Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Int J Cancer ; 130(7): 1577-89, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-21618510

ABSTRACT

Cetuximab is a human/mouse chimeric IgG1 monoclonal antibody (mAb) to epidermal growth factor receptor, approved for colorectal carcinoma treatment in combination with chemotherapy. The immune-mediated effects elicited by its human fraction of crystallization moiety might critically contribute to the overall anti-tumor effectiveness of the antibody. We therefore investigated cetuximab ability to promote colon cancer cell opsonization and phagocytosis by human dendritic cells (DCs) that are subsequently engaged in antigen-cross presentation to cytotoxic T-lymphocyte (CTL) precursors. Human colon cancer cell lines were evaluated for susceptibility to DC-mediated phagocytosis before and after treatment with chemotherapy ± cetuximab in vitro. Human DCs loaded with control or drug-treated cetuximab-coated colon cancer cells were used to in vitro generate cytotoxic T cell clones from peripheral blood mononuclear cells of human leucocyte antigen-A(*)02.01(+) donors. T-cell cultures were characterized for immune-phenotype and tumor-antigen specific CTL activity. The results confirmed that treatment of tumor cells with irinotecan + L-folinate + 5-flurouracil (ILF) or with gemcitabine + ILF increased tumor antigen expression. Moreover, malignant cells exposed to chemotherapy and cetuximab were highly susceptible to phagocytosis by human DCs and were able to promote their activation. The consequent DC-mediated cross-priming of antigens derived from mAb-covered/drug-treated cancer cells elicited a robust CTL anti-tumor response. On the basis of our data, we suggest a possible involvement of CTL-dependent immunity in cetuximab anti-cancer effects.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Dendritic Cells/drug effects , Phagocytosis/drug effects , T-Lymphocytes, Cytotoxic/drug effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/immunology , Cell Line, Tumor , Cetuximab , Cross-Priming/drug effects , Cross-Priming/immunology , Dendritic Cells/immunology , HT29 Cells , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Phagocytosis/immunology , T-Lymphocytes, Cytotoxic/immunology
2.
J Chemother ; 6(5): 328-36, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7532216

ABSTRACT

Previous studies have shown that treatment of leukemia-bearing mice with triazene compounds results in a profound alteration of the immunological properties of leukemic cells. These cells become highly immunogenic and susceptible to natural immunity (NI). Moreover, in a pilot clinical study, dacarbazine was found to suppress bone marrow blasts in patients with acute non-lymphoblastic leukemias. The cytotoxic mechanism involved could be of biochemical and immunological origin as well. Therefore experiments were carried out to test whether triazenes could influence the susceptibility of blast cells to human NI effector lymphocytes (represented, at least in part, by NK cells). The results obtained with target Epstein-Barr virus (EBV)-immortalized B cells and effector cells of different donors, showed that: (a) multiple in vitro treatments of lymphoblastoid cells with methyl-triazene-benzoic acid (MTBA, a triazene compound active in vitro), gave origin to lines that were more resistant than the parental lines to the antitumor effects of MTBA; (b) MTBA-treated lines were more susceptible (37.5% of cases), or less susceptible (31.2% of cases) to NI than parental cells. Effector lymphocytes of various donors recognized different changes in susceptibility to natural killer (NK)-mediated lysis; (c) treatment of parental or MTBA-treated lines with interferon-beta reduced target cell susceptibility to NK-mediated cytolysis, but increased NK activity and lymphoblast chemosensitivity to MTBA.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/immunology , Triazenes/pharmacology , B-Lymphocytes/drug effects , Cell Line , Humans , Interferons/pharmacology , Methyltransferases/metabolism , O(6)-Methylguanine-DNA Methyltransferase
3.
J Chemother ; 14(4): 412-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12420861

