Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
Haemophilia ; 23(1): 67-76, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27480487

ABSTRACT

INTRODUCTION: Surgery in patients with haemophilia B carries a high risk of excessive bleeding and requires adequate haemostatic control until wound healing. Nonacog beta pegol, a long-acting recombinant glycoPEGylated factor IX (FIX), was used in the perioperative management of patients undergoing major surgery. AIM: To evaluate the efficacy and safety of nonacog beta pegol in patients with haemophilia B who undergo major surgery. METHODS: This was an open-label, multicentre, non-controlled surgery trial aimed at assessing peri- and postoperative efficacy and safety of nonacog beta pegol in 13 previously treated patients with haemophilia B. All patients received a preoperative nonacog beta pegol bolus injection of 80 IU kg-1 . Postoperatively, the patients received fixed nonacog beta pegol doses of 40 IU kg-1 , repeated at the investigator's discretion. Safety assessments included monitoring of immunogenicity and adverse events. RESULTS: Intraoperative haemostatic effect was rated 'excellent' or 'good' in all 13 cases. Apart from the preoperative injection, none of the patients needed additional doses of nonacog beta pegol on the day of surgery. The median number of postoperative doses of nonacog beta pegol was 2.0 from days 1 to 6 and 1.5 from days 7 to 13. No unexpected intra- or postoperative complications were observed including deaths or thromboembolic events. No patients developed inhibitors. CONCLUSIONS: These results indicated that nonacog beta pegol was safe and effective in the perioperative setting, allowing major surgical interventions in patients with haemophilia B with minimal peri- and postoperative concentrate consumption and infrequent injections as reported with standard FIX products.


Subject(s)
Factor IX/therapeutic use , Hemophilia B/drug therapy , Hemophilia B/surgery , Hemostatics/therapeutic use , Recombinant Proteins/therapeutic use , Adolescent , Adult , Aged , Disease Management , Humans , Male , Middle Aged , Young Adult
3.
Apoptosis ; 9(2): 193-204, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15004516

ABSTRACT

Inhibition of the proteasome, a multicatalytic proteinase complex, is an attractive approach to cancer therapy. Here we report that a selective inhibitor of the chymotrypsin-like activity of the proteasome, PSI (N-benzyloxycarbonyl-Ile-Glu(O-t-butyl)-Ala-leucinal) may inhibit growth of solid tumors not only through apoptosis induction, but also indirectly--through inhibition of angiogenesis. Two murine tumors: colon adenocarcinoma (C-26) and Lewis lung carcinoma (3LL) were chosen to study the antitumor effect of PSI. In an in vivo model of local tumor growth, PSI exerted significant antitumor effects against C-26 colon carcinoma, but not against 3LL lung carcinoma. Retardation of tumor growth was observed in mice treated with both 10 nmoles and 100 nmoles doses of PSI and in the latter group prolongation of the survival time of tumor-bearing mice was observed. PSI inhibited angiogenesis in the C-26 growing tumors with no such effect in 3LL tumors. Unexpectedly, that activity was associated with upregulation of vascular endothelial growth factor (VEGF) at the level of mRNA expression and protein production in C-26 tumors treated with PSI. C-26 cells treated with PSI produced increased amounts of VEGF in vitro in a dose- and time-dependent manner. We demonstrated that in C-26 colon adenocarcionoma higher VEGF production may render endothelial cells susceptible to the proapoptotic activity of PSI and is associated with inhibition of tumor growth.


Subject(s)
Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Oligopeptides/pharmacology , Proteasome Inhibitors , Vascular Endothelial Growth Factor A/metabolism , Animals , Carcinoma, Lewis Lung/drug therapy , Mice , Oligopeptides/therapeutic use
4.
Leukemia ; 15(4): 613-20, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11368364

ABSTRACT

In this study we investigated the efficacy of a combination of IL-12 and 5-FU, a chemotherapeutic exerting several immunomodulatory effects, in murine L1210 leukemia. Mice inoculated with 1 x 10(5) leukemia cells were treated with a single dose of 5-FU (50 mg/kg) and seven daily doses of IL-12 (100 ng/dose), and were observed for survival. Treatment with IL-12 or 5-FU given alone produced moderate anti-leukemic effects. However, combination of both drugs resulted in a significant prolongation of mouse survival time. Importantly, there were 70% of long-term (>60 days) survivors among mice treated with both agents simultaneously. Moreover, we observed 100% of long-term survivors when mice were treated with a minimally increased dose of IL-12 (170 ng) in combination with 5-FU (50 mg/kg). The antileukemic effects were completely abrogated in scid/scid mice and in mice depleted of peritoneal macrophages and significantly decreased after administration of anti-CD3+, anti-CD4+ or anti-CD8+ monoclonal antibodies. Administration of anti-NK1.1 antibodies did not decrease the antileukemic effects indicating that NK cells are not important effectors of this treatment regimen. Collectively, these results indicate that the combination of IL-12 and 5-FU is inducing strong antileukemic responses that are dependent on the presence and activity of macrophages and T lymphocytes and warrant further studies of combined chemo-immunotherapy with IL-12.


Subject(s)
Fluorouracil/administration & dosage , Interleukin-12/administration & dosage , Leukemia L1210/therapy , Animals , Female , Interferon-gamma/blood , Killer Cells, Natural/immunology , Leukemia L1210/immunology , Leukemia L1210/mortality , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, SCID
5.
Cancer Lett ; 164(2): 143-8, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11179828

ABSTRACT

Butyric acid has been known to inhibit growth and to induce differentiation of a variety of tumor cells. Butyrate-treated tumor cells have also been observed to undergo apoptosis. Although butyrate compounds have demonstrated antitumor activity in murine tumor models and have already been admitted to clinical trials in tumor patients, the exact mechanism of their antitumor effects has not been elucidated. The results of our study showed antitumor activity of tributyrin, a butyric acid prodrug, in murine melanoma model and are strongly suggestive that antiangiogenic effects could participate in antitumor effects of butyrate compounds in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma, Experimental/prevention & control , Prodrugs/pharmacology , Triglycerides/pharmacology , Animals , Blotting, Western , Butyric Acid/metabolism , Butyric Acid/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Neovascularization, Pathologic , Time Factors , Tumor Cells, Cultured
6.
Anticancer Res ; 21(6A): 4001-4, 2001.
Article in English | MEDLINE | ID: mdl-11911283

ABSTRACT

Butyric acid (NaBut) and its derivatives are well-known agents eliciting tumor cell differentiation and apoptosis. In experimental models, NaBut is also used to enhance the efficacy of viral vectors. With the use of B78 murine melanoma cells transduced with the retroviral vector containing human tumor necrosis factor alpha (hTNF-alpha) gene, we investigated the ability of NaBut to increase the cytokine expression. We observed an increase in hTNF-alpha expression in vitro after incubation with NaBut. We also describe that the NaBut pro-drug tributyrin is able to increase hTNF-alpha expression in transduced B78 cells in a tumor vaccination model in mice. This observation strongly suggests a novel potential role for NaBut and its derivatives in tumor therapy. It could be used not only as a therapeutic directly acting on tumor cells but, in parallel, as a genetic vaccine "enhancer".


Subject(s)
Butyrates/pharmacology , Melanoma, Experimental/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Division/drug effects , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Retroviridae/genetics , Transduction, Genetic , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
7.
Neoplasma ; 48(5): 358-61, 2001.
Article in English | MEDLINE | ID: mdl-11845979

ABSTRACT

Systemic administration of cytokines has not found broad application in cancer immunotherapy due to its toxicity and lack of effectiveness in a broad spectrum of tumors. Among the most promising cytokines used often in pre-clinical and clinical trials are interferon alpha and interleukin 12. We have shown in our study that combining IL-12 with IFN-alpha in a dose which alone does not show antitumor activity results in potentiated antitumor effects without inducing toxicity.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Interferon-alpha/therapeutic use , Interleukin-12/therapeutic use , Melanoma, Experimental/drug therapy , Animals , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation , Time Factors , Tumor Cells, Cultured
8.
Int J Biochem Cell Biol ; 32(9): 957-65, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11084375

ABSTRACT

Lovastatin and simvastatin are HMG-CoA reductase inhibitors widely used as antihyperlipidemic drugs, which also display antiproliferative properties. In the present paper, we provide evidence that both lovastatin and simvastatin are modulators of the purified bovine pituitary 20 S proteasome, since they mildly stimulate the chymotrypsin-like activity and inhibit the peptidylglutamylpeptide hydrolyzing activity without interfering with the trypsin-like activity. However, those effects are only observed when the closed ring forms of the drugs are used, while the opened ring form of lovastatin acts as a mild inhibitor of the chymotrypsin like activity. The closed ring form of lovastatin is much more potent as a cytotoxic agent on the Colon-26 (C-26) colon carcinoma cell line than the opened ring form, which is only mildly cytostatic. Moreover, neither the cytotoxic effects nor the effects on 20 S proteasome activities are prevented by mevalonate, which by itself inhibits the trypsin-like activity of the proteasome. Neither the opened ring nor the closed ring form of lovastatin induces an accumulation of ubiquitin-protein conjugates, which is observed after treatment with lactacystin, a selective proteasome inhibitor. In contrast with the opened ring form of lovastatin, the closed ring form induces the disappearance of detectable p27(kip1) from C-26 cells. Altogether, our results indicate that the closed ring form of lovastatin induces cytotoxic effects independent of its HMG-CoA inhibiting activity, however, those effects are mediated by a complex modulation of proteasome activity rather than by inhibition of the 20 S proteasome.


Subject(s)
Cysteine Endopeptidases/metabolism , Lovastatin/pharmacology , Multienzyme Complexes/metabolism , Simvastatin/pharmacology , Animals , Antineoplastic Agents/toxicity , Blotting, Western , Growth Inhibitors/toxicity , Lovastatin/toxicity , Mice , Proteasome Endopeptidase Complex , Simvastatin/toxicity , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
9.
Clin Cancer Res ; 6(5): 2044-52, 2000 May.
Article in English | MEDLINE | ID: mdl-10815931

ABSTRACT

Lovastatin, a drug commonly used in the clinic to treat hypercholesterolemia, has previously been reported to exert antitumor effects in rodent tumor models and to strengthen the antitumor effects of immune response modifiers (tumor necrosis factor alpha and IFN-gamma) or chemotherapeutic drugs (cisplatin). In the present report, we show in three murine tumor cell lines (Colon-26 cells, v-Ha-ras-transformed NIH-3T3 sarcoma cells, and Lewis lung carcinoma cells) that lovastatin can also effectively potentiate the cytostatic/cytotoxic activity of doxorubicin. In three tumor models (Co-ion-26 cells, v-Ha-ras-transformed NIH-3T3 sarcoma cells, and Lewis lung carcinoma cells) in vivo, we have demonstrated significantly increased sensitivity to the combined treatment with both lovastatin (15 mg/kg for 10 days) and doxorubicin (3 x 2.5 mg/kg; cumulative dose, 7.5 mg/kg) as compared with either agent acting alone. Lovastatin treatment also resulted in a significant reduction of troponin T release by cardiomyocytes in doxorubicin-treated mice. This observation is particularly interesting because lovastatin is known to reduce doxorubicin-induced cardiac injury.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Heart Diseases/prevention & control , Neoplasms, Experimental/drug therapy , 3T3 Cells , Animals , Cell Division/drug effects , Cell Line, Transformed , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Female , Heart Diseases/blood , Heart Diseases/chemically induced , Lovastatin/administration & dosage , Lovastatin/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation , Neoplasms, Experimental/blood , Neoplasms, Experimental/pathology , Time Factors , Troponin T/blood , Troponin T/drug effects , Tumor Cells, Cultured
10.
Br J Cancer ; 82(8): 1485-91, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10780531

ABSTRACT

Photofrin-based photodynamic therapy (PDT) has recently been approved for palliative and curative purposes in cancer patients. It has been demonstrated that neutrophils are indispensable for its anti-tumour effectiveness. We decided to evaluate the extent of the anti-tumour effectiveness of PDT combined with administration of granulocyte colony-stimulating factor (G-CSF) as well as the influence of Photofrin and G-CSF on the myelopoiesis and functional activity of neutrophils in mice. An intensive treatment with G-CSF significantly potentiated anti-tumour effectiveness of Photofrin-based PDT resulting in a reduction of tumour growth and prolongation of the survival time of mice bearing two different tumours: colon-26 and Lewis lung carcinoma. Moreover, 33% of C-26-bearing mice were completely cured of their tumours after combined therapy and developed a specific and long-lasting immunity. The tumours treated with both agents contained more infiltrating neutrophils and apoptotic cells then tumours treated with either G-CSF or PDT only. Importantly, simultaneous administration of Photofrin and G-CSF stimulated bone marrow and spleen myelopoiesis that resulted in an increased number of neutrophils demonstrating functional characteristics of activation. Potentiated anti-tumour effects of Photofrin-based PDT combined with G-CSF observed in two murine tumour models suggest that clinical trials using this tumour therapy protocol would be worth pursuing.


Subject(s)
Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Dihematoporphyrin Ether/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Photochemotherapy , Adenocarcinoma/pathology , Animals , Bone Marrow Cells/pathology , Colonic Neoplasms/pathology , Colony-Forming Units Assay , Combined Modality Therapy , Filgrastim , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Recombinant Proteins , Spleen/pathology
11.
Anticancer Res ; 20(1A): 391-4, 2000.
Article in English | MEDLINE | ID: mdl-10769685

ABSTRACT

The application of antiangiogenic agents in cancer therapy has been studied extensively. Combination of agents with antiangiogenic properties could possibly enhance antitumor effects. Interleukin 12 is a cytokine with potent antitumor activity mediated also via antiangiogenic mechanisms. These effects are attributed to IFN-gamma production stimulated by IL-12. Since IFN-gamma has been reported to augment antitumor effects when combined with one of the metalloproteinase inhibitors--batimastat (BB-94), we have examined a combined treatment with IL-12 and BB-94 in a murine melanoma model. The administration of both agents showed potentiated antitumor activity. Furthermore, we have shown in a tumor-induced angiogenesis model that the combined application of IL-12 and batimastat inhibits the formation of new blood vessels to a greater extent than either agent alone. Our observations show that antiangiogenic effects are at least partly responsible for the enhanced antitumor effects of the combined treatment with IL-12 and BB-94.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Interleukin-12/therapeutic use , Melanoma, Experimental/drug therapy , Metalloendopeptidases/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Phenylalanine/analogs & derivatives , Protease Inhibitors/therapeutic use , Thiophenes/therapeutic use , Adjuvants, Immunologic/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Drug Synergism , Interleukin-12/pharmacology , Melanoma, Experimental/blood supply , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation , Phenylalanine/pharmacology , Phenylalanine/therapeutic use , Protease Inhibitors/pharmacology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thiophenes/pharmacology , Tumor Cells, Cultured/transplantation
12.
Life Sci ; 66(13): 1223-30, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10737417

ABSTRACT

Nonsteroidal anti-inflammatory drugs have been shown to reduce the incidence and mortality from colorectal cancer. It has recently been demonstrated that these drugs are capable of suppressing the production of pro-angiogenic factors from tumor cells. The mechanisms of antitumor action of interleukin 12 include the enforced secretion of anti-angiogenic factors and stimulation of antitumor immunity. Therefore, we hypothesized that the combination of a model nonsteroidal anti-inflammatory drug--indomethacin and interleukin 12--would result in enhanced angiogenesis-dependent antitumor effects against a colon-26 carcinoma cells transplanted into syngeneic mice. As expected the combined administration of both agents simultaneously resulted in a strengthened antitumor activity that was manifested as a retardation of tumor growth and prolongation of mouse survival. Importantly some mice were completely cured after the combined treatment. As administration of interleukin 12 and indomethacin resulted in enhanced inhibition of angiogenesis it seems possible that prevention of new blood vessel formation is one of the mechanisms responsible for the observed antitumor effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Indomethacin/therapeutic use , Interleukin-12/therapeutic use , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic , Animals , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/therapeutic use , Drug Synergism , Drug Therapy, Combination , Female , Indomethacin/administration & dosage , Interleukin-12/administration & dosage , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Tumor Cells, Cultured
13.
Oncol Rep ; 7(1): 177-81, 2000.
Article in English | MEDLINE | ID: mdl-10601614

ABSTRACT

In previous studies we have shown that combined chemo-immunotherapy of L1210 leukemia with IL-12 and doxorubicin results in striking anti-tumor effects producing 100% of long-term survivors. In this study we investigated the efficacy of a combination of IL-12 and mitoxantrone in murine L1210 leukemia. Mice inoculated with 1x105 leukemia cells were treated with a single dose of mitoxantrone and seven daily doses of IL-12, and were daily observed for survival. Treatment with IL-12 or mitoxantrone given alone produced moderate anti-leukemic effects. However, combination of both drugs resulted in a significant prolongation of mouse survival time. Importantly, there were almost 50% of long-term (>60 days) survivors among the mice treated with both agents. This therapeutic effect was completely abrogated by sub-lethal, whole-body X-irradiation, and significantly reduced after macrophage depletion.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interleukin-12/administration & dosage , Leukemia L1210/drug therapy , Mitoxantrone/administration & dosage , Animals , Drug Synergism , Macrophages/physiology , Mice
14.
Eur Cytokine Netw ; 10(3): 345-56, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10477391

ABSTRACT

Interleukin-12 (IL-12) is a potent immunoregulatory cytokine that exhibits antitumor activity in many experimental tumor models. In the present study, we investigated the ability of IL-15, a cytokine sharing many functions of IL-2, to modulate antitumor effectiveness of IL-12 against B16F10 melanoma in mice. In a model of locally growing tumor, intratumoral (i.t.) administration of IL-12, in three cycles of five consecutive daily injections (0.1 mug) followed by 2 days of rest, led to considerable delay of tumor development but no curative response was achieved. When combined with IL-12, subtherapeutic doses of IL-15 (0.4 mug) pontentiated the antitumor effects of IL-12 and induced complete tumor regressions in 50% of mice. Similar results were obtained in a model in which tumor-bearing mice were intravenously co-injected with melanoma cells to induce metastases. Combined administration of IL-12 and IL-15 yielded greater antitumor activity than injections of either cytokine alone and resulted in prolonged survival of mice bearing locally growing tumor and metastases. Studies of immunological parameters in mice treated with both IL-12 and IL-15 have shown enhanced NK activity (against YAC-1 cells) in the spleen and stimulation of both NK activity and specific anti-B16F10 cytotoxic effector cells in tumor-draining lymph nodes (LN). The strong antitumor effect of the IL-12 + IL-15 combination correlated with a high serum level of IFN-gamma in the treated mice. Moreover, increased expression of IL-15Ralpha was demonstrated in LN lymphocytes isolated from mice injected with IL-12. This result together with findings of other authors showing enhanced expression of IL-12 receptor by IL-15 [1] suggests that the augmentation of the antitumor effect during the course of IL-12/IL-15-based therapy could result from reciprocal upregulation of receptors by both cytokines and synergistic effects on IFN-gamma induction.


Subject(s)
Interleukin-12/pharmacology , Interleukin-15/pharmacology , Melanoma, Experimental/immunology , Animals , Disease Models, Animal , Drug Synergism , Interferon-gamma/blood , Interleukin-12/administration & dosage , Interleukin-15/administration & dosage , Macrophages, Peritoneal/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Metastasis/immunology , Receptors, Interleukin-15 , Receptors, Interleukin-2/metabolism
16.
Int J Cancer ; 81(4): 560-7, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10225445

ABSTRACT

Lovastatin, a drug commonly used in the treatment of hypercholesterolemia, has previously been reported to exert potentiated antitumor activity when combined with either tumor necrosis factor-alpha (TNF-alpha), cisplatin or doxorubicin in a melanoma model in mice. Since lovastatin interferes with the function of ras oncogene-encoded (Ras) proteins, we have investigated the antitumor activity of lovastatin and TNF-alpha using a Ha-ras-transformed murine tumor model. In in vitro studies, lovastatin inhibited the growth of cells transformed with Ha-ras oncogene (Ras-3T3 and HBL100-ras cells) more effectively than control NIH-3T3 and HBL100-neo cells. In in vivo experiments, the Ras-3T3 tumor demonstrated significantly increased sensitivity to combined treatment with both lovastatin (50 mg/kg) and TNF-alpha (1 microg/day) compared with either agent alone. Combined treatment with both agents also resulted in greater inhibition of blood-vessel formation. Ras-3T3 tumor cells produced increased amounts of vascular endothelial growth factor (VEGF) and lovastatin effectively suppressed VEGF production by these cells. Our results suggest that lovastatin increases antitumor activity of TNF-alpha against tumor cells transformed with v-Ha-ras oncogene via inhibition of tumor-induced blood-vessel formation.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic , Genes, ras , Lovastatin/toxicity , Lovastatin/therapeutic use , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Tumor Necrosis Factor-alpha/toxicity , Tumor Necrosis Factor-alpha/therapeutic use , 3T3 Cells , Animals , Antineoplastic Agents/toxicity , Cell Division/drug effects , Cell Survival/drug effects , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Lymphokines/biosynthesis , Lymphokines/genetics , Macrophages/cytology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/pathology , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Cancer Lett ; 147(1-2): 67-75, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10660091

ABSTRACT

In our recent study we described a significant antileukemic efficacy of a combination therapy with interleukin-12 (IL-12) and doxorubicin (DOX) in the L1210 leukemia model. This therapeutic effect was abrogated by elimination of activated macrophages. Activated macrophages produce a variety of factors that can contribute to the elimination of tumor cells in vivo, including proteases, TNF, reactive oxygen intermediates, and nitric oxide (NO). Based on the results of previous reports, the contribution of NO in potentiated antileukemic effects of IL-12 + DOX combination seemed to be highly possible. Both DOX and IL-12 given alone increased the production of NO by peritoneal macrophages, however, macrophages derived from the mice treated with the combination of those agents produced significantly less NO than macrophages from IL-12-alone-treated mice. Production of NO by spleen macrophages after IL-12 + DOX treatment was higher than it was in controls, IL-12-alone or DOX-alone-treated groups. In serum, concentrations of NOx- in IL-12- or IL-12 + DOX-treated mice were significantly higher in comparison with controls, however not significantly different from each other. Addition of L-NAME treatment to the IL-12 + DOX therapy in leukemia-bearing mice did not significantly change the antileukemic efficacy of this therapy. Thus, our results indicate that the augmented antileukemic effects of IL-12 + DOX combination therapy in L1210 model are NO-independent. Therefore, further studies on the possible mechanisms of potentiated antileukemic activity of combination of IL-12 and DOX would be worth pursuing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia L1210/drug therapy , Leukemia L1210/metabolism , Nitric Oxide/biosynthesis , Adjuvants, Immunologic/administration & dosage , Animals , Antibiotics, Antineoplastic/administration & dosage , Cells, Cultured , Crosses, Genetic , Doxorubicin/administration & dosage , Drug Synergism , Enzyme Inhibitors/pharmacology , Female , Interleukin-12/administration & dosage , Leukemia L1210/immunology , Leukemia L1210/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , NG-Nitroarginine Methyl Ester/pharmacology , Neoplasm Transplantation , Nitrates/blood , Nitric Oxide Synthase/antagonists & inhibitors , Nitrites/blood , Spleen/drug effects , Spleen/metabolism , Survival Rate
19.
Int J Cancer ; 77(5): 720-7, 1998 Aug 31.
Article in English | MEDLINE | ID: mdl-9688305

ABSTRACT

It has been well established that chemo-immunotherapy using cytotoxic drugs and appropriate cytokines offers a new approach to increasing the therapeutic index in the treatment of neoplastic diseases. This study investigates the efficacy of combinations of interleukin-12 with cyclophosphamide, paclitaxel, cisplatin or doxorubicin in the murine L1210 leukemia model. Mice inoculated i.p. with 1 x 10(3) or 1 x 10(5) leukemia cells were treated with interleukin-12 and/or chemotherapeutics, and were observed daily for survival. Immunosuppression with X-irradiation or macrophage depletion with injections of silica were used to examine the dependence of the therapeutic effects on the efficiency of the immune system. Treatment with interleukin-12 or one of the studied chemotherapeutics given alone resulted in moderate antileukemic effects. Combination of interleukin-12 with cyclophosphamide or paclitaxel produced no augmentation of anti-leukemic effects in comparison with these agents given alone. Combination of interleukin-12 with cisplatin resulted in prolongation of the survival time; however, in the experiment with mice inoculated with 1 x 10(5) leukemia cells, no long-term survivors (>60 days) were observed; on the contrary, combination of interleukin-12 with doxorubicin resulted in 100% long-term survivors. This effect was completely abrogated either by X-irradiation of mice or by macrophage depletion. We also found that doxorubicin augments IL-12-stimulated production of interferon-gamma in vivo. Our observations demonstrating potentiation of the antileukemic effects of the IL-12 and doxorubicin combination suggest that the combined use of these 2 agents could be beneficial in leukemia therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Interleukin-12/therapeutic use , Leukemia L1210/therapy , Animals , Cisplatin/therapeutic use , Combined Modality Therapy , Cyclophosphamide/therapeutic use , Female , Immunotherapy , Interferon-gamma/immunology , Leukemia L1210/blood , Leukemia L1210/immunology , Macrophage Activation , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Paclitaxel/therapeutic use , Recombinant Proteins/therapeutic use , Survival Analysis , Time Factors , Whole-Body Irradiation
20.
Neoplasma ; 45(1): 35-9, 1998.
Article in English | MEDLINE | ID: mdl-9605000

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) was found to exert antitumor activity against murine MmB16 melanoma when administered intratumorally. However, subcutaneous administration of this cytokine at a site distant from the growing tumor did not show any antitumor effects. G-CSF did not influence the proliferative activity of MmB16 in vitro. Intraperitoneal administration of G-CSF resulted in decreased secretion of nitric oxide (NO) by peritoneal macrophages and their decreased tumoricidal activity against MmB16.


Subject(s)
Antineoplastic Agents/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Melanoma, Experimental/drug therapy , Animals , Cell Division/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nitric Oxide/biosynthesis , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...