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










Publication year range
1.
Bioorg Chem ; 140: 106820, 2023 11.
Article in English | MEDLINE | ID: mdl-37672952

ABSTRACT

4-(3-Alkyl-2-oxoimidazolidin-1-yl)-N-phenylbenzenesulfonamides (PAIB-SAs) are members of a new family of prodrugs bioactivated by cytochrome P450 1A1 (CYP1A1) in breast cancer cells into their potent 4-(2-oxoimidazolidin-1-yl)-N-phenylbenzenesulfonamide metabolites (PIB-SAs). One of the predominant problems for the galenic formulation and administration of PAIB-SAs in animal studies is their poor hydrosolubility. To circumvent that difficulty, we report the design, the synthesis, the chemical characterization, the evaluation of the aqueous solubility, the antiproliferative activity and the mechanism of action of 18 new Na+, K+ and Li+ salts of PAIB-SAs. Our results evidenced that the latter exhibited highly selective antiproliferative activity toward MCF7 and MDA-MB-468 breast cancer cells expressing endogenously CYP1A1 compared to insensitive MDA-MB-231 and HaCaT cells. Moreover, PAIB-SA salts 1-18 are significantly more hydrosoluble (3.9-9.4 mg/mL) than their neutral counterparts (< 0.0001 mg/mL). In addition, the most potent PAIB-SA salts 1-3 and 10-12 arrested the cell cycle progression in the G2/M phase and disrupted the cytoskeleton's dynamic assembly. Finally, PAIB-SA salts are N-dealkylated by CYP1A1 into their corresponding PIB-SA metabolites, which are potent antimitotics. In summary, our results show that our water-soluble PAIB-SA salts, notably the sodium salts, still exhibit potent antiproliferative efficacy and remain prone to CYP1A1 bioactivation. In addition, these PAIB-SA salts will allow the development of galenic formulations suitable for further biopharmaceutical and pharmacodynamic studies.


Subject(s)
Antimitotic Agents , Breast Neoplasms , Cytochrome P-450 CYP1A1 , Prodrugs , Animals , Antimitotic Agents/chemistry , Antimitotic Agents/pharmacokinetics , Antimitotic Agents/pharmacology , Cytochrome P-450 CYP1A1/metabolism , Prodrugs/chemistry , Prodrugs/pharmacology , Salts , Humans
2.
Cell Death Dis ; 12(3): 268, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712556

ABSTRACT

Targeting cell division by chemotherapy is a highly effective strategy to treat a wide range of cancers. However, there are limitations of many standard-of-care chemotherapies: undesirable drug toxicity, side-effects, resistance and high cost. New small molecules which kill a wide range of cancer subtypes, with good therapeutic window in vivo, have the potential to complement the current arsenal of anti-cancer agents and deliver improved safety profiles for cancer patients. We describe results with a new anti-cancer small molecule, WEHI-7326, which causes cell cycle arrest in G2/M, cell death in vitro, and displays efficacious anti-tumor activity in vivo. WEHI-7326 induces cell death in a broad range of cancer cell lines, including taxane-resistant cells, and inhibits growth of human colon, brain, lung, prostate and breast tumors in mice xenografts. Importantly, the compound elicits tumor responses as a single agent in patient-derived xenografts of clinically aggressive, treatment-refractory neuroblastoma, breast, lung and ovarian cancer. In combination with standard-of-care, WEHI-7326 induces a remarkable complete response in a mouse model of high-risk neuroblastoma. WEHI-7326 is mechanistically distinct from known microtubule-targeting agents and blocks cells early in mitosis to inhibit cell division, ultimately leading to apoptotic cell death. The compound is simple to produce and possesses favorable pharmacokinetic and toxicity profiles in rodents. It represents a novel class of anti-cancer therapeutics with excellent potential for further development due to the ease of synthesis, simple formulation, moderate side effects and potent in vivo activity. WEHI-7326 has the potential to complement current frontline anti-cancer drugs and to overcome drug resistance in a wide range of cancers.


Subject(s)
Antimitotic Agents/pharmacology , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Animals , Antimitotic Agents/pharmacokinetics , Antimitotic Agents/toxicity , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mitosis/drug effects , Neoplasms/pathology , PC-3 Cells , Rats, Sprague-Dawley , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Biochem Pharmacol ; 184: 114364, 2021 02.
Article in English | MEDLINE | ID: mdl-33310050

ABSTRACT

Eg5, the product of Kif11 gene, also known as kinesin spindle protein, is a motor protein involved in the proper establishment of a bipolar mitotic spindle. Eg5 is one of the 45 different kinesins coded in the human genome of the kinesin motor protein superfamily. Over the last three decades Eg5 has attracted great interest as a promising new mitotic target. The identification of monastrol as specific inhibitor of the ATPase activity of the motor domain of Eg5 inhibiting the Eg5 microtubule motility in vitro and in cellulo sparked an intense interest in academia and industry to pursue the identification of novel small molecules that target Eg5 in order to be used in cancer chemotherapy based on the anti-mitotic strategy. Several Eg5 inhibitors entered clinical trials. Currently the field is faced with the problem that most of the inhibitors tested exhibited only limited efficacy. However, one Eg5 inhibitor, Arry-520 (clinical name filanesib), has demonstrated clinical efficacy in patients with multiple myeloma and is scheduled to enter phase III clinical trials. At the same time, new trends in Eg5 inhibitor research are emerging, including an increased interest in novel inhibitor binding sites and a focus on drug synergy with established antitumor agents to improve chemotherapeutic efficacy. This review presents an updated view of the structure and function of Eg5-inhibitor complexes, traces the possible development of resistance to Eg5 inhibitors and their potential therapeutic applications, and surveys the current challenges and future directions of this active field in drug discovery.


Subject(s)
Antimitotic Agents/pharmacology , Antineoplastic Agents/pharmacology , Kinesins/antagonists & inhibitors , Kinesins/metabolism , Adenosine Triphosphate/metabolism , Animals , Antimitotic Agents/chemistry , Antimitotic Agents/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Binding Sites , Biological Products/chemistry , Biological Products/pharmacology , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Humans , Kinesins/chemistry , Molecular Targeted Therapy/methods
4.
Eur J Pharm Biopharm ; 157: 183-190, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33222770

ABSTRACT

Cancer drugs which are specifically targeted at mitosis have generally under-delivered as a class. One likely reason is that only a small percentage of cancer cells in a tumor are actually dividing at any moment. If this is the case, then prolonged bioavailability in the tumor should significantly increase the efficacy of antimitotic agents. Here, we show that if the Plk1 inhibitor BI 2536 is co-encapsulated in a liposome with a pair of anions, its release rate is dependent on both the identity and stoichiometry of the anions. We created a library of liposomes with varying release rates using this approach and found that liposomal drug release rates correlated inversely with in vitro cancer cell killing. Xenografted mice treated with a single dose of slow-releasing liposomal BI 2536 experienced tumor volume decreases lasting 12 days and complete responses in 20% of mice. Treatment with two doses a week apart increased the response rate to 75%. This approach, which we termed Paired Anion Calibrated Release (PACeR), has the potential to revive the clinical utility of antimitotic cancer drugs which have failed clinical trials.


Subject(s)
Antimitotic Agents/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Lipids/chemistry , Mitosis/drug effects , Pteridines/pharmacology , Animals , Antimitotic Agents/chemistry , Antimitotic Agents/pharmacokinetics , Colonic Neoplasms/pathology , Drug Compounding , Drug Liberation , Female , HCT116 Cells , Humans , Kinetics , Liposomes , Mice, Nude , Pteridines/chemistry , Pteridines/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
J Hematol Oncol ; 12(1): 9, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30642372

ABSTRACT

BACKGROUND: Chiauranib is a novel orally active multi-target inhibitor that simultaneously inhibits the angiogenesis-related kinases (VEGFR2, VEGFR1, VEGFR3, PDGFRα, and c-Kit), mitosis-related kinase Aurora B, and chronic inflammation-related kinase CSF-1R. This phase I dose-escalation study was to determine the maximum tolerated dose (MTD), safety, pharmacokinetics, and preliminary antitumor activity of chiauranib in patients with refractory advanced solid tumor and lymphoma. METHODS: Eighteen patients were treated with continuous dosing of chiauranib from 10 to 65 mg once daily in a dose-escalation 3 + 3 design and evaluated in 28-day cycles. Pharmacokinetic profile of plasma chiauranib was analyzed in both single and multiple dose studies. RESULTS: Dose-limiting toxicity (DLT) as of grade 3 hypertension occurred in two patients at 65 mg/day, and one dose level below as MTD was 50 mg/day. The most common treatment-related adverse events included fatigue (61.1%), proteinuria (44.4%), hematuria (38.9%), hypothyroidism (38.9%), hypertriglyceridemia (33.3%), and hypertension (33.3%). A linear and dose-dependent pharmacokinetic profile of chiauranib was characterized with rapid absorption and slow elimination feature in both single and multiple dose studies. The accumulative exposure of chiauranib reached the steady state within 8 days and was approximately increased by twofold as those in the single dose study. No complete or partial response was observed, and 12 patients (66.7%) achieved stable disease (SD). CONCLUSIONS: Chiauranib demonstrated an acceptable safety and favorable pharmacokinetic profile with potential antitumor activity. Several phase Ib/II clinical studies are currently under further investigation. TRIAL REGISTRATION: NCT, NCT02122809 . Registered 25 April 2014.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Anti-Inflammatory Agents/adverse effects , Antimitotic Agents/adverse effects , Antineoplastic Agents/adverse effects , Lymphoma, Non-Hodgkin/drug therapy , Naphthalenes/adverse effects , Protein Kinase Inhibitors/adverse effects , Quinolines/adverse effects , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacokinetics , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Antimitotic Agents/administration & dosage , Antimitotic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cohort Studies , Dose-Response Relationship, Drug , Fatigue/etiology , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Naphthalenes/administration & dosage , Naphthalenes/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Proteinuria/etiology , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Treatment Outcome , Young Adult
6.
J Med Chem ; 60(12): 4963-4982, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28535350

ABSTRACT

Prodrug-mediated utilization of the cytochrome P450 (CYP) 1A1 to obtain the selective release of potent anticancer products within cancer tissues is a promising approach in chemotherapy. We herein report the rationale, preparation, biological evaluation, and mechanism of action of phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) that are antimicrotubule prodrugs activated by CYP1A1. Although PAIB-SOs are inert in most cells tested, they are highly cytocidal toward several human breast cancer cells, including hormone-independent and chemoresistant types. PAIB-SOs are N-dealkylated into cytotoxic phenyl 4-(2-oxo-3-imidazolidin-1-yl)benzenesulfonates (PIB-SOs) in CYP1A1-positive cancer cells, both in vitro and in vivo. In conclusion, PAIB-SOs are novel chemotherapeutic prodrugs with no equivalent among current antineoplastics and whose selective action toward breast cancer is tailored to the characteristic pattern of CYP1A1 expression observed in a large percentage of human breast tumors.


Subject(s)
Antimitotic Agents/pharmacology , Benzenesulfonates/chemistry , Breast Neoplasms/drug therapy , Cytochrome P-450 CYP1A1/metabolism , Prodrugs/pharmacology , Animals , Antimitotic Agents/pharmacokinetics , Benzenesulfonates/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Chick Embryo , Cytochrome P-450 CYP1A1/genetics , Drug Screening Assays, Antitumor/methods , Female , Humans , Prodrugs/pharmacokinetics
7.
Br J Clin Pharmacol ; 75(2): 507-15, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22803519

ABSTRACT

AIM: Eribulin mesylate is a non-taxane microtubule dynamics inhibitor that was recently approved for treatment of metastatic breast cancer. The aim of this study was to determine the effect of rifampicin, a CYP3A4 inducer, on the plasma pharmacokinetics of eribulin in patients with solid tumours. METHODS: An open-label, non-randomized phase I study was carried out. Patients received intravenous 1.4 mg m(-2) eribulin mesylate on days 1 and 15 and oral rifampicin 600 mg on days 9 to 20 of a 28 day cycle. Pharmacokinetic sampling for determination of eribulin plasma concentrations was performed up to 144 h following administration. AUC(0,∞) and C(max) for eribulin exposure without or with co-administration of rifampicin were subjected to an analysis of variance (anova) and corresponding 90% confidence intervals (CI) were calculated. Subsequently, patients were allowed to continue eribulin mesylate treatment with 1.4 mg m(-2) eribulin mesylate on days 1 and 8 of a 21 day cycle. Also the adverse event profile and anti-tumour activity were assessed. RESULTS: Fourteen patients were included and 11 patients were evaluable for pharmacokinetic analysis. Co-administration of rifampicin had no effect on single dose exposure to eribulin (geometric least square means ratio: AUC(0,∞) = 1.10, 90% CI 0.91, 1.34 and C(max) = 0.97, 90% 0.81, 1.17). The most common treatment-related grade ≥3 adverse events were grade 3 neutropenia (4/14, 29%), leucopenia and fatigue (both 3/14, 21%). CONCLUSIONS: These results indicate that eribulin mesylate may be safely co-administered with compounds that are CYP3A4 inducers.


Subject(s)
Antimitotic Agents/pharmacokinetics , Enzyme Inhibitors/administration & dosage , Furans/pharmacokinetics , Ketones/pharmacokinetics , Neoplasms/metabolism , Rifampin/administration & dosage , Administration, Oral , Adult , Aged , Antimitotic Agents/administration & dosage , Area Under Curve , Asian People , Drug Interactions , Female , Furans/administration & dosage , Humans , Ketones/administration & dosage , Male , Microtubules/drug effects , Microtubules/metabolism , Middle Aged , White People
8.
Invest New Drugs ; 30(3): 1107-15, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21494838

ABSTRACT

Eg5 (kinesin spindle protein) is a microtubule motor protein, essential for centrosome separation during mitosis. This Phase I/II, open-label, multicenter, two-part study investigated AZD4877, a potent Eg5 inhibitor, in patients with acute myeloid leukemia. Primary objectives were to determine the maximum tolerated dose (MTD) (part A), assess efficacy (part B) and determine the pharmacokinetic profile (parts A and B). Secondary objectives included assessment of safety and tolerability. AZD4877 was administered at a range of doses (2, 4, 7, 10, 13, 16 and 18 mg/day) as a 1-hour intravenous infusion on three consecutive days of a continuous 2-week schedule. The MTD in part A was defined as 16 mg/day based on dose-limiting stomatitis at 16 and 18 mg/day, hyperbilirubinemia at 16 mg/day and palmar-plantar erythrodysesthesia syndrome at 18 mg/day. Systemic exposure to AZD4877 generally increased with increasing dose whereas half-life was not dose dependent. No evaluable patients experienced a complete remission (CR) or CR with incomplete blood count recovery (CRi), demonstrating no evidence of AZD4877 efficacy in this population. Evidence of monoasters in all but the 4 mg/day dose group provided proof of mechanism for AZD4877. This study was terminated due to lack of efficacy. (ClinicalTrials.gov identifier NCT00486265).


Subject(s)
Antimitotic Agents/administration & dosage , Benzamides/administration & dosage , Kinesins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Pyrimidinones/administration & dosage , Adult , Aged , Aged, 80 and over , Antimitotic Agents/adverse effects , Antimitotic Agents/pharmacokinetics , Benzamides/adverse effects , Benzamides/pharmacokinetics , Female , Humans , Leukemia, Myeloid, Acute/blood , Male , Middle Aged , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , Young Adult
9.
Cancer Chemother Pharmacol ; 69(3): 733-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22020315

ABSTRACT

PURPOSE: GSK923295 is an inhibitor of CENP-E, a key cellular protein important in the alignment of chromosomes during mitosis. This was a Phase I, open-label, first-time-in-human, dose-escalation study, to determine the maximum-tolerated dose (MTD), safety, and pharmacokinetics of GSK923295. PATIENTS AND METHODS: Adult patients with previously treated solid tumors were enrolled in successive cohorts at GSK923295 doses ranging from 10 to 250 mg/m(2). GSK923295 was administered by a 1-h intravenous infusion, once weekly for three consecutive weeks, with treatment cycles repeated every 4 weeks. RESULTS: A total of 39 patients were enrolled. The MTD for GSK923295 was determined to be 190 mg/m(2). Observed dose-limiting toxicities (all grade 3) were as follows: fatigue (n = 2, 5%), increased AST (n = 1, 2.5%), hypokalemia (n = 1, 2.5%), and hypoxia (n = 1, 2.5%). Across all doses, fatigue was the most commonly reported drug-related adverse event (n = 13; 33%). Gastrointestinal toxicities of diarrhea (n = 12, 31%), nausea (n = 8, 21%), and vomiting (n = 7, 18%) were generally mild. Frequency of neutropenia was low (13%). There were two reports of neuropathy and no reports of mucositis or alopecia. GSK923295 exhibited dose-proportional pharmacokinetics from 10 to 250 mg/m(2) and did not accumulate upon weekly administration. The mean terminal elimination half-life of GSK923295 was 9-11 h. One patient with urothelial carcinoma experienced a durable partial response at the 250 mg/m(2) dose level. CONCLUSIONS: The novel CENP-E inhibitor, GSK923295, had dose-proportional pharmacokinetics and a low number of grade 3 or 4 adverse events. The observed incidence of myelosuppression and neuropathy was low. Further investigations may provide a more complete understanding of the potential for GSK923295 as an antiproliferative agent.


Subject(s)
Antimitotic Agents/administration & dosage , Antimitotic Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Neoplasms/drug therapy , Sarcosine/analogs & derivatives , Adult , Aged , Antimitotic Agents/adverse effects , Antimitotic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Sarcosine/administration & dosage , Sarcosine/adverse effects , Sarcosine/pharmacokinetics , Sarcosine/therapeutic use , Treatment Outcome , Young Adult
10.
Clin Exp Pharmacol Physiol ; 37(5-6): 636-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20132238

ABSTRACT

1. It has been shown that tubulin-binding agents can destabilize cellular microtubules and suppress tumour growth; but it has also become apparent that some compounds can exert anti-vascular effects within the neovasculature of a solid tumour. To date, the difficulty with these targets has been the ability to selectivity induce vascular damage to the tumour while leaving normal vasculature unaffected. The data presented here characterizes the in vivo, tumour selective, anti-vascular effects of the novel tubulin-binding agent A-318315. 2. To that purpose, we have used an anaesthetized in vivo rat model designed to quantify acute changes in regional vascular resistance (VR) in both tumour and non-tumour vascular beds, simultaneously. Tissue-isolated tumours (approximately 1.25 gm) with blood flow supplied by a single epigastric artery were grown in the hindlimb of adult male rats. Blood flow to the tumour, mesenteric, renal and normal (non-tumour epigastric) arteries was measured pre-dose and post-dose under anaesthesia. 3. A-318315 was tested at 3, 10 and 30 mg/kg, i.v. These doses produced modest, transient increases in mean arterial pressure with little to no effect on heart rate. At peak effect, tumour VR increased to 175 +/- 47, 337 +/- 77 and 751 +/- 151% above the baseline, for the 3, 10 and 30 mg/kg doses, respectively, whereas VR was only modestly and transiently increased in normal epigastric (88 +/- 19%), mesenteric (33 +/- 3.3%) and renal arteries (17 +/- 8.6%). 4. These data demonstrate that A-318315 produces marked reductions in tumour blood flow in the rat at doses that exert minor effects on normal vascular function.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antimitotic Agents/therapeutic use , Hemodynamics/drug effects , Indoles/therapeutic use , Neovascularization, Pathologic/drug therapy , Sulfonamides/therapeutic use , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Antimitotic Agents/adverse effects , Antimitotic Agents/pharmacokinetics , Antimitotic Agents/pharmacology , Blood Pressure/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Heart Rate/drug effects , Indoles/adverse effects , Indoles/pharmacokinetics , Indoles/pharmacology , Male , Molecular Structure , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Rats , Rats, Inbred F344 , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Tubulin/metabolism , Vascular Resistance/drug effects
11.
J Control Release ; 143(1): 2-12, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-19925835

ABSTRACT

Taxanes are potent antimitotic agents that have demonstrated efficacy in a wide range of malignancies. Due to their poor water-solubility, these cytostatic drugs were first formulated with low molecular weight surfactants, e.g. Cremophor EL (CrEL) and Tween 80 (polysorbate 80), which are known to exhibit serious adverse effects in humans. In recent years, there has been growing interest in the design of more biocompatible formulations for both paclitaxel and docetaxel. Polymer-based drug carriers represent an attractive venue given the diversity in the array of existing polymers. Most notably, biopolyesters are vastly employed in the field of biomedical research given their biocompatibility and biodegradability. Polyester-based micelles and nanoparticles have been applied to the parenteral delivery of taxanes with varying degrees of success. Block copolymer micelles possess a unique core-shell structure generated through the self-assembly of amphiphilic copolymers in aqueous media. Although these systems have shown greatly enhanced tolerability compared to formulations based on low molecular weight surfactants, in some cases their failure to retain their cargo following parenteral administration has hindered their capacity to target taxanes to solid tumours. While polyester-based nanoparticles possess comparatively greater stability and drug targeting capacity, they frequently display a significant burst effect whereby a major portion of the cargo is immediately discarded from the carrier upon injection. This review focuses on the current application of polyester-based micelles and nanoparticles to the tumour targeting of taxanes. The preparation, loading efficiencies, release kinetics, cytotoxicity and in vivo behaviour of these systems is discussed in detail.


Subject(s)
Antimitotic Agents/administration & dosage , Drug Carriers , Micelles , Nanoparticles , Neoplasms/drug therapy , Polyesters/chemistry , Taxoids/administration & dosage , Animals , Antimitotic Agents/chemistry , Antimitotic Agents/pharmacokinetics , Biological Availability , Chemistry, Pharmaceutical , Drug Compounding , Drug Stability , Humans , Infusions, Parenteral , Models, Molecular , Molecular Structure , Neoplasms/metabolism , Polyesters/toxicity , Solubility , Taxoids/chemistry , Taxoids/pharmacokinetics , Technology, Pharmaceutical/methods , Tissue Distribution
12.
Methods Find Exp Clin Pharmacol ; 31(7): 443-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19907719

ABSTRACT

Antimitotic agents are among the most effective drugs for the treatment of solid tumors and metastatic cancer. These drugs promote cell death by interfering with the crucial structural and regulatory function of microtubules in cells. Most of the agents of clinical relevance are natural products or semisynthetic derivatives thereof, and they fall into two major classes: microtubule stabilizers such as the taxanes, which enhance tubulin polymerization, and microtubule destabilizers such as the Vinca alkaloids, which lead to the depolymerization of existing microtubules. While these drugs are effective in inhibiting the progression of certain types of tumors, their utility is limited in part by incomplete tumor responses and/or significant side effects. In addition, inherent resistance is encountered in many tumor types, or acquired resistance may occur as a result of multiple cycles of therapy. Cevipabulin (TTI-237) is a novel, small synthetic molecule with an unusual biological mode of action. It appears to bind at the vinca site, but exhibits some properties similar to those of taxane-site ligands, such as enhancing tubulin polymerization. The compound works against a variety of tumors, including those resistant to paclitaxel and vincristine. Furthermore, cevipabulin is stable and water-soluble, and can be administered i.v. or p.o. in saline. It can be synthesized in bulk quantities efficiently. Based on these properties, cevipabulin was selected for clinical development.


Subject(s)
Antimitotic Agents/therapeutic use , Hydrocarbons, Halogenated/therapeutic use , Microtubules/drug effects , Triazoles/therapeutic use , Animals , Antimitotic Agents/adverse effects , Antimitotic Agents/metabolism , Antimitotic Agents/pharmacokinetics , Antimitotic Agents/pharmacology , Clinical Trials as Topic , Clinical Trials, Phase I as Topic , Drug Evaluation, Preclinical , Humans , Hydrocarbons, Halogenated/adverse effects , Hydrocarbons, Halogenated/metabolism , Hydrocarbons, Halogenated/pharmacokinetics , Hydrocarbons, Halogenated/pharmacology , Mice , Neoplasms/drug therapy , Rats , Triazoles/adverse effects , Triazoles/metabolism , Triazoles/pharmacokinetics , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL