Size | Price | Stock | Qty |
---|---|---|---|
100mg |
|
||
250mg |
|
||
500mg |
|
||
1g |
|
||
Other Sizes |
|
Ceritinib diHCl (LDK-378; LDK378; Zykadia; Spexib), the dihydrochloride salt of ceritinib, is an ALK (anaplastic lymphoma kinase positive) inhibitor approved in 2014 for the treatment of non-small cell lung cancer (NSCLC).
Targets |
ALK (IC50 = 0.2 nM); IGF-1R (IC50 = 8 nM); InsR (IC50 = 7 nM); STK22D (IC50 = 23 nM)
|
|
---|---|---|
ln Vitro |
|
|
ln Vivo |
|
|
Enzyme Assay |
All kinases are expressed using the baculovirus expression technology as either GST- or histidine-tagged fusion proteins, with the exception of untagged ERK2, which is made in E. coli. Using the LabChip mobility shift assay, the kinase activity is determined. For sixty minutes, the assay is run at 30°C. It is routinely possible to determine the effect of LDK378 on the enzymatic activity from a single reading (end point measurement) by analyzing the linear progress curves in both the presence and absence of LDK378.
Enzymatic Kinase Profiling Description[1] All kinases were expressed as either histidine- or GST-tagged fusion proteins using the baculovirus expression technology except for the untagged ERK2 which was produced in E. coli. AURORA-A, JAK2, MK2, SYK, ERK2 and PKA were purchased commercially, and all other kinases were supplied in-house. The kinase activity was measured in the LabChip mobility-shift assay. The assay was performed at 30 °C for 60 min. The effect of compound on the enzymatic activity was obtained from the linear progress curves in the absence and presence of compound and routinely determined from one reading (end point measurement). GSH-Trapping Assay[1] For the characterization of the metabolic activation, 10 mmol/L DMSO stock solutions of the compounds were incubated at 37 °C for up to 60 min with human liver microsomes (50 μL), containing 1 mg protein/mL with phosphate buffer. Four microliters of 0.5 mM of 20 μL of DMSO stock solution of compound in 180 μL of a mixture of acetonitrile/water (ratio 1:1) was added to 50 μL of 1 mg/mL liver microsomes in phosphate buffer and preincubated for 3 min at 37 °C. After preincubation, the reaction was started by addition of 50 μL of the NADPH (1 mmol/L), UDPGA (1 mmol/L), MgCl (2 mmol/L), and ethyl ester GSH reduced (2 mmol/L). After 60 min, the reaction was stopped with 200 μL of ice-cold acetonitrile. The reaction mixture was stored at −20 °C. The mixture was centrifuged (10000g, 5 min), and 250 μL of 300 μL supernatant was transferred. From this solution, 20 μL aliquots were used for analysis. The liquid chromatographic separation was performed using an Agilent HP1100 pump and a Phenyl HexylRP column, 150 mm × 2.0 mm, particle size 4.6 μm. Gradient mobile phase programming was used with a flow rate of 350 μL/min. Eluent A was Milli-Q water with 0.1% formic acid. Eluent B was acetonitrile with 0.1% formic acid. The mobile phase was a linear gradient from 5% B to 95% B over 6 min and held for 2 min at 95% B for a total run time of 10 min. The column effluent was introduced directly into the ion source of a triple quadrupole MS instrument or ion trap MS instrument. The ionization technique employed was positive electrospray (ES). The TS-Quantum was used in a product ion scan mode, utilizing collision induced dissociation in Q2 (collision chamber). Collision gas was argon. The collision energy was set at 30 eV. Solubility Assays[1] One-hundred microliter aliquots of 1 mM DMSO solutions were added to each of three glass vials and evaporated to dryness prior to addition of 500 μL of pH 6.8 buffer. Following 24 h of shaking, the solutions were vacuum filtered through MultiScreen Solubility 96-well plates with 0.4 μm modified PCTE membrane, and an aliquot of each filtrate is transferred to a UV plate for quantification as described in Uvarova et al. Metabolic Clearance Assays[1] The metabolic clearance assays were conducted using the method described in Richmond et al. CYP Inhibition Assays[1] The samples were prepared as 10 mM solutions in DMSO, then assayed and analyzed using the general LC–MS/MS method described by Bell et al. |
|
Cell Assay |
LDK378 or DMSO are serially diluted and incubated with luciferase-expressing cells for two to three days. Using the Bright-Glo Luciferase Assay System, luciferase expression is measured as a proxy for cell proliferation and survival. The XLFit program is used to generate IC50 values.
GI50 determination[3] In order to calculate the half maximal growth inhibitory concentration (GI50) of individual compounds [Ceritinib (LDK-378)], neuroblastoma tumor cells were seeded into 96‐well plates in a total volume of 100 μL and allowed to attach overnight. Compound (e.g. Ceritinib (LDK-378) dissolved in DMSO) was added to wells in six replicates of 100 μL, across a concentration gradient including a DMSO‐only control, the next day. The cells were exposed to drug for 72 h. Thereafter, the cell number in treated versus control wells was estimated after cell fixation with 10% trichloroacetic acid and staining with sulforhodamine B in 1% acetic acid. The GI50 was calculated as the drug concentration that inhibits cell growth by 50% compared with control growth, according to nonlinear regression analysis, using graphpad prism. Preparation of protein lysates[3] Cell lines were harvesting by scraping, spun at 500 g for 5 min, and washed once in phosphate‐buffered saline, and the cell pellets were resuspended in CHAPS lysis buffer [50 mm Tris/HCl pH 8.0, 1 mm EDTA, 150 mm NaCl, 1% CHAPS, 0.2 mm PMSF, 1 : 50 Phosphatase Inhibitor Cocktail 2 and 3, 1 : 100 Protease Inhibitor Cocktail. Frozen tissue samples were homogenized in CHAPS lysis buffer prepared as for cell lysates. After incubation for 30 min on ice, lysates were spun at 16 000 g for 15 min and the supernatant was collected. Protein concentrations were determined using BCA protein assay by comparison with bovine serine albumin standard. |
|
Animal Protocol |
RNU nude rats bearing the Karpas299/H2228 tumors
~50 mg/kg o.g. Studies on in vivo PK are carried out on dogs, rats, mice, and cynomolgus monkeys. Male Balb/c mice are given cetinib (LDK378) (HCl salt) orally via gavage at a dose of 20 mg/kg (n=3) and intravenously via tail vein at a dose of 5 mg/kg (n= 3). Sprague-Dawley rats are dosed with Ceritinib (LDK378) (HCl salt) intravenously via the tail vein at 3 mg/kg (n=3) and orally via gavage at 10 mg/kg (n=3) using the same formulation. Serial blood samples are taken over the course of 24 hours following dosage at prearranged times. Ceritinib (phosphate salt) is given as a single intravenous (n = 2) or oral (n = 3) dose to male beagle dogs. The intravenous solution has a dosage of 5 mg/kg, while the oral suspension has a dosage of 20 mg/kg. A single intravenous (n = 2) or oral (n = 3) dose of Ceritinib (free base) is given to male Cynomologus monkeys. The intravenous solution has a dose of 5 mg/kg, while the oral suspension has a dose of 60 mg/kg. For plasma, blood is drawn at prearranged intervals over a period of 144 hours following dosage. PK Studies[1] In vivo PK studies were conducted in mice, rats, dogs, and cynomolgus monkeys. 15b (HCl salt) was formulated as a solution in 75% PEG300/25% D5W and administered to male Balb/c mice intravenously via tail vein at 5 mg/kg (n = 3) and orally via gavage at 20 mg/kg (n = 3). By use of the same formulation, 15b (HCl salt) was dosed to Sprague–Dawley rats intravenously via the tail vein at 3 mg/kg (n = 3) and orally via gavage at 10 mg/kg (n = 3). Blood samples were collected serially at scheduled times over 24 h after dosing. Male beagle dogs received a single intravenous (n = 2) or oral (n = 3) dose of 15b (phosphate salt) as an intravenous solution in 30% propylene glycol/5% Solutol buffer at 5 mg/kg and an oral suspension in suspension in 0.5% (w/v) aqueous methylcellulose/0.5% Tween 80 at 20 mg/kg, respectively. Male cynomologus monkeys received single intravenous (n = 2) or oral (n = 3) dose of 15b (free base) as an intravenous solution in 30% propylene glycol/5% solutol at 5 mg/kg and an oral suspension in 0.5% (w/v) methylcellulose at 60 mg/kg, respectively. Blood samples for plasma were collected at prescheduled times over 144 h after dosing. In Vivo Experiments[1] RNU nude rats bearing the Karpas299 tumors were randomized into five groups (n = 6 rats per group) with an average tumor size of 326 ± 128 mm3. 15b (phosphate salt) was formulated in 0.5% MC/0.5% Tween 80 and administered by oral gavage at a dosing volume of 10 μL/g of an animal body weight. Animals in each group received vehicle or 6.25, 12.5, 25, 50 mg/kg 15b every day for 14 consecutive days. RNU nude rats bearing the H2228 tumors were randomized into four groups (n = 4 rats per group) with an average tumor volume of 371 ± 139 mm3. 15b (phosphate salt) was formulated in 0.5% MC/0.5% Tween 80 and administered by oral gavage at a dosing volume of 10 μL/g of an animal body weight. Animals in each group received vehicle or 5, 10, 25 mg/kg 15b (phosphate salt) every day for 14 consecutive days. RNU nude rats bearing the Karpas299 tumors were dosed with 15b (phosphate salt) at 50 mg/kg. Tumor and plasma samples were collected 7, 24, 48, and 72 h after dosing. Two tumor pieces were collected from each animal, one piece for protein extraction and the other for PK analysis. Proteins were extracted from tumor samples and then subjected to SDS–PAGE followed by Western blot with phospho-STAT3 antibody (pSTAT3, Tyr705). HOMA-IR[1] Homeostatic model assessment (HOMA) of insulin resistance (IR) is a technical method for assessing IR from basal (fasting) glucose and insulin or C-peptide concentrations. The model is widely used to estimate insulin resistance. Groups of wild-type mice (n = 8 mice per group) were randomized into treatment groups based on their initial body weight. Mice were housed four per cage and dosed with vehicle orally, or ALK inhibitor (15b or Ceritinib (LDK-378)), orally once per day for 7 days. On day 7, compound was administered 180 min prior to a 3 g/kg glucose bolus. OGTT evaluations were performed in conscious mice that were 11 weeks of age. The mice were fasted by removing food at 6 p.m. the day before. A baseline blood sample was taken at t = −180 min, and the mice were then dosed orally with the compounds. A baseline blood sample was taken at t = 0 min, and the animals were then administered an oral glucose bolus (3 g/kg) immediately. Blood was obtained (via tail bleeding) to measure blood glucose (using glucometer). A single drop of blood from the tail was measured for glucose using a glucometer at t = −180, 0, 20, 40, 60, 120 min. Approximately 40 μL samples of blood were collected separately for insulin analysis 3 h prior to dosing (on day 0 and day 7) into chilled collection tubes containing EDTA. Plasma was isolated and stored at −70 °C until further analysis. The homeostatic model assessment-insulin resistance index (HOMA-IR) was used as a measure of insulin resistance and was calculated using the formula HOMA-IR = (FPG × FPI)/22.5 where FPG (mM) is the fasting plasma glucose concentration and FPI (μU/mL) is the fasting plasma insulin concentration. Insulin levels were determined using a detection assay kit from Mesoscale Discovery (MSD): catalog no. K112BZC-2. Higher values indicate insulin resistance. |
|
ADME/Pharmacokinetics |
Absorption, Distribution and Excretion
After oral administration of ceritinib, peak concentrations were achieved after approximately 4 to 6 hours. Following oral administration of a single 750 mg radiolabeled ceritinib dose, 92.3% of the administered dose was recovered in the feces (with 68% as unchanged parent compound) while 1.3% of the administered dose was recovered in the urine. The apparent volume of distribution (Vd/F) is 4230 L following a single 750 mg dose. The geometric mean apparent clearance (CL/F) of ceritinib was lower at steady-state (33.2 L/h) after 750 mg daily dosing than after a single 750 mg dose (88.5 L/h). Metabolism / Metabolites In vitro studies demonstrated that CYP3A was the major enzyme involved in the metabolic clearance of ceritinib. Following oral administration of a single 750 mg radiolabeled ceritinib dose, ceritinib as the parent compound was the main circulating component (82%) in human plasma. Biological Half-Life The terminal half life is 41 hours. |
|
Toxicity/Toxicokinetics |
Hepatotoxicity
Elevations in serum aminotransferase levels are common during ceritinib therapy occurring in 20% to 50% of patients, but rising above 5 times the upper limit of the normal range in only 1% to 2%. Hepatic failure is said to have occurred in 0.2% of patients and to have resulted in several fatalities. Hepatotoxicity appears to be a class effect among ALK inhibitors, although liver injury appears to be more frequent and more severe with crizotinib than ceritinib or alectinib. Specific details of the liver injury associated with ceritinib such as latency, serum enzyme pattern, clinical features and course, have not been published. Other ALK inhibitors typically cause liver injury arising within days or weeks of starting therapy, and presenting abruptly with hepatocellular enzyme elevations and a moderate-to-severe course. Immunoallergic and autoimmune features are not common. The rate of clinically significant liver injury and hepatic failure is increased in patients with preexisting cirrhosis or hepatic impairment due to liver tumor burden. Recurrence upon reexposure has been reported. Likelihood score: D (possible cause of clinically apparent liver injury). Effects During Pregnancy and Lactation ◉ Summary of Use during Lactation No information is available on the clinical use of ceritinib during breastfeeding. Because ceritinib is 97% bound to plasma proteins, the amount in milk is likely to be low. The manufacturer recommends that breastfeeding be discontinued during ceritinib therapy and for 2 weeks after the final dose. ◉ Effects in Breastfed Infants Relevant published information was not found as of the revision date. ◉ Effects on Lactation and Breastmilk Relevant published information was not found as of the revision date. Protein Binding Ceritinib is 97% bound to human plasma proteins, independent of drug concentration. |
|
References |
|
|
Additional Infomation |
Ceritinib is a member of the class of aminopyrimidines that is 2,6-diamino-5-chloropyrimidine in which the amino groups at positions 2 and 6 are respectively carrying 2-methoxy-4-(piperidin-4-yl)-5-methylphenyl and 2-(isopropylsulfonyl)phenyl substituents. Used for the treatment of ALK-positive metastatic non-small cell lung cancer. It has a role as an antineoplastic agent and an EC 2.7.10.1 (receptor protein-tyrosine kinase) inhibitor. It is an aminopyrimidine, an aromatic ether, an organochlorine compound, a secondary amino compound, a member of piperidines and a sulfone.
Ceritinib is used for the treatment of adults with anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer (NSCLC) following failure (secondary to resistance or intolerance) of prior crizotinib therapy. About 4% of patients with NSCLC have a chromosomal rearrangement that generates a fusion gene between EML4 (echinoderm microtubule-associated protein-like 4) and ALK (anaplastic lymphoma kinase), which results in constitutive kinase activity that contributes to carcinogenesis and seems to drive the malignant phenotype. Ceritinib exerts its therapeutic effect by inhibiting autophosphorylation of ALK, ALK-mediated phosphorylation of the downstream signaling protein STAT3, and proliferation of ALK-dependent cancer cells. Following treatment with crizotinib (a first-generation ALK inhibitor), most tumours develop drug resistance due to mutations in key "gatekeeper" residues of the enzyme. This occurrence led to development of novel second-generation ALK inhibitors such as ceritinib to overcome crizotinib resistance. The FDA approved ceritinib in April 2014 due to a surprisingly high response rate (56%) towards crizotinib-resistant tumours and has designated it with orphan drug status. Ceritinib is a Kinase Inhibitor. The mechanism of action of ceritinib is as a Tyrosine Kinase Inhibitor, and Cytochrome P450 3A Inhibitor, and Cytochrome P450 2C9 Inhibitor. Ceritinib is a small molecule tyrosine kinase receptor inhibitor and antineoplastic agent that is used in the therapy of selected forms of advanced non-small cell lung cancer (NSCLC). Ceritinib is associated with a moderate rate of serum aminotransferase elevations during therapy and rare instances of clinically apparent acute liver injury. Ceritinib is an orally available inhibitor of the receptor tyrosine kinase activity of anaplastic lymphoma kinase (ALK) with antineoplastic activity. Upon administration, ceritinib binds to and inhibits wild-type ALK kinase, ALK fusion proteins and ALK point mutation variants. Inhibition of ALK leads to both the disruption of ALK-mediated signaling and the inhibition of cell growth in ALK-overexpressing tumor cells. ALK belongs to the insulin receptor superfamily and plays an important role in nervous system development. ALK dysregulation and gene rearrangements are associated with a variety of tumor cell types. Drug Indication Ceritinib is a kinase inhibitor indicated for the treatment of patients with anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer (NSCLC) who have progressed on or are intolerant to crizotinib. This indication is approved under accelerated approval based on tumor response rate and duration of response. An improvement in survival or disease-related symptoms has not been established. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials. FDA Label Zykadia is indicated for the treatment of adult patients with anaplastic lymphoma kinase (ALK) positive advanced non small cell lung cancer (NSCLC) previously treated with crizotinib. Mechanism of Action Ceritinib inhibits Anaplastic lymphoma kinase (ALK) also known as ALK tyrosine kinase receptor or CD246 (cluster of differentiation 246), which is an enzyme that in humans is encoded by the ALK gene. About 4-5% of NSCLCs have a chromosomal rearrangement that generates a fusion gene between EML4 (echinoderm microtubule-associated protein-like 4) and ALK (anaplastic lymphoma kinase), which results in constitutive kinase activity that contributes to carcinogenesis and seems to drive the malignant phenotype. Ceritinib exerts its therapeutic effect by inhibiting autophosphorylation of ALK, ALK-mediated phosphorylation of the downstream signaling protein STAT3, and proliferation of ALK-dependent cancer cells. Ceritinib has been shown to inhibit in vitro proliferation of cell lines expressing EML4-ALK and NPM-ALK fusion proteins and demonstrated dose-dependent inhibition of EML4-ALK-positive NSCLC xenograft growth in mice and rats. |
Molecular Formula |
C28H38CL3N5O3S
|
---|---|
Molecular Weight |
631.0570
|
Exact Mass |
629.176
|
Elemental Analysis |
C, 53.29; H, 6.07; Cl, 16.85; N, 11.10; O, 7.61; S, 5.08
|
CAS # |
1380575-43-8
|
Related CAS # |
Ceritinib;1032900-25-6;Ceritinib-d7;1632484-77-5
|
PubChem CID |
67973512
|
Appearance |
Light yellow to yellow solid powder
|
LogP |
8.87
|
Hydrogen Bond Donor Count |
5
|
Hydrogen Bond Acceptor Count |
8
|
Rotatable Bond Count |
9
|
Heavy Atom Count |
40
|
Complexity |
835
|
Defined Atom Stereocenter Count |
0
|
SMILES |
ClC1=C([H])N=C(N=C1N([H])C1=C([H])C([H])=C([H])C([H])=C1S(C([H])(C([H])([H])[H])C([H])([H])[H])(=O)=O)N([H])C1C([H])=C(C([H])([H])[H])C(=C([H])C=1OC([H])(C([H])([H])[H])C([H])([H])[H])C1([H])C([H])([H])C([H])([H])N([H])C([H])([H])C1([H])[H].Cl[H].Cl[H]
|
InChi Key |
WNCJOPLFICTLPT-UHFFFAOYSA-N
|
InChi Code |
InChI=1S/C28H36ClN5O3S.2ClH/c1-17(2)37-25-15-21(20-10-12-30-13-11-20)19(5)14-24(25)33-28-31-16-22(29)27(34-28)32-23-8-6-7-9-26(23)38(35,36)18(3)4;;/h6-9,14-18,20,30H,10-13H2,1-5H3,(H2,31,32,33,34);2*1H
|
Chemical Name |
5-chloro-2-N-(5-methyl-4-piperidin-4-yl-2-propan-2-yloxyphenyl)-4-N-(2-propan-2-ylsulfonylphenyl)pyrimidine-2,4-diamine;dihydrochloride
|
Synonyms |
Ceritinib dihydrochloride; LDK-378; Ceritinib HCl; 1380575-43-8; Ceritinib dihydrochloride; LDK378 dihydrochloride; LDK378 (dihydrochloride); Ceritinib 2hcl; LDK378 2HCl (Ceritinib); LDK-378 hydrochloride; 2,4-Pyrimidinediamine, 5-chloro-N4-[2-[(1-methylethyl)sulfonyl]phenyl]-N2-[5-methyl-2-(1-methylethoxy)-4-(4-piperidinyl)phenyl]-, hydrochloride (1:2); LDK 378; LDK378
|
HS Tariff Code |
2934.99.03.00
|
Storage |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month Note: Please store this product in a sealed and protected environment, avoid exposure to moisture. |
Shipping Condition |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
Solubility (In Vitro) |
DMSO: 13~100 mg/mL (20.60~158.5 mM)
Ethanol: ~10 mg/mL(~15.9 mM) |
---|---|
Solubility (In Vivo) |
Solubility in Formulation 1: 2.5 mg/mL (3.96 mM) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (add these co-solvents sequentially from left to right, and one by one), clear solution; with sonication.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 400 μL PEG300 and mix evenly; then add 50 μL Tween-80 to the above solution and mix evenly; then add 450 μL normal saline to adjust the volume to 1 mL. Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution. Solubility in Formulation 2: ≥ 2.5 mg/mL (3.96 mM) (saturation unknown) in 10% DMSO + 90% (20% SBE-β-CD in Saline) (add these co-solvents sequentially from left to right, and one by one), clear solution. For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 900 μL of 20% SBE-β-CD physiological saline solution and mix evenly. Preparation of 20% SBE-β-CD in Saline (4°C,1 week): Dissolve 2 g SBE-β-CD in 10 mL saline to obtain a clear solution. View More
Solubility in Formulation 3: ≥ 2.5 mg/mL (3.96 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution. Solubility in Formulation 4: 33.33 mg/mL (52.82 mM) in PBS (add these co-solvents sequentially from left to right, and one by one), clear solution; with ultrasonication. |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 1.5846 mL | 7.9232 mL | 15.8464 mL | |
5 mM | 0.3169 mL | 1.5846 mL | 3.1693 mL | |
10 mM | 0.1585 mL | 0.7923 mL | 1.5846 mL |
*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.
Calculation results
Working concentration: mg/mL;
Method for preparing DMSO stock solution: mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.
Method for preparing in vivo formulation::Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.
(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
(2) Be sure to add the solvent(s) in order.
NCT Number | Recruitment | interventions | Conditions | Sponsor/Collaborators | Start Date | Phases |
NCT03501368 | Active Recruiting |
Drug: Ceritinib Drug: Trametinib |
Melanoma Advanced Melanoma |
H. Lee Moffitt Cancer Center and Research Institute |
June 27, 2018 | Phase 1 |
NCT02321501 | Active Recruiting |
Drug: Ceritinib Drug: Everolimus |
ALK Positive ROS1 Gene Rearrangement |
M.D. Anderson Cancer Center | June 22, 2016 | Phase 1 |
NCT03611738 | Active Recruiting |
Drug: Ceritinib Drug: Docetaxel |
Lung Cancer | H. Lee Moffitt Cancer Center and Research Institute |
February 1, 2019 | Phase 1 |
NCT01828099 | Active Recruiting |
Drug: Ceritinib Drug: Pemetrexed |
Non-Small Cell Lung Cancer/td> | Novartis Pharmaceuticals | July 9, 2013 | Phase 3 |
NCT02584933 | Recruiting | Drug: ceritinib | ALK Positive Malignancies | Novartis Pharmaceuticals | December 11, 2015 | Phase 4 |