PeptideDB

Dexrazoxane xHCl (ICRF-187) 149003-01-0

Dexrazoxane xHCl (ICRF-187) 149003-01-0

CAS No.: 149003-01-0

Dexrazoxane xHCl (formerly also known as ICRF-187, ADR-529) acts as an intracellular iron chelator, which decreases the
Sales Email:peptidedb@qq.com

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

Dexrazoxane xHCl (formerly also known as ICRF-187, ADR-529) acts as an intracellular iron chelator, which decreases the formation of superoxide radicals, and is mainly used as a cardioprotective agent. Dextrazoxane, a derivative of EDTA, chelates iron; however, its exact mechanism of action to protect the heart is unknown. The purpose of this medication is to shield the heart from cardiotoxic side effects. Rat cardiac myocytes exposed to higher anthracycline concentrations do not experience necrosis; instead, daunorubicin-induced myocyte apoptosis is prevented by dexrazoxane (10 mM), which is known clinically to limit anthracycline cardiac toxicity.


Physicochemical Properties


Molecular Formula C11H16N4O4.XHCL
Molecular Weight N/A
Exact Mass 304.094
Elemental Analysis C, 43.36; H, 5.62; Cl, 11.63; N, 18.39; O, 21.00
CAS # 149003-01-0
Related CAS # 1263283-43-7 (HCl); 24584-09-6; 149003-01-0 (HCl)
PubChem CID 6918223
Appearance White to off-white solid powder
Boiling Point 531.5ºC at 760 mmHg
Melting Point 193ºC
Flash Point 275.3ºC
Vapour Pressure 2.22E-11mmHg at 25°C
Hydrogen Bond Donor Count 3
Hydrogen Bond Acceptor Count 6
Rotatable Bond Count 3
Heavy Atom Count 20
Complexity 404
Defined Atom Stereocenter Count 1
SMILES

Cl[H].O=C1C([H])([H])N(C([H])([H])C(N1[H])=O)[C@@]([H])(C([H])([H])[H])C([H])([H])N1C([H])([H])C(N([H])C(C1([H])[H])=O)=O

InChi Key BIFMNMPSIYHKDN-FJXQXJEOSA-N
InChi Code

InChI=1S/C11H16N4O4.ClH/c1-7(15-5-10(18)13-11(19)6-15)2-14-3-8(16)12-9(17)4-14;/h7H,2-6H2,1H3,(H,12,16,17)(H,13,18,19);1H/t7-;/m0./s1
Chemical Name

4-[(2S)-2-(3,5-dioxopiperazin-1-yl)propyl]piperazine-2,6-dione;hydrochloride
Synonyms

ICRF-187 (ADR-529) HCl; (+)-Razoxane hydrochloride; ADR-529 hydrochloride; Cardioxan; Dexrazoxane HCl, Dexrazoxane hydrochloride; 149003-01-0; Dexrazoxane HCl; Totect; Cardioxane; Cardioxan; Savene; Zinecard; Dexrazoxane hydrochloride; ICRF-187 hydrochloride; Savene; ADR529; ADR-529; ADR 529; ICRF-187; ICRF187; ICRF 187; NSC169780; NSC-169780; NSC 169780; Cardioxan; Cardioxane; US brand names: Totect; Zinecard. Foreign brand names: Cardioxane Savene.
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

Note: Please store this product in a sealed and protected environment (e.g. under nitrogen), avoid exposure to moisture and light.
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 Iron chelator
ln Vitro

Dexrazoxane (10 mM), which is known to clinically limit the cardiac toxicity of anthracyclines, inhibits daunorubicin-induced myocyte apoptosis but not necrosis brought on by higher anthracycline concentrations in rat cardiac myocytes. It is likely that dexrazoxane's cardioprotective effects come from binding iron, either loosely bound or bound to doxorubicin, which prevents or reduces the production of oxygen radicals that damage specific cell components at a given site. In H9C2 cardiomyocytes, doxorubicin-induced DNA damage signal gamma-H2AX is specifically eliminated by dexrazoxane, but not by camptothecin or hydrogen peroxide. Additionally, doxorubicin-induced DNA damage is reduced in tandem with the fast degradation of Top2beta induced by dexrazoxane. Dexrazoxane interferes with Top2beta to counteract doxorubicin-induced DNA damage, which may be related to Top2beta's role in doxorubicin cardiotoxicity.

