yingweiwo

Tosedostat (CHR-2797)

Alias: CHR2797; CHR-2797; CHR 2797; KZK563J2UW; C21H30N2O6; Tosedostat (CHR2797); Tosedostat.
Cat No.:V5082 Purity: ≥98%
Tosedostat (CHR2797) is a novel, orally bioavailable and potent inhibitor of the M1 family of aminopeptidases with potential antineoplastic activity.
Tosedostat (CHR-2797)
Tosedostat (CHR-2797) Chemical Structure CAS No.: 238750-77-1
Product category: Aminopeptidase
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
5mg
10mg
25mg
50mg
100mg
250mg
500mg
Other Sizes
Official Supplier of:
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text

 

  • Business Relationship with 5000+ Clients Globally
  • Major Universities, Research Institutions, Biotech & Pharma
  • Citations by Top Journals: Nature, Cell, Science, etc.
Top Publications Citing lnvivochem Products
Purity & Quality Control Documentation

Purity: ≥98%

Product Description

Tosedostat (CHR2797) is a novel, orally bioavailable and potent inhibitor of the M1 family of aminopeptidases with potential antineoplastic activity. Tosedostat is converted intracellularly into a poorly membrane-permeable active metabolite (CHR-79888) which inhibits the M1 family of aminopeptidases, particularly puromycin-sensitive aminopeptidase (PuSA), and leukotriene A4 (LTA4) hydrolase; inhibition of these aminopeptidases in tumor cells may result in amino acid deprivation, inhibition of protein synthesis due to a decrease in the intracellular free amino acid pool, an increase in the level of the proapoptotic protein Noxa, and cell death.

Biological Activity I Assay Protocols (From Reference)
Targets
Aminopeptidase
ln Vitro
The HL-60 cell growth media has more stanniocalcin 2 (STC2) protein when tosedostat (CHR-2797) is used. After two hours of treatment with Tosedostat (60 nM), SLC7A11 expression rose. U-937 and HuT 78 cell lines are blocked by tosedostat treatment, which also causes U-937 cells but not HuT 78 cells to express more of the incremental response (AADR) gene [1]. The mean MCA production of untreated control cells was lowered to 77.8% by tosedostat (0.01 μM; the IC50 for 24 hours was 10 nM and >10 μM, respectively); likewise, MCA production was decreased to 51.3% at 1 μM and 51.3% at 5 μM, and to 38.5% at μM and 35.3% at 10 μM [2].
ln Vivo
In animal cancer models that are pregnant, tosedostat (CHR-2797) is active and exhibited a dose-response relationship in both models; the effect of tosedostat is less pronounced when the tumor burden is at its highest prior to treatment [1].
Enzyme Assay
In Vitro Enzyme Assays [2]
LAP[2]
LAP activity was determined by measuring the hydrolysis of the tripeptide, LGG, which was detected by the derivatization reagent OPA in the presence of βmercaptoethanol. The enzyme was resuspended in H2O, then buffer exchanged on a PD-10 colum into 50mM HEPES, pH 8.0. Protein concentration was determined by BCA assay, and the concentration was adjusted to 10µg/ml. LGG was diluted to 0.5mM in 50mM HEPES. The assay was carried out in 96-well assay plates. Wells contained diluted inhibitor/vehicle (5µl), 10µg/ml LAP (5µl) and 40µl of 0.5mM LGG. Samples were assayed in triplicate. The plate was shaken briefly, and incubated for 90min at 37o C. The reaction was terminated by addition of 200µl of OPA/β-Mercaptoethanol per well. The plate was read on the Victor Wallac3 plate reader: excitation, 355nm and emission, 460nm, and IC50 values calculated.
PuSA[2]
PuSA activity was determined using the fluorogenic substrate Ala-AMC. Incubations contained diluted inhibitor/vehicle (20µl), substrate (125µM Ala-AMC in 0.125M TrisHCl buffer, pH 7.5; 40µl) and enzyme (40µl). After incubation for 2h at 37o C, the reaction was stopped by addition of 100µl 3% (v/v) acetic acid. Fluorescence was measured using a SLT Fluostar fluorimeter.
LTA4 Hydrolase[2]
LTA4 hydrolase was assayed using the fluorogenic substrate Arg-AMC. Incubations contained diluted inhibitor/vehicle (10µl), substrate (285µM Arg-AMC in PBS-0.01% (v/v) Brij-35; 70µl) and enzyme (20µl). After incubation for 1h at 37o C, the reaction was stopped by addition of 100µl 3% (v/v) acetic acid. Fluorescence was measured using a SLT Fluostar fluorimeter.
PILSAP[2]
2 PILSAP activity was determined using the colorimetric substrate l-leucine pnitroanilide according to the manufacturer’s instructions.
Aminopeptidase N Aminopeptidase N was assayed using the fluorogenic substrate, Ala-AMC. Incubations contained diluted inhibitor/vehicle (20µl), substrate (40µl; final concentration, 60µM) and enzyme (40µl, 1:8000 dilution) and were incubated for 60min at 37o C prior to addition of 100µl 3% (v/v) acetic acid, to stop the reaction. Fluorescence was measured using a SLT Fluostar fluorimeter.
Aminopeptidase B[2]
Aminopeptidase B was assayed using the fluorescent substrate, Arg-AMC. Incubations contained diluted inhibitor/vehicle (20µl), substrate (40µl; final concentration 200µM) and enzyme (40µl) and were incubated for 120min at 37o C prior to addition of 100µl 3% (v/v) acetic acid, to stop the reaction. Fluorescence was measured using a SLT Fluostar fluorimeter.
MetAP-2[2]
MetAP-2 was assayed using Met-Ala-Ser as substrate, and released Met was detected by OPA in the presence of reducing agents to generate a fluorescent product. Incubations contained substrate Met-Ala-Ser-OH (1.67mM) in assay buffer (83mM sodium phosphate buffer pH 7.2, 83mM NaCl, 0.266mM CoCl2; 60µl), diluted inhibitor/vehicle (20µl) and enzyme (20µl). They were incubated for 90min at 37o C prior to addition of 150µl stop reagent (0.2mM OPA/0.2mM β-mercaptoethanol in 50mM borax pH 9.5). Fluorescence was measured using a SLT Fluostar fluorimeter.
Cell Assay
Cell proliferation, survival and cell cycle assays[1]
Inhibition of proliferation was measured using a WST-1 assay as per the manufacturer's instructions. Cell death was measured by flow cytometry using the AnnexinV:FITC Apoptosis Detection Kit I on a FACSCaliburTM. Cell cycle status was measured by fixing cells on ice in 70% ethanol, re-suspending in PBS and treated for 30 minutes at 37°C with 100 μg/ml RNase, then stained with 50 μg/ml Propidium Iodide (PI) and analysed by flow cytometry.
Cell Cycle Analysis[2]
Cytokinetic investigations into the effects of CHR-2797 or CHR-5346 were analysed on U-937 and HuT 78 tumour cell lines using a bivariate BrdUrd-PI staining FACS protocol, based on the method of Dolbeare et al (Proc Natl Acad Sci USA 1983;80:5573-77). This method also identified the presence of sub-G1 apoptotic DNA profiles. For annexin analysis, the presence of externalised phosphatidylserine was detected using the Annexin V-FITC staining kit-1. Samples were analysed by flow cytometry on a FACS Canto using a 488nm excitation laser, with a 530nm/30nm bandpass filter for FITC, and a 670nmLP filter for PI fluorescence emission.
CHR-2797 is a novel metalloenzyme inhibitor that is converted into a pharmacologically active acid product (CHR-79888) inside cells. CHR-79888 is a potent inhibitor of a number of intracellular aminopeptidases, including leucine aminopeptidase. CHR-2797 exerts antiproliferative effects against a range of tumor cell lines in vitro and in vivo and shows selectivity for transformed over nontransformed cells. Its antiproliferative effects are at least 300 times more potent than the prototypical aminopeptidase inhibitor, bestatin. However, the mechanism by which inhibition of these enzymes leads to proliferative changes is not understood. Gene expression microarrays were used to profile changes in mRNA expression levels in the human promyelocytic leukemia cell line HL-60 treated with CHR-2797. This analysis showed that CHR-2797 treatment induced a transcriptional response indicative of amino acid depletion, the amino acid deprivation response, which involves up-regulation of amino acid synthetic genes, transporters, and tRNA synthetases. These changes were confirmed in other leukemic cell lines sensitive to the antiproliferative effects of CHR-2797. Furthermore, CHR-2797 treatment inhibited phosphorylation of mTOR substrates and reduced protein synthesis in HL-60 cells, both also indicative of amino acid depletion. Treatment with CHR-2797 led to an increase in the concentration of intracellular small peptides, the substrates of aminopeptidases. It is suggested that aminopeptidase inhibitors, such as CHR-2797 and bestatin, deplete sensitive tumor cells of amino acids by blocking protein recycling, and this generates an antiproliferative effect. CHR-2797 is orally bioavailable and currently undergoing phase II clinical investigation in the treatment of myeloid leukemia.[2]
Animal Protocol
Rat mammary (HOSP.1P) carcinoma model: lung colonisation [2]
Female rats (CBH/cbi) were randomised into groups (n=8-10/group) to receive CHR2797 (3, 10 or 30mg/kg/day) or vehicle. HOSP.1P cells (passage 6/7, 30,000 in 0.4 ml) were injected i.v. into all rats under halothane anaesthesia. Rats were dosed daily p.o. commencing 2 days after tumour cell inoculation and continuing until day 32. Lungs were removed from all rats 24 hours after the final dose. After tumour dissection and fixation in Methacarn, their number, mean weights and total tumour burden were determined.[2]
Rat chondrosarcoma (HSN LV10) model: liver colonisation [2]
Male (CBH/cbi) rats were randomised into groups (n=9-10/group) to receive CHR2797 (10 or 100mg/kg/day) or vehicle. Animals were injected with HSN LV10 cells (3,000 tumour cells in 0.2ml; passage 17, propagated in vitro) via the mesenteric vein under halothane anaesthesia, by exteriorising a small portion of the small intestine. The wound was closed using a silk suture for the musculature and a Michel clip for the skin. Animals were dosed daily p.o. commencing 2 days after tumour cell inoculation and continuing until day 20. Blood was taken from all drug-treated rats 24 hours after the last dose, for estimation of drug and CHR-79888 concentrations. Tumours in the liver were dissected and ten tumours (where possible), of representative sizes, from each drug-treated rat were weighed and snap-frozen in liquid nitrogen for analyte estimation by LC/MS (see Figure 1D). The remaining tumours were fixed in Methacarn and their number, mean weight and total tumour burden determined, including those removed for analysis. [2]
Human tumour xenograft model: MDA-MB-435 breast carcinoma [2]
Nude mice (MF1 (nu/nu); Harlan, Bicester, UK) were inoculated sub-cutaneously into the mammary fat pad with MDA-MB 435 cells (106 cells/mouse) on day 0. The tumours were allowed to grow to an average volume of 350µl/animal. On day 23, the mice were divided into two treatment groups based on a composite of tumour volume and the rate of tumour growth. Animals were dosed with either vehicle or CHR-2797 (100 mg/kg/day p.o.) from day 23 until day 37. The primary tumour was resected on day 28 and the study ended on day 61. Lungs were dissected out at the end of the study and fixed in Bouin’s solution. Under a dissecting microscope, the diameter of all tumours was measured using callipers. Lung tumour numbers were also counted for each animal and the total tumour burden assessed.
Human tumour xenograft model: MDA-MB-468 breast carcinoma [2]
Nude mice (MF1 (nu/nu) were inoculated sub-cutaneously into the mammary fat pad with MDA-MB-468 cells (2x106 cells/mouse) on day 0. Either on day 22 (Supplementary Figure S4, bottom) or day 39 (Figure S4, top), they were randomised into groups (n ≥ 14 per group) based on a composite of tumour mass and % change in tumour mass/unit time. From days 22-49 or days 40-69, mice were treated i.p., u.i.d. with either CHR-2797 (100mg/kg) or vehicle. Every 3 or 4 days, tumour volume was assessed using callipers. Mice were culled on day 50 or 70.
References
[1]. Emma M Smith, et al. The combination of HDAC and aminopeptidase inhibitors is highly synergistic in myeloma and leads to disruption of the NFκB signalling pathway. Oncotarget. 2015 Jul 10;6(19):17314-27.
[2]. Krige D, et al. CHR-2797: an antiproliferative aminopeptidase inhibitor that leads to amino acid deprivation in human leukemic cells. Cancer Res. 2008 Aug 15;68(16):6669-79.
[3]. Jenkins C, et al. Aminopeptidase inhibition by the novel agent CHR-2797 (tosedostat) for the therapy of acute myeloid leukemia. Leuk Res. 2011 May;35(5):677-81.
[4]. Smith EM, et al. The combination of HDAC and aminopeptidase inhibitors is highly synergistic in myeloma and leads to disruption of the NFκB signalling pathway. Oncotarget. 2015 Jul 10;6(19):17314-27
Additional Infomation
(2S)-2-[[(2R)-2-[(1S)-1-hydroxy-2-(hydroxyamino)-2-oxoethyl]-4-methyl-1-oxopentyl]amino]-2-phenylacetic acid cyclopentyl ester is a secondary carboxamide, a hydroxamic acid and a carboxylic ester. ChEBI
Tosedostat has been used in trials studying the treatment and supportive care of AML, Leukemia, Pancreas Cancer, Multiple Myeloma, and Pancreatic Cancer, among others. Tosedostat is an inhibitor of the M1 family of aminopeptidases, in particular PuSA, and LTA4 hydrolase. It has demonstrated anti-tumour activity in a number of models of cancer, both as a single agent and in synergy with cytotoxic agents such as carboplatin and paclitaxel. It entered the clinical trial in patients with haematological malignancies. DrugBank
Tosedostat is a proprietary orally bioavailable inhibitor of the M1 family of aminopeptidases with potential antineoplastic activity. Aminopeptidase inhibitor CHR-2797 is converted intracellularly into a poorly membrane-permeable active metabolite (CHR-79888) which inhibits the M1 family of aminopeptidases, particularly puromycin-sensitive aminopeptidase (PuSA), and leukotriene A4 (LTA4) hydrolase; inhibition of these aminopeptidases in tumor cells may result in amino acid deprivation, inhibition of protein synthesis due to a decrease in the intracellular free amino acid pool, an increase in the level of the proapoptotic protein Noxa, and cell death. Noxa is a member of the BH3 (Bcl-2 homology 3)-only subgroup of the proapoptotic Bcl-2 (B-cell CLL/lymphoma 2) protein family.
Tosedostat has pleiotropic effects against a range of human tumor cell lines originating from diverse tumor types in vitro and in vivo.
Tosedostat is anti-proliferative agent which induces apoptosis in leukemic cell lines in vitro. The mechanism underlying these anti-cancer actions is unclear, particularly since normal cells are much less sensitive to the agents than transformed cells. It exerts potent anti-proliferative, pro-apoptotic and anti-angiogenic effects in vitro and shows selectivity for transformed over non-transformed cells. It inhibits a number of M1 aminopeptidase enzyme family members in vitro (eg puromycin-sensitive aminopeptidase (PSA), leukotriene A4 hydrolase (LTA4H)).
There is a growing body of evidence supporting the use of epigenetic therapies in the treatment of multiple myeloma. We show the novel HDAC inhibitor CHR-3996 induces apoptosis in myeloma cells at concentrations in the nanomolar range and with apoptosis mediated by p53 and caspase pathways. In addition, HDAC inhibitors are highly synergistic, both in vitro and in vivo, with the aminopeptidase inhibitor tosedostat (CHR-2797). We demonstrate that the basis for this synergy is a consequence of changes in the levels of NFκB regulators BIRC3/cIAP2, A20, CYLD, and IκB, which were markedly affected by the combination. When co-administered the HDAC and aminopeptidase inhibitors caused rapid nuclear translocation of NFκB family members p65 and p52, following activation of both canonical and non-canonical NFκB signalling pathways. The subsequent up-regulation of inhibitors of NFκB activation (most significantly BIRC3/cIAP2) turned off the cytoprotective effects of the NFκB signalling response in a negative feedback loop. These results provide a rationale for combining HDAC and aminopeptidase inhibitors clinically for the treatment of myeloma patients and support the disruption of the NFκB signalling pathway as a therapeutic strategy[1].
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C21H30N2O6
Molecular Weight
406.4727
Exact Mass
406.21038
Elemental Analysis
C, 62.05; H, 7.44; N, 6.89; O, 23.62
CAS #
238750-77-1
PubChem CID
15547703
Appearance
White to off-white solid powder
Density
1.2±0.1 g/cm3
Index of Refraction
1.566
LogP
2.23
tPSA
124.96
SMILES
O=C(OC1CCCC1)[C@@H](NC([C@@H]([C@H](O)C(NO)=O)CC(C)C)=O)C2=CC=CC=C2
InChi Key
FWFGIHPGRQZWIW-SQNIBIBYSA-N
InChi Code
InChI=1S/C21H30N2O6/c1-13(2)12-16(18(24)20(26)23-28)19(25)22-17(14-8-4-3-5-9-14)21(27)29-15-10-6-7-11-15/h3-5,8-9,13,15-18,24,28H,6-7,10-12H2,1-2H3,(H,22,25)(H,23,26)/t16-,17+,18+/m1/s1
Chemical Name
(S)-cyclopentyl 2-((R)-2-((S)-1-hydroxy-2-(hydroxyamino)-2-oxoethyl)-4-methylpentanamido)-2-phenylacetate
Synonyms
CHR2797; CHR-2797; CHR 2797; KZK563J2UW; C21H30N2O6; Tosedostat (CHR2797); Tosedostat.
HS Tariff Code
2934.99.9001
Storage

Powder      -20°C    3 years

                     4°C     2 years

In solvent   -80°C    6 months

                  -20°C    1 month

Shipping Condition
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
Solubility Data
Solubility (In Vitro)
DMSO : ~25 mg/mL (~61.51 mM)
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 2.5 mg/mL (6.15 mM) (saturation unknown) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% 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 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 (6.15 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 (6.15 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (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 corn oil and mix evenly.


 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 2.4602 mL 12.3010 mL 24.6021 mL
5 mM 0.4920 mL 2.4602 mL 4.9204 mL
10 mM 0.2460 mL 1.2301 mL 2.4602 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.

Calculator

Molarity Calculator allows you to calculate the mass, volume, and/or concentration required for a solution, as detailed below:

  • Calculate the Mass of a compound required to prepare a solution of known volume and concentration
  • Calculate the Volume of solution required to dissolve a compound of known mass to a desired concentration
  • Calculate the Concentration of a solution resulting from a known mass of compound in a specific volume
An example of molarity calculation using the molarity calculator is shown below:
What is the mass of compound required to make a 10 mM stock solution in 5 ml of DMSO given that the molecular weight of the compound is 350.26 g/mol?
  • Enter 350.26 in the Molecular Weight (MW) box
  • Enter 10 in the Concentration box and choose the correct unit (mM)
  • Enter 5 in the Volume box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 17.513 mg appears in the Mass box. In a similar way, you may calculate the volume and concentration.

Dilution Calculator allows you to calculate how to dilute a stock solution of known concentrations. For example, you may Enter C1, C2 & V2 to calculate V1, as detailed below:

What volume of a given 10 mM stock solution is required to make 25 ml of a 25 μM solution?
Using the equation C1V1 = C2V2, where C1=10 mM, C2=25 μM, V2=25 ml and V1 is the unknown:
  • Enter 10 into the Concentration (Start) box and choose the correct unit (mM)
  • Enter 25 into the Concentration (End) box and select the correct unit (mM)
  • Enter 25 into the Volume (End) box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 62.5 μL (0.1 ml) appears in the Volume (Start) box
g/mol

Molecular Weight Calculator allows you to calculate the molar mass and elemental composition of a compound, as detailed below:

Note: Chemical formula is case sensitive: C12H18N3O4  c12h18n3o4
Instructions to calculate molar mass (molecular weight) of a chemical compound:
  • To calculate molar mass of a chemical compound, please enter the chemical/molecular formula and click the “Calculate’ button.
Definitions of molecular mass, molecular weight, molar mass and molar weight:
  • Molecular mass (or molecular weight) is the mass of one molecule of a substance and is expressed in the unified atomic mass units (u). (1 u is equal to 1/12 the mass of one atom of carbon-12)
  • Molar mass (molar weight) is the mass of one mole of a substance and is expressed in g/mol.
/

Reconstitution Calculator allows you to calculate the volume of solvent required to reconstitute your vial.

  • Enter the mass of the reagent and the desired reconstitution concentration as well as the correct units
  • Click the “Calculate” button
  • The answer appears in the Volume (to add to vial) box
In vivo Formulation Calculator (Clear solution)
Step 1: Enter information below (Recommended: An additional animal to make allowance for loss during the experiment)
Step 2: Enter in vivo formulation (This is only a calculator, not the exact formulation for a specific product. Please contact us first if there is no in vivo formulation in the solubility section.)
+
+
+

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.

Clinical Trial Information
Crossover Trial of the Effect of a High-Fat Meal on the PK of Oral CHR 2797 in Healthy Male Subjects
CTID: NCT01638442
Phase: Phase 1
Status: Completed
Date: 2023-09-18
Tosedostat and Cytarabine or Azacitidine in Treating Older Participants With Acute Myeloid Leukemia or High Risk Myelodysplastic Syndrome
CTID: NCT01636609
Phase: Phase 1
Status: Terminated
Date: 2020-04-13
Tosedostat With Capecitabine in Patients With Metastatic Pancreatic Adenocarcinoma
CTID: NCT02352831
Phase: Phase 1/Phase 2
Status: Terminated
Date: 2019-08-28
Study of Clinical Efficacy and Safety of Tosedostat in MDS
CTID: NCT02452346
Phase: Phase 2
Status: Completed
Date: 2018-06-12
Tosedostat in Combination With Cytarabine or Decitabine in Treating Patients With Newly Diagnosed Acute Myeloid Leukemia or High-Risk Myelodysplastic Syndrome
CTID: NCT01567059
Phase: Phase 2
Status: Completed
Date: 2017-02-15
Contact Us