Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Scand J Surg ; 108(3): 194-200, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30249173

ABSTRACT

BACKGROUND AND AIMS: Patients with liver tumors involving the inferior vena cava have a poor outcome without surgery. Liver resection en bloc with inferior vena cava resection and reconstruction is now performed in many centers. The purpose of this study is to investigate the safety and efficacy of inferior vena cava reconstruction during hepatic resection. MATERIALS AND METHODS: A review of 12 centers reporting 240 patients with combined hepatectomy and inferior vena cava resection and reconstruction for malignant tumors was performed. Sample size, patient characteristics, histological type of the tumor, method of reconstruction, complications, and long-term survival (1-, 2-, and 5-year survival) were evaluated. RESULTS: A total of 240 patients from 12 institutions (male 58%) with mean age 54 years underwent combined liver resection and inferior vena cava resection and reconstruction for colorectal liver metastases (43%), cholangiocarcinomas (26%), hepatocellular carcinomas (19%), leiomyosarcomas (4%), and other tumors (7.9%). Reconstruction included primary closure (35.8%), patch repair (13.3%), or interposition graft (50.8%) In-hospital mortality was 6.25% and overall morbidity was 42.1%. 1- and 10-year survival rates were 79.7% and 28.9%, respectively. CONCLUSION: Tumors arising in or extending to inferior vena cava that require liver resection should be considered for surgery as it can be performed with an acceptable mortality and morbidity in centers with liver transplantation and hepato-pancreato-biliary facilities.


Subject(s)
Hepatectomy , Liver Neoplasms/surgery , Vascular Surgical Procedures , Vena Cava, Inferior/surgery , Humans , Postoperative Complications , Vena Cava, Inferior/pathology
2.
Eur J Pediatr ; 177(3): 285-294, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29243189

ABSTRACT

Congenital portosystemic venous shunts are rare developmental anomalies resulting in diversion of portal flow to the systemic circulation and have been divided into extra- and intrahepatic shunts. They occur during liver and systemic venous vascular embryogenesis and are associated with other congenital abnormalities. They carry a higher risk of benign and malignant liver tumors and, if left untreated, can result in significant medical complications including systemic encephalopathy and pulmonary hypertension. CONCLUSION: This article reviews the various types of congenital portosystemic shunts and their anatomy, pathogenesis, symptomatology, and timing and options of treatment. What is Known: • The natural history and basic management of this rare congenital anomaly are presented. What is New: • This paper is a comprehensive review; highlights important topics in pathogenesis, clinical symptomatology, and treatment options; and proposes an algorithm in the management of congenital portosystemic shunt disease in order to provide a clear idea to a pediatrician. An effort has been made to emphasize the indications for treatment in the children population and link to the adult group by discussing the consequences of lack of treatment or delayed diagnosis.


Subject(s)
Portal Vein/abnormalities , Vascular Malformations , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/embryology , Abnormalities, Multiple/therapy , Endovascular Procedures , Hepatectomy , Humans , Ligation , Liver Transplantation , Portal Vein/embryology , Vascular Malformations/diagnosis , Vascular Malformations/embryology , Vascular Malformations/therapy
3.
Ann Oncol ; 27(7): 1241-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27029708

ABSTRACT

BACKGROUND: AE37 is the Ii-Key hybrid of the MHC class II peptide, AE36 (HER2 aa:776-790). Phase I studies showed AE37 administered with granulocyte macrophage colony-stimulating factor (GM-CSF) to be safe and highly immunogenic. A prospective, randomized, multicenter phase II adjuvant trial was conducted to evaluate the vaccine's efficacy. METHODS: Clinically disease-free node-positive and high-risk node-negative breast cancer patients with tumors expressing any degree of HER2 [immunohistochemistry (IHC) 1-3+] were enrolled. Patients were randomized to AE37 + GM-CSF versus GM-CSF alone. Toxicity was monitored. Clinical recurrences were documented and disease-free survival (DFS) analyzed. RESULTS: The trial enrolled 298 patients; 153 received AE37 + GM-CSF and 145 received GM-CSF alone. The groups were well matched for clinicopathologic characteristics. Toxicities have been minimal. At the time of the primary analysis, the recurrence rate in the vaccinated group was 12.4% versus 13.8% in the control group [relative risk reduction 12%, HR 0.885, 95% confidence interval (CI) 0.472-1.659, P = 0.70]. The Kaplan-Meier estimated 5-year DFS rate was 80.8% in vaccinated versus 79.5% in control patients. In planned subset analyses of patients with IHC 1+/2+ HER2-expressing tumors, 5-year DFS was 77.2% in vaccinated patients (n = 76) versus 65.7% in control patients (n = 78) (P = 0.21). In patients with triple-negative breast cancer (HER2 IHC 1+/2+ and hormone receptor negative) DFS was 77.7% in vaccinated patients (n = 25) versus 49.0% in control patients (n = 25) (P = 0.12). CONCLUSION: The overall intention-to-treat analysis demonstrates no benefit to vaccination. However, the results confirm that the vaccine is safe and suggest that vaccination may have clinical benefit in patients with low HER2-expressing tumors, specifically TNBC. Further evaluation in a randomized trial enrolling TNBC patients is warranted.


