Peripheral burst of tumor-specific cytotoxic T lymphocytes and infiltration of metastatic lesions by memory CD8+ T cells in melanoma patients receiving interleukin 12
- Author
-
Andrea Anichini, Bajetta, E., Bersani, I., Borri, A., Cerundolo, V., Mortarini, R., Pilotti, S., Tragni, G., and Vegetti, C.
15. Multiple Melanoma-Associated Epitopes Recognized by HLA-A3-Restricted CTLs and Shared by Melanomas but Not Melanocytes
- Author
-
Mazzocchi A, Wj, Storkus, Traversari C, Tarsini P, Markus Maeurer, Rivoltini L, Vegetti C, Belli F, Anichini A, Parmiani G, and Castelli C
- Subjects
Cytotoxicity, Immunologic ,Antigen Presentation ,Receptors, Antigen, T-Cell, alpha-beta ,Immunology ,HLA-A3 Antigen ,Transfection ,Epitopes ,Interferon-gamma ,Antigens, Neoplasm ,Chlorocebus aethiops ,Immunology and Allergy ,Animals ,Humans ,Melanocytes ,Melanoma ,Cell Line, Transformed ,T-Lymphocytes, Cytotoxic - Abstract
The molecular characterization of melanoma-associated Ags allowed the definition of several HLA class I-presented peptides recognized by T cells. However, no HLA-A3.1-restricted melanoma epitopes have been identified to date. To gain insight into the HLA-A3.1-restricted T cell epitope repertoire of human melanoma, we analyzed the immunologic reactivity of CTLs isolated from tumor-involved or tumor-free lymph nodes in two HLA-A3.1+ melanoma patients. Three CTL lines, clonal or highly oligoclonal in their TCR composition, and two CTL clones were selected for HLA class I-restricted lysis of the autologous tumor and then tested for the recognition of HLA-A3+ and HLA-A3- normal or neoplastic cells of the melanocyte lineage. One CTL recognized a unique HLA-A3.1-restricted Ag expressed only by the autologous tumors, while all the other CTLs defined three HLA-A3.1 epitopes shared by melanomas, but not by melanocytes. Moreover, the epitopes of two CTL lines with different specificity were reconstituted by nonoverlapping fractions of HLA-A3+ melanoma-derived peptides resolved by reverse phase-HPLC, indicating that distinct naturally processed peptides were specifically recognized on melanoma cells in association with HLA-A3.1 molecules. These novel lineage-unrelated HLA-A3.1-restricted melanoma epitopes do not derive from MAGE, BAGE, or GAGE gene families, as evaluated by the COS-7 transfection assay. Our data show that CTLs may recognize HLA-A3.1-class 1 complexes presenting melanoma (but not melanocyte)-associated epitopes that are either unique to a given patient's tumor or that are shared between multiple melanomas.
16. Early effector T lymphocytes coexpress multiple inhibitory receptors in primary non-small cell lung cancer
- Author
-
Tassi E, Grazia G, Vegetti C, Bersani I, Bertolini G, Molla A, Baldassari P, Andriani F, Roz L, Sozzi G, Pastorino U, Mortarini R, and Andrea Anichini
17. Multiple melanoma-associated epitopes recognized by HLA-A3-restricted CTLs and shared by melanomas but not melanocytes
- Author
-
Mazzocchi, A., Storkus, W.J., Traversari, C., Tarsini, P., Maeurer, M.J., Rivoltini, L., Vegetti, C., Belli, F., Anichini, A., Parmiani, G., and Castelli, C.
- Subjects
HLA histocompatibility antigens -- Research ,Killer cells -- Research ,Melanoma -- Research ,Histocompatibility antigens -- Research ,Health ,Research - Abstract
Mazzocchi, A.; Storkus, W.J.; Traversari, C.; Tarsini, P.; Maeurer, M.J.; Rivoltini, L.; Vegetti, C.; Belli, F.; Anichini, A.; Parmiani, G.; Castelli, C. 'Multiple Melanoma-Associated Epitopes Recognized by HLA-A3-Restricted CTLs and [...]
- Published
- 1996
18. A microRNA Prognostic Signature in Patients with Diffuse Intrinsic Pontine Gliomas through Non-Invasive Liquid Biopsy.