ln Vivo Dexrazoxane binds iron to stop the production of superhydroxide radicals, thereby averting mitochondrial damage. It is hydrolyzed to its active form inside cells.
Advances in cancer treatment utilizing multiple chemotherapies have dramatically increased cancer survivorship. Female cancer survivors treated with doxorubicin (DXR) chemotherapy often suffer from an acute impairment of ovarian function, which can persist as long-term, permanent ovarian insufficiency. Dexrazoxane (Dexra) pretreatment reduces DXR-induced insult in the heart, and protects in vitro cultured murine and non-human primate ovaries, demonstrating a drug-based shield to prevent DXR insult. The present study tested the ability of Dexra pretreatment to mitigate acute DXR chemotherapy ovarian toxicity in mice through the first 24 hours post-treatment, and improve subsequent long-term fertility throughout the reproductive lifespan. Adolescent CD-1 mice were treated with Dexra 1 hour prior to DXR treatment in a 1:1 mg or 10:1 mg Dexra:DXR ratio. During the acute injury period (2-24 hours post-injection), Dexra pretreatment at a 1:1 mg ratio decreased the extent of double strand DNA breaks, diminished γH2FAX activation, and reduced subsequent follicular cellular demise caused by DXR. In fertility and fecundity studies, dams pretreated with either Dexra:DXR dose ratio exhibited litter sizes larger than DXR-treated dams, and mice treated with a 1:1 mg Dexra:DXR ratio delivered pups with birth weights greater than DXR-treated females. While DXR significantly increased the "infertility index" (quantifying the percentage of dams failing to achieve pregnancy) through 6 gestations following treatment, Dexra pretreatment significantly reduced the infertility index following DXR treatment, improving fecundity. Low dose Dexra not only protected the ovaries, but also bestowed a considerable survival advantage following exposure to DXR chemotherapy. Mouse survivorship increased from 25% post-DXR treatment to over 80% with Dexra pretreatment. These data demonstrate that Dexra provides acute ovarian protection from DXR toxicity, improving reproductive health in a mouse model, suggesting this clinically available drug may provide ovarian protection for cancer patients[3].
Cell Assay Cells were treated with 0.1% DMSO (solvent control) or 200 μmol/L Dexrazoxane for 5 h followed by coincubation with doxorubicin for 1 day or VP-16 for 2 days. MTT (0.1 mg) was then added to each well and cells were incubated for an additional 4 h at 37°C. After removal of medium, DMSO was added and absorbance at 570 nm was measured using the Microplate Reader. Average IC50 values (mean ± SE) were determined in triplicate or quadruplicate.[1]
Neutral comet assay. Primary MEFs were treated with DMSO or doxorubicin for 1.5 h in a CO2 incubator at 37°C followed by additional 30-min incubation in fresh medium to reverse Top2 cleavage complexes. H9C2 cells were treated with DMSO or Dexrazoxane (100 μmol/L) for 3 h, washed, and replenished with fresh medium. Cells were then treated with DMSO or doxorubicin for 1.5 h followed by additional 30-min incubation in fresh medium to reverse Top2 cleavage complexes. Cells were then washed and trypsinized using 0.005% trypsin and resuspended in DMEM supplemented with 10% FetalPlex animal serum complex (10,000/mL). Cell suspension (50 μL) was then mixed with 500 μL 0.5% low-melting point agarose at 37°C. Cell/agarose mixture (75 μL) was transferred onto glass slides. Slides were then immersed in prechilled lysis buffer [2.5 mol/L NaCl, 100 mmol/L EDTA, 10 mmol/L Tris (pH 10.0), 1% Triton X-100, 10% DMSO] for 1 h followed by equilibration in 1× Tris-borate EDTA (TBE) buffer for 30 min. Slides were electrophoresed in 1× TBE at 1.0 V/cm for 10 min and stained with Vistra Green. Images were visualized under a fluorescence microscope and captured with a charge-coupled device camera. The average comet tail moment was determined from measuring at least 100 cells for each treatment group as described previously. Statistical analysis of the mean comet tail moments was done using Student's t test.[1]
Band depletion assay. H9C2 cells (1.2 × 105) were treated with 250 μmol/L VP-16 in the presence or absence of Dexrazoxane (150 μmol/L) for 15 min. Cells were either lysed immediately or incubated in drug-free medium for another 30 min at 37°C (to reverse Top2 cleavage complexes) before lysis. Cell lysates were analyzed by Western blotting using the anti-Top2α/Top2β and anti–α-tubulin antibody. The amount of Top2 cleavage complexes can be estimated from the difference between the amount of free Top2 after reversal and the amount of free Top2 without reversal[1].
Animal Protocol Mice [3]
All surgery was performed under Ketamine and isofluorane anesthesia. Female CD-1 mice were allowed to acclimate to the laboratory environment for one week prior to the start of an experiment under the supervision and care of the animal facility staff. At 4 weeks of age, the adolescent mice were injected with Dexrazoxane/Dexra or vehicle control (0.0167 M lactate in saline) via intraperitoneal injection using ≤ 200 μL/injection 1 hour prior to DXR injection. DXR or vehicle (saline) was subsequently administered via intraperitoneal injection.
Acute treatment [3]
At 4 weeks of age, mice were treated with 1) Vehicle for Dexrazoxane/Dexra + Vehicle for DXR, 2) Vehicle for Dexra + 20 mg/kg DXR, 3) 20 mg/kg Dexra + Vehicle for DXR, or 4) 20 mg/kg Dexra + 20 mg/kg DXR; doses were calculated based on the average weight of a 4-week-old CD-1 mouse. The 20 mg/kg DXR dose represents twice the maximum human equivalent DXR dose and was chosen in order to engage ample acute DXR toxicity. The 20 mg/kg Dexra dose represents a 1:1 Dexra/DXR mg ratio, providing a significant dose reduction from that used in cardioprotection to limit potential side effects of Dexra. The chosen Dexra dose was based on our previous in vitro study demonstrating a 2 μM Dexra dose, 100-folds lower than that used in in vitro cardiac protection studies, preserved granulosa cell viability against DXR. Animals were euthanized with CO2 followed by cervical dislocation and ovaries removed surgically 0, 2, 4, 10, 12 or 24 h after the second injection. Experiments were carried out in 4 biological replicates in which 3 mice were treated per drug group and harvested for each time point per biological replicate; in sum, n = 12 animals per treatment were totaled across all replicates. Ovaries were placed in 2 mL phosphate buffered saline, pH 7.4, and cleared of fat and attached bursa. For each ovarian pair, one was fixed in 10% formalin and processed for TUNEL assay, and the second was processed for a neutral comet assay. Separate mice were treated to provide ovaries utilized for protein extraction followed by Western blot analysis as previously described.
Breeding trial [3]
Female CD-1 mice were housed in Innovive system cages from 3 weeks until 8 months of age. At 4 weeks of age, mice were treated with: 1) Vehicle for Dexrazoxane/Dexra + Vehicle for DXR, 2) Vehicle for Dexra + 10mg/kg DXR, 3) 10mg/kg Dexra (1:1 mg ratio) + 10mg/kg DXR, 4) 100 mg/kg Dexra (10:1 mg ratio) + 10mg/kg DXR, 5) 10mg/kg Dexra (1:1 mg ratio) + Vehicle for DXR, or 6) 100mg/kg Dexra (10:1 mg ratio) + Vehicle for DXR. DXR was administered at 10 mg/kg body weight (a human equivalent dose of 30mg/m2) to minimize long-term cardiotoxicity. Dexra dose is expressed as a ratio to DXR dose throughout the manuscript. Dexra was administered at either a 1:1 mg ratio (labeled as Dexra1:DXR1, groups 3 above) or 10:1 mg ratio (labeled as Dexra10:DXR1, group 4 above, currently used in cardioprotective protocols) to DXR as indicated. Dexra control-treated animals (groups 5 and 6, above) are labeled as DexraC (DexraC1 and DexraC10 