ABSTRACT

We describe a novel method for detecting micrometastasis in the blood stream of cancer patients based on RT-PCR amplification of tumor-associated carcinoembryonic antigen (CEA) mRNA. To increase sensitivity and specificity of RT-PCR, CEA transcript was selectively up-regulated in cancer cells by exposure of peripheral blood to non-toxic concentrations of staurosporine (ST). Thereafter, polyA(+) RNA was extracted from tumor cells captured by means of magnetic beads coated with a monoclonal antibody against a common human epithelial antigen. Finally, RNA was subjected to RT-PCR analysis of CEA transcript. Using this approach, we demonstrated an ST-mediated increase in CEA transcript in blood specimens collected from a patient with metastatic colon cancer before receiving treatment with 5-fluorouracil/leucovorin. After a few cycles of chemotherapy, CEA-positive tumor cells were no longer detected. Clinical follow-up of this patient indicated that treatment with chemotherapy induced a dramatic reduction in liver metastasis. Therefore, it can be hypothesized that lack of CEA transcript detection might be consistent with disappearance or at least marked reduction of circulating tumor cells.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoembryonic Antigen/genetics , Colonic Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Neoplastic Cells, Circulating , RNA, Messenger/analysis , Adenocarcinoma/blood , Adenocarcinoma/secondary , Carcinoembryonic Antigen/metabolism , Colonic Neoplasms/blood , Colonic Neoplasms/pathology , DNA Primers/chemistry , Female , Fluorouracil/administration & dosage , Humans , Immunomagnetic Separation , Leucovorin/administration & dosage , Liver Neoplasms/blood , Liver Neoplasms/secondary , Middle Aged , Prognosis , RNA, Neoplasm/blood , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity
4.
J Chemother ; 15(2): 173-83, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12797396

ABSTRACT

O6-alkylguanine-DNA alkyltransferase (OGAT) and the mismatch repair system (MRS) play a crucial role in the susceptibility of tumor cells to the cytotoxic effects of agents that generate O6-methylguanine in DNA, including the triazene compound temozolomide (TMZ). Studies performed with peripheral blood mononuclear cells (MNC) showed that TMZ was scarcely active on lymphocyte functions not dependent on cell proliferation (e.g. NK activity and cytokine-mediated induction of CD1b molecule in adherent MNC). In contrast, TMZ depressed proliferation and lymphokine activated killer (LAK) cell generation in response to IL-2. In this case, a reasonably good inverse relationship was found between OGAT levels of MNC and their susceptibility to TMZ. This study also analyzed the ratio of the toxic effect of TMZ on MNC and on tumor cells (i.e. "Tumor-Immune Function Toxicity Index", TIFTI). A particularly favorable TIFTI can be obtained when OGAT levels are extremely high in MNC and markedly low in tumor cells. This holds true for MRS-proficient neoplastic cells, but not for MRS-deficient tumors. In conclusion, strategies aimed at modulating OGAT and MRS may improve the clinical response to TMZ. However, the use of OGAT inhibitors to potentiate the antitumor activity of TMZ might result in a concomitant increase of the immunosuppressive effects of the drug, thus reducing the relative TIFTI.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Repair , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Leukocytes, Mononuclear/drug effects , O(6)-Methylguanine-DNA Methyltransferase/pharmacology , Burkitt Lymphoma/pathology , Cell Division , DNA Damage , Drug Resistance, Neoplasm , Humans , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated , Leukemia, Erythroblastic, Acute/pathology , Leukocytes, Mononuclear/physiology , Lymphocytes/physiology , Melanoma/pathology , O(6)-Methylguanine-DNA Methyltransferase/drug effects , Skin Neoplasms/pathology , Temozolomide , Tumor Cells, Cultured
5.
J Chemother ; 16(5): 479-86, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15565916

ABSTRACT

Preclinical studies based on a "simulation design", were performed with cultured melanoma cells prelabeled with 51Cr, added to normal blood and subjected to separation and recognition steps. Mononuclear cells (MNC) were isolated on ficollhypaque gradient, and melanoma cells were separated from lymphocytes using anti-CD45 immunomagnetic beads. Malignant cells were then recognized by measuring telomerase activity (TRAP and TRAP-ELISA assays). It was found that: (a)recovery of prelabeled cells present in MNC did not exceed 75%; (b) further recovery of prelabeled cells after separation from lymphocytes did not exceed 68%. Therefore, the overall recovery of prelabeled cells did not exceed 48%; (c) the entire procedure was able to reliably detect as few as 30 malignant cells added to normal blood, providing a telomerase signal significantly higher than that found in absence of melanoma cells. These results furnish the technical bases for developing a tumor detection assay in the blood of melanoma patients.


Subject(s)
Biomarkers, Tumor/blood , Melanoma/diagnosis , Neoplastic Cells, Circulating , Skin Neoplasms/diagnosis , Telomerase/blood , Cell Line, Tumor , Humans , Melanoma/pathology , Predictive Value of Tests , Sensitivity and Specificity , Skin Neoplasms/pathology
6.
Curr Med Chem ; 20(19): 2389-401, 2013.
Article in English | MEDLINE | ID: mdl-23521681

ABSTRACT

Acute myeloid leukemia (AML) is a highly lethal disease, especially in old patients. Chemoresistance and the absence of host immune responses against autochthonous malignancy play a major role in the poor prognosis of AML. The triazene compounds Dacarbazine and Temozolomide are monofunctional alkylators that donate methyl groups to many sites in DNA, including the O(6)-position of guanine producing O(6)-methylguanine (O(6)-MeG). If not repaired, O(6)-MeG frequently mispairs with thymine during DNA duplication. O(6)-MeG:T mismatches can be recognized by the mismatch repair (MMR) system which activates a cascade of molecular events leading to cell cycle arrest and cell death. If MMR is defective, cells continue to divide and GC → AT transition mutations occur. In preclinical models, such mutations can lead to the appearance of abnormal proteins containing non-self peptides ("chemical xenogenization" CX) that can be recognized by host cell-mediated immunity. Repair of O(6)-MeG is achieved by the DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), which removes the methyl adduct in an autoinactivating stoichiometric reaction. High MGMT levels attenuate the pharmacodynamic effects of triazenes. In the last few years, triazenes, alone or with MGMT inhibitors, have been tested in AML. In view of their potential activity as CX inducers, triazenes could offer the additional advantage of host anti-leukemia immune responses. The present paper describes several studies of leukemia treatment with triazenes and a case of acute refractory leukemia with massive skin infiltration by malignant cells. Treatment with Temozolomide and Lomeguatrib, a potent MGMT inhibitor, produced a huge, although transient, blastolysis and complete disappearance of all skin lesions.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Triazenes/therapeutic use , Animals , Antineoplastic Agents, Alkylating/chemistry , Dacarbazine/chemistry , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Temozolomide , Triazenes/chemistry
8.
Pharmacol Res ; 53(4): 317-23, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16412662

ABSTRACT

Anti-tumour activity of triazene compounds of clinical interest [i.e. dacarbazine and temozolomide (TMZ)] relies mainly on the generation of methyl adducts to purine bases of DNA. Two DNA repair enzyme systems, i.e. the O6-guanine-alkyl-transferase (MGMT) and mismatch repair (MMR), play a predominant role in conditioning the cytotoxic effects of triazenes. In particular, high levels of MGMT associated with target cells are responsible of resistance to triazenes. On the contrary, the presence of MMR is required for the cytotoxic effects of these compounds. Previous studies performed by our group and a more recent clinical investigation reported by Karen Seiter, pointed out that triazene compounds could play an important role in the treatment of refractory acute leukaemia. Leukaemia blasts, especially of lymphoblastic leukaemia, show frequently high levels of MGMT activity. Therefore, it reasonable to hypothesize that combined treatment of leukaemia patients with triazene compounds along with MGMT inhibitors could lead to a better control of the disease. PaTrin-2 (O6-(4-bromothenyl)guanine, PAT) is a potent and scarcely toxic MGMT inhibitor recently introduced in clinical trials. This drug is used in combination with triazene compounds in order to augment their anti-tumour efficacy against neoplastic cells endowed with high MGMT activity. The present report describes, for the first time, pre-clinical in vitro studies on the cytotoxic activity of combined treatment with PAT+TMZ against long-term cultured leukaemia cells and primary leukaemia blasts obtained from patients with acute lymphoblastic leukaemia or acute myeloblastic leukaemia. The results point out that, both in long-term cultured leukaemia cell lines and in primary blast samples, PAT could improve dramatically the sensitivity of malignant cells to the cytotoxic effects of TMZ. This sensitizing effect is detectable when leukaemia cells show resistance mechanisms based on a MGMT-proficient phenotype. On the contrary, when resistance to TMZ is dependent on MMR deficiency, no influence of PAT can be detected in various experimental conditions. In conclusion, these results appear to provide disease-oriented rational basis to design novel clinical protocols for the treatment of acute leukaemia with combined administration of PAT and triazene compounds.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dacarbazine/analogs & derivatives , Guanine/analogs & derivatives , Leukemia, Myeloid/drug therapy , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Acute Disease , Dacarbazine/pharmacology , Drug Synergism , Guanine/pharmacology , HL-60 Cells , Humans , Leukemia, Myeloid/blood , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/pathology , Temozolomide , Tumor Cells, Cultured
9.
Immunopharmacol Immunotoxicol ; 16(4): 695-715, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7876468

ABSTRACT

It was shown that Dacarbazine and other triazene compounds render murine leukemias highly immunogenic and susceptible to natural immunity (NI). In addition a pilot clinical study revealed that Dacarbazine can be cytotoxic for bone marrow blasts in patients with acute non-lymphoblastic leukemias through a mechanism that could be, at least in part, of immunological origin. However triazenes depress antigen-dependent responses and NI, whereas interferons, including interferon-beta (IFN), antagonize drug-induced impairment of NI. Therefore the complex interaction between triazenes and IFN on NI effector (i.e. NK) lymphocytes and human target lymphoblastoid cells has been investigated. The results show that: (a) IFN increases NK activity and antagonizes the depressive effects of methyl-triazene-benzoic acid (MTBA, an in vitro active triazene compound) on the NK function; (b) a lymphoblastoid cell line exposed to multiple in vitro treatments with MTBA, shows increased growth rate, augmented chemoresistance to MTBA, and higher susceptibility to NI than parental cells; (c) as expected IFN pretreatment down-regulates the susceptibility of lymphoblastoid cells to NK effectors; (d) however a net "therapeutic gain" was found if the overall influence of MTBA+IFN on target and effector cells is considered.


Subject(s)
Interferon-beta/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes/immunology , Triazenes/pharmacology , Antibodies, Monoclonal/immunology , Cell Line , Cytotoxicity Tests, Immunologic , Flow Cytometry , Humans , Methyltransferases/metabolism , O(6)-Methylguanine-DNA Methyltransferase
10.
Proc Natl Acad Sci U S A ; 98(13): 7164-9, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11416201

ABSTRACT

Members of hereditary nonpolyposis colon cancer (HNPCC) families harboring heterozygous germline mutations in the DNA mismatch repair genes hMSH2 or hMLH1 present with tumors generally two to three decades earlier than individuals with nonfamilial sporadic colon cancer. We searched for phenotypic features that might predispose heterozygous cells from HNPCC kindreds to malignant transformation. hMSH2(+/-) lymphoblastoid cell lines were found to be on average about 4-fold more tolerant than wild-type cells to killing by the methylating agent temozolomide, a phenotype that is invariably linked with impairment of the mismatch repair system. This finding was associated with an average 2-fold decrease of the steady-state level of hMSH2 protein in hMSH2(+/-) cell lines. In contrast, hMLH1(+/-) heterozygous cells were indistinguishable from normal controls in these assays. Thus, despite the fact that HNPCC families harboring mutations in hMSH2 or hMLH1 cannot be distinguished clinically, the early stages of the carcinogenic process in hMSH2 and hMLH1 mutation carriers may be different. Should hMSH2(+/-) colonocytes and lymphoblasts harbor a similar phenotype, the increased tolerance of the former to DNA-damaging agents present in the human colon may play a key role in the initiation of the carcinogenic process.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , DNA Repair , DNA-Binding Proteins , Dacarbazine/toxicity , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Carrier Proteins , Cell Line , Cell Survival/drug effects , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Dacarbazine/analogs & derivatives , Heterozygote , Humans , Lymphocytes , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins , Temozolomide , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL