Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 182: 394-401, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33839182

ABSTRACT

The methionine dependence is a well known phenomenon in metabolism of cancer cells. Methionine γ-lyase (EC 4.4.1.11, MGL) catalyzes the γ-elimination reaction of L-methionine and thus could effectively inhibit the growth of malignant cells. Recently we have demonstrated that the mutant form of the enzyme C115H MGL can be used as a component of the pharmacological pair enzyme/S-(allyl/alkyl)-L-cysteine sulfoxides to yield thiosulfinates in situ. Thiosulfinates were shown to be toxic to various cancer cell lines. Therefore the application of the enzyme in enzyme pro-drug therapy may be promising. The conjugates of MGL and C115H MGL with polysialic acid were obtained and their kinetic and pharmacokinetic parameters were determined. The formation of polysialic shell around the enzyme was confirmed by atomic force microscopy. The half-life of conjugated enzymes increased 3-6 times compared to the native enzyme. The cytotoxic effect of conjugated MGL against methionine dependent cancer cell lines was increased two times compared to the values for the native enzymes. The anticancer efficiency of thiosulfinates produced by pharmacological pair C115H MGL/S-(allyl/alkyl)-L-cysteine sulfoxides was demonstrated in vitro. The results indicate that the conjugates of MGL with polysialic acid could be new antitumor drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Carbon-Sulfur Lyases/chemistry , Neoplasms/drug therapy , Sialic Acids/chemistry , Animals , Antineoplastic Agents/therapeutic use , Carbon-Sulfur Lyases/metabolism , Carbon-Sulfur Lyases/pharmacokinetics , Carbon-Sulfur Lyases/pharmacology , Cell Line, Tumor , Female , Humans , Kinetics , MCF-7 Cells , Mice , Mice, Inbred BALB C , Neoplasms/therapy , Sialic Acids/pharmacology , Sialic Acids/therapeutic use
2.
Clin Pharmacol Drug Dev ; 9(8): 1003-1014, 2020 11.
Article in English | MEDLINE | ID: mdl-32627420

ABSTRACT

ABP 798 is a proposed biosimilar to rituximab reference product (RP), an anti-CD20 monoclonal antibody. Pharmacokinetics (PK), pharmacodynamics (PD), and safety results from the comparative clinical study that evaluated the PK, PD, safety, efficacy, and immunogenicity of ABP 798 versus rituximab RP are presented here. Subjects with moderate to severe rheumatoid arthritis (RA) received 2 doses of ABP 798, United States-sourced RP (rituximab US) or European Union-sourced RP (rituximab EU), each consisting of two 1000-mg infusions 2 weeks apart. For the second dose (week 24), ABP 798- and rituximab EU-treated subjects received the same treatment; rituximab US-treated subjects transitioned to ABP 798. End points included area under the serum concentration-time curve from time 0 extrapolated to infinity and maximum observed serum concentration following the second infusion of the first dose (PK) and percentage of subjects with complete CD19+ cell depletion days 1-33 (PD). Primary analysis established PK similarity between ABP 798 and rituximab RP based on 90% confidence intervals of the adjusted geometric mean ratios being within a prespecified equivalence margin of 0.8 and 1.25. Complete CD19+ B-cell depletion on day 3 among groups confirmed PD similarity. These findings demonstrated PK/PD similarity between ABP 798 and rituximab RP in subjects with moderate to severe RA.


Subject(s)
Antirheumatic Agents/pharmacokinetics , Arthritis, Rheumatoid/drug therapy , Biosimilar Pharmaceuticals/pharmacokinetics , Carbon-Sulfur Lyases/pharmacokinetics , Rituximab/pharmacokinetics , Adult , Aged , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/blood , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/blood , Antirheumatic Agents/pharmacology , Area Under Curve , Biosimilar Pharmaceuticals/administration & dosage , Biosimilar Pharmaceuticals/blood , Biosimilar Pharmaceuticals/pharmacology , Carbon-Sulfur Lyases/administration & dosage , Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/pharmacology , Double-Blind Method , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Rituximab/administration & dosage , Rituximab/blood , Rituximab/pharmacology , Safety , Severity of Illness Index , Therapeutic Equivalency , Treatment Outcome
3.
Int J Biol Macromol ; 140: 1277-1283, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31470057

ABSTRACT

Citrobacter freundii methionine γ-lyase (MGL), in addition to the physiological reaction, catalyzes the ß-elimination reaction of S-alk(en)yl-L-cysteine sulfoxides to yield thiosulfinates, which have antibacterial activity. We have obtained the mutant form C115H MGL, which cleaves S-alk(en)yl-L-cysteine sulfoxides more effectively than the wild type enzyme does. The binary system MGL/S-alk(en)yl-L-cysteine sulfoxides may be considered as a new pharmacological pair in enzyme prodrug therapy (EPT). Despite of the successful application of this pair in antibacterial studies in vitro, in vivo experiments may lead to several problems typical of therapeutic proteins including a relatively short-lasting biological activity. To circumvent these problems, we have investigated several approaches to improve safety and efficacy of the enzyme component of the pharmacological pair. This included covalent attachment of poly(ethylene glycol) to the enzyme, its encapsulation in liposomes and polymeric vesicles (PICsomes). The steady-state and pharmacokinetic parameters of modified/encapsulated enzyme were determined. It was demonstrated that the encapsulation in PICsomes prolongs in vivo stability of C115H MGL to over 42 h compared to PEGylated enzyme (3 h). Antibacterial activity of binary system ("pharmacological pair") modified/encapsulated enzyme/S-alk(en)yl-L-cysteine sulfoxides was tested and remained the same as for the naked enzyme. Thus, the usage of MGL-loaded PICsomes as enzymatic nanoreactors in ETP to produce antimicrobial thiosulfinates is promising.


Subject(s)
Carbon-Sulfur Lyases/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Anti-Infective Agents/pharmacology , Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/pharmacology , Citrobacter freundii/enzymology , Female , Liposomes , Mice, Inbred BALB C , Microbial Sensitivity Tests , Polyethylene Glycols/chemistry , Prodrugs/pharmacology
4.
Methods Mol Biol ; 1866: 231-242, 2019.
Article in English | MEDLINE | ID: mdl-30725419

ABSTRACT

Methionine (MET) has been shown to be a tumor-selective therapeutic target for cancer, since cancer cells require higher amounts of MET to divide and survive than normal cells. This phenomena is known as MET dependence and is probably due to MET overuse by cancer cells. A pilot clinical trial was initially carried out with non-recombinant METase (METase) produced from Pseudomonas putida and subsequently highly purified. No acute clinical toxicity was observed for any criteria measured in the three patients. The depletion of serum MET started within 30 min of the infusion and was maintained for 4 h after the infusion was completed in patient 1 and patient 2. The lowest serum MET levels were 35% and 19% of the pretreatment level, respectively, in patient 1 and patient 2. Patient 3 received a 10 h i.v. infusion of METase without any sign of side effects. MET was depleted over 200-fold from 23.1 to 0.1 µM by the 10-h infusion of patient 3. No clinical toxicity was observed in any criteria measured in patient 3. Subsequently, another pilot Phase I clinical trial was carried out of serum MET depletion in cancer patients by recombinant METase (rMETase) cloned from Pseudomonas putida and produced in E. coli. Patients with advanced breast cancer, lung cancer, renal cancer, and lymphoma were given a single rMETase treatment at doses ranging from 5000 to 20,000 units by i.v. infusion over 6-24 h. No clinical toxicity was observed in any patient after rMETase treatment. rMETase levels were measured at 0.1 to 0.4 units per ml of serum in the patients which correspond to therapeutic levels in vitro. The lowest serum MET levels in rMETase-treated patients were 0.1% of the pretreatment levels corresponding to approximately 0.1 µM, which also correlates to therapeutic levels in vitro as well as in vivo. The results of the METase and rMETase pilot Phase I clinical trials therefore indicate that i.v. infusion of rMETase is safe and effectively depletes its biochemical target of serum MET, suggesting potential efficacy in future clinical trials.


Subject(s)
Carbon-Sulfur Lyases/therapeutic use , Methionine/blood , Neoplasms/drug therapy , Neoplasms/pathology , Carbon-Sulfur Lyases/isolation & purification , Carbon-Sulfur Lyases/pharmacokinetics , Fermentation , Humans , Infusions, Intravenous , Neoplasm Staging , Neoplasms/blood , Pilot Projects
5.
Cancer Med ; 6(6): 1437-1452, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28544589

ABSTRACT

Erymet is a new therapy resulting from the encapsulation of a methionine gamma-lyase (MGL; EC number 4.4.1.11) in red blood cells (RBC). The aim of this study was to evaluate erymet potential efficacy in methionine (Met)-dependent cancers. We produced a highly purified MGL using a cGMP process, determined the pharmacokinetics/pharmacodynamics (PK/PD) properties of erymet in mice, and assessed its efficacy on tumor growth prevention. Cytotoxicity of purified MGL was tested in six cancer cell lines. CD1 mice were injected with single erymet product supplemented or not with vitamin B6 vitamer pyridoxine (PN; a precursor of PLP cofactor). NMRI nude mice were xenografted in the flank with U-87 MG-luc2 glioblastoma cells for tumor growth study following five intravenous (IV) injections of erymet with daily PN oral administration. Endpoints included efficacy and event-free survival (EFS). Finally, a repeated dose toxicity study of erymet combined with PN cofactor was conducted in CD1 mice. Recombinant MGL was cytotoxic on 4/6 cell lines tested. MGL half-life was increased from <24 h to 9-12 days when encapsulated in RBC. Conversion of PN into PLP by RBC was demonstrated. Combined erymet + PN treatment led to a sustained Met depletion in plasma for several days with a 85% reduction of tumor volume after 45 days following cells implantation, and a significant EFS prolongation for treated mice. Repeated injections in mice exhibited a very good tolerability with only minor impact on clinical state (piloerection, lean aspect) and a slight decrease in hemoglobin and triglyceride concentrations. This study demonstrated that encapsulation of methioninase inside erythrocyte greatly enhanced pharmacokinetics properties of the enzyme and is efficacy against tumor growth. The perspective on these results is the clinical evaluation of the erymet product in patients with Met starvation-sensitive tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Carbon-Sulfur Lyases/administration & dosage , Drug Delivery Systems , Erythrocytes , Neoplasms/drug therapy , Pyridoxine/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Carbon-Sulfur Lyases/pharmacokinetics , Carbon-Sulfur Lyases/therapeutic use , Carbon-Sulfur Lyases/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Male , Methionine/blood , Methionine/metabolism , Mice, Nude , Neoplasms/blood , Neoplasms/metabolism , Neoplasms/pathology , Pyridoxal Phosphate/blood , Pyridoxine/pharmacokinetics , Pyridoxine/therapeutic use , Pyridoxine/toxicity , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Tumor Burden/drug effects
6.
Oncotarget ; 8(22): 35630-35638, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28404944

ABSTRACT

Methionine dependence is due to the overuse of methionine for aberrant transmethylation reactions in cancer. Methionine dependence may be the only general metabolic defect in cancer. In order to exploit methionine dependence for therapy, our laboratory previously cloned L-methionine α-deamino-γ-mercaptomethane lyase [EC 4.4.1.11]). The cloned methioninase, termed recombinant methioninase, or rMETase, has been tested in mouse models of human cancer cell lines. Ewing's sarcoma is recalcitrant disease even though development of multimodal therapy has improved patients'outcome. Here we report efficacy of rMETase against Ewing's sarcoma in a patient-derived orthotopic xenograft (PDOX) model. The Ewing's sarcoma was implanted in the right chest wall of nude mice to establish a PDOX model. Eight Ewing's sarcoma PDOX mice were randomized into untreated control group (n = 4) and rMETase treatment group (n = 4). rMETase (100 units) was injected intraperitoneally (i.p.) every 24 hours for 14 consecutive days. All mice were sacrificed on day-15, 24 hours after the last rMETase administration. rMETase effectively reduced tumor growth compared to untreated control. The methionine level both of plasma and supernatants derived from sonicated tumors was lower in the rMETase group. Body weight did not significantly differ at any time points between the 2 groups. The present study is the first demonstrating rMETase efficacy in a PDOX model, suggesting potential clinical development, especially in recalcitrant cancers such as Ewing's sarcoma.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carbon-Sulfur Lyases/pharmacology , Recombinant Proteins/pharmacology , Sarcoma, Ewing/pathology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Biopsy , Carbon-Sulfur Lyases/administration & dosage , Carbon-Sulfur Lyases/pharmacokinetics , Disease Models, Animal , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Sarcoma, Ewing/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Biomed Pharmacother ; 88: 978-984, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28178629

ABSTRACT

PK studies were carried out after a single i.v. administration of 500 and 1000 U/kg by measuring of MGL activity in plasma samples. L-methionine concentration was measured by mass spectrometry. After single i.v. injection of 500U/kg the circulating T1/2 of enzymes in mice varies from 73 to 123min. The AUC0-tinf values determined for MGL 500U/kg from C. freundii, C. tetani and C. sporogenes are 8.21±0.28, 9.04±0.33 and 13.88±0.39U/(ml×h), respectively. Comparison of PK parameters of three MGL sources in the dose of 500U/kg indicated the MGL C. sporogenes to have better PK parameters: clearance 0.83(95%CI: 0.779-0.871) - was lower than C. tetanii 1.27(95%CI: 1.18-1.36) and C. freundii 1.39(95%CI: 1.30-1.49). Mice plasma methionine decreased to undetectable level 10min after MGL 1000 U/kg injection. After MGL C. sporogenes 500U/kg injection plasma methionine level completely omitted after 10min till 6h, assuming the sustainability of negligible levels of methionine (<5µM) in plasma of mice for about 6h. The recovery of methionine concentration showed the advantageous efficiency of MGL from C. sporogenes: 95% 0.010-0.022 vs 0.023-0.061 for MGL C. freundii and 0.036-0.056 for MGL C. tetani. There are no significant differences between methionine cleavage after MGL C. tetani and MGL C. sporogenes i.v. injection at all doses. MGL from C. sporogenes may be considered as promising enzyme for further investigation as potential anticancer agent.


Subject(s)
Carbon-Sulfur Lyases/pharmacokinetics , Citrobacter freundii/enzymology , Clostridium/enzymology , Methionine/blood , Methionine/pharmacokinetics , Animals , Carbon-Sulfur Lyases/administration & dosage , Carbon-Sulfur Lyases/blood , Female , Mice, Inbred C57BL , Nonlinear Dynamics , Regression Analysis
8.
Enzyme Microb Technol ; 51(4): 200-10, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22883554

ABSTRACT

Methionine starvation can powerfully modulate DNA methylation, cell cycle transition, polyamines and antioxidant synthesis of tumor cells, in contrary to normal ones. Aspergillus flavipesl-methioninase was previously characterized by our studies, displaying affordable biochemical properties comparing to Pseudomonas putida enzyme (ONCASE). Thus, the objective of current study was to evaluate the catalytic properties of Af-METase in New Zealand rabbits, exploring its antitumor efficacy. In vivo, Af-METase (40.8 U/ml) have T(1/2) 19.8 h, elimination constant 0.088 U/h and apparent volume distribution 85 U/ml. Also, Af-METase has two maxima one at A(280 nm) (apo-enzyme) and at A(420 nm) (internal Schiff base of PLP), unlike control plasma (without enzyme). The two peaks of absorption spectra were detected maximally at 15 min then the absorbance at 420 nm was subsequently decreased with circulation time, due to dissociation of the co-enzyme. The A280/420 ratio was increased from 1.69 to 5.81 with circulation time from 15 to 30 h. Rabbits plasma methionine was depleted from 18.7 µM (control) to 8.8 µM after 1h of enzyme injection and completely omitted after 2 h till 19 h, assuming the sustainability of negligible levels of methionine (< 2 µM) in plasma of rabbits, for about 17 h. Upon infusion of PLP, the T(½) of Af-METase was significantly prolonged by 3.2 fold, assuming the fully reconstitution of the enzyme. The holo-AfMETase still retained its co-enzyme, completely, till 33 h of PLP infusion. From spectral studies, the internal aldimine linkage of apo-Af-METase was constructed upon PLP infusion, with fully catalytic structure after less than 4h of its infusion, the A280/420 ratio being not relatively changed till 45 h. After 25 days of last enzyme dose, the titer of IgG was increase by about 1.66 fold comparing to control (without enzyme). However, IgM was not detected along the tested challenge points. In vitro, plasma anti-Af-METase neutralizing antibodies (NAb) were assessed, with no significant reduction on activity of Af-METase by Nab. All the hematological parameters were in normal range, otherwise, the RBCs titer and platelet level was slightly increased, after 25 days of Af-METase injection, comparing to control. There is no obvious negative effect on chemistry of liver, kidney, glucose, lipids, and other electrolytes. Additionally, the anticancer activity of Af-METase was evaluated against five types of human cancer cell lines, in vitro. The enzyme showed a powerful activity against prostate (PC3), liver (HEPG2) and breast (MCF7) cancers, with IC50 0.001 U/ml, 0.26 U/ml and 0.37 U/ml, respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Aspergillus/enzymology , Carbon-Sulfur Lyases/immunology , Carbon-Sulfur Lyases/pharmacokinetics , MCF-7 Cells/drug effects , Methionine/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Aspergillus/classification , Aspergillus/metabolism , Carbon-Sulfur Lyases/isolation & purification , Carbon-Sulfur Lyases/pharmacology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Half-Life , Humans , Lung Neoplasms/drug therapy , Male , Prostatic Neoplasms/drug therapy , Rabbits
9.
J Drug Target ; 19(5): 326-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20678009

ABSTRACT

Human ovarian cancer cells specifically bind the isoflavone daidzein. A chemical conjugate between daidzein and the garlic enzyme alliinase was prepared. The conjugate specifically bound to ovarian cancer cells and upon addition of the prodrug alliin, it effectively produced cytotoxic allicin molecules which killed the cancer cells. In vivo targeting and antitumor effect was confirmed by NIR and bioluminescence imaging using daidzein-alliinase-CyTE-777 conjugates and luciferase-expressing ovarian cancer cells. Co-localization of the fluorescent conjugate with bioluminescence was observed for intraperitoneal tumors while nonconjugated alliinase did not accumulate. Biodistribution studies with Europium-labeled conjugate revealed a five fold higher uptake in tumors as compared to other tissues. Treatment of tumor bearing mice with daidzein-alliinase and alliin effectively attenuated tumor progression during the first 12 days while a 5-fold increase in bioluminescence was detected in placebo-treated animals. Autopsy revealed only small individual foci of luminescence at the site of tumor cells inoculation. Histological examination of organs and tissues did not reveal any additional foci of carcinoma or signs of toxicity. These results suggest that the targeted alliinase conjugates in the presence of alliin, generated therapeutically effective levels of allicin which were capable of suppressing tumor progression of intraperitoneal ovarian cancer in an animal model.


Subject(s)
Antineoplastic Agents/pharmacology , Carbon-Sulfur Lyases/pharmacology , Cysteine/analogs & derivatives , Isoflavones/pharmacology , Ovarian Neoplasms/drug therapy , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/pharmacokinetics , Carbon-Sulfur Lyases/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cysteine/chemistry , Cysteine/pharmacokinetics , Cysteine/pharmacology , Cysteine/therapeutic use , Drug Compounding , Female , Fluorescent Dyes/chemistry , Humans , Isoflavones/chemistry , Isoflavones/pharmacokinetics , Isoflavones/therapeutic use , Luciferases/genetics , Mice , Mice, Nude , Molecular Imaging , Ovarian Neoplasms/metabolism , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Tissue Distribution , Transfection , Treatment Outcome , Xenograft Model Antitumor Assays
10.
Cancer Res ; 66(5): 2807-14, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510603

ABSTRACT

A highly potent recombinant L-methionine gamma-lyase (METase) conjugated with polyethylene glycol (PEG) was characterized physicochemically and pharmacokinetically in vivo and in vitro. Pegylated METase (PEG-METase), which contains pyridoxal 5'-phosphate (PLP) as a cofactor in the molecule, is a potent anticancer agent that can deplete L-methionine from plasma. Although pegylation decreased its specific activity, dithiothreitol (DTT) treatment increased it over three times with the detachment of one PEG moiety modified with a cysteine residue. We can produce DTT-treated PEG-METase on a large scale in sufficient quality for therapeutic use. The superiority of DTT-treated PEG-METase was confirmed by the enhancement of L-methionine depletion and amelioration of pharmacokinetics in mice. The holoenzyme of DTT-treated PEG-METase gave a several times larger area under the plasma concentration curve than that of DTT-untreated PEG-METase, not because of an increase of the half-life but because of high specific activity. Conversely, simultaneous PLP infusion led to a greatly increased half-life of the holoenzyme. DTT-treated PEG-METase administration with PLP infusion was the most useful combination for maximizing the potency of the enzyme. We showed that serum albumin interfered with holoenzyme activity in vitro. The decrease of holoenzyme activity was dependent on the type of serum albumin. We concluded that PLP was released from PEG-METase by serum albumin in vivo and in vitro. The deleterious effect of PLP dissociation from PEG-METase could be improved by supplementing PLP and oleic acid. Their synergistic effect in preventing a decrease of the holoenzyme activity was also observed.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carbon-Sulfur Lyases/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/pharmacology , Dithiothreitol/pharmacology , Female , Half-Life , Humans , Methionine/metabolism , Mice , Mice, Inbred BALB C , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Serum Albumin/metabolism , Serum Albumin/pharmacology , gamma-Globulins/metabolism , gamma-Globulins/pharmacology
11.
Cancer Res ; 64(18): 6673-8, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374983

ABSTRACT

Methionine depletion by recombinant methioninase (rMETase) has been demonstrated previously to be highly effective in tumor-bearing mouse models. However, the therapeutic potential of rMETase has been limited by its short plasma half-life and immunologic effects, including high antibody production in mice and monkeys and anaphylactic reactions in monkeys. To overcome these limits of rMETase, the enzyme has been coupled to methoxypolyethylene glycol succinimidyl glutarate (MEGC-PEG-5000). In this study, we evaluated the pharmacokinetics, antigenicity and toxicity of MEGC-PEG-rMETase in Macaca fascicularis monkeys using an escalating-dose strategy. Dose ranging studies at 1,000, 4,000, and 8,000 units/kg i.v. determined that a single dose of 4,000 units/kg was sufficient to reduce plasma methionine to <5 micromol/L for 12 hours. Pharmacokinetic analysis with the single 4,000 units/kg dose showed that MEGC-PEG-rMETase holoenzyme activity was eliminated with a biological half-life of 1.3 hours, and the MEGC-PEG-rMETase apoenzyme was eliminated with a biological half-life of 90 hours, an approximately 36-fold increase compared with non-PEGylated rMETase. A single dose at 2,000 units/kg of MEGC-PEG-rMETase resulted in an apoenzyme half-life of 143 hours. A seven-day i.v. administration of 4,000 units/kg every 12 hours resulted in a steady-state depletion of plasma methionine to <5 micromol/L. The only manifest toxicity was decreased food intake and slight weight loss. Red cell values and hemoglobin declined transiently during treatment but recovered after cessation of treatment. Subsequent challenges on days 29, 50 and, 71 did not result in any immunologic reactions. This result is in contrast to non-PEGylated rMETase, which elicited anaphylactic reactions in monkeys. Anti-MEGC-PEG-rMETase antibodies (at 10(-2)) were found on day 29, and these increased to 10(-3) to 10(4) on day 71, 100 to 1,000-fold less than antibodies elicited by naked rMETase. Although anti-MEGC-PEG-rMETase antibodies were produced, no neutralizing antibody was identified, and each challenge dose was effective in depleting plasma methionine levels. The results of the present study demonstrate that PEGylation greatly prolongs serum half-life of the rMETase apoenzyme and eliminated anaphylactic reactions. The results indicate a profile with respect to serum half-life, toxicity, and antigenicity that suggest clinical potential of MEGC-PEG-rMETase.


Subject(s)
Carbon-Sulfur Lyases/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Animals , Antibodies/blood , Body Weight/drug effects , Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/immunology , Carbon-Sulfur Lyases/pharmacology , Dose-Response Relationship, Drug , Drug Carriers , Eating/drug effects , Half-Life , Macaca fascicularis , Male , Methionine/deficiency , Methionine/metabolism , Polyethylene Glycols/pharmacology , Recombinant Proteins/blood , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
12.
Cancer Res ; 64(16): 5775-8, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313919

ABSTRACT

Recombinant methioninase (rMETase) has been shown to target the elevated methionine (MET) dependence of tumor cells and arrest their growth as well as make tumors more sensitive to standard chemotherapy agents. Polyethylene glycol (PEG)-modified rMETase (PEG-rMETase) has reduced antigenicity compared with unmodified rMETase. However, PEG-rMETase has a limited active circulating half-life due to rapid in vivo dissociation of its cofactor pyridoxal-5'-phosphate (PLP), a surprising finding, because PLP is tightly bound to PEG-rMETase in buffer. The question asked in the current study was on the effect of increasing doses of PLP to extend the circulating half-life of active PEG-rMETase holoenzyme in vivo. rMETase was conjugated with methoxypolyethylene glycol succinimidyl glutarate 5000 (MEGC-PEG). Miniosmotic pumps containing various concentrations of PLP were implanted in BALB-C mice. PLP-infused mice were then injected with a single dose of 4000 or 8000 units/kg PEG-rMETase. Mice infused with 5, 50, 100, 200, and 500 mg/ml PLP-containing miniosmotic pumps increased plasma PLP to 7, 24, 34, 60, and 95 microm, respectively, from the PLP baseline of 0.3 microm. PLP increased the half-life of MEGC-PEG-rMETase holoenzyme in a dose-dependent manner. Pumps containing 500 mg/ml PLP increased the half-life of MEGC-PEG-rMETase holoenzyme 4.5-fold from 1.5 to 7 h. Infused PLP did not extend the half-life of MEGC-PEG-rMETase apoenzyme. With a dose of 4000 units/kg MEGC-PEG-rMETase in the mice infused with 5, 50, 200, and 500 mg/ml PLP, plasma MET was depleted from 50 microm to < or = 5 microm for 8, 24, 72, and 72 h, respectively. Thus, PLP infusion could extend the period of MET depletion by MEGC-PEG-rMETase by approximately 10-fold in a dose-dependent manner. The mice given 8000 units/kg MEGC-PEG-rMETase showed a similar plasma MET depletion time course, indicating that the limiting factor for MEGC-PEG-rMETase-mediated MET depletion in vivo was PLP. The extended time of MET depletion by MEGC-PEG-rMETase was due to the maintenance of active MEGC-PEG-rMETase holoenzyme by infused PLP. The infused PLP either bound to apo-MEGC-PEG-rMETase and/or inhibited dissociation of PLP from holo-PEG-rMETase, thereby maintaining the holoenzyme form of MEGC-PEG-rMETase in vivo. The combination of MEGC-PEG-rMETase treatment with PLP infusion suggests an effective clinical strategy for long-term MET depletion to arrest cancer growth.


Subject(s)
Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Pyridoxal Phosphate/pharmacology , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics , Dose-Response Relationship, Drug , Half-Life , Mice , Mice, Inbred BALB C , Mice, Nude , Polyethylene Glycols/chemistry , Recombinant Proteins/blood , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics
13.
Clin Cancer Res ; 10(6): 2131-8, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15041734

ABSTRACT

Pharmacokinetics, methionine depletion, antigenicity, and toxicity of recombinant methioninase (rMETase), which has shown efficacy in achieving cell kill in a broad range of human tumor models, were examined in macaque monkeys. Dose-ranging studies at 1000, 2000, and 4000 units/kg i.v. identified the 4000 units/kg dose as able to reduce plasma methionine to an undetectable level (less than 0.5 microM) by 30 min, and the level so remained for 8 h. Pharmacokinetic analysis showed that rMETase was eliminated with a T(1/2) of 2.49 h. A 2-week i.v. administration of 4000 units/kg every 8 h/day for 2 weeks resulted in a steady-state depletion of plasma methionine to less than 2 microM. The only manifest toxicity was decreased food intake and slight weight loss. Serum albumin and red cell values declined transiently during treatment, which may be related to extensive blood sampling. Re-challenge on day 28 resulted in anaphylactic shock and death in one animal. Subsequent pretreatment with hydrocortisone prevented the anaphylactic reaction, although vomiting was frequently observed. Re-challenge was carried out at days 66, 86, and 116. Anti-rMETase antibodies (at 10(-3)) were found after the first challenge, and these increased to 10(-6) after the fourth challenge and decreased to 10(-2) by 2 months post therapy. The main rMETase antibody was IgG, and although it has some in vitro features of being a neutralizing antibody, each challenge dose was effective in depleting plasma methionine levels. Thus, rMETase was able to effectively deplete plasma methionine levels with minimal toxicity in a primate model. These data provide the bases for alteration by polyethyleneglycol conjugation (PEGylation) of the enzyme to increase its duration of effect and reduce its immunogenicity.


Subject(s)
Carbon-Sulfur Lyases/pharmacokinetics , Methionine/blood , Animals , Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/metabolism , Half-Life , Humans , Kinetics , Macaca fascicularis , Male , Primates , Recombinant Proteins/blood , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics
14.
Ceska Slov Farm ; 50(1): 12-20, 2001 Jan.
Article in Czech | MEDLINE | ID: mdl-11242829

ABSTRACT

The paper sums up new experimental knowledge concerning the individual groups of organic sulfurous substances of the garlic: sulfoxides, thiosulfinate, ajoens, vinyldithiines, alkyl and alkene sulfides and glutamylpeptides of sulfurous amino acids, their transformation reactions (based on the temperature, pH, extraction medium, and time) and the final products of transformations (Scheme 1, 2). It deals with the activity of the enzyme alliinase necessary for the transformation of sulfoxides present in the whole garlic, its isolation and stability as well as the stability of the dominant thiosulfinate allicin in various media and simulated body fluids. It refers to the studies of the metabolism and transformations of the most important sulfurous components performed in vitro on the hepatocytes and on the isolated rat liver, and those carried out in vivo on the rats and including the examination of the composition of the exhaled air. It follows from published papers that all different degradation products of thiosulfinates, mainly the prevailing allicin, are carriers of various biological activities. The paper also lists the types of commercial preparations prepared from the garlic, their differences, and considerable variability of their contents of active principles.


Subject(s)
Garlic/chemistry , Plants, Medicinal , Sulfur Compounds/analysis , Animals , Biotransformation , Carbon-Sulfur Lyases/pharmacokinetics , Garlic/therapeutic use , Humans , Liver/metabolism , Phytotherapy , Sulfur Compounds/chemistry , Sulfur Compounds/pharmacokinetics
15.
Anticancer Res ; 17(5B): 3857-60, 1997.
Article in English | MEDLINE | ID: mdl-9427792

ABSTRACT

The tumor-specific increased minimal requirement for methionine has been shown to be a highly promising therapeutic target. To attack this target we have previously cloned the methioninase gene from Pseudomonas putida and produced recombinant methioninase (rMETase). A pilot Phase I clinical trial has been carried out to determine rMETase toxicity, rMETase pharmacokinetics, and serum MET-depletion in cancer patients. Patients with advanced breast cancer, lung cancer, renal cancer and lymphoma were given a single rMETase treatment at doses ranging from 5,000 to 20,000 units by i.v. infusion over 6-24 hours. No clinical toxicity was observed in any patient after rMETase treatment. rMETase levels reached 0.1 to 0.4 units per ml of serum in the patients which correspond to therapeutic levels in vitro. The lowest serum methionine levels in rMETase-treated patients were 0.1% of the pre-treatment levels corresponding to approximately 0.1 microM, which also correlates to therapeutic levels in vitro. The results of the rMETase pilot Phase I clinical trial therefore indicate that i.v. infusion of rMETase is safe and effectively depletes its biochemical target of serum methionine suggesting potential efficacy in future clinical trials.


Subject(s)
Antimetabolites, Antineoplastic/blood , Carbon-Sulfur Lyases/blood , Methionine/blood , Neoplasms/blood , Neoplasms/drug therapy , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Carbon-Sulfur Lyases/administration & dosage , Carbon-Sulfur Lyases/biosynthesis , Carbon-Sulfur Lyases/pharmacokinetics , Humans , Neoplasm Staging , Neoplasms/pathology , Pilot Projects , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...