Size | Price | Stock | Qty |
---|---|---|---|
50mg |
|
||
100mg |
|
||
250mg |
|
||
1g |
|
||
Other Sizes |
|
Targets |
Topoisomerase
|
---|---|
ln Vitro |
Biochemical and Structural Basis of Exatecan as a Higher Potency TOP1 inhibitor [1]
DXd was derived from exatecan by the chemical modification of NH2 on the F-ring by a 2-hydroxyacetyl (COCH2OH) group (Supplementary Fig. S1A). Exatecan showed an average of 10 to 20 times more in vitro cytotoxic potency than DXd with subnanomolar IC50 (Supplementary Fig. S1B; Supplementary Table S1) in a panel of cancer cell lines. Cell-based TOP1 inhibition assays and structural modeling provided a mechanism of higher cytotoxic potency of exatecan. The level of DNA-trapped TOP1 (TOP1ccs) of exatecan in comparison with DXd and SN-38 was examined by a modified rapid approach to DNA adduct recovery (RADAR) assay (21). Exatecan was the most potent drug and induced TOP1ccs at a lower concentration than DXd and SN-38 (Fig. 1A; Supplementary Fig. S1C). DNA-trapped TOP1 is typically removed from DNA and degraded. To measure the rate of TOP1 degradation, cells were treated with exatecan or DXd/SN-38 for 2 hours and then allowed to grow without drugs for 30 minutes for the reversal of TOP1ccs and TOP1 degradation. Exatecan induced TOP1 degradation in a dose-dependent manner and was more effective than DXd and SN-38 (Fig. 1B). Exatecan Shows Less Sensitivity to MDR Mechanisms than DXd and SN38 [1] The MDR transporter ABCG2- and P-gp–mediated efflux of exatecan, DXd, and SN-38 was determined in Caco-2 cells (Supplementary Fig. S1E). DXd showed an order of magnitude higher efflux ratios than exatecan without or with the ABCG2 inhibitor novobiocin, the P-gp inhibitor verapamil, or the dual inhibitor GF120918 (Fig. 1D). Interestingly, SN-38 efflux ratios were higher than exatecan but lower than DXd. Immunofluorescence (IF) examination of the intracellular accumulation of exatecan and DXd showed that DXd accumulation was lower than exatecan in the ABCG2 high expression cell line NCI-H460 (Fig. 1E; Supplementary Fig. S1F–1J). Consistent with MDR substrate experiment results, the cytotoxic potency difference between DXd and exatecan showed a correlation with endogenous ABCG2/P-gp expression (mRNA and protein; Supplementary Fig. S2A–S2C). In general, the IC50 ratio of DXd/exatecan was higher in cells with a higher ABCG2 or P-gp expression (Fig. 1F; Supplementary Fig. S2D and S2E). The inhibition of ABCG2 or P-gp improved the cytotoxicity of DXd/SN-38 but had a much smaller effect on exatecan (Fig. 1G). Even with an improved IC50 with inhibitor for DXd/SN-38, exatecan remained more potent (Supplementary Fig. S2F and S2G). Collectively, exatecan demonstrated less sensitivity to the MDR genes than DXd/SN38, potentially conferring higher cytotoxicity in cancer cells along with a more prevalent resistance mechanism. |
ln Vivo |
Antibody-drug conjugates (ADC) using DNA topoisomerase I inhibitor DXd/SN-38 have transformed cancer treatment, yet more effective ADCs are needed for overcoming resistance. We have designed an ADC class using a novel self-immolative T moiety for traceless conjugation and release of exatecan, a more potent topoisomerase I inhibitor with less sensitivity to multidrug resistance (MDR). Characterized by enhanced therapeutic indices, higher stability, and improved intratumoral pharmacodynamic response, antibody-T moiety-exatecan conjugates targeting HER2, HER3, and TROP2 overcome the intrinsic or treatment resistance of equivalent DXd/SN-38 ADCs in low-target-expression, large, and MDR+ tumors. T moiety-exatecan ADCs display durable antitumor activity in patient-derived xenograft and organoid models representative of unmet clinical needs, including EGFR ex19del/T790M/C797S triple-mutation lung cancer and BRAF/KRAS-TP53 double-mutant colon cancer, and show synergy with PARP/ATR inhibitor and anti-PD-1 treatment. High tolerability of the T moiety-exatecan ADC class in nonhuman primates supports its potential to expand the responding patient population and tumor types beyond current ADCs.
Significance: ADCs combining a novel self-immolative moiety and topoisomerase I inhibitor exatecan as payload show deep and durable response in low-target-expressing and MDR+ tumors resistant to DXd/SN-38 ADCs without increasing toxicity. This new class of ADCs has the potential to benefit an additional patient population beyond current options. See related commentary by Gupta et al., p. 817. This article is highlighted in the In This Issue feature, p. 799 [1].
|
Cell Assay |
In Vitro Stability of MTX-1000/DS-8201a in Plasma [1]
The release rate of exatecan/DXd from MTX-1000/DS-8201a at the concentration of 10 mg/mL at 37°C up to 21 days was evaluated in mouse, rat, monkey, and human plasma. ELISA [1] For a binding assay, immune plates were coated with 2.5 mg/mL His-tagged HER2-ECD protein in coating buffer and kept overnight at 4°C. After washing, the plates were blocked, and each serially diluted substance was added to the wells. After incubation for 1.5 hours at 37°C, the plates were washed and incubated with HRP-conjugated anti-human IgG secondary antibody for 1 hour at 37°C. After washing, TMB solution was added and A450 in each well was measured with a microplate reader. For the detection of phosphorylated Akt (pAkt), SK-BR-3 cells were preincubated in a 96-well plate for 4 days and then incubated with each substance for 24 hours. After incubation, the cells were lysed and intercellular pAkt and total Akt were detected using a PathScan Phospho-Akt1 (Ser473) Sandwich ELISA Kit and PathScan Total-Akt1 Sandwich ELISA Kit according to the manufacturer's instructions. The relative pAkt of each sample well was calculated by dividing treated normalized pAkt values by untreated normalized pAkt values. Immunoblotting [1] KPL-4 cells were treated with each substance. After 24, 48, or 72 hours, the cells were harvested and lysed with M-PER lysis buffer containing Halt Protease and Phosphatase Inhibitor Cocktail. The samples were loaded and separated by SDS-PAGE and blotted onto polyvinylidene difluoride membranes. The membranes were blocked and probed overnight with anti–phospho-Chk1 (Ser345; 133D3) rabbit mAb, anti-Chk1 (2G1D5) mouse mAb, anti–cleaved PARP (Asp214) antibody, anti–β-actin (8H10D10) mouse mAb, anti–phospho-Histone H2A.X (Ser139) antibody, and anti–Histone H2A.X antibody (Abcam) at 4°C. Then, the membranes were washed and incubated with fluorescence-labeled secondary antibodies for 10 minutes using SNAP intradermally. The fluorescence signal was detected using an Odyssey imaging system. |
Animal Protocol |
Cell Line and PDX Studies [1]
All tumor-bearing models were established in female BALB/c nude mice that were purchased from the Shanghai Family Planning Research Institute. All in vivo studies were performed in accordance with the local guidelines of the Institutional Animal Care and Use Committee of Multitude Therapeutics and with the approval of the committee. Briefly, 4- to 6-week-old mice were housed together in sterilized cages and maintained under required pathogen-free conditions. Each cell suspension or tumor fragment was inoculated subcutaneously into female nude mice. When the tumor had grown to an appropriate volume, the tumor-bearing mice were randomized into treatment and control groups based on the tumor volumes, and dosing was started. Antibodies and ADCs were administered intravenously to the mice. Small-molecule drugs were administered orally. Tumor volume defined as 1/2 × length × width2 was measured twice (3–4 days) a week. TGI (%) was calculated as follows: TGI (%) = [1 – (mean of treatment group tumor volume on evaluation day)/(mean of control group tumor volume on evaluation day)] × 100. The mice were euthanized with CO2 gas when they reached endpoints (tumor volume exceeding 3,000 mm3, >10% reduction of body weight, or clinical signs indicating that mice should be euthanized for ethical reasons). Syngeneic Model [1] Female BALB/c mice, weighing approximately 18 to 20 g, were maintained with 5 mice per cage in an individually ventilated cage in a specific pathogen–free animal facility with autoclaved cages. After 9 days of acclimation, animals were subcutaneously inoculated with CT26-hHER2 cells (3 × 105 cells/0.1 mL/mouse) on the right flank for tumor development. Ten days after inoculation, 36 mice with tumor sizes ranging from 100 to 200 mm3 (average tumor size 178 mm3) were enrolled into the efficacy study and randomized into six therapy groups, and the treatments were initiated on the randomization day (defined as D0). MTX-1000, DS-8201a (10 mg/kg), and anti–PD-1 antibody (5 mg/kg) were administered intravenously at a volume of 10 mL/kg to mice. As a control, ABS buffer (10 mmol/L acetate buffer, 5% sorbitol, pH 5.5) was administered at the same volume as the drugs. MTX-1000 and DS-8201a were administered on days 0 and 7. Anti–PD-1 antibody was administered on days 0, 3, 7, and 10. For flow-cytometric analysis of T cells, dendritic cells, and tumor cells, when the average volume of tumors reached approximately 250 to 400 mm3 (8 days after tumor inoculation), the mice were treated with vehicle or DS-8201a (10 mg/kg, once, intravenously; day 0). The mice were euthanized with CO2 asphyxiation on day 8, and tumors were cut into small pieces and dissociated with the Tumor Dissociation Kit by the gentleMACS Octo Dissociator with Heaters. The resultant single cells were blocked with Mouse BD Fc Block reagent and stained with antibodies against mouse CD3, CD4, CD8, CD11c, CD45, CD86, granzyme B, MHC class I, MHC class II, and PD-L1 and human HER2. Tissue Multiplex IHC [1] Sections from formalin-fixed, paraffin-embedded tumor tissue blocks were stained using Opal multiplex according to the manufacturer's protocol for DAPI, CD45, CD4, and CD8. Slide scanning was performed on the Vectra 3.0 instrument. All the images were analyzed with the HALOTM Image Analysis platform. The whole section was analyzed, and the big necrosis area and stroma area were excluded. Single-positive cells were counted separately. Images were spectrally unmixed, evaluated for staining intensities and morphology, tissue segmented based on tissue markers, cell segmented based on nuclear and membrane markers, and phenotypically scored. PK of MTX-1000/DS-8201a in Rat/Mouse [1] Concentrations of ADC and the total antibody in plasma were determined with a validated ligand-binding assay; the lower limit of quantitation was 0.02 μg/mL. Briefly, immunoplates were coated with 1 μg/mL Human HER2 Protein, His Tag in coating buffer and kept overnight at 4°C. After washing, the plates were blocked, and each serially diluted sample was added to the wells. After incubation for 1 to 2 hours at 37°C, the plates were washed and incubated with HRP-conjugated anti-human IgG Fc secondary antibody for total antibody measurement or a biotin-labeled anti-exatecan/DXd antibody for ADC measurement. After reaction at 37°C for 1 to 2 hours, TMB solution was added directly or after incubation with Streptavidin Protein, HRP for 40 to 60 minutes at 37°C. A450 in each well was measured with a microplate reader. Concentrations of payload exatecan/DXd in plasma were determined with a validated LC/MS-MS method; the lower limit of quantitation was 0.05 ng/mL. MTX-1000/DS-8201a was intravenously administered at 4.0 mg/kg to rats. Plasma concentrations of ADC, total antibody, and DXd were measured up to 21 days after dose. |
ADME/Pharmacokinetics |
T Moiety Conjugation Translates Exatecan into a Superior ADC: Physicochemical and Pharmacologic Profile, Stability, and Toxicity [1]
T Moiety Design: Hydrophilic Modulation of the PABC Spacer for Traceless Conjugation [1] To enable a traceless conjugation and release of exatecan, we used a dipeptide VA linker and a modified self-immolative spacer p-amino benzyl (pAB) called T moiety (Fig. 2A; S1 in Supplementary Fig. S4A). VA was selected because it creates more hydrophilic high-DAR ADCs compared with other peptide linkers VC or Gly–Gly–Phe–Gly (GGFG; S0 in Supplementary Fig. S4A; refs. 29–30). Direct conjugation of HER2-targeting Tras and exatecan using the unmodified peptide linker VA/VC/GGFG caused high aggregation (Supplementary Fig. S4B). Integrating T moiety with a PEG group (T900; S3 in Supplementary Fig. S4A) significantly reduced aggregation to an acceptable level of 2%. However, modification of MC with the same PEG (called M moiety, M900; S2 in Supplementary Fig. S4A) led to a high aggregation of >50% (Supplementary Fig. S4B), highlighting the importance of the judicious selection of modification position. Considering the potential lethal adverse reactions caused by PEG-based biological drugs (31), we decided to use polysarcosine (pSAR) due to its higher solubility and biocompatibility (Supplementary Fig. S4C; ref. 32). Polysarcosine with 10 units (pSAR10) modified pAB (T1000) resulted in homogeneous ADC with a negligible level of aggregation (Supplementary Fig. S4B). Remarkably, an intermediate structure T800 with m-methylaminomethyl modification of pAB for attaching pSAR10 produced homogeneous ADC of high DAR without aggregation. In comparison, the attachment of the same m-methylaminomethyl group to the VA linker (VK-pABC, M800, CLogP = 4.98) caused 10% aggregation with exatecan even when T800 (CLogP = 4.73) and M800 are considered to be chemically equivalent, supporting pAB selection as the optimal modification site. The conjugation yield for T moiety ADCs was >90%. Replacing the VA linker with GGFG in T1000 also yielded homogeneous ADC without aggregation (T1001; Supplementary Fig. S4B). Tras–T1000–Exatecan (MTX-1000) Shows a Superior Physicochemical Profile [1] To compare the physicochemical parameters of Tras–T1000–exatecan (MTX-1000) with those of DS-8201a, we used either purchased DS-8201a or internally manufactured Tras–GGFG–DXd with closely matched physicochemical profiles (Supplementary Fig. S5A and S5B), in vivo potency (Supplementary Fig. S5C–S5E), and pharmacokinetics (PK) in mice (Supplementary Fig. S5F). The attachment of exatecan to the HER2 antibody did not affect HER2 antibody binding to target by flow cytometry or ELISA, similar to Tras–GGFG–DXd (Supplementary Fig. S6A and S6B). Tras–exatecan conjugates using T800 or T900 also resulted in more hydrophilic ADCs than Tras–GGFG–DXd (Supplementary Fig. S6C and S6D). Both DAR8 and DAR4 ADCs were readily generated (Supplementary Fig. S6E). Tras–T moiety–exatecan ADCs (Tras–T800–exatecan/MTX-800 and Tras–T1000–exatecan/MTX-1000, both DAR8) demonstrated similar or better stability than Tras–GGFG–DXd. This was measured by aggregation formed for an extended incubation time at 37°C and under repeat freeze–thaw cycles (Supplementary Fig. S6F). T moiety–exatecan ADCs also showed better thermostability and photostability than DS-8201a (Supplementary Fig. S6G and S6H). Notably, VA and pSAR ADC (T1000) showed better stability and less aggregation than other linker (GGFG) and modification (PEG; T900 or T1001; Supplementary Fig. S6C, S6D, S6F, and S6G), highlighting the impact of linker and conjugation chemistry choice on the physicochemical function of an ADC. |
Toxicity/Toxicokinetics |
Exatecan Mesylate Is Well Tolerated in Rats[1]
We conducted a 4-week (days 1, 8, 15, 22, and 29) intermittent intravenous dose toxicity study of exatecan mesylate in rats (6 animals/group) with a 4-week recovery period (3 of the 6 animals). Exatecan mesylate was well tolerated in rats at doses up to 10 mg/kg (calculated as exatecan-free base). At 30 mg/kg, exatecan mesylate resulted in decreased animal body weight, morbidity (decreased food consumption, abnormal clinical signs), and/or mortality (occurred 5 or 6 days after dose). The dose-dependent transient suppression of body weight gain (Supplementary Fig. S3A), decrease of blood cell counts (Supplementary Fig. S3B–S3E), and increase of key serum enzyme levels (Supplementary Fig. S3F and S3G) were observed in 3 mg/kg and 10 mg/kg groups but largely reversible by the end of the 4-week recovery period (Supplementary Table S2), suggesting that exatecan was similarly tolerated to DXd or SN-38. |
References |
|
Additional Infomation |
T moiety–exatecan ADCs maintained a higher potency without increasing toxicity with HNSTDs in nonhuman primate studies at a level that was similar to or higher than that of other TOP1 inhibitor ADCs. Notably, side effects from the administration of T moiety–exatecan ADC were often more prominently observed in the GI tract (diarrhea) and the hematologic system (reticulocyte reduction), though symptoms in both were fully reversible. The hematologic and digestive organ toxicities for T moiety–exatecan ADCs mirrored those of exatecan as a single agent in human trials. However, T moiety–exatecan ADC achieved more favorable safety signals in the GI tract and minimal myelotoxicity than exatecan, possibly reflecting the ADC design's superior physicochemical features. Improved stability, longer ADC half-life, lower free payload release, and the reduced myelotoxicity potential of ADC in comparison with free payload may all contribute to the favorable toxicity profile of T moiety–exatecan ADC. Interstitial lung disease occurred in just over 10% of DXd-based ADC–treated patients during clinical trials. Although we did not observe any structural damage or signs of inflammation in the lungs of animals dosed with T moiety–exatecan ADCs, it remains uncertain whether prolonged treatment will yield additional pulmonary toxicity. Nevertheless, it is conceivable that T moiety–exatecan ADCs could have different target-independent tissue distributions than DXd-based ADCs, resulting in the manifestation of diverse symptoms.
For tumors less responsive or resistant to TOP1 inhibitors, payload classes with different mechanisms may be necessary. T moiety is a modular structure compatible with diverse linker chemistry and is amenable to building ADCs with dual payloads. Taken together, T moiety–exatecan ADCs have the potential to address patient needs unfulfilled by current ADCs, whereas the versatility and scalability of T moiety can facilitate the introduction of payloads of completely different MOAs to meet the continuous challenges of drug resistance.[1] |
Molecular Formula |
C13H13NO5
|
---|---|
Molecular Weight |
263.25
|
Exact Mass |
263.079
|
CAS # |
102978-40-5
|
Related CAS # |
Exatecan Intermediate 1;110351-94-5;(R)-Exatecan Intermediate 1;110351-91-2
|
PubChem CID |
359849
|
Appearance |
White to light yellow solid powder
|
LogP |
0.089
|
Hydrogen Bond Donor Count |
1
|
Hydrogen Bond Acceptor Count |
5
|
Rotatable Bond Count |
1
|
Heavy Atom Count |
19
|
Complexity |
574
|
Defined Atom Stereocenter Count |
0
|
InChi Key |
IGKWOGMVAOYVSJ-UHFFFAOYSA-N
|
InChi Code |
InChI=1S/C13H13NO5/c1-2-13(18)8-5-9-10(15)3-4-14(9)11(16)7(8)6-19-12(13)17/h5,18H,2-4,6H2,1H3
|
Chemical Name |
4-ethyl-4-hydroxy-7,8-dihydro-1H-pyrano[3,4-f]indolizine-3,6,10-trione
|
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 | 3.7987 mL | 18.9934 mL | 37.9867 mL | |
5 mM | 0.7597 mL | 3.7987 mL | 7.5973 mL | |
10 mM | 0.3799 mL | 1.8993 mL | 3.7987 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.