PeptideDB

Saruparib (AZD5305) 2589531-76-8

Saruparib (AZD5305) 2589531-76-8

CAS No.: 2589531-76-8

Saruparib (AZD 5305; AZD-5305; AZD5305) is a novel, potent, selective and orally bioactive PARP [Poly(ADP-ribose)polymer
Sales Email:peptidedb@qq.com

This product is for research use only, not for human use. We do not sell to patients.

Saruparib (AZD 5305; AZD-5305; AZD5305) is a novel, potent, selective and orally bioactive PARP [Poly(ADP-ribose)polymerase-1] inhibitor with anticancer activity. Its IC50 values for PARP1/2 inhibition are 3 nM and 1400 nM, respectively.


Physicochemical Properties


Molecular Formula C22H26N6O2
Molecular Weight 406.4808
Exact Mass 406.21
Elemental Analysis C, 65.01; H, 6.45; N, 20.68; O, 7.87
CAS # 2589531-76-8
PubChem CID 155586901
Appearance Off-white to light yellow solid powder
LogP 1.1
Hydrogen Bond Donor Count 2
Hydrogen Bond Acceptor Count 6
Rotatable Bond Count 5
Heavy Atom Count 30
Complexity 660
Defined Atom Stereocenter Count 0
InChi Key WQAVGRAETZEADU-UHFFFAOYSA-N
InChi Code

InChI=1S/C22H26N6O2/c1-3-16-11-19-20(26-21(16)29)10-15(12-24-19)14-27-6-8-28(9-7-27)17-4-5-18(25-13-17)22(30)23-2/h4-5,10-13H,3,6-9,14H2,1-2H3,(H,23,30)(H,26,29)
Chemical Name

5-[4-[(7-ethyl-6-oxo-5H-1,5-naphthyridin-3-yl)methyl]piperazin-1-yl]-N-methylpyridine-2-carboxamide
Synonyms

AZD-5305; AZD5305; 2589531-76-8; Saruparib; 16MZ1V3RBT; AZD-5305 [WHO-DD]; example 4 [WO2021013735]; 5-(4-((7-Ethyl-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)piperazin-1-yl)-N-methylpicolinamide; AZD 5305
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)

Biological Activity


Targets PARP1 ( IC50 = 3 nM ); PARP2 ( IC50 = 1400 nM )
ln Vitro AZD5305 has excellent pharmacokinetics in preclinical species, good secondary pharmacology, good physicochemical properties, and is a highly selective inhibitor for PARP1 over other PARP family members. In vitro, AZD5305 lessens the anti-proliferation effects on human bone marrow progenitor cells.
ln Vivo AZD5305 exhibits excellent in vivo efficacy as a potent and selective PARP1 inhibitor and PARP1-DNA trapper in a BRCA mutant HBCx-17 PDX model.[2]
AZD5305 demonstrates sustained antitumor activity in BRCAm xenograft and PDX models in vivo. Antitumor efficacy of AZD5305 correlates with PK and PD in BRCA1m tumor models. AZD5305 and carboplatin show combination benefit in HBCx-9 PDX and SUM149PT xenograft model. AZD5305 demonstrates reduced hematologic toxicity when compared with dual PARP1/2 inhibitors in rat preclinical models. [3]
AZD5305 demonstrates sustained antitumor activity in BRCAm xenograft and PDX models in vivo[3]
The antitumor efficacy of Saruparib (AZD5305) monotherapy was evaluated in vivo in BRCAm xenografts and PDX models. In the MDA-MB-436 BRCA1m TNBC model, daily treatment with ≥ 0.1 mg/kg of Saruparib (AZD5305) resulted in profound regressions (≥90%). Further reductions of AZD5305 dose levels to 0.03 mg/kg and 0.01 mg/kg resulted in diminished antitumor efficacy (40% regression and not efficacious, respectively; Fig. 3A). In Capan-1 xenografts, a BRCA2m pancreatic model, daily treatment with 1 or 10 mg/kg of Saruparib (AZD5305) resulted in tumor stasis, while a 0.1 mg/kg oral daily dose led to 52% tumor growth inhibition[3].
With suitable oral exposure in the mouse, compound 25/Saruparib (AZD5305) was tested for efficacy in vivo in a BRCA1m triple-negative breast cancer (TNBC) patient-derived explant (PDX) model, HBCx-17 (Xentech France) (Figure 4). Tumor fragments were implanted subcutaneously into mice, and the mice were randomized into treatment groups when the mean tumor volume reached approximately 0.1 cm3. Animals were treated daily for 5 weeks from the day after the randomization with the vehicle or compound 1 at 100 mg/kg or compound 25 at 10, 1, 0.1, and 0.03 mg/kg. Compound 25 dosed daily at 10 mg/kg and 1 mg/kg delivered over 80% tumor regression, which was comparable to the effect of compound 1 at 100 mg/kg, while 0.1 mg/kg of compound 25 resulted in 73% regression. Compound 25 dosed as low as 0.03 mg/kg also resulted in tumor regressions (25%)[2].
Enzyme Assay Compound coupling and competition affinity pulldowns [2]
PARP affinity probe was immobilized on sepharose beads through covalent linkage using primary amino compound 101 (see synthetic experimental section for preparation) and carboxyl groups as described previously.10 One mL of NHS-activated sepharose and compound 101 (2μmol/mL) were equilibrated in DMSO. 15 μL of triethylamine was added to start the coupling reaction and the mixture was incubated on an end-over-end shaker for 20 h in the dark. Free NHS-groups on the beads were blocked by adding 50 μL amino ethanol and incubation on an end-over-end shaker for 16-20 h in the dark. Coupled beads were washed and stored in isopropanol at 4 °C in the dark. Competition affinity pulldowns were performed as described previously10. Briefly, MDA-MB-436 cells were lysed in 0.8 % NP40, 50 mM Tris-HCl pH 7.5, 5 % glycerol, 1.5 mM MgCl2,150 mM NaCl, 1 mM Na3VO4, 25 mM NaF, 1 mM DTT, HALT protease inhibitor cocktail. Lysate (5 mg total protein each) was pre-incubated with increasing competitor (test compounds 1-5 or Saruparib (AZD5305)) concentrations (DMSO vehicle, 0.64 nM, 3.2 nM, 16 nM, 80 nM, 400 nM, 2 µM, 10 µM and 50 µM) on an end-over-end shaker for 45 min at 4°C. Subsequently, lysates were incubated with beads for 30 min at 4°C. The beads were washed and collected by centrifugation. To assess the degree of protein depletion from the lysates, a second pulldown was performed with fresh beads and the flow through