Subject(s)
Cancer Vaccines/administration & dosage , Receptor, ErbB-2/immunology , Triple Negative Breast Neoplasms/prevention & control , Adjuvants, Immunologic/administration & dosage , Adult , Aged , Cancer Vaccines/adverse effects , Disease-Free Survival , Female , Humans , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , Receptor, ErbB-2/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology
4.
Br J Cancer ; 92(1): 72-9, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15583693

ABSTRACT

Unfractionated peptides (MW: up to 10 kDa), derived from HLA-A2.1 positive (+) HER-2/neu-overexpressing primary tumour cell acid cell extracts (ACE), were successfully used to generate in vitro cytotoxic T lymphocytes (CTL). Primary tumour cells were collected from peritoneal malignant effusions of patients with ovarian cancer. Acid cell extracts-induced CTL specifically lysed in an HLA-A2-restricted manner HER-2/neu+ autologous primary tumour cells as well as HER-2/neu+ tumour cell lines. In addition, adoptive transfer of such CTL significantly prolonged the survival of SCID mice xenografted with HLA-A2.1+, HER-2/neu+ human breast and ovarian tumour cell lines. Acid cell extracts collected from HLA-A2.1+ HER-2/neu negative (-) primary ovarian tumours induced HLA-A2.1-restricted CTL with weak in vitro and in vivo antitumour capacity, suggesting that HER-2/neu peptides within ACE from HER-2/neu-overexpressing primary ovarian tumour cells are immunodominant. The results presented herein serve as a rationale for the initiation of vaccination studies in patients with HER-2/neu-overexpressing ovarian tumours utilising autologous tumour-derived ACE.


Subject(s)
Ovarian Neoplasms/metabolism , Peptides/immunology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Cell Extracts , Cell Line, Tumor , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Female , Humans , Immunodominant Epitopes/immunology , Mice , Mice, SCID , Neoplasm Transplantation , Ovarian Neoplasms/immunology , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Br J Cancer ; 89(6): 1055-61, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12966425

ABSTRACT

HER-2/neu oncoprotein contains several major histocompatibility complex class I-restricted epitopes, which are recognised by cytotoxic T lymphocyte (CTL) on autologous tumours and therefore can be used in immune-based cancer therapies. Of these, the most extensively studied is HER-2(9(369)). In the present report, we used dendritic cells pulsed with HER-2(9(369)) to stimulate, in the presence of IL-7 and IL-12, the production of IFN-gamma by patients' CTL detected by the enzyme-linked immunosorbent spot-assay. Frequencies of peptide-specific precursors were estimated in HLA-A2, HLA-A3 and HLA-A26 patients with HER-2/neu-positive (+) breast, ovarian, lung, colorectal and prostate cancers and healthy individuals. We found increased percentages of such precursors in HLA-A2 (25%) and HLA-A26 (30%) patients, which were significantly higher (60%) in HLA-A3 patients. Our results demonstrate for the first time that pre-existing immunity to HER-2(9(369)) occurs in patients with colorectal, lung and prostate cancer. They also suggest that HER-2(9(369)) can be recognised by CTL, besides HLA-A2, also in the context of HLA-A3 and HLA-A26, thus increasing the applicability of HER-2(9(369))-based vaccinations in a considerably broader patients' population.


Subject(s)
Dendritic Cells/immunology , Neoplasms/immunology , Peptide Fragments/immunology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Cell Division , Cytokines/metabolism , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , HLA-A Antigens/immunology , HLA-A2 Antigen/immunology , HLA-A3 Antigen/immunology , Humans , Immunization , Immunophenotyping , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Middle Aged
6.
Br J Cancer ; 88(8): 1292-300, 2003 Apr 22.
Article in English | MEDLINE | ID: mdl-12698199

