Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
Methods Mol Biol ; 1866: 107-131, 2019.
Article in English | MEDLINE | ID: mdl-30725412

ABSTRACT

The elevated requirement for methionine (MET) of cancer cells is termed MET dependence. To selectively target the MET dependence of tumors for treatment on a large-scale preclinical and clinical basis, the L-methionine α-deamino-γ-mercaptomethane-lyase (EC 4.4.1.11) (methioninase, [METase]) gene from Pseudomonas putida has been cloned in Escherichia coli using the polymerase chain reaction (PCR). Purification using two DEAE Sepharose FF ion-exchange column and one ActiClean Etox endotoxin-affinity chromatography column has been established. Plasmid pMGLTrc03, which has a trc promoter and a spacing of 12 nucleotides between the Shine-Dalgarno sequence and the ATG translation initiation codon, was selected as the most suitable plasmid. The recombinant bacteria produced rMETase at 43% of the total proteins in soluble fraction by simple batch fermentation using a 500 L fermentor. Crystals were directly obtained from crude enzyme with 87% yield by a crystallization in the presence of 9.0% polyethylene glycol 6000, 3.6% ammonium sulfate, and 0.18 M sodium chloride using a 100 L crystallizer. After recrystallization, the enzyme was purified by anion-exchange column chromatography to remove endotoxins and by gel filtration for polishing. Purified rMETase is stable to lyophilization. In order to prevent immunological reactions which might be produced by multiple dosing of rMETase and to prolong the serum half-life of rMETase, the N-hydroxysuccinimidyl ester of methoxypolyethylene glycol propionic acid (M-SPA-PEG 5000) has been coupled to rMETase. The PEGylated molecules (PEG-rMETase) were purified from unreacted PEG with Amicon 30 K centriprep concentrators or by Sephacryl S-300 HR gel-filtration chromatography. Unreacted rMETase was removed by DEAE Sepharose FF anion-exchange chromatography. The resulting PEG-rMETase subunit, produced from a PEG/rMETase ratio of 30/1 in the synthetic reaction, had a molecular mass of approximately 53 kda determined by matrix-assisted laser desorption/ionization mass spectrometry, indicating the conjugation of two PEG molecules per subunit of rMETase and eight per tetramer. PEG-rMETase molecules obtained from reacting ratios of PEG/rMETase of 30/1 had an enzyme activity of 70% of unmodified rMETase. PEGylation of rMETase increased the serum half-life of the enzyme in rats to approximately 160 min compared to 80 min for unmodified rMETase. PEG-rMETase could deplete serum MET levels to less than 0.1 µM for approximately 8 h compared to 2 h for rMETase in rats. A significant prolongation of in vivo activity and effective MET depletion by the PEG-rMETase were achieved by the simultaneous administration of pyridoxal 5'-phosphate. rMETase was also 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 µM, respectively, from the PLP baseline of 0.3 µM. PLP increased the half-life of MEGC-PEG-rMETase holoenzyme in a dose-dependent manner. The extended time of MET depletion by MEGC-PEG-rMETase was due to the maintenance of active MEGC-PEG-rMETase holoenzyme by infused PLP.


Subject(s)
Carbon-Sulfur Lyases/therapeutic use , Neoplasms/drug therapy , Recombinant Proteins/therapeutic use , Animals , Apoenzymes/metabolism , Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/isolation & purification , Crystallization , Escherichia coli/metabolism , Fermentation , Mice, Inbred BALB C , Polyethylene Glycols/chemistry , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Pyridoxal Phosphate/administration & dosage , Pyridoxal Phosphate/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
4.
Methods Mol Biol ; 1866: 149-161, 2019.
Article in English | MEDLINE | ID: mdl-30725414

ABSTRACT

Methionine (MET) is a general target in cancer due to the excess requirement of MET by cancer cells. MET has been effectively restricted by recombinant methioninase (rMETase) in mouse models of cell-line tumors. This chapter reviews the efficacy of rMETase on patient-derived orthotopic xenograft (PDOX) mouse models of human cancer. Ewing's sarcoma is a recalcitrant disease even though development of multimodal therapy has improved patients' outcome. A Ewing's sarcoma was implanted in the right chest wall of nude mice to establish a PDOX model. rMETase effectively reduced tumor growth compared to the untreated control. The MET level both of plasma and supernatants derived from sonicated tumors was lower in the rMETase treatment group. Body weight did not significantly differ at any time points between the two groups. A PDOX nude mouse model of a BRAF V600E-mutant melanoma was established in the chest wall of nude mice and also tested with rMETase in combination with a first-line melanoma drug, temozolomide (TEM). Combination therapy of TEM and rMETase was significantly more efficacious than either monotherapy. The results reviewed in this chapter demonstrate the clinical potential of rMETase.


Subject(s)
Carbon-Sulfur Lyases/therapeutic use , Neoplasms/drug therapy , Recombinant Proteins/therapeutic use , Xenograft Model Antitumor Assays , Animals , Body Weight , Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/pharmacology , Cell Proliferation/drug effects , Humans , Mice, Nude , Mutation/genetics , Neoplasms/blood , Neoplasms/pathology , Proto-Oncogene Proteins B-raf/metabolism , Recombinant Proteins/biosynthesis , Temozolomide/pharmacology , Temozolomide/therapeutic use , Time Factors
5.
Cell Cycle ; 17(3): 356-361, 2018.
Article in English | MEDLINE | ID: mdl-29187018

ABSTRACT

The elevated methionine (MET) use by cancer cells is termed MET dependence and may be the only known general metabolic defect in cancer. Targeting MET by recombinant methioninase (rMETase) can arrest the growth of cancer cells in vitro and in vivo. We previously reported that rMETase, administrated by intra-peritoneal injection (ip-rMETase), could inhibit tumor growth in a patient-derived orthotopic xenograft (PDOX) model of a BRAF-V600E mutant melanoma. In the present study, we compared ip-rMETase and oral rMETase (o-rMETase) for efficacy on the melanoma PDOX. Melanoma PDOX nude mice were randomized into four groups of 5 mice each: untreated control; ip-rMETase (100 units, i.p., 14 consecutive days); o-rMETase (100 units, p.o., 14 consecutive days); o-rMETase+ip-rMETase (100 units, p.o.+100 units, i.p., 14 consecutive days). All treatments inhibited tumor growth on day 14 after treatment initiation, compared to untreated control (ip-rMETase, p<0.0001; o-rMETase, p<0.0001; o-rMETase+ip-rMETase, p<0.0001). o-rMETase was significantly more effective than ip-rMETase (p = 0.0086). o-rMETase+ip-rMETase was significantly more effective than either mono-therapy: ip-rMETase, p = 0.0005; or o-rMETase, p = 0.0367. The present study is the first demonstrating that o-rMETase is effective as an anticancer agent. The results of the present study indicate the potential of clinical development of o-rMETase as an agent for chronic cancer therapy and for cancer prevention and possibly for life extension since dietary MET reduction extends life span in many animal models.


Subject(s)
Carbon-Sulfur Lyases/administration & dosage , Carbon-Sulfur Lyases/therapeutic use , Melanoma/drug therapy , Melanoma/prevention & control , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Recombinant Proteins/therapeutic use , Xenograft Model Antitumor Assays , Administration, Oral , Aged , Animals , Carbon-Sulfur Lyases/blood , Carbon-Sulfur Lyases/pharmacology , Female , Humans , Melanoma/genetics , Mice, Nude , Recombinant Proteins/administration & dosage
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Anticancer Res ; 13(5A): 1465-8, 1993.
Article in English | MEDLINE | ID: mdl-8239522

ABSTRACT

Methionine dependence is a tumor-specific metabolic defect found in human cancer cell lines as well as in fresh human tumor specimens. Methionine dependent tumors cease growing when deprived of methionine, unlike normal cells which can substitute homocysteine for methionine for their growth requirement. We have previously purified a stable, endotoxin-free methioninase from the bacterium, Pseudomonas putida. We demonstrate in this report that purified methioninase can lower the serum levels of methionine in normal and nude mice from 60 microM to approximately 5 microM within 1 hour. The circulating half-life of methioninase is approximately 100 minutes in mice after i.v. injection. The enzyme therefore seems to be a good candidate as an antitumor agent for methionine-dependent tumors.


Subject(s)
Carbon-Sulfur Lyases/pharmacology , Methionine/blood , Animals , Carbon-Sulfur Lyases/administration & dosage , Carbon-Sulfur Lyases/blood , Half-Life , Injections, Intravenous , Methionine/administration & dosage , Mice , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...