of the DMSO control.10 Bound proteins were subjected to on-bead digestion for 12h at 37°C with trypsin (2ug) in the presence of 50mM tetraethylammonium tetrahydroborate (TEAB). Peptides were labeled with TMT 10-plex, combined into a single sample and then subjected to offline fractionation by high pH reverse-phase HPLC (Agilent 1200) on an Gemini C18 column (5 uM, 110 Å, 150 x 2 mm; Phenomenex) with 20mM ammonium hydroxide in water as mobile phase A and 20 mM ammonium hydroxide in acetonitrile. The 96 resulting fractions were pooled in a non-continuous manner into 8 fractions for subsequent mass spectrometry analysis.
Binding assay using TIRF microscopy (M&M) [2]
For surface-immobilization, full-length PARP1 and PARP2 was modified by chemical biotinylation. In brief, protein was incubated with NHS-PEG12-biotin in a 1:4 ratio for 4h. Residual reagent was removed by SEC. Microscopy-compatible 384-wells glass plates was cleaned using a S21 1:1:5 mixture of NH4:H2O2:H2O and heated to ~80 °C, before extensively washed with MQ. The surface was functionalized with PLL-g-PEG-biotin (SuSoS) at 10 ug/mL (overnight incubation) to eliminate unspecific binding of protein and probe before subsequent addition of pre-mixed biotinylated PARP1/2 and Neutravidin in a 1:1 ratio at 100 nM (>2h incubation). For affinity determination, dilution series (3-fold) of all compounds were mixed with Alexa647-probe (in-house custom synthesis) at 2.5 nM (final concentration) and added to the wells. Due to the slow binding kinetics and high potency of compounds, all samples were incubated for >4h to reach equilibrium. Dilution series (3-fold) of probe only was used to determine Kd of probe to enable Ki calculations. For determination of residence time, 500 nM of compound was added to each well and allowed to equilibrate before extensive washing using plate washer (BioTek) and addition of probe. The plate was rapidly transferred to the TIRF microscope for time-lapse imaging with an acquisition rate of 5 min/frame to minimize bleaching. Samples were imaged using a 60x oil immersion objective (NA= 1.49) mounted on a Ti Eclipse inverted microscope, equipped with a Cy5 filter cube, CoolLED pE4000 illumination system (635 nm illumination) and Orca Flash 4.0 CMOS camera. Multiple images (200×200 µm2 ) were acquired per sample using identical illumination (100 ms exposure time) and focus (via perfect focus system). Quantification of image intensity was used to determine steady state binding levels, which were fitted to a 1:1 binding model to determine IC50.
Cell Assay BRCA2 and DLD-1(-/-) Using a Multidrop Combi, 40 μL of DLD-1 cells per well are seeded into 384-well plates at a density of 5000 cells/mL and 2.5 × 104 cells/mL, respectively, in complete media. The plates are then incubated for an overnight period at 37 °C with 5% CO2. Day 0 plate was incubated for more than three hours at room temperature (RT) after sytox green (5 μL, 2 μM) and saponin (10 μL, 0.25% stock) were added using a Multidrop Combi. The plate was then sealed with a black adhesive lid. Cell Insight Focused with a 4× objective is used to image cells. Using an Echo 555, AZD5305 is added, and the mixture is then incubated for seven days at 37 °C with 5% CO2. Day 8: Plates are filled with sytox green (5 μL, 2 μM) and saponin (10 μL, 0.25% stock). A black adhesive lid is used to seal the plate, and it is incubated for more than three hours at room temperature. On a 4× Objective Cell Insight, every cell is read.
Hematotoxicity Assay[1]
All inhibitors were diluted in DMSO to a concentration of 3 mM (talazoparib) or 10 mM (compound 25/Saruparib (AZD5305)) and stored under nitrogen. CD34+ hematopoietic stem and progenitor cells were cultured overnight in StemSpan SFEM II media supplemented with 25 ng/mL SCF, 50 ng/mL TPO, and 50 ng/ml Flt3-L at 37 °C, 5% CO2. The following day, cells were resuspended in CellExpand Suspension Expansion Culture BFU, plated at 500 cells per well in 96-well plates, and technical replicates were treated with a vehicle or a concentration range of the compound. After 5 days of incubation, the number of viable cells per well was determined using CellTiter-Glo 2.0 with the luminescence readout being performed on an Envision plate reader. Data were normalized to vehicle controls and dose–response curves generated using non-linear regression (curve fit) analysis using GraphPad Prism (v9).
Animal Protocol Animals were treated from the day after the randomization. Saruparib (AZD5305) and olaparib (AZD2281) were administered by oral gavage once daily (QD) at 10 mL/kg final dose volume. Olaparib was formulated in 10% DMSO (Sigma), 30% Kleptose. Saruparib (AZD5305) was formulated in water/HCl pH 3.5–4. Carboplatin was prepared fresh on the day of dosing and administered intraperitoneally at 10 mL/kg final dose volume. Carboplatin was formulated in 0.85% physiologic saline. For Xentech studies carboplatin stock (Teva or Sandoz) was diluted to the final concentration in 0.9% NaCl.[3]
During monotherapy studies, rats received either 1 mg/kg Saruparib (AZD5305) or vehicle [0.5% w/v hydroxypropyl methylcellulose (HPMC)/0.1% Tween80 in water, adjusted for pH 3–3.2], 57 mg/kg niraparib or vehicle (0.5% w/v HPMC) or 100 mg/kg olaparib (co-administered with 0.5% w/v HPMC, 0.1% Tween 80 because this was a monotherapy arm from a combination study), or vehicle (0.5% w/v HPMC/0.1% Tween 80 in water) orally, daily for 14 days.
In combination studies, rats were dosed with 1 mg/kg Saruparib (AZD5305), 100 mg/kg olaparib, or vehicle daily. In the 14-day study, rats were dosed in combination with a single administration of 30 mg/kg carboplatin or vehicle (0.9% w/v physiologic saline) on day 1. In the 42-day two-cycle study, rats were dosed in combination with 40 mg/kg carboplatin or vehicle on day 1 and day 22. Carboplatin or vehicle control were delivered via a single slow (over 1 min) intravenous bolus injection into the lateral tail vein followed immediately with either vehicle control or test article.[3]
For toxicokinetic analysis of Saruparib (AZD5305), niraparib, and olaparib, and hematology analyses, blood samples were collected via tail vein prick at various time points. All animals were monitored throughout the study and sacrificed on day 15 or day 42, approximately 24 hours after administration of the final dose. Bone marrow (left femurs) was fixed in 10% neutral buffered formalin prior to processing, then sectioned and stained with hematoxylin and eosin (H&E) for histopathologic evaluation. Right femurs were processed for bone marrow progenitor cell population analysis by flow cytometry.[3]
ADME/Pharmacokinetics To correlate the antitumor efficacy of Saruparib (AZD5305) with PK of the compound at steady state in the MDA-MB-436 model (Fig. 3A), plasma samples were collected at various time points post the day 7 dose. In the maximally efficacious dose groups (≥0.1 mg/kg, achieving >90% tumor regression; Fig. 3A), the unbound plasma levels of Saruparib (AZD5305) were above the DLD-1 BRCA2−/− cells in vitro clonogenic assay IC95 (0.0064 μmol/L) for the duration of the dosing interval of 24 hours (Fig. 4B). In contrast, in the 0.03 mg/kg group, which achieved 40% regression, the unbound plasma levels of Saruparib (AZD5305) were above IC95 for only approximately 17 hours, while in the nonefficacious group (0.01 mg/kg), the unbound plasma concentration of Saruparib (AZD5305) did not reach the IC95 value. These data were consistent with the requirement to maintain high and sustained target engagement in order to achieve maximal efficacy.[2]
The in vivo preclinical pharmacokinetic profile of compound 25 [Saruparib (AZD5305)] was determined in the mouse, rat, dog, and cynomolgus monkey (Table 7). Compound 25 exhibited very low plasma clearances (CLp) of 0.23, 1.1, 0.33, and 0.84 mL/min/kg and generally low steady-state volumes of distribution (Vss) of 0.17, 0.38, 0.30, and 0.38 L/kg in the mouse, rat, dog, and monkey, respectively. The resulting plasma half-lives were 8.0, 4.6, 10, and 7.1 h, respectively. The oral bioavailability of 25 was high across all species, consistent with the low hepatic clearance and a high fraction absorbed. The unbound clearance of ≤34 mL/min/kg, in combination with high potency, was expected to drive low efficacious doses in vivo which was predicted to carry through to humans based on the equally low in vitro CLint values.[1]
The metabolic clearance routes of compound 25 were investigated in vitro in hepatocytes from preclinical species and humans. Consistent with the low hepatocyte CLint, metabolite formation was limited; however, several metabolites were observed, principally resulting from likely sequential oxidation on the methylcarboxamide resulting in the loss of the methyl group and subsequent further metabolism to the carboxylic acid. Other non-localizable oxidations were also observed (see the Supporting Information details).[1]
Toxicity/Toxicokinetics Because bone marrow toxicity and negative effects on the peripheral blood are common adverse events in the clinic for dual PARP1/2 inhibitors, we sought to get an early indication of the potential of compound 25 [Saruparib (AZD5305)] to cause hematotoxicity. To assess this, we used an in vitro hematotoxicity assay, and compared the effects of compound 25 [Saruparib (AZD5305)]and compound 4 on the viability of proliferating and differentiating hematopoietic stem/progenitor cells (HSPCs) following 5 days of compound treatment. Compound 4 showed a dose dependent reduction in viability of HSPCs, starting at 14 nM, with an IC50 of 27 nM and a maximum effect of 1% viability (Figure 5, red data). In contrast, despite compound 25 causing an initial loss of viability at concentrations as low as 3 nM, a dose-dependent effect was not observed and an IC50 could not be determined. At 100 nM, 46% of cells remained viable (in contrast to 10% with compound 4), with little further loss of viability at concentrations up to 10 μM, where a maximal effect of 38% was observed. Thus, these data suggest that compound 25 exerts less toxicity on human in vitro HSPCs than compound 4.[2]
Saruparib (AZD5305) demonstrates reduced hematologic toxicity when compared with dual PARP1/2 inhibitors in rat preclinical models[3]
To determine whether the PARP1 selectivity of Saruparib (AZD5305) would mitigate the hematologic toxicity observed for dual PARP1/2 inhibitors in rat preclinical models, we carried out comparative rat studies (16, 17). Saruparib (AZD5305) and clinical non–PARP1-selective PARPi were dosed once daily for 14 days: AZD5305 1 mg/kg once daily, olaparib 100 mg/kg once daily, or niraparib at 57 mg/kg once daily. AZD5305 and olaparib exposures were targeted to cover their respective cell IC95 for approximately 24 hours (Fig. 5A; dotted lines indicating the IC95). The 57 mg/kg niraparib dose resulted in an unbound AUC(0–24) of 13.7 μmol/L/hour, approximately matching the reported clinical free AUC of 11.4 μmol/L/hour from a 300 mg clinical dose (18). Sequential peripheral blood hematology analysis was performed (Fig. 5B; Supplementary Fig. S5A–S5C) and flow cytometry used to directly assess bone marrow lineage precursors at termination (Fig. 5C; Supplementary Fig. S5D). Consistent with clinical anemia reported for olaparib (14, 19, 20), we observed a sustained reduction in reticulocytes (immature red blood cells) in peripheral blood compared with vehicle controls, although overall red cell mass and hemoglobin were only beginning to show effects within this 14-day study (Fig. 5B; Supplementary Fig. S5A). There were no other significant effects noted in other blood lineages (Supplementary Fig. S5A). Consistent with the effects observed on the peripheral blood, flow cytometry measurement of bone marrow lineage precursor cells revealed reduced erythroid precursor numbers, while myeloid and platelet precursors were unaffected (Fig. 5C; Supplementary Fig. S5D).
References

[1]. Cancer J . 2021 Nov-Dec;27(6):521-528.

[2]. J Med Chem . 2021 Oct 14;64(19):14498-14512.

[3]. Clin Cancer Res . 2022 Nov 1;28(21):4724-4736.

Additional Infomation Saruparib is an orally bioavailable inhibitor of the nuclear enzyme poly(ADP-ribose) polymerase (PARP), with potential chemo/radiosensitizing and antineoplastic activities. Upon administration, saruparib selectively targets and binds to PARP and prevents PARP-mediated DNA repair of single-strand DNA breaks via the base-excision repair pathway. This enhances the accumulation of DNA strand breaks and promotes genomic instability and eventually leads to apoptosis. This may enhance the cytotoxicity of DNA-damaging agents. PARP catalyzes post-translational ADP-ribosylation of nuclear proteins that signal and recruit other proteins to repair damaged DNA and is activated by single-strand DNA breaks. The PARP-mediated repair pathway is dysregulated in a variety of cancer cell types.
Poly(ADP-ribose) polymerase (PARP) inhibitors have transformed the therapeutic landscape for advanced ovarian cancer and expanded treatment options for other tumor types, including breast, pancreas, and prostate cancer. Yet, despite the success of PARP inhibitors in our current therapeutic armamentarium, not all patients benefit because of primary resistance, whereas different acquired resistance mechanisms can lead to disease progression on therapy. In addition, the toxicity profile of PARP inhibitors, primarily myelosuppression, has led to adverse events in a proportion of patients as monotherapy, and has limited the use of PARP inhibitors for certain rational combination strategies, such as chemotherapy and targeted therapy regimens. Currently approved PARP inhibitors are essentially equipotent against PARP1 and PARP2 enzymes. In this review, we describe the development of next-generation PARP1-selective inhibitors that have entered phase I clinical trials. These inhibitors have demonstrated increased PARP1 inhibitory potency and exquisitely high PARP1 selectivity in preclinical studies-features that may lead to improved clinical efficacy and a wider therapeutic window. First-in-human clinical trials seeking to establish the safety, tolerability, and recommended phase II dose, as well as antitumor activity of these novel agents, have commenced. If successful, this next-generation of PARP1-selective agents promises to build on the succeses of current PARP inhibitor treatment paradigms in cancer medicine.[1]
Poly-ADP-ribose-polymerase (PARP) inhibitors have achieved regulatory approval in oncology for homologous recombination repair deficient tumors including BRCA mutation. However, some have failed in combination with first-line chemotherapies, usually due to overlapping hematological toxicities. Currently approved PARP inhibitors lack selectivity for PARP1 over PARP2 and some other 16 PARP family members, and we hypothesized that this could contribute to toxicity. Recent literature has demonstrated that PARP1 inhibition and PARP1-DNA trapping are key for driving efficacy in a BRCA mutant background. Herein, we describe the structure- and property-based design of 25 (AZD5305), a potent and selective PARP1 inhibitor and PARP1-DNA trapper with excellent in vivo efficacy in a BRCA mutant HBCx-17 PDX model. Compound 25 is highly selective for PARP1 over other PARP family members, with good secondary pharmacology and physicochemical properties and excellent pharmacokinetics in preclinical species, with reduced effects on human bone marrow progenitor cells in vitro.[2]
Purpose: We hypothesized that inhibition and trapping of PARP1 alone would be sufficient to achieve antitumor activity. In particular, we aimed to achieve selectivity over PARP2, which has been shown to play a role in the survival of hematopoietic/stem progenitor cells in animal models. We developed AZD5305 with the aim of achieving improved clinical efficacy and wider therapeutic window. This next-generation PARP inhibitor (PARPi) could provide a paradigm shift in clinical outcomes achieved by first-generation PARPi, particularly in combination. Experimental design: AZD5305 was tested in vitro for PARylation inhibition, PARP-DNA trapping, and antiproliferative abilities. In vivo efficacy was determined in mouse xenograft and PDX models. The potential for hematologic toxicity was evaluated in rat models, as monotherapy and combination. Results: AZD5305 is a highly potent and selective inhibitor of PARP1 with 500-fold selectivity for PARP1 over PARP2. AZD5305 inhibits growth in cells with deficiencies in DNA repair, with minimal/no effects in other cells. Unlike first-generation PARPi, AZD5305 has minimal effects on hematologic parameters in a rat pre-clinical model at predicted clinically efficacious exposures. Animal models treated with AZD5305 at doses ≥0.1 mg/kg once daily achieved greater depth of tumor regression compared to olaparib 100 mg/kg once daily, and longer duration of response. Conclusions: AZD5305 potently and selectively inhibits PARP1 resulting in excellent antiproliferative activity and unprecedented selectivity for DNA repair deficient versus proficient cells. These data confirm the hypothesis that targeting only PARP1 can retain the therapeutic benefit of nonselective PARPi, while reducing potential for hematotoxicity. AZD5305 is currently in phase I trials[3]

Solubility Data


Solubility (In Vitro) DMSO: 12.5~20 mg/mL (30.8~49.2 mM)
Ethanol: 2 mg/mL
Solubility (In Vivo) Solubility in Formulation 1: ≥ 0.56 mg/mL (1.38 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 5.6 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: ≥ 0.56 mg/mL (1.38 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 5.6 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.

Solubility in Formulation 3: ≥ 0.56 mg/mL (1.38 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 5.6 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly.

Solubility in Formulation 4: 5%DMSO+ 40%PEG300+ 5%Tween 80+ 50%ddH2O: 0.8mg/ml (1.97mM)

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 2.4601 mL 12.3007 mL 24.6015 mL
5 mM 0.4920 mL 2.4601 mL 4.9203 mL
10 mM 0.2460 mL 1.2301 mL 2.4601 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.