ABSTRACT

Chimeric receptors comprising of the T-cell receptor-zeta cytoplasmic signalling chain fused to an extracellular ligand-binding domain of a single-chain antibody (scFv) have served as effective tools for redirecting cytotoxic T lymphocytes (CTL) against tumour cells. In this report, we constructed a chimeric scFv/zeta gene composed of the variable regions of an HER-2/neu-specific monoclonal antibody (MAb) joined to the TCR-zeta chain. The scFv(anti-HER-2/neu)/zeta chimeric gene was successfully expressed as a functional surface receptor in the MD.45 CTL hybridoma (MD.45-HER/zeta). More importantly, the scFv(anti-HER-2/neu)/zeta receptor was functionally active, since it triggered cytokine secretion by the MD.45-HER/zeta cells upon recognition of HER-2/neu-positive (+) tumour cell lines, or primary tumour cells from patients with HER-2/neu(+) cancers. The MD.45-HER/zeta-transduced cells also lysed HER-2/neu(+) target cells in vitro with high specificity. We tested the antitumour efficacy of scFv(anti-HER-2/neu)/zeta expressing MD.45 cells in severe combined immunodeficiency disease mice/human and murine tumour models. The adoptively transferred MD.45-HER/zeta cells both slowed significantly the growth of human FM3 melanoma or murine ALC leukaemic cells both transfected to express HER-2/neu. Our data demonstrate the feasibility of redirecting MD.45 CTL with the scFv(anti-HER-2/neu)/zeta chimeric receptor to respond specifically against HER-2/neu expressing tumour cells in vitro and in vivo. Moreover, they make it likely that T cells transduced with the same chimeric gene might be utilised in the treatment of patients with HER-2/neu(+) tumours.


Subject(s)
Breast Neoplasms/immunology , Ovarian Neoplasms/immunology , Receptor, ErbB-2/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Flow Cytometry , Humans , Hybridomas/immunology , Interferon-gamma/biosynthesis , Mice , Mice, SCID , Molecular Sequence Data , Transfection
7.
Cancer Immunol Immunother ; 51(8): 440-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12202905

ABSTRACT

We have developed culture conditions for the efficient expansion of cytotoxic effector cells from peripheral blood mononuclear cells (PBMC) by the timed addition of cytokine-rich supernatants collected from allogeneic PBMC cultures stimulated with anti-CD3 monoclonal antibody (mAb) (allogeneic CD3 supernatants; ACD3S). These cytotoxic effectors belonged primarily to CD56(+) natural killer (NK) cells, and the cell subset with the greatest cytotoxic activity was an otherwise rare population of CD3(+)CD56(+) cells (NKT cells) that expand dramatically under these conditions. CD3(+)CD56(+) cytotoxic effectors were generated from the PBMC of 16 patients with several types of cancer. The CD3(+)CD56(+) cell subset expanded significantly and efficiently lysed NK- as well as lymphokine-activated killer (LAK)-sensitive targets. More importantly, ACD3S-activated CD3(+)CD56(+) cells were capable of efficiently lysing autologous tumor cells including metastatic colorectal, ovarian, breast, lung and pancreatic tumor cells as well as melanoma cells. ACD3S-expanded CD3(+)CD56(+) cells exhibited increased levels of cytoplasmic interleukin-2 (IL-2), tumor necrosis factor-alpha (TNF-alpha), gamma-interferon (IFN-gamma) and perforin. CD3(+)CD56(+) cell-mediated cytotoxicity was not restricted by major histocompatibility complex (MHC) gene products, since it was not blocked by anti-MHC class I mAb but was highly inhibited in the presence of CD2- and CD18-specific mAb. These data suggest that CD3(+)CD56(+) cells expanded under the presence of ACD3S may be utilized in clinical protocols for cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD3 Complex/biosynthesis , CD56 Antigen/biosynthesis , Cytokines/biosynthesis , Immunotherapy/methods , Neoplasms/therapy , CD18 Antigens/biosynthesis , CD2 Antigens/biosynthesis , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , K562 Cells , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/metabolism , Neoplasms/blood , Neoplasms/immunology , Tumor Cells, Cultured
8.
J Biol Regul Homeost Agents ; 16(4): 289-302, 2002.
Article in English | MEDLINE | ID: mdl-12597645

ABSTRACT

Several techniques have been proposed for flow cytometric evaluation of intracellular antigens. This approach is particularly important for detection at the single cell level of proteins which correlate to tumour progression. Bcl-2 and p53 are two of the most relevant proteins. In the present study we have compared five different cell fixation-permeabilisation protocols and nine fluorochrome-conjugated (FITC or PE) monoclonal antibodies (mAb): four mAb directed against Bcl-2 and five against p53. For detection of Bcl-2 we have analysed three Bcl-2 positive cell lines (K562, Daudi and MCF-7), and peripheral blood samples obtained from nine healthy subjects. To distinguish internal positive (lymphocytes) and negative control cells (granulocytes), it was necessary to perform simultaneous detection of surface and intracellular antigens. For detection of p53 three cell lines, two p53 positive (Raji and CEM) and one p53 negative (HL-60), were analysed. Using these cells we have performed a combined analysis of the efficiency of monoclonal antibodies and sample preparation techniques. In conclusion, clones 124-FITC and Bcl-2/100-PE (Bcl-2), and clones BP53,12-FITC and G59-12-PE (p53) provided the highest specific fluorescence intensity of the respective markers independent of cell preparation protocols. Importantly, our results show that mAb background may depend on the specific fixation/permeabilisation kit and that mAb titration using negative and positive control cells is essential to determine the specificity and the sensitivity of the mAb used.


Subject(s)
Antibodies, Monoclonal/immunology , Flow Cytometry/methods , Proto-Oncogene Proteins c-bcl-2/analysis , Tumor Suppressor Protein p53/analysis , Fluorescence , Humans , Light , Proto-Oncogene Proteins c-bcl-2/immunology , Reference Values , Scattering, Radiation , Sensitivity and Specificity , Specimen Handling , Tumor Suppressor Protein p53/immunology
9.
Br J Cancer ; 85(10): 1527-34, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11720440

ABSTRACT

HER2/neu-derived peptides inducing MHC class II-restricted CD4+ T helper lymphocyte (Th) responses, although critical for tumour rejection, are not thoroughly characterized. Here, we report the generation and characterization of CD4+ T cell clones specifically recognizing a HER-2/neu-derived peptide (776-788) [designated HER2(776-788)]. Such clones yielded specific proliferative and cytokine [gamma-interferon(IFN)-gamma] responses when challenged with autologous dendritic cells (DCs) loaded with HER2(776-788). By performing blocking studies with monoclonal antibodies (MAbs) and by using DCs from allogeneic donors sharing certain HLA-DR alleles, we found that HER2(776-788) is a promiscuous peptide presented, at least, by DRB5*0101, DRB1*0701 and DRB1*0405 alleles. One TCRV beta 6.7+ clone recognized the HLA-DRB5*0101+ FM3 melanoma cell line transfected with a full length HER-2/neu cDNA. Moreover, this clone recognized the HER-2/neu+ SKBR3 breast cancer cell line induced to express HLA-DR, thus demonstrating that HER2(776-788) represents a naturally processed and presented epitope. Our data demonstrate that helper peptide HER2(776-788) represents a promiscuous epitope binding to at least three HLA-DR alleles, thus offering a broad population coverage. The use of antigenic peptides presented by major histocompatibility complex (MHC) class II in addition to those presented by class I may improve the therapeutic efficacy of active immunization.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte/immunology , HLA-DR Antigens/physiology , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Cell Line , Cells, Cultured , Clone Cells , Humans , Interferon-gamma/biosynthesis , Lymphocyte Activation , Neoplasms/immunology , Peptides/immunology , Tumor Cells, Cultured
11.
Anticancer Res ; 21(2A): 1199-206, 2001.
Article in English | MEDLINE | ID: mdl-11396164

ABSTRACT

BACKGROUND: We recently demonstrated that supernatants from cultures of peripheral blood mononuclear cells (PBMC) activated with anti-CD3-specific antibody (ACD3S) can induce, upon brief exposure, tumor-reactive lymphocytes in cancer patients. Here, we report that ACD3S can also induce rapid and stable maturation of dendritic cells (DC) which can be used as antigen presenting cells in in vitro protocols and for cancer immunotherapy in vivo. MATERIALS AND METHODS: A short (4-day) priming of CD14+ monocytes with granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4) followed by only a 24 hour-incubation in ACD3S, is sufficient to generate fully mature and stable DC. RESULTS: These DC (i) stimulated strong T cell proliferative responses in the mixed lymphocyte reaction, (ii) when pulsed with unfractionated peptides from autologous tumor membrane extracts activated CD4+ T cells which proliferated in response to the autologous tumor and CD8+ cytotoxic T cells (CTL) which specifically lyse autologous tumor targets and (iii) produced high levels of IL-12. CONCLUSION: ACD3S-treated DC are functionally superior to monocyte-conditioned medium (MCM)-treated DC generated under the same short-term protocol and as efficient as DC induced by the standard 10-day protocol. Our data present an efficient and effective method for generating in a very short period of time, highly mature and functionally competent DC.


Subject(s)
Cell Culture Techniques/methods , Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Lipopolysaccharide Receptors , Aged , Aged, 80 and over , Breast Neoplasms/pathology , CD3 Complex/immunology , CD4-Positive T-Lymphocytes , Cell Differentiation , Cells, Cultured , Female , Humans , Immunophenotyping , Leukocytes, Mononuclear/cytology , Lung Neoplasms/pathology , Male , Middle Aged , Ovarian Neoplasms/pathology , T-Lymphocytes, Cytotoxic
12.
Schizophr Res ; 47(1): 13-25, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11163541

ABSTRACT

A number of immunological parameters were studied in 82 DSM-III-R diagnosed schizophrenic patients (53 first drug-naive and 29 medicated chronic patients) as well as 62 healthy blood donors. The serum levels of interleukin-2 (IL-2), interleukin-1 beta (IL-1beta) and tumour necrosis factor-alpha (TNF-alpha) were measured and correlated with cellular immunity, as assessed by the autologous mixed lymphocyte reaction (AMLR). T lymphocyte subsets were also examined. The above immune parameters were reassessed in a subgroup of 11 first-episode, drug-naive patients 1month after neuroleptic medication. IL-2 serum levels were significantly lower, and IL-1beta and TNF-alpha were significantly higher in schizophrenic patients compared with healthy donors (P<0.001); no significant difference was observed between the two patient groups (medicated and not medicated). Abnormal cytokine serum levels were associated with decreased AMLR responses in vitro. Increased percentages of activated CD4+ and CD16+ natural killer cells, as well as cells expressing ICAM-1 adhesion molecules and IL-2 specific receptors, were detected in the patients. Immunophenotype studies revealed a higher percentage of cells expressing IL-2 receptors in medicated chronic schizophrenic patients compared with drug-naive patients. The abnormal cytokine production in vivo, along with the low AMLR responses in vitro, and the high percentage of activated CD4+ lymphocytes presented in this study suggest alterations in the immune system of schizophrenic patients (medicated or not medicated) consistent with immune activation.


Subject(s)
Antipsychotic Agents/therapeutic use , Cytokines/blood , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Schizophrenia/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Antipsychotic Agents/adverse effects , Chronic Disease , Female , Humans , Lymphocyte Activation/drug effects , Male , Middle Aged , Reference Values , Schizophrenia/drug therapy , T-Lymphocyte Subsets/drug effects
13.
Cancer Immunol Immunother ; 49(8): 449-58, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11043852

ABSTRACT

Peripheral blood mononuclear cells (PBMC) from cancer patients were cultured in vitro with irradiated autologous tumor cells isolated from malignant effusions (mixed lymphocyte tumor cultures, MLTC) and low-dose (50 IU/ml) recombinant interleukin-2 (IL-2). The combination of IL-2 and prothymosin alpha (ProTalpha) resulted in a greater PBMC-induced response to the autologous tumor than that brought about by IL-2 alone. In particular, ProTalpha specifically enhanced the CD4+ T-cell-mediated proliferation against the autologous tumor. CD4+ T cells seemed to recognize tumor antigens presented by HLA-DR molecules expressed on the autologous monocytes, since preincubation of the latter with an anti-HLA-DR monoclonal antibody (mAb) abrogated the response. In addition, MLTC set up with IL-2 and ProTalpha also generated more MHC-class-I-restricted cytotoxic T lymphocytes (CTL) against the autologous tumor than did MLTC set up with IL-2 alone. The MLTC-induced CTL contained high levels of cytoplasmic perforin and their development was strictly dependent on the presence of both autologous CD4+ T cells and monocytes. In the absence of either population there was a strong impairment of both proliferative and cytotoxic responses which was not restored by the presence of ProTalpha. In contrast, when both cell populations were present, ProTalpha exerted optimal enhancement of CD4+ T cell proliferation, which was associated with potentiated CTL responses. Our data emphasize the role of ProTalpha for the enhancement of IL-2-induced CTL responses against autologous tumor cells. Such responses require collaborative interactions between CD4+, CD8+ T cells and monocytes as antigen-presenting cells. Our data are relevant for adoptive immunotherapeutic settings utilizing IL-2 and ProTalpha-induced autologous-tumor-specific CTL.


Subject(s)
Carcinoma/therapy , Interleukin-2/therapeutic use , Protein Precursors/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Thymosin/analogs & derivatives , Thymosin/therapeutic use , Aged , Animals , Breast Neoplasms/therapy , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cattle , Cell Communication , Cell Division/drug effects , Cell Division/immunology , Cell Separation , Cells, Cultured , Female , Humans , Lung Neoplasms/therapy , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Ovarian Neoplasms/therapy , Phenotype , Thymus Gland/chemistry , Time Factors , Tumor Cells, Cultured
14.
Eur J Immunol ; 30(7): 1957-66, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10940885

ABSTRACT

The response of lymphokine-activated killer (LAK) and natural killer (NK) cells from mice lacking tumor necrosis factor-alpha (TNF-alpha-/- mice) was impaired in cytotoxicity assays against various tumor cell targets. Furthermore, allogeneic cytotoxic T lymphocyte (CTL) responses were also impaired as compared to TNF-alpha+/+ littermates (control mice). Cytotoxicity was restored both upon in vitro incubation of TNF-alpha-/- lymphocytes with recombinant TNF-alpha (rTNF-alpha) or upon in vivo treatment of TNF-alpha-/- mice with rTNF-alpha. Using combinations of monoclonal antibodies we were able to show that TNF-alpha-/- effector lymphocytes exhibit both perforin- and Fas ligand-based cytotoxicity. Furthermore, upon in vivo administration of rTNF-alpha these effectors, in addition to perforin and Fas ligand, are also armed with TNF-alpha cytotoxic molecules, thus resembling to the cytotoxic effectors from control mice. In a tumor model, immunized TNF-alpha-/- mice failed to reject the syngeneic fibrosarcoma MC57X, but did so when injected with rTNF-alpha. In vivo administration of anti-TNF-alpha mAb neutralized the effect of rTNF-alpha supporting the growth of MC57X cells. Our data provide novel evidence for TNF-alpha as an essential factor in (i) controlling cytotoxicity in vitro and in vivo and (ii) promoting tumor rejection in vivo.


Subject(s)
Fibrosarcoma/immunology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Killer Cells, Lymphokine-Activated/cytology , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Neoplasms, Experimental/immunology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes, Cytotoxic/cytology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology
15.
J Immunol ; 164(7): 3902-12, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10725753

ABSTRACT

This study focuses on the specific CD4+ T cell requirement for optimal induction of cytotoxicity against MHC class II negative autologous tumors (AuTu) collected from patients with various types of cancer at advanced stages. CD4+ T cells were induced in cultures of cancer patients' malignant effusion-associated mononuclear cells with irradiated AuTu (mixed lymphocyte tumor cultures (MLTC)) in the presence of recombinant IL-2 and recombinant IL-7. Tumor-specific CD4+ T cells did not directly recognize the AuTu cells, but there was an MHC class II-restricted cross-priming by autologous dendritic cells (DCs), used as APC. CD8+ CTL, also induced during the MLTC, lysed specifically AuTu cells or DCs pulsed with AuTu peptide extracts (acid wash extracts (AWE)) in an MHC class I-restricted manner. Removal of CD4+ T cells or DCs from the MLTC drastically reduced the CD8+ CTL-mediated cytotoxic response against the AuTu. AWE-pulsed DCs preincubated with autologous CD4+ T cells were able, in the absence of CD4+ T cells, to stimulate CD8+ T cells to lyse autologous tumor targets. Such activated CD8+ T cells produced IL-2, IFN-gamma, TNF-alpha, and GM-CSF. The process of the activation of AWE-pulsed DCs by CD4+ T cells could be inhibited with anti-CD40 ligand mAb. Moreover, the role of CD4+ T cells in activating AWE-pulsed DCs was undertaken by anti-CD40 mAb. Our data demonstrate for the first time in patients with metastatic cancer the essential role of CD4+ Th cell-activated DCs for optimal CD8+ T cell-mediated killing of autologous tumors and provide the basis for the design of novel protocols in cellular adoptive immunotherapy of cancer, utilizing synthetic peptides capable of inducing T cell help in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Aged , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cell Extracts/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Female , Humans , Immunotherapy, Adoptive/methods , Lymphocyte Culture Test, Mixed , Male , Middle Aged , Neoplasms/pathology , Trifluoroacetic Acid , Tumor Cells, Cultured
16.
Cancer Immunol Immunother ; 48(2-3): 71-84, 1999.
Article in English | MEDLINE | ID: mdl-10414460

ABSTRACT

Anti-CD3 monoclonal antibody (mAb) activates in vitro peripheral blood mononuclear cells (PBMC) to lyse a variety of tumor cell lines in a non-major histocompatibility-complex(MHC)-restricted manner [subsequently referred to as anti-CD3-activated killer (AAK) cytotoxicity]. Prothymosin alpha (ProTalpha) is a biological response modifier that exerts its effects primarily on mononuclear cells, especially when these cells' effector functions are impaired. In this study, we report that ProTalpha enhances the AAK cytotoxicity in PBMC from healthy donors. This effect was more profound with cancer patients' PBMC, which were deficient in their ability to respond with enhanced AAK cytotoxicity upon in vitro stimulation with anti-CD3. Thus, cancer patients' PBMC, activated with a combination of anti-CD3 and ProTalpha, exhibited increased AAK activity and efficiently lysed both autologous tumor and Daudi targets. The ProTalpha effect on PBMC was demonstrated to involve stimulation of adhesion molecules (CD2, CD18, CD54, CD49f) and CD25 expression, up-regulation of perforin mRNA transcription, increased numbers of perforin-positive (+) cells and elevated production of interleukin-2 (IL-2), interleukin-1beta (IL-1beta) and tumor necrosis factor alpha (TNFalpha). Moreover, effector CD8+ and CD56+ cells pretreated with anti-CD3 and ProTalpha contained high cytoplasmic perforin levels and increased expression of IL-1beta- and TNFalpha-specific receptors. The induction of autologous-tumor-reactive CD8+ and CD56+ lymphocytes in anti-CD3-activated PBMC by ProTalpha provides an alternative protocol aimed at the improvement of clinical results in cellular adoptive immunotherapy of cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD3 Complex/immunology , Neoplasms/therapy , Protein Precursors/pharmacology , T-Lymphocytes/immunology , Thymosin/analogs & derivatives , Aged , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Female , Humans , Immunotherapy, Adoptive , Male , Membrane Glycoproteins/biosynthesis , Middle Aged , Neoplasms/immunology , Perforin , Pore Forming Cytotoxic Proteins , Receptors, Interleukin-2/analysis , Thymosin/pharmacology
17.
Immunopharmacol Immunotoxicol ; 20(3): 355-72, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9736441

ABSTRACT

This report demonstrates that in vitro activation of human cells with the beta-galactoside-specific lectin from mistletoe (ML-I) or interleukin-2 (IL-2) results in different patterns of activation and function of cytotoxic cells. It is now well established that natural killer (NK) and lymphokine-activated killer (LAK) cytotoxicity is mainly mediated by resting (NK) and IL-2-activated (LAK) CD56-positive (+) cells respectively. Culture of peripheral blood lymphocytes (PBL) for 3 days with ML-I led to expansion and activation of T cells which demonstrated NK- and LAK-like cytotoxicity. T lymphocyte subset analysis revealed that in total PBL, ML-I preferentially stimulated and expanded CD8+ T cells which mediated the cytotoxic effect. Incubation of highly purified CD8+ T cells alone with ML-I did not lead to induction of cytotoxicity, which required the presence of both CD4+ and CD14+ (monocytes) cells, suggesting that ML-I does not exert a direct effect on CD8+ T cells. Activation of PBL with both ML-I and IL-2 resulted in simultaneous induction of T and CD56+ cell-mediated NK and LAK cytotoxicity. These data suggest that treatment with ML-I and IL-2 might provide an approach to induce maximum cytotoxicity against tumors and to recruit both T and NK cells for tumor therapy.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Plant Preparations , Plant Proteins , Toxins, Biological/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Drug Synergism , Humans , Ribosome Inactivating Proteins, Type 2
18.
Anticancer Res ; 18(3A): 1501-8, 1998.
Article in English | MEDLINE | ID: mdl-9673361

ABSTRACT

The prognosis of breast cancer is of major clinical importance and several histopathological, biochemical and immunological variables have been reported to be useful prognostic factors. In the present study, we investigated the clinical significance of the levels of alpha-thymosins in relation to established prognostic factors, both in breast cancer and non-malignant breast lesions, alpha-thymosin levels were measured in breast tissue extracts by specific radioimmunoassays (RIAs) developed for human prothymosin alpha (ProT alpha) and parathymosin alpha (ParaT alpha) and were found to be significantly higher (up to 17.2-fold) in malignant but not in benign breast lesions, as compared to the values of the neighbouring tissues. When alpha-thymosin levels of the tumor samples were correlated with various known prognostic parameters a statistically significant correlation (p < 0.05) was observed between the levels of ProT alpha in malignant tissues to the grade of cancer and the lymph node status of the patient. An association between ProT alpha levels with increase in risk of death from breast cancer was also noticed. These results suggest that the expression of alpha-thymosins in human breast cancer a) depends on the proliferation status of the tumor, b) associates with established prognostic factors describing the metastatic potential of the tumor and c) is related to the overall survival of the patient. The fact that such relationships hold only for cancer tissues encourages the future use of alpha-thymosins as potent prognostic factors in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/surgery , Thymosin/analysis , Adult , Aged , Aged, 80 and over , Antibody Specificity , Breast Neoplasms/mortality , Cross Reactions , Female , Humans , Lymphatic Metastasis , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Predictive Value of Tests , Prognosis , Radioimmunoassay , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Survival Rate , Thymosin/analogs & derivatives
19.
Eur J Cancer ; 33(8): 1202-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9301443

ABSTRACT

In this report, we studied the immunorestorative properties of subcutaneously administered granulocyte-macrophage colony-stimulating factor (GM-CSF) in patients with refractory solid tumours receiving second-line chemotherapy. Such patients exhibit abnormal immune responses in vivo and in vitro and, therefore, it was of interest to examine the effect of GM-CSF-induced immunomodulation on clinical response. We examined patients with primary malignant carcinomas (head and neck, n = 10; urogenital tract, n = 17; penis n = 6; colorectal, n = 8) who were treated with carboplatin (JM8), 300 ng/m2 on days 1 and 22, leucovorin (LV), 200 mg/m2 plus 5-fluoracil (5-FU), 500 mg/m2 on days 8, 15 and 29 and four cycles of daily injections with placebo or GM-CSF, 300 micrograms/day on days 3-6, 10-13, 17-20 and 24-27. Peripheral blood was collected from the patients one day after the end of each of the four-cycle injections with placebo or GM-CSF, namely on days 7, 14, 21 and 28. Peripheral blood mononuclear cells (PBMC) were tested in the autologous mixed lymphocyte reaction (AMLR) and for natural killer (NK) or lymphokine-activated killer (LAK) cell activity. Cytokine levels in serum were measured by immunoenzymatic (ELISA) assay. A total of 21 patients received a four-cycle regimen with GM-CSF (Group 1) and 20 were similarly treated with placebo (Group 2). All received standard chemotherapy as outlined above. Before GM-CSF treatment, all patients exhibited increased serum levels of interleukin-1 (IL-1 beta), tumour necrosis factor-alpha (TNF-alpha), IL-6 and prostaglandin E2 (PGE2) and decreased serum levels of IL-2. Cellular immune responses (AMLR, NK- and LAK-cytotoxicity) were also low in all patients. Five patients from Group 1 had a PR (partial response), 2 patients had CR (complete response), and 14 patients had stable disease. Seven patients from Group 2 showed progressive disease, 3 had a PR and 10 had stable disease. All immune parameters were significantly improved during treatment in Group 1 but remained unchanged or even deteriorated in Group 2. Administration of GM-CSF during treatment of cancer patients with conventional chemotherapeutic drugs results in a marked potentiation of deficient cellular immune responses in vitro and a change towards normalisation of cytokine serum levels. The results reported herein support the use of GM-CSF as immunopotentiator during chemotherapy, but more patients must be studied before definite conclusions can be drawn.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Adult , Aged , Carboplatin/administration & dosage , Cytokines/blood , Cytotoxicity, Immunologic , Female , Fluorouracil/administration & dosage , Humans , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Lymphocyte Culture Test, Mixed , Male , Middle Aged , Neoplasms/drug therapy
20.
Eur J Clin Invest ; 27(4): 308-15, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9134379

ABSTRACT

Our objective was to investigate the initial levels of circulating proinflammatory cytokines, such as interleukin 1 beta (IL-1 beta), interleukin 6 (IL-6), and tumour necrosis factor alpha (TNF-alpha), of certain acute-phase proteins, such as C-reactive protein (CRP), fibrinogen (FBN) and albumin, and of the glycoprotein fibronectin at presentation and their daily variation during the clinical course of community-acquired pneumonia (CAP) in relation to clinical and laboratory indices of infection. Thirty otherwise healthy hospitalized patients aged 48 +/- 3 years (mean +/- SEM) and with bacteriologically confirmed CAP were studied prospectively. IL-1 beta and IL-6 were found to be 15-fold higher on admission (122 +/- 9 pg mL-1 and 60 +/- 4 pg mL-1 respectively), whereas TNF-alpha was three-fold higher (102 +/- 5 pg mL-1) than those of controls, all of them showing a decline towards normal. Initial CRP levels were increased 90-fold (416 +/- 1 mg L-1), whereas fibronectin levels were reduced (242 +/- 9 mg dL-1). The presence of parapneumonic effusion was associated with a higher TNF-alpha serum level (127 +/- 7 vs. 86 +/- 4 pg mL-1, P = 0.0002), a more rapid daily decline in TNF-alpha (-7.2 +/- 0.7 vs. -3.8 +/- 0.5 pg mL-1 day-1, P = 0.0005), a slower rate of decline in CRP (-42.8 +/- 3.0 vs. -54.6 +/- 3.0 mg L-1 day-1, P = 0.02) and a slower rate of increase in FBN (5.9 +/- 1.0 vs. 11.7 +/- 1.0 mg dL-1 day-1), P = 0.001]. Furthermore, daily progression of serum levels of cytokines and acute-phase proteins correlated strongly with pyrexia, erythrocyte sedimentation rate (ESR), neutrophil count, alveolar-arterial oxygen difference and radiographic resolution, clinically manifested by improvement in the patients' condition.


Subject(s)
Acute-Phase Proteins/analysis , Community-Acquired Infections/blood , Cytokines/blood , Pneumonia/blood , Adult , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...