respectively) throughout the manuscript. At 6 weeks of age and prior to breeding, animals were treated for two weeks with drinking water medicated with enrofloxacin (22.7 mg/ml) at a calculated dose of 5 mg/kg (0.5 mL/300 mL ddH2O bottle) as a prophylactic to mitigate the side effects of a compromised immune system brought on by DXR treatment. At 8 weeks of age, females were moved to breeder cages where two females were paired with one male. Females were continuously mated from 8 weeks of age to 8 months of age or until 6 litters were achieved. Males were rotated following each breeding round to minimize any potential male-specific infertility effect. Animals within the breeder cage were fed a maintenance chow diet with protein: 24%; Fat: 4%; Fiber: 4.5% as well as irradiated sunflower seeds. Bi-weekly assessment of animal health was conducted, and additional nutritive support via DietGel® and sunflower seeds was given to females having difficulty maintaining body condition. Females remained within the breeder cage until they showed visual or palpable signs of pregnancy, at which point they were separated and maintained on a breeder irradiated diet (Protein: 19%; Fat: 9%; Fiber: 5%) until parturition. The health of the breeding mice was monitored at least three times daily when the mice were near parturition. [3]
br> Following delivery, pups were separated and the females were returned to the breeder cage within 24 h post-partum. The pups were counted, weighed, and euthanized on post-natal day 1 (PND1). At 8 months of age, the now non-pregnant dams were weighed, anesthetized with isoflurane (confirmed with limb pinch) and sacrificed via terminal blood draw followed by cervical dislocation. A terminal blood draw was carried out for future studies. Ovaries were removed from each female and weighed. Mice that did not survive to breeding age or that displayed signs of deteriorating health were removed from the breeding trial to minimize any suffering. The breeding trial was carried out in 4 replicates, with 3–6 mice per group per replicate, where the total number of female mice in each group at the start of breeding was 16 control, 16 DXR, 21 Dexrazoxane/Dexra1:DXR1, 16 Dexra10:DXR1, 12 DexraC1, and 12 DexraC10 across all 4 replicates. Data for survival analysis, pup weights, and litter sizes were included for analysis at the intervals for which the dam was present in the trial. Infertility index was conducted on mice that gave birth at each mating round and ovarian weight analysis was conducted at 8 months.
ADME/Pharmacokinetics Absorption, Distribution and Excretion
IV administration results in complete bioavailability.
Urinary excretion plays an important role in the elimination of dexrazoxane. Forty-two percent of the 500 mg/m2 dose of dexrazoxane was excreted in the urine.
9 to 22.6 L/m^2
7.88 L/h/m2 [dose of 50 mg/m2 Doxorubicin and 500 mg/m2 Dexrazoxane]
6.25 L/h/m2 [dose of 60 mg/m2 Doxorubicin and 600 mg/m2 Dexrazoxane]
After intravenous administration, the drug is rapidly distributed into tissue fluids, the highest concentrations of the parent drug and its hydrolysis product being found in hepatic and renal tissues.
The mean peak plasma concentration of dexrazoxane was 36.5 mcg/mL at the end of the 15-minute infusion of a 500 mg/sq m doxorubicin dose. Following a rapid distributive phase, dexrazoxane reaches post-distributive equilibrium within 2 to 4 hours.
The estimated steady-state volume of distribution of dexrazoxane suggests its distribution primarily in the total body water (25 L/sq m ).
In vitro studies have shown that /dexrazoxane/ is not bound to plasma proteins.
For more Absorption, Distribution and Excretion (Complete) data for DEXRAZOXANE (9 total), please visit the HSDB record page.
Metabolism / Metabolites
Dexrazoxane is hydrolysed by the enzyme dihydropyrimidine amidohydrolase in the liver and kidney to active metabolites that are capable of binding to metal ions.
Metabolic products include the unchanged drug, a diacid-diamide cleavage product, and two monoacid-monoamide ring products of unknown concentrations.
In vitro studies have shown dexrazoxane to be hydrolysed by DHPase in liver and kidney, but not heart extracts.
/This/ study was undertaken to determine the metabolism of dexrazoxane (ICRF-187) to its one-ring open hydrolysis products and its two-rings opened metal-chelating product ADR-925 in cancer patients with brain metastases treated with high-dose etoposide. In this phase I/II trial dexrazoxane was used as a rescue agent to reduce the extracerebral toxicity of etoposide. Dexrazoxane and its one-ring open hydrolysis products were determined by HPLC and ADR-925 was determined by a fluorescence flow injection assay. The two one-ring open hydrolysis intermediates of dexrazoxane appeared in the plasma at low levels upon completion of dexrazoxane infusion and then rapidly decreased with half-lives of 0.6 and 2.5 hr. A plasma concentration of 10 micro M ADR-925 was also detected at the completion of the dexrazoxane i.v. infusion period, indicating that dexrazoxane was rapidly metabolized in vivo. A plateau level of 30 micro M ADR-925 was maintained for 4 hr and then slowly decreased. The pharmacokinetics of dexrazoxane were found to be similar to other reported data in other settings and at lower doses. The rapid appearance of ADR-925 in plasma may make ADR-925 available to be taken up by heart tissue and bind free iron. These results suggest that the dexrazoxane intermediates are enzymatically metabolized to ADR-925 and provide a pharmacodynamic basis for the antioxidant cardioprotective activity of dexrazoxane.
Dexrazoxane is hydrolysed by the enzyme dihydropyrimidine amidohydrolase in the liver and kidney to active metabolites that are capable of binding to metal ions.
Route of Elimination: Urinary excretion plays an important role in the elimination of dexrazoxane. Forty-two percent of the 500 mg/m2 dose of dexrazoxane was excreted in the urine.
Half Life: 2.5 hours
Biological Half-Life
2.5 hours
The distribution half-life has ranged from about 12 to 60 minutes ...
Elimination - 2.5 hours.
Toxicity/Toxicokinetics Toxicity Summary
The mechanism by which dexrazoxane exerts its cardioprotective activity is not fully understood. Dexrazoxane is a cyclic derivative of EDTA that readily penetrates cell membranes. Results of laboratory studies suggest that dexrazoxane (a prodrug) is converted intracellularly to a ring-opened bidentate chelating agent that chelates to free iron and interferes with iron-mediated free radical generation thought to be responsible, in part, for anthracycline-induced cardiomyopathy. It should be noted that dexrazoxane may also be protective through its inhibitory effect on topoisomerase II.
Effects During Pregnancy and Lactation
◉ Summary of Use during Lactation
No information is available on the use of dexrazoxane during breastfeeding. The manufacturer recommends that women not breastfeed during treatment and for 2 weeks following the final dose of dexrazoxane. However, because dexrazoxane is used with doxorubicin, the abstinence period might be longer, depending on the doxorubicin dose.
◉ Effects in Breastfed Infants
Relevant published information was not found as of the revision date.
◉ Effects on Lactation and Breastmilk
Relevant published information was not found as of the revision date.
Protein Binding
Very low (< 2%)
Toxicity Data
Man(iv): TDLo: 383 mg/kg
Mouse(ip): LDLo 800 mg/kg
Dog(iv): LDLo: 2 gm/kg
Intraperitoneal, mouse LD10 = 500 mg/kg. Intravenous, dog LD10 = 2 gm/kg.
Interactions
There was no significant change in the pharmacokinetics of doxorubicin (50 mg/sq m ) and its predominant metabolite, doxorubicinol, in the presence of dexrazoxane (500 mg/sq m ) in a crossover study in cancer patients.
References

[1]. Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Res. 2007 Sep 15;67(18):8839-46.

[2]. Daunorubicin-induced apoptosis in rat cardiac myocytes is inhibited by dexrazoxane. Circ Res. 1999 Feb 19;84(3):257-65.

[3]. Dexrazoxane Diminishes Doxorubicin-Induced Acute Ovarian Damage and Preserves Ovarian Function and Fecundity in Mice. PLoS One . 2015 Nov 6;10(11):e0142588.

Additional Infomation Dexrazoxane Hydrochloride is the hydrochloride salt of a bisdioxopiperazine with iron-chelating, chemoprotective, cardioprotective, and antineoplastic activities. After hydrolysis to an active form that is similar to ethylenediaminetetraacetic acid (EDTA), dexrazoxane chelates iron, limiting the formation of free radical-generating anthracycline-iron complexes, which may minimize anthracycline-iron complex-mediated oxidative damage to cardiac and soft tissues. This agent also inhibits the catalytic activity of topoisomerase II, which may result in tumor cell growth inhibition.
The (+)-enantiomorph of razoxane.
See also: Dexrazoxane (has active moiety).
Drug Indication
Savene is indicated for the treatment of anthracycline extravasation.
Doxorubicin is among the most effective and widely used anticancer drugs in the clinic. However, cardiotoxicity is one of the life-threatening side effects of doxorubicin-based therapy. Dexrazoxane (Zinecard, also known as ICRF-187) has been used in the clinic as a cardioprotectant against doxorubicin cardiotoxicity. The molecular basis for doxorubicin cardiotoxicity and the cardioprotective effect of dexrazoxane, however, is not fully understood. In the present study, we showed that dexrazoxane specifically abolished the DNA damage signal gamma-H2AX induced by doxorubicin, but not camptothecin or hydrogen peroxide, in H9C2 cardiomyocytes. Doxorubicin-induced DNA damage was also specifically abolished by the proteasome inhibitors bortezomib and MG132 and much reduced in top2beta(-/-) mouse embryonic fibroblasts (MEF) compared with TOP2beta(+/+) MEFs, suggesting the involvement of proteasome and DNA topoisomerase IIbeta (Top2beta). Furthermore, in addition to antagonizing Top2 cleavage complex formation, dexrazoxane also induced rapid degradation of Top2beta, which paralleled the reduction of doxorubicin-induced DNA damage. Together, our results suggest that dexrazoxane antagonizes doxorubicin-induced DNA damage through its interference with Top2beta, which could implicate Top2beta in doxorubicin cardiotoxicity. The specific involvement of proteasome and Top2beta in doxorubicin-induced DNA damage is consistent with a model in which proteasomal processing of doxorubicin-induced Top2beta-DNA covalent complexes exposes the Top2beta-concealed DNA double-strand breaks. [1]
The clinical efficacy of anthracycline antineoplastic agents is limited by a high incidence of severe and usually irreversible cardiac toxicity, the cause of which remains controversial. In primary cultures of neonatal and adult rat ventricular myocytes, we found that daunorubicin, at concentrations /=10 micromol/L induced necrotic cell death within 24 hours, with no changes characteristic of apoptosis. To determine whether reactive oxygen species play a role in daunorubicin-mediated apoptosis, we monitored the generation of hydrogen peroxide with dichlorofluorescein (DCF). However, daunorubicin (1 micromol/L) did not increase DCF fluorescence, nor were the antioxidants N-acetylcysteine or the combination of alpha-tocopherol and ascorbic acid able to prevent apoptosis. In contrast, dexrazoxane (10 micromol/L), known clinically to limit anthracycline cardiac toxicity, prevented daunorubicin-induced myocyte apoptosis, but not necrosis induced by higher anthracycline concentrations (>/=10 micromol/L). The antiapoptotic action of dexrazoxane was mimicked by the superoxide-dismutase mimetic porphyrin manganese(II/III)tetrakis(1-methyl-4-peridyl)porphyrin (50 micromol/L). The recognition that anthracycline-induced cardiac myocyte apoptosis, perhaps mediated by superoxide anion generation, occurs at concentrations well below those that result in myocyte necrosis, may aid in the design of new therapeutic strategies to limit the toxicity of these drugs.[2]
Dexrazoxane/Dexra mitigated acute DXR-induced ovarian toxicity and improved the fertility window as shown by increased fecundity, pup weight, litters size, and number of deliveries post-DXR therapy. The 1:1 Dexra:DXR dose conferred ovarian protection. Easy-to-administer Dexra may provide a timely, cost effective and safe, drug-based method for ovarian protection, particularly for prepubertal and adolescent girls for whom oocyte and embryo freezing are not viable fertility preservation options.[3]

Solubility Data


Solubility (In Vitro)
DMSO: ~60 mg/mL (~196.9 mM)
Water: ~60 mg/mL (~196.9 mM)
Ethanol: <1 mg/mL
Solubility (In Vivo) Solubility in Formulation 1: ≥ 3 mg/mL (8.79 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 30.0 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: ≥ 3 mg/mL (8.79 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 30.0 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.

Solubility in Formulation 3: ≥ 3 mg/mL (8.79 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 30.0 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly.

Solubility in Formulation 4: 130 mg/mL (381.02 mM) in PBS (add these co-solvents sequentially from left to right, and one by one), clear solution; with ultrasonication.

 (Please use freshly prepared in vivo formulations for optimal results.)