- Author
-
Iannó MF, Biassoni V, Schiavello E, Carenzo A, Boschetti L, Gandola L, Diletto B, Marchesi E, Vegetti C, Molla A, Kramm CM, van Vuurden DG, Gasparini P, Gianno F, Giangaspero F, Modena P, Bison B, Anichini A, Vennarini S, Pignoli E, Massimino M, and De Cecco L
- Abstract
Diffuse midline gliomas (DMGs) originate in the thalamus, brainstem, cerebellum and spine. This entity includes tumors that infiltrate the pons, called diffuse intrinsic pontine gliomas (DIPGs), with a rapid onset and devastating neurological symptoms. Since surgical removal in DIPGs is not feasible, the purpose of this study was to profile circulating miRNA expression in DIPG patients in an effort to identify a non-invasive prognostic signature with clinical impact. Using a high-throughput platform, miRNA expression was profiled in serum samples collected at the time of MRI diagnosis and prior to radiation and/or systemic therapy from 47 patients enrolled in clinical studies, combining nimotuzumab and vinorelbine with concomitant radiation. With progression-free survival as the primary endpoint, a semi-supervised learning approach was used to identify a signature that was also tested taking overall survival as the clinical endpoint. A signature comprising 13 circulating miRNAs was identified in the training set ( n = 23) as being able to stratify patients by risk of disease progression (log-rank p = 0.00014; HR = 7.99, 95% CI 2.38-26.87). When challenged in a separate validation set ( n = 24), it confirmed its ability to predict progression (log-rank p = 0.00026; HR = 5.51, 95% CI 2.03-14.9). The value of our signature was also confirmed when overall survival was considered (log-rank p = 0.0021, HR = 4.12, 95% CI 1.57-10.8). We have identified and validated a prognostic marker based on the expression of 13 circulating miRNAs that can shed light on a patient's risk of progression. This is the first demonstration of the usefulness of nucleic acids circulating in the blood as powerful, easy-to-assay molecular markers of disease status in DIPG. This study provides Class II evidence that a signature based on 13 circulating miRNAs is associated with the risk of disease progression.
- Published
- 2022
- Full Text
- View/download PDF
19. Early Effector T Lymphocytes Coexpress Multiple Inhibitory Receptors in Primary Non-Small Cell Lung Cancer.
- Author
-
Tassi E, Grazia G, Vegetti C, Bersani I, Bertolini G, Molla A, Baldassari P, Andriani F, Roz L, Sozzi G, Pastorino U, Mortarini R, and Anichini A
- Subjects
- Antigens, CD analysis, CTLA-4 Antigen analysis, Forkhead Transcription Factors analysis, HLA-DR Antigens analysis, Hepatitis A Virus Cellular Receptor 2 analysis, Humans, Interferon-gamma biosynthesis, Interleukin-2 biosynthesis, Lymphocyte Activation, Lymphocytes, Tumor-Infiltrating immunology, Programmed Cell Death 1 Receptor analysis, Receptors, Immunologic analysis, Lymphocyte Activation Gene 3 Protein, CD8-Positive T-Lymphocytes immunology, Carcinoma, Non-Small-Cell Lung immunology, Lung Neoplasms immunology
- Abstract
Clinical efficacy of PD-1/PD-L1 targeting relies upon the reactivation of tumor-specific but functionally impaired PD-1
+ T cells present before therapy. Thus, analyzing early-stage primary tumors may reveal the presence of T cells that are not yet functionally impaired. In this study, we report that activated (HLA-DR+ ) T cells with an effector memory (TEM ) profile are enriched in such lesions. Tumor-infiltrating lymphocytes coexpressed PD-1 with the inhibitory receptors TIM-3, CTLA-4, LAG-3, and TIGIT, but also displayed a recently activated, nonexhausted phenotype. We also identified a subset of CD8+ PD-1+ FOXP3+ T lymphocytes at the earliest phase of functional differentiation after priming, termed "early effector cells" (EEC), which also exhibited an activated nonexhausted phenotype, but was less differentiated and associated with coexpression of multiple inhibitory receptors. In response to autologous tumor, EECs upregulated CD107a, produced IL2 and IFNγ, and were competent for differentiation. The identification of EECs marked by inhibitory receptor expression at tumor sites will enable investigations of early stages of adaptive antitumor immunity, as well as support the rationale for administering immunotherapy in early-stage non-small cell lung cancer. Cancer Res; 77(4); 851-61. ©2016 AACR ., (©2016 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
20. NFATc2 is a potential therapeutic target in human melanoma.
- Author
-
Perotti V, Baldassari P, Bersani I, Molla A, Vegetti C, Tassi E, Dal Col J, Dolcetti R, Anichini A, and Mortarini R
- Subjects
- Apoptosis drug effects, Calcineurin metabolism, Caspase 2 metabolism, Caspase 3 metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cyclosporine pharmacology, Cysteine Endopeptidases metabolism, Down-Regulation drug effects, Down-Regulation physiology, Enzyme Inhibitors pharmacology, Humans, Inhibitor of Apoptosis Proteins metabolism, MAP Kinase Signaling System drug effects, Melanoma drug therapy, Melanoma pathology, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf metabolism, Skin Neoplasms drug therapy, Skin Neoplasms pathology, TNF-Related Apoptosis-Inducing Ligand metabolism, Apoptosis physiology, MAP Kinase Signaling System physiology, Melanoma metabolism, NFATC Transcription Factors metabolism, Skin Neoplasms metabolism
- Abstract
The identification of intracellular signaling pathways that promote cell proliferation and resistance to cell death may lead to the development of improved treatment for advanced melanoma. Here we show that the calcineurin/nuclear factor of activated T cells c2 (NFATc2) pathway has an antiapoptotic role in melanoma cells. Expression of NFATc2 was constitutive in vitro and in vivo in human melanoma, and cyclosporin A (CsA) treatment of melanoma cells led to downmodulation of NFATc2. Inhibition of the calcineurin/NFAT pathway by CsA, or by NFATc2 silencing, led to modulation of cell cycle inhibitors and apoptosis-related proteins such as Apollon, and promoted caspase-dependent apoptosis of neoplastic cells. Calcineurin/NFATc2 targeting significantly enhanced melanoma cell death induced by antitumor agents, such as MEK- or BRAF(V600E)-specific inhibitors, and tumor necrosis factor-related apoptosis-inducing ligand, which trigger the intrinsic or extrinsic pathway of apoptosis, respectively. These findings identify NFATc2 as a potential therapeutic target in melanoma.
- Published
- 2012
- Full Text
- View/download PDF
21. AMPK activators inhibit the proliferation of human melanomas bearing the activated MAPK pathway.
- Author
-
Petti C, Vegetti C, Molla A, Bersani I, Cleris L, Mustard KJ, Formelli F, Hardie GD, Sensi M, and Anichini A
- Subjects
- AMP-Activated Protein Kinases genetics, Amino Acid Sequence, Aminoimidazole Carboxamide analogs & derivatives, Aminoimidazole Carboxamide pharmacology, Animals, Cell Cycle drug effects, Cell Growth Processes drug effects, Cell Line, Tumor, Female, HEK293 Cells, Humans, Immunohistochemistry, Melanoma enzymology, Melanoma genetics, Melanoma pathology, Mice, Mice, Nude, Molecular Sequence Data, Phenformin pharmacology, Phosphorylation, Ribonucleotides pharmacology, Signal Transduction drug effects, Skin Neoplasms enzymology, Skin Neoplasms genetics, AMP-Activated Protein Kinases metabolism, Enzyme Activators pharmacology, Melanoma drug therapy, Skin Neoplasms drug therapy
- Abstract
Raf/MEK/ERK signaling can inhibit the liver kinase B1-AMP-activated protein kinase (LKB1-AMPK) pathway, thus rendering melanoma cells resistant to energy stress conditions. We evaluated whether pharmacological reactivation of the AMPK function could exert antitumor effects on melanoma cells bearing this pathway constitutively active because of a mutation in NRAS or BRAF genes. Nine melanoma cell lines were treated with the AMPK activators 5-aminoimidazole-4-carboxamide-ribonucleoside (AICAR) and phenformin. The activation of AMPK enzymatic activity, phosphorylation of AMPK and acetyl-CoA carboxylase kinase, in-vitro proliferation, cell cycle, and in-vivo growth of xenografts in nude mice were evaluated. AICAR and phenformin promoted phosphorylation and enzymatic activity of AMPK, as well as phosphorylation of the AMPK downstream target acetyl-CoA carboxylase. Drug treatment of either BRAF-mutant or NRAS-mutant melanomas, at doses not inducing cell death, was accompanied by a dose-dependent decrease in melanoma cell proliferation because of cell cycle arrest in either the G0/G1 or the S phase, associated with an increased expression of the p21 cell cycle inhibitor. Melanomas isolated from subcutaneously implanted mice, 25 days from treatment with AICAR, showed increased staining of the senescence-associated marker β-galactosidase, high p21 expression, and evidence of necrosis. Altogether, these results indicate that pharmacological activators of AMPK-dependent pathways inhibit the cell growth of melanoma cells with active Raf/MEK/ERK signaling and provide a rationale for further investigation on their use in combination therapies.
- Published
- 2012
- Full Text
- View/download PDF
22. Role of Apollon in human melanoma resistance to antitumor agents that activate the intrinsic or the extrinsic apoptosis pathways.
- Author
-
Tassi E, Zanon M, Vegetti C, Molla A, Bersani I, Perotti V, Pennati M, Zaffaroni N, Milella M, Ferrone S, Carlo-Stella C, Gianni AM, Mortarini R, and Anichini A
- Subjects
- Antibodies, Monoclonal immunology, Antineoplastic Agents pharmacology, Caspase 2 metabolism, Caspase 3 biosynthesis, Caspase 8 metabolism, Caspase 9 metabolism, Cell Line, Transformed, Cell Polarity, Down-Regulation, Enzyme Activation, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Gene Expression Profiling, Humans, Inhibitor of Apoptosis Proteins genetics, Melanoma metabolism, Mitochondria metabolism, Proto-Oncogene Proteins B-raf antagonists & inhibitors, RNA Interference, RNA, Small Interfering, TOR Serine-Threonine Kinases antagonists & inhibitors, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Inhibitor of Apoptosis Proteins metabolism, Melanoma drug therapy
- Abstract
Purpose: To assess the role of Apollon in melanoma resistance to intrinsic and extrinsic pathways of apoptosis and to identify strategies to reduce its expression., Experimental Design: Apollon expression was assessed in melanoma cells in vitro and in vivo. Apollon modulation and melanoma apoptosis were evaluated by Western blot and/or flow cytometry in response to cytotoxic drugs, mitogen-activated protein/extracellular signal-regulated kinase (MEK)-, BRAF(V600E)-, and mTOR-specific inhibitors, TRAIL and anti-HLA class II monoclonal antibodies (mAb). Mitochondrial depolarization, caspase activation, apoptosis assays, and gene expression profiling were used to test effects of Apollon silencing, by siRNA, on melanoma response to antitumor agents., Results: Apollon was constitutively expressed by melanoma cells, in vitro and in vivo, and at higher levels than in benign melanocytic lesions. Melanoma apoptosis correlated significantly with Apollon protein downmodulation in response to cytotoxic drugs, MEK, or BRAF(V600E)-specific inhibitors. Combinatorial treatment with MEK and mTOR inhibitors and HLA class II ligation, by a specific mAb, promoted Apollon downmodulation and enhanced melanoma apoptosis. Apollon downmodulation induced by antitumor agents was caspase independent, but proteasome dependent. Knockdown of Apollon, by siRNA, triggered apoptosis and/or significantly enhanced melanoma cell death in response to cytotoxic drugs, MEK- and BRAF(V600E)-specific inhibitors, and soluble or membrane-bound TRAIL. Apollon silencing promoted mitochondrial depolarization and caspase-2, caspase-8, caspase-9, and caspase-3 activation in response to different antitumor agents and altered the profile of genes modulated by MEK or BRAF(V600E)-specific inhibitors., Conclusions: Targeting of Apollon may significantly improve melanoma cell death in response to antitumor agents that trigger the intrinsic or the extrinsic apoptosis pathways., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
23. Tumor-reactive CD8+ early effector T cells identified at tumor site in primary and metastatic melanoma.
- Author
-
Anichini A, Molla A, Vegetti C, Bersani I, Zappasodi R, Arienti F, Ravagnani F, Maurichi A, Patuzzo R, Santinami M, Pircher H, Di Nicola M, and Mortarini R
- Subjects
- Blotting, Western, CD8-Positive T-Lymphocytes pathology, Cell Differentiation, Cells, Cultured, Cytokines metabolism, Flow Cytometry, Forkhead Transcription Factors metabolism, Humans, Immunoenzyme Techniques, Immunologic Memory immunology, Lymph Nodes immunology, Lymphatic Metastasis, Lymphocyte Activation physiology, Melanoma secondary, Phenotype, Skin Neoplasms pathology, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, T-Lymphocytes, Regulatory pathology, CD8-Positive T-Lymphocytes immunology, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Skin Neoplasms immunology
- Abstract
CD8(+) T cells at the earliest stage of effector generation have not been identified at tumor site of melanoma patients. Such early effectors, if present, should be characterized by a specific phenotype, distinct from that expressed at later stages of the antigen-induced differentiation program, by short-lived effector cells, memory precursors, and terminal effectors. Here, we show that neoplastic tissues from primary and metastatic lesions of melanoma patients contain a subset of CD8(+) T cells expressing FOXP3. CD8(+) FOXP3(+) CD25(+) T lymphocytes were found in tumor-invaded lymph nodes (TILN), s.c. metastases, and advanced primary lesions. Their frequency was significantly higher in TILN compared with tumor-free lymph nodes or with peripheral blood and in primary tumors compared with TILN. CD8(+) FOXP3(+) T cells did not express markers of regulatory [CTLA-4, CCL4, interleukin-10 (IL-10), transforming growth factor-β1], exhausted (PD-1), or senescent (CD57) CD8(+) T lymphocytes. Instead, this subset showed an antigen-experienced "EM1" phenotype (CCR7(-) CD45RA(-) CD28(+) CD27(+)) and exhibited a CD127(-), KLRG1(-), HLA-DR(+), CD38(+), T-bet(+), perforin(+) "early effector" profile predicted by current models. CD8(+) FOXP3(+) T cells produced IFN-γ on short in vitro activation, recognized autologous tumor by CD107a mobilization, and expressed Ki-67 on ex vivo analysis. In response to autologous tumor plus IL-2/IL-15, the CD8(+) FOXP3(+) T cells proliferated promptly and showed competence for differentiation (downregulation of CD27 and upregulation of T-bet). These results suggest development of early phases of antitumor immunity even in advanced melanoma. Moreover, the CD8(+) FOXP3(+) "early effector" subset may be an invaluable tool for monitoring immunity at tumor site., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
24. Impaired STAT phosphorylation in T cells from melanoma patients in response to IL-2: association with clinical stage.
- Author
-
Mortarini R, Vegetti C, Molla A, Arienti F, Ravagnani F, Maurichi A, Patuzzo R, Santinami M, and Anichini A
- Subjects
- CD3 Complex immunology, CD3 Complex metabolism, Cell Proliferation drug effects, Humans, Interleukin-15 pharmacology, Janus Kinase 3 immunology, Janus Kinase 3 metabolism, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Melanoma pathology, Neoplasm Staging, Phosphorylation drug effects, Receptors, Cytokine immunology, Receptors, Cytokine metabolism, Skin Neoplasms pathology, T-Lymphocytes metabolism, Interleukin-2 pharmacology, Killer Cells, Natural drug effects, Melanoma immunology, STAT1 Transcription Factor metabolism, STAT5 Transcription Factor metabolism, Skin Neoplasms immunology, T-Lymphocytes drug effects
- Abstract
Purpose: To assess the extent of signal transducer and activator of transcription (STAT) activation in response to interleukin 2 (IL-2) in melanoma patients' T cells, along with clinical stage of tumor progression., Experimental Design: T lymphocytes from peripheral blood of healthy donors and of American Joint Committee on Cancer stage I to IV melanoma patients, as well as from metastatic lymph nodes of patients, were evaluated for responsiveness to IL-2. CFSE assays and single-cell phospho-STAT-specific flow cytometry screening were used. Results. T cells from advanced melanoma patients, in comparison with healthy donors, showed reduced proliferation to IL-2 and IL-15, but not to anti-CD3 monoclonal antibody. Impaired response occurred in CCR7(+) and CCR7(-) T-cell subsets, but not in CD3(-) CD8(+) natural killer (NK) cells, and was not explained by induction of apoptosis, increased cytokine consumption, or altered IL-2R subunit expression in patients' T lymphocytes. By phospho-specific flow cytometry, defective STAT1 and STAT5 activation in response to IL-2 was found mainly in T lymphocytes from peripheral blood and/or tumor site of American Joint Committee on Cancer stage III and IV patients, compared with stage I and II patients and to donors, and in melanoma antigen-specific T cells isolated from metastatic lymph nodes. At tumor site, impaired STAT activation in T cells did not correlate with frequency of CD4(+) CD25(+) Foxp3(+) T cells. Serum from advanced melanoma patients inhibited IL-2-dependent STAT activation in donors' T cells and a neutralizing monoclonal antibody to transforming growth factor beta1 counteracted such inhibition., Conclusions: These results provide evidence for development of impaired STAT signaling in response to IL-2, along with clinical evolution of the disease, in melanoma patients' T cells.
- Published
- 2009
- Full Text
- View/download PDF
25. Coexpression of NRASQ61R and BRAFV600E in human melanoma cells activates senescence and increases susceptibility to cell-mediated cytotoxicity.
- Author
-
Petti C, Molla A, Vegetti C, Ferrone S, Anichini A, and Sensi M
- Subjects
- Alleles, Antigen Presentation, Cell Line, Tumor, Cellular Senescence physiology, Cytotoxicity, Immunologic, Doxycycline pharmacology, Enzyme Activation, Gene Expression Regulation, Neoplastic, Histocompatibility Antigens Class I immunology, Humans, Melanoma genetics, Melanoma pathology, Mitogen-Activated Protein Kinase Kinases metabolism, Mutation, Phosphorylation, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins p21(ras) genetics, Melanoma immunology, Melanoma metabolism, Proto-Oncogene Proteins B-raf biosynthesis, Proto-Oncogene Proteins p21(ras) biosynthesis, T-Lymphocytes, Cytotoxic immunology
- Abstract
Activating mutations in BRAF and NRAS oncogenes in human melanomas are mutually exclusive. This finding has suggested an epistatic relationship but is consistent even with synthetic lethality. To evaluate the latter possibility, a mutated NRAS(Q61R) oncogene was expressed, under a constitutive or a doxycycline-regulated promoter, in a metastatic melanoma clone (clone 21) harboring an activated BRAF(V600E) oncogene. After the first 10 to 12 in vitro passages, the constitutive NRAS(Q61R) transfectant displayed progressive accumulation in G(0)-G(1) phase of the cell cycle and stained for the senescence-associated beta-galactosidase activity (SA-beta-Gal). Inducible expression of NRAS(Q61R), by the Tet-Off system, in clone 21 cells (21NRAS(61ON)) led to overactivation of the RAS/RAF/mitogen-activated protein kinase signaling pathway and, after the 10th in vitro passage, led to promotion of senescence. This was documented by reduced proliferation, flattened cell morphology, reduced growth in Matrigel, positive staining for SA-beta-Gal, and expression of AMP-activated protein kinase and of the cell cycle inhibitor p21(waf1/Cip1). These effects were detected neither in 21 cells with silenced NRAS(Q61R) (21NRAS(61OFF)) nor in cells transfected with an inducible wild-type NRAS gene (21NRAS(WTON)). In addition, when compared with parental 21 cells, or with 21NRAS(61OFF), 21NRAS(61ON) and constitutive NRAS(Q61R) transfectants cells showed increased susceptibility to cytotoxicity by both HLA class I antigen-restricted and nonspecific T cells and up-regulation of several MHC class I antigen processing machinery components. These results suggest a relationship of synthetic lethality between NRAS and BRAF oncogenes, leading to selection against "double-mutant" cells.
- Published
- 2006
- Full Text
- View/download PDF
26. Association of antigen-processing machinery and HLA antigen phenotype of melanoma cells with survival in American Joint Committee on Cancer stage III and IV melanoma patients.
- Author
-
Anichini A, Mortarini R, Nonaka D, Molla A, Vegetti C, Montaldi E, Wang X, and Ferrone S
- Subjects
- Animals, Cell Line, Tumor, HLA Antigens biosynthesis, Humans, Mice, Mice, Inbred BALB C, Neoplasm Staging, Antigen Presentation immunology, HLA Antigens immunology, Melanoma immunology, Melanoma pathology
- Abstract
Because changes in the expression level of antigen-processing machinery (APM) components and HLA class I and II antigens in melanoma cells are expected to affect their interactions with the immune system of the host, we assessed the clinical relevance of quantitative variations in the expression of these molecules in melanoma lesions. Short-term (<10 in vitro passages) melanoma cell lines isolated from 85 American Joint Committee on Cancer (AJCC) stage III and IV patients were stained with APM component and HLA class I antigen-specific and HLA class II antigen-specific monoclonal antibodies and analyzed by flow cytometry. The phenotype of all tumors was characterized by intertumor and intratumor heterogeneity in the expression of all the markers and by significant correlations in the level of expression of markers belonging to the HLA class I antigen-processing and presentation pathway. Hierarchical clustering of the mean fluorescence intensity data defined two main clusters of tumors. The corresponding groups of patients differed significantly in the overall survival but not in other relevant clinical variables, including AJCC stage and therapy received after surgery. Cox regression analysis showed that beta2-microglobulin and HLA class II antigen expression were significantly associated with patients' survival. This evidence was corroborated by the immunohistochemical analysis for HLA class II antigen expression of melanoma lesions from an unrelated group of 52 AJCC stage III and IV patients. These results suggest that quantitative variations in APM component and HLA expression in melanoma lesions from AJCC stage III and IV patients may have an effect on the clinical course of the disease.
- Published
- 2006
- Full Text
- View/download PDF
27. Apoptosis protease activator protein-1 expression is dispensable for response of human melanoma cells to distinct proapoptotic agents.
- Author
-
Zanon M, Piris A, Bersani I, Vegetti C, Molla A, Scarito A, and Anichini A
- Subjects
- Amidines pharmacology, Apoptosis physiology, Apoptotic Protease-Activating Factor 1, Benzylamines pharmacology, Camptothecin pharmacology, Caspase 9, Caspase Inhibitors, Caspases metabolism, Celecoxib, Cell Line, Tumor, Enzyme Activation, Enzyme Inhibitors pharmacology, Humans, Immunohistochemistry, Melanoma metabolism, Protein Biosynthesis drug effects, Proteins genetics, Pyrazoles, RNA, Messenger biosynthesis, RNA, Messenger genetics, Sulfonamides pharmacology, Antineoplastic Agents pharmacology, Apoptosis drug effects, Melanoma drug therapy, Melanoma pathology, Proteins metabolism
- Abstract
Loss of expression of the apoptosis protease activator protein-1 (APAF-1) in human melanoma is thought to promote resistance to programmed cell death by preventing caspase-9 activation. However, the role of the APAF-1-dependent pathway in apoptosis activated by cellular stress and/or DNA damage has been recently questioned. We investigated APAF-1 expression in a large panel of human melanomas and assessed cellular response to several proapoptotic agents in tumors expressing or lacking APAF-1 protein. In two melanomas with wild-type p53 but with differential expression of APAF-1, treatment with camptothecin, celecoxib, or an nitric oxide synthase inhibitor (1400W) significantly modulated expression of 36 of 96 genes in an apoptosis-specific cDNA macroarray, but APAF-1 mRNA levels were not induced (in APAF-1(-) cells) nor up-regulated (in APAF-1(+) cells), a finding confirmed at the protein level. Treatment with cisplatin, camptothecin, etoposide, betulinic acid, celecoxib, 1400W, and staurosporine promoted enzymatic activity not only of caspases -2, -8, and -3 but also of caspase-9 in both APAF-1(+) and APAF-1(-) tumor cells. Moreover, drug-induced caspase-9 enzymatic activity could be not only partially but significantly reduced by caspase-2, -3, and -8 -specific inhibitors in both APAF-1(+) and APAF-1(-) tumor cells. In response to 1 to 100 micromol/L of cisplatin, camptothecin, or celecoxib, APAF-1(+) melanomas (n = 12) did not show significantly increased levels of apoptosis compared with APAF-1(-) tumors (n = 7), with the exception of enhanced apoptosis in response to a very high dose (100 micromol/L) of etoposide. These results suggest that the response of human melanoma cells to different proapoptotic agents may be independent of their APAF-1 phenotype.
- Published
- 2004
- Full Text
- View/download PDF
28. The paradox of T-cell-mediated antitumor immunity in spite of poor clinical outcome in human melanoma.
- Author
-
Anichini A, Vegetti C, and Mortarini R
- Subjects
- Antigens, Differentiation immunology, Antigens, Neoplasm immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines therapeutic use, Cell Differentiation, Clinical Trials as Topic, Disease Progression, Humans, Lymphocytes, Tumor-Infiltrating immunology, Melanoma pathology, Melanoma therapy, Models, Immunological, Neoplasm Metastasis, Neoplasm Staging, Treatment Failure, Melanoma immunology, T-Lymphocyte Subsets immunology, Tumor Escape immunology
- Abstract
Human melanoma is hardly ever curable at an advanced stage, but overwhelming evidence from untreated or vaccinated patients indicates that this tumor is highly antigenic and frequently immunogenic. Here, we review recent results indicating that CD8(+) T cell-mediated antitumor immunity is activated at the systemic and tumor level in the early clinical stages (AJCC stages I and II) and continues to be promoted, in a fraction of patients, even in metastatic disease (stages III and IV). This evidence was obtained by looking at frequency, differentiation phenotype, and function of antitumor T cells in periphery and tumor site of melanoma patients. On the other hand, the paradox of immunity in spite of poor clinical evolution of the disease, points toward a model of concurrent evolution of immunity and tumor escape. As melanoma progresses to metastatic disease, powerful mechanisms of tumor evasion from immune recognition, and of immunosuppression, are activated, thus tilting the balance between immunity and escape in favor of tumor resistance to host defense. Nevertheless, recent developments in our understanding of regulation of T cell-mediated immunity can provide clues to the prospects for improved immunotherapy approaches. By integrating the information from basic research in immunology, from murine tumor models, and from trials of immunotherapy, we discuss how the most relevant steps of the antitumor response should be manipulated with greater efficacy by future clinical trials.
- Published
- 2004
- Full Text
- View/download PDF
29. Peripheral burst of tumor-specific cytotoxic T lymphocytes and infiltration of metastatic lesions by memory CD8+ T cells in melanoma patients receiving interleukin 12.
- Author
-
Mortarini R, Borri A, Tragni G, Bersani I, Vegetti C, Bajetta E, Pilotti S, Cerundolo V, and Anichini A
- Subjects
- Antigens, Neoplasm analysis, CD18 Antigens analysis, HLA-A Antigens immunology, Humans, Immunohistochemistry, Lymphatic Metastasis, MART-1 Antigen, Melanoma pathology, Neoplasm Metastasis, Neoplasm Proteins analysis, Pilot Projects, Recombinant Proteins therapeutic use, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Interleukin-12 therapeutic use, Lymphocytes, Tumor-Infiltrating immunology, Melanoma drug therapy, Melanoma immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
Systemic effects on T-cell-mediated antitumor immunity, on expression of T-cell adhesion/homing receptors, and on the promotion of T-cell infiltration of neoplastic tissue may represent key steps for the efficacy of immunological therapies of cancer. In this study, we investigated whether these processes can be promoted by s.c. administration of low-dose (0.5 microg/kg) recombinant human interleukin-12 (rHuIL-12) to metastatic melanoma patients. A striking burst of HLA-restricted CTL precursors (CTLp) directed to autologous tumor was documented in peripheral blood by a high-efficiency limiting dilution analysis technique within a few days after rHuIL-12 injection. A similar burst in peripheral CTLp frequency was observed even when looking at response to a single tumor-derived peptide, as documented by an increase in Melan-A/Mart-1(27-35)-specific CTLp in two HLA-A*0201+ patients by limiting dilution analysis and by staining peripheral blood lymphocytes (PBLs) with HLA-A*0201-melanoma antigen-A/melanoma antigen recognized by T cells (Melan-A/Mart)-1 tetrameric complexes. The CTLp burst was associated, in PBLs, with enhanced expression of T-cell adhesion/homing receptors CD11a/CD18, CD49d, CD44, and with increased proportion of cutaneous lymphocyte antigen (CLA)-positive T cells. This was matched by a marked increase, in serum, of soluble forms of the endothelial cell adhesion molecules E-selectin, vascular cell adhesion molecules (VCAM)-1 and intercellular adhesion molecules (ICAM)-1. Infiltration of neoplastic tissue by CDS+ T cells with a memory and cytolytic phenotype was found by immunohistochemistry in eight of eight posttreatment metastatic lesions but not in five of five pretreatment metastatic lesions from three patients. Increased tumor necrosis and/or fibrosis were also found in several posttherapy lesions of two of three patients in comparison with pretherapy metastases. These results provide the first evidence that rHuIL-12 can boost the frequency of circulating antitumor CTLp in tumor patients, enhances expression of ligand receptor pairs contributing to the lymphocyte function-associated antigen-1/ICAM-1, very late antigen-4/VCAM-1, and CLA/E-selectin adhesion pathways, and promotes infiltration of neoplastic lesions by CD8+ memory T cells in a clinical setting.
- Published
- 2000
30. Differential loss of T cell signaling molecules in metastatic melanoma patients' T lymphocyte subsets expressing distinct TCR variable regions.
- Author
-
Maccalli C, Pisarra P, Vegetti C, Sensi M, Parmiani G, and Anichini A
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal pharmacology, CD28 Antigens immunology, CD3 Complex biosynthesis, Cell-Free System immunology, Disease Progression, Down-Regulation immunology, Female, HLA Antigens metabolism, Histocompatibility Antigens Class I metabolism, Humans, Immunophenotyping, Lymphocyte Activation immunology, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) biosynthesis, Male, Melanoma metabolism, Melanoma pathology, Membrane Proteins biosynthesis, Middle Aged, Protein-Tyrosine Kinases biosynthesis, Receptors, Antigen, T-Cell biosynthesis, Solubility, Suppressor Factors, Immunologic metabolism, Tumor Cells, Cultured, ZAP-70 Protein-Tyrosine Kinase, Melanoma immunology, Melanoma secondary, Receptors, Antigen, T-Cell, alpha-beta biosynthesis, Signal Transduction immunology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism
- Abstract
In this study we tested the hypothesis that loss of T cell signaling molecules in metastatic melanoma patients' T cells may affect differently T cell subsets characterized by distinct TCR variable regions. By a two-color immunofluorescence technique, expression of zeta-chain, lck, and ZAP-70 was evaluated in CD3+ T cells and in three representative T cell subsets expressing TCRAV2, TCRBV2, or TCRBV18. Partial loss of lck and ZAP-70 was found in CD3+ T cells from PBL of most melanoma patients, but not of healthy donors. The extent of zeta-chain, lck, and ZAP-70 loss depended on the TCRV region expressed by the T cells, and this association was maintained or increased during progression of disease. Coculture of patients' or donors' T cell with melanoma cells, or with their supernatants, but not with normal fibroblasts or their supernatants, down-modulated expression of zeta-chain, lck, and ZAP-70 in a TCRV region-dependent way. Immunodepletion of soluble HLA class I molecules present in tumor supernatants, but not of soluble ICAM-1, blocked the suppressive effect on T cell signaling molecule expression. T cell activation with mAbs to a single TCRV region and to CD28 led to significant and TCRV region-specific re-induction of zeta-chain expression. These findings indicate that extent of TCR signaling molecules loss in T lymphocytes from metastatic melanoma patients depends on the TCRV region and suggest that tumor-derived HLA class I molecules may contribute to induce such alterations.
- Published
- 1999
31. Frequency of cytotoxic T lymphocyte precursors (CTLp) interacting with autologous tumor via the T-cell receptor: limiting dilution analysis of specific CTLp in peripheral blood and tumor-invaded lymph nodes of melanoma patients.
- Author
-
Mazzocchi A, Belli F, Mascheroni L, Vegetti C, Parmiani G, and Anichini A
- Subjects
- Adult, Aged, Female, Humans, Lymphatic Metastasis, Lymphocyte Activation, Lymphocyte Subsets, Male, Melanoma secondary, Middle Aged, Phenotype, Sensitivity and Specificity, Hematopoietic Stem Cells cytology, Lymph Nodes pathology, Melanoma blood, Melanoma pathology, Receptors, Antigen, T-Cell physiology, T-Lymphocytes, Cytotoxic cytology
- Abstract
The frequencies of cytotoxic T-lymphocyte precursors (CTLp) that lyse autologous tumor by a T-cell receptor (TCR)-dependent mechanism (specific CTLp) were evaluated by limiting dilution analysis (LDA) using lymphocytes from peripheral blood (PBL) and from surgically resected, tumor-invaded lymph nodes (LNL) in 9 melanoma patients. The frequency of specific CTLp was determined in PBLs and/or LNIs of all patients by a modified LDA assay, enabling us to measure lytic activity on the autologous tumor that could be significantly inhibited by an anti-CD3 monoclonal antibody (MAb). This assay allowed us to detect frequencies of specific CTLp ranging from 1/720 to 1/32,037 in peripheral blood and from 1/328 to 1/22,061 in tumor-invaded lymph nodes. These frequencies indicated that lymphoid populations from PBLs or LNLs of melanoma patients may contain as low as 30 to as much as 3,000 specific CTLp/10(6) lymphocytes. In addition, comparison of wells containing specific CTLp with those showing no inhibition by anti-CD3 MAb indicated that specific CTLp represent between 3 and 88% of all precursors with lytic activity on the tumor. In 6 of 9 patients, no marked differences between PBLs and LNIs in specific CTLp frequencies were found. A 10-fold increase of specific CTLp, in comparison to PBL and LNL, was found only in lymphocytes isolated from a subcutaneous metastasis of one patient. Our results indicate that CTLp interacting with autologous tumor by a TCR-dependent mechanism exist in PBL and LNL of most melanoma patients, although a wide variation in their absolute number is evident among different patients.
- Published
- 1994
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.