Size | Price | |
---|---|---|
500mg | ||
1g | ||
Other Sizes |
Targets |
Topoisomerase; Camptothecins
|
---|---|
ln Vitro |
Antibody-drug conjugates deliver anticancer agents selectively and efficiently to tumor tissue and have significant antitumor efficacy with a wide therapeutic window[2]. The antitumor activity of [fam-] trastuzumab deruxtecan for CRC with five CRC cell lines that possess different biological characteristics was investigated. The expression of HER2 at both mRNA and protein levels in these various cell lines was first examined. Immunoblot analysis and RT and real-time polymerase chain reaction (PCR) analysis revealed that the amounts of HER2 protein and HER2 mRNA were much smaller in all the CRC cell lines than in NCI-N87 cells. [fam-] trastuzumab deruxtecan attenuated the viability of NCI-N87 cells, consistent with previous results, whereas all five CRC cell lines showed resistance to this agent. These findings suggested that the expression level of HER2 protein might determine sensitivity to [fam-] trastuzumab deruxtecan.
|
ln Vivo |
Antitumor activity of DS-8201a in low HER2–expressing tumors [3]
T-DM1 has been approved for HER2-positive metastatic breast cancer patients, defined as being HER2 IHC 3+ or IHC 2+/FISH–positive according to the current guidelines, and there are still clinical unmet needs in FISH-negative, HER2 1+ and 2+ populations for HER2-targeting therapies. Therefore, the antitumor activity of DS-8201a was evaluated in various mice xenograft models with different HER2 expression levels; KPL-4 (strong positive), JIMT-1 (moderate positive), Capan-1 (weak positive), and GCIY (negative) (Fig. 4A and B). Anti-HER2 ADC with the same drug-linker as DS-8201a and about half the DAR (DAR 3.4) was also evaluated to investigate the effect of DAR on antitumor activity. While T-DM1 was effective against only the KPL4 model, DS-8201a was effective against all HER2-positive models with KPL4, JIMT-1, and Capan-1. Both ADCs were not effective in the GCIY model. Anti-HER2 ADC (DAR 3.4) inhibited tumor growth against all HER2-positive models, and the efficacy was HER2 expression–dependent. A stronger efficacy was apparently observed for DS-8201a than anti-HER2 ADC (DAR 3.4) in the HER2 weak–positive Capan-1 model. These results suggest that the high DAR ADC, DS-8201a, enables the delivery of sufficient payload amounts into cancer cells, indicating cytotoxicity even with low HER2 levels. In case of HER2 strong –positive models, even a low DAR ADC is able to deliver a sufficient amount of payload for cell death. DS-8201a was effective in tumors with broader HER2 levels due to its high DAR, approximately 8. To confirm HER2-specificity of DS-8201a in a HER2 low–expressing model, a competitive inhibition study was performed in a HER2 low CFPAC-1 model (Fig. 4C). The efficacy of DS-8201a was cancelled by the prior treatment of the anti-HER2 Ab, and the control IgG-ADC did not inhibit tumor growth at a 3-fold higher dose than DS-8201a. From these results, the HER2 specificity of DS-8201a in a HER2 low–expressing model was confirmed. |
Enzyme Assay |
Parallel artificial membrane permeability assay (PAMPA) was carried out using a Freedom EVO200 system. The filter membrane of the acceptor plate was coated with GIT‐0 lipid solution. Each compound solution in DMSO (10 mM) was added to Prisma HT buffer (Pion) to obtain 5‐μM donor solutions (containing 0.05% DMSO, pH 5.0 and pH 7.4), and then placed on a donor plate. The acceptor plate was filled with an acceptor sink buffer. The donor plate was stacked onto the acceptor plate and incubated for 4 h at 25°C. After incubation, the concentrations of compounds in both plates were measured by an LC‐MS/MS system (API 4000). The permeability coefficient (Peff; 10−6 cm/s) was calculated using PAMPA Evolution DP software (Pion).[1]
|
Cell Assay |
Cells were seeded in a 96‐well plate at 1000 cells/well for KPL‐4 and 2000 cells/well for MDA‐MB‐468. After overnight incubation, a serially diluted solution of each ADC was added. Cell viability was evaluated after 5 days using a CellTiter‐Glo luminescent cell viability assay from Promega according to the manufacturer's instructions. For coculture study, KPL‐4 and MDA‐MB‐468 cells were seeded in a 6‐well plate at 1 × 105 cells and 3 × 105 cells, respectively, in 2 mL/well culture medium. After overnight incubation, the supernatant was removed from the plate and each ADC diluent (10 nM) was added at 6 mL/well. Viable cells were detached from the plate after 5 days of culture, and the cell number in each well was determined using a cell counter. In order to determine the ratio of KPL‐4 and MDA‐MD‐468 cells of the total viable cells, the cells were stained with anti‐HER2/nue FITC and incubated on ice for 20 min. After washing, fluorescent signals of 2 × 104 stained cells were measured using a flow cytometer. Based on the number and ratio of HER2‐positive and HER2‐negative cells in each treatment well, the number of KPL‐4 or MDA‐MB‐468 cells was calculated.[1]
|
Animal Protocol |
In vivo xenograft studies
All in vivo studies were carried out in accordance with the local guidelines of the Institutional Animal Care and Use Committee. Specific pathogen‐free female CAnN.Cg‐Foxn1nu/CrlCrlj mice (BALB/c nude mice) aged 5 weeks were used. All models were established by s.c. inoculation in the flanks of the mice. NCI‐N87 and MDA‐MB‐468‐Luc models were established by injecting 5 × 106 and 1 × 107 cells suspended in a Matrigel matrix, respectively. After 6 days for NCI‐N87, and 9 days for MDA‐MB‐468‐Luc models, the tumor‐bearing mice were randomized into treatment and control groups based on the tumor volume, and dosing initiated (day 0). Each ADC was given i.v. to the mice at a dose of 3 or 10 mg/kg, and a volume of 10 mL/kg. As a vehicle, ABS buffer (10 mM acetate buffer, 5% sorbitol, pH 5.5) was given at the same volume as the ADCs. The tumor volume was defined as 1/2 × length × width2.[1]
|
ADME/Pharmacokinetics |
Pharmacokinetics in cynomolgus monkeys [3]
The plasma DS-8201a concentrations decreased exponentially after a single intravenous administration of DS-8201a. The volume of distribution at steady state (Vss) of DS-8201a and total antibody was close to the plasma volume (data not shown). No clear difference was observed in the pharmacokinetic profile between DS-8201a and the total antibody, indicating that the peptide-linker of DS-8201a is stable in plasma even at DAR 8 (Fig. 2E). A low level of DXd was detected only at the limited time points (Fig. 2E).
|
Toxicity/Toxicokinetics |
Safety profile of DS-8201a [3]
A repeated intravenous dosing (every 3 weeks for 3 doses) study was conducted in cynomolgus monkeys, the cross-reactive species for DS-8201a, and in rats (antigen–non-binding species; Table 1). In the rat study, no deaths or life-threatening toxicities were found at dose levels up to 197 mg/kg, the maximum dose. Therefore, the severely toxic dose of 10% in animals (STD10) was considered to be >197 mg/kg. In the monkey study, one female at the highest dose of 78.8 mg/kg was euthanized due to moribundity on day 26. The cause of the moribundity appeared to be the deteriorated condition of the animal, which included decreased body weight and food consumption, as well as bone marrow toxicity and intestinal toxicity. Microscopic findings in the intestines, bone marrow and lungs in the surviving monkeys are shown in Supplementary Table S1. Gastrointestinal toxicity and bone marrow toxicity are typical dose-limiting factors in the clinical use of topoisomerase I inhibitors. The effects of DS-8201a on the intestines were very slight, and severe changes were not pronounced in any animal at up to 78.8 mg/kg. The bone marrow toxicity was produced only at 78.8 mg/kg, and was accompanied by decreases in reticulocyte ratios. No abnormalities in leukocyte and erythrocyte counts were observed in monkeys at 10 and 30 mg/kg. The repeated dose of DS-8201a caused moderate pulmonary toxicity in monkeys at 78.8 mg/kg, and findings graded as slight or very slight after the 6-week recovery period at ≥30 mg/kg. On the basis of the mortality and severity of the findings above, the highest non-severely toxic dose (HNSTD) for monkeys was considered to be 30 mg/kg. DS-8201a was well tolerated at the doses up to 197 mg/kg in rats and 30 mg/kg in monkeys following the repeated administration corresponding to the clinical regimen, and the nonclinical safety profile was acceptable for entry into human trials.
|
References | |
Additional Infomation |
Antibody-drug conjugates deliver anticancer agents selectively and efficiently to tumor tissue and have significant antitumor efficacy with a wide therapeutic window. DS-8201a is a human epidermal growth factor receptor 2 (HER2)-targeting antibody-drug conjugate prepared using a novel linker-payload system with a potent topoisomerase I inhibitor, exatecan derivative (DX-8951 derivative, DXd). It was effective against trastuzumab emtansine (T-DM1)-insensitive patient-derived xenograft models with both high and low HER2 expression. In this study, the bystander killing effect of DS-8201a was evaluated and compared with that of T-DM1. We confirmed that the payload of DS-8201a, DXd (1), was highly membrane-permeable whereas that of T-DM1, Lys-SMCC-DM1, had a low level of permeability. Under a coculture condition of HER2-positive KPL-4 cells and negative MDA-MB-468 cells in vitro, DS-8201a killed both cells, whereas T-DM1 and an antibody-drug conjugate with a low permeable payload, anti-HER2-DXd (2), did not. In vivo evaluation was carried out using mice inoculated with a mixture of HER2-positive NCI-N87 cells and HER2-negative MDA-MB-468-Luc cells by using an in vivo imaging system. In vivo, DS-8201a reduced the luciferase signal of the mice, indicating suppression of the MDA-MB-468-Luc population; however, T-DM1 and anti-HER2-DXd (2) did not. Furthermore, it was confirmed that DS-8201a was not effective against MDA-MB-468-Luc tumors inoculated at the opposite side of the NCI-N87 tumor, suggesting that the bystander killing effect of DS-8201a is observed only in cells neighboring HER2-positive cells, indicating low concern in terms of systemic toxicity. These results indicated that DS-8201a has a potent bystander effect due to a highly membrane-permeable payload and is beneficial in treating tumors with HER2 heterogeneity that are unresponsive to T-DM1.[1]
|
Molecular Formula |
C52H56FN9O13
|
---|---|
Molecular Weight |
1034.099
|
Exact Mass |
1033.39816
|
CAS # |
2270986-87-1
|
Related CAS # |
Exatecan mesylate dihydrate;197720-53-9
|
PubChem CID |
169450545
|
Appearance |
Brown to dark brown solid powder
|
LogP |
-0.4
|
Hydrogen Bond Donor Count |
7
|
Hydrogen Bond Acceptor Count |
15
|
Rotatable Bond Count |
22
|
Heavy Atom Count |
75
|
Complexity |
2360
|
Defined Atom Stereocenter Count |
3
|
SMILES |
CC[C@@]1(C2=C(COC1=O)C(=O)N3CC4=C5[C@@H](CCC6=C5C(=CC(=C6C)F)N=C4C3=C2)NC(=O)COCNC(=O)CNC(=O)[C@H](CC7=CC=CC=C7)NC(=O)CNC(=O)CNC(=O)CCCCCN8C(=O)C=CC8=O)O
|
InChi Key |
WXNSCLIZKHLNSG-WAJAXPBZSA-N
|
InChi Code |
InChI=1S/C52H56FN9O13/c1-3-52(73)33-19-38-48-31(24-62(38)50(71)32(33)25-75-51(52)72)47-35(14-13-30-28(2)34(53)20-36(60-48)46(30)47)58-43(67)26-74-27-57-41(65)22-56-49(70)37(18-29-10-6-4-7-11-29)59-42(66)23-55-40(64)21-54-39(63)12-8-5-9-17-61-44(68)15-16-45(61)69/h4,6-7,10-11,15-16,19-20,35,37,73H,3,5,8-9,12-14,17-18,21-27H2,1-2H3,(H,54,63)(H,55,64)(H,56,70)(H,57,65)(H,58,67)(H,59,66)/t35-,37+,52+/m1/s1
|
Chemical Name |
6-(2,5-dioxopyrrol-1-yl)-N-[2-[[2-[[(2S)-1-[[2-[[2-[[(10S,23R)-10-ethyl-18-fluoro-10-hydroxy-19-methyl-5,9-dioxo-8-oxa-4,15-diazahexacyclo[14.7.1.02,14.04,13.06,11.020,24]tetracosa-1,6(11),12,14,16,18,20(24)-heptaen-23-yl]amino]-2-oxoethoxy]methylamino]-2-oxoethyl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-2-oxoethyl]amino]-2-oxoethyl]hexanamide
|
Synonyms |
(1R)-Deruxtecan; SCHEMBL26807942;
|
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) |
May dissolve in DMSO (in most cases), if not, try other solvents such as H2O, Ethanol, or DMF with a minute amount of products to avoid loss of samples
|
---|---|
Solubility (In Vivo) |
Note: Listed below are some common formulations that may be used to formulate products with low water solubility (e.g. < 1 mg/mL), you may test these formulations using a minute amount of products to avoid loss of samples.
Injection Formulations
Injection Formulation 1: DMSO : Tween 80: Saline = 10 : 5 : 85 (i.e. 100 μL DMSO stock solution → 50 μL Tween 80 → 850 μL Saline)(e.g. IP/IV/IM/SC) *Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH ₂ O to obtain a clear solution. Injection Formulation 2: DMSO : PEG300 :Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL DMSO → 400 μLPEG300 → 50 μL Tween 80 → 450 μL Saline) Injection Formulation 3: DMSO : Corn oil = 10 : 90 (i.e. 100 μL DMSO → 900 μL Corn oil) Example: Take the Injection Formulation 3 (DMSO : Corn oil = 10 : 90) as an example, if 1 mL of 2.5 mg/mL working solution is to be prepared, you can take 100 μL 25 mg/mL DMSO stock solution and add to 900 μL corn oil, mix well to obtain a clear or suspension solution (2.5 mg/mL, ready for use in animals). View More
Injection Formulation 4: DMSO : 20% SBE-β-CD in saline = 10 : 90 [i.e. 100 μL DMSO → 900 μL (20% SBE-β-CD in saline)] Oral Formulations
Oral Formulation 1: Suspend in 0.5% CMC Na (carboxymethylcellulose sodium) Oral Formulation 2: Suspend in 0.5% Carboxymethyl cellulose Example: Take the Oral Formulation 1 (Suspend in 0.5% CMC Na) as an example, if 100 mL of 2.5 mg/mL working solution is to be prepared, you can first prepare 0.5% CMC Na solution by measuring 0.5 g CMC Na and dissolve it in 100 mL ddH2O to obtain a clear solution; then add 250 mg of the product to 100 mL 0.5% CMC Na solution, to make the suspension solution (2.5 mg/mL, ready for use in animals). View More
Oral Formulation 3: Dissolved in PEG400  (Please use freshly prepared in vivo formulations for optimal results.) |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 0.9670 mL | 4.8351 mL | 9.6702 mL | |
5 mM | 0.1934 mL | 0.9670 mL | 1.9340 mL | |
10 mM | 0.0967 mL | 0.4835 mL | 0.9670 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 |
NCT04644237 | Active, not recruiting | Drug: Trastuzumab deruxtecan |
Non-Small Cell Lung Cancer | Daiichi Sankyo,Inc. | March 19, 2021 | Phase 2 |
NCT04619004 | Active, not recruiting | Drug: Patritumab Deruxtecan (Fixed dose) |
Non-Small Cell Lung Cancer Metastatic |
Daiichi Sankyo,Inc. | February 2, 2021 | Phase 2 |
NCT05458401 | Recruiting | Drug: Trastuzumab deruxtecan |
HER2-positive Breast Cancer | Daiichi Sankyo,Inc. | November 11, 2022 |