Size | Price | Stock | Qty |
---|---|---|---|
10mg |
|
||
25mg |
|
||
50mg |
|
||
100mg |
|
||
250mg |
|
||
500mg |
|
||
1g |
|
||
Other Sizes |
|
Purity: ≥98%
Galunisertib (formerly known as LY2157299; LY-2157299) is a selective, ATP-mimetic, and orally bioavailable small molecule inhibitor of the TGFβ receptor I (TβRI) with potential anticancer activity. It inhibits TGFβ with an IC50 of 56 nM in cell-free assays. Galunisertib inhibited HCC cell migration on Laminin-5, Fibronectin, Vitronectin, Fibrinogen and Collagen-I and de novo phosphorylation of pSMAD2. Galunisertib inhibited HCC migration and cell growth independently of the expression levels of TGF-βRII. Galunisertib has antitumor activity in tumor-bearing animal models such as breast, colon, lung cancers,and hepatocellular carcinoma. Galunisertib was evaluated by Lilly Pharmaceuticals in multiple clinical trials either as a single agent or in combination with various agents such as gemcitabine, paclitaxel, sorafenib, or durvalumab. Galunisertib was in a phase II clinical trial for treatment of hepatocellular carcinoma but was discontinued in January 2020.
Targets |
TGF-β receptor type I (TGF-βRI) kinase (IC50 = 56 nM)
|
||
---|---|---|---|
ln Vitro |
In SK-Sora, HepG2, and Hep3B cell lines, galunisertib (LY2157299) (0.1, 1, 10, and 100 μM) somewhat potentiates Bay 43-9006 in a dose-dependent manner; however, this effect is not observed in JHH6, SK-HEP1, or HuH7 cell lines[2].
Galunisertib (LY2157299) is a selective ATP-mimetic inhibitor of TGF-β receptor (TβR)-I activation currently under clinical investigation in hepatocellular carcinoma (HCC) patients. Our study explored the effects of galunisertib in vitro in HCC cell lines and ex vivo on patient samples. Galunisertib was evaluated in HepG2, Hep3B, Huh7, JHH6 and SK-HEP1 cells as well as in SK-HEP1-derived cells tolerant to sorafenib (SK-Sora) and sunitinib (SK-Suni). Exogenous stimulation of all HCC cell lines with TGF-β yielded downstream activation of p-Smad2 and p-Smad3 that was potently inhibited with galunisertib treatment at micromolar concentrations. Despite limited antiproliferative effects, galunisertib yielded potent anti-invasive properties. Tumor slices from 13 patients with HCC surgically resected were exposed ex vivo to 1 µM and 10 µM galunisertib, 5 µM sorafenib or a combination of both drugs for 48 hours. Galunisertib but not sorafenib decreased p-Smad2/3 downstream TGF-β signaling. Immunohistochemistry analysis of galunisertib and sorafenib-exposed samples showed a significant decrease of the proliferative marker Ki67 and increase of the apoptotic marker caspase-3. In combination, galunisertib potentiated the effect of sorafenib efficiently by inhibiting proliferation and increasing apoptosis. |
||
ln Vivo |
Subcutaneous implants of human xenografts, Calu6 (non-small cell lung cancer) and MX1 (breast cancer), are made in nude mice. Galunisertib (LY2157299) causes a 70% reduction in pSmad for both kinds of cell lines when taken orally at a dose of 75 mg/kg. The recovery of pSmad to 80% of baseline occurred about 6 hours post-administration [3].
Human xenografts Calu6 (non-small cell lung cancer) and MX1 (breast cancer) were implanted subcutaneously in nude mice and LY2157299, a new type I receptor TGF-beta kinase antagonist, was administered orally. Plasma levels of LY2157299, percentage of phosphorylated Smad2,3 (pSmad) in tumour, and tumour size were used to establish a semi-mechanistic pharmacokinetic/pharmacodynamic model. An indirect response model was used to relate plasma concentrations with pSmad. The model predicts complete inhibition of pSmad and rapid turnover rates [t(1/2) (min)=18.6 (Calu6) and 32.0 (MX1)]. Tumour growth inhibition was linked to pSmad using two signal transduction compartments characterised by a mean signal propagation time with estimated values of 6.17 and 28.7 days for Calu6 and MX1, respectively. The model provides a tool to generate experimental hypothesis to gain insights into the mechanisms of signal transduction associated to the TGF-beta membrane receptor type I.[3] |
||
Enzyme Assay |
Recently, kinase inhibitors have shown great potential against fibrotic diseases and, specifically, the transforming growth factor-β receptor (TGF-βR) was found as a new and promising target for scleroderma therapy. In the current study, we propose that the large pool of existing kinase inhibitors could be exploited for inhibiting the TGF-βR to suppress scleroderma. In this respect, we developed a modeling protocol to systematically profile the inhibitory activities of 169 commercially available kinase inhibitors against the TGF-βR, from which five promising candidates were selected and tested using a standard kinase assay protocol. Consequently, two molecular entities, namely the PKB inhibitor MK-2206 and the mTOR C1/C2 inhibitor AZD8055, showed high potency when bound to the TGF-βR, with IC50 values of 97 and 86 nM, respectively, which are close to those of the recently developed TGF-βR selective inhibitors SB525334 and galunisertib/LY2157299 (IC50 = 14.3 and 56 nM, respectively). We also performed atomistic molecular dynamics simulations and post-molecular mechanics/Poisson–Boltzmann surface area analyses to dissect the structural basis and energetic properties of intermolecular interactions between the TGF-βR kinase domain and these potent compounds, highlighting intensive nonbonded networks across the tightly packed interface of non-cognate TGF-βR-inhibitor complexes[1].
|
||
Cell Assay |
Cell cytotoxicity assay[2]
Cell survival was determined using the MTT assay (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide). The conversion of yellow water-soluble tetrazolium MTT into purple insoluble formazan is catalyzed by mitochondrial dehydrogenases and used to estimate the number of viable cells. In brief, cells were seeded in 96-well tissue culture plates at a density of 2 × 103 cells/well. After drug exposure, cells were incubated with 0.4 mg/mL MTT for 4 hours at 37°C. After incubation, the supernatant was discarded, insoluble formazan precipitates were dissolved in 0.1 mL of DMSO, and the absorbance was measured at 560 nm by use of a microplate reader. Wells with untreated cells or with drug-containing medium without cells were used as positive and negative controls respectively. For proliferation assay, MTT assay was done daily to determine the number of viable cells in untreated control and galunisertib-treated group. Ex Vivo tissue profiling (TIPCAN®)[2] The effects of galunisertib were tested on freshly resected tumors from HCC patients which can be cultured alive in specific conditions of culture medium and atmosphere, depending on available tumor resection from the surgical department. After pathological evaluation by the hospital pathologist, the tumor samples were extemporaneously sliced using Tissue Slicer® instrument into 300μm-thick slices and cultured “alive” at 37°C into the William’s E medium, complemented with in-house proprietary dedicated components including foetal calf serum, glucose, gentamicin and HEPES, under normoxic conditions. The samples were prepared using tissue-slicer technology and treated for 24 to 72 hours with 1 and 10 μM galunisertib or 5 μM sorafenib. After 24 to 72 hours treatment, the explanted HCC was paraffin embedded and assessed for expression of selected markers. The tests comprised assessment of cancer cell proliferation (MIB1/Ki67), death (active caspase- 3), and several changes in cell signalling (phospho-kinases). Tissue quality was assessed by a pathologist. If tissue integrity was not maintained over time (>20% necrosis induction), tissues were discarded. |
||
Animal Protocol |
|
||
ADME/Pharmacokinetics |
Pharmacokinetic parameters were determined for patients administered galunisertib during the first 14 days of the 28-day intermittent treatment cycle (2 weeks on/2 weeks off schedule). The PK profile of galunisertib was characterized by rapid absorption, with median t max ranging from 0.5 to 2 h following oral dosing with 80 or 150 mg BID (Fig. 2). At steady state, on Day 14 in Cycle 1, the mean t 1/2 was 8.90 h and the mean CLss/F and Vz,ss/F during the terminal phase were 30.2 L/h and 388 L, respectively, for 150 mg BID (Table 3). Although the number of patients in the 2 cohorts was small and imbalanced (Cohort 1, n = 3; Cohort 2, n = 9), high interpatient variability for galunisertib exposure [AUC(0−48) coefficient of variation (CV) %] was observed (Cohort 1 CV % = 35 %; Cohort 2 CV % = 88 %). [5]
|
||
Toxicity/Toxicokinetics |
Galunisertib administered to 12 Japanese patients with advanced solid tumors was well tolerated and had a favorable safety profile; no DLTs or cardiovascular toxicities were reported. Dose escalation was successfully performed within the 2 dosing cohorts (80 and 150 mg BID) and galunisertib exposure data confirmed that exposure could be maintained within the predefined therapeutic window for the majority of patients during treatment with galunisertib. All patients completed at least one cycle of galunisertib treatment before discontinuing due to disease progression; no patients had a clinical response to treatment, however, two patients had stable disease. [5]
The favorable tolerability and safety profile of 80 and 150 mg BID doses of galunisertib in Japanese patients was confirmed based on the TEAE profile reported during the study. Overall, there were no CTCAE Grade ≥3 study drug-related toxicities reported. Possible drug-related TEAEs included two patients with increased BNP levels, two patients with leukopenia, and two patients with rash. The two patients with increased BNP (Grade 1) did not experience any cardiotoxicities, and no febrile neutropenia was reported for any patient. Possible study drug-related leukopenia (n = 3 events) was also reported in the FHD study in patients who received a combination of galunisertib and lomustine; however, causality could not be specifically attributed to either drug. Therefore, it is unclear if the reported leukopenia was related to galunisertib treatment. |
||
References |
|
||
Additional Infomation |
LY-2157299 is a pyrrolopyrazole that is 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole which is substituted at positions 2 and 3 by 6-methylpyridin-2-yl and 6-(aminocarbonyl)quinolin-4-yl groups, respectively. A Transforming growth factor-betaRI (TGF-betaRI) kinase inhibitor, it blocks TGF-beta-mediated tumor growth in glioblastoma. It has a role as a TGFbeta receptor antagonist and an antineoplastic agent. It is a member of quinolines, a pyrrolopyrazole, a member of methylpyridines, an aromatic amide and a monocarboxylic acid amide.
Galunisertib has been used in trials studying the basic science and treatment of Glioma, Neoplasms, Solid Tumor, GLIOBLASTOMA, and Prostate Cancer, among others. Galunisertib is an orally available, small molecule antagonist of the tyrosine kinase transforming growth factor-beta (TGF-b) receptor type 1 (TGFBR1), with potential antineoplastic activity. Upon administration, galunisertib specifically targets and binds to the kinase domain of TGFBR1, thereby preventing the activation of TGF-b-mediated signaling pathways. This may inhibit the proliferation of TGF-b-overexpressing tumor cells. Dysregulation of the TGF-b signaling pathway is seen in a number of cancers and is associated with increased cancer cell proliferation, migration, invasion and tumor progression. Galunisertib (LY2157299) is a selective ATP-mimetic inhibitor of TGF-β receptor (TβR)-I activation currently under clinical investigation in hepatocellular carcinoma (HCC) patients. Our study explored the effects of galunisertib in vitro in HCC cell lines and ex vivo on patient samples. Galunisertib was evaluated in HepG2, Hep3B, Huh7, JHH6 and SK-HEP1 cells as well as in SK-HEP1-derived cells tolerant to sorafenib (SK-Sora) and sunitinib (SK-Suni). Exogenous stimulation of all HCC cell lines with TGF-β yielded downstream activation of p-Smad2 and p-Smad3 that was potently inhibited with galunisertib treatment at micromolar concentrations. Despite limited antiproliferative effects, galunisertib yielded potent anti-invasive properties. Tumor slices from 13 patients with HCC surgically resected were exposed ex vivo to 1 µM and 10 µM galunisertib, 5 µM sorafenib or a combination of both drugs for 48 hours. Galunisertib but not sorafenib decreased p-Smad2/3 downstream TGF-β signaling. Immunohistochemistry analysis of galunisertib and sorafenib-exposed samples showed a significant decrease of the proliferative marker Ki67 and increase of the apoptotic marker caspase-3. In combination, galunisertib potentiated the effect of sorafenib efficiently by inhibiting proliferation and increasing apoptosis. Our data suggest that galunisertib may be active in patients with HCC and could potentiate the effects of sorafenib.[2] Human xenografts Calu6 (non-small cell lung cancer) and MX1 (breast cancer) were implanted subcutaneously in nude mice and LY2157299, a new type I receptor TGF-beta kinase antagonist, was administered orally. Plasma levels of LY2157299, percentage of phosphorylated Smad2,3 (pSmad) in tumour, and tumour size were used to establish a semi-mechanistic pharmacokinetic/pharmacodynamic model. An indirect response model was used to relate plasma concentrations with pSmad. The model predicts complete inhibition of pSmad and rapid turnover rates [t(1/2) (min)=18.6 (Calu6) and 32.0 (MX1)]. Tumour growth inhibition was linked to pSmad using two signal transduction compartments characterised by a mean signal propagation time with estimated values of 6.17 and 28.7 days for Calu6 and MX1, respectively. The model provides a tool to generate experimental hypothesis to gain insights into the mechanisms of signal transduction associated to the TGF-beta membrane receptor type I.[3] Transforming growth factor-beta (TGF-β) signaling regulates a wide range of biological processes. TGF-β plays an important role in tumorigenesis and contributes to the hallmarks of cancer, including tumor proliferation, invasion and metastasis, inflammation, angiogenesis, and escape of immune surveillance. There are several pharmacological approaches to block TGF-β signaling, such as monoclonal antibodies, vaccines, antisense oligonucleotides, and small molecule inhibitors. Galunisertib (LY2157299 monohydrate) is an oral small molecule inhibitor of the TGF-β receptor I kinase that specifically downregulates the phosphorylation of SMAD2, abrogating activation of the canonical pathway. Furthermore, galunisertib has antitumor activity in tumor-bearing animal models such as breast, colon, lung cancers, and hepatocellular carcinoma. Continuous long-term exposure to galunisertib caused cardiac toxicities in animals requiring adoption of a pharmacokinetic/pharmacodynamic-based dosing strategy to allow further development. The use of such a pharmacokinetic/pharmacodynamic model defined a therapeutic window with an appropriate safety profile that enabled the clinical investigation of galunisertib. These efforts resulted in an intermittent dosing regimen (14 days on/14 days off, on a 28-day cycle) of galunisertib for all ongoing trials. Galunisertib is being investigated either as monotherapy or in combination with standard antitumor regimens (including nivolumab) in patients with cancer with high unmet medical needs such as glioblastoma, pancreatic cancer, and hepatocellular carcinoma. The present review summarizes the past and current experiences with different pharmacological treatments that enabled galunisertib to be investigated in patients.[4] |
Molecular Formula |
C22H19N5O
|
---|---|
Molecular Weight |
369.42
|
Exact Mass |
369.158
|
Elemental Analysis |
C, 71.53; H, 5.18; N, 18.96; O, 4.33
|
CAS # |
700874-72-2
|
Related CAS # |
700874-72-2;924898-09-9 (hydrate);
|
PubChem CID |
10090485
|
Appearance |
White to yellow solid powder
|
Density |
1.4±0.1 g/cm3
|
Boiling Point |
619.0±55.0 °C at 760 mmHg
|
Flash Point |
328.2±31.5 °C
|
Vapour Pressure |
0.0±1.8 mmHg at 25°C
|
Index of Refraction |
1.751
|
LogP |
1.73
|
Hydrogen Bond Donor Count |
1
|
Hydrogen Bond Acceptor Count |
4
|
Rotatable Bond Count |
3
|
Heavy Atom Count |
28
|
Complexity |
585
|
Defined Atom Stereocenter Count |
0
|
InChi Key |
IVRXNBXKWIJUQB-UHFFFAOYSA-N
|
InChi Code |
InChI=1S/C22H19N5O/c1-13-4-2-5-18(25-13)21-20(19-6-3-11-27(19)26-21)15-9-10-24-17-8-7-14(22(23)28)12-16(15)17/h2,4-5,7-10,12H,3,6,11H2,1H3,(H2,23,28)
|
Chemical Name |
4-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-6-carboxamide
|
Synonyms |
LY2157299; LY2157299; 4-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-6-carboxamide; UNII-3OKH1W5LZE; ly2157299(galunisertib); LY 2157299
|
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 (In Vitro) |
|
|||
---|---|---|---|---|
Solubility (In Vivo) |
Solubility in Formulation 1: ≥ 5.75 mg/mL (15.56 mM) (saturation unknown) in 5% DMSO + 40% PEG300 + 5% Tween80 + 50% Saline (add these co-solvents sequentially from left to right, and one by one), clear solution.
Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution. Solubility in Formulation 2: ≥ 2.08 mg/mL (5.63 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 20.8 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. View More
Solubility in Formulation 3: ≥ 2.08 mg/mL (5.63 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. Solubility in Formulation 4: ≥ 2.08 mg/mL (5.63 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 20.8 mg/mL clear DMSO stock solution to 900 μL corn oil and mix evenly. Solubility in Formulation 5: 2% DMSO+30% PEG 300+ddH2O:5 mg/mL |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 2.7069 mL | 13.5347 mL | 27.0695 mL | |
5 mM | 0.5414 mL | 2.7069 mL | 5.4139 mL | |
10 mM | 0.2707 mL | 1.3535 mL | 2.7069 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.