Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Peptides ; 175: 171111, 2024 May.
Article in English | MEDLINE | ID: mdl-38036098

ABSTRACT

Endogenous peptide inhibitor for CXCR4 (EPI-X4) is a CXCR4 antagonist with potential for cancer therapy. It is a processed fragment of serum albumin from the hemofiltrate of dialysis patients. This study reports the efficacy of fifteen EPI-X4 derivatives in pancreatic cancer and lymphoma models. In vitro, the peptides were investigated for antiproliferation (cytotoxicity) by MTT assay. The mRNA expression for CXCR4 and CXCL12 was determined by RT-PCR, chip array and RNA sequencing. Chip array analysis yielded 634 genes associated with CXCR4/CXCL12 signaling. About 21% of these genes correlated with metastasis in the context of cell motility, proliferation, and survival. Expression levels of these genes were altered in pancreatic cancer (36%), lymphoma models (53%) and in patients' data (58%). EPI-X4 derivatives failed to inhibit cell proliferation due to low expression of CXCR4 in vitro, but inhibited tumor growth in the bioassays with significant efficacy. In the pancreatic cancer model, EPI-X4a, f and k inhibited mean tumor growth by > 50% and even caused complete remissions. In the lymphoma model, EPI-X4b, n and p inhibited mean tumor growth by > 70% and caused stable disease. Given the non-toxic and non-immunogenic properties of EPI-X4, these findings underscore its status as a promising therapy of pancreatic cancer and lymphoma and warrant further studies. SIMPLE SUMMARY: This study examined the value of chemokine receptor CXCR4 as an antineoplastic target for the endogenous peptide inhibitor of CXCR4 (EPI-X4), a 12-meric peptide derived from serum albumin. EPI-X4 inhibits CXCR4 interaction with its natural ligand, CXCL12 (SDF1). Therefore, malignancies (including pancreatic cancer and lymphoma) that depend on the CXCR4/CXCL12 pathway for progression can be targeted with EPI-X4. Of 634 genes that were linked to the CXCR4/CXCL12 pathway, 21% were associated with metastasis. In cultured human Suit2-007 pancreatic cancer cells, CXCR4 showed low to undetectable expression, which was why EPI-X4 did not inhibit pancreatic cancer cell proliferation. These findings were different in vivo, where CXCR4 was highly expressed and EPI-X4 inhibited tumor growth in rodents harboring pancreatic cancer or lymphoma. In the pancreatic cancer model, EPI-X4 derivatives a, f and k caused complete remissions, while in lymphomas EPI-X4 derivatives b, n and p caused stable disease.


Subject(s)
Lymphoma , Pancreatic Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Lymphoma/drug therapy , Lymphoma/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Peptides/chemistry , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Signal Transduction
2.
Cancer Lett ; 474: 82-93, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31954769

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease, and novel therapeutic strategies are urgently needed. Recently, expression of the C-C chemokine receptor 5 (CCR5) and its ligands has been found to play an important role in cancer progression and metastasis. In this study, we blocked the CCR5 receptor by the FDA approved antagonist maraviroc (MVC) in Suit2-007 and MIA-PaCa-2 human PDAC cells. The treatment significantly inhibited their proliferation and induced apoptosis of exposed cells as evidenced by caspases activation and increased Bax levels. Moreover, MVC inhibited the cell cycle by down regulating the proteins of the complexes of cyclin dependent kinase (CDK) 4/6 - Cyclin D and CDK2 - Cyclin E, as well as by increasing the protein levels of CDK inhibitors p18, p21 and p27. In line with this, MVC caused significant retardation of Suit2-007 cells growing in a PDAC liver metastasis xenograft model (p < 0.05). These results suggest that maraviroc could be a promising treatment strategy for PDAC patients with liver metastases.


Subject(s)
Apoptosis , Carcinoma, Pancreatic Ductal/drug therapy , Cell Cycle , Liver Neoplasms/drug therapy , Maraviroc/pharmacology , Pancreatic Neoplasms/drug therapy , Receptors, CCR5/chemistry , Animals , CCR5 Receptor Antagonists/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Movement , Cell Proliferation , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Rats , Rats, Nude , Receptors, CCR5/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Oncol Lett ; 15(2): 1441-1448, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29434835

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has one of the poorest prognoses of all malignancy types. To improve the survival of patients with PDAC, the development of novel anticancer agents is warranted. Riproximin (Rpx) is a newly identified plant lectin, which was isolated from Ximenia americana. The ribosome inactivating protein of type II exhibits potent anticancer activity as recently demonstrated. The rat PDAC cell line ASML was used for in vitro and in vivo studies. The antiproliferative effect of Rpx was assessed using an MTT assay. The modulation of proteins involved in apoptosis was evaluated using western blotting. Tumor-bearing nude rats were treated with Rpx, gemcitabine (GEM) or dinaline (DIN) as single agents, or a combination of Rpx with GEM, or DIN. Rpx was administered intraperitoneally at doses of 1.7-5.4 µg/kg, three times/week, GEM was administered intravenously (50 mg/kg/week) and DIN perorally (10 mg/kg, 5 times/week). Rpx inhibited ASML cell proliferation at IC50-values of 0.8-172 pM, caused apoptosis and reduced tumor growth significantly by 90% (P<0.05). The survival rate of rats was significantly increased (21.8 days for Rpx treated vs. 17.6 days for control rats; P=0.05). Higher doses of Rpx caused no further reduction in tumor size when compared with the low dose of Rpx or a combination of Rpx with GEM, or DIN. The standard drug GEM alone was less effective compared with Rpx. In addition, DIN was ineffective, and in combination, reduced the activity of Rpx. These results suggest that Rpx has an evident potential for use in pancreatic cancer treatment. Further experiments are required in order to elucidate its affinity for certain cancer cells and to optimize the combination therapy with other antineoplastic agents.

4.
Afr Health Sci ; 18(4): 1303-1310, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30766596

ABSTRACT

BACKGROUND: Metastasis is the leading cause of cancer deaths. Migration of tumor cells is an important stage in metastasis. Therefore, recent studies have focused on clarifying migration and migration-dependent cell functions such as angiogenesis, wound healing, and invasion. OBJECTIVES: In the present study, we aimed to investigate the effect of acetazolamide, which is a classical carbonic anhydrase inhibitor, on the cell viability, migration, and colony forming capacity of human LS174T colorectal cancer cells. METHODS: Three different cell culture techniques (MTT test, wound healing and clonogenic assay) were performed in this in vitro study on colorectal cancer cells. RESULTS: Acetazolamide reduced the cell viability, migration and colony formation ability of cells depending on dose. There was no significant difference between the cells treated with acetazolamide with 1 µM dose and the control. However, it can be concluded that acetazolamide exerts its effect on human colorectal cancer cells at 10-1000 µM concentrations. CONCLUSION: Acetazolamide was observed to significantly inhibit the cell viability, colony forming capacity, and migration ability in the culture medium of LS174T cells. This inhibitor effect of acetazolamide was observed to be dependent on the concentration in medium.


Subject(s)
Acetazolamide/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Acetazolamide/administration & dosage , Carbonic Anhydrase Inhibitors/administration & dosage , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans
5.
Oncol Rep ; 28(6): 2177-87, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23007550

ABSTRACT

Owing to aggressiveness and chemoresistance, pancreatic ductal adenocarcinoma (PDAC) is characterised by a poor prognosis. To address this disease-spe-cific dilemma we aimed to establish animal models, which can be used for identifying new specific tumor markers, as well as serving as tools for potential therapeutic approaches. From a panel of sixteen pancreatic cancer cell lines, two human (Suit2-007 and Suit2-013) and a rat (ASML) cell line were selected for their properties to grow in the liver of male RNU rats and mimic liver metastasis of PDAC. For better monitoring of metastatic tumor growth in vivo, all three pancreatic cancer cell lines were stably transfected with eGFP and luciferase marker genes. In addition, the mRNA expression profile of 13 human PDAC cell lines was analyzed by BeadChip array analysis. Only 33 genes and 5 signaling pathways were identified as significantly associated with the ability of the cell lines to grow initially and/or consistently in rat liver. Only a minority of these genes (osteopontin, matrix metalloproteinase-1 and insulin-like growth factor 1) has been intensively studied and shown to be closely related to cancer progression. The function of the remaining 30 genes ranges from moderate to poorly investigated, and their function in cancer progression is still unclear. The ensuing three pancreatic cancer liver metastasis models vary in their aggressiveness and macroscopic growth. They will be used for preclinical evaluation of new therapeutic approaches aiming at the genes identified.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/secondary , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Humans , Liver/pathology , Luciferases/genetics , Male , Neoplasm Metastasis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Signal Transduction
6.
J Cell Mol Med ; 16(2): 260-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21388515

ABSTRACT

Terminal progression of colorectal cancer (CRC) culminates in liver metastasis. To identify genes that are involved in the metastatic phenotype, cDNA microarrays were used to analyse mRNA expression profiles of colorectal carcinoma (CC)531 rat colon adenocarcinoma cells for changes related to their homing into the liver. Briefly, CC531 cells were intraportally implanted into the liver of Wag-Rij rats and re-isolated after 3, 6, 9, 14 and 21 days. Compared to control CC531 cells, claudin1 and claudin4 were among the ≥8-fold initially down-regulated genes. The co-culture of tumour cells with isolated rat hepatocytes and Kupffer cells did not induce down-regulation of either claudin1 or 4. When the environment effective on circulating tumour cells was simulated by cell culture conditions favouring their adhesion, only claudin4 showed augmented expression. Knockdown of claudin1 and claudin4 mediated by small interfering RNA caused significantly increased migration and decreased clonogenic growth of tumour cells (P < 0.05), but had no effect on their proliferation. These experimental results were paralleled by increased claudin1 and claudin4 expression in human CRC samples in Union for International Cancer Control (UICC) stages I-III, as evaluated by real-time PCR. Increased claudin4 levels were correlated with significantly reduced overall survival (log-rank test, P= 0.018). Further, significantly (P < 0.05) reduced expression of claudin1 and claudin4 was observed in stage IV and liver metastasis by immunohistochemistry. In conclusion, sequential biphasic changes in claudin1 and claudin4 expression occur during the homing of rat CC531 CRC cells to the liver. This modulation is reflected by significant changes in claudin expression in human primary and metastatic CRC.


Subject(s)
Claudins/metabolism , Colorectal Neoplasms/genetics , Liver Neoplasms/secondary , Membrane Proteins/metabolism , Adenocarcinoma/genetics , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Claudin-1 , Claudin-4 , Claudins/genetics , Coculture Techniques , Colorectal Neoplasms/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , Hepatocytes/metabolism , Humans , Kupffer Cells/metabolism , Liver Neoplasms/genetics , Male , Membrane Proteins/genetics , Neoplasm Metastasis/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Rats , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Int J Oncol ; 37(2): 249-56, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20596651

ABSTRACT

Colorectal cancer accounts for 11% of all cancers and is the second most frequent cause of cancer-related death, with the majority of deaths attributable to hepatic metastasis. The main aim of the study was to investigate changes which occur in CC531 rat colon adenocarcinoma cells and are instrumental to the metastatic phenotype after homing to the liver. RT-PCR and Western blotting were used to detect the expression of certain proteins, transcription factors and enzymes, which are intimately linked to colorectal metastasis. These included osteopontin (OPN), bone sialoprotein ll (BSP ll), Runx2, Hoxc8, matrix metalloprotease-7 (MMP-7) and matrix-metalloprotease-9 (MMP-9). Subsequently, in order to detect the role of the hepatocytes in these changes seen in tumor cells, two models of co-culturing hepatocytes with CC531 cells were established. OPN, Runx2 and MMP-7 were found to be highly expressed in CC531 metastases explanted from the liver, but showed subsequent down-regulation and/or disappearance in cell culture. The inverse regulation of Hoxc8, OPN and Runx2 suggests that these genes may be regulated in a feed-back loop manner. MMP-9 mRNA and active MMP-7 protein were expressed in tumor cells themselves. The presence of hepatocytes was insufficient to induce induction of OPN and Runx2 in tumor cells in vitro, so was the addition of OPN or TGF-beta1. Whereas TGF-beta1 induced over-expression of OPN and Runx2 in hepatocytes, it did not exert the same effect on hepatocytes co-cultured with CC531 cells, indicating that this response was abrogated by CC531 cells.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Osteopontin/genetics , Sialoglycoproteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Neoplastic , Hepatocytes/metabolism , Hepatocytes/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Integrin-Binding Sialoprotein , Male , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Neoplasm Metastasis , Osteopontin/metabolism , Rats , Sialoglycoproteins/metabolism
8.
Cancer Biol Ther ; 10(1): 54-64, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20495387

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death in western countries and among the malignancies with the worst prognosis. Osteonectin and osteopontin, two proteins of the extracellular matrix, have been found to be upregulated in PDAC. In the present study the expression of osteopontin mRNA as determined in a panel of 14 human pancreatic cancer cell lines was significantly related to the growth of these cell lines in the liver of nude rats (p = 0.001); whereas osteonectin showed a trend of being negatively related to pancreatic cancer cell growth in vivo (p = 0.10). In an in vitro co-culture model of human Suit2-007 and rat AsML PDAC cells with rat hepatocytes, a clearly increased expression of OPN mRNA was found in the tumor cells. In addition, both downregulation of osteopontin with specific antisense oligonucleotides and treatment with exogenous rh-osteonectin were associated with reduced cell proliferation. In accordance with the latter finding downregulation of osteonectin was coupled with increased proliferation. This evidence supports a protumorigenic role of osteopontin and points to an antitumorigenic role of osteonectin in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/secondary , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/secondary , Osteonectin/genetics , Osteopontin/genetics , Pancreatic Neoplasms/pathology , Animals , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Coculture Techniques , Disease Progression , Down-Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Male , Oligonucleotides, Antisense/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA, Messenger/genetics , Rats , Rats, Nude , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Clin Exp Metastasis ; 25(3): 273-82, 2008.
Article in English | MEDLINE | ID: mdl-18259882

ABSTRACT

Systemic chemotherapy has limited success in treating liver metastasis of colorectal cancer. Alternative approaches such as hepatic arterial infusion or trans arterial chemoembolisation aim to deliver the chemotherapy locally to address the predominant liver disease. Chemoembolisation with drug eluting beads (DEB) designed to deliver drug at the target over a protracted period of time is a new strategy to reduce the tumor burden of liver metastases. To test this hypothesis, DEB possessing anionic groups capable of ionically complexing with cationic drugs were synthesised by a suspension polymerisation method and were fractionated to produce an average size of 75 microm. The DEB were loaded with the desired concentration of either doxorubicin hydrochloride or irinotecan hydrochloride prior to administration by immersion in the drug solution, yielding essentially 100% loading efficiency. To determine their effect in vivo, a transplantable orthotopic and isogenic rat liver metastasis model was used which is based on intraportal injection of 4 x 10(6) beta-galactosidase transfected CC531 rat colorectal cancer cells into male WAG/Rij rats. By MTT assay, the cells were shown to be sensitive to both drugs in vitro with the IC(50) being by two orders of magnitude lower for doxorubicin (110 nM after 72 h) compared to irinotecan (25 microM after 72 h). For the in vivo phase, a differential expression of the ERK MAP kinase between tumor cells cultured in vitro and those inoculated in vivo was noted using Western blotting techniques. This was considered to be indicative of passage-induced cell senescence that reduced the sensitivity of the tumor cells to DEB chemoembolisation. This notwithstanding, administration of DEB loaded with irinotecan or doxorubicin by single injection into the hepatic artery showed significant anticancer activity, as measured by a reduction in the tumor burden of the liver and a corresponding reduction in liver weight. Comparing the two agents, irinotecan appears more advantageous because of its significant activity and excellent tolerability following administration at two dosages of either 20 or 30 mg/kg. Doxorubicin showed a narrower window of activity, being effective at 4 mg/kg but ineffective at the lower dose of 2 mg/kg. We conclude that chemoembolisation with DEB with either agent may have potential for treating patients with colorectal liver metastasis, although irinotecan DEB appeared to have a more favourable safety profile.


Subject(s)
Adenocarcinoma/therapy , Camptothecin/analogs & derivatives , Chemoembolization, Therapeutic , Colorectal Neoplasms/therapy , Doxorubicin/administration & dosage , Liver Neoplasms/therapy , Adenocarcinoma/secondary , Animals , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Blotting, Western , Camptothecin/administration & dosage , Cell Proliferation , Colorectal Neoplasms/pathology , Drug Delivery Systems , Irinotecan , Liver Neoplasms/secondary , Male , Microspheres , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Prodrugs , Rats , beta-Galactosidase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
FASEB J ; 20(8): 1194-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16641197

ABSTRACT

The aim of this study was to identify and characterize the active component(s) of Ximenia americana plant material used to treat cancer in African traditional medicine. By a combination of preextraction, extraction, ion exchange and affinity chromatography, a mixture of two cytotoxic proteins was isolated. Using degenerated primers designed on the de novo sequence of two tryptic peptides from one of these proteins, a DNA fragment was amplified and the sequence obtained was used to determine the complete cDNA sequence by the RACE method. Sequence analysis and molecular modeling showed that the new protein, riproximin, belongs to the family of type II ribosome inactivating proteins. These results are in good agreement with the ability of riproximin to inhibit protein synthesis in a cell-free system, as well as with the cytotoxicity of riproximin, as demonstrated by its IC50 value of 0.5 pM in MCF7, 1.1 pM in HELA and 0.6 pM in CC531-lacZ cells. To assess the antineoplastic efficacy of the purified riproximin in vivo, the CC531-lacZ colorectal cancer rat metastasis model was used. Significant anticancer activity was found after administration of total dosages of 100 (perorally) and 10 (intraperitoneally) pmol riproximin/kg. These results suggest that riproximin has distinct potential for cancer treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Medicine, African Traditional , Olacaceae/chemistry , Plant Proteins/pharmacology , Protein Biosynthesis/drug effects , Amino Acid Sequence , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Line, Tumor , Galactose/chemistry , Glycosylation , Humans , Molecular Sequence Data , Plant Proteins/chemistry , Plant Proteins/isolation & purification , Rats , Sequence Analysis, Protein , Structural Homology, Protein
11.
Toxicol Appl Pharmacol ; 211(3): 177-87, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16005923

ABSTRACT

The antineoplastic activity of a plant powder used in African traditional medicine for treating cancer was investigated by analyzing the activity of various extracts in vitro. The most active, aqueous extract was subsequently subjected to a detailed investigation in a panel of 17 tumor cell lines, showing an average IC50 of 49 mg raw powder/ml medium. The sensitivity of the cell lines varied by two orders of magnitude, from 1.7 mg/ml in MCF7 breast cancer cells to 170 mg/ml in AR230 chronic-myeloid leukemia cells. Immortalized, non-tumorigenic cell lines showed a marginal sensitivity. In addition, kinetic and recovery experiments performed in MCF7 and U87-MG cells and a comparison with the antineoplastic activity of miltefosine, gemcitabine, and cisplatinum in MCF7, U87-MG, HEp2, and SAOS2 cells revealed no obvious similarity between the sensitivity profiles of the extract and the three standard agents, suggesting a different mechanism of cytotoxicity. The in vivo antitumor activity was determined in the CC531 colorectal cancer rat model. Significant anticancer activity was found following administration of equitoxic doses of 100 (perorally) and 5 (intraperitoneally) mg raw powder/kg, indicating a 95% reduced activity following intestinal absorption. By sequencing the mitochondrial gene for the large subunit of the ribulose bis-phosphate carboxylase (rbcL) in DNA from the plant material, the source plant was identified as Ximenia americana. A physicochemical characterization showed that the active antineoplastic component(s) of the plant material are proteins with galactose affinity. Moreover, by mass spectrometry, one of these proteins was shown to contain a stretch of 11 amino acids identical to a tryptic peptide from the ribosome-inactivating protein ricin.


Subject(s)
Antineoplastic Agents, Phytogenic , Cell Proliferation/drug effects , Liver Neoplasms, Experimental/drug therapy , Medicine, African Traditional , Olacaceae/chemistry , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Neoplasm Transplantation , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Inbred Strains , Water , Xenograft Model Antitumor Assays
12.
J Cancer Res Clin Oncol ; 131(5): 289-99, 2005 May.
Article in English | MEDLINE | ID: mdl-15657768

ABSTRACT

PURPOSE: The aim of this study was to evaluate the combination effect of pemetrexed disodium (MTA; Alimta; LY 231514) and gemcitabine (GEM) administered by hepatic artery and portal vein chemoembolization (HACE and PVCE) in a colorectal cancer rat liver metastasis model. MATERIALS AND METHODS: Proliferation studies on CC531-lac-Z rat colon cancer cells were performed using the MTT assay to obtain the optimal combination schedule of the two antineoplastic agents. To generate diffuse liver metastasis, 4 x 10(6) tumor cells were implanted into the portal vein of male WAG/Rij rats. MTA (30 mg/kg, 60 mg/kg, and 90 mg/kg) was administered locoregionally by portal vein chemoembolization (PVCE) and compared with repeated systemic intravenous injection. GEM (50 mg/kg) was also given locoregionally by hepatic artery chemoembolization (HACE) as well as systemically. All routes of administration were examined alone as well as in combination. Efficacy of treatment in terms of liver metastases burden was determined at the end of the experiment by measuring the beta-galactosidase activity of CC531-lac-Z cells with a chemoluminescence assay. RESULTS: Combination experiments in vitro showed a more than additive tumor cell reduction after sequential exposure to MTA preceding GEM (observed/expected ratio [O/E] = 0.73). Experiments with the reverse sequence (GEM-->MTA) resulted only in additive combination effects (O/E ratio = 1.08). Simultaneous drug exposure showed less than additive combination effects (O/E ratios > or = 1.25). In vivo, locoregional administration by HACE with GEM was significantly more effective than systemic intravenous bolus treatment (P = 0.03). Portal vein chemoembolization with MTA performed immediately after tumor cell inoculation was ineffective. Repeated systemic treatment with MTA yielded a slight reduction in tumor cell load that was significant versus control at the medium and high doses (60 mg/kg, P = 0.009; 90 mg/kg, P = 0.046) but not versus intraportal chemoembolization. The combination treatment of systemic (60 and 90 mg/kg) or locoregional (60 mg/kg) MTA with HACE using GEM (50 mg/kg) resulted in more than 80% tumor growth inhibition; this antineoplastic combination effect was maximally additive. CONCLUSION: A regimen-dependent synergistic combination effect of both drugs was found in vitro. In animals, hepatic artery chemoembolization with GEM was superior to systemic intravenous bolus treatment. Portal vein chemoembolization with MTA was ineffective. The optimal in vitro regimen of MTA (intravenous or PVCE) preceding GEM (HACE) resulted in a maximally additive tumor growth inhibition. The results indicate that MTA and GEM can successfully be combined and favor further evaluation in patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Glutamates/therapeutic use , Guanine/analogs & derivatives , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chemoembolization, Therapeutic , Colorectal Neoplasms , Combined Modality Therapy , Death , Deoxycytidine/therapeutic use , Disease Models, Animal , Guanine/therapeutic use , Liver Neoplasms/drug therapy , Male , Neoplasm Metastasis , Pemetrexed , Rats , Rats, Inbred